3.A.1 The ATP-binding Cassette (ABC) Superfamily

The ABC superfamily contains both uptake and efflux transport systems, and the members of these two porter groups generally cluster loosely together with just a few exceptions (Saurin et al., 1999). ATP hydrolysis without protein phosphorylation energizes transport. There are dozens of families within the ABC superfamily, and family generally correlates with substrate specificity. However there are exceptions. The high resolution X-ray structures of several ABC transporters, both uptake and efflux systems, have been determined, and specific details of the transport mechanisms have been proposed (Davidson and Maloney, 2007; Lee et al., 2007). Several multidrug resistance (MDR) transporters catalyze lipid, lipopolysaccharide, and/or lipoprotein export. This can occur by a 'flip-flop' mechanism or a 'projection' mechanism (Nagao et al., 2010). Pagès et al. (2011) have described several classes of efflux pump inhibitors that counteract MDR. Known structures have been discussed by Zolnerciks et al. (2011) and by Wilkens 2015. Mechanistic models for the transport cycle have been compared (Szöllősi et al. 2017). Transporters can be modified posttranslationally by phsophorylation, ubiquitination, glycosylation and/or palmitoylation (Czuba et al. 2018). Ford and Beis 2019 reviewed  progress on structural determination of eukaryotic and bacterial ABC transporters as well as novel proposed mechanisms.  There are 48 genes encoding ABC transporters in humans (Sakamoto et al. 2019).  1,4-dihydropyridines are inhibitors of the transmembrane efflux pump ABCB1 in cancer cells, and they potentiate the inhibitory effect of clofazimine in Mycobacterium tuberculosis (Lentz et al. 2019). Phospholipids and cholesterol are inducers of cancer multidrug resistance and therapeutic targets (Kopecka et al. 2019). Multiple positions (residues) in the membrane domains of ABC transporters influence their substrate selectivities (Srikant et al. 2020). An involvement of ABC porters in cancer multidrug resistance (MDR) has been reviewed (Juan-Carlos et al. 2021). The functional dynamics (substrate transport) of ABC transporters are realized by concerted motions, such as NBD dimerization, mechanical transmission via coupling helices, and the translocation of substrates through TMDs, which are induced by the binding and/or hydrolysis of ATP molecules and substrates (Furuta 2021). Automatic algorithms have been derived that permit the large-scale identification of the solute binding protein (SBP) repertoire in proteomes (Ortega et al. 2022).  An alternating access mechanism for many bacterial and mammalian ABC-type multidrug resistance (MDR) transporters has been reviewed (Badiee et al. 2023). It has been proposed that ATP binding cassette transporters  evolved roles in herbicide resistance through exaptation (Caygill and Dolan 2023).

Switch and constant contact models have been presented (George and Jones, 2012).  The prevailing paradigm for the ABC transport mechanism is the Switch Model, in which the nucleotide binding domains are proposed to dimerise upon binding of two ATP molecules, and thence dissociate upon sequential hydrolysis of the ATP. This idea appears consistent with crystal structures of both isolated subunits and whole transporters, as well as with the biochemical data. Nonetheless, an alternative Constant Contact Model has been proposed, in which the nucleotide binding domains do not fully dissociate, and ATP hydrolysis occurs alternately at each of the two active sites (Jones and George 2012).  In this model, one of the sites remains closed and contains an occluded nucleotide at all times. The cassettes remain in contact, and the active sites swing open in an alternately seesawing motion. Whilst the concept of NBD association/dissociation in the Switch Model is naturally compatible with a single alternating-access channel, the asymmetric functioning proposed by the Constant Contact model suggests an alternating or reciprocating function in the TMDs. A new model for the function of ABC transporters has been proposed by Jones and George 2014 in which the sequence of ATP binding, hydrolysis, and product release in each active site is directly coupled to the analogous sequence of substrate binding, translocation and release in one of two functionally separate substrate translocation pathways. Each translocation pathway functions 180 degrees out of phase. 

The solute-binding proteins of oceanic SAR11 bacteria have extremely high binding affinities (dissociation constants near 20 pM) and high binding specificities, revealing molecular mechanisms of oligotrophic adaptation (Clifton et al. 2024). These functional data have also uncovered new carbon sources for SAR11 bacteria and enable accurate biogeographical analysis of SAR11 substrate uptake capabilities throughout the ocean. This study provides a comprehensive view of the substrate uptake capabilities of ubiquitous marine bacteria, providing a necessary foundation for understanding their contribution to assimilation of dissolved organic matter in marine ecosystems (Clifton et al. 2024).

As noted above, the porters of the ABC superfamily consist of two integral membrane domains/proteins and two cytoplasmic domains/proteins. The uptake systems (but not the efflux systems) usually possess extracytoplasmic solute-binding receptors (one or more per system) which in Gram-negative bacteria are found in the periplasm, and in Gram-positive bacteria is present either as a lipoprotein, tethered to the external surface of the cytoplasmic membrane, or as a cell surface-associated protein, bound to the external membrane surface via electrostatic interactions. For those systems with two or more extracytoplasmic solute binding receptors, the receptors may interact in a cooperative fashion (Biemans-Oldehinkel and Poolman, 2003). These binding proteins fall into six phylogenetic clusters (Berntsson et al., 2010). Both the integral membrane channel constituent(s) and the cytoplasmic ATP-hydrolyzing constituent(s) may be present as homodimers or heterodimers. Two families of ABC transporters have members in which one or two receptors are fused to either the N- or C-terminus of the translocating membrane protein. This suggests that two or even four substrate-binding sites may function in the complex. Possibly multiple receptors in proximity to the translocator enhances the transport rate. Multiple receptors may also broaden the substrate specificity of the system (van der Heide and Poolman, 2002). These systems with covalent receptor domains linked to the transmembrane translocators are found in the PAAT family (TC #3.A.1.3) and the QAT family (TC #3.A.1.12) (van der Heide and Poolman, 2002). Some high affinity ABC uptake systems specific for vitamins, minerals and other small molecules, called ECF systems, lack an extracytoplasm receptor and function by a mechanisms as discussed by Slotboom 2014. Both ECF (type I) and binding protein receptor-dependent uptake systems have been found in eukaryotes, especially in chloroplasts of algae and lower plants (Choi and Ford 2021). The diversity of ABC transporter genes across the phylum Nematoda has been examined (Langeland et al. 2021). Most of the ABCB and ABCG subfamily members are actively involved in heavy metal responses, and the functions of specific ABC transporters in Cd2+ and Hg2+ stress responses is likely in soybean (Glycine max L.) (Naaz et al. 2023). Some ABCG transporters in plants are likely to transport plant terpenes (Salgado et al. 2023).

ABC transporters always have two nucleotide binding domains (NBDs or NBSs). ATP-bound NBDs dimerize in a head-to-tail arrangement, with two nucleotides sandwiched at the dimer interface. Upon the binding of ATP molecules to nucleotide binding domains (NBDs), ATP-binding cassette (ABC) exporters undergo a conformational transition from an inward-facing (IF) to an outward-facing (OF) state via chemo-mechanical coupling. The ATP binding energy is converted into distortion energy of several transmembrane helices (Arai et al. 2017). Both NBDs contribute residues to each of the two nucleotide-binding sites (NBSs) in the dimer. The prototypical NBD MJ0796 from Methanocaldococcus jannaschii forms ATP-bound dimers that dissociate completely following hydrolysis of one of the two bound ATP molecules.  ATP hydrolysis at one nucleotide-binding site drives NBD dissociation, but two binding sites are required to form the ATP-sandwich NBD dimer necessary for hydrolysis (Zoghbi and Altenberg 2013). Single versus dimeric subunit ATPase have been discussed (Ford and Hellmich 2020). Transporters with two active NBSs are called canonical transporters, while ABC exporters from eukaryotic organisms frequently have a degenerate NBS1 containing noncanonical residues that strongly impair ATP hydrolysis. The latter have been reviewed (Stockner et al. 2020). A summary of the structures available and an overview of recent structure-based studies have been discussed, while the artificial intelligence (AI)-based predictions from AlphaFold-2 are considered (Huang and Ecker 2023). Eight families of MDR effux pump inhibitors, active against Escherichia coli or Pseudomonas aeruginosa have been described and reviewed (Compagne et al. 2023).  A total of 159 ABC genes were identified in the legume, Cajanus cajan, that belong to eight canonical classes, CcABCA to CcABCG and CcABCI (Mall et al. 2023). Different cis-acting regulatory elements like stress, hormone, and cellular responsive were identified, and expression profiling of CcABC genes at various developmental stages of different anatomical tissues was performed.

Kinases can affect the activities of ABC transporters as do protein-protein interactions. Crawford et al. 2018 reviewed the effects of such interactions on the ABC transporters ABCB1, ABCB11, ABCC1, ABCC4, and ABCG2 of humans, showing how kinases and protein-protein interactions regulate these transporters. Yeast have all of these types of ABC transporters, and a comprehensive overview of these proteins of the drug-resistant human fungal pathogen, Candida glabrata (Kumari et al. 2018). A comprehensive review of the classes of efflux pump inhibitors from various sources, highlighting their structure-activity relationships, which can be useful for medicinal chemists in the pursuit of novel efflux pump inhibitors has appeared (Durães et al. 2018). Protein expression and functional relevance of various primary and secondary drug efflux and uptake porters at the Blood-Brain Barrier of human brain and glioblastoma have been investigated (Bao et al. 2019). ABC transporter gene expression and mutational status not surprisingly affect survival rates of cancer patients (Kadioglu et al. 2020). Copper N-(2-hydroxy acetophenone) glycinate (CuNG) exerted different modulatory activities towards ABC-transporter-expressing cells. While CuNG-mediated ABC-transporter inhibition may improve tumor chemotherapy, CuNG-mediated enhanced ABC-transport may be a new strategy to ameliorate inflammatory diseases associated with decreased ABC-transporter expression as in ulcerative colitis (Hartinger et al. 2023).

Acar et al. 2020 investigated the allosteric networks of three representative ABC transporters using a hybrid molecular simulations approach validated by experiments. Each of the three transporters uses a different allosteric network. In the constitutive B12 importer BtuCD, ATP binding is the main driver of allostery and docking/undocking of the substrate-binding protein (SBP), which is the driven event. The allosteric signal originates at the cytoplasmic side of the membrane before propagating to the extracellular side. In the substrate-controlled maltose transporter, the SBP is the main driver of allostery, ATP binding is the driven event, and the allosteric signal propagates from the extracellular to the cytoplasmic side of the membrane. In the lipid flippase PglK, a cyclic crosstalk between ATP and substrate binding underlies allostery. These results demonstrate that speciation of biological functions may arise from variations in allosteric connectivity (Acar et al. 2020).  RNA thermometers are temperature-sensing non-coding RNAs that regulate the expression of downstream genes, ABC porter genes are among them (Tong et al. 2024).

Using mass spectrometry, Al-Majdoub et al. 2020 identified and quantified the amounts of 32 (out of 48) ABC transporters in several human tissues. ABCD3 was the most abundant in liver, whereas ABCA8, ABCB2/TAP1, and ABCE1 were detected in all tissues.They created an atlas which revealed that ABCB2/TAP1 may have TAP2-independent functions in the brain, and that livers from biliary atresia patients have quite different ABC-transporter profiles than controls. Nearly a third to half of patients with noninfectious uveitis (NIU) fail to achieve control with immunomodulatory therapy due to MDR (Tagirasa et al. 2020). Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation (ROHHAD) syndrome is a rare neurocristopathy.  ATPase transmembrane transporters, acetylglucosaminyltransferases, and phagocytic vesicle membrane proteins may contribute to the ROHHAD phenotype (Victor et al. 2023).  Mutations in ABC genes cause 21 monogenic diseases, and polymorphisms in these genes are associated with susceptibility to complex diseases (Duvivier et al. 2024). ABC transporters also play a major role in drug bioavailability, and they mediate multidrug resistance in cancer. At least 13 ABC transporters were shown to be involved in drug resistance in vitro.

The involvement of ABC transporters in transport of secondary metabolites in medicinal plants has been reviewed (Liu et al. 2021). 49 ABC transporter genes have been divided into eight subfamilies (ABCA-ABCH), including seven ABCAs, seven ABCBs, 10 ABCCs, two ABCDs, one ABCE, three ABCFs, 16 ABCGs, and three ABCHs according to phylogenetic analysis in Zeugodacus cucurbitae, a highly destructive insect pest of cucurbitaceous and other related crops (Xu et al. 2021). Transporter-mediated natural product-drug interactions have been reviewed for transporters in several different families (Bi et al. 2022). The new AlphaFold protein structure prediction tool predicts a new type of transmembrane domain fold within the ABC transporters that is associated with cation exporters of bacteria and plants (Younus et al. 2022).

Plant MRPs, or GS-X pumps (glutathione-conjugate or multispecific organic anion Mg2+-ATPases,  participate in the transport of exogenous and endogenous amphipathic anions and glutathionated compounds, usually from the cytosol into the vacuole. Encoded by a multigene family and possessing a unique domain organization, the types of processes that likely converge and depend on plant MRPs include herbicide detoxification, cell pigmentation, the alleviation of oxidative damage, and the storage of antimicrobial compounds. Additional functional capabilities might include channel regulation of activity and/or the transport of heavy metal chelates (Rea et al. 1998).

The TC system of classification uses the integral membrane proteins, not the energy coupling proteins, receptor, or auxiliary subunits to classify the system into families (Saier, 1994; 2000). The exception to this rule was the ABC superfamily which by engrained tradition was classified based on the use of an ATP-binding cassette (ABC) ATPase for energy coupling before the TC system was designed. Since then it has become known that the membrane proteins of ABC export systems fall into three evolutionarily distinct families that followed three different pathways of origin (Wang et al., 2009). These have been designated ABC1, ABC2 and ABC3. ABC1 porters arose by triplication of a primordial 2 TMS element; ABC2 porters arose by duplication of a 3 TMS element, and ABC3 porters arose from a 4 TMS precursor that either remained as two 4 TMS proteins (a homo or hetero dimer) or internally duplicated to give 8 or 10 proteins, the extra two appear to be in the center between the two 4 TMS repeat units (Khwaja et al. 2005). The ABC functional superfamily therefore consists of at least three true superfamilies. The ABC subfamiies or clusters that belong to each of these three superfamilies are listed below. In addition, the LPS effluxing transport system family with TC# 3.A.152 may be unrelated to the other three exporter superfamilies (Thomas et al. 2020). Based on 3-D structures, there may also be three types of integral membrane ABC uptake systems, bringing the total to 7 types (Thomas et al. 2020).

Energy-coupling factor (ECF)-type ABC importers appear to fulfil important roles in both algae and plants where they form the ABCI sub-family. Choi and Ford 2021 discussed whether bacterial Type I and Type II ABC importers are present in extant eukaryotes. Type I importers exist in algae and the liverwort family of primitive non-vascular plants, but not in higher plants. The existence of eukaryotic Type II importers is also supported: a transmembrane protein homologous to vitamin B12 import system transmembrane protein (BtuC), hemin transport system transmembrane protein (HmuU) and high-affinity zinc uptake system membrane protein (ZnuB) is encoded within the Cyanophora paradoxa genome. This last protein has homologs within the genomes of red algae. Furthermore, its candidate nucleotide-binding domain (NBD) shows closest similarity to other bacterial Type II importer NBDs such as BtuD. Functional studies suggest that Type I importers have roles in maintaining sulphate levels in the chloroplast, while Type II importers probably act as importers of Mn2+ or Zn2+ , as inferred by comparisons with bacterial homologs. Possible explanations for the presence of these transporters in simple plants, but not in other eukaryotic organisms, are considered (Choi and Ford 2021). In order to utilise the existing nomenclature for eukaryotic ABC proteins, it was proposed that eukaryotic Type I and II importers be classified as ABCJ and ABCK transporters, respectively.

ABC1:
3.A.1.106 The Lipid Exporter (LipidE) Family
3.A.1.108 The β-Glucan Exporter (GlucanE) Family
3.A.1.109 The Protein-1 Exporter (Prot1E) Family
3.A.1.110 The Protein-2 Exporter (Prot2E) Family
3.A.1.111 The Peptide-1 Exporter (Pep1E) Family
3.A.1.112 The Peptide-2 Exporter (Pep2E) Family
3.A.1.113 The Peptide-3 Exporter (Pep3E) Family
3.A.1.117 The Drug Exporter-2 (DrugE2) Family
3.A.1.118 The Microcin J25 Exporter (McjD) Family
3.A.1.119 The Drug/Siderophore Exporter-3 (DrugE3) Family
3.A.1.123 The Peptide-4 Exporter (Pep4E) Family
3.A.1.127 The AmfS Peptide Exporter (AmfS-E) Family
3.A.1.129 The CydDC Cysteine Exporter (CydDC-E) Family
3.A.1.135 The Drug Exporter-4 (DrugE4) Family
3.A.1.139 The UDP-Glucose Exporter (U-GlcE) Family (UPF0014 Family)
3.A.1.201 The Multidrug Resistance Exporter (MDR) Family (ABCB)
3.A.1.202 The Cystic Fibrosis Transmembrane Conductance Exporter (CFTR) Family (ABCC)
3.A.1.203 The Peroxysomal Fatty Acyl CoA Transporter (P-FAT) Family (ABCD)
3.A.1.206 The a-Factor Sex Pheromone Exporter (STE) Family (ABCB)
3.A.1.208 The Drug Conjugate Transporter (DCT) Family (ABCC) (Dębska et al., 2011)
3.A.1.209 The MHC Peptide Transporter (TAP) Family (ABCB)
3.A.1.210 The Heavy Metal Transporter (HMT) Family (ABCB)
3.A.1.212 The Mitochondrial Peptide Exporter (MPE) Family (ABCB)
3.A.1.21   The Siderophore-Fe3+ Uptake Transporter (SIUT) Family

ABC2:
3.A.1.101 The Capsular Polysaccharide Exporter (CPSE) Family
3.A.1.102 The Lipooligosaccharide Exporter (LOSE) Family
3.A.1.103 The Lipopolysaccharide Exporter (LPSE) Family
3.A.1.104 The Teichoic Acid Exporter (TAE) Family
3.A.1.105 The Drug Exporter-1 (DrugE1) Family
3.A.1.107 The Putative Heme Exporter (HemeE) Family
3.A.1.115 The Na+ Exporter (NatE) Family
3.A.1.116 The Microcin B17 Exporter (McbE) Family
3.A.1.124 The 3-component Peptide-5 Exporter (Pep5E) Family
3.A.1.126 The β-Exotoxin I Exporter (βETE) Family
3.A.1.128 The SkfA Peptide Exporter (SkfA-E) Family
3.A.1.130 The Multidrug/Hemolysin Exporter (MHE) Family
3.A.1.131 The Bacitracin Resistance (Bcr) Family
3.A.1.132 The Gliding Motility ABC Transporter (Gld) Family
3.A.1.133 The Peptide-6 Exporter (Pep6E) Family
3.A.1.138 The Unknown ABC-2-type (ABC2-1) Family
3.A.1.141 The Ethyl Viologen Exporter (EVE) Family (DUF990 Family)
3.A.1.142 The Glycolipid Flippase (G.L.Flippase) Family
3.A.1.143 The Exoprotein Secretion System (EcsAB(C))
3.A.1.144:  Functionally Uncharacterized ABC2-1 (ABC2-1) Family
3.A.1.145:  Peptidase Fused Functionally Uncharacterized ABC2-2 (ABC2-2) Family
3.A.1.146:  The actinorhodin (ACT) and undecylprodigiosin (RED) exporter (ARE) family
3.A.1.147:  Functionally Uncharacterized ABC2-2 (ABC2-2) Family
3.A.1.148:  Functionally Uncharacterized ABC2-3 (ABC2-3) Family
3.A.1.149:  Functionally Uncharacterized ABC2-4 (ABC2-4) Family
3.A.1.150:  Functionally Uncharacterized ABC2-5 (ABC2-5) Family
3.A.1.151:  Functionally Uncharacterized ABC2-6 (ABC2-6) Family
3.A.1.152:  The lipopolysaccharide export (LptBFG) Family
3.A.1.204 The Eye Pigment Precursor Transporter (EPP) Family (ABCG)
3.A.1.205 The Pleiotropic Drug Resistance (PDR) Family (ABCG)
3.A.1.211 The Cholesterol/Phospholipid/Retinal (CPR) Flippase Family (ABCA)
9.B.74      The Phage Infection Protein (PIP) Family
 

ABC3:
3.A.1.114 The Probable Glycolipid Exporter (DevE) Family
3.A.1.122 The Macrolide Exporter (MacB) Family
3.A.1.125 The Lipoprotein Translocase (LPT) Family
3.A.1.134 The Peptide-7 Exporter (Pep7E) Family
3.A.1.136 The Uncharacterized ABC-3-type (U-ABC3-1) Family
3.A.1.137 The Uncharacterized ABC-3-type (U-ABC3-2) Family
3.A.1.140 The FtsX/FtsE Septation (FtsX/FtsE) Family
3.A.1.207 The Eukaryotic ABC3 (E-ABC3) Family

Uptake systems (TC#s 3.A.1.1 - 3.A.1.34) may be largely of the ABC2 type, but they have undergone extensive sequence and topological diversification. They even have different 3-dimensional structural differences, suggesting that they may have different origins. The only exception is ABC family 3.A.1.21, the Siderophore-Fe3+ Uptake Transporter (SIUT) Family which is clearly of the ABC1 type. It has no extracytoplasmic receptor as well. 

ABC importers have been divided into two classes: type I importers follow an alternating access mechanism driven by the presence of the substrate, while type II importers accept substrates in a nucleotide-free state, with hydrolysis driving an inward-facing conformation.  RbsABC2 (TC#3.A.1.2.1) seems to share functional traits with both type I and type II importers, as well as possessing unique features, and may employ a distinct mechanism relative to other ABC transporters (Clifton et al. 2014).

Unlike most uptake systems which have one or two functionally equivalent membrane subunits that form a homo- or hetero-dimer and an extracytoplasmic receptor, a subset of these porters have two functionally dissimilar membrane subunits, called S (substrate recognition) and T (energy transducer) that are very divergent in sequence, and they lack extracytoplasmic receptors (Erkens et al. 2012). This group of ABC porters represent a subfamily within the ABC uptake systems. This subset, called Energy-coupling Factor 'ECF' porters, includes:

ECF:
3.A.1.18 The Cobalt Uptake Transporter (CoT) Family 
3.A.1.22 The Nickel Uptake Transporter (NiT) Family
3.A.1.23 The Nickel/Cobalt Uptake Transporter (NiCoT) Family
3.A.1.25 The Biotin Uptake Transporter (BioMNY) Family
3.A.1.26 The Putative Thiamine Uptake Transporter (ThiW) Family
3.A.1.28 The Queuosine (Queuosine) Family
3.A.1.29 The Methionine Precursor (Met-P) Family
3.A.1.30 The Thiamin Precursor (Thi-P) Family
3.A.1.31 The Unknown-ABC1 (U-ABC1) Family
3.A.1.32 The Cobalamin Precursor (B12-P) Family
3.A.1.33 The Methylthioadenosine (MTA) Family
S-subunits are homologous to:
2.A.87 The Prokaryotic Riboflavin Transporter (P-RFT) Family
2.A.88 The Vitamin Uptake Transporter (VUT or ECF) Family

Karpowich and Wang (2013) characterized the ECF transporters from Thermotoga maritima and Streptococcus thermophilus and determined a subunit stoichiometry of 2S:2T:1A:1A'.  They concluded that S subunits for different substrates can be incorporated into the same transporter complex simultaneously. In the crystal structure of the A-A' heterodimer, each subunit contains a novel motif called the Q-helix that plays a role in subunit coupling with the T subunits. A mechanism for coupling ATP binding and hydrolysis to transmembrane transport by ECF transporters was proposed. ECF porters mediate the uptake of micronutrients in prokaryotes, and consist of two ATP-binding-cassette family ATPases, a transmembrane coupling protein (T component) and a substrate-binding membrane protein (S component), as noted above. ECF transporters for Co2+ and Ni2+ ions have one or two additional proteins with extracytoplasmic regions. Homologs of T components with predicted localization in plastids are widespread in plants, but S components in eukaryotes are rare and restricted to biotin-specific variants (Finkenwirth and Eitinger 2019). Apart from a potential contribution to the export of flavins to serve the assembly of extracytoplasmic electron transfer chains, ECF transporters function as importers.

The maltose import transporter is composed of two TM subunits, MalF and MalG, and two subunits of a cytoplasmic ATPase, MalK. Like many uptake systems in Gram-negative bacteria, the periplasmic maltose-binding protein (MBP), is required to stimulate the ATPase activity of the transporter. In the absence of maltose, MBP exists in equilibrium between an open and closed conformation, and binding of maltose stabilizes the closed conformation. Two structures of MalFGK2 have been determined by x-ray crystallography. In the absence of MBP, MalFGK2 forms an inward-facing conformation with the TM maltose-binding site exposed to the cytoplasm. An outward-facing conformation, crystallized in complex with open MBP and ATP, shows that closure of the NBDs of MalK is concomitant with transfer of maltose from MBP to the TM subunits. These structures capture two states in the transport cycle: The inward-facing conformation represents the resting state where the transporter has a very low ATPase activity, and the outward-facing conformation represents a catalytic intermediate where ATP is poised for hydrolysis. Because MBP stimulates ATP hydrolysis and initiates the transport process, it must interact with the resting state conformation to form a 'pretranslocation' complex that is metastable in order to advance to the outward-facing conformation in the presence of ATP (25). Oldham and Chen (2011) presented the crystal structure of the initial complex formed between closed MBP and MalFGK2. As an essential intermediate between the inward- and outward-facing conformations, this structure suggests a mechanism by which substrate bound on the periplasmic surface influences the conformation of the NBDs at the intracellular surface. The same investigators suggested that ABC transporters catalyze ATP hydrolysis via a general base mechanism (Oldham and Chen, 2011).

The homodimeric LmrA drug efflux pump (TC #3.A.1.117.1) of Lactococcus lactis appears to function by an alternating site (half of sites) type mechanism. In many of these porters, the various domains are fused in a variety of combinations. Uptake porters generally have their constituents as distinct polypeptide chains, while efflux systems usually have them fused. ABC-type uptake systems have not been identified in eukaryotes, but ABC-type efflux systems abound in both prokaryotes and eukaryotes. The eukaryotic efflux systems often have the four domains (two cytoplasmic domains and two integral membrane domains) fused into either one or two polypeptide chains. The integral membrane porter domains each usually possesses 5 (uptake) or 6 (efflux) transmembrane spanners, but exceptions exist. For example, the MntB protein (TC #3.A.1.15.1) exhibits 9 established TMSs. The 3-dimensional structure of the E. coli MsbA protein (TC #3.A.1.106.1) has been solved to a resolution of 3.7 Å (Ward et al., 2007), that of the Staphylococcus aureus Sav1866 protein (TC #3.A.1.106.2) has been solved to a resolution of 3.0 Å (Dawson and Locher, 2006), that of the Archaeoglobus fulgidus ModABC complex has been solved at 3.1 Å resolution (Hollenstein et al., 2007), that of the E. coli BtuCDF Vitamin B12 transporter was solved at 2.6 Å resolution (Hvorup et al., 2007), and the maltose transporter has been solved at 2.8 Å resolution (Oldham et al., 2007). These structures are very different, but the two transmembrane domains form a single barrel 5-6 nm in diameter and about 5 nm deep with an entral pore open either to the external or internal surface spanning much of the membrane (Rosenberg et al., 2003). A model has been proposed allowing the channel to open up to the lipid bilayer. A half of sites model in which the two nucleotide binding domains interact in a fashion controlled by substrate binding has also been proposed (Hou et al., 2003; Loo et al., 2003).

Hollenstein et al. (2007) presented the 3.1 Å crystal structure of a putative molybdate transporter (ModB2C2) from Archaeoglobus fulgidus in complex with its binding protein (ModA). Twelve transmembrane helices of the ModB subunits provide an inward-facing conformation, with a closed gate near the external membrane boundary. The ATP-hydrolyzing ModC subunits reveal a nucleotide-free, open conformation, whereas the attached binding protein aligns the substrate-binding cleft with the entrance to the presumed translocation pathway. Structural comparison of ModB2C2A with Sav1866 suggests a common alternating access and release mechanism, with binding of ATP promoting an outward-facing conformation and dissociation of the hydrolysis products promoting an inward-facing conformation. ATP hydrolysis at one of the two sites in ABC transporters initiates transport-related conformational transitions (Gyimesi et al., 2011).

Smriti et al., 2009 mapped residues proximal to the daunorubicin (DNR)-binding site in MsbA (TC#3.A.1.106.1) using site-specific, ATP-dependent quenching of DNR intrinsic fluorescence by spin labels. In the nucleotide-free MsbA intermediate, DNR-binding residues cluster at the cytoplasmic end of helices 3 and 6 at a site accessible from the membrane/water interface and extending into an aqueous chamber formed at the interface between the two transmembrane domains. Binding of a nonhydrolyzable ATP analog inverts the transporter to an outward-facing conformation. DNR may thus enter near an elbow helix parallel to the water/membrane interface, partitioning into the open chamber, and then translocating toward the periplasm upon ATP binding.

The turnover rates of some transporters are inhibited by their substrates in a process termed trans-inhibition. Gerber et al. (2008) presented the crystal structure of a molybdate/tungstate ABC transporter (ModBC) from Methanosarcina acetivorans in a trans-inhibited state. The regulatory domains of the nucleotide-binding subunits proved to be in close contact, providing two oxyanion binding pockets at the shared interface. By specifically binding to these pockets, molybdate or tungstate prevent adenosine triphosphatase activity and lock the transporter in an inward-facing conformation, with the catalytic motifs of the nucleotide-binding domains separated. This allosteric effect prevents the transporter from switching between the inward-facing and the outward-facing states, thus interfering with the alternating access and release mechanism.

The cystic fibrosis transmembrane conductance regulator (CFTR; 3.A.1.202.1) is an ATP-dependent chloride channel. Jordan et al., 2008 compared CFTR protein sequences to those of ABCC4 proteins (the closest mammalian paralogs) to identify the evolutionary transition from transporter to channel activity. R352 in the sixth transmembrane helix interacts with D993 in TM9 to stabilize the open-channel state; D993 is absolutely conserved between CFTRs and ABCC4s. Thus CFTR channel activity evolved, at least in part, by converting the conformational changes associated with binding and hydrolysis of ATP, as are found in true ABC transporters, into an open permeation pathway by means of intraprotein interactions that stabilize the open state.  In general, plant ABC transport systems are more numerous than those in animals.  The maize systems have been categorized and their expression profiles have been determined (Pang et al. 2013).

The LolCDE complex of Escherichia coli (TC# 3.A.1.125.1) initiates the lipoprotein sorting to the outer membrane by catalysing their release from the inner membrane. LolC and/or LolE, membrane subunits, recognize lipoproteins anchored to the outer surface of the inner membrane, while LolD hydrolyses ATP on its inner surface. The ligand-bound LolCDE has been purified from the inner membrane in the absence of ATP (Ito et al., 2006). Liganded LolCDE represents an intermediate of the release reaction and exhibits higher affinity for ATP than the unliganded form. ATP binding to LolD weakens the interaction between LolCDE and lipoproteins and causes their dissociation in a detergent solution, while lipoprotein release from membranes requires ATP hydrolysis. A single molecule of lipoprotein is found to bind per molecule of the LolCDE complex.

The three structurally dissimilar constituents of the ABC uptake porters have generally arisen from a common ancestral porter system with minimal shuffling of constituents between/domain constituents is almost always the same. However the rates of sequence divergences differ drastically with the extracytoplasmic solute-binding receptors diverging most rapidly, the integral-membrane, channel-forming constituents diverging at an intermediate rate, and the cytoplasmic ATP-hydrolyzing constituents diverging most slowly. Thus, all ATP-hydrolyzing constituents are demonstrably homologous, but this is not true for the integral membrane constituents or the receptors. Nevertheless, clustering patterns are generally the same for all three types of proteins, and 3-dimensional structural data suggest that, in spite of their extensive sequence divergence, the extracytoplasmic solute-binding receptors are homologous to each other.

Unlike most of the known ABC transporters, ABCC1 (TC #3.A.1.208.8) has an additional membrane-spanning domain (MSD) at its amino terminus with a domain arrangement of MSD0-MSD1-NBD1-MSD2-NBD2. The additional MSD0 domain consists of five putative transmembrane segments with a predicted extracellular amino terminus. It has a U-shaped folding with the bottom of the U-structure facing cytoplasm and both ends in extracellular space. This U-shaped amino terminus probably functions as a gate to regulate the drug transport activity of human ABCC1 (Chen et al., 2006).

Polar lipid trafficking is essential in eukaryotic cells as membranes of lipid assembly are often distinct from final destination membranes. A striking example is the biogenesis of the photosynthetic membranes (thylakoids) in plastids of plants. Lipid biosynthetic enzymes at the endoplasmic reticulum and the inner and outer plastid envelope membranes are involved. This compartmentalization requires extensive lipid trafficking. Mutants of Arabidopsis disrupt the incorporation of endoplasmic reticulum-derived lipid precursors into thylakoid lipids. Two proteins affected in two of these mutants, trigalactosyldiacylglycerol 1 (TGD1) and TGD2, encode the permease and substrate binding component, respectively, of a proposed lipid translocator at the inner chloroplast envelope membrane. A third protein, TGD3, a small ABC-type ATPase, energizer transport. As in the tgd1 and tgd2 mutants, triacylglycerols and trigalactolipids accumulate in a tgd3 mutant. The TGD3 protein shows basal ATPase activity and is localized inside the chloroplast beyond the inner chloroplast envelope membrane. Proteins orthologous to TGD1, -2, and -3 are predicted to be present in Gram-negative bacteria, and the respective genes are organized in operons suggesting a common biochemical role for the gene products. The Tgd1,2,3 system (TC#3.A.1.27.2) probably transfers ER-derived lipids to the thylakoid membrane (Lu et al., 2007). It is one of the few known eukaryotic uptake systems.

Rodionov et al., 2009 identified 21 families of these substrate capture proteins, each with a different specificity predicted by genome context analyses. Roughly half of the substrate capture proteins (335 cases) examined by Rodionov et al., 2009 have a dedicated energizing module, but in 459 cases distributed among almost 100 gram-positive bacteria, different and unrelated substrate capture proteins share the same energy-coupling module. The shared use of energy-coupling modules was experimentally confirmed for folate, thiamine, and riboflavin transporters. Rodionov et al., 2009 proposed the name energy-coupling factor transporters for the new class of putative ABC membrane transporters. These membrane proteins are homologues to ABC-2 exporters. When evidence is minimal for association with an ABC-type ATP-hydrolyzing subunit, these porters are placed in category 2.A (secondary carriers; e.g., 2.A.88).

Canonical ABC importers play important roles in cell integrity, environmental stresses, cell-to-cell communication, cell differentiation and pathogenicity. An ABC sub-superfamily of micronutrient importers, the 'energy-coupling factor' (ECF) transporters, use ABC ATPases. Fundamental differences between tranditional ABC and ECF porters include the modular architecture and the independence of ECF systems of extracytoplasmic solute-binding proteins. Eitinger et al. (2011) review the roles of both types of transporters in diverse physiological processes including pathogenesis. They also point out the differences and similarities in modular assembly and their traits.

The uptake porters of the ABC superfamily and of the vitamin/small molecule transporters described by Rodionov et al., 2009 are homologous to the porters in the VUT family (2.A.88). In fact, studies indicated that all uptake porters of the ABC superfamily are of the ABC2 type (Zheng et al. 2013). When evidence suggests that homologous membrane transport proteins of the ABC2 type couple transport to ATP hydrolysis using a homologue of the ABC-type ATPases, we list these proteins in the ABC superfamily. If there is no such evidence, (e.g., experimental evidence and the occurrence of the gene for the membrane transporter protein is in an operon that lacks the ATPase and auxillary subunit) then the porter is placed into family 2.A.88.

Ter Beek et al. (2011) have determined the subunit stoichiometry and functional unit of the energy coupling factor (ECF)-type of ABC transporters (Rodionov et al., 2009). ECF transporters consist of a conserved energizing module (two peripheral ATPases and the integral membrane protein EcfT) and an integral membrane protein responsible for substrate specificity (S-component). S-components for different substrates  may associate with the same energizing module. The energizing module from Lactococcus lactis has been shown to form stable complexes with each of the eight predicted S-components found in this organism. Using light scattering, EcfT, the two ATPases (EcfA and EcfA'), and the S-component were found to be present in a stoichiometric 1:1:1:1 ratio. The complexes were reconstituted in proteoliposomes and shown to mediate ATP-dependent transport. ECF-type transporters are the smallest known ABC transporters.

Energy coupling factor (ECF) transporters are a subgroup of ATP-binding cassette (ABC) transporters involved in the uptake of vitamins and micronutrients in prokaryotes. In contrast to classical ABC importers, ECF transporters do not make use of water-soluble substrate binding proteins or domains but instead employ integral membrane proteins for substrate binding (S-components or EcfS). S-components form active translocation complexes with the ECF module, an assembly of two nucleotide-binding domains (NBDs, or EcfA) and a second transmembrane 'energy transducer' protein, EcfT. In many cases, the ECF module can interact with several different S-components that bind diverse substrates. The modular organization with exchangeable S-components on a single ECF module allows the transport of chemically different substrates via a common route. The determination of the crystal structures of the S-components that recognize thiamin and riboflavin provided clues about the mechanism of S-component exchange. Erkens et al. (2012) described current views of the transport mechanism by ECF transporters.

Some ABC exporters act on protein substrates. Export depends on the ABC transporter, a periplasmic 'adapter', the membrane fusion proteins (MFPs; 8.A.1) and an outer membrane factor (OMF; 1.B.17). Assembly of the tripartite complex can be transient and induced upon binding of the substrate to the ABC protein. Masi & Wandersman (2010) showed that in addition to the C-terminal targeting sequence, many additional signals throughout the substrate protein facilitate secretion. Interaction of the C-terminal 'targeting' signal activates the ATPase activity, causing disassembly of the complex. Thus, the proposed 'targeting' motif may signal dissociation rather than targeting (Masi & Wandersman et al., 2010). Dassa and Bouige (2001) have devised a phylogenetic/functional classification system for ABC transporters that overlaps the TC system. In their system, several of the TC families are included in single families. These reveal the closer phylogenetic relationship of TC families as follows:

Jones & George (2012) reported molecular dynamics simulations of the ATP/apo and ATP/ADP states of the bacterial ABC exporter Sav1866 (TC#3.A.1.106.2). Conformers of the active site have a canonical geometry for an in-line nucleophilic attack on the ATP γ-phosphate. The conserved glutamate immediately downstream of the Walker B motif is the catalytic base, forming a dyad with the H-loop histidine, while the Q-loop glutamine has an organising role. Each D-loop provides a coordinating residue of the attacking water, and comparison with the simulation of the ATP/ADP state suggested that via their flexibility, the D-loops modulate formation of the hydrolysis-competent state. A global switch involving a coupling helix delineates the signal transmission route by which allosteric control of ATP hydrolysis in ABC transporters is mediated. 

Binding of ATP to the nucleotide binding domains (NBDs) of ABC proteins drives the dimerization of NBDs, which, in turn, causes large conformational changes within the transmembrane domains.  NBD dimerization proceeds with a large gain of water entropy when ATP molecules bind to the NBDs. The energetic gain arising from direct NBD-NBD interactions is canceled by the dehydration penalty and the configurational-entropy loss. ATP hydrolysis induces a loss of the shape complementarity between the NBDs, which leads to the dissociation of the dimer, due to a decrease in the water-entropy gain and an increase in the configurational-entropy loss (Hayashi et al. 2014).

ABC proteins play critical roles in maintaining lipid and sterol homeostasis in higher eukaryotes. In human, several subfamily-A and -G members function as cholesterol transporters across the cellular membranes. Deficiencies of these ABC proteins can cause dyslipidemia that is associated with health conditions such as atherosclerosis, diabetes, fatty liver disease, and neurodegeneration (Xavier et al. 2018). The physiological roles of ABC cholesterol transporters have been implicated in mediating cholesterol efflux for reverse cholesterol transport and in maintaining membrane integrity for cell survival.  The membrane constituents of ABC transporters may play key roles in determining the transport substrates and the translocation mechanisms via the transmembrane domains. High resolution structures of human sterol transporter ABCG5/G8 and its functional homologs have shed light on the structural features of ABC transporters. Xavier et al. 2018 outlined what is known about ABCG cholesterol transporters, addressed key structural features in the putative sterol translocation pathway on the transmembrane domains, and concluded by proposing a mechanistic model of ABC cholesterol transporters.

ABC peptide/protein exporters are usuallly believed to export their substrates directly from the cytoplasm to the extracellular medium without a periplasmic intermediate.  However, a subgroup of systems, linked with a bacterial transglutaminase-like cysteine proteinase (BTLCP) apparently uses a two-step secretion mechanism. BTLCP-linked T1SSs transport a class of repeats-in-toxin (RTX) adhesins that are critical for biofilm formation. The prototype of this RTX adhesin group, LapA of Pseudomonas fluorescens Pf0-1, uses a novel N-terminal retention module to anchor the adhesin at the cell surface as a secretion intermediate threaded through the outer membrane-localized TolC-like protein LapE. This secretion intermediate is posttranslationally cleaved by the BTLCP family LapG protein to release LapA from its cognate T1SS pore. Thus, the secretion of LapA and related RTX adhesins into the extracellular environment appears to be a T1SS-mediated two-step process that involves a periplasmic intermediate (Smith et al. 2018).

The ABC-F family comprises an extremely diverse set of cytoplasmic ATPase proteins. All of the proteins in the ABC-F family characterized act on the ribosome and are translation factors (Ousalem et al. 2019). Their common function is ATP-dependent modulation of the stereochemistry of the peptidyl transferase center (PTC) in the ribosome coupled to changes in its global conformation and P-site tRNA binding geometry. Ousalem et al. 2019 presented an overview of the function, structure, and theories for the mechanisms-of-action of microbial ABC-F proteins including those involved in mediating resistance to ribosome-binding antibiotics.

Xiao et al. 2022 identified 87 ABC transporter genes in the genomes of Tenebrio molitor as well as those from Asbolus verrucosus (104), Hycleus cichorii (65), and Hycleus phaleratus (80). Combining these genes (336 in total) with published genes reported for Tribolium castaneum, the phylogeny of ABC transporter genes in all five Tenebrionids were analyzed. They were assigned to eight subfamilies (ABCA-H). In comparison to other species, the ABCC subfamily in this group of beetles appears expanded. The expression profiles of the T. molitor genes at different life stages and in various tissues were also investigated using transcriptomic analysis. Most of them display developmental specific expression patterns, suggesting their possible roles in development. Most of them are highly expressed in detoxification-related tissues including the gut and Malpighian tubule, from which roles in insecticide resistance were suggested. We detected specific or abundant expression of many ABC transporter genes in various tissues such as salivary gland, ovary, testis, and antenna (Xiao et al. 2022).

Dassa and Bouige (2001) summarized the protein and domain organization of each of the various family-type proteins (see Table 1). Based on the structures, a general mechanism for ABC transporters has been proposed, known as the Switch or Alternating Access Model, which holds that the NBDs are widely separated, with the TMDs and NBDs together forming an intracellular-facing inverted 'V' shape. Binding of two ATPs and the substrate to the inward-facing conformation induces a transition to an outward conformation. Despite this apparent progress, certainty around the transport mechanism for any given ABC remains elusive (Jones and George 2023). How substrate binding and transport is coupled to ATP binding and hydrolysis is not known, and there is a large body of biochemical and biophysical data that is at odds with widely separated NBDs being a functional/physiological state. An alternative Constant Contact model has been proposed in which the two NBSs operate 180 degrees out of phase with respect to ATP hydrolysis, with the NBDs remaining in close proximity throughout the transport cycle and operating in an asymmetric allosteric manner (Jones and George 2023).

 

Table 1
D&B Family TC Families
Uptake
MOI SulT, + PhoT + MolT + FeT + POPT + ThiT
OTCN QAT + NitT + TauT
ISVH VB12 + FeCT
Export
DPL Lipid E + Glucan E + Prot1E + Prot2E + Pep1E + Pep2E + Pep3E + DrugE2 + DrugE3 + MDR + CFTR + Ste + TAP + HMT + MPE
OAD CT1 + CT2
EPD EPP + PDR
DRA DrugE1 + CPR
DRI NatE
CLS CPSE + LPSE + TAE

The generalized transport reaction for ABC-type uptake systems is:

Solute (out) + ATP → Solute (in) + ADP + Pi.

The generalized transport reaction for ABC-type efflux systems is:

Substrate (in) + ATP → Substrate (out) + ADP + Pi.



This family belongs to the ArsA ATPase (ArsA) Superfamily.

 

References:

and Akiyama M. (2014). The roles of ABCA12 in epidermal lipid barrier formation and keratinocyte differentiation. Biochim Biophys Acta. 1841(3):435-40.

and Tarling EJ. (2013). Expanding roles of ABCG1 and sterol transport. Curr Opin Lipidol. 24(2):138-46.

Świątek, M.A., J. Gubbens, G. Bucca, E. Song, Y.H. Yang, E. Laing, B.G. Kim, C.P. Smith, and G.P. van Wezel. (2013). The ROK family regulator Rok7B7 pleiotropically affects xylose utilization, carbon catabolite repression, and antibiotic production in streptomyces coelicolor. J. Bacteriol. 195: 1236-1248.

Abd-Ellatef, G.E.F., E. Gazzano, A.H. El-Desoky, A.R. Hamed, J. Kopecka, D.C. Belisario, C. Costamagna, M.A. S Marie, S.R. Fahmy, A.Z. Abdel-Hamid, and C. Riganti. (2021). Glabratephrin reverses doxorubicin resistance in triple negative breast cancer by inhibiting P-glycoprotein. Pharmacol Res 175: 105975. [Epub: Ahead of Print]

Abele, R. and R. Tampé. (1999). Function of the transport complex TAP in cellular immune recognition. Biochim. Biophys. Acta. 1461: 405-419.

Abellón-Ruiz, J., S.S. Kaptan, A. Baslé, B. Claudi, D. Bumann, U. Kleinekathöfer, and B. van den Berg. (2017). Structural basis for maintenance of bacterial outer membrane lipid asymmetry. Nat Microbiol 2: 1616-1623.

Acar, B., J. Rose, B. Aykac Fas, N. Ben-Tal, O. Lewinson, and T. Haliloglu. (2020). Distinct Allosteric Networks Underlie Mechanistic Speciation of ABC Transporters. Structure. [Epub: Ahead of Print]

Adebesin, F., J.R. Widhalm, B. Boachon, F. Lefèvre, B. Pierman, J.H. Lynch, I. Alam, B. Junqueira, R. Benke, S. Ray, J.A. Porter, M. Yanagisawa, H.Y. Wetzstein, J.A. Morgan, M. Boutry, R.C. Schuurink, and N. Dudareva. (2017). Emission of volatile organic compounds from petunia flowers is facilitated by an ABC transporter. Science 356: 1386-1388.

Adorni, M.P., M. Galetti, S. La Monica, M. Incerti, A. Ruffoni, L. Elviri, I. Zanotti, B. Papotti, D. Cavallo, R. Alfieri, P.G. Petronini, and F. Bernini. (2023). A New ABCB1 Inhibitor Enhances the Anticancer Effect of Doxorubicin in Both In Vitro and In Vivo Models of NSCLC. Int J Mol Sci 24:.

Afonso de Lima, C., I.L. de Souza Bueno, S. Nunes Siqueira Vasconcelos, J.M. Sciani, A.L.T.G. Ruiz, M.A. Foglio, J.E. Carvalho, and G. Barbarini Longato. (2020). Reversal of Ovarian Cancer Cell Lines Multidrug Resistance Phenotype by the Association of Apiole with Chemotherapies. Pharmaceuticals (Basel) 13:.

Agarwal, S., D.W. Hunnicutt, and M.J. McBride. (1997). Cloning and characterization of the Flavobacterium johnsoniae (Cytophaga johnsonae) gliding motility gene, gldA. Proc. Natl. Acad. Sci. USA 94: 12139-12144.

Aguilar-Bryan, L. and J. Bryan. (1999). Molecular biology of adenosine triphosphate-sensitive potassium channels. Endocr Rev 20: 101-135.

Agustiandari, H., J. Lubelski, H.B. van den Berg van Saparoea, O.P. Kuipers, and A.J. Driessen. (2008). LmrR is a transcriptional repressor of expression of the multidrug ABC transporter LmrCD in Lactococcus lactis. J. Bacteriol. 190: 759-763.

Ahn, S., S. Moniot, M. Elias, E. Chabriere, D. Kim, and K. Scott. (2007). Structure-function relationships in a bacterial DING protein. FEBS Lett. 581: 3455-3460.

Akabas, M.H. (2000). Cystic fibrosis transmembrane conductance regulator. Structure and function of an epithelial chloride channel. J. Biol. Chem. 275: 3729-3732.

Akanuma, G., M. Ueki, M. Ishizuka, Y. Ohnishi, and S. Horinouchi. (2011). Control of aerial mycelium formation by the BldK oligopeptide ABC transporter in Streptomyces griseus. FEMS Microbiol. Lett. 315: 54-62.

Akatsuka, H., E. Kawai, K. Omori, and T. Shibatani. (1995). The three genes lipB, lipC, and lipD involved in the extracellular secretion of the Serratia marcescens lipase which lacks an N-terminal signal peptide. J. Bacteriol. 177: 6381-6389.

Akatsuka, H., R. Binet, E. Kawai, C. Wandersman, and K. Omori. (1997). Lipase secretion by bacterial hybrid ATP-binding cassette exporters: molecular recognition of the LipBCD, PrtDEF, and HasDEF exporters. J. Bacteriol. 179: 4754-4760.

Akiyama, M. (2011). The roles of ABCA12 in keratinocyte differentiation and lipid barrier formation in the epidermis. Dermatoendocrinol 3: 107-112.

Aktas, M., K.A. Jost, C. Fritz, and F. Narberhaus. (2011). Choline Uptake in Agrobacterium tumefaciens by the High-Affinity ChoXWV Transporter. J. Bacteriol. 193: 5119-5129.

Al-Majdoub, Z.M., B. Achour, N. Couto, M. Howard, Y. Elmorsi, D. Scotcher, S. Alrubia, E. El-Khateeb, A.M. Vasilogianni, N. Alohali, S. Neuhoff, L. Schmidt, A. Rostami-Hodjegan, and J. Barber. (2020). Mass Spectrometry-Based Abundance Atlas of ABC Transporters in Human Liver, Gut, Kidney, Brain, and Skin. FEBS Lett. [Epub: Ahead of Print]

Alaimo, C., I. Catrein, L. Morf, C.L. Marolda, N. Callewaert, M.A. Valvano, M.F. Feldman, and M. Aebi. (2006). Two distinct but interchangeable mechanisms for flipping of lipid-linked oligosaccharides. EMBO. J. 25: 967-976.

Alam, A., J. Kowal, E. Broude, I. Roninson, and K.P. Locher. (2019). Structural insight into substrate and inhibitor discrimination by human P-glycoprotein. Science 363: 753-756.

Alam, A., R. Küng, J. Kowal, R.A. McLeod, N. Tremp, E.V. Broude, I.B. Roninson, H. Stahlberg, and K.P. Locher. (2018). Structure of a zosuquidar and UIC2-bound human-mouse chimeric ABCB1. Proc. Natl. Acad. Sci. USA. [Epub: Ahead of Print]

Alejandro, S., Y. Lee, T. Tohge, D. Sudre, S. Osorio, J. Park, L. Bovet, Y. Lee, N. Geldner, A.R. Fernie, and E. Martinoia. (2012). AtABCG29 is a monolignol transporter involved in lignin biosynthesis. Curr. Biol. 22: 1207-1212.

Alenbawi, J., Y.A. Al-Sarraj, U.I. Umlai, A. Kadhi, N.N. Hendi, G. Nemer, and O.M.E. Albagha. (2024). Genome-wide association study and meta-analysis of phytosterols identifies a novel locus for serum levels of campesterol. Hum Genomics 18: 85.

Aleo, M.F., F. Morandini, F. Bettoni, R. Giuliani, F. Rovetta, N. Steimberg, P. Apostoli, G. Parrinello, and G. Mazzoleni. (2005). Endogenous thiols and MRP transporters contribute to Hg2+ efflux in HgCl2-treated tubular MDCK cells. Toxicology 206: 137-151.

Alhumaidi, M., L.M. Nentwig, H. Rahman, L. Schmitt, A. Rudrow, A. Harris, C. Dillon, L. Restrepo, E. Lamping, N. Arya, S.V. Ambudkar, J.S. Choy, and J. Golin. (2022). Residues forming the gating regions of asymmetric multidrug transporter Pdr5 also play roles in conformational switching and protein folding. J. Biol. Chem. 102689. [Epub: Ahead of Print]

Aller, S.G., J. Yu, A. Ward, Y. Weng, S. Chittaboina, R. Zhuo, P.M. Harrell, Y.T. Trinh, Q. Zhang, I.L. Urbatsch, and G. Chang. (2009). Structure of P-glycoprotein reveals a molecular basis for poly-specific drug binding. Science 323: 1718-1722.

Alloing, G., I. Travers, B. Sagot, D. Le Rudulier, and L. Dupont. (2006). Proline betaine uptake in Sinorhizobium meliloti: Characterization of Prb, an opp-like ABC transporter regulated by both proline betaine and salinity stress. J. Bacteriol. 188: 6308-6317.

Alvarez, F.J., C. Orelle, Y. Huang, R. Bajaj, R.M. Everly, C.S. Klug, and A.L. Davidson. (2015). Full engagement of liganded maltose-binding protein stabilizes a semi-open ATP-binding cassette dimer in the maltose transporter. Mol. Microbiol. [Epub: Ahead of Print]

Ammendola, S., P. Pasquali, C. Pistoia, P. Petrucci, P. Petrarca, G. Rotilio, and A. Battistoni. (2007). High-affinity Zn2+ uptake system ZnuABC is required for bacterial zinc homeostasis in intracellular environments and contributes to the virulence of Salmonella enterica. Infect. Immun. 75: 5867-5876.

Anantharaman, V. and L. Aravind. (2002). The PRC-barrel: a widespread, conserved domain shared by photosynthetic reaction center subunits and proteins of RNA metabolism. Genome Biol 3: RESEARCH0061.

Andersen, J.M., R. Barrangou, M.A. Hachem, S.J. Lahtinen, Y.J. Goh, B. Svensson, and T.R. Klaenhammer. (2012). Transcriptional analysis of prebiotic uptake and catabolism by Lactobacillus acidophilus NCFM. PLoS One 7: e44409.

Anderson, D.S., P. Adhikari, A.J. Nowalk, C.Y. Chen, and T.A. Mietzner. (2004). The hFbpABC transporter from Haemophilus influenzae functions as a binding-protein-dependent ABC transporter with high specificity and affinity for ferric iron. J. Bacteriol. 186: 6220-6229.

Anderson, L.N., P.K. Koech, A.E. Plymale, E.V. Landorf, A. Konopka, F.R. Collart, M.S. Lipton, M.F. Romine, and A.T. Wright. (2015). Live Cell Discovery of Microbial Vitamin Transport and Enzyme-Cofactor Interactions. ACS Chem Biol. [Epub: Ahead of Print]

Andrade, A.C., G. Del Sorbo, J.G. Van Nistelrooy, and M.A. Waard. (2000). The ABC transporter AtrB from Aspergillus nidulans mediates resistance to all major classes of fungicides and some natural toxic compounds. Microbiology 146(Pt8): 1987-1997.

Andreesen, J.R. and K. Makdessi. (2008). Tungsten, the surprisingly positively acting heavy metal element for prokaryotes. Ann. N.Y. Acad. Sci. 1125: 215-229.

Andreoletti, P., Q. Raas, C. Gondcaille, M. Cherkaoui-Malki, D. Trompier, and S. Savary. (2017). Predictive Structure and Topology of Peroxisomal ATP-Binding Cassette (ABC) Transporters. Int J Mol Sci 18:.

Ángeles-Arvizu, A., S. Enriquez-Flores, A. Jiménez-Gutiérrez, A. Pérez-Rangel, J.P. Luna-Arias, A. Castillo-Romero, J.M. Hernández, and G. León-Avila. (2021). MDR1 protein (ABC-C1) Over Expression in Giardia Intestinalis Incubated with Albendazole and Nitazoxanide. Acta Parasitol. [Epub: Ahead of Print]

Angelis, I., V. Moussis, D.C. Tsoukatos, and V. Tsikaris. (2021). Multidrug Resistance Protein 4 (MRP4/ABCC4): A Suspected Efflux Transporter for Human''s Platelet Activation. Protein Pept Lett. [Epub: Ahead of Print]

Angerer, A., B. Klupp, and V. Braun. (1992). Iron transport systems of Serratia marcescens. J. Bacteriol. 174: 1378-1387.

Angerer, A., S. Gaisser, and V. Braun. (1990). Nucleotide sequences of the sfuA, sfuB, and sfuC genes of Serratia marcescens suggest a periplasmic-binding-protein-dependent iron transport mechanism. J. Bacteriol. 172: 572-578.

Anjard, C., W.F. Loomis, and. (2002). Evolutionary analyses of ABC transporters of Dictyostelium discoideum. Eukaryot. Cell. 1: 643-652.

Arai, N., T. Furuta, and M. Sakurai. (2017). Analysis of an ATP-induced conformational transition of ABC transporter MsbA using a coarse-grained model. Biophys Physicobiol 14: 161-171.

Aranda, R., 4th, C.E. Worley, M. Liu, E. Bitto, M.S. Cates, J.S. Olson, B. Lei, and G.N. Phillips, Jr. (2007). Bis-methionyl coordination in the crystal structure of the heme-binding domain of the streptococcal cell surface protein Shp. J. Mol. Biol. 374: 374-383.

Aranda-Caraballo, J., R.A. Saenz, A.A. López-Zavala, B. Velazquez-Cruz, L. Espinosa-Barrera, Y. Cárdenas-Conejo, A. Zárate-Romero, O. Linares-Vergara, J.A. Osuna-Castro, E. Bonales-Alatorre, S. Centeno-Leija, and H. Serrano-Posada. (2023). Binding Specificity of a Novel Cyclo/Maltodextrin-Binding Protein and Its Role in the Cyclodextrin ABC Importer System from Thermoanaerobacterales. Molecules 28:.

Ardelli, B.F. (2013). Transport proteins of the ABC systems superfamily and their role in drug action and resistance in nematodes. Parasitol Int 62: 639-646.

Arend, C., I.L. Grothaus, M. Waespy, L.C. Ciacchi, and R. Dringen. (2024). Modulation of Multidrug Resistance Protein 1-mediated Transport Processes by the Antiviral Drug Ritonavir in Cultured Primary Astrocytes. Neurochem Res 49: 66-84.

Arends, S.J., R.J. Kustusch, and D.S. Weiss. (2009). ATP-binding site lesions in FtsE impair cell division. J. Bacteriol. 191: 3772-3784.

Arendt, M., A. Ambrosen, T. Fall, M. Kierczak, K. Tengvall, J.R.S. Meadows, &.#.1.9.7.;. Karlsson, A.S. Lagerstedt, T. Bergström, G. Andersson, K. Lindblad-Toh, and R. Hagman. (2021). The ABCC4 gene is associated with pyometra in golden retriever dogs. Sci Rep 11: 16647.

Arlanov, R., T. Lang, G. Jedlitschky, E. Schaeffeler, T. Ishikawa, M. Schwab, and A.T. Nies. (2015). Functional characterization of common protein variants in the efflux transporter ABCC11 and identification of T546M as functionally damaging variant. Pharmacogenomics J. [Epub: Ahead of Print]

Arnold, F.M., M.S. Weber, I. Gonda, M.J. Gallenito, S. Adenau, P. Egloff, I. Zimmermann, C.A.J. Hutter, L.M. Hürlimann, E.E. Peters, J. Piel, G. Meloni, O. Medalia, and M.A. Seeger. (2020). The ABC exporter IrtAB imports and reduces mycobacterial siderophores. Nature 580: 413-417.

Arya, N., H. Rahman, A. Rudrow, M. Wagner, L. Schmitt, S.V. Ambudkar, and J. Golin. (2019). An A666G mutation in transmembrane helix 5 of the yeast multidrug transporter Pdr5 increases drug efflux by enhancing cooperativity between transport sites. Mol. Microbiol. [Epub: Ahead of Print]

Asai, T., K. Okamoto-Shibayama, Y. Kikuchi, and K. Ishihara. (2018). Characterization of a novel potential peptide import system in Treponema denticola. Microb. Pathog. 123: 467-472.

Asakuma, S., E. Hatakeyama, T. Urashima, E. Yoshida, T. Katayama, K. Yamamoto, H. Kumagai, H. Ashida, J. Hirose, and M. Kitaoka. (2011). Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J. Biol. Chem. 286: 34583-34592.

Asmus, E., W. Karle, M.C. Brack, C. Wittig, F. Behrens, L. Reinshagen, M. Pfeiffer, S. Schulz, B. Mandzimba-Maloko, L. Erfinanda, P.L. Perret, L. Michalick, P.J. Smeele, E.H.T. Lim, C.E. van den Brom, A.B.A. Vonk, T. Kaiser, N. Suttorp, S. Hippenstiel, L.E. Sander, F. Kurth, U. Rauch, U. Landmesser, A. Haghikia, R. Preissner, H.J. Bogaard, M. Witzenrath, W.M. Kuebler, R. Szulcek, and S. Simmons. (2023). Cystic fibrosis transmembrane conductance regulator modulators attenuate platelet activation and aggregation in blood of healthy donors and COVID-19 patients. Eur Respir J 61:.

Assaraf, Y.G. The role of multidrug resistance efflux transporters in antifolate resistance and folate homeostasis. Drug Resist Updat 9: 227-246.

Assaraf, Y.G., I. Ifergan, W.N. Kadry, and R.Y. Pinter. (2006). Computer modelling of antifolate inhibition of folate metabolism using hybrid functional petri nets. J Theor Biol 240: 637-647.

Asthana, P., D. Singh, J.S. Pedersen, M.J. Hynönen, R. Sulu, A.V. Murthy, M. Laitaoja, J. Jänis, L.W. Riley, and R. Venkatesan. (2021). Structural insights into the substrate-binding proteins Mce1A and Mce4A from. IUCrJ 8: 757-774.

Atsumi, S., K. Miyamoto, K. Yamamoto, J. Narukawa, S. Kawai, H. Sezutsu, I. Kobayashi, K. Uchino, T. Tamura, K. Mita, K. Kadono-Okuda, S. Wada, K. Kanda, M.R. Goldsmith, and H. Noda. (2012). Single amino acid mutation in an ATP-binding cassette transporter gene causes resistance to Bt toxin Cry1Ab in the silkworm, Bombyx mori. Proc. Natl. Acad. Sci. USA 109: E1591-1598.

Aucher, W., C. Lacombe, A. Héquet, J. Frère, and J.M. Berjeaud. (2005). Influence of amino acid substitutions in the leader peptide on maturation and secretion of mesentericin Y105 by Leuconostoc mesenteroides. J. Bacteriol. 187: 2218-2223.

Aucher, W., V. Simonet, C. Fremaux, K. Dalet, L. Simon, Y. Cenatiempo, J. Frère, and J.M. Berjeaud. (2004). Differences in mesentericin secretion systems from two Leuconostoc strains. FEMS Microbiol. Lett. 232: 15-22.

Aung, T., B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann, M. Lahmann, T. Weinhage, K. Menck, T. Hupfeld, R. Koch, L. Trümper, and G.G. Wulf. (2011). Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 108: 15336-15341.

Austermuhle, M.I., J.A. Hall, C.S. Klug, and A.L. Davidson. (2004). Maltose-binding protein is open in the catalytic transition state for ATP hydrolysis during maltose transport. J. Biol. Chem. 279: 28243-28250.

Awram, P. and J. Smit. (1998). The Caulobacter crescentus paracrystalline S-layer protein is secreted by an ABC transporter (type I) secretion apparatus. J. Bacteriol. 180: 3062-3069.

Börnke, F., M. Hajirezaei, and U. Sonnewald. (2001). Cloning and characterization of the gene cluster for palatinose metabolism from the phytopathogenic bacterium Erwinia rhapontici. J. Bacteriol. 183: 2425-2430.

Baarine, M., K. Ragot, A. Athias, T. Nury, Z. Kattan, E.C. Genin, P. Andreoletti, F. Ménétrier, J.M. Riedinger, M. Bardou, and G. Lizard. (2012). Incidence of Abcd1 level on the induction of cell death and organelle dysfunctions triggered by very long chain fatty acids and TNF-α on oligodendrocytes and astrocytes. Neurotoxicology 33: 212-228.

Babu, M., C. Bundalovic-Torma, C. Calmettes, S. Phanse, Q. Zhang, Y. Jiang, Z. Minic, S. Kim, J. Mehla, A. Gagarinova, I. Rodionova, A. Kumar, H. Guo, O. Kagan, O. Pogoutse, H. Aoki, V. Deineko, J.H. Caufield, E. Holtzapple, Z. Zhang, A. Vastermark, Y. Pandya, C.C. Lai, M. El Bakkouri, Y. Hooda, M. Shah, D. Burnside, M. Hooshyar, J. Vlasblom, S.V. Rajagopala, A. Golshani, S. Wuchty, J. F Greenblatt, M. Saier, P. Uetz, T. F Moraes, J. Parkinson, and A. Emili. (2018). Global landscape of cell envelope protein complexes in Escherichia coli. Nat Biotechnol 36: 103-112.

Badiee, S.A., U.H. Isu, E. Khodadadi, and M. Moradi. (2023). The Alternating Access Mechanism in Mammalian Multidrug Resistance Transporters and Their Bacterial Homologs. Membranes (Basel) 13:.

Badri, D.V., J.M. Chaparro, D.K. Manter, E. Martinoia, and J.M. Vivanco. (2012). Influence of ATP-Binding Cassette Transporters in Root Exudation of Phytoalexins, Signals, and in Disease Resistance. Front Plant Sci 3: 149.

Baghel, P., M.K. Rawal, M.F. Khan, S. Sen, M.H. Siddiqui, V. Chaptal, P. Falson, and R. Prasad. (2017). Multidrug ABC transporter Cdr1 of Candida albicans harbors specific and overlapping binding sites for human steroid hormones transport. Biochim. Biophys. Acta. 1859: 1778-1789. [Epub: Ahead of Print]

Bagnat, M., A. Navis, S. Herbstreith, K. Brand-Arzamendi, S. Curado, S. Gabriel, K. Mostov, J. Huisken, and D.Y. Stainier. (2010). Cse1l is a negative regulator of CFTR-dependent fluid secretion. Curr. Biol. 20: 1840-1845.

Baharara, H., P. Kesharwani, T.P. Johnston, and A. Sahebkar. (2023). Therapeutic potential of phytochemicals for cystic fibrosis. Biofactors. [Epub: Ahead of Print]

Bai, M., Q. Shen, Y. Wu, Z. Ma, Y. Wang, M. Chen, D. Liu, and L. Zhou. (2024). Evaluation of transport mechanisms of methotrexate in human choriocarcinoma cell lines by LC-MS/MS. J Pharm Biomed Anal 247: 116268.

Bai, Y., M. Li, and T.C. Hwang. (2010). Dual roles of the sixth transmembrane segment of the CFTR chloride channel in gating and permeation. J Gen Physiol 136: 293-309.

Bai, Y., M. Li, and T.C. Hwang. (2011). Structural basis for the channel function of a degraded ABC transporter, CFTR (ABCC7). J Gen Physiol 138: 495-507.

Bai, Y., Z. Zhang, L. Jin, Y. Zhu, L. Zhao, B. Shi, J. Li, G. Guo, B. Guo, D.P. McManus, S. Wang, and W. Zhang. (2020). Dynamic Changes in the Global Transcriptome and MicroRNAome Reveal Complex miRNA-mRNA Regulation in Early Stages of the Bi-Directional Development of Protoscoleces. Front Microbiol 11: 654.

Bajaj, R., K.E. Bruce, A.L. Davidson, B.E. Rued, C.V. Stauffacher, and M.E. Winkler. (2016). Biochemical characterization of essential cell division proteins FtsX and FtsE that mediate peptidoglycan hydrolysis by PcsB in Streptococcus pneumoniae. Microbiologyopen 5: 738-752.

Bakhsheshian, J., B.R. Wei, K.E. Chang, S. Shukla, S.V. Ambudkar, R.M. Simpson, M.M. Gottesman, and M.D. Hall. (2013). Bioluminescent imaging of drug efflux at the blood-brain barrier mediated by the transporter ABCG2. Proc. Natl. Acad. Sci. USA 110: 20801-20806.

Bale, S., K.R. Rajashankar, K. Perry, T.P. Begley, and S.E. Ealick. (2010). HMP binding protein ThiY and HMP-P synthase THI5 are structural homologues. Biochemistry 49: 8929-8936.

Balibar, C.J., F.H. Vaillancourt, and C.T. Walsh. (2005). Generation of D amino acid residues in assembly of arthrofactin by dual condensation/epimerization domains. Chem Biol 12: 1189-1200.

Balzi, E., M. Wang, S. Leterme, L. Van Dyck, and A. Goffeau. (1994). PDR5, a novel yeast multidrug resistance conferring transporter controlled by the transcription regulator PDR1. J. Biol. Chem. 269: 2206-2214.

Ban, N., M. Sasaki, H. Sakai, K. Ueda, and N. Inagaki. (2005). Cloning of ABCA17, a novel rodent sperm-specific ABC (ATP-binding cassette) transporter that regulates intracellular lipid metabolism. Biochem. J. 389: 577-585.

Ban, N., Y. Matsumura, H. Sakai, Y. Takanezawa, M. Sasaki, H. Arai, and N. Inagaki. (2007). ABCA3 as a lipid transporter in pulmonary surfactant biogenesis. J. Biol. Chem. 282: 9628-9634.

Banasiak J., Biala W., Staszkow A., Swarcewicz B., Kepczynska E., Figlerowicz M. and Jasinski M. (2013). A Medicago truncatula ABC transporter belonging to subfamily G modulates the level of isoflavonoids. J Exp Bot. 64(4):1005-15.

Banerjee, A., A. Moreno, M.F. Khan, R. Nair, S. Sharma, S. Sen, A.K. Mondal, J. Pata, C. Orelle, P. Falson, and R. Prasad. (2019). Cdr1p highlights the role of the non-hydrolytic ATP-binding site in driving drug translocation in asymmetric ABC pumps. Biochim. Biophys. Acta. Biomembr 183131. [Epub: Ahead of Print]

Banerjee, A., J. Pata, V. Chaptal, A. Boumendjel, P. Falson, and R. Prasad. (2023). Structure, function, and inhibition of catalytically asymmetric ABC transporters: Lessons from the PDR subfamily. Drug Resist Updat 71: 100992. [Epub: Ahead of Print]

Banks, D.B., S.L. Lierz, R.E. Cannon, and K.S. Korach. (2024). Nongenomic ERα-AMPK Signaling Regulates Sex-Dependent Bcrp Transport Activity at the Blood-Brain Barrier. Endocrinology 165:.

Banks, W.A. (2023). Viktor Mutt lecture: Peptides can cross the blood-brain barrier. Peptides 169: 171079. [Epub: Ahead of Print]

Bao, X., J. Wu, Y. Xie, S. Kim, S. Michelhaugh, J. Jiang, S. Mittal, N. Sanai, and J. Li. (2019). Protein Expression and Functional Relevance of Efflux and Uptake Drug Transporters at the Blood-Brain Barrier of Human Brain and Glioblastoma. Clin Pharmacol Ther. [Epub: Ahead of Print]

Barik, S., M. Saini, M. Rana, and P.K. Gupta. (2019). Multidrug resistance protein 4 (MRP4) is expressed as transcript variants in both Gallus domesticus and Gyps himalyanesis. Gene 689: 172-182.

Barona-Gómez, F., S. Lautru, F.X. Francou, P. Leblond, J.L. Pernodet, and G.L. Challis. (2006). Multiple biosynthetic and uptake systems mediate siderophore-dependent iron acquisition in Streptomyces coelicolor A3(2) and Streptomyces ambofaciens ATCC 23877. Microbiology 152: 3355-3366.

Barrangou, R., E. Altermann, R. Hutkins, R. Cano, and T.R. Klaenhammer. (2003). Functional and comparative genomic analyses of an operon involved in fructooligosaccharide utilization by Lactobacillus acidophilus. Proc. Natl. Acad. Sci. USA 100: 8957-8962.

Barreca, M., M. Renda, V. Spanò, A. Montalbano, M.V. Raimondi, S. Giuffrida, R. Bivacqua, T. Bandiera, L.J.V. Galietta, and P. Barraja. (2024). Identification of 6,9-dihydro-5H-pyrrolo[3,2-h]quinazolines as a new class of F508del-CFTR correctors for the treatment of cystic fibrosis. Eur J Med Chem 276: 116691.

Barth, K., M. Rudolph, T. Diederichs, T.F. Prisner, R. Tampé, and B. Joseph. (2020). Thermodynamic Basis for Conformational Coupling in an ATP-Binding Cassette Exporter. J Phys Chem Lett 11: 7946-7953.

Barth, K., S. Hank, P.E. Spindler, T.F. Prisner, R. Tampé, and B. Joseph. (2018). Conformational Coupling and trans-Inhibition in the Human Antigen Transporter Ortholog TmrAB Resolved with Dipolar EPR Spectroscopy. J. Am. Chem. Soc. 140: 4527-4533.

Bartsevich, V.V. and H.B. Pakrasi. (1999). Membrane topology of MntB, the transmembrane protein component of an ABC transporter system for manganese in the cyanobacterium Synechocystis sp. strain PCC 6803. J. Bacteriol. 181: 3591-3593.

Basavanna, S., S. Khandavilli, J. Yuste, J.M. Cohen, A.H. Hosie, A.J. Webb, G.H. Thomas, and J.S. Brown. (2009). Screening of Streptococcus pneumoniae ABC transporter mutants demonstrates that LivJHMGF, a branched-chain amino acid ABC transporter, is necessary for disease pathogenesis. Infect. Immun. 77: 3412-3423.

Basso LR Jr., Gast CE., Mao Y. and Wong B. (2010). Fluconazole transport into Candida albicans secretory vesicles by the membrane proteins Cdr1p, Cdr2p, and Mdr1p. Eukaryot Cell. 9(6):960-70.

Bauer, B.E., H. Wolfger, and K. Kuchler. (1999). Inventory and function of yeast ABC proteins: about sex, stress, pleiotropic drug and heavy metal resistance. Biochim. Biophys. Acta. 1461: 217-236.

Bearden, S.W. and R.D. Perry. (1999). The Yfe system of Yersinia pestis transports iron and manganese and is required for full virulence of plague. Mol. Microbiol. 32: 403-414.

Beasley, F.C., E.D. Vinés, J.C. Grigg, Q. Zheng, S. Liu, G.A. Lajoie, M.E. Murphy, and D.E. Heinrichs. (2009). Characterization of staphyloferrin A biosynthetic and transport mutants in Staphylococcus aureus. Mol. Microbiol. 72: 947-963.

Bechara, C., A. Nöll, N. Morgner, M.T. Degiacomi, R. Tampé, and C.V. Robinson. (2015). A subset of annular lipids is linked to the flippase activity of an ABC transporter. Nat Chem 7: 255-262.

Becker, J.P., F. Van Bambeke, P.M. Tulkens, and M. Prévost. (2010). Dynamics and structural changes induced by ATP binding in SAV1866, a bacterial ABC exporter. J Phys Chem B 114: 15948-15957.

Becker, P., R. Hakenbeck, and B. Henrich. (2009). An ABC transporter of Streptococcus pneumoniae involved in susceptibility to vancoresmycin and bacitracin. Antimicrob. Agents Chemother. 53: 2034-2041.

Beckers, G., A.K. Bendt, R. Krämer, and A. Burkovski. (2004). Molecular identification of the urea uptake system and transcriptional analysis of urea transporter- and urease-encoding genes in Corynebacterium glutamicum. J. Bacteriol. 186: 7645-7652.

Beesley, N.J., K. Cwiklinski, K. Allen, R.C. Hoyle, T.W. Spithill, E.J. La Course, D.J.L. Williams, S. Paterson, and J.E. Hodgkinson. (2023). A major locus confers triclabendazole resistance in Fasciola hepatica and shows dominant inheritance. PLoS Pathog 19: e1011081. [Epub: Ahead of Print]

Behmard, E., E. Barzegari, S. Najafipour, A. Kouhpayeh, Y. Ghasemi, and A.A. Asadi-Pooya. (2022). Efflux dynamics of the antiseizure drug, levetiracetam, through the P-glycoprotein channel revealed by advanced comparative molecular simulations. Sci Rep 12: 13674.

Bellamy, W.T. (1996). P-glycoproteins and multidrug resistance. Annu Rev Pharmacol Toxicol 36: 161-183.

Benoit, S.L., S. Seshadri, R. Lamichhane-Khadka, and R.J. Maier. (2013). Helicobacter hepaticus NikR controls urease and hydrogenase activities via the NikABDE and HH0418 putative nickel import proteins. Microbiology 159: 136-146.

Bera, K., P. Rani, G. Kishor, S. Agarwal, A. Kumar, and D.V. Singh. (2017). Structural elucidation of transmembrane domain zero (TMD0) of EcdL: A multidrug resistance-associated protein (MRP) family of ATP-binding cassette transporter protein revealed by atomistic simulation. J Biomol Struct Dyn 1-13. [Epub: Ahead of Print]

Bernard, D.G., Y. Cheng, Y. Zhao, and J. Balk. (2009). An allelic mutant series of ATM3 reveals its key role in the biogenesis of cytosolic iron-sulfur proteins in Arabidopsis. Plant Physiol. 151: 590-602.

Bernard, R., A. Guiseppi, M. Chippaux, M. Foglino, and F. Denizot. (2007). Resistance to bacitracin in Bacillus subtilis: unexpected requirement of the BceAB ABC transporter in the control of expression of its own structural genes. J. Bacteriol. 189: 8636-8642.

Bernard, R., P. Joseph, A. Guiseppi, M. Chippaux, and F. Denizot. (2003). YtsCD and YwoA, two independent systems that confer bacitracin resistance to Bacillus subtilis. FEMS Microbiol. Lett. 228: 93-97.

Bernecic, N.C., S.P. Graaf, T. Leahy, and B.M. Gadella. (2021). HDL mediates reverse cholesterol transport from ram spermatozoa and induces hyperactivated motility. Biol Reprod. [Epub: Ahead of Print]

Berntsson RP., Smits SH., Schmitt L., Slotboom DJ. and Poolman B. (2010). A structural classification of substrate-binding proteins. FEBS Lett. 584(12):2606-17.

Berntsson, R.P., A.M. Thunnissen, B. Poolman, and D.J. Slotboom. (2011). Importance of a hydrophobic pocket for peptide binding in lactococcal OppA. J. Bacteriol. 193: 4254-4256.

Berntsson, R.P., J. ter Beek, M. Majsnerowska, R.H. Duurkens, P. Puri, B. Poolman, and D.J. Slotboom. (2012). Structural divergence of paralogous S components from ECF-type ABC transporters. Proc. Natl. Acad. Sci. USA 109: 13990-13995.

Berntsson, R.P., M.K. Doeven, F. Fusetti, R.H. Duurkens, D. Sengupta, S.J. Marrink, A.M. Thunnissen, B. Poolman, and D.J. Slotboom. (2009). The structural basis for peptide selection by the transport receptor OppA. EMBO. J. 28: 1332-1340.

Bessadok, A., E. Garcia, H. Jacquet, S. Martin, A. Garrigues, N. Loiseau, F. André, S. Orlowski, and M. Vivaudou. (2011). Recognition of sulfonylurea receptor (ABCC8/9) ligands by the multidrug resistance transporter P-glycoprotein (ABCB1): functional similarities based on common structural features between two multispecific ABC proteins. J. Biol. Chem. 286: 3552-3569.

Bessire, M., S. Borel, G. Fabre, L. Carraça, N. Efremova, A. Yephremov, Y. Cao, R. Jetter, A.C. Jacquat, J.P. Métraux, and C. Nawrath. (2011). A member of the PLEIOTROPIC DRUG RESISTANCE family of ATP binding cassette transporters is required for the formation of a functional cuticle in Arabidopsis. Plant Cell 23: 1958-1970.

Bevers, L.E., P.L. Hagedoorn, G.C. Krijger, and W.R. Hagen. (2006). Tungsten transport protein A (WtpA) in Pyrococcus furiosus: the first member of a new class of tungstate and molybdate transporters. J. Bacteriol. 188: 6498-6505.

Bhattacharya, S. and A. Palillo. (2021). Structural and dynamic studies of the peptidase domain from Clostridium thermocellum PCAT1. Protein. Sci. [Epub: Ahead of Print]

Bi, Y., E. Mann, C. Whitfield, and J. Zimmer. (2018). Architecture of a channel-forming O-antigen polysaccharide ABC transporter. Nature 553: 361-365.

Bi, Y., X. Wang, H. Ding, F. He, L. Han, and Y. Zhang. (2022). Transporter-mediated natural product-drug interactions. Planta Med. [Epub: Ahead of Print]

Biala, W., J. Banasiak, K. Jarzyniak, A. Pawela, and M. Jasinski. (2017). Medicago truncatula ABCG10 is a transporter of 4-coumarate and liquiritigenin in the medicarpin biosynthetic pathway. J Exp Bot 68: 3231-3241.

Bian, J., H. Shen, Y. Tu, A. Yu, and C. Li. (2011). The Riboswitch Regulates a Thiamine Pyrophosphate ABC Transporter of the Oral Spirochete Treponema denticola. J. Bacteriol. 193: 3912-3922.

Bibb, L.A. and M.P. Schmitt. (2010). The ABC transporter HrtAB confers resistance to hemin toxicity and is regulated in a hemin-dependent manner by the ChrAS two-component system in Corynebacterium diphtheriae. J. Bacteriol. 192: 4606-4617.

Bickers, S.C., S. Benlekbir, J.L. Rubinstein, and V. Kanelis. (2021). Structure of Ycf1p reveals the transmembrane domain TMD0 and the regulatory region of ABCC transporters. Proc. Natl. Acad. Sci. USA 118:.

Bidossi, A., L. Mulas, F. Decorosi, L. Colomba, S. Ricci, G. Pozzi, J. Deutscher, C. Viti, and M.R. Oggioni. (2012). A functional genomics approach to establish the complement of carbohydrate transporters in Streptococcus pneumoniae. PLoS One 7: e33320.

Bieler, S., F. Silva, C. Soto, and D. Belin. (2006). Bactericidal activity of both secreted and nonsecreted microcin E492 requires the mannose permease. J. Bacteriol. 188: 7049-7061.

Biemans-Oldehinkel, E. and B. Poolman. (2003). On the role of the two extracytoplasmic substrate-binding domains in the ABC transporter OpuA. EMBO. J. 22: 5983-5993.

Biemans-Oldehinkel, E., N.A. Mahmood, and B. Poolman. (2006). A sensor for intracellular ionic strength. Proc. Natl. Acad. Sci. USA 103: 10624-10629.

Binet, R., S. Létoffé, J.M. Ghigo, P. Delepelaire, and C. Wandersman. (1997). Protein secretion by Gram-negative bacterial ABC exporters--a review. Gene 192: 7-11.

Bird, D., F. Beisson, A. Brigham, J. Shin, S. Greer, R. Jetter, L. Kunst, X. Wu, A. Yephremov, and L. Samuels. (2007). Characterization of Arabidopsis ABCG11/WBC11, an ATP binding cassette (ABC) transporter that is required for cuticular lipid secretion. Plant J. 52: 485-498.

Biswas-Fiss, E.E. (2006). Interaction of the nucleotide binding domains and regulation of the ATPase activity of the human retina specific ABC transporter, ABCR. Biochemistry 45: 3813-3823.

Blakeslee, J.J., A. Bandyopadhyay, O.R. Lee, J. Mravec, B. Titapiwatanakun, M. Sauer, S.N. Makam, Y. Cheng, R. Bouchard, J. Adamec, M. Geisler, A. Nagashima, T. Sakai, E. Martinoia, J. Friml, W.A. Peer, and A.S. Murphy. (2007). Interactions among PIN-FORMED and P-glycoprotein auxin transporters in Arabidopsis. Plant Cell 19: 131-147.

Blanco, F.C., M.V. Bianco, S. Garbaccio, V. Meikle, M.J. Gravisaco, V. Montenegro, E. Alfonseca, M. Singh, S. Barandiaran, A. Canal, L. Vagnoni, B.M. Buddle, F. Bigi, and A. Cataldi. (2013). Mycobacterium bovis Δmce2 double deletion mutant protects cattle against challenge with virulent M. bovis. Tuberculosis (Edinb) 93: 363-372.

Bobrov, A.G., O. Kirillina, J.D. Fetherston, M.C. Miller, J.A. Burlison, and R.D. Perry. (2014). The Yersinia pestis siderophore, yersiniabactin, and the ZnuABC system both contribute to zinc acquisition and the development of lethal septicaemic plague in mice. Mol. Microbiol. 93: 759-775.

Bock, C., F. Löhr, F. Tumulka, K. Reichel, J. Würz, G. Hummer, L. Schäfer, R. Tampé, B. Joseph, F. Bernhard, V. Dötsch, and R. Abele. (2018). Structural and functional insights into the interaction and targeting hub TMD0 of the polypeptide transporter TAPL. Sci Rep 8: 15662.

Bonito, C.A., R.J. Ferreira, M.U. Ferreira, J.P. Gillet, M.N.D.S. Cordeiro, and D.J.V.A. Dos Santos. (2020). Theoretical insights on helix repacking as the origin of P-glycoprotein promiscuity. Sci Rep 10: 9823.

Bordignon, E., M. Grote, and E. Schneider. (2010). The maltose ATP-binding cassette transporter in the 21st century--towards a structural dynamic perspective on its mode of action. Mol. Microbiol. 77: 1354-1366.

Borst, P. and R.O. Elferink. (2002). Mammalian ABC transporters in health and disease. Annu. Rev. Biochem. 71: 537-592.

Borst, P., R. Evers, M. Kool, and J. Wijnholds. (1999). The multidrug resistance protein family. Biochim. Biophys. Acta. 1461: 347-357.

BoseDasgupta, S., A. Ganguly, A. Roy, T. Mukherjee, and H.K. Majumder. (2008). A novel ATP-binding cassette transporter, ABCG6 is involved in chemoresistance of Leishmania. Mol Biochem Parasitol 158: 176-188.

Bossé, J.T., H.D. Gilmour, and J.I. MacInnes. (2001). Novel genes affecting urease acivity in Actinobacillus pleuropneumoniae. J. Bacteriol. 183: 1242-1247.

Bossennec, M., A. Di Roio, C. Caux, and C. Ménétrier-Caux. (2018). MDR1 in immunity: friend or foe? Oncoimmunology 7: e1499388.

Bouchard, R., A. Bailly, J.J. Blakeslee, S.C. Oehring, V. Vincenzetti, O.R. Lee, I. Paponov, K. Palme, S. Mancuso, A.S. Murphy, B. Schulz, and M. Geisler. (2006). Immunophilin-like TWISTED DWARF1 modulates auxin efflux activities of Arabidopsis P-glycoproteins. J. Biol. Chem. 281: 30603-30612.

Boucher, N. and K.M. Noll. (2011). Ligands of thermophilic ABC transporters encoded in a newly sequenced genomic region of Thermotoga maritima MSB8 screened by differential scanning fluorimetry. Appl. Environ. Microbiol. 77: 6395-6399.

Braakman, R., M.J. Follows, and S.W. Chisholm. (2017). Metabolic evolution and the self-organization of ecosystems. Proc. Natl. Acad. Sci. USA 114: E3091-E3100.

Braibant, M., P. Gilot, and J. Content. (2000). The ATP binding cassette (ABC) transport systems of Mycobacterium tuberculosis. FEMS Microbiol. Rev. 24: 449-467.

Braun, V. and C. Herrmann. (2007). Docking of the periplasmic FecB binding protein to the FecCD transmembrane proteins in the ferric citrate transport system of Escherichia coli. J. Bacteriol. 189: 6913-6918.

Brautigam CA., Ouyang Z., Deka RK. and Norgard MV. (2014). Sequence, biophysical, and structural analyses of the PstS lipoprotein (BB0215) from Borrelia burgdorferi reveal a likely binding component of an ABC-type phosphate transporter. Protein Sci. 23(2):200-12.

Brayboy, L.M., L.O. Knapik, S. Long, M. Westrick, and G.M. Wessel. (2018). Ovarian hormones modulate multidrug resistance transporters in the ovary. Contracept Reprod Med 3: 26.

Brechbuhl, H.M., N. Gould, R. Kachadourian, W.R. Riekhof, D.R. Voelker, and B.J. Day. (2010). Glutathione transport is a unique function of the ATP-binding cassette protein ABCG2. J. Biol. Chem. 285: 16582-16587.

Breedveld, P., D. Pluim, G. Cipriani, F. Dahlhaus, M.A. van Eijndhoven, C.J. de Wolf, A. Kuil, J.H. Beijnen, G.L. Scheffer, G. Jansen, P. Borst, and J.H. Schellens. (2007). The effect of low pH on breast cancer resistance protein (ABCG2)-mediated transport of methotrexate, 7-hydroxymethotrexate, methotrexate diglutamate, folic acid, mitoxantrone, topotecan, and resveratrol in in vitro drug transport models. Mol Pharmacol 71: 240-249.

Brem, D., C. Pelludat, A. Rakin, C.A. Jacobi, and J. Heesemann. (2001). Functional analysis of yersiniabactin transport genes of Yersinia enterocolitica. Microbiology 147: 1115-1127.

Brickman, T.J., C.K. Vanderpool, and S.K. Armstrong. (2006). Heme transport contributes to in vivo fitness of Bordetella pertussis during primary infection in mice. Infect. Immun. 74: 1741-1744.

Brodhagen, M., I. Paulsen, and J.E. Loper. (2005). Reciprocal regulation of pyoluteorin production with membrane transporter gene expression in Pseudomonas fluorescens Pf-5. Appl. Environ. Microbiol. 71: 6900-6909.

Brünker, P., J. Altenbuchner, and R. Mattes. (1998). Structure and function of the genes involved in mannitol, arabitol and glucitol utilization from Pseudomonas fluorescens DSM50106. Gene 206: 117-126.

Brunner, R., C.L. Ng, H. Aissaoui, M.H. Akabas, C. Boss, R. Brun, P.S. Callaghan, O. Corminboeuf, D.A. Fidock, I.J. Frame, B. Heidmann, A. Le Bihan, P. Jenö, C. Mattheis, S. Moes, I.B. Müller, M. Paguio, P.D. Roepe, R. Siegrist, T. Voss, R.W. Welford, S. Wittlin, and C. Binkert. (2013). UV-triggered affinity capture identifies interactions between the Plasmodium falciparum multidrug resistance protein 1 (PfMDR1) and antimalarial agents in live parasitized cells. J. Biol. Chem. 288: 22576-22583.

Brunner, R., H. Aissaoui, C. Boss, Z. Bozdech, R. Brun, O. Corminboeuf, S. Delahaye, C. Fischli, B. Heidmann, M. Kaiser, J. Kamber, S. Meyer, P. Papastogiannidis, R. Siegrist, T. Voss, R. Welford, S. Wittlin, and C. Binkert. (2012). Identification of a new chemical class of antimalarials. J Infect Dis 206: 735-743.

Bryan, E.J., Q. Qiao, Y. Wang, J.Y. Roberge, E.J. LaVoie, and D.S. Pilch. (2024). A FtsZ Inhibitor That Can Utilize Siderophore-Ferric Iron Uptake Transporter Systems for Activity against Gram-Negative Bacterial Pathogens. Antibiotics (Basel) 13:.

Bryan, J. and L. Aguilar-Bryan. (1999). Sulfonylurea receptors: ABC transporters that regulate ATP-sensitive K+ channels. Biochim. Biophys. Acta. 1461: 285-303.

Bukowska, M.A., M. Hohl, E.R. Geertsma, L.M. Hürlimann, M.G. Grütter, and M.A. Seeger. (2015). A Transporter Motor Taken Apart: Flexibility in the Nucleotide Binding Domains of a Heterodimeric ABC Exporter. Biochemistry 54: 3086-3099.

Burguière, P., J. Fert, I. Guillouard, S. Auger, A. Danchin, and I. Martin-Verstraete. (2005). Regulation of the Bacillus subtilis ytmI operon, involved in sulfur metabolism. J. Bacteriol. 187: 6019-6030.

Burguière, P., S. Auger, M.F. Hullo, A. Danchin, and I. Martin-Verstraete. (2004). Three different systems participate in L-cystine uptake in Bacillus subtilis. J. Bacteriol. 186: 4875-4884.

Burkhard, K.A. and A. Wilks. (2008). Functional characterization of the Shigella dysenteriae heme ABC transporter. Biochemistry 47: 7977-7979.

Burnat, M., B. Li, S.H. Kim, A.J. Michael, and E. Flores. (2018). Homospermidine biosynthesis in the cyanobacterium Anabaena requires a deoxyhypusine synthase homologue and is essential for normal diazotrophic growth. Mol. Microbiol. 109: 763-780.

Burns, V., L.J. Sharpe, I.C. Gelissen, and A.J. Brown. (2013). Species variation in ABCG1 isoform expression: Implications for the use of animal models in elucidating ABCG1 function. Atherosclerosis 226: 408-411.

Butcher, B.G. and J.D. Helmann. (2006). Identification of Bacillus subtilis sigma-dependent genes that provide intrinsic resistance to antimicrobial compounds produced by Bacilli. Mol. Microbiol. 60: 765-782.

Butcher, B.G., Y.P. Lin, and J.D. Helmann. (2007). The yydFGHIJ operon of Bacillus subtilis encodes a peptide that induces the LiaRS two-component system. J. Bacteriol. 189: 8616-8625.

Cabral, M., C. Anjard, W.F. Loomis, and A. Kuspa. (2006). Genetic evidence that the acyl coenzyme A binding protein AcbA and the serine protease/ABC transporter TagA function together in Dictyostelium discoideum cell differentiation. Eukaryot. Cell. 5: 2024-2032.

Caceres G., Robey RW., Sokol L., McGraw KL., Clark J., Lawrence NJ., Sebti SM., Wiese M. and List AF. (2012). HG-829 is a potent noncompetitive inhibitor of the ATP-binding cassette multidrug resistance transporter ABCB1. Cancer Res. 72(16):4204-13.

Caffalette, C.A. and J. Zimmer. (2021). Cryo-EM structure of the full-length WzmWzt ABC transporter required for lipid-linked O antigen transport. Proc. Natl. Acad. Sci. USA 118:.

Caffalette, C.A., R.A. Corey, M.S.P. Sansom, P.J. Stansfeld, and J. Zimmer. (2019). A lipid gating mechanism for the channel-forming O antigen ABC transporter. Nat Commun 10: 824.

Cai, S.Y., L. Wang, N. Ballatori, and J.L. Boyer. (2001). Bile salt export pump is highly conserved during vertebrate evolution and its expression is inhibited by PFIC type II mutations. Am. J. Physiol. Gastrointest Liver Physiol 281: G316-322.

Cai, X., Z. Bikadi, Z. Ni, E.W. Lee, H. Wang, M.F. Rosenberg, and Q. Mao. (2010). Role of basic residues within or near the predicted transmembrane helix 2 of the human breast cancer resistance protein in drug transport. J Pharmacol Exp Ther 333: 670-681.

Campbell, J.L. and H.A. Nash. (2001). Volatile general anesthetics reveal a neurobiological role for the white and brown genes of Drosophila melanogaster. J Neurobiol 49: 339-349.

Cannon, R.D., F.J. Fischer, K. Niimi, M. Niimi, and M. Arisawa. (1998). Drug pumping mechanisms in Candida albicans. Nippon Ishinkin Gakkai Zasshi 39: 73-78.

Cao, Q.T., J.A. Aguiar, B.J. Tremblay, N. Abbas, N. Tiessen, S. Revill, N. Makhdami, A. Ayoub, G. Cox, K. Ask, A.C. Doxey, and J.A. Hirota. (2020). ABCF1 Regulates dsDNA-induced Immune Responses in Human Airway Epithelial Cells. Front Cell Infect Microbiol 10: 487.

Carey Hulyer, A.R., D.A. Briggs, M.L. O''Mara, I.D. Kerr, J.R. Harmer, and R. Callaghan. (2020). Cross-linking, DEER-spectroscopy and molecular dynamics confirm the inward facing state of P-glycoprotein in a lipid membrane. J Struct Biol 211: 107513.

Carraro, N., T.E. Tisdale-Orr, R.M. Clouse, A.S. Knöller, and R. Spicer. (2012). Diversification and Expression of the PIN, AUX/LAX, and ABCB Families of Putative Auxin Transporters in Populus. Front Plant Sci 3: 17.

Carrasco-Torres, G., S. Fattel-Fazenda, G.S. López-Alvarez, R. García-Román, S. Villa-Treviño, and V.R. Vásquez-Garzón. (2015). The transmembrane transporter ABCC3 participates in liver cancer progression and is a potential biomarker. Tumour Biol. [Epub: Ahead of Print]

Carrier, D.J., C.W.T. van Roermund, T.A. Schaedler, H.L. Rong, L. IJlst, R.J.A. Wanders, S.A. Baldwin, H.R. Waterham, F.L. Theodoulou, and A. Baker. (2019). Mutagenesis separates ATPase and thioesterase activities of the peroxisomal ABC transporter, Comatose. Sci Rep 9: 10502.

Carrier, M.C., D. Lalaouna, and E. Massé. (2023). Hfq protein and GcvB small RNA tailoring of target mRNA to levels allowing translation activation by MicF small RNA in. RNA Biol 20: 59-76.

Casemayou, A., J. Belliere, E. Letavernier, E. Colliou, H. El Hachem, J. Zarowski, D. Bazin, C. Kounde, A. Piedrafita, G. Feuillet, J.P. Schanstra, and S. Faguer. (2023). Abcc6 deficiency prevents rhabdomyolysis-induced acute kidney injury. Sci Rep 13: 21513.

Castanys-Muñoz, E., N. Alder-Baerens, T. Pomorski, F. Gamarro, and S. Castanys. (2007). A novel ATP-binding cassette transporter from Leishmania is involved in transport of phosphatidylcholine analogues and resistance to alkyl-phospholipids. Mol. Microbiol. 64: 1141-1153.

Castranio, E.L., C.M. Wolfe, K.N. Nam, F. Letronne, N.F. Fitz, I. Lefterov, and R. Koldamova. (2018). ABCA1 haplodeficiency affects the brain transcriptome following traumatic brain injury in mice expressing human APOE isoforms. Acta Neuropathol Commun 6: 69.

Cavazza, C., L. Martin, E. Laffly, H. Lebrette, M.V. Cherrier, L. Zeppieri, P. Richaud, M. Carrière, and J.C. Fontecilla-Camps. (2011). Histidine 416 of the periplasmic binding protein NikA is essential for nickel uptake in Escherichia coli. FEBS Lett. 585: 711-715.

Caygill, S. and L. Dolan. (2023). ATP binding cassette transporters and uridine diphosphate glycosyltransferases are ancient protein families that evolved roles in herbicide resistance through exaptation. PLoS One 18: e0287356.

Chacon-Camacho, O.F., N. Xilotl-de Jesús, E. Calderón-Martínez, V. Ordoñez-Labastida, M.I. Neria-Gonzalez, R. Villafuerte-de la Cruz, A. Martinez-Rojas, and J.C. Zenteno. (2024). Genotypic spectrum of ABCA4-associated retinal degenerations in 211 unrelated Mexican patients: identification of 22 novel disease-causing variants. Mol. Genet. Genomics 299: 79.

Chahine, S., S. Seabrooke, and M.J. O''Donnell. (2012). Effects of genetic knock-down of organic anion transporter genes on secretion of fluorescent organic ions by Malpighian tubules of Drosophila melanogaster. Arch Insect Biochem Physiol 81: 228-240.

Chan, Y.K., W.A. McCormick, and R.J. Watson. (1997). A new nos gene downstream from nosDFY is essential for dissimilatory reduction of nitrous oxide by Rhizobium (Sinorhizobium) meliloti. Microbiology 143(Pt8): 2817-2824.

Chandravanshi, M., A. Sharma, P. Dasgupta, S.K. Mandal, and S.P. Kanaujia. (2019). Identification and characterization of ABC transporters for carbohydrate uptake in Thermus thermophilus HB8. Gene 696: 135-148.

Chandravanshi, M., P. Gogoi, and S.P. Kanaujia. (2016). Computational characterization of TTHA0379: A potential glycerophosphocholine binding protein of Ugp ATP-binding cassette transporter. Gene 592: 260-268.

Chandravanshi, M., P. Gogoi, and S.P. Kanaujia. (2019). Structural and thermodynamic correlation illuminates the selective transport mechanism of disaccharide α-glycosides through ABC transporter. FEBS J. [Epub: Ahead of Print]

Chang HK., Dennis JJ. and Zylstra GJ. (2009). Involvement of two transport systems and a specific porin in the uptake of phthalate by Burkholderia spp. J Bacteriol. 191(14):4671-3.

Chang, G. (2003). Structure of MsbA from Vibrio cholera: a multidrug resistance ABC transporter homolog in a closed conformation. J. Mol. Biol. 330: 419-430.

Chang, G. and C.B. Roth. (2001). Structure of MsbA from E. coli: a homolog of the multidrug resistance ATP binding cassette (ABC) transporters. Science 293: 1793-1800.

Chang, G., C.B. Roth, C.L. Reyes, O. Pornillos, Y.J. Chen, and A.P. Chen. (2006). Retraction. Science 314: 1875.

Chang, Y.C., W.H. Sheu, Y.S. Chien, P.C. Tseng, W.J. Lee, and A.N. Chiang. (2013). Hyperglycemia accelerates ATP-binding cassette transporter A1 degradation via an ERK-dependent pathway in macrophages. J. Cell. Biochem. 114: 1364-1373.

Chantemargue, B., F. Di Meo, K. Berka, N. Picard, H. Arnion, M. Essig, P. Marquet, M. Otyepka, and P. Trouillas. (2018). Structural patterns of the human ABCC4/MRP4 exporter in lipid bilayers rationalize clinically observed polymorphisms. Pharmacol Res. [Epub: Ahead of Print]

Chater, K.F. and S. Horinouchi. (2003). Signalling early developmental events in two highly diverged Streptomyces species. Mol. Microbiol. 48: 9-15.

Chaudhuri, B.N., J. Ko, C. Park, T.A. Jones, and S.L. Mowbray. (1999). Structure of D-allose binding protein from Escherichia coli bound to D-allose at 1.8 Å resolution. J. Mol. Biol. 286: 1519-1531.

Checroun, C. and C. Gutierrez. (2004). Sigma(s)-dependent regulation of yehZYXW, which encodes a putative osmoprotectant ABC transporter of Escherichia coli. FEMS Microbiol. Lett. 236: 221-226.

Cheema, Y., Y.S. Kiani, K.J. Linton, and I. Jabeen. (2023). Identification and Empiric Evaluation of New Inhibitors of the Multidrug Transporter P-Glycoprotein (ABCB1). Int J Mol Sci 24:.

Chen ZS. and Tiwari AK. (2011). Multidrug resistance proteins (MRPs/ABCCs) in cancer chemotherapy and genetic diseases. FEBS J. 278(18):3226-45.

Chen, C. and G.A. Beattie. (2007). Characterization of the osmoprotectant transporter OpuC from Pseudomonas syringae and demonstration that cystathionine-β-synthase domains are required for its osmoregulatory function. J. Bacteriol. 189: 6901-6912.

Chen, C., A.A. Malek, M.J. Wargo, D.A. Hogan, and G.A. Beattie. (2010). The ATP-binding cassette transporter Cbc (choline/betaine/carnitine) recruits multiple substrate-binding proteins with strong specificity for distinct quaternary ammonium compounds. Mol. Microbiol. 75: 29-45.

Chen, D.Q., Y. Xie, L.Q. Cao, J.S. Fleishman, Y. Chen, T. Wu, and D.H. Yang. (2024). The role of ABCC10/MRP7 in anti-cancer drug resistance and beyond. Drug Resist Updat 73: 101062. [Epub: Ahead of Print]

Chen, F., H. Hu, Z. Li, J. Huang, X. Cai, C. Wang, Q. He, and J. Cao. (2015). Deletion of HAPS_2096 Increases Sensitivity to Cecropin B in Haemophilus parasuis. J. Mol. Microbiol. Biotechnol. 25: 284-291.

Chen, J., S. Han, S. Li, H. Zhu, S. Li, J. Yan, and T. Zhu. (2022). Tandem Mass Tags Quantitative Proteome Identification and Function Analysis of ABC Transporters in. Int J Mol Sci 23:.

Chen, L., J. Wei, C. Liu, W. Zhang, B. Wang, L. Niu, and G. Liang. (2018). Specific Binding Protein ABCC1 Is Associated With Cry2Ab Toxicity in. Front Physiol 9: 745.

Chen, M.M., G.H. Shi, Y. Dai, W.X. Fang, and Q. Wu. (2023). Identifying genetic variants associated with amphotericin B (AMB) resistance in via merbased GWAS. Front Genet 14: 1133593.

Chen, Q., Y. Yang, L. Li, and J.T. Zhang. (2006). The amino terminus of the human multidrug resistance transporter ABCC1 has a U-shaped folding with a gating function. J. Biol. Chem. 281: 31152-31163.

Chen, S., R. Sánchez-Fernández, E.R. Lyver, A. Dancis, and P.A. Rea. (2007). Functional characterization of AtATM1, AtATM2, and AtATM3, a subfamily of Arabidopsis half-molecule ATP-binding cassette transporters implicated in iron homeostasis. J. Biol. Chem. 282: 21561-21571.

Chen, W., M. Zhou, B. Guan, B. Xie, Y. Liu, J. He, J. Zhao, Q. Zhao, and D. Yan. (2024). Tumour-associated macrophage-derived DOCK7-enriched extracellular vesicles drive tumour metastasis in colorectal cancer via the RAC1/ABCA1 axis. Clin Transl Med 14: e1591.

Chen, Y.Y. and R.A. Burne. (2003). Identification and characterization of the nickel uptake system for urease biogenesis in Streptococcus salivarius 57.I. J. Bacteriol. 185: 6773-6779.

Chen, Z.P., D. Xu, L. Wang, Y.X. Mao, Y. Li, M.T. Cheng, C.Z. Zhou, W.T. Hou, and Y. Chen. (2022). Structural basis of substrate recognition and translocation by human very long-chain fatty acid transporter ABCD1. Nat Commun 13: 3299.

Cheng, J., A.A. Guffanti, and T.A. Krulwich. (1997). A two-gene ABC-type transport system that extrudes Na+ in Bacillus subtilis is induced by ethanol or protonophore. Mol. Microbiol. 23: 1107-1120.

Cheong, N., H. Zhang, M. Madesh, M. Zhao, K. Yu, C. Dodia, A.B. Fisher, R.C. Savani, and H. Shuman. (2007). ABCA3 is critical for lamellar body biogenesis in vivo. J. Biol. Chem. 282: 23811-23817.

Chevance, F.F., M. Erhardt, C. Lengsfeld, S.J. Lee, and W. Boos. (2006). Mlc of Thermus thermophilus: a glucose-specific regulator for a glucose/mannose ABC transporter in the absence of the phosphotransferase system. J. Bacteriol. 188: 6561-6571.

Chin, N., J. Frey, C.F. Chang, and Y.F. Chang. (1996). Identification of a locus involved in the utilization of iron by Actinobacillus pleuropneumoniae. FEMS Microbiol. Lett. 143: 1-6.

Cho, H. and H. Kang. (2012). The PseEF efflux system is a virulence factor of Pseudomonas syringae pv. syringae. J Microbiol 50: 79-90.

Cho, J., S.K. Costa, R.M. Wierzbicki, W.F.C. Rigby, and A.L. Cheung. (2021). The extracellular loop of the membrane permease VraG interacts with GraS to sense cationic antimicrobial peptides in Staphylococcus aureus. PLoS Pathog 17: e1009338.

Cho, M., Z.W. Lee, and H.T. Cho. (2012). ATP-binding cassette B4, an auxin-efflux transporter, stably associates with the plasma membrane and shows distinctive intracellular trafficking from that of PIN-FORMED proteins. Plant Physiol. 159: 642-654.

Cho, S., M. Lu, X. He, P.L. Ee, U. Bhat, E. Schneider, L. Miele, and W.T. Beck. (2011). Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proc. Natl. Acad. Sci. USA 108: 20778-20783.

Choi, C.C. and R.C. Ford. (2021). ATP binding cassette importers in eukaryotic organisms. Biol Rev Camb Philos Soc. [Epub: Ahead of Print]

Chong, Z.S., W.F. Woo, and S.S. Chng. (2015). Osmoporin OmpC forms a complex with MlaA to maintain outer membrane lipid asymmetry in Escherichia coli. Mol. Microbiol. 98: 1133-1146.

Choudhury, H.G., Z. Tong, I. Mathavan, Y. Li, S. Iwata, S. Zirah, S. Rebuffat, H.W. van Veen, and K. Beis. (2014). Structure of an antibacterial peptide ATP-binding cassette transporter in a novel outward occluded state. Proc. Natl. Acad. Sci. USA 111: 9145-9150.

Chow, V., G. Nong, and J.F. Preston. (2007). Structure, function, and regulation of the aldouronate utilization gene cluster from Paenibacillus sp. strain JDR-2. J. Bacteriol. 189: 8863-8870.

Christensen, O., E.M. Harvat, L. Thöny-Meyer, S.J. Ferguson, and J.M. Stevens. (2007). Loss of ATP hydrolysis activity by CcmAB results in loss of c-type cytochrome synthesis and incomplete processing of CcmE. FEBS J. 274: 2322-2332.

Clay, N.K., A.M. Adio, C. Denoux, G. Jander, and F.M. Ausubel. (2009). Glucosinolate metabolites required for an Arabidopsis innate immune response. Science 323: 95-101.

Clifton MC., Simon MJ., Erramilli SK., Zhang H., Zaitseva J., Hermodson MA. and Stauffacher CV. (2015). In vitro reassembly of the ribose ATP-binding cassette transporter reveals a distinct set of transport complexes. J Biol Chem. 290(9):5555-65.

Clifton, B.E., U. Alcolombri, G.I. Uechi, C.J. Jackson, and P. Laurino. (2024). The ultra-high affinity transport proteins of ubiquitous marine bacteria. Nature. [Epub: Ahead of Print]

Coelho, D., J.C. Kim, I.R. Miousse, S. Fung, M. du Moulin, I. Buers, T. Suormala, P. Burda, M. Frapolli, M. Stucki, P. Nürnberg, H. Thiele, H. Robenek, W. Höhne, N. Longo, M. Pasquali, E. Mengel, D. Watkins, E.A. Shoubridge, J. Majewski, D.S. Rosenblatt, B. Fowler, F. Rutsch, and M.R. Baumgartner. (2012). Mutations in ABCD4 cause a new inborn error of vitamin B12 metabolism. Nat. Genet. 44: 1152-1155.

Collins, B., N. Curtis, P.D. Cotter, C. Hill, and R.P. Ross. (2010). The ABC transporter AnrAB contributes to the innate resistance of Listeria monocytogenes to nisin, bacitracin, and various β-lactam antibiotics. Antimicrob. Agents Chemother. 54: 4416-4423.

Compagne, N., A. Vieira Da Cruz, R.T. Müller, R.C. Hartkoorn, M. Flipo, and K.M. Pos. (2023). Update on the Discovery of Efflux Pump Inhibitors against Critical Priority Gram-Negative Bacteria. Antibiotics (Basel) 12:.

Conners, S.B., C.I. Montero, D.A. Comfort, K.R. Shockley, M.R. Johnson, S.R. Chhabra, and R.M. Kelly. (2005). An expression-driven approach to the prediction of carbohydrate transport and utilization regulons in the hyperthermophilic bacterium Thermotoga maritima. J. Bacteriol. 187: 7267-7282.

Conseil, G. and S.P.C. Cole. (2021). The First Cytoplasmic Loop in the Core Structure of the ABCC1 (Multidrug Resistance Protein 1; MRP1) Transporter Contains Multiple Amino Acids Essential for Its Expression. Int J Mol Sci 22:.

Conseil, G., R.G. Deeley, and S.P. Cole. (2005). Role of two adjacent cytoplasmic tyrosine residues in MRP1 (ABCC1) transport activity and sensitivity to sulfonylureas. Biochem Pharmacol 69: 451-461.

Consonni, C., M.E. Humphry, H.A. Hartmann, M. Livaja, J. Durner, L. Westphal, J. Vogel, V. Lipka, B. Kemmerling, P. Schulze-Lefert, S.C. Somerville, and R. Panstruga. (2006). Conserved requirement for a plant host cell protein in powdery mildew pathogenesis. Nat. Genet. 38: 716-720.

Cortese, A., S.J. Beecroft, S. Facchini, R. Curro, M. Cabrera-Serrano, I. Stevanovski, S.R. Chintalaphani, H. Gamaarachchi, B. Weisburd, C. Folland, G. Monahan, C.K. Scriba, L. Dofash, M. Johari, B.R. Grosz, M. Ellis, L.G. Fearnley, R. Tankard, J. Read, A. Merve, N. Dominik, E. Vegezzi, R.P. Schnekenberg, G. Fernandez-Eulate, M. Masingue, D. Giovannini, M.B. Delatycki, E. Storey, M. Gardner, D.J. Amor, G. Nicholson, S. Vucic, R.D. Henderson, T. Robertson, J. Dyke, V. Fabian, F. Mastaglia, M.R. Davis, M. Kennerson, , R. Quinlivan, S. Hammans, A. Tucci, M. Bahlo, C.A. McLean, N.G. Laing, T. Stojkovic, H. Houlden, M.G. Hanna, I.W. Deveson, P.J. Lockhart, P.J. Lamont, M.C. Fahey, E. Bugiardini, and G. Ravenscroft. (2024). A CCG expansion in ABCD3 causes oculopharyngodistal myopathy in individuals of European ancestry. Nat Commun 15: 6327.

Coudray, N., G.L. Isom, M.R. MacRae, M.N. Saiduddin, G. Bhabha, and D.C. Ekiert. (2020). Structure of bacterial phospholipid transporter MlaFEDB with substrate bound. Elife 9:. [Epub: Ahead of Print]

Cox, M.H., P. Kapoor, D.A. Briggs, and I.D. Kerr. (2018). Residues contributing to drug transport by ABCG2 are localised to multiple drug-binding pockets. Biochem. J. 475: 1553-1567.

Crawford, R.R., P.K. Potukuchi, E.G. Schuetz, and J.D. Schuetz. (2018). Beyond Competitive Inhibition: Regulation of ABC Transporters by Kinases and Protein-Protein Interactions as Potential Mechanisms of Drug-Drug Interactions. Drug Metab Dispos 46: 567-580.

Crouch, M.L., M. Castor, J.E. Karlinsey, T. Kalhorn, and F.C. Fang. (2008). Biosynthesis and IroC-dependent export of the siderophore salmochelin are essential for virulence of Salmonella enterica serovar Typhimurium. Mol. Microbiol. 67: 971-983.

Crouzet, J., T. Trombik, A.S. Fraysse, and M. Boutry. (2006). Organization and function of the plant pleiotropic drug resistance ABC transporter family. FEBS Lett. 580: 1123-1130.

Crow, A., N.P. Greene, E. Kaplan, and V. Koronakis. (2017). Structure and mechanotransmission mechanism of the MacB ABC transporter superfamily. Proc. Natl. Acad. Sci. USA 114: 12572-12577.

Cruz-Ramos, H., G.M. Cook, G. Wu, M.W. Cleeter, and R.K. Poole. (2004). Membrane topology and mutational analysis of Escherichia coli CydDC, an ABC-type cysteine exporter required for cytochrome assembly. Microbiology 150: 3415-3427.

Csanády, L., P. Vergani, and D.C. Gadsby. (2010). Strict coupling between CFTR's catalytic cycle and gating of its Cl- ion pore revealed by distributions of open channel burst durations. Proc. Natl. Acad. Sci. USA 107: 1241-1246.

Csanády, L., P. Vergani, and D.C. Gadsby. (2019). STRUCTURE, GATING, AND REGULATION OF THE CFTR ANION CHANNEL. Physiol. Rev. 99: 707-738.

Cui, Y., S. Liang, S. Zhang, C. Zhang, Y. Zhao, D. Wu, J. Wang, R. Song, J. Wang, D. Yin, Y. Liu, S. Pan, X. Liu, Y. Wang, J. Han, F. Meng, B. Zhang, H. Guo, Z. Lu, and L. Liu. (2020). ABCA8 is regulated by miR-374b-5p and inhibits proliferation and metastasis of hepatocellular carcinoma through the ERK/ZEB1 pathway. J Exp Clin Cancer Res 39: 90.

Cuthbertson, L., M.S. Kimber, and C. Whitfield. (2007). Substrate binding by a bacterial ABC transporter involved in polysaccharide export. Proc. Natl. Acad. Sci. USA 104: 19529-19534.

Cuyx, S., A.S. Ramalho, S. Fieuws, N. Corthout, M. Proesmans, M. Boon, K. Arnauts, M.S. Carlon, S. Munck, L. Dupont, K. De Boeck, F. Vermeulen, and. (2024). Rectal organoid morphology analysis (ROMA) as a novel physiological assay for diagnostic classification in cystic fibrosis. Thorax 79: 834-841.

Czuba, L.C., K.M. Hillgren, and P.W. Swaan. (2018). Post-translational modifications of transporters. Pharmacol Ther 192: 88-99.

Dębska, S., A. Owecka, U. Czernek, K. Szydłowska-Pazera, M. Habib, and P. Potemski. (2011). [Transmembrane transporters ABCC - structure, function and role in multidrug resistance of cancer cells]. Postepy Hig Med Dosw (Online) 65: 552-561.

da Costa, K.M., L. Freire-de-Lima, L.M. da Fonseca, J.O. Previato, L. Mendonça-Previato, and R.D.C. Valente. (2023). ABCB1 and ABCC1 Function during TGF-β-Induced Epithelial-Mesenchymal Transition: Relationship between Multidrug Resistance and Tumor Progression. Int J Mol Sci 24:.

Dai, J., Y. He, J. Fang, H. Wang, L. Chao, L. Zhao, Z. Hong, and Y. Chai. (2022). In Vitro Evaluation of the Interaction of Seven Biologically Active Components in with P-gp. Molecules 27:.

Dale, S.E., M.T. Sebulsky, and D.E. Heinrichs. (2004). Involvement of SirABC in iron-siderophore import in Staphylococcus aureus. J. Bacteriol. 186: 8356-8362.

Dalmas, O., M.A. Do Cao, M.R. Lugo, F.J. Sharom, A. Di Pietro, and J.M. Jault. (2005). Time-resolved fluorescence resonance energy transfer shows that the bacterial multidrug ABC half-transporter BmrA functions as a homodimer. Biochemistry 44: 4312-4321.

Dalton, T.L., J.T. Collins, T.C. Barnett, and J.R. Scott. (2006). RscA, a member of the MDR1 family of transporters, is repressed by CovR and required for growth of Streptococcus pyogenes under heat stress. J. Bacteriol. 188: 77-85.

Damiani, D. and M. Tiribelli. (2024). ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target? Biomedicines 12:.

Damodaran, S. and L.C. Strader. (2019). Indole 3-Butyric Acid Metabolism and Transport in. Front Plant Sci 10: 851.

Daniel, J., L. Abraham, A. Martin, X. Pablo, and S. Reyes. (2018). Rv2477c is an antibiotic-sensitive manganese-dependent ABC-F ATPase in Mycobacterium tuberculosis. Biochem. Biophys. Res. Commun. 495: 35-40.

Daniel, P., K. Balušíková, R. Václavíková, K. Šeborová, &.#.3.5.2.;. Ransdorfová, M. Valeriánová, L. Wei, M. Jelínek, T. Tlapáková, T. Fleischer, V.N. Kristensen, P. Souček, I. Ojima, and J. Kovář. (2023). Amplicon Contains Cyclic AMP Response Element-Driven Gene in Taxane-Resistant MCF-7 Breast Cancer Sublines. Genes (Basel) 14:.

Danilchanka, O., C. Mailaender, and M. Niederweis. (2008). Identification of a novel multidrug efflux pump of Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 52: 2503-2511.

Darnell, R.L., Y. Nakatani, M.K. Knottenbelt, S. Gebhard, and G.M. Cook. (2019). Functional characterization of BcrR: a one-component transmembrane signal transduction system for bacitracin resistance. Microbiology. [Epub: Ahead of Print]

Darvari, R. and M. Boroujerdi. (2004). Concentration dependency of modulatory effect of amlodipine on P-glycoprotein efflux activity of doxorubicin--a comparison with tamoxifen. J Pharm Pharmacol 56: 985-991.

Dasgupta, A., K. Sureka, D. Mitra, B. Saha, S. Sanyal, A.K. Das, P. Chakrabarti, M. Jackson, B. Gicquel, M. Kundu, and J. Basu. (2010). An oligopeptide transporter of Mycobacterium tuberculosis regulates cytokine release and apoptosis of infected macrophages. PLoS One 5: e12225.

Dassa, E. and P. Bouige. The ABC of ABCS: a phylogenetic and functional classification of ABC systems in living organisms. Res. Microbiol. 152: 211-229.

Dastvan, R., S. Mishra, Y.B. Peskova, R.K. Nakamoto, and H.S. Mchaourab. (2019). Mechanism of allosteric modulation of P-glycoprotein by transport substrates and inhibitors. Science 364: 689-692.

Davidson, A.L. and P.C. Maloney. (2007). ABC transporters: how small machines do a big job. Trends Microbiol. 15: 448-455.

Davies, B.W. and G.C. Walker. (2007). Disruption of sitA compromises Sinorhizobium meliloti for manganese uptake required for protection against oxidative stress. J. Bacteriol. 189: 2101-2109.

Dawson, R.J. and K.P. Locher. (2006). Structure of a bacterial multidrug ABC transporter. Nature 443: 180-185.

De Costa, D.M., K. Suzuki, and K. Yoshida. (2003). Structural and functional analysis of a putative gene cluster for palatinose transport on the linear chromosome of Agrobacterium tumefaciens MAFF301001. J. Bacteriol. 185: 2369-2373.

De Greef, J., M. Akue, N. Panin, K.A. Delongie, M. André, G. Mahieu, E. Hoste, L. Elens, L. Belkhir, and V. Haufroid. (2024). Effect of ABCB1 most frequent polymorphisms on the accumulation of bictegravir in recombinant HEK293 cell lines. Sci Rep 14: 16290.

De la Torre, L.I., J.G. Vergara Meza, S. Cabarca, A.G. Costa-Martins, and A. Balan. (2021). Comparison of carbohydrate ABC importers from Mycobacterium tuberculosis. BMC Genomics 22: 841.

De Marcos Lousa, C., C.W. van Roermund, V.L. Postis, D. Dietrich, I.D. Kerr, R.J. Wanders, S.A. Baldwin, A. Baker, and F.L. Theodoulou. (2013). Intrinsic acyl-CoA thioesterase activity of a peroxisomal ATP binding cassette transporter is required for transport and metabolism of fatty acids. Proc. Natl. Acad. Sci. USA 110: 1279-1284.

de Miranda, R., B.J. Cuthbert, T. Klevorn, A. Chao, J. Mendoza, M. Arbing, P.J. Sieminski, K. Papavinasasundaram, S. Abdul-Hafiz, S. Chan, C.M. Sassetti, S. Ehrt, and C.W. Goulding. (2023). Differentiating the roles of Mycobacterium tuberculosis substrate binding proteins, FecB and FecB2, in iron uptake. PLoS Pathog 19: e1011650. [Epub: Ahead of Print]

De Muyt, A., D. Vezon, G. Gendrot, J.L. Gallois, R. Stevens, and M. Grelon. (2007). AtPRD1 is required for meiotic double strand break formation in Arabidopsis thaliana. EMBO. J. 26: 4126-4137.

De Vilder, E.Y., M.J. Hosen, and O.M. Vanakker. (2015). The ABCC6 Transporter as a Paradigm for Networking from an Orphan Disease to Complex Disorders. Biomed Res Int 2015: 648569.

de Wet, H., M.G. Rees, K. Shimomura, J. Aittoniemi, A.M. Patch, S.E. Flanagan, S. Ellard, A.T. Hattersley, M.S. Sansom, and F.M. Ashcroft. (2007). Increased ATPase activity produced by mutations at arginine-1380 in nucleotide-binding domain 2 of ABCC8 causes neonatal diabetes. Proc. Natl. Acad. Sci. USA 104: 18988-18992.

de Wet, H., M.V. Mikhailov, C. Fotinou, M. Dreger, T.J. Craig, C. Vénien-Bryan, and F.M. Ashcroft. (2007). Studies of the ATPase activity of the ABC protein SUR1. FEBS J. 274: 3532-3544.

Dean, M. and R. Allikmets. (2001). Complete characterization of the human ABC gene family. J. Bioenerg. Biomembr. 33: 475-479.

Decottignies, A. and A. Goffeau. (1997). Complete inventory of the yeast ABC proteins. Nat. Genet. 15: 137-145.

Deeley, R.G., C. Westlake, and S.P. Cole. (2006). Transmembrane transport of endo- and xenobiotics by mammalian ATP-binding cassette multidrug resistance proteins. Physiol. Rev. 86: 849-899.

Degiorgio, D., C. Colombo, M. Seia, L. Porcaro, L. Costantino, L. Zazzeron, D. Bordo, and D.A. Coviello. (2007). Molecular characterization and structural implications of 25 new ABCB4 mutations in progressive familial intrahepatic cholestasis type 3 (PFIC3). Eur J Hum Genet 15: 1230-1238.

Dehghan, A., R.C. Pinto, I. Karaman, J. Huang, B.R. Durainayagam, M. Ghanbari, A. Nazeer, Q. Zhong, S. Liggi, L. Whiley, R. Mustafa, M. Kivipelto, A. Solomon, T. Ngandu, T. Kanekiyo, T. Aikawa, C.I. Radulescu, S.J. Barnes, G. Graça, E. Chekmeneva, S. Camuzeaux, M.R. Lewis, M.R. Kaluarachchi, M.A. Ikram, E. Holmes, I. Tzoulaki, P.M. Matthews, J.L. Griffin, and P. Elliott. (2022). Metabolome-wide association study on indicates a role of ceramide metabolism in Alzheimer''s disease. Proc. Natl. Acad. Sci. USA 119: e2206083119.

Deka, R.K., C.A. Brautigam, B.A. Biddy, W.Z. Liu, and M.V. Norgard. (2013). Evidence for an ABC-type riboflavin transporter system in pathogenic spirochetes. MBio 4: e615-61512.

Deka, R.K., C.A. Brautigam, X.F. Yang, J.S. Blevins, M. Machius, D.R. Tomchick, and M.V. Norgard. (2006). The PnrA (Tp0319; TmpC) lipoprotein represents a new family of bacterial purine nucleoside receptor encoded within an ATP-binding cassette (ABC)-like operon in Treponema pallidum. J. Biol. Chem. 281: 8072-8081.

del Castillo, T., E. Duque, and J.L. Ramos. (2008). A set of activators and repressors control peripheral glucose pathways in Pseudomonas putida to yield a common central intermediate. J. Bacteriol. 190: 2331-2339.

Delepelaire, P. (2019). Bacterial ABC transporters of iron containing compounds. Res. Microbiol. [Epub: Ahead of Print]

Delgado, M.A., P.A. Vincent, R.N. Farías, and R.A. Salomón. (2005). YojI of Escherichia coli functions as a microcin J25 efflux pump. J. Bacteriol. 187: 3465-3470.

Demirel O., Jan I., Wolters D., Blanz J., Saftig P., Tampe R. and Abele R. (2012). The lysosomal polypeptide transporter TAPL is stabilized by interaction with LAMP-1 and LAMP-2. J Cell Sci. 125(Pt 18):4230-40.

Demirel, O., Z. Waibler, U. Kalinke, F. Grünebach, S. Appel, P. Brossart, A. Hasilik, R. Tampé, and R. Abele. (2007). Identification of a lysosomal peptide transport system induced during dendritic cell development. J. Biol. Chem. 282: 37836-37843.

Demissie, Z.A., M. Tarnowycz, A.M. Adal, L.S. Sarker, and S.S. Mahmoud. (2018). A lavender ABC transporter confers resistance to monoterpene toxicity in yeast. Planta. [Epub: Ahead of Print]

Demmer, A., H. Thole, M. Raida, and B. Tümmler. (2021). Lysine 268 adjacent to transmembrane helix 5 of hamster P-glycoprotein is the major photobinding site of iodomycin in CHO B30 cells. FEBS Open Bio. [Epub: Ahead of Print]

Demurtas, O.C., R. de Brito Francisco, E. Martinoia, and G. Giuliano. (2020). Transportomics for the Characterization of Plant Apocarotenoid Transmembrane Transporters. Methods Mol Biol 2083: 89-99.

Deng, F., L. Ghemtio, E. Grazhdankin, P. Wipf, H. Xhaard, and H. Kidron. (2020). Binding site interactions of modulators of Breast Cancer Resistance Protein, Multidrug Resistance Associated Protein 2 and P-glycoprotein activity. Mol Pharm. [Epub: Ahead of Print]

Dergunov, A.D., E.V. Savushkin, L.V. Dergunova, and D.Y. Litvinov. (2018). Significance of Cholesterol-Binding Motifs in ABCA1, ABCG1, and SR-B1 Structure. J. Membr. Biol. [Epub: Ahead of Print]

Deutch, C.E., I. Spahija, and C.E. Wagner. (2014). Susceptibility of Escherichia coli to the toxic L-proline analogue L-selenaproline is dependent on two L-cystine transport systems. J Appl Microbiol 117: 1487-1499.

Deutschbauer, A., M.N. Price, K.M. Wetmore, W. Shao, J.K. Baumohl, Z. Xu, M. Nguyen, R. Tamse, R.W. Davis, and A.P. Arkin. (2011). Evidence-based annotation of gene function in Shewanella oneidensis MR-1 using genome-wide fitness profiling across 121 conditions. PLoS Genet 7: e1002385.

Dezi M., Di Cicco A., Bassereau P. and Levy D. (2013). Detergent-mediated incorporation of transmembrane proteins in giant unilamellar vesicles with controlled physiological contents. Proc Natl Acad Sci U S A. 110(18):7276-81.

Di Diego, J.M., B. Patocskai, H. Barajas-Martinez, V. Borbáth, M.J. Ackerman, A. Burashnikov, J. Clatot, G.R. Li, V.M. Robinson, D. Hu, and C. Antzelevitch. (2020). Acacetin suppresses the electrocardiographic and arrhythmic manifestations of the J wave syndromes. PLoS One 15: e0242747.

Diederichs, K., J. Diez, G. Greller, C. Müller, J. Breed, C. Schnell, C. Vonrhein, W. Boos, and W. Welte. (2000). Crystal structure of MalK, the ATPase subunit of the trehalose/maltose ABC transporter of the archaeon Thermococcus litoralis. EMBO. J. 19: 5951-5961.

Dietrich, D., H. Schmuths, C.d.e.M. Lousa, J.M. Baldwin, S.A. Baldwin, A. Baker, F.L. Theodoulou, and M.J. Holdsworth. (2009). Mutations in the Arabidopsis peroxisomal ABC transporter COMATOSE allow differentiation between multiple functions in planta: insights from an allelic series. Mol. Biol. Cell 20: 530-543.

Ding, D., J.X. Wu, X. Duan, S. Ma, L. Lai, and L. Chen. (2022). Structural identification of vasodilator binding sites on the SUR2 subunit. Nat Commun 13: 2675.

Ding, H., C.B. Yip, B.A. Geddes, I.J. Oresnik, and M.F. Hynes. (2012). Glycerol utilization by Rhizobium leguminosarum requires an ABC transporter and affects competition for nodulation. Microbiology 158: 1369-1378.

Dintilhac, A., G. Alloing, C. Granadel, and J.P. Claverys. (1997). Competence and virulence of Streptococcus pneumoniae: Adc and PsaA mutants exhibit a requirement for Zn and Mn resulting from inactivation of putative ABC metal permeases. Mol. Microbiol. 25: 727-739.

Dintner, S., R. Heermann, C. Fang, K. Jung, and S. Gebhard. (2014). A Sensory Complex Consisting of an ATP-binding Cassette Transporter and a Two-component Regulatory System Controls Bacitracin Resistance in Bacillus subtilis. J. Biol. Chem. 289: 27899-27910.

Do, Y., M. Yagi, H. Hirai, K. Miki, Y. Fukahori, D. Setoyama, M. Yamamoto, T. Furukawa, Y. Kunisaki, D. Kang, and T. Uchiumi. (2024). Cardiomyocyte-specific deletion of the mitochondrial transporter Abcb10 causes cardiac dysfunction via lysosomal-mediated ferroptosis. Biosci Rep 44:.

Doerrler, W.T., M.C. Reedy, and C.R. Raetz. (2001). An Escherichia coli mutant defective in lipid export. J. Biol. Chem. 276: 11461-11464.

Doeven, M.K., G. van den Bogaart, V. Krasnikov, and B. Poolman. (2008). Probing receptor-translocator interactions in the oligopeptide ABC transporter by fluorescence correlation spectroscopy. Biophys. J. 94: 3956-3965.

Doeven, M.K., R. Abele, R. Tampé, and B. Poolman. (2004). The binding specificity of OppA determines the selectivity of the oligopeptide ATP-binding cassette transporter. J. Biol. Chem. 279: 32301-32307.

Domenech, P., H. Kobayashi, K. LeVier, G.C. Walker, and C.E. Barry, 3rd. (2009). BacA, an ABC transporter involved in maintenance of chronic murine infections with Mycobacterium tuberculosis. J. Bacteriol. 191: 477-485.

Domingos, L.T.S., M.F.C. Santos, D.C. de Moraes, L.F.R. de Sá, V.A.D. da Silva, L.M. Meuren, R.G.S. Berlinck, and A. Ferreira-Pereira. (2020). Batzelladine D and norbatzelladine L purified from marine sponge Monanchora arbuscula induce the reversal of fluconazole. Bioorg Chem 105: 104402. [Epub: Ahead of Print]

Dong, H., X. Tang, Z. Zhang, and C. Dong. (2017). Structural insight into lipopolysaccharide transport from the Gram-negative bacterial inner membrane to the outer membrane. Biochim. Biophys. Acta. 1862: 1461-1467.

Dong, H., Z. Zhang, X. Tang, N.G. Paterson, and C. Dong. (2017). Structural and functional insights into the lipopolysaccharide ABC transporter LptB2FG. Nat Commun 8: 222.

Dong, J., R. Lai, K. Nielsen, C.A. Fekete, H. Qiu, and A.G. Hinnebusch. (2004). The essential ATP-binding cassette protein RLI1 functions in translation by promoting preinitiation complex assembly. J. Biol. Chem. 279: 42157-42168.

Dong, W., Z.G. Liao, G.W. Zhao, X.J. Guan, J. Zhang, X.L. Liang, and M. Yang. (2018). Reversal Effect of Oxypeucedanin on P-glycoprotein-mediated Drug Transport. Molecules 23:.

Döring, H., D. Kreutzer, C. Ritter, and A. Hilgeroth. (2020). Discovery of Novel Symmetrical 1,4-Dihydropyridines as Inhibitors of Multidrug-Resistant Protein (MRP4) Efflux Pump for Anticancer Therapy. Molecules 26:.

Dou, W., J. Zhu, T. Wang, W. Wang, H. Li, X. Chen, and W. Guan. (2016). Mutations of charged amino acids at the cytoplasmic end of transmembrane helix 2 affect transport activity of the budding yeast multidrug resistance protein Pdr5p. FEMS Yeast Res 16:.

Downie, M.J., K. El Bissati, A.M. Bobenchik, L. Nic Lochlainn, A. Amerik, R. Zufferey, K. Kirk, and C. Ben Mamoun. (2010). PfNT2, a permease of the equilibrative nucleoside transporter family in the endoplasmic reticulum of Plasmodium falciparum. J. Biol. Chem. 285: 20827-20833.

Draper, L.A., P.D. Cotter, C. Hill, and R.P. Ross. (2015). Lantibiotic resistance. Microbiol. Mol. Biol. Rev. 79: 171-191.

Duan, C., M. Yu, J. Xu, B.Y. Li, Y. Zhao, and R.K. Kankala. (2023). Overcoming Cancer Multi-drug Resistance (MDR): Reasons, mechanisms, nanotherapeutic solutions, and challenges. Biomed Pharmacother 162: 114643. [Epub: Ahead of Print]

Duanmu, D., A.R. Miller, K.M. Horken, D.P. Weeks, and M.H. Spalding. (2009). Knockdown of limiting-CO2-induced gene HLA3 decreases HCO3- transport and photosynthetic Ci affinity in Chlamydomonas reinhardtii. Proc. Natl. Acad. Sci. USA 106: 5990-5995.

Dugourd, D., C. Martin, C.R. Rioux, M. Jacques, and J. Harel. (1999). Characterization of a periplasmic ATP-binding cassette iron import system of Brachyspira (Serpulina) hyodysenteriae. J. Bacteriol. 181: 6948-6957.

Dupont, L., I. Garcia, M.C. Poggi, G. Alloing, K. Mandon, and D. Le Rudulier. (2004). The Sinorhizobium meliloti ABC transporter Cho is highly specific for choline and expressed in bacteroids from Medicago sativa nodules. J. Bacteriol. 186: 5988-5996.

Durães, F., M. Pinto, and E. Sousa. (2018). Medicinal Chemistry Updates on Bacterial Efflux Pump Modulators. Curr. Med. Chem. 25: 6030-6069.

Durmus, S., J.J. Hendrikx, and A.H. Schinkel. (2015). Apical ABC transporters and cancer chemotherapeutic drug disposition. Adv Cancer Res 125: 1-41.

Duvivier, L., L. Gerard, A. Diaz, and J.P. Gillet. (2024). Linking ABC transporters to the hallmarks of cancer. Trends Cancer 10: 124-134.

Ecker, G.F., K. Pleban, S. Kopp, E. Csaszar, G.J. Poelarends, M. Putman, D. Kaiser, W.N. Konings, and P. Chiba. (2004). A three-dimensional model for the substrate binding domain of the multidrug ATP binding cassette transporter LmrA. Mol Pharmacol 66: 1169-1179.

Eckford PD. and Sharom FJ. (2010). The reconstituted Escherichia coli MsbA protein displays lipid flippase activity. Biochem J. 429(1):195-203.

Ehses, S., R.M. Leonhardt, G. Hansen, and M.R. Knittler. (2005). Functional role of C-terminal sequence elements in the transporter associated with antigen processing. J Immunol 174: 328-339.

Eichhorn, E. and T. Leisinger. (2001). Escherichia coli utilizes methanesulfonate and L-cysteate as sole sulfur sources for growth. FEMS Microbiol. Lett. 205: 271-275.

Eitinger, T., D.A. Rodionov, M. Grote, and E. Schneider. (2011). Canonical and ECF-type ATP-binding cassette importers in prokaryotes: diversity in modular organization and cellular functions. FEMS Microbiol. Rev. 35: 3-67.

Ejby, M., F. Fredslund, J.M. Andersen, A. Vujičić Žagar, J.R. Henriksen, T.L. Andersen, B. Svensson, D.J. Slotboom, and M. Abou Hachem. (2016). An ATP Binding Cassette Transporter Mediates the Uptake of α-(1,6)-Linked Dietary Oligosaccharides in Bifidobacterium and Correlates with Competitive Growth on These Substrates. J. Biol. Chem. 291: 20220-20231.

Ekiert, D.C., G. Bhabha, G.L. Isom, G. Greenan, S. Ovchinnikov, I.R. Henderson, J.S. Cox, and R.D. Vale. (2017). Architectures of Lipid Transport Systems for the Bacterial Outer Membrane. Cell 169: 273-285.e17.

Elferink, M.G., S.V. Albers, W.N. Konings, and A.J. Driessen. (2001). Sugar transport in Sulfolobus solfataricus is mediated by two families of binding protein-dependent ABC transporters. Mol. Microbiol. 39: 1494-1503.

Elias, M., A. Wellner, K. Goldin-Azulay, E. Chabriere, J.A. Vorholt, T.J. Erb, and D.S. Tawfik. (2012). The molecular basis of phosphate discrimination in arsenate-rich environments. Nature 491: 134-137.

Elliott, A.M. and M.A. Al-Hajj. (2009). ABCB8 mediates doxorubicin resistance in melanoma cells by protecting the mitochondrial genome. Mol Cancer Res 7: 79-87.

Endo, R., Y. Ohtsubo, M. Tsuda, and Y. Nagata. (2007). Identification and characterization of genes encoding a putative ABC-type transporter essential for utilization of γ-hexachlorocyclohexane in Sphingobium japonicum UT26. J. Bacteriol. 189: 3712-3720.

Engelen, M., R. Ofman, P.A. Mooijer, B.T. Poll-The, R.J. Wanders, and S. Kemp. (2008). Cholesterol-deprivation increases mono-unsaturated very long-chain fatty acids in skin fibroblasts from patients with X-linked adrenoleukodystrophy. Biochim. Biophys. Acta. 1781: 105-111.

Eom, G.T., J.Y. Oh, J.H. Park, H.J. Lim, S.J. Lee, E.Y. Kim, J.E. Choi, J. Jegal, B.K. Song, J.H. Yu, and J.K. Song. (2016). Alleviation of temperature-sensitive secretion defect of Pseudomonas fluorescens ATP-binding cassette (ABC) transporter, TliDEF, by a change of single amino acid in the ABC protein, TliD. J Biosci Bioeng. [Epub: Ahead of Print]

Erbeznik, M., S.E. Hudson, A.B. Herrman, and H.J. Strobel. (2004). Molecular analysis of the xylFGH operon, coding for xylose ABC transport, in Thermoanaerobacter ethanolicus. Curr. Microbiol. 48: 295-299.

Erkens, G.B., M. Majsnerowska, J. Ter Beek, and D.J. Slotboom. (2012). Energy Coupling Factor-Type ABC Transporters for Vitamin Uptake in Prokaryotes. Biochemistry 51: 4390-4396.

Escudero, J.A., A. San Millan, B. Gutierrez, L. Hidalgo, R.M. La Ragione, M. AbuOun, M. Galimand, M.J. Ferrándiz, L. Domínguez, A.G. de la Campa, and B. Gonzalez-Zorn. (2011). Fluoroquinolone efflux in Streptococcus suis is mediated by SatAB and not by SmrA. Antimicrob. Agents Chemother. 55: 5850-5860.

Espinasse, S., M. Gohar, D. Lereclus, and V. Sanchis. (2002). An ABC transporter from Bacillus thuringiensis is essential for β-exotoxin I production. J. Bacteriol. 184: 5848-5854.

Esser, L., F. Zhou, K.M. Pluchino, J. Shiloach, J. Ma, W.K. Tang, C. Gutierrez, A. Zhang, S. Shukla, J.P. Madigan, T. Zhou, P.D. Kwong, S.V. Ambudkar, M.M. Gottesman, and D. Xia. (2016). Structures of the Multidrug Transporter P-glycoprotein Reveal Asymmetric ATP Binding and the Mechanism of Polyspecificity. J. Biol. Chem. [Epub: Ahead of Print]

Eswarappa, S.M., K.K. Panguluri, M. Hensel, and D. Chakravortty. (2008). The yejABEF operon of Salmonella confers resistance to antimicrobial peptides and contributes to its virulence. Microbiology 154: 666-678.

Fachin, A.L., M.S. Ferreira-Nozawa, W. Maccheroni, Jr, and N.M. Martinez-Rossi. (2006). Role of the ABC transporter TruMDR2 in terbinafine, 4-nitroquinoline N-oxide and ethidium bromide susceptibility in Trichophyton rubrum. J. Med. Microbiol. 55: 1093-1099.

Fahmy, S.N., L.H. Khedr, S.A. Wahdan, E.T. Menze, S.S. Azab, and E. El-Demerdash. (2024). Effect of carvedilol on pharmacokinetics of sofosbuvir and its metabolite GS-331007: role of P-glycoprotein. J Pharm Pharmacol 76: 1051-1064.

Falcón-Pérez, J.M., M.J. Mazón, J. Molano, and P. Eraso. (1999). Functional domain analysis of the yeast ABC transporter Ycf1p by site-directed mutagenesis. J. Biol. Chem. 274: 23584-23590.

Falord, M., G. Karimova, A. Hiron, and T. Msadek. (2012). GraXSR proteins interact with the VraFG ABC transporter to form a five-component system required for cationic antimicrobial peptide sensing and resistance in Staphylococcus aureus. Antimicrob. Agents Chemother. 56: 1047-1058.

Fan, C., J.T. Kaiser, and D.C. Rees. (2020). A structural framework for unidirectional transport by a bacterial ABC exporter. Proc. Natl. Acad. Sci. USA. [Epub: Ahead of Print]

Fan, F., Y.X. Zhu, M.Y. Wu, W.X. Yin, G.Q. Li, M. Hahn, M.S. Hamada, and C.X. Luo. (2023). Mitochondrial Inner Membrane ABC Transporter Bcmdl1 Is Involved in Conidial Germination, Virulence, and Resistance to Anilinopyrimidine Fungicides in Botrytis cinerea. Microbiol Spectr e0010823. [Epub: Ahead of Print]

Fan, N., X. Li, W. Xie, X. Wei, Q. Fang, J. Xu, and C.F. Huang. (2024). Modulation of external and internal aluminum resistance by ALS3-dependent STAR1-mediated promotion of STOP1 degradation. New Phytol. [Epub: Ahead of Print]

Fan, Y.F., W. Zhang, L. Zeng, Z.N. Lei, C.Y. Cai, P. Gupta, D.H. Yang, Q. Cui, Z.D. Qin, Z.S. Chen, and L.D. Trombetta. (2018). Dacomitinib antagonizes multidrug resistance (MDR) in cancer cells by inhibiting the efflux activity of ABCB1 and ABCG2 transporters. Cancer Lett. [Epub: Ahead of Print]

Farhat, D., F. Rezaei, M. Ristovski, Y. Yang, A. Stancescu, L. Dzimkova, S. Samnani, J.F. Couture, and J.Y. Lee. (2022). Structural Analysis of Cholesterol Binding and Sterol Selectivity by ABCG5/G8. J. Mol. Biol. 434: 167795.

Farman, S., A. Javed, Arshia, K.M. Khan, A. Nasir, A.U. Khan, M.A. Lodhi, H. Gul, F. Khan, M. Asad, and Z. Parveen. (2020). Benzophenone Sulfonamide Derivatives as Interacting Partners and Inhibitors of Human P-glycoprotein. Anticancer Agents Med Chem 20: 1739-1751.

Fath, M.J. and R. Kolter. (1993). ABC transporters: bacterial exporters. Microbiol Rev 57: 995-1017.

Fay, A., P. Meyer, and J. Dworkin. (2010). Interactions between late-acting proteins required for peptidoglycan synthesis during sporulation. J. Mol. Biol. 399: 547-561.

Federici, L., B. Woebking, S. Velamakanni, R.A. Shilling, B. Luisi, and H.W. van Veen. (2007). New structure model for the ATP-binding cassette multidrug transporter LmrA. Biochem Pharmacol 74: 672-678.

Feissner, R.E., C.L. Richard-Fogal, E.R. Frawley, and R.G. Kranz. (2006). ABC transporter-mediated release of a haem chaperone allows cytochrome c biogenesis. Mol. Microbiol. 61: 219-231.

Fendley, G.A., I.L. Urbatsch, R.B. Sutton, M.E. Zoghbi, and G.A. Altenberg. (2016). Nucleotide dependence of the dimerization of ATP binding cassette nucleotide binding domains. Biochem. Biophys. Res. Commun. 480: 268-272.

Feng, L., S.N. Senchenkova, J. Yang, A.S. Shashkov, J. Tao, H. Guo, J. Cheng, Y. Ren, Y.A. Knirel, P.R. Reeves, and L. Wang. (2004). Synthesis of the heteropolysaccharide O antigen of Escherichia coli O52 requires an ABC transporter: structural and genetic evidence. J. Bacteriol. 186: 4510-4519.

Feng, Y., Q. Sun, G. Zhang, T. Wu, X. Zhang, X. Xu, Z. Han, and Y. Wang. (2019). Genome-Wide Identification and Characterization of ABC Transporters in Nine Rosaceae Species Identifying as a Possible Cytokinin Transporter linked to Dwarfing. Int J Mol Sci 20:.

Fernández-López, M., W. D''Haeze, P. Mergaert, C. Verplancke, J.C. Promé, M. Van Montagu, and M. Holsters. (1996). Role of nodl and nodJ in lipo-chitooligosaccharide secretion in Azorhizobium caulinodans and Escherichia coli. Mol. Microbiol. 20: 993-1000.

Ferreira, M.J. and I. Sá-Nogueira. (2010). A multitask ATPase serving different ABC-type sugar importers in Bacillus subtilis. J. Bacteriol. 192: 5312-5318.

Ferreira, M.J., A.L. Mendes, and I. de Sá-Nogueira. (2017). The MsmX ATPase plays a crucial role in pectin mobilization by Bacillus subtilis. PLoS One 12: e0189483.

Ferrero, S., D. Grimaldi, E. Arrigoni, M. Pironti, G.M. Zaccaria, B. Alessandria, E. Genuardi, G. De Luca, M. Ghislieri, R. Tavarozzi, A. Di Rocco, A. Re, V. Stefoni, F. Cavallo, C. Boccomini, M. Balzarotti, V.R. Zilioli, F. Moita, L. Arcaini, E. Lucchini, F. Ballerini, A.J.M. Ferreri, B. Puccini, G.A. Palumbo, S. Galimberti, S. Cortelazzo, A. Di Paolo, and M. Ladetto. (2023). Candidate germline biomarkers of lenalidomide efficacy in mantle cell lymphoma: the FIL MCL0208 trial. Blood Adv. [Epub: Ahead of Print]

Fetherston, J.D., V.J. Bertolino, and R.D. Perry. (1999). YbtP and YbtQ: two ABC transporters required for iron uptake in Yersinia pestis. Mol. Microbiol. 32: 289-299.

Fiedler, G., A.M. Muro-Pastor, E. Flores, and I. Maldener. (2001). NtcA-dependent expression of the devBCA operon, encoding a heterocyst-specific ATP-binding cassette transporter in Anabaena spp. J. Bacteriol. 183: 3795-3799.

Fiedler, G., M. Arnold, S. Hannus, and I. Maldener. (1998). The DevBCA exporter is essential for envelope formation in heterocysts of the cyanobacterium Anabaena sp. strain PCC 7120. Mol. Microbiol. 27: 1193-1202.

Fiedler, G., M. Pajatsch, and A. Böck. (1996). Genetics of a novel starch utilisation pathway present in Klebsiella oxytoca. J. Mol. Biol. 256: 279-291.

Finkenwirth F., Sippach M., Landmesser H., Kirsch F., Ogienko A., Grunzel M., Kiesler C., Steinhoff HJ., Schneider E. and Eitinger T. (2015). ATP-dependent Conformational Changes Trigger Substrate Capture and Release by an ECF-type Biotin Transporter. J Biol Chem. 290(27):16929-42.

Finkenwirth, F. and T. Eitinger. (2019). ECF-type ABC transporters for uptake of vitamins and transition metal ions into prokaryotic cells. Res. Microbiol. 170: 358-365.

Finkenwirth, F., M. Sippach, S.N. Pecina, M. Gäde, J. Ruta, A. Ricke, E. Bondarenko, J.P. Klare, M. Zinke, S. Lange, A. Lange, H.J. Steinhoff, and T. Eitinger. (2019). Dynamic interactions of CbiN and CbiM trigger activity of a cobalt energy-coupling-factor transporter. Biochim. Biophys. Acta. Biomembr 183114. [Epub: Ahead of Print]

Finkenwirth, F., O. Neubauer, J. Gunzenhäuser, J. Schoknecht, S. Scolari, M. Stöckl, T. Korte, A. Herrmann, and T. Eitinger. (2010). Subunit composition of an energy-coupling-factor-type biotin transporter analysed in living bacteria. Biochem. J. 431: 373-380.

Fitzpatrick, A.W.P., S. Llabrés, A. Neuberger, J.N. Blaza, X.C. Bai, U. Okada, S. Murakami, H.W. van Veen, U. Zachariae, S.H.W. Scheres, B.F. Luisi, and D. Du. (2017). Structure of the MacAB-TolC ABC-type tripartite multidrug efflux pump. Nat Microbiol 2: 17070.

Flores, E., M. Nieves-Morión, and C.W. Mullineaux. (2018). Cyanobacterial Septal Junctions: Properties and Regulation. Life (Basel) 9:.

Ford, R.C. and K. Beis. (2019). Learning the ABCs one at a time: structure and mechanism of ABC transporters. Biochem Soc Trans. [Epub: Ahead of Print]

Ford, R.C. and U.A. Hellmich. (2020). What monomeric nucleotide binding domains can teach us about dimeric ABC proteins. FEBS Lett. [Epub: Ahead of Print]

Formstone, A., R. Carballido-López, P. Noirot, J. Errington, and D.J. Scheffers. (2008). Localization and interactions of teichoic acid synthetic enzymes in Bacillus subtilis. J. Bacteriol. 190: 1812-1821.

Fourcroy, P., P. Sisó-Terraza, D. Sudre, M. Savirón, G. Reyt, F. Gaymard, A. Abadía, J. Abadia, A. Alvarez-Fernández, and J.F. Briat. (2014). Involvement of the ABCG37 transporter in secretion of scopoletin and derivatives by Arabidopsis roots in response to iron deficiency. New Phytol 201: 155-167.

Francis, G.A., M. Tsujita, and T.L. Terry. (1999). Apolipoprotein AI efficiently binds to and mediates cholesterol and phospholipid efflux from human but not rat aortic smooth muscle cells. Biochemistry 38: 16315-16322.

Francisco, R.M., A. Regalado, A. Ageorges, B.J. Burla, B. Bassin, C. Eisenach, O. Zarrouk, S. Vialet, T. Marlin, M.M. Chaves, E. Martinoia, and R. Nagy. (2013). ABCC1, an ATP binding cassette protein from grape berry, transports anthocyanidin 3-O-Glucosides. Plant Cell 25: 1840-1854.

Frank, N.Y., A. Margaryan, Y. Huang, T. Schatton, A.M. Waaga-Gasser, M. Gasser, M.H. Sayegh, W. Sadee, and M.H. Frank. (2005). ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res 65: 4320-4333.

Frank, N.Y., S.S. Pendse, P.H. Lapchak, A. Margaryan, D. Shlain, C. Doeing, M.H. Sayegh, and M.H. Frank. (2003). Regulation of progenitor cell fusion by ABCB5 P-glycoprotein, a novel human ATP-binding cassette transporter. J. Biol. Chem. 278: 47156-47165.

Frelet-Barrand, A., H.U. Kolukisaoglu, S. Plaza, M. Rüffer, L. Azevedo, S. Hörtensteiner, K. Marinova, B. Weder, B. Schulz, and M. Klein. (2008). Comparative mutant analysis of Arabidopsis ABCC-type ABC transporters: AtMRP2 contributes to detoxification, vacuolar organic anion transport and chlorophyll degradation. Plant Cell Physiol. 49: 557-569.

Fremaux, C., Y. Héchard, and Y. Cenatiempo. (1995). Mesentericin Y105 gene clusters in Leuconostoc mesenteroides Y105. Microbiology 141(Pt7): 1637-1645.

Frías, J.E., E. Flores, and A. Herrero. (1997). Nitrate assimilation gene cluster from the heterocyst-forming cyanobacterium Anabaena sp. strain PCC 7120. J. Bacteriol. 179: 477-486.

Fribourg PF., Chami M., Sorzano CO., Gubellini F., Marabini R., Marco S., Jault JM. and Levy D. (2014). 3D cryo-electron reconstruction of BmrA, a bacterial multidrug ABC transporter in an inward-facing conformation and in a lipidic environment. J Mol Biol. 426(10):2059-69.

Frock, A.D., S.R. Gray, and R.M. Kelly. (2012). Hyperthermophilic Thermotoga species differ with respect to specific carbohydrate transporters and glycoside hydrolases. Appl. Environ. Microbiol. 78: 1978-1986.

Frossard, S.M., A.A. Khan, E.C. Warrick, J.M. Gately, A.D. Hanson, M.L. Oldham, D.A. Sanders, and L.N. Csonka. (2012). Identification of a third osmoprotectant transport system, the osmU system, in Salmonella enterica. J. Bacteriol. 194: 3861-3871.

Fujita M. and Shinozaki K. (2014). Identification of polyamine transporters in plants: paraquat transport provides crucial clues. Plant Cell Physiol. 55(5):855-61.

Fujita, K. and K. Ichida. (2018). ABCG2 as a therapeutic target candidate for gout. Expert Opin Ther Targets 22: 123-129.

Fujiwara, T. and H. Harigae. (2013). Pathophysiology and genetic mutations in congenital sideroblastic anemia. Pediatr Int 55: 675-679.

Fukuda, S., H. Toh, T.D. Taylor, H. Ohno, and M. Hattori. (2012). Acetate-producing bifidobacteria protect the host from enteropathogenic infection via carbohydrate transporters. Gut Microbes 3: 449-454.

Fukushima, S., M. Yoshimura, T. Chibazakura, T. Sato, and H. Yoshikawa. (2006). The putative ABC transporter YheH/YheI is involved in the signalling pathway that activates KinA during sporulation initiation. FEMS Microbiol. Lett. 256: 90-97.

Fulyani, F., G.K. Schuurman-Wolters, A.V. Zagar, A. Guskov, D.J. Slotboom, and B. Poolman. (2013). Functional diversity of tandem substrate-binding domains in ABC transporters from pathogenic bacteria. Structure 21: 1879-1888.

Fulyani, F., G.K. Schuurman-Wolters, D.J. Slotboom, and B. Poolman. (2015). Relative rates of amino acid import via the ABC transporter GlnPQ determine the growth performance of Lactococcus lactis. J. Bacteriol. [Epub: Ahead of Print]

Furuta, T. (2021). Structural dynamics of ABC transporters: molecular simulation studies. Biochem Soc Trans 49: 405-414.

Futamata, R., F. Ogasawara, T. Ichikawa, A. Kodan, Y. Kimura, N. Kioka, and K. Ueda. (2020). FRET analyses reveal a role of ATP hydrolysis-associated conformational changes in human P-glycoprotein. J. Biol. Chem. [Epub: Ahead of Print]

Gaballa, A. and J.D. Helmann. (2007). Substrate induction of siderophore transport in Bacillus subtilis mediated by a novel one-component regulator. Mol. Microbiol. 66: 164-173.

Gaballa, A., T. Wang, R.W. Ye, and J.D. Helmann. (2002). Functional analysis of the Bacillus subtilis Zur regulon. J. Bacteriol. 184: 6508-6514.

Gadsby, D.C., P. Vergani, and L. Csanády. (2006). The ABC protein turned chloride channel whose failure causes cystic fibrosis. Nature 440: 477-483.

Gál, Z., C. Hegedüs, G. Szakács, A. Váradi, B. Sarkadi, and C. Özvegy-Laczka. (2015). Mutations of the central tyrosines of putative cholesterol recognition amino acid consensus (CRAC) sequences modify folding, activity, and sterol-sensing of the human ABCG2 multidrug transporter. Biochim. Biophys. Acta. 1848: 477-487.

Galazzo, L., G. Meier, D. Januliene, K. Parey, D. De Vecchis, B. Striednig, H. Hilbi, L.V. Schäfer, I. Kuprov, A. Moeller, E. Bordignon, and M.A. Seeger. (2022). The ABC transporter MsbA adopts the wide inward-open conformation in cells. Sci Adv 8: eabn6845.

Galián, C., F. Manon, M. Dezi, C. Torres, C. Ebel, D. Lévy, and J.M. Jault. (2011). Optimized purification of a heterodimeric ABC transporter in a highly stable form amenable to 2-D crystallization. PLoS One 6: e19677.

Ganapathy, V. and S. Miyauchi. (2005). Transport systems for opioid peptides in mammalian tissues. AAPS J 7: E852-856.

Gao, B., H. Vorwerk, C. Huber, M. Lara-Tejero, J. Mohr, A.L. Goodman, W. Eisenreich, J.E. Galán, and D. Hofreuter. (2017). Metabolic and fitness determinants for in vitro growth and intestinal colonization of the bacterial pathogen Campylobacter jejuni. PLoS Biol 15: e2001390.

Gao, H., Y. Wang, X. Fei, D.A. Wright, and M.H. Spalding. (2015). Expression activation and functional analysis of HLA3, a putative inorganic carbon transporter in Chlamydomonas reinhardtii. Plant J. 82: 1-11.

Garault, P., D. Le Bars, C. Besset, and V. Monnet. (2002). Three oligopeptide-binding proteins are involved in the oligopeptide transport of Streptococcus thermophilus. J. Biol. Chem. 277: 32-39.

Garces, F.A., J.F. Scortecci, and R.S. Molday. (2020). Functional Characterization of ABCA4 Missense Variants Linked to Stargardt Macular Degeneration. Int J Mol Sci 22:.

Garrigues, A., A.E. Escargueil, and S. Orlowski. (2002). The multidrug transporter, P-glycoprotein, actively mediates cholesterol redistribution in the cell membrane. Proc. Natl. Acad. Sci. USA 99: 10347-10352.

Gebhard, S. and G.M. Cook. (2008). Differential regulation of high-affinity phosphate transport systems of Mycobacterium smegmatis: identification of PhnF, a repressor of the phnDCE operon. J. Bacteriol. 190: 1335-1343.

Geddes, B.A., B.S. Pickering, N.J. Poysti, H. Collins, H. Yudistira, and I.J. Oresnik. (2010). A locus necessary for the transport and catabolism of erythritol in Sinorhizobium meliloti. Microbiology 156: 2970-2981.

Geller, B.L., R.G. Ivey, J.E. Trempy, and B. Hettinger-Smith. (1993). Cloning of a chromosomal gene required for phage infection of Lactococcus lactis subsp. lactis C2. J. Bacteriol. 175: 5510-5519.

George AM. and Jones PM. (2012). Perspectives on the structure-function of ABC transporters: the Switch and Constant Contact models. Prog Biophys Mol Biol. 109(3):95-107.

Gerber, S., M. Comellas-Bigler, B.A. Goetz, and K.P. Locher. (2008). Structural basis of trans-inhibition in a molybdate/tungstate ABC transporter. Science 321: 246-250.

Gerwe, B., L.L. Kelley, B.D. Dillard, T. Lai, Z.J. Liu, W. Tempel, L. Chen, J. Habel, D. Lee, F.E. Jenney, Jr, F.J. Sugar, J.S. Richardson, D.C. Richardson, M.G. Newton, B.C. Wang, M.W. Adams, and J.P. Rose. (2007). Structural and transcriptional analyses of a purine nucleotide-binding protein from Pyrococcus furiosus: a component of a novel, membrane-bound multiprotein complex unique to this hyperthermophilic archaeon. J Struct Funct Genomics 8: 1-10.

Ghssein, G., C. Brutesco, L. Ouerdane, C. Fojcik, A. Izaute, S. Wang, C. Hajjar, R. Lobinski, D. Lemaire, P. Richaud, R. Voulhoux, A. Espaillat, F. Cava, D. Pignol, E. Borezée-Durant, and P. Arnoux. (2016). Biosynthesis of a broad-spectrum nicotianamine-like metallophore in Staphylococcus aureus. Science 352: 1105-1109.

Gibson, K., J.K. Chu, S. Zhu, D. Nguyen, J. Mrázek, J. Liu, and T.R. Hoover. (2022). A Tripartite Efflux System Affects Flagellum Stability in. Int J Mol Sci 23:.

Gil, J.P. and S. Krishna. (2017). pfmdr1 (Plasmodium falciparum multidrug drug resistance gene 1): a pivotal factor in malaria resistance to artemisinin combination therapies. Expert Rev Anti Infect Ther. [Epub: Ahead of Print]

Gilson, E., C.F. Higgins, M. Hofnung, G. Ferro-Luzzi Ames, and H. Nikaido. (1982). Extensive homology between membrane-associated components of histidine and maltose transport systems of Salmonella typhimurium and Escherichia coli. J. Biol. Chem. 257: 9915-9918.

Gimmestad, M., M. Steigedal, H. Ertesvåg, S. Moreno, B.E. Christensen, G. Espín, and S. Valla. (2006). Identification and characterization of an Azotobacter vinelandii type I secretion system responsible for export of the AlgE-type mannuronan C-5-epimerases. J. Bacteriol. 188: 5551-5560.

Glaser, P., H. Sakamoto, J. Bellalou, A. Ullmann, and A. Danchin. (1988). Secretion of cyclolysin, the calmodulin-sensitive adenylate cyclase-haemolysin bifunctional protein of Bordetella pertussis. EMBO. J. 7: 3997-4004.

Gochenauer, A.E., D.L. Dreger, B.W. Davis, S. Cook, K.E. Barber, and K.J. Ekenstedt. (2022). Polymorphisms and Predictive Fluoroquinolone Phototoxicity in Nondomestic Felids. Genes (Basel) 13:.

Godinho, C.P., C.S. Prata, S.N. Pinto, C. Cardoso, N.M. Bandarra, F. Fernandes, and I. Sá-Correia. (2018). Pdr18 is involved in yeast response to acetic acid stress counteracting the decrease of plasma membrane ergosterol content and order. Sci Rep 8: 7860.

Gokirmak T., Campanale JP., Shipp LE., Moy GW., Tao H. and Hamdoun A. (2012). Localization and substrate selectivity of sea urchin multidrug (MDR) efflux transporters. J Biol Chem. 287(52):43876-83.

Golin, J., Z.N. Kon, C.P. Wu, J. Martello, L. Hanson, S. Supernavage, S.V. Ambudkar, and Z.E. Sauna. (2007). Complete inhibition of the Pdr5p multidrug efflux pump ATPase activity by its transport substrate clotrimazole suggests that GTP as well as ATP may be used as an energy source. Biochemistry 46: 13109-13119.

Gominet, M., N. Seghezzi, and P. Mazodier. (2011). Acyl depsipeptide (ADEP) resistance in Streptomyces. Microbiology 157: 2226-2234.

Gompf, S., A. Zutz, M. Hofacker, W. Haase, C. van der Does, and R. Tampé. (2007). Switching of the homooligomeric ATP-binding cassette transport complex MDL1 from post-translational mitochondrial import to endoplasmic reticulum insertion. FEBS J. 274: 5298-5310.

Gonsorcikova, L., M. Vaxillaire, S. Pruhova, A. Dechaume, P. Dusatkova, O. Cinek, O. Pedersen, P. Froguel, T. Hansen, and J. Lebl. (2011). Familial mild hyperglycemia associated with a novel ABCC8-V84I mutation within three generations. Pediatr Diabetes 12: 266-269.

González-Guerrero, M., K. Benabdellah, A. Valderas, C. Azcón-Aguilar, and N. Ferrol. (2010). GintABC1 encodes a putative ABC transporter of the MRP subfamily induced by Cu, Cd, and oxidative stress in Glomus intraradices. Mycorrhiza 20: 137-146.

Gonzalez-Lobato, L., V. Chaptal, J. Molle, and P. Falson. (2016). Leishmania tarentolae as a Promising Tool for Expressing Polytopic and Multi-Transmembrane Spans Eukaryotic Membrane Proteins: The Case of the ABC Pump ABCG6. Methods Mol Biol 1432: 119-131.

González-Pastor, J.E., E.C. Hobbs, and R. Losick. (2003). Cannibalism by sporulating bacteria. Science 301: 510-513.

González-Pons, M., A.C. Szeto, R. González-Méndez, and A.E. Serrano. (2009). Identification and bioinformatic characterization of a multidrug resistance associated protein (ABCC) gene in Plasmodium berghei. Malar J 8: 1.

Good, J.R., M. Cabral, S. Sharma, J. Yang, N. Van Driessche, C.A. Shaw, G. Shaulsky, and A. Kuspa. (2003). TagA, a putative serine protease/ABC transporter of Dictyostelium that is required for cell fate determination at the onset of development. Development 130: 2953-2965.

Goodman, C.D., P. Casati, and V. Walbot. (2004). A multidrug resistance-associated protein involved in anthocyanin transport in Zea mays. Plant Cell 16: 1812-1826.

Gopisetty, M.K., D.I. Adamecz, F.I. Nagy, &.#.1.9.3.;. Baji, V. Lathira, M.R. Szabó, R. Gáspár, T. Csont, &.#.2.0.1.;. Frank, and M. Kiricsi. (2021). Androstano-arylpyrimidines: Novel small molecule inhibitors of MDR1 for sensitizing multidrug-resistant breast cancer cells. Eur J Pharm Sci 156: 105587.

Gottesman, M.M., C.A. Hrycyna, P.V. Schoenlein, U.A. Germann, and I. Pastan. (1995). Genetic analysis of the multidrug transporter. Annu Rev Genet 29: 607-649.

Gottschalk, B., G. Bröker, M. Kuhn, S. Aymanns, U. Gleich-Theurer, and B. Spellerberg. (2006). Transport of multidrug resistance substrates by the Streptococcus agalactiae hemolysin transporter. J. Bacteriol. 188: 5984-5992.

Graab, P., C. Bock, K. Weiss, A. Hirth, N. Koller, M. Braner, J. Jung, F. Loehr, R. Tampé, C. Behrends, and R. Abele. (2019). Lysosomal targeting of the ABC transporter TAPL is determined by membrane localized charged residues. J. Biol. Chem. [Epub: Ahead of Print]

Graf, S.A., S.E. Haigh, E.D. Corson, and O.S. Shirihai. (2004). Targeting, import, and dimerization of a mammalian mitochondrial ATP binding cassette (ABC) transporter, ABCB10 (ABC-me). J. Biol. Chem. 279: 42954-42963.

Grasekamp, K.P., B. Beaud Benyahia, N. Taib, B. Audrain, B. Bardiaux, Y. Rossez, N. Izadi-Pruneyre, M. Lejeune, X. Trivelli, Z. Chouit, Y. Guerardel, J.M. Ghigo, S. Gribaldo, and C. Beloin. (2023). The Mla system of diderm Firmicute Veillonella parvula reveals an ancestral transenvelope bridge for phospholipid trafficking. Nat Commun 14: 7642.

Grec, S., D. Vanham, J.C. de Ribaucourt, B. Purnelle, and M. Boutry. (2003). Identification of regulatory sequence elements within the transcription promoter region of NpABC1, a gene encoding a plant ABC transporter induced by diterpenes. Plant J. 35: 237-250.

Green, R.M., F. Hoda, and K.L. Ward. (2000). Molecular cloning and characterization of the murine bile salt export pump. Gene 241: 117-123.

Greene, N.P., E. Kaplan, A. Crow, and V. Koronakis. (2018). Antibiotic Resistance Mediated by the MacB ABC Transporter Family: A Structural and Functional Perspective. Front Microbiol 9: 950.

Greiner, T., A. Moroni, J.L. Van Etten, and G. Thiel. (2018). Genes for Membrane Transport Proteins: Not So Rare in Viruses. Viruses 10:.

Gribenko, A., L. Mosyak, S. Ghosh, K. Parris, K. Svenson, J. Moran, L. Chu, S. Li, T. Liu, V.L. Woods, Jr, K.U. Jansen, B.A. Green, A.S. Anderson, and Y.V. Matsuka. (2013). Three-dimensional structure and biophysical characterization of Staphylococcus aureus cell surface antigen-manganese transporter MntC. J. Mol. Biol. 425: 3429-3445.

Grigoras, I., M. Lazard, P. Plateau, and S. Blanquet. (2008). Functional characterization of the Saccharomyces cerevisiae ABC-transporter Yor1p overexpressed in plasma membranes. Biochim. Biophys. Acta. 1778: 68-78.

Gronkowska, K., S. Michlewska, and A. Robaszkiewicz. (2023). Activity of Lysosomal ABCC3, ABCC5 and ABCC10 is Responsible for Lysosomal Sequestration of Doxorubicin and Paclitaxel-Oregongreen488 in Paclitaxel-Resistant Cancer Cell Lines. Cell Physiol Biochem 57: 360-378.

Grunden, A.M., R.M. Ray, J.K. Rosentel, F.G. Healy, and K.T. Shanmugam. (1996). Repression of the Escherichia coli modABCD (molybdate transport) operon by ModE. J. Bacteriol. 178: 735-744.

Gu RX., Corradi V., Singh G., Choudhury HG., Beis K. and Tieleman DP. (2015). Conformational Changes of the Antibacterial Peptide ATP Binding Cassette Transporter McjD Revealed by Molecular Dynamics Simulations. Biochemistry. 54(38):5989-98.

Guefrachi, I., O. Pierre, T. Timchenko, B. Alunni, Q. Barrière, P. Czernic, J.A. Villaécija-Aguilar, C. Verly, M. Bourge, J. Fardoux, M. Mars, E. Kondorosi, E. Giraud, and P. Mergaert. (2015). Bradyrhizobium BclA Is a Peptide Transporter Required for Bacterial Differentiation in Symbiosis with Aeschynomene Legumes. Mol. Plant Microbe Interact. 28: 1155-1166.

Guffick, C., P.Y. Hsieh, A. Ali, W. Shi, J. Howard, D.K. Chinthapalli, A.C. Kong, I. Salaa, L.I. Crouch, M.R. Ansbro, S.C. Isaacson, H. Singh, N.P. Barrera, A.V. Nair, C.V. Robinson, M.J. Deery, and H.W. van Veen. (2022). Drug-dependent inhibition of nucleotide hydrolysis in the heterodimeric ABC multidrug transporter PatAB from Streptococcus pneumoniae. FEBS J. [Epub: Ahead of Print]

Gul N. and Poolman B. (2013). Functional reconstitution and osmoregulatory properties of the ProU ABC transporter from Escherichia coli. Mol Membr Biol. 30(2):138-48.

Gul, N., G. Schuurman-Wolters, A. Karasawa, and B. Poolman. (2012). Functional characterization of amphipathic α-helix in the osmoregulatory ABC transporter OpuA. Biochemistry 51: 5142-5152.

Guo, D., M.G. Bowden, R. Pershad, and H.B. Kaplan. (1996). The Myxococcus xanthus rfbABC operon encodes an ATP-binding cassette transporter homolog required for O-antigen biosynthesis and multicellular development. J. Bacteriol. 178: 1631-1639.

Guo, Y., E. Kotova, Z.S. Chen, K. Lee, E. Hopper-Borge, M.G. Belinsky, and G.D. Kruh. (2003). MRP8, ATP-binding cassette C11 (ABCC11), is a cyclic nucleotide efflux pump and a resistance factor for fluoropyrimidines 2',3'-dideoxycytidine and 9'-(2'-phosphonylmethoxyethyl)adenine. J. Biol. Chem. 278: 29509-29514.

Guo, Z., Y. Shi, B. Jiang, X. Peng, L. Zhang, C. Tu, and T. Wang. (2023). Psoraleae Fructus Ethanol Extract Induced Hepatotoxicity via Impaired Lipid Metabolism Caused by Disruption of Fatty Acid -Oxidation. Oxid Med Cell Longev 2023: 4202861.

Gupta, D., K.E. Shalvarjian, and D.D. Nayak. (2022). An Archaea-specific -type cytochrome maturation machinery is crucial for methanogenesis in. Elife 11:.

Gustot, A., Smriti, J.M. Ruysschaert, H. McHaourab, and C. Govaerts. (2010). Lipid composition regulates the orientation of transmembrane helices in HorA, an ABC multidrug transporter. J. Biol. Chem. 285: 14144-14151.

Guthmiller, J.M., D. Kolodrubetz, and E. Kraig. (1995). Mutational analysis of the putative leukotoxin transport genes in Actinobacillus actinomycetemcomitans. Microb. Pathog. 18: 307-321.

Gutiérrez-Preciado, A., A.G. Torres, E. Merino, H.R. Bonomi, F.A. Goldbaum, and V.A. García-Angulo. (2015). Extensive Identification of Bacterial Riboflavin Transporters and Their Distribution across Bacterial Species. PLoS One 10: e0126124.

Gyimesi, G., S. Ramachandran, P. Kota, N.V. Dokholyan, B. Sarkadi, and T. Hegedus. (2011). ATP hydrolysis at one of the two sites in ABC transporters initiates transport related conformational transitions. Biochim. Biophys. Acta. 1808: 2954-2964.

Hafiane, A., K. Gasbarrino, and S.S. Daskalopoulou. (2019). Adiponectin and cholesterol efflux. Metabolism 153953. [Epub: Ahead of Print]

Haider, A.J., M.H. Cox, N. Jones, A.J. Goode, K.S. Bridge, K. Wong, D. Briggs, and I.D. Kerr. (2015). Identification of residues in ABCG2 affecting protein trafficking and drug transport, using co-evolutionary analysis of ABCG sequences. Biosci Rep 35:.

Haimeur, A., R.G. Deeley, and S.P. Cole. (2002). Charged amino acids in the sixth transmembrane helix of multidrug resistance protein 1 (MRP1/ABCC1) are critical determinants of transport activity. J. Biol. Chem. 277: 41326-41333.

Halenius, A., F. Momburg, H. Reinhard, D. Bauer, M. Lobigs, and H. Hengel. (2006). Physical and functional interactions of the cytomegalovirus US6 glycoprotein with the transporter associated with antigen processing. J. Biol. Chem. 281: 5383-5390.

Hammond, C.L., R. Marchan, S.M. Krance, and N. Ballatori. (2007). Glutathione export during apoptosis requires functional multidrug resistance-associated proteins. J. Biol. Chem. 282: 14337-14347.

Hanekop, N., M. Höing, L. Sohn-Bösser, M. Jebbar, L. Schmitt, and E. Bremer. (2007). Crystal structure of the ligand-binding protein EhuB from Sinorhizobium meliloti reveals substrate recognition of the compatible solutes ectoine and hydroxyectoine. J. Mol. Biol. 374: 1237-1250.

Haney, D.Q. (1999). New class of antibiotics may help doctors overcome resistant germs. Assoc. Press.

Hanks, T.S., M. Liu, M.J. McClure, and B. Lei. (2005). ABC transporter FtsABCD of Streptococcus pyogenes mediates uptake of ferric ferrichrome. BMC Microbiol 5: 62.

Haque, A., G.A. Baig, A.S. Alshawli, K.H.W. Sait, B.B. Hafeez, M.K. Tripathi, B.S. Alghamdi, H.S.H. Mohammed Ali, and M. Rasool. (2022). Interaction Analysis of MRP1 with Anticancer Drugs Used in Ovarian Cancer: In Silico Approach. Life (Basel) 12:.

Harakalova M., van Harssel JJ., Terhal PA., van Lieshout S., Duran K., Renkens I., Amor DJ., Wilson LC., Kirk EP., Turner CL., Shears D., Garcia-Minaur S., Lees MM., Ross A., Venselaar H., Vriend G., Takanari H., Rook MB., van der Heyden MA., Asselbergs FW., Breur HM., Swinkels ME., Scurr IJ., Smithson SF., Knoers NV., van der Smagt JJ., Nijman IJ., Kloosterman WP., van Haelst MM., van Haaften G. and Cuppen E. (2012). Dominant missense mutations in ABCC9 cause Cantu syndrome. Nat Genet. 44(7):793-6.

Harders, R.H., T.H. Morthorst, L.E. Landgrebe, A.D. Lande, M.S. Fuglsang, S.B. Mortensen, V. Feteira-Montero, H.H. Jensen, J.B. Wesseltoft, and A. Olsen. (2024). CED-6/GULP and components of the clathrin-mediated endocytosis machinery act redundantly to correctly display CED-1 on the cell membrane in Caenorhabditis elegans. G3 (Bethesda) 14:.

Hartinger, E.M., A. Mahringer, S.K. Choudhuri, G. Fricker, and T. Efferth. (2023). Modulatory Activity of the Copper Chelate, Copper N-(2-Hydroxy Acetophenone) Glycinate, in ABC-transporter-expressing Cell Lines. Anticancer Res 43: 1031-1041.

Haschka, T., F. Lamari, F. Mochel, and V. Zujovic. (2024). Innovative tree-based method for sampling molecular conformations: exploring the ATP-binding cassette subfamily D member 1 (ABCD1) transporter as a case study. Front Mol Biosci 11: 1440529.

Hashimoto, Y., N. Li, H. Yokoyama, and T. Ezaki. (1993). Complete nucleotide sequence and molecular characterization of ViaB region encoding Vi antigen in Salmonella typhi. J. Bacteriol. 175: 4456-4465.

Hayashi T., Chiba S., Kaneta Y., Furuta T. and Sakurai M. (2014). ATP-induced conformational changes of nucleotide-binding domains in an ABC transporter. Importance of the water-mediated entropic force. J Phys Chem B. 118(44):12612-20.

Hazlett, K.R., F. Rusnak, D.G. Kehres, S.W. Bearden, C.J. La Vake, M.E. La Vake, M.E. Maguire, R.D. Perry, and J.D. Radolf. (2003). The Treponema pallidum tro operon encodes a multiple metal transporter, a zinc-dependent transcriptional repressor, and a semi-autonomously expressed phosphoglycerate mutase. J. Biol. Chem. 278: 20687-20694.

He, Q., C.R. Vossbrinck, Q. Yang, X.Z. Meng, J. Luo, G.Q. Pan, Z.Y. Zhou, and T. Li. (2018). Evolutionary and functional studies on microsporidian ATP-binding cassettes: Insights into the adaptation of microsporidia to obligated intracellular parasitism. Infect Genet Evol 68: 136-144. [Epub: Ahead of Print]

He, S.M., R. Li, J.R. Kanwar, and S.F. Zhou. (2011). Structural and functional properties of human multidrug resistance protein 1 (MRP1/ABCC1). Curr. Med. Chem. 18: 439-481.

He, Y., Y. Liu, and S. Zeng. (2010). Stereoselective and multiple carrier-mediated transport of cetirizine across Caco-2 cell monolayers with potential drug interaction. Chirality 22: 684-692.

Hebbeln, P., D.A. Rodionov, A. Alfandega, and T. Eitinger. (2007). Biotin uptake in prokaryotes by solute transporters with an optional ATP-binding cassette-containing module. Proc. Natl. Acad. Sci. USA 104: 2909-2914.

Hegyi, Z. and L. Homolya. (2016). Functional Cooperativity between ABCG4 and ABCG1 Isoforms. PLoS One 11: e0156516.

Hemmer, M., S. Krawczyk, I. Simon, H. Lage, and A. Hilgeroth. (2015). Discovery of substituted 1,4-dihydroquinolines as novel class of ABCB1 modulators. Bioorg Med Chem 23: 5015-5021.

Henrich, A., N. Kuhlmann, A.W. Eck, R. Krämer, and G.M. Seibold. (2013). Maltose uptake by the novel ABC transport system MusEFGK2I causes increased expression of ptsG in Corynebacterium glutamicum. J. Bacteriol. 195: 2573-2584.

Herget, M., C. Baldauf, C. Schölz, D. Parcej, K.H. Wiesmüller, R. Tampé, R. Abele, and E. Bordignon. (2011). Conformation of peptides bound to the transporter associated with antigen processing (TAP). Proc. Natl. Acad. Sci. USA 108: 1349-1354.

Herget, M., N. Kreissig, C. Kolbe, C. Schölz, R. Tampé, and R. Abele. (2009). Purification and reconstitution of the antigen transport complex TAP: a prerequisite for determination of peptide stoichiometry and ATP hydrolysis. J. Biol. Chem. 284: 33740-33749.

Herrou, J. and S. Crosson. (2013). myo-Inositol and D-Ribose Ligand Discrimination in an ABC Periplasmic Binding Protein. J. Bacteriol. 195: 2379-2388.

Hettema, E.H., C.W. van Roermund, B. Distel, M. van den Berg, C. Vilela, C. Rodrigues-Pousada, R.J. Wanders, and H.F. Tabak. (1996). The ABC transporter proteins Pat1 and Pat2 are required for import of long-chain fatty acids into peroxisomes of Saccharomyces cerevisiae. EMBO. J. 15: 3813-3822.

Heuveling, J., H. Landmesser, and E. Schneider. (2018). One intact transmembrane substrate binding site is sufficient for function of the homodimeric type I ATP-binding cassette importer for positively charged amino acids Art(MP)of. J. Bacteriol. [Epub: Ahead of Print]

Heuveling, J., H. Landmesser, and E. Schneider. (2019). Evidence from Mutational Analysis for a Single Transmembrane Substrate Binding Site in the Histidine ATP-Binding Cassette Transporter of Salmonella enterica Serovar Typhimurium. J. Bacteriol. 201:.

Heuveling, J., V. Frochaux, J. Ziomkowska, R. Wawrzinek, P. Wessig, A. Herrmann, and E. Schneider. (2014). Conformational changes of the bacterial type I ATP-binding cassette importer HisQMP2 at distinct steps of the catalytic cycle. Biochim. Biophys. Acta. 1838: 106-116.

Higgins, C.F. (1995). The ABC of channel regulation. Cell 82: 693-696.

Hillebrand, M., S.E. Verrier, A. Ohlenbusch, A. Schäfer, H.D. Söling, F.S. Wouters, and J. Gärtner. (2007). Live cell FRET microscopy: homo- and heterodimerization of two human peroxisomal ABC transporters, the adrenoleukodystrophy protein (ALDP, ABCD1) and PMP70 (ABCD3). J. Biol. Chem. 282: 26997-27005.

Hillerich, B. and J. Westpheling. (2006). A new GntR family transcriptional regulator in streptomyces coelicolor is required for morphogenesis and antibiotic production and controls transcription of an ABC transporter in response to carbon source. J. Bacteriol. 188: 7477-7487.

Hinsa, S.M., M. Espinosa-Urgel, J.L. Ramos, and G.A. O'Toole. (2003). Transition from reversible to irreversible attachment during biofilm formation by Pseudomonas fluorescens WCS365 requires an ABC transporter and a large secreted protein. Mol. Microbiol. 49: 905-918.

Hinz, A. and R. Tampé. (2012). ABC Transporters and Immunity: Mechanism of Self-Defense. Biochemistry 51: 4981-4989.

Hiron, A., B. Posteraro, M. Carrière, L. Remy, C. Delporte, M. La Sorda, M. Sanguinetti, V. Juillard, and E. Borezée-Durant. (2010). A nickel ABC-transporter of Staphylococcus aureus is involved in urinary tract infection. Mol. Microbiol. 77: 1246-1260.

Hiron, A., E. Borezée-Durant, J.C. Piard, and V. Juillard. (2007). Only one of four oligopeptide transport systems mediates nitrogen nutrition in Staphylococcus aureus. J. Bacteriol. 189: 5119-5129.

Hirose, T. and H.R. Horvitz. (2014). The translational regulators GCN-1 and ABCF-3 act together to promote apoptosis in C. elegans. PLoS Genet 10: e1004512.

Hirota, R., Z.I. Katsuura, N. Momokawa, H. Murakami, S. Watanabe, T. Ishida, T. Ikeda, H. Funabashi, and A. Kuroda. (2022). Gatekeeper Residue Replacement in a Phosphite Transporter Enhances Mutational Robustness of the Biocontainment Strategy. ACS Synth Biol 11: 3397-3404.

Hitti-Malin, R.J., D.M. Panneman, Z. Corradi, E.G.M. Boonen, G. Astuti, C.M. Dhaenens, H. Stöhr, B.H.F. Weber, D. Sharon, E. Banin, M. Karali, S. Banfi, T. Ben-Yosef, D. Glavač, G.J. Farrar, C. Ayuso, P. Liskova, L. Dudakova, M. Vajter, M. Ołdak, J.P. Szaflik, A. Matynia, M.B. Gorin, K. Kämpjärvi, M. Bauwens, E. De Baere, C.B. Hoyng, C.H.Z. Li, C.C.W. Klaver, C.F. Inglehearn, K. Fujinami, C. Rivolta, R. Allikmets, J. Zernant, W. Lee, O.L. Podhajcer, A. Fakin, J. Sajovic, A. AlTalbishi, S. Valeina, G. Taurina, A.L. Vincent, L. Roberts, R. Ramesar, G. Sartor, E. Luppi, S.M. Downes, L.I. van den Born, T.L. McLaren, J.N. De Roach, T.M. Lamey, J.A. Thompson, F.K. Chen, A.M. Tracewska, S. Kamakari, J.M.F. Sallum, H.J. Bolz, H. Kayserili, S. Roosing, and F.P.M. Cremers. (2024). Towards Uncovering the Role of Incomplete Penetrance in Maculopathies through Sequencing of 105 Disease-Associated Genes. Biomolecules 14:.

Ho, H., A. Miu, M.K. Alexander, N.K. Garcia, A. Oh, I. Zilberleyb, M. Reichelt, C.D. Austin, C. Tam, S. Shriver, H. Hu, S.S. Labadie, J. Liang, L. Wang, J. Wang, Y. Lu, H.E. Purkey, J. Quinn, Y. Franke, K. Clark, M.H. Beresini, M.W. Tan, B.D. Sellers, T. Maurer, M.F.T. Koehler, A.T. Wecksler, J.R. Kiefer, V. Verma, Y. Xu, M. Nishiyama, J. Payandeh, and C.M. Koth. (2018). Structural basis for dual-mode inhibition of the ABC transporter MsbA. Nature. [Epub: Ahead of Print]

Hohl, M., C. Briand, M.G. Grütter, and M.A. Seeger. (2012). Crystal structure of a heterodimeric ABC transporter in its inward-facing conformation. Nat Struct Mol Biol 19: 395-402.

Hohl, M., L.M. Hürlimann, S. Böhm, J. Schöppe, M.G. Grütter, E. Bordignon, and M.A. Seeger. (2014). Structural basis for allosteric cross-talk between the asymmetric nucleotide binding sites of a heterodimeric ABC exporter. Proc. Natl. Acad. Sci. USA 111: 11025-11030.

Holland, I.B. and M.A. Blight. (1999). ABC-ATPases, adaptable energy generators fuelling transmembrane movement of a variety of molecules in organisms from bacteria to humans. J. Mol. Biol. 293: 381-399.

Holland, I.B., S.P.C. Cole, K. Kuchler, and C.F. Higgins (Eds.). (2003). ABC Proteins: From Bacteria to Man. London: Academic Press/Elsevier Science.

Hollenstein, K., D.C. Frei, and K.P. Locher. (2007). Structure of an ABC transporter in complex with its binding protein. Nature 446: 213-216.

Holmes, A.R., S. Tsao, S.W. Ong, E. Lamping, K. Niimi, B.C. Monk, M. Niimi, A. Kaneko, B.R. Holland, J. Schmid, and R.D. Cannon. (2006). Heterozygosity and functional allelic variation in the Candida albicans efflux pump genes CDR1 and CDR2. Mol. Microbiol. 62: 170-186.

Holmes, A.R., Y.H. Lin, K. Niimi, E. Lamping, M. Keniya, M. Niimi, K. Tanabe, B.C. Monk, and R.D. Cannon. (2008). ABC transporter Cdr1p contributes more than Cdr2p does to fluconazole efflux in fluconazole-resistant Candida albicans clinical isolates. Antimicrob. Agents Chemother. 52: 3851-3862.

Holyoake, L.V., S. Hunt, G. Sanguinetti, G.M. Cook, M.J. Howard, M.L. Rowe, R.K. Poole, and M. Shepherd. (2016). CydDC-mediated reductant export in Escherichia coli controls the transcriptional wiring of energy metabolism and combats nitrosative stress. Biochem. J. 473: 693-701.

Honorat, M., R. Terreux, P. Falson, A. Di Pietro, C. Dumontet, and L. Payen. (2013). Localization of putative binding sites for cyclic guanosine monophosphate and the anti-cancer drug 5-fluoro-2'-deoxyuridine-5'-monophosphate on ABCC11 in silico models. BMC Struct Biol 13: 7.

Horazdovsky, B.F. and R.W. Hogg. (1989). Genetic reconstitution of the high-affinity L-arabinose transport system. J. Bacteriol. 171: 3053-3059.

Horlacher, R., K.B. Xavier, H. Santos, J. DiRuggiero, M. Kossmann, and W. Boos. (1998). Archaeal binding protein-dependent ABC transporter: molecular and biochemical analysis of the trehalose/maltose transport system of the hyperthermophilic archaeon Thermococcus litoralis. J. Bacteriol. 180: 680-689.

Hoshino, T. and K. Kose. (1990). Cloning, nucleotide sequences, and identification of products of the Pseudomonas aeruginosa PAO bra genes, which encode the high-affinity branched-chain amino acid transport system. J. Bacteriol. 172: 5531-5539.

Hosie, A.H., D. Allaway, C.S. Galloway, H.A. Dunsby, and P.S. Poole. (2002). Rhizobium leguminosarum has a second general amino acid permease with unusually broad substrate specificity and high similarity to branched-chain amino acid transporters (Bra/LIV) of the ABC family. J. Bacteriol. 184: 4071-4080.

Hou, Y.X., J.R. Riordan, and X.B. Chang. (2003). ATP binding, not hydrolysis, at the first nucleotide-binding domain of multidrug resistance-associated protein MRP1 enhances ADP.Vi trapping at the second domain. J. Biol. Chem. 278: 3599-3605.

Howlett, R.M., B.M. Hughes, A. Hitchcock, and D.J. Kelly. (2012). Hydrogenase activity in the foodborne pathogen Campylobacter jejuni depends upon a novel ABC-type nickel transporter (NikZYXWV) and is SlyD-independent. Microbiology 158: 1645-1655.

Hsu, W.L., T. Furuta, and M. Sakurai. (2017). The mechanism of nucleotide-binding domain dimerization in the intact maltose transporter as studied by all-atom molecular dynamics simulations. Proteins. [Epub: Ahead of Print]

Hu D., Barajas-Martinez H., Terzic A., Park S., Pfeiffer R., Burashnikov E., Wu Y., Borggrefe M., Veltmann C., Schimpf R., Cai JJ., Nam GB., Deshmukh P., Scheinman M., Preminger M., Steinberg J., Lopez-Izquierdo A., Ponce-Balbuena D., Wolpert C., Haissaguerre M., Sanchez-Chapula JA. and Antzelevitch C. (2014). ABCC9 is a novel Brugada and early repolarization syndrome susceptibility gene. Int J Cardiol. 171(3):431-42.

Huang, C.F., N. Yamaji, N. Mitani, M. Yano, Y. Nagamura, and J.F. Ma. (2009). A bacterial-type ABC transporter is involved in aluminum tolerance in rice. Plant Cell 21: 655-667.

Huang, J. and G.F. Ecker. (2023). A Structure-Based View on ABC-Transporter Linked to Multidrug Resistance. Molecules 28:.

Huang, X., A. Yan, X. Zhang, and Y. Xu. (2006). Identification and characterization of a putative ABC transporter PltHIJKN required for pyoluteorin production in Pseudomonas sp. M18. Gene 376: 68-78.

Huang, X., X. Zhang, N. An, M. Zhang, M. Ma, Y. Yang, L. Jing, Y. Wang, Z. Chen, and P. Zhang. (2023). Cryo-EM structure and molecular mechanism of abscisic acid transporter ABCG25. Nat Plants. [Epub: Ahead of Print]

Huang, Y., C. Xue, R. Bu, C. Wu, J. Li, J. Zhang, J. Chen, Z. Shi, Y. Chen, Y. Wang, and Z. Liu. (2024). Inhibition and transport mechanisms of the ABC transporter hMRP5. Nat Commun 15: 4811.

Huang, Y., P. Anderle, K.J. Bussey, C. Barbacioru, U. Shankavaram, Z. Dai, W.C. Reinhold, A. Papp, J.N. Weinstein, and W. Sadée. (2004). Membrane transporters and channels: role of the transportome in cancer chemosensitivity and chemoresistance. Cancer Res 64: 4294-4301.

Hudek, L., L.A. Pearson, A. Michalczyk, B.A. Neilan, and M.L. Ackland. (2013). Molecular and cellular characterisation of the zinc uptake (Znu) system of Nostoc punctiforme. FEMS Microbiol Ecol 86: 149-171.

Hugouvieux-Cotte-Pattat, N. and S. Reverchon. (2001). Two transporters, TogT and TogMNAB, are responsible for oligogalacturonide uptake in Erwinia chrysanthemi 3937. Mol. Microbiol. 41: 1125-1132.

Hugouvieux-Cotte-Pattat, N., N. Blot, and S. Reverchon. (2001). Identification of TogMNAB, an ABC transporter which mediates the uptake of pectic oligomers in Erwinia chrysanthemi 3937. Mol. Microbiol. 41: 1113-1123.

Hullo, M.F., S. Auger, E. Dassa, A. Danchin, and I. Martin-Verstraete. (2004). The metNPQ operon of Bacillus subtilis encodes an ABC permease transporting methionine sulfoxide, D- and L-methionine. Res. Microbiol. 155: 80-86.

Hunnicutt, D.W., M.J. Kempf, and M.J. McBride. (2002). Mutations in Flavobacterium johnsoniae gldF and gldG disrupt gliding motility and interfere with membrane localization of GldA. J. Bacteriol. 184: 2370-2378.

Hürlimann, L.M., V. Corradi, M. Hohl, G.V. Bloemberg, D.P. Tieleman, and M.A. Seeger. (2016). The heterodimeric ABC transporter EfrCD mediates multidrug efflux in Enterococcus faecalis. Antimicrob. Agents Chemother. [Epub: Ahead of Print]

Hvorup, R.N., B.A. Goetz, M. Niederer, K. Hollenstein, E. Perozo, and K.P. Locher. (2007). Asymmetry in the structure of the ABC transporter-binding protein complex BtuCD-BtuF. Science 317: 1387-1390.

Hyink, O., P.A. Wescombe, M. Upton, N. Ragland, J.P. Burton, and J.R. Tagg. (2007). Salivaricin A2 and the novel lantibiotic salivaricin B are encoded at adjacent loci on a 190-kilobase transmissible megaplasmid in the oral probiotic strain Streptococcus salivarius K12. Appl. Environ. Microbiol. 73: 1107-1113.

Ibanez-Ruiz, M., V. Robbe-Saule, D. Hermant, S. Labrude, and F. Norel. (2000). Identification of RpoS (σS)-regulated genes in Salmonella enterica serovar typhimurium. J. Bacteriol. 182: 5749-5756.

Ibba, R., S. Sestito, F.A. Ambrosio, E. Marchese, G. Costa, F.P. Fiorentino, F. Fusi, I. Marchesi, B. Polini, G. Chiellini, S. Alcaro, S. Piras, and A. Carta. (2024). Discovery of pyridoquinoxaline-based new P-gp inhibitors as coadjutant against Multi Drug Resistance in cancer. Eur J Med Chem 276: 116647.

Ibrahim, M.A.A., K.A.A. Abdeljawaad, A.H.M. Abdelrahman, P.A. Sidhom, A.M. Tawfeek, G.A.H. Mekhemer, M.K. Abd El-Rahman, E. Dabbish, and T. Shoeib. (2023). In-Silico Mining of the Toxins Database (T3DB) towards Hunting Prospective Candidates as ABCB1 Inhibitors: Integrated Molecular Docking and Lipid Bilayer-Enhanced Molecular Dynamics Study. Pharmaceuticals (Basel) 16:.

Igarashi, K. and K. Kashiwagi. (1996). Polyamine transport inEscherichia coli. Amino Acids 10: 83-97.

Igarashi, Y., K.F. Aoki, H. Mamitsuka, K. Kuma, and M. Kanehisa. (2004). The evolutionary repertoires of the eukaryotic-type ABC transporters in terms of the phylogeny of ATP-binding domains in eukaryotes and prokaryotes. Mol Biol Evol 21: 2149-2160.

Iliás, A., Z. Urbán, T.L. Seidl, O. Le Saux, E. Sinkó, C.D. Boyd, B. Sarkadi, and A. Váradi. (2002). Loss of ATP-dependent transport activity in pseudoxanthoma elasticum-associated mutants of human ABCC6 (MRP6). J. Biol. Chem. 277: 16860-16867.

Immadisetty, K., J. Hettige, and M. Moradi. (2019). Lipid-Dependent Alternating Access Mechanism of a Bacterial Multidrug ABC Exporter. ACS Cent Sci 5: 43-56.

Inoue, Y., T. Yamaguchi, T. Otsuka, Y. Utsunomiya, D. Pan, H. Ogawa, and H. Kato. (2022). Structure-based alteration of tryptophan residues of the multidrug transporter CmABCB1 to assess substrate binding using fluorescence spectroscopy. Protein. Sci. 31: e4331.

Iqbal, J., M.T. Walsh, and M.M. Hussain. (2022). ATP-Binding Cassette Transporter Family C Protein 10 Participates in the Synthesis and Efflux of Hexosylceramides in Liver Cells. Nutrients 14:.

Irobalieva, R.N., I. Manolaridis, S.M. Jackson, D. Ni, E. Pardon, H. Stahlberg, J. Steyaert, and K.P. Locher. (2023). Structural Basis of the Allosteric Inhibition of Human ABCG2 by Nanobodies. J. Mol. Biol. 435: 168234.

Ishii, S., T. Yano, and H. Hayashi. (2006). Expression and characterization of the peptidase domain of Streptococcus pneumoniae ComA, a bifunctional ATP-binding cassette transporter involved in quorum sensing pathway. J. Biol. Chem. 281: 4726-4731.

Isom, G.L., N. Coudray, M.R. MacRae, C.T. McManus, D.C. Ekiert, and G. Bhabha. (2020). LetB Structure Reveals a Tunnel for Lipid Transport across the Bacterial Envelope. Cell 181: 653-664.e19.

Issitt, T., E. Bosseboeuf, N. De Winter, N. Dufton, G. Gestri, V. Senatore, A. Chikh, A.M. Randi, and C. Raimondi. (2018). Neuropilin-1 Controls Endothelial Homeostasis by Regulating Mitochondrial Function and Iron-Dependent Oxidative Stress. iScience 11: 205-223. [Epub: Ahead of Print]

Ito, H. and W.M. Gray. (2006). A gain-of-function mutation in the Arabidopsis pleiotropic drug resistance transporter PDR9 confers resistance to auxinic herbicides. Plant Physiol. 142: 63-74.

Ito, Y., K. Kanamaru, N. Taniguchi, S. Miyamoto, and H. Tokuda. (2006). A novel ligand bound ABC transporter, LolCDE, provides insights into the molecular mechanisms underlying membrane detachment of bacterial lipoproteins. Mol. Microbiol. 62: 1064-1075.

Iwaki, T., Y. Giga-Hama, and K. Takegawa. (2006). A survey of all 11 ABC transporters in fission yeast: two novel ABC transporters are required for red pigment accumulation in a Schizosaccharomyces pombe adenine biosynthetic mutant. Microbiology 152: 2309-2321.

Jackson, S.M., I. Manolaridis, J. Kowal, M. Zechner, N.M.I. Taylor, M. Bause, S. Bauer, R. Bartholomaeus, G. Bernhardt, B. Koenig, A. Buschauer, H. Stahlberg, K.H. Altmann, and K.P. Locher. (2018). Structural basis of small-molecule inhibition of human multidrug transporter ABCG2. Nat Struct Mol Biol 25: 333-340.

Jacobo-Albavera, L., M. Domínguez-Pérez, D.J. Medina-Leyte, A. González-Garrido, and T. Villarreal-Molina. (2021). The Role of the ATP-Binding Cassette A1 (ABCA1) in Human Disease. Int J Mol Sci 22:.

Jacquet, E., J.M. Girard, O. Ramaen, O. Pamlard, H. Lévaique, J.M. Betton, E. Dassa, and O. Chesneau. (2008). ATP hydrolysis and pristinamycin IIA inhibition of the Staphylococcus aureus Vga(A), a dual ABC protein involved in streptogramin A resistance. J. Biol. Chem. 283: 25332-25339.

Janas, E., M. Hofacker, M. Chen, S. Gompf, C. van der Does, and R. Tampé. (2003). The ATP hydrolysis cycle of the nucleotide-binding domain of the mitochondrial ATP-binding cassette transporter Mdl1p. J. Biol. Chem. 278: 26862-26869.

Janke, D., S. Mehralivand, D. Strand, U. Gödtel-Armbrust, A. Habermeier, U. Gradhand, C. Fischer, M.R. Toliat, P. Fritz, U.M. Zanger, M. Schwab, M.F. Fromm, P. Nürnberg, L. Wojnowski, E.I. Closs, and T. Lang. (2008). 6-mercaptopurine and 9-(2-phosphonyl-methoxyethyl) adenine (PMEA) transport altered by two missense mutations in the drug transporter gene ABCC4. Hum Mutat 29: 659-669.

Janvilisri, T., H. Venter, S. Shahi, G. Reuter, L. Balakrishnan, and H.W. van Veen. (2003). Sterol transport by the human breast cancer resistance protein (ABCG2) expressed in Lactococcus lactis. J. Biol. Chem. 278: 20645-20651.

Jedlitschky, G., B. Burchell, and D. Keppler. (2000). The multidrug resistance protein 5 functions as an ATP-dependent export pump for cyclic nucleotides. J. Biol. Chem. 275: 30069-30074.

Jensen, J.B., N.K. Peters, and T.V. Bhuvaneswari. (2002). Redundancy in periplasmic binding protein-dependent transport systems for trehalose, sucrose, and maltose in Sinorhizobium meliloti. J. Bacteriol. 184: 2978-2986.

Jha, R.M., T.A. Koleck, A.M. Puccio, D.O. Okonkwo, S.Y. Park, B.E. Zusman, R.S.B. Clark, L.A. Shutter, J.S. Wallisch, P.E. Empey, P.M. Kochanek, and Y.P. Conley. (2018). Regionally clustered polymorphisms in a prospective cohort predict cerebral oedema and outcome in severe traumatic brain injury. J Neurol Neurosurg Psychiatry. [Epub: Ahead of Print]

Ji, N., H. Li, Y. Zhang, Y. Li, P. Wang, X. Chen, Y.N. Liu, J.Q. Wang, Y. Yang, Z.S. Chen, Y. Li, R. Wang, and D. Kong. (2024). Lansoprazole (LPZ) reverses multidrug resistance (MDR) in cancer through impeding ATP-binding cassette (ABC) transporter-mediated chemotherapeutic drug efflux and lysosomal sequestration. Drug Resist Updat 76: 101100.

Jia, Y., Y. Zhang, W. Wang, J. Lei, Z. Ying, and G. Yang. (2022). Structural and functional insights of the human peroxisomal ABC transporter ALDP. Elife 11:.

Jiang, H., L. Shang, S.H. Yoon, S.Y. Lee, and Z. Yu. (2006). Optimal production of poly-γ-glutamic acid by metabolically engineered Escherichia coli. Biotechnol Lett 28: 1241-1246.

Jiang, P., Z. Dai, C. Yang, L. Ding, S. Li, X. Xu, C. Cheng, J. Wang, and S. Liu. (2024). CFTR Inhibitors Display Antiviral Activity against Herpes Simplex Virus. Viruses 16:.

Jiang, Y., W. Wang, Q. Xie, N. Liu, L. Liu, D. Wang, X. Zhang, C. Yang, X. Chen, D. Tang, and E. Wang. (2017). Plants transfer lipids to sustain colonization by mutualistic mycorrhizal and parasitic fungi. Science 356: 1172-1175.

Jin MS., Oldham ML., Zhang Q. and Chen J. (2012). Crystal structure of the multidrug transporter P-glycoprotein from Caenorhabditis elegans. Nature. 490(7421):566-9.

Jin, H., D. Liu, K. Zuo, Y. Gong, Z. Miao, Y. Chen, W. Ren, X. Sun, and K. Tang. (2007). Molecular cloning and characterization of Crmdr1, a novel MDR-type ABC transporter gene from Catharanthus roseus. DNA Seq 18: 316-325.

Johnsen, U., M. Ortjohann, J.M. Sutter, S. Geweke, and P. Schönheit. (2019). Uptake of D-xylose and L-arabinose in Haloferax volcanii involves an ABC transporter of the CUT1 subfamily. FEMS Microbiol. Lett. 366:.

Johnson, D.A., S.G. Tetu, K. Phillippy, J. Chen, Q. Ren, and I.T. Paulsen. (2008). High-throughput phenotypic characterization of Pseudomonas aeruginosa membrane transport genes. PLoS Genet 4: e1000211.

Jones PM. and George AM. (2013). Mechanism of the ABC transporter ATPase domains: catalytic models and the biochemical and biophysical record. Crit Rev Biochem Mol Biol. 48(1):39-50.

Jones PM. and George AM. (2014). A reciprocating twin-channel model for ABC transporters. Q Rev Biophys. 47(3):189-220.

Jones, P.M. and A.M. George. (2011). Molecular-dynamics simulations of the ATP/apo state of a multidrug ATP-binding cassette transporter provide a structural and mechanistic basis for the asymmetric occluded state. Biophys. J. 100: 3025-3034.

Jones, P.M. and A.M. George. (2012). Role of the D-loops in allosteric control of ATP hydrolysis in an ABC transporter. J Phys Chem A 116: 3004-3013.

Jones, P.M. and A.M. George. (2017). How Intrinsic Dynamics Mediates the Allosteric Mechanism in the ABC Transporter Nucleotide Binding Domain Dimer. J Chem Theory Comput 13: 1712-1722.

Jones, P.M. and A.M. George. (2023). The Switch and Reciprocating Models for the Function of ABC Multidrug Exporters: Perspectives on Recent Research. Int J Mol Sci 24:.

Jordan, I.K., K.C. Kota, G. Cui, C.H. Thompson, and N.A. McCarty. (2008). Evolutionary and functional divergence between the cystic fibrosis transmembrane conductance regulator and related ATP-binding cassette transporters. Proc. Natl. Acad. Sci. USA 105: 18865-18870.

Joseph, P., G. Fichant, Y. Quentin, and F. Denizot. (2002). Regulatory relationship of two-component and ABC transport systems and clustering of their genes in the Bacillus/Clostridium group, suggest a functional link between them. J. Mol. Microbiol. Biotechnol. 4: 503-513.

Joshi, S.M., A.K. Pandey, N. Capite, S.M. Fortune, E.J. Rubin, and C.M. Sassetti. (2006). Characterization of mycobacterial virulence genes through genetic interaction mapping. Proc. Natl. Acad. Sci. USA 103: 11760-11765.

Josts, I., Y. Gao, D.C.F. Monteiro, S. Niebling, J. Nitsche, K. Veith, T.W. Gräwert, C.E. Blanchet, M.A. Schroer, N. Huse, A.R. Pearson, D.I. Svergun, and H. Tidow. (2019). Structural Kinetics of MsbA Investigated by Stopped-Flow Time-Resolved Small-Angle X-Ray Scattering. Structure. [Epub: Ahead of Print]

Juan-Carlos, P.M., P.P. Perla-Lidia, M.M. Stephanie-Talia, A.M. Mónica-Griselda, and T.E. Luz-María. (2021). ABC transporter superfamily. An updated overview, relevance in cancer multidrug resistance and perspectives with personalized medicine. Mol Biol Rep 48: 1883-1901.

Kadaba, N.S., J.T. Kaiser, E. Johnson, A. Lee, and D.C. Rees. (2008). The high-affinity E. coli methionine ABC transporter: structure and allosteric regulation. Science 321: 250-253.

Kadioglu, O., M.E.M. Saeed, M. Munder, A. Spuller, H.J. Greten, and T. Efferth. (2020). Effect of ABC transporter expression and mutational status on survival rates of cancer patients. Biomed Pharmacother 131: 110718. [Epub: Ahead of Print]

Kagawa, T., N. Watanabe, K. Mochizuki, A. Numari, Y. Ikeno, J. Itoh, H. Tanaka, I.M. Arias, and T. Mine. (2008). Phenotypic differences in PFIC2 and BRIC2 correlate with protein stability of mutant Bsep and impaired taurocholate secretion in MDCK II cells. Am. J. Physiol. Gastrointest Liver Physiol 294: G58-67.

Kahnert, A., P. Vermeij, C. Wietek, P. James, T. Leisinger, and M.A. Kertesz. (2000). The ssu locus plays a key role in organosulfur metabolism in Pseudomonas putida S-313. J. Bacteriol. 182: 2869-2878.

Kala, S.V., M.W. Neely, G. Kala, C.I. Prater, D.W. Atwood, J.S. Rice, and M.W. Lieberman. (2000). The MRP2/cMOAT transporter and arsenic-glutathione complex formation are required for biliary excretion of arsenic. J. Biol. Chem. 275: 33404-33408.

Kalashnikova, T.V., L.V. Sutormina, Z.Y. Samoilova, O.N. Oktyabrsky, and G.V. Smirnova. (2023). Effect of Changes in the Redox Status on Biofilm Formation in Escherichia coli. Bull Exp Biol Med 174: 451-454.

Kallabi, F., I. Hadj Salem, G. Ben Salah, H. Ben Turkia, A. Ben Chehida, N. Tebib, F. Fakhfakh, and H. Kamoun. (2013). Molecular characterization of X-linked adrenoleukodystrophy in a Tunisian family: identification of a novel missense mutation in the ABCD1 gene. Neurodegener Dis 12: 207-211.

Kallenberg F., Dintner S., Schmitz R. and Gebhard S. (2013). Identification of regions important for resistance and signalling within the antimicrobial peptide transporter BceAB of Bacillus subtilis. J Bacteriol. 195(14):3287-97.

Kalscheuer, R., B. Weinrick, U. Veeraraghavan, G.S. Besra, and W.R. Jacobs, Jr. (2010). Trehalose-recycling ABC transporter LpqY-SugA-SugB-SugC is essential for virulence of Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA 107: 21761-21766.

Kaltak, M., R. Blanco-Garavito, L.L. Molday, C.M. Dhaenens, E.E. Souied, G. Platenburg, J. Swildens, R.S. Molday, and F.P.M. Cremers. (2023). Stargardt disease-associated in-frame ABCA4 exon 17 skipping results in significant ABCA4 function. J Transl Med 21: 546.

Kamakura, A., Y. Fujimoto, Y. Motohashi, K. Ohashi, A. Ohashi-Kobayashi, and M. Maeda. (2008). Functional dissection of transmembrane domains of human TAP-like (ABCB9). Biochem. Biophys. Res. Commun. 377: 847-851.

Kanamaru, K., N. Taniguchi, S. Miyamoto, S. Narita, and H. Tokuda. (2007). Complete reconstitution of an ATP-binding cassette transporter LolCDE complex from separately isolated subunits. FEBS J. 274: 3034-3043.

Kang, X.L., M. Zhang, K. Wang, X.F. Qiao, and M.H. Chen. (2016). MOLECULAR CLONING, EXPRESSION PATTERN OF MULTIDRUG RESISTANCE ASSOCIATED PROTEIN 1 (MRP1, ABCC1) GENE, AND THE SYNERGISTIC EFFECTS OF VERAPAMIL ON TOXICITY OF TWO INSECTICIDES IN THE BIRD CHERRY-OAT APHID. Arch Insect Biochem Physiol 92: 65-84.

Kapoor, K., S. Pant, and E. Tajkhorshid. (2021). Active participation of membrane lipids in inhibition of multidrug transporter P-glycoprotein. Chem Sci 12: 6293-6306.

Kapoor, P., A.J. Horsey, M.H. Cox, and I.D. Kerr. (2018). ABCG2: does resolving its structure elucidate the mechanism? Biochem Soc Trans. [Epub: Ahead of Print]

Kapoor, P., D.A. Briggs, M.H. Cox, and I.D. Kerr. (2020). Disruption of the Unique ABCG-Family NBD:NBD Interface Impacts Both Drug Transport and ATP Hydrolysis. Int J Mol Sci 21:.

Kappes, R.M. and E. Bremer. (1998). Response of Bacillus subtilis to high osmolarity: uptake of carnitine, crotonobetaine and ?-butyrobetaine via the ABC transport system OpuC. Microbiology 144: 83-90.

Kaproń, B., R. Czarnomysy, D. Radomska, K. Bielawski, and T. Plech. (2023). Thiosemicarbazide Derivatives Targeting Human TopoIIα and IDO-1 as Small-Molecule Drug Candidates for Breast Cancer Treatment. Int J Mol Sci 24:.

Karasawa, A., L.J. Swier, M.C. Stuart, J. Brouwers, B. Helms, and B. Poolman. (2013). Physicochemical factors controlling the activity and energy coupling of an ionic strength-gated ATP-binding cassette (ABC) transporter. J. Biol. Chem. 288: 29862-29871.

Karlikowska, M., A. Singh, A. Bhatt, S. Ott, A.R. Bottrill, G.S. Besra, and E. Fullam. (2021). Biochemical and phenotypic characterisation of the transporter UspABC. Cell Surf 7: 100052.

Karlinsey, J.E., M.E. Maguire, L.A. Becker, M.L. Crouch, and F.C. Fang. (2010). The phage shock protein PspA facilitates divalent metal transport and is required for virulence of Salmonella enterica sv. Typhimurium. Mol. Microbiol. 78: 669-685.

Karpouzas, G.A., B. Papotti, S.R. Ormseth, M. Palumbo, E. Hernandez, M.P. Adorni, F. Zimetti, M.J. Budoff, and N. Ronda. (2023). ATP-binding cassette G1 membrane transporter-mediated cholesterol efflux capacity influences coronary atherosclerosis and cardiovascular risk in Rheumatoid Arthritis. J Autoimmun 136: 103029.

Karpouzas, G.A., B. Papotti, S.R. Ormseth, M. Palumbo, E. Hernandez, M.P. Adorni, F. Zimetti, M.J. Budoff, and N. Ronda. (2023). Inflammation and immunomodulatory therapies influence the relationship between ATP-binding cassette A1 membrane transporter-mediated cholesterol efflux capacity and coronary atherosclerosis in rheumatoid arthritis. J Transl Autoimmun 7: 100209.

Karpowich, N.K. and D.N. Wang. (2013). Assembly and mechanism of a group II ECF transporter. Proc. Natl. Acad. Sci. USA 110: 2534-2539.

Karpowich, N.K., J. Song, and D.N. Wang. (2016). An aromatic cap seals the substrate binding site in an ECF-type S subunit for riboflavin. J. Mol. Biol. [Epub: Ahead of Print]

Karpowich, N.K., J.M. Song, N. Cocco, and D.N. Wang. (2015). ATP binding drives substrate capture in an ECF transporter by a release-and-catch mechanism. Nat Struct Mol Biol 22: 565-571.

Kashiwagi, K., M.H. Tsuhako, K. Sakata, T. Saisho, A. Igarashi, S.O. da Costa, and K. Igarashi. (1998). Relationship between spontaneous aminoglycoside resistance in Escherichia coli and a decrease in oligopeptide binding protein. J. Bacteriol. 180: 5484-5488.

Kashiwayama, Y., M. Seki, A. Yasui, Y. Murasaki, M. Morita, Y. Yamashita, M. Sakaguchi, Y. Tanaka, and T. Imanaka. (2009). 70-kDa peroxisomal membrane protein related protein (P70R/ABCD4) localizes to endoplasmic reticulum not peroxisomes, and NH2-terminal hydrophobic property determines the subcellular localization of ABC subfamily D proteins. Exp Cell Res 315: 190-205.

Katzir, H., D. Yeheskely-Hayon, R. Regev, and G.D. Eytan. (2010). Role of the plasma membrane leaflets in drug uptake and multidrug resistance. FEBS J. 277: 1234-1244.

Kaur, H., A. Lakatos-Karoly, R. Vogel, A. Nöll, R. Tampé, and C. Glaubitz. (2016). Coupled ATPase-adenylate kinase activity in ABC transporters. Nat Commun 7: 13864.

Kawai, E., H. Akatsuka, A. Idei, T. Shibatani, and K. Omori. (1998). Serratia marcescens S-layer protein is secreted extracellularly via an ATP-binding cassette exporter, the Lip system. Mol. Microbiol. 27: 941-952.

Kawai, H., T. Tanji, H. Shiraishi, M. Yamada, R. Iijima, T. Inoue, Y. Kezuka, K. Ohashi, Y. Yoshida, K. Tohyama, K. Gengyo-Ando, S. Mitani, H. Arai, A. Ohashi-Kobayashi, and M. Maeda. (2009). Normal formation of a subset of intestinal granules in Caenorhabditis elegans requires ATP-binding cassette transporters HAF-4 and HAF-9, which are highly homologous to human lysosomal peptide transporter TAP-like. Mol. Biol. Cell 20: 2979-2990.

Kawanobe, T., S. Kogure, S. Nakamura, M. Sato, K. Katayama, J. Mitsuhashi, K. Noguchi, and Y. Sugimoto. (2012). Expression of human ABCB5 confers resistance to taxanes and anthracyclines. Biochem. Biophys. Res. Commun. 418: 736-741.

Kazanov MD., Li X., Gelfand MS., Osterman AL. and Rodionov DA. (2013). Functional diversification of ROK-family transcriptional regulators of sugar catabolism in the Thermotogae phylum. Nucleic Acids Res. 41(2):790-803.

Kehres, D.G., A. Janakiraman, J.M. Slauch, and M.E. Maguire. (2002). SitABCD is the alkaline Mn2+ transporter of Salmonella enterica serovar Typhimurium. J. Bacteriol. 184: 3159-3166.

Kell, D.B. (2021). The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 26:.

Kelly, J., H. Jarrell, L. Millar, L. Tessier, L.M. Fiori, P.C. Lau, B. Allan, and C.M. Szymanski. (2006). Biosynthesis of the N-linked glycan in Campylobacter jejuni and addition onto protein through block transfer. J. Bacteriol. 188: 2427-2434.

Kelly, S.D., D.M. Williams, S. Zhu, T. Kim, M. Jana, J. Nothof, V.N. Thota, T.L. Lowary, and C. Whitfield. (2024). Klebsiella pneumoniae O-polysaccharide biosynthesis highlights the diverse organization of catalytic modules in ABC transporter-dependent glycan assembly. J. Biol. Chem. 300: 107420. [Epub: Ahead of Print]

Kempf, B. and E. Bremer. (1998). Uptake and synthesis of compatible solutes as microbial stress responses to high-osmolality environments. Arch. Microbiol. 170: 319-330.

Keppler, D. (1999). Export pumps for glutathione S-conjugates. Free Radic Biol Med 27: 985-991.

Keppler, D. and J. Konig. (1997). Hepatic canalicular membrane 5: Expression and localization of the conjugate export pump encoded by the MRP2 (cMRP/cMOAT) gene in liver. FASEB J. 11: 509-516.

Khan, S.A., M. Reichelt, and D.G. Heckel. (2017). Functional analysis of the ABCs of eye color in Helicoverpa armigera with CRISPR/Cas9-induced mutations. Sci Rep 7: 40025.

Khare, D., H. Choi, S.U. Huh, B. Bassin, J. Kim, E. Martinoia, K.H. Sohn, K.H. Paek, and Y. Lee. (2017). Arabidopsis ABCG34 contributes to defense against necrotrophic pathogens by mediating the secretion of camalexin. Proc. Natl. Acad. Sci. USA 114: E5712-E5720.

Khare, D., M.L. Oldham, C. Orelle, A.L. Davidson, and J. Chen. (2009). Alternating access in maltose transporter mediated by rigid-body rotations. Mol. Cell 33: 528-536.

Khassanova, G., S. Jatayev, A. Gabdola, M. Kuzbakova, A. Zailasheva, G. Kylyshbayeva, C. Schramm, K. Schleyer, L. Philp-Dutton, C. Sweetman, P. Anderson, C.L.D. Jenkins, K.L. Soole, and Y. Shavrukov. (2024). Haplotypes of ATP-Binding Cassette in Chickpea from Kazakhstan Are Associated with Salinity Tolerance and Leaf Necrosis via Oxidative Stress. Biomolecules 14:.

Khot, M.I., C.L. Downey, G. Armstrong, H.S. Svavarsdottir, F. Jarral, H. Andrew, and D.G. Jayne. (2019). The role of ABCG2 in modulating responses to anti-cancer photodynamic therapy. Photodiagnosis Photodyn Ther. [Epub: Ahead of Print]

Khunweeraphong, N. and K. Kuchler. (2020). The first intracellular loop is essential for the catalytic cycle of the human ABCG2 multidrug resistance transporter. FEBS Lett. [Epub: Ahead of Print]

Khwaja, M., Q. Ma, and M.H. Saier, Jr. (2005). Topological analysis of integral membrane constituents of prokaryotic ABC efflux systems. Res. Microbiol. 156: 270-277.

Kieuvongngam, V. and J. Chen. (2022). Structures of the peptidase-containing ABC transporter PCAT1 under equilibrium and nonequilibrium conditions. Proc. Natl. Acad. Sci. USA 119:.

Kieuvongngam, V., P.D.B. Olinares, A. Palillo, M.L. Oldham, B.T. Chait, and J. Chen. (2020). Structural basis of substrate recognition by a polypeptide processing and secretion transporter. Elife 9:.

Kikuchi, H., M. Aichi, I. Suzuki, and T. Omato. (1996). Positive regulation by nitrite of the nitrate assimilation operon in the cyanobacteria Synechococcus sp. strain PCC 7942 and Plectonema boryanum. J. Bacteriol. 178: 5822-5825.

Kim, C., S. Song, and C. Park. (1997). The D-allose operon of Escherichia coli K-12. J. Bacteriol. 179: 7631-7637.

Kim, D.Y., L. Bovet, M. Maeshima, E. Martinoia, and Y. Lee. (2007). The ABC transporter AtPDR8 is a cadmium extrusion pump conferring heavy metal resistance. Plant J. 50: 207-218.

Kim, D.Y., L. Bovet, S. Kushnir, E.W. Noh, E. Martinoia, and Y. Lee. (2006). AtATM3 is involved in heavy metal resistance in Arabidopsis. Plant Physiol. 140: 922-932.

Kim, H. and S.K. Farrand. (1997). Characterization of the acc operon from the nopaline-type Ti plasmid pTiC58, which encodes utilization of agrocinopines A and B and susceptibility to agrocin 84. J. Bacteriol. 179: 7559-7572.

Kim, J., S. Wu, T.M. Tomasiak, C. Mergel, M.B. Winter, S.B. Stiller, Y. Robles-Colmanares, R.M. Stroud, R. Tampé, C.S. Craik, and Y. Cheng. (2015). Subnanometre-resolution electron cryomicroscopy structure of a heterodimeric ABC exporter. Nature 517: 396-400.

Kim, S., A.K. Sharma, and O.K. Vatamaniuk. (2018). N-Terminal Extension and C-Terminal Domains Are Required for ABCB6/HMT-1 Protein Interactions, Function in Cadmium Detoxification, and Localization to the Endosomal-Recycling System in. Front Physiol 9: 885.

Kim, S., D.S. Selote, and O.K. Vatamaniuk. (2010). The N-terminal extension domain of the C. elegans half-molecule ABC transporter, HMT-1, is required for protein-protein interactions and function. PLoS One 5: e12938.

Kim, W. and W.B. Whitman. (1999). Isolation of acetate auxotrophs of the methane-producing archaeon Methanococcus maripaludis by random insertional mutagenesis. Genetics 152: 1429-1437.

Kim, Y. and J. Chen. (2018). Molecular structure of human P-glycoprotein in the ATP-bound, outward-facing conformation. Science 359: 915-919.

Kinch, L.N., S. Cheek, and N.V. Grishin. (2005). EDD, a novel phosphotransferase domain common to mannose transporter EIIA, dihydroxyacetone kinase, and DegV. Protein. Sci. 14: 360-367.

Kirsch F. and Eitinger T. (2014). Transport of nickel and cobalt ions into bacterial cells by S components of ECF transporters. Biometals. 27(4):653-60.

Kiss K., Kucsma N., Brozik A., Tusnady GE., Bergam P., van Niel G. and Szakacs G. (2015). Role of the N-terminal transmembrane domain in the endo-lysosomal targeting and function of the human ABCB6 protein. Biochem J. 467(1):127-39.

Kittichai, V., W. Nguitragool, H.G. Ngassa Mbenda, J. Sattabongkot, and L. Cui. (2018). Genetic diversity of the Plasmodium vivax multidrug resistance 1 gene in Thai parasite populations. Infect Genet Evol 64: 168-177.

Kjaerulff, S., S. Müller, and M.R. Jensen. (2005). Alternative protein secretion: the Mam1 ABC transporter supports secretion of M-factor linked GFP in fission yeast. Biochem. Biophys. Res. Commun. 338: 1853-1859.

Klein, I., B. Sarkadi, and A. Váradi. (1999). An inventory of the human ABC proteins. Biochim. Biophys. Acta. 1461: 237-262.

Kluth M., Stindt J., Droge C., Linnemann D., Kubitz R. and Schmitt L. (2015). A mutation within the extended X loop abolished substrate-induced ATPase activity of the human liver ATP-binding cassette (ABC) transporter MDR3. J Biol Chem. 290(8):4896-907.

Ko, T.P., S.T. Tseng, S.J. Lai, S.C. Chen, H.H. Guan, C.S. Yang, C.J. Chen, and Y. Chen. (2016). SH3-Like Motif-Containing C-terminal Domain of Staphylococcal Teichoic Acid Transporter Suggests Possible Function. Proteins. [Epub: Ahead of Print]

Kobae, Y., T. Sekino, H. Yoshioka, T. Nakagawa, E. Martinoia, and M. Maeshima. (2006). Loss of AtPDR8, a plasma membrane ABC transporter of Arabidopsis thaliana, causes hypersensitive cell death upon pathogen infection. Plant Cell Physiol. 47: 309-318.

Kobayashi N., Kobayashi N., Yamaguchi A. and Nishi T. (2009). Characterization of the ATP-dependent sphingosine 1-phosphate transporter in rat erythrocytes. J Biol Chem. 284(32):21192-200.

Kobayashi, N., K. Nishino, and A. Yamaguchi. (2001). Novel macrolide-specific ABC-type efflux transporter in Escherichia coli. J. Bacteriol. 183: 5639-5644.

Koch, D.J., C. Rückert, D.A. Rey, A. Mix, A. Pühler, and J. Kalinowski. (2005). Role of the ssu and seu genes of Corynebacterium glutamicum ATCC 13032 in utilization of sulfonates and sulfonate esters as sulfur sources. Appl. Environ. Microbiol. 71: 6104-6114.

Koch, J., R. Guntrum, and R. Tampé. (2006). The first N-terminal transmembrane helix of each subunit of the antigenic peptide transporter TAP is essential for independent tapasin binding. FEBS Lett. 580: 4091-4096.

Kochan, T.J. and S. Dawid. (2013). The HtrA Protease of Streptococcus pneumoniae Controls Density-Dependent Stimulation of the Bacteriocin blp Locus via Disruption of Pheromone Secretion. J. Bacteriol. 195: 1561-1572.

Kodan, A., T. Yamaguchi, T. Nakatsu, K. Matsuoka, Y. Kimura, K. Ueda, and H. Kato. (2019). Inward- and outward-facing X-ray crystal structures of homodimeric P-glycoprotein CmABCB1. Nat Commun 10: 88.

Kodan, A., T. Yamaguchi, T. Nakatsu, K. Sakiyama, C.J. Hipolito, A. Fujioka, R. Hirokane, K. Ikeguchi, B. Watanabe, J. Hiratake, Y. Kimura, H. Suga, K. Ueda, and H. Kato. (2014). Structural basis for gating mechanisms of a eukaryotic P-glycoprotein homolog. Proc. Natl. Acad. Sci. USA 111: 4049-4054.

Kogan, I., M. Ramjeesingh, C. Li, J.F. Kidd, Y. Wang, E.M. Leslie, S.P. Cole, and C.E. Bear. (2003). CFTR directly mediates nucleotide-regulated glutathione flux. EMBO. J. 22: 1981-1989.

Koh, E.I., C.S. Hung, and J.P. Henderson. (2016). The Yersiniabactin-Associated ATP Binding Cassette Proteins YbtP and YbtQ Enhance Escherichia coli Fitness during High-Titer Cystitis. Infect. Immun. 84: 1312-1319.

Koh-Tan, H.H., E. Strachan, K. Cooper, L. Bell-Sakyi, and N.N. Jonsson. (2016). Identification of a novel β-adrenergic octopamine receptor-like gene (βAOR-like) and increased ATP-binding cassette B10 (ABCB10) expression in a Rhipicephalus microplus cell line derived from acaricide-resistant ticks. Parasit Vectors 9: 425.

Köhler, S.C., S. Vahdati, M.S. Scholz, and M. Wiese. (2018). Structure activity relationships, multidrug resistance reversal and selectivity of heteroarylphenyl ABCG2 inhibitors. Eur J Med Chem 146: 483-500.

Koide, A. and J.A. Hoch. (1994). Identification of a second oligopeptide transport system in Bacillus subtilis and determination of its role in sporulation. Mol. Microbiol. 13: 417-426.

Kolenbrander, P.E., R.N. Andersen, R.A. Baker, and H.F. Jenkinson. (1998). The adhesion-associated sca operon in Streptococcus gordonii encodes an inducible high-affinity ABC transporter for Mn2+ uptake. J. Bacteriol. 180: 290-295.

Kopecka, J., P. Trouillas, A.&.#.2.6.8.;. Gašparović, E. Gazzano, Y.G. Assaraf, and C. Riganti. (2019). Phospholipids and cholesterol: Inducers of cancer multidrug resistance and therapeutic targets. Drug Resist Updat 49: 100670. [Epub: Ahead of Print]

Korhonen, J.T., V.M. Olkkonen, R. Lahesmaa, and M. Puolakkainen. (2013). ABC-cassette transporter 1 (ABCA1) expression in epithelial cells in Chlamydia pneumoniae infection. Microb. Pathog. 61-62: 57-61.

Korkhov, V.M., S.A. Mireku, R.N. Hvorup, and K.P. Locher. (2012). Asymmetric states of vitamin B₁₂ transporter BtuCD are not discriminated by its cognate substrate binding protein BtuF. FEBS Lett. 586: 972-976.

Kotlyarov, S. and A. Kotlyarova. (2021). Bioinformatic Analysis of Gene Expression in Smoking and Chronic Obstructive Pulmonary Disease. Membranes (Basel) 11:.

Kowal, L., J. Huang, H. Luo, J. Singh, A.E. Snook, J. Uitto, and Q. Li. (2021). Functional Assessment of Missense Variants in the ABCC6 Gene Implicated in Pseudoxanthoma Elasticum, a Heritable Ectopic Mineralization Disorder. J Invest Dermatol. [Epub: Ahead of Print]

Koyanagi, T., T. Katayama, H. Suzuki, and H. Kumagai. (2004). Identification of the LIV-I/LS system as the third phenylalanine transporter in Escherichia coli K-12. J. Bacteriol. 186: 343-350.

Krause, A., J. Dingemanse, A. Mathis, L. Marquart, J.J. Möhrle, and J.S. McCarthy. (2016). Pharmacokinetic/pharmacodynamic modelling of the antimalarial effect of Actelion-451840 in an induced blood stage malaria study in healthy subjects. Br J Clin Pharmacol 82: 412-421.

Krehenbrink, M. and J.A. Downie. (2008). Identification of protein secretion systems and novel secreted proteins in Rhizobium leguminosarum bv. viciae. BMC Genomics 9: 55.

Krejcík, Z., K. Denger, S. Weinitschke, K. Hollemeyer, V. Paces, A.M. Cook, and T.H. Smits. (2008). Sulfoacetate released during the assimilation of taurine-nitrogen by Neptuniibacter caesariensis: purification of sulfoacetaldehyde dehydrogenase. Arch. Microbiol. 190: 159-168.

Kretzschmar, T., W. Kohlen, J. Sasse, L. Borghi, M. Schlegel, J.B. Bachelier, D. Reinhardt, R. Bours, H.J. Bouwmeester, and E. Martinoia. (2012). A petunia ABC protein controls strigolactone-dependent symbiotic signalling and branching. Nature 483: 341-344.

Kreutzer, D., H. Döring, P. Werner, C.A. Ritter, and A. Hilgeroth. (2021). Novel Symmetrical Cage Compounds as Inhibitors of the Symmetrical MRP4-Efflux Pump for Anticancer Therapy. Int J Mol Sci 22:.

Krishnamurthy, P.C., G. Du, Y. Fukuda, D. Sun, J. Sampath, K.E. Mercer, J. Wang, B. Sosa-Pineda, K.G. Murti, and J.D. Schuetz. (2006). Identification of a mammalian mitochondrial porphyrin transporter. Nature 443: 586-589.

Krumpochova, P., S. Sapthu, J.F. Brouwers, M. de Haas, R. de Vos, P. Borst, and K. van de Wetering. (2012). Transportomics: screening for substrates of ABC transporters in body fluids using vesicular transport assays. FASEB J. 26: 738-747.

Kuai, Z., Y. Xu, Q. Zhao, J. Liu, S. Guan, Y. Qiao, X. Gong, J. Nie, P. Li, D. Liu, Y. Xing, H. Li, Z. Sun, W. Wang, C. Ning, Y. Shi, W. Kong, and Y. Shan. (2018). Effects of insulin on transcriptional response and permeability in an in vitro model of human blood-brain barrier. J. Cell. Biochem. [Epub: Ahead of Print]

Kuan, G., E. Dassa, W. Saurin, M. Hofnung, and M.H. Saier, Jr. (1995). Phylogenetic analyses of the ATP-binding constituents of bacterial extracytoplasmic receptor-dependent ABC-type nutrient uptake permeases. Res. Microbiol. 146: 271-278.

Kubo, Y., S. Sekiya, M. Ohigashi, C. Takenaka, K. Tamura, S. Nada, T. Nishi, A. Yamamoto, and A. Yamaguchi. (2005). ABCA5 resides in lysosomes, and ABCA5 knockout mice develop lysosomal disease-like symptoms. Mol. Cell Biol. 25: 4138-4149.

Kumar, J.S., B.R. Wei, J.P. Madigan, R.M. Simpson, M.D. Hall, and M.M. Gottesman. (2016). Bioluminescent imaging of ABCG2 efflux activity at the blood-placenta barrier. Sci Rep 6: 20418.

Kumari, S., M. Kumar, N.K. Khandelwal, P. Kumari, M. Varma, P. Vishwakarma, G. Shahi, S. Sharma, A.M. Lynn, R. Prasad, and N.A. Gaur. (2018). ABC transportome inventory of human pathogenic yeast Candida glabrata: Phylogenetic and expression analysis. PLoS One 13: e0202993.

Kunzelmann, K., J. Ousingsawat, A. Kraus, J.H. Park, T. Marquardt, R. Schreiber, and B. Buchholz. (2023). Pathogenic Relationships in Cystic Fibrosis and Renal Diseases: CFTR, SLC26A9 and Anoctamins. Int J Mol Sci 24:.

Kuznetsova, A., G. Masrati, E. Vigonsky, N. Livnat-Levanon, J. Rose, M. Grupper, A. Baloum, J.G. Yang, D.C. Rees, N. Ben-Tal, and O. Lewinson. (2021). Titratable transmembrane residues and a hydrophobic plug are essential for manganese import via the Bacillus anthracis ABC transporter MntBC-A. J. Biol. Chem. 101087. [Epub: Ahead of Print]

Kwan, B.W., D.M. Lord, W. Peti, R. Page, M.J. Benedik, and T.K. Wood. (2015). The MqsR/MqsA toxin/antitoxin system protects Escherichia coli during bile acid stress. Environ Microbiol 17: 3168-3181.

Kym, P.R., X. Wang, M. Pizzonero, and S.E. Van der Plas. (2018). Recent Progress in the Discovery and Development of Small-Molecule Modulators of CFTR. Prog Med Chem 57: 235-276.

Légaré, D., D. Richard, R. Mukhopadhyay, Y.D. Stierhof, B.P. Rosen, A. Haimeur, B. Papadopoulou, and M. Ouellette. (2001). The Leishmania ATP-binding cassette protein PGPA is an intracellular metal-thiol transporter ATPase. J. Biol. Chem. 276: 26301-26307.

Lages, M.A., L. Ageitos, J. Rodríguez, C. Jiménez, M.L. Lemos, and M. Balado. (2022). Identification of Key Functions Required for Production and Utilization of the Siderophore Piscibactin Encoded by the High-Pathogenicity Island -HPI in. Int J Mol Sci 23:.

Lagos, R., J.E. Villanueva, and O. Monasterio. (1999). Identification and properties of the genes encoding microcin E492 and its immunity protein. J. Bacteriol. 181: 212-217.

Lagos, R., M. Baeza, G. Corsini, C. Hetz, E. Strahsburger, J.A. Castillo, C. Vergara, and O. Monasterio. (2001). Structure, organization and characterization of the gene cluster involved in the production of microcin E492, a channel-forming bacteriocin. Mol. Microbiol. 42: 229-243.

Laiolo, J., D.G. Graikioti, C.L. Barbieri, M.B. Joray, A.I. Antoniou, D.M.A. Vera, C.M. Athanassopoulos, and M.C. Carpinella. (2024). Novel betulin derivatives as multidrug reversal agents targeting P-glycoprotein. Sci Rep 14: 70.

Laiolo, J., P.A. Lanza, O. Parravicini, C. Barbieri, D. Insuasty, J. Cobo, D.M.A. Vera, R.D. Enriz, and M.C. Carpinella. (2021). Structure activity relationships and the binding mode of quinolinone-pyrimidine hybrids as reversal agents of multidrug resistance mediated by P-gp. Sci Rep 11: 16856.

Lakli, M., J. Dumont, V. Vauthier, J. Charton, V. Crespi, M. Banet, Y. Riahi, A. Ben Saad, E. Mareux, M. Lapalus, E. Gonzales, E. Jacquemin, F. Di Meo, B. Deprez, F. Leroux, and T. Falguières. (2024). Identification of new correctors for traffic-defective ABCB4 variants by a high-content screening approach. Commun Biol 7: 898.

Lambert, A., M. Østerås, K. Mandon, M.C. Poggi, and D. Le Rudulier. (2001). Fructose uptake in Sinorhizobium meliloti is mediated by a high-affinity ATP-binding cassette transport system. J. Bacteriol. 183: 4709-4717.

Lamping, E., P.V. Baret, A.R. Holmes, B.C. Monk, A. Goffeau, and R.D. Cannon. (2010). Fungal PDR transporters: Phylogeny, topology, motifs and function. Fungal Genet Biol 47: 127-142.

Lan, Y.L., X. Wang, J.C. Lou, J.S. Xing, S. Zou, Z.L. Yu, X.C. Ma, H. Wang, and B. Zhang. (2018). Marinobufagenin inhibits glioma growth through sodium pump α1 subunit and ERK signaling-mediated mitochondrial apoptotic pathway. Cancer Med 7: 2034-2047.

Lange, H., G. Kispal, and R. Lill. (1999). Mechanism of iron transport to the site of heme synthesis inside yeast mitochondria. J. Biol. Chem. 274: 18989-18996.

Langeland, A., H. Jetter, and D.M. O''Halloran. (2021). The diversity of ABC transporter genes across the Phylum Nematoda. Parasitol Int 83: 102357. [Epub: Ahead of Print]

Lansky, S., R. Salama, S. Shulami, N. Lavid, S. Sen, I. Schapiro, Y. Shoham, and G. Shoham. (2020). Carbohydrate-Binding Capability and Functional Conformational Changes of AbnE, an Arabino-oligosaccharide Binding Protein. J. Mol. Biol. 432: 2099-2120.

Larrazabal, C., L.M.R. Silva, L. Pervizaj-Oruqaj, S. Herold, C. Hermosilla, and A. Taubert. (2021). P-Glycoprotein Inhibitors Differently Affect , and Proliferation in Bovine Primary Endothelial Cells. Pathogens 10:.

Larsen, P.B., J. Cancel, M. Rounds, and V. Ochoa. (2007). Arabidopsis ALS1 encodes a root tip and stele localized half type ABC transporter required for root growth in an aluminum toxic environment. Planta 225: 1447-1458.

Larsen, P.B., M.J. Geisler, C.A. Jones, K.M. Williams, and J.D. Cancel. (2005). ALS3 encodes a phloem-localized ABC transporter-like protein that is required for aluminum tolerance in Arabidopsis. Plant J. 41: 353-363.

Larue, K., R.C. Ford, L.M. Willis, and C. Whitfield. (2011). Functional and structural characterization of polysaccharide co-polymerase proteins required for polymer export in ATP-binding cassette transporter-dependent capsule biosynthesis pathways. J. Biol. Chem. 286: 16658-16668.

László, L., B. Sarkadi, and T. Hegedűs. (2016). Jump into a New Fold-A Homology Based Model for the ABCG2/BCRP Multidrug Transporter. PLoS One 11: e0164426.

Latunde-Dada, G.O., R.J. Simpson, and A.T. McKie. (2006). Recent advances in mammalian haem transport. Trends. Biochem. Sci. 31: 182-188.

Lavilla Lerma, L., N. Benomar, A.S. Valenzuela, M.d.e.l.C. Casado Muñoz, A. Gálvez, and H. Abriouel. (2014). Role of EfrAB efflux pump in biocide tolerance and antibiotic resistance of Enterococcus faecalis and Enterococcus faecium isolated from traditional fermented foods and the effect of EDTA as EfrAB inhibitor. Food Microbiol 44: 249-257.

Lazard, M., N.T. Ha-Duong, S. Mounié, R. Perrin, P. Plateau, and S. Blanquet. (2011). Selenodiglutathione uptake by the Saccharomyces cerevisiae vacuolar ATP-binding cassette transporter Ycf1p. FEBS J. 278: 4112-4121.

Le Hir, R., C. Sorin, D. Chakraborti, T. Moritz, H. Schaller, F. Tellier, S. Robert, H. Morin, L. Bako, and C. Bellini. (2013). ABCG9, ABCG11 and ABCG14 ABC transporters are required for vascular development in Arabidopsis. Plant J. 76: 811-824.

Le Roy, F., C. Bisbal, M. Silhol, C. Martinand, B. Lebleu, and T. Salehzada. (2001). The 2-5A/RNase L/RNase L inhibitor (RLI) [correction of (RNI)] pathway regulates mitochondrial mRNAs stability in interferon α-treated H9 cells. J. Biol. Chem. 276: 48473-48482.

Le, L.T.M., J.R. Thompson, P.X. Dang, J. Bhandari, and A. Alam. (2022). Structures of the human peroxisomal fatty acid transporter ABCD1 in a lipid environment. Commun Biol 5: 7.

Le, L.T.M., J.R. Thompson, S. Dehghani-Ghahnaviyeh, S. Pant, P.X. Dang, J.B. French, T. Kanikeyo, E. Tajkhorshid, and A. Alam. (2023). Cryo-EM structures of human ABCA7 provide insights into its phospholipid translocation mechanisms. EMBO. J. 42: e111065.

Lecher J., Schwarz CK., Stoldt M., Smits SH., Willbold D. and Schmitt L. (2012). An RTX transporter tethers its unfolded substrate during secretion via a unique N-terminal domain. Structure. 20(10):1778-87.

Lecher, J., M. Stoldt, C.K. Schwarz, S.H. Smits, L. Schmitt, and D. Willbold. (2011). 1H, 15N and 13C resonance assignment of the N-terminal C39 peptidase-like domain of the ABC transporter Haemolysin B (HlyB). Biomol NMR Assign 5: 199-201.

Lee, E.M., S.H. Ahn, J.H. Park, J.H. Lee, S.C. Ahn, and I.S. Kong. (2004). Identification of oligopeptide permease (opp) gene cluster in Vibrio fluvialis and characterization of biofilm production by oppA knockout mutation. FEMS Microbiol. Lett. 240: 21-30.

Lee, E.W., M.N. Huda, T. Kuroda, T. Mizushima, and T. Tsuchiya. (2003). EfrAB, an ABC multidrug efflux pump in Enterococcus faecalis. Antimicrob. Agents Chemother. 47: 3733-3738.

Lee, J.Y., J.G. Yang, D. Zhitnitsky, O. Lewinson, and D.C. Rees. (2014). Structural basis for heavy metal detoxification by an Atm1-type ABC exporter. Science 343: 1133-1136.

Lee, J.Y., L.N. Kinch, D.M. Borek, J. Wang, J. Wang, I.L. Urbatsch, X.S. Xie, N.V. Grishin, J.C. Cohen, Z. Otwinowski, H.H. Hobbs, and D.M. Rosenbaum. (2016). Crystal structure of the human sterol transporter ABCG5/ABCG8. Nature 533: 561-564.

Lee, M., K. Lee, J. Lee, E.W. Noh, and Y. Lee. (2005). AtPDR12 contributes to lead resistance in Arabidopsis. Plant Physiol. 138: 827-836.

Lee, S.J., A. Böhm, M. Krug, and W. Boos. (2007). The ABC of binding-protein-dependent transport in Archaea. Trends Microbiol. 15: 389-397.

Lee, S.K., S. Mo, and J.W. Suh. (2012). An ABC transporter complex containing S-adenosylmethionine (SAM)-induced ATP-binding protein is involved in antibiotics production and SAM signaling in Streptomyces coelicolor M145. Biotechnol Lett 34: 1907-1914.

Lee, S.W., D.A. Mitchell, A.L. Markley, M.E. Hensler, D. Gonzalez, A. Wohlrab, P.C. Dorrestein, V. Nizet, and J.E. Dixon. (2008). Discovery of a widely distributed toxin biosynthetic gene cluster. Proc. Natl. Acad. Sci. USA 105: 5879-5884.

Lefèvre, F. and M. Boutry. (2018). Towards Identification of the Substrates of ATP-Binding Cassette Transporters. Plant Physiol. 178: 18-39.

Lehman, M.K., N.A. Sturd, F. Razvi, D.L. Wellems, S.D. Carson, and P.D. Fey. (2023). Proline transporters ProT and PutP are required for Staphylococcus aureus infection. PLoS Pathog 19: e1011098.

Leisico, F., L.M. Godinho, I.C. Gonçalves, S.P. Silva, B. Carneiro, M.J. Romão, T. Santos-Silva, and I. de Sá-Nogueira. (2020). Multitask ATPases (NBDs) of bacterial ABC importers type I and their interspecies exchangeability. Sci Rep 10: 19564.

Lentz, F., N. Reiling, G. Spengler, A. Kincses, A. Csonka, J. Molnár, and A. Hilgeroth. (2019). Dually Acting Nonclassical 1,4-Dihydropyridines Promote the Anti-Tuberculosis (Tb) Activities of Clofazimine. Molecules 24:.

Lentzas, A., M.C. de Gooijer, S. Zuidema, A. Meurs, C.H. Çitirikkaya, N. Venekamp, J.H. Beijnen, and O. van Tellingen. (2024). ATP-binding cassette transporter inhibitor potency and substrate drug affinity are critical determinants of successful drug delivery enhancement to the brain. Fluids Barriers CNS 21: 62.

Leon-Kempis, M.d.e.l.R., E. Guccione, F. Mulholland, M.P. Williamson, and D.J. Kelly. (2006). The Campylobacter jejuni PEB1a adhesin is an aspartate/glutamate-binding protein of an ABC transporter essential for microaerobic growth on dicarboxylic amino acids. Mol. Microbiol. 60: 1262-1275.

Leonhardt, R.M., P. Abrahimi, S.M. Mitchell, and P. Cresswell. (2014). Three Tapasin Docking Sites in TAP Cooperate To Facilitate Transporter Stabilization and Heterodimerization. J Immunol 192: 2480-2494.

Leskelä, S., E. Wahlström, H.L. Hyyryläinen, M. Jacobs, A. Palva, M. Sarvas, and V.P. Kontinen. (1999). Ecs, an ABC transporter of Bacillus subtilis: dual signal transduction functions affecting expression of secreted proteins as well as their secretion. Mol. Microbiol. 31: 533-543.

Leskelä, S., V.P. Kontinen, and M. Sarvas. (1996). Molecular analysis of an operon in Bacillus subtilis encoding a novel ABC transporter with a role in exoprotein production, sporulation and competence. Microbiology 142(Pt1): 71-77.

Leslie, E.M., A. Haimeur, and M.P. Waalkes. (2004). Arsenic transport by the human multidrug resistance protein 1 (MRP1/ABCC1). Evidence that a tri-glutathione conjugate is required. J. Biol. Chem. 279: 32700-32708.

Létoffé, S., P. Delepelaire, and C. Wandersman. (2008). Functional differences between heme permeases: Serratia marcescens HemTUV permease exhibits a narrower substrate specificity (restricted to heme) than the Escherichia coli DppABCDF peptide-heme permease. J. Bacteriol. 190: 1866-1870.

Levring, J. and J. Chen. (2024). Structural identification of a selectivity filter in CFTR. Proc. Natl. Acad. Sci. USA 121: e2316673121.

Li, C., P.C. Krishnamurthy, H. Penmatsa, K.L. Marrs, X.Q. Wang, M. Zaccolo, K. Jalink, M. Li, D.J. Nelson, J.D. Schuetz, and A.P. Naren. (2007). Spatiotemporal coupling of cAMP transporter to CFTR chloride channel function in the gut epithelia. Cell 131: 940-951.

Li, H., M. Wang, K. Qu, R. Xu, and H. Zhu. (2023). MP Allosterically Activates AMPK to Enhance ABCA1 Stability by Retarding the Calpain-Mediated Degradation Pathway. Int J Mol Sci 24:.

Li, H., N. Li, Q. Xu, C. Xiao, H. Wang, Z. Guo, J. Zhang, X. Sun, and Q.Y. He. (2013). Lipoprotein FtsB in Streptococcus pyogenes binds ferrichrome in two steps with residues Tyr137 and Trp204 as critical ligands. PLoS One 8: e65682.

Li, J., C. Wang, G. Yang, Z. Sun, H. Guo, K. Shao, Y. Gu, W. Jiang, and P. Zhang. (2017). Molecular mechanism of environmental d-xylose perception by a XylFII-LytS complex in bacteria. Proc. Natl. Acad. Sci. USA 114: 8235-8240.

Li, J., M. Yue, D. Zhou, M. Wang, and H. Zhang. (2017). Abcb1a but not Abcg2 played a predominant role in limiting the brain distribution of Huperzine A in mice. Food Chem Toxicol 107: 68-73.

Li, J., W. Zheng, M. Gu, L. Han, Y. Luo, K. Yu, M. Sun, Y. Zong, X. Ma, B. Liu, E.P. Lowder, D.L. Mendez, R.G. Kranz, K. Zhang, and J. Zhu. (2022). Structures of the CcmABCD heme release complex at multiple states. Nat Commun 13: 6422.

Li, L., Y. Xiong, Y. Wang, S. Wu, C. Xiao, S. Wang, S. Cheng, and H. Cheng. (2023). Effect of Nano-Selenium on Nutritional Quality of Cowpea and Response of ABCC Transporter Family. Molecules 28:.

Li, S., K.M. Paulsson, S. Chen, H.O. Sjögren, and P. Wang. (2000). Tapasin is required for efficient peptide binding to transporter associated with antigen processing. J. Biol. Chem. 275: 1581-1586.

Li, S., Y. Ren, X. Lu, Y. Shen, and X. Yang. (2021). Cryo-EM structure of human ABCB8 transporter in nucleotide binding state. Biochem. Biophys. Res. Commun. 557: 187-191.

Li, S.X., Y.J. Song, L. Jiang, Y.J. Zhao, H. Guo, D.M. Li, K.J. Zhu, and H. Zhang. (2017). Synergistic Effects of Tetrandrine with Posaconazole Against Aspergillus fumigatus. Microb Drug Resist 23: 674-681.

Li, Y. and W.A. Prinz. (2004). ATP-binding cassette (ABC) transporters mediate nonvesicular, raft-modulated sterol movement from the plasma membrane to the endoplasmic reticulum. J. Biol. Chem. 279: 45226-45234.

Licht, A., M. Bommer, T. Werther, K. Neumann, C. Hobe, and E. Schneider. (2018). Structural and functional characterization of a maltose/maltodextrin ABC transporter comprising a single solute binding domain (MalE) fused to the transmembrane subunit MalF. Res. Microbiol. [Epub: Ahead of Print]

Light, S.H., L. Su, R. Rivera-Lugo, J.A. Cornejo, A. Louie, A.T. Iavarone, C.M. Ajo-Franklin, and D.A. Portnoy. (2018). A flavin-based extracellular electron transfer mechanism in diverse Gram-positive bacteria. Nature 562: 140-144.

Lill, R., B. Hoffmann, S. Molik, A.J. Pierik, N. Rietzschel, O. Stehling, M.A. Uzarska, H. Webert, C. Wilbrecht, and U. Mühlenhoff. (2012). The role of mitochondria in cellular iron-sulfur protein biogenesis and iron metabolism. Biochim. Biophys. Acta. 1823: 1491-1508.

Lim, K.H., C.E. Jones, R.N. vanden Hoven, J.L. Edwards, M.L. Falsetta, M.A. Apicella, M.P. Jennings, and A.G. McEwan. (2008). Metal binding specificity of the MntABC permease of Neisseria gonorrhoeae and its influence on bacterial growth and interaction with cervical epithelial cells. Infect. Immun. 76: 3569-3576.

Lin, D.Y., S. Huang, and J. Chen. (2015). Crystal structures of a polypeptide processing and secretion transporter. Nature 523: 425-430.

Lin, H.T., V.N. Bavro, N.P. Barrera, H.M. Frankish, S. Velamakanni, H.W. van Veen, C.V. Robinson, M.I. Borges-Walmsley, and A.R. Walmsley. (2009). MacB ABC transporter is a dimer whose ATPase activity and macrolide-binding capacity are regulated by the membrane fusion protein MacA. J. Biol. Chem. 284: 1145-1154.

Linka, N. and C. Esser. (2012). Transport proteins regulate the flux of metabolites and cofactors across the membrane of plant peroxisomes. Front Plant Sci 3: 3.

Linton, K.J. and C.F. Higgins. (1998). The Escherichia coli ATP-binding cassette (ABC) proteins. Mol. Microbiol. 28: 5-13.

Lisher, J.P., K.A. Higgins, M.J. Maroney, and D.P. Giedroc. (2013). Physical characterization of the manganese-sensing pneumococcal surface antigen repressor from Streptococcus pneumoniae. Biochemistry 52: 7689-7701.

Liu, F., Y. Yang, Y. Zheng, Y.H. Liang, and K. Zeng. (2018). Mutation and expression of ABCA12 in keratosis pilaris and nevus comedonicus. Mol Med Rep 18: 3153-3158.

Liu, F., Z. Zhang, A. Levit, J. Levring, K.K. Touhara, B.K. Shoichet, and J. Chen. (2019). Structural identification of a hotspot on CFTR for potentiation. Science 364: 1184-1188.

Liu, G., R. Sánchez-Fernández, Z.S. Li, and P.A. Rea. (2001). Enhanced multispecificity of arabidopsis vacuolar multidrug resistance-associated protein-type ATP-binding cassette transporter, AtMRP2. J. Biol. Chem. 276: 8648-8656.

Liu, G., W. Liu, R. Zhao, J. He, Z. Dong, L. Chen, W. Wan, Z. Chang, W. Wang, and X. Li. (2021). Genome-wide identification and gene-editing of pigment transporter genes in the swallowtail butterfly Papilio xuthus. BMC Genomics 22: 120.

Liu, H., R.N. Irobalieva, J. Kowal, D. Ni, K. Nosol, R. Bang-Sørensen, L. Lancien, H. Stahlberg, B. Stieger, and K.P. Locher. (2023). Structural basis of bile salt extrusion and small-molecule inhibition in human BSEP. Nat Commun 14: 7296.

Liu, J., L. Zhao, L. Shi, Y. Yuan, D. Fu, Z. Ye, Q. Li, Y. Deng, X. Liu, Q. Lv, Y. Cheng, Y. Xu, X. Jiang, G. Wang, L. Wang, and Z. Wang. (2020). A Sequentially Responsive Nanosystem Breaches Cascaded Bio-barriers and Suppresses P-Glycoprotein Function for Reversing Cancer Drug Resistance. ACS Appl Mater Interfaces 12: 54343-54355.

Liu, M., W. Zhang, W. Zhang, X. Zhou, M. Li, and J. Miao. (2017). Prenylflavonoid Isoxanthohumol Sensitizes MCF-7/ADR Cells to Doxorubicin Cytotoxicity via Acting as a Substrate of ABCB1. Toxins (Basel) 9:.

Liu, M., W.N. Tanaka, H. Zhu, G. Xie, D.M. Dooley, and B. Lei. (2008). Direct hemin transfer from IsdA to IsdC in the iron-regulated surface determinant (Isd) heme acquisition system of Staphylococcus aureus. J. Biol. Chem. 283: 6668-6676.

Liu, M., X. Lu, J. Zhang, X. Zhao, W. Zhang, and X. Lin. (2019). Teratogenic jervine increases the activity of doxorubicin in MCF-7/ADR cells by inhibiting ABCB1. Biomed Pharmacother 117: 109059.

Liu, N., G. Yang, M. Hu, Y. Cai, Z. Hu, C. Jia, and M. Zhang. (2018). Association of ABCC2  polymorphism and gender with high-density lipoprotein cholesterol response to simvastatin. Pharmacogenomics 19: 1125-1132.

Liu, N., H. Sun, Z.B. Liu, C. Shao, and L.L. Zhang. (2021). [Progress of studies on ATP-binding cassette transporters and transportation of secondary metabolites in medicinal plants]. Zhongguo Zhong Yao Za Zhi 46: 52-56.

Liu, Y., W. Li, Y. Wei, Y. Jiang, and X. Tan. (2013). Efficient preparation and metal specificity of the regulatory protein TroR from the human pathogen Treponema pallidum. Metallomics 5: 1448-1457.

Locher, K.P., A.T. Lee, and D.C. Rees. (2002). The E. coli BtuCD structure: a framework for ABC transporter architecture and mechanism. Science 296: 1091-1098.

Loo, T.W., M.C. Bartlett, and D.M. Clarke. (2003). Drug binding in human P-glycoprotein causes conformational changes in both nucleotide-binding domains. J. Biol. Chem. 278: 1575-1578.

López-Solanilla, E., F. García-Olmedo, and P. Rodríguez-Palenzuela. (1998). Inactivation of the sapA to sapF locus of Erwinia chrysanthemi reveals common features in plant and animal bacterial pathogenesis. Plant Cell 10: 917-924.

Lu S. and Zgurskaya HI. (2012). Role of ATP binding and hydrolysis in assembly of MacAB-TolC macrolide transporter. Mol Microbiol. 86(5):1132-43.

Lu, B., C. Xu, K. Awai, A.D. Jones, and C. Benning. (2007). A small ATPase protein of Arabidopsis, TGD3, involved in chloroplast lipid import. J. Biol. Chem. 282: 35945-35953.

Lu, J.F., E. Barron-Casella, R. Deering, A.K. Heinzer, A.B. Moser, K.L. deMesy Bentley, G.S. Wand, M. C McGuinness, Z. Pei, P.A. Watkins, A. Pujol, K.D. Smith, and J.M. Powers. (2007). The role of peroxisomal ABC transporters in the mouse adrenal gland: the loss of Abcd2 (ALDR), Not Abcd1 (ALD), causes oxidative damage. Lab Invest 87: 261-272.

Lu, Y.P., Z.S. Li, and P.A. Rea. (1997). AtMRP1 gene of Arabidopsis encodes a glutathione S-conjugate pump: isolation and functional definition of a plant ATP-binding cassette transporter gene. Proc. Natl. Acad. Sci. USA 94: 8243-8248.

Lubelski, J., A. de Jong, R. van Merkerk, H. Agustiandari, O.P. Kuipers, J. Kok, and A.J. Driessen. (2006). LmrCD is a major multidrug resistance transporter in Lactococcus lactis. Mol. Microbiol. 61: 771-781.

Lubelski, J., P. Mazurkiewicz, R. van Merkerk, W.N. Konings, and A.J. Driessen. (2004). ydaG and ydbA of Lactococcus lactis encode a heterodimeric ATP-binding cassette-type multidrug transporter. J. Biol. Chem. 279: 34449-34455.

Luckenbach, T. and D. Epel. (2008). ABCB- and ABCC-type transporters confer multixenobiotic resistance and form an environment-tissue barrier in bivalve gills. Am. J. Physiol. Regul Integr Comp Physiol 294: R1919-1929.

Luckenbach, T. and K. Burkhardt-Medicke. (2024). Differing temperature dependencies of functional homologs zebrafish Abcb4 and human ABCB1. Front Pharmacol 15: 1426040.

Luginbuehl, L.H., G.N. Menard, S. Kurup, H. Van Erp, G.V. Radhakrishnan, A. Breakspear, G.E.D. Oldroyd, and P.J. Eastmond. (2017). Fatty acids in arbuscular mycorrhizal fungi are synthesized by the host plant. Science 356: 1175-1178.

Lundstedt, E.A., B.W. Simpson, and N. Ruiz. (2020). Lipopolysaccharide transport involves long-range coupling between cytoplasmic and periplasmic domains of the LptBFGC extractor. J. Bacteriol. [Epub: Ahead of Print]

Luo, B., X.Y. Xue, W.L. Hu, L.J. Wang, and X.Y. Chen. (2007). An ABC transporter gene of Arabidopsis thaliana, AtWBC11, is involved in cuticle development and prevention of organ fusion. Plant Cell Physiol. 48: 1790-1802.

Luo, Q., H. Shi, and X. Xu. (2021). Cryo-EM structures of LptBFG and LptBFGC from Klebsiella pneumoniae in complex with lipopolysaccharide. Biochem. Biophys. Res. Commun. 571: 20-25.

Luo, Q., X. Yang, S. Yu, H. Shi, K. Wang, L. Xiao, G. Zhu, C. Sun, T. Li, D. Li, X. Zhang, M. Zhou, and Y. Huang. (2017). Structural basis for lipopolysaccharide extraction by ABC transporter LptB2FG. Nat Struct Mol Biol 24: 469-474.

Lüttmann, D., Y. Göpel, and B. Görke. (2012). The phosphotransferase protein EIIA(Ntr) modulates the phosphate starvation response through interaction with histidine kinase PhoR in Escherichia coli. Mol. Microbiol. 86: 96-110.

Lycklama A Nijeholt, J.A., R. Vietrov, G.K. Schuurman-Wolters, and B. Poolman. (2018). Energy Coupling Efficiency in the Type I ABC Transporter GlnPQ. J. Mol. Biol. [Epub: Ahead of Print]

Lyssenko, N.N. and D. Praticò. (2020). ABCA7 and the altered lipidostasis hypothesis of Alzheimer''s disease. Alzheimers Dement. [Epub: Ahead of Print]

Lyu, C., L. Wang, B. Stadlbauer, A. Buchner, and H. Pohla. (2022). A Pan-Cancer Landscape of ABCG2 across Human Cancers: Friend or Foe? Int J Mol Sci 23:.

Méndez, C. and J.A. Salas. (1998). ABC transporters in antibiotic-producing actinomycetes. FEMS Microbiol. Lett. 158: 1-8.

Mühlenhoff, U. and R. Lill. (2000). Biogenesis of iron-sulfur proteins in eukaryotes: a novel task of mitochondria that is inherited from bacteria. Biochim. Biophys. Acta. 1459: 370-382.

Macalou, S., R.W. Robey, G. Jabor Gozzi, S. Shukla, I. Grosjean, T. Hegedus, S.V. Ambudkar, S.E. Bates, and A. Di Pietro. (2015). The linker region of breast cancer resistance protein ABCG2 is critical for coupling of ATP-dependent drug transport. Cell Mol Life Sci. [Epub: Ahead of Print]

Mächtel, R., A. Narducci, D.A. Griffith, T. Cordes, and C. Orelle. (2019). An integrated transport mechanism of the maltose ABC importer. Res. Microbiol. [Epub: Ahead of Print]

Mackenzie, S.M., A.J. Howells, G.B. Cox, and G.D. Ewart. (2000). Sub-cellular localisation of the white/scarlet ABC transporter to pigment granule membranes within the compound eye of Drosophila melanogaster. Genetica 108: 239-252.

Maclean, A.M., W. Haerty, G.B. Golding, and T.M. Finan. (2011). The LysR-type PcaQ protein regulates expression of a protocatechuate-inducible ABC-type transport system in Sinorhizobium meliloti. Microbiology 157: 2522-2533.

MacRae, M.R., D. Puvanendran, M.A.B. Haase, N. Coudray, L. Kolich, C. Lam, M. Baek, G. Bhabha, and D.C. Ekiert. (2023). Protein-protein interactions in the Mla lipid transport system probed by computational structure prediction and deep mutational scanning. J. Biol. Chem. 104744. [Epub: Ahead of Print]

Madani, G., E. Lamping, and R.D. Cannon. (2021). Engineering a Cysteine-Deficient Functional Candida albicans Cdr1 Molecule Reveals a Conserved Region at the Cytosolic Apex of ABCG Transporters Important for Correct Folding and Trafficking of Cdr1. mSphere 6:.

Maeda, S. and T. Omata. (1997). Substrate-binding lipoprotein of the cyanobacterium Synechococcus sp. strain PCC 7942 involved in the transport of nitrate and nitrite. J. Biol. Chem. 272: 3036-3041.

Maeda, S. and T. Omata. (2009). Nitrite transport activity of the ABC-type cyanate transporter of the cyanobacterium Synechococcus elongatus. J. Bacteriol. 191: 3265-3272.

Maggi, S., O. Massidda, G. Luzi, D. Fadda, L. Paolozzi, and P. Ghelardini. (2008). Division protein interaction web: identification of a phylogenetically conserved common interactome between Streptococcus pneumoniae and Escherichia coli. Microbiology 154: 3042-3052.

Mahé, Y., Y. Lemoine, and K. Kuchler. (1996). The ATP binding cassette transporters Pdr5 and Snq2 of Saccharomyces cerevisiae can mediate transport of steroids in vivo. J. Biol. Chem. 271: 25167-25172.

Mahmood, N.A., E. Biemans-Oldehinkel, J.S. Patzlaff, G.K. Schuurman-Wolters, and B. Poolman. (2006). Ion specificity and ionic strength dependence of the osmoregulatory ABC transporter OpuA. J. Biol. Chem. 281: 29830-29839.

Makdessi, K., J.R. Andreesen, and A. Pich. (2001). Tungstate Uptake by a highly specific ABC transporter in Eubacterium acidaminophilum. J. Biol. Chem. 276: 24557-24564.

Malinverni, J.C. and T.J. Silhavy. (2009). An ABC transport system that maintains lipid asymmetry in the gram-negative outer membrane. Proc. Natl. Acad. Sci. USA 106: 8009-8014.

Mall, M.S., S. Shah, S. Singh, N. Singh, N. Singh, S. Vaish, and D. Gupta. (2023). Genome-wide identification and characterization of ABC transporter superfamily in the legume Cajanus cajan. J Appl Genet. [Epub: Ahead of Print]

Mallack, E.J., K. Gao, M. Engelen, and S. Kemp. (2022). Structure and Function of the Variant Database: 20 Years, 940 Pathogenic Variants, and 3400 Cases of Adrenoleukodystrophy. Cells 11:.

Malmström, A., M. Łysiak, L. Åkesson, I. Jakobsen, M. Mudaisi, P. Milos, M. Hallbeck, V. Fomichov, H. Broholm, K. Grunnet, H.S. Poulsen, C. Bratthäll, M. Strandeus, A. Papagiannopoulou, M. Stenmark-Askmalm, H. Green, and P. Söderkvist. (2019). ABCB1 single-nucleotide variants and survival in patients with glioblastoma treated with radiotherapy concomitant with temozolomide. Pharmacogenomics J. [Epub: Ahead of Print]

Mandal, S.K., R. Adhikari, A. Sharma, M. Chandravanshi, P. Gogoi, and S.P. Kanaujia. (2019). Designating ligand specificities to metal uptake ABC transporters in Thermus thermophilus HB8. Metallomics. [Epub: Ahead of Print]

Mandal, S.K., S.G. Nayak, and S.P. Kanaujia. (2021). Identification and characterization of metal uptake ABC transporters in Mycobacterium tuberculosis unveil their ligand specificity. Int J Biol Macromol 185: 324-337. [Epub: Ahead of Print]

Manolaridis, I., S.M. Jackson, N.M.I. Taylor, J. Kowal, H. Stahlberg, and K.P. Locher. (2018). Cryo-EM structures of a human ABCG2 mutant trapped in ATP-bound and substrate-bound states. Nature 563: 426-430.

Mao, Q., R.G. Deeley, and S.P. Cole. (2000). Functional reconstitution of substrate transport by purified multidrug resistance protein MRP1 (ABCC1) in phospholipid vesicles. J. Biol. Chem. 275: 34166-34172.

Mao, X., Z. He, F. Zhou, Y. Huang, and G. Zhu. (2019). Prognostic significance and molecular mechanisms of adenosine triphosphate-binding cassette subfamily C members in gastric cancer. Medicine (Baltimore) 98: e18347.

Marassi, M., M.L. Morieri, V. Sanga, G. Ceolotto, A. Avogaro, and G.P. Fadini. (2024). The Elusive Nature of ABCC8-related Maturity-Onset Diabetes of the Young (ABCC8-MODY). A Review of the Literature and Case Discussion. Curr Diab Rep 24: 197-206.

Marcoux J., Wang SC., Politis A., Reading E., Ma J., Biggin PC., Zhou M., Tao H., Zhang Q., Chang G., Morgner N. and Robinson CV. (2013). Mass spectrometry reveals synergistic effects of nucleotides, lipids, and drugs binding to a multidrug resistance efflux pump. Proc Natl Acad Sci U S A. 110(24):9704-9.

Marek, M., S. Milles, G. Schreiber, D.L. Daleke, G. Dittmar, A. Herrmann, P. Müller, and T.G. Pomorski. (2011). The yeast plasma membrane ATP binding cassette (ABC) transporter Aus1: purification, characterization, and the effect of lipids on its activity. J. Biol. Chem. 286: 21835-21843.

Mares, L., F. Vilchis, B. Chávez, and L. Ramos. (2019). Expression and regulation of ABCG2/BCRP1 by sex steroids in the Harderian gland of the Syrian hamster (Mesocricetus auratus). C R Biol 342: 279-289.

Mareux, E., M. Lapalus, A. Ben Saad, R. Zelli, M. Lakli, Y. Riahi, M. Almes, M. Banet, I. Callebaut, J.L. Decout, T. Falguières, E. Jacquemin, and E. Gonzales. (2022). In Vitro Rescue of the Bile Acid Transport Function of ABCB11 Variants by CFTR Potentiators. Int J Mol Sci 23:.

Margolles, A., A.B. Flórez, J.A. Moreno, D. van Sinderen, and C.G. de los Reyes-Gavilán. (2006). Two membrane proteins from Bifidobacterium breve UCC2003 constitute an ABC-type multidrug transporter. Microbiology 152: 3497-3505.

Margoni, M., F. Soli, A. Sangalli, M. Bellizzi, E. Cecchini, and M. Buganza. (2017). A novel mutation in ABCD1 unveils different clinical phenotypes in a family with adrenoleukodystrophy. J Clin Neurosci 43: 175-177.

Martinand, C., C. Montavon, T. Salehzada, M. Silhol, B. Lebleu, and C. Bisbal. (1999). RNase L inhibitor is induced during human immunodeficiency virus type 1 infection and down regulates the 2-5A/RNase L pathway in human T cells. J. Virol. 73: 290-296.

Martínez-García, M., J. Campos-Salinas, M. Cabello-Donayre, E. Pineda-Molina, F.J. Gálvez, L.M. Orrego, M.P. Sánchez-Cañete, S. Malagarie-Cazenave, D.M. Koeller, and J.M. Pérez-Victoria. (2016). LmABCB3, an atypical mitochondrial ABC transporter essential for Leishmania major virulence, acts in heme and cytosolic iron/sulfur clusters biogenesis. Parasit Vectors 9: 7.

Martins, M.P., A.C. Franceschini, T.R. Jacob, A. Rossi, and N.M. Martinez-Rossi. (2016). Compensatory expression of multidrug-resistance genes encoding ABC transporters in dermatophytes. J. Med. Microbiol. 65: 605-610.

Martinussen, J., C. Sørensen, C.B. Jendresen, and M. Kilstrup. (2010). Two nucleoside transporters in Lactococcus lactis with different substrate specificities. Microbiology 156: 3148-3157.

Maruyama Y., Itoh T., Kaneko A., Nishitani Y., Mikami B., Hashimoto W. and Murata K. (2015). Structure of a Bacterial ABC Transporter Involved in the Import of an Acidic Polysaccharide Alginate. Structure. 23(9):1643-54.

Masi, M. and C. Wandersman. (2010). Multiple signals direct the assembly and function of a type 1 secretion system. J. Bacteriol. 192: 3861-3869.

Mason, K.M., M.E. Bruggeman, R.S. Munson, and L.O. Bakaletz. (2006). The non-typeable Haemophilus influenzae Sap transporter provides a mechanism of antimicrobial peptide resistance and SapD-dependent potassium acquisition. Mol. Microbiol. 62: 1357-1372.

Matano, L.M., H.G. Morris, A.R. Hesser, S.E.S. Martin, W. Lee, T.W. Owens, E. Laney, H. Nakaminami, D. Hooper, T.C. Meredith, and S. Walker. (2017). Antibiotic That Inhibits the ATPase Activity of an ATP-Binding Cassette Transporter by Binding to a Remote Extracellular Site. J. Am. Chem. Soc. 139: 10597-10600.

Mathur, H., P.M. O''Connor, P.D. Cotter, C. Hill, and R.P. Ross. (2014). Heterologous expression of thuricin CD immunity genes in Listeria monocytogenes. Antimicrob. Agents Chemother. 58: 3421-3428.

Matsuhashi, A., H. Tahara, Y. Ito, J. Uchiyama, S. Ogawa, and H. Ohta. (2015). Slr2019, lipid A transporter homolog, is essential for acidic tolerance in Synechocystis sp. PCC6803. Photosynth Res 125: 267-277.

Matsumoto-Nakano, M. and H.K. Kuramitsu. (2006). Role of bacteriocin immunity proteins in the antimicrobial sensitivity of Streptococcus mutans. J. Bacteriol. 188: 8095-8102.

Matsumura, Y., N. Ban, and N. Inagaki. (2008). Aberrant catalytic cycle and impaired lipid transport into intracellular vesicles in ABCA3 mutants associated with nonfatal pediatric interstitial lung disease. Am. J. Physiol. Lung Cell Mol Physiol 295: L698-707.

Matsumura, Y., N. Ban, K. Ueda, and N. Inagaki. (2006). Characterization and classification of ATP-binding cassette transporter ABCA3 mutants in fatal surfactant deficiency. J. Biol. Chem. 281: 34503-34514.

Matsuo, T., J. Chen, Y. Minato, W. Ogawa, T. Mizushima, T. Kuroda, and T. Tsuchiya. (2008). SmdAB, a heterodimeric ABC-Type multidrug efflux pump, in Serratia marcescens. J. Bacteriol. 190: 648-654.

Matsuoka, K., T. Nakatsu, and H. Kato. (2021). The crystal structure of the CmABCB1 G132V mutant, which favors the outward-facing state, reveals the mechanism of the pivotal joint between TM1 and TM3. Protein. Sci. [Epub: Ahead of Print]

Mattle, D., A. Zeltina, J.S. Woo, B.A. Goetz, and K.P. Locher. (2010). Two stacked heme molecules in the binding pocket of the periplasmic heme-binding protein HmuT from Yersinia pestis. J. Mol. Biol. 404: 220-231.

Matzneller, P., M. Kussmann, S. Eberl, A. Maier-Salamon, W. Jäger, M. Bauer, O. Langer, M. Zeitlinger, and W. Poeppl. (2018). Pharmacokinetics of the P-gp Inhibitor Tariquidar in Rats After Intravenous, Oral, and Intraperitoneal Administration. Eur J Drug Metab Pharmacokinet. [Epub: Ahead of Print]

Maupin-Furlow, J.A., J.K. Rosentel, J.H. Lee, U. Deppenmeier, R.P. Gunsalus, and K.T. Shanmugam. (1995). Genetic analysis of the modABCD (molybdate transport) operon of Escherichia coli. J. Bacteriol. 177: 4851-4856.

May, J.M., T.W. Owens, M.D. Mandler, B.W. Simpson, M.B. Lazarus, D.J. Sherman, R.M. Davis, S. Okuda, W. Massefski, N. Ruiz, and D. Kahne. (2017). The Antibiotic Novobiocin Binds and Activates the ATPase That Powers Lipopolysaccharide Transport. J. Am. Chem. Soc. 139: 17221-17224.

McAuliffe, O., R.P. Ross, and C. Hill. (2001). Lantibiotics: structure, biosynthesis and mode of action. FEMS Microbiol. Rev. 25: 285-308.

McBride, M.J. and Y. Zhu. (2013). Gliding motility and Por secretion system genes are widespread among members of the phylum bacteroidetes. J. Bacteriol. 195: 270-278.

McBride, S.M. and A.L. Sonenshein. (2011). Identification of a genetic locus responsible for antimicrobial peptide resistance in Clostridium difficile. Infect. Immun. 79: 167-176.

McDonald, C.M., E.K. Reid, J.F. Pohl, T.K. Yuzyuk, L.M. Padula, K. Vavrina, and K. Altman. (2024). Cystic fibrosis and fat malabsorption: Pathophysiology of the cystic fibrosis gastrointestinal tract and the impact of highly effective CFTR modulator therapy. Nutr Clin Pract 39Suppl1: S57-S77.

McDonald, E.F., K.E. Oliver, J.P. Schlebach, J. Meiler, and L. Plate. (2024). Benchmarking AlphaMissense pathogenicity predictions against cystic fibrosis variants. PLoS One 19: e0297560.

McFarlane, H.E., J.J. Shin, D.A. Bird, and A.L. Samuels. (2010). Arabidopsis ABCG transporters, which are required for export of diverse cuticular lipids, dimerize in different combinations. Plant Cell 22: 3066-3075.

McKee, A.G., E.F. McDonald, W.D. Penn, C.P. Kuntz, K. Noguera, L.M. Chamness, F.J. Roushar, J. Meiler, K.E. Oliver, L. Plate, and J.P. Schlebach. (2023). General trends in the effects of VX-661 and VX-445 on the plasma membrane expression of clinical CFTR variants. Cell Chem Biol. [Epub: Ahead of Print]

McQuerry, J.A., J. Chen, J.T. Chang, and A.H. Bild. (2021). Tepoxalin increases chemotherapy efficacy in drug-resistant breast cancer cells overexpressing the multidrug transporter gene ABCB1. Transl Oncol 14: 101181.

Medeot, A.C., A.C. Boaglio, G. Salas, P.M. Maidagan, G.S. Miszczuk, I.R. Barosso, E.J. Sánchez Pozzi, F.A. Crocenzi, and M.G. Roma. (2024). Tauroursodeoxycholate prevents estradiol 17β-d-glucuronide-induced cholestasis and endocytosis of canalicular transporters by switching off pro-cholestatic signaling pathways. Life Sci 352: 122839.

Medrano, M.S., Y. Ding, X.G. Wang, P. Lu, J. Coburn, and L.T. Hu. (2007). Regulators of expression of the oligopeptide permease A proteins of Borrelia burgdorferi. J. Bacteriol. 189: 2653-2659.

Melis, A. and H.C. Chen. (2005). Chloroplast sulfate transport in green algae--genes, proteins and effects. Photosynth Res 86: 299-307.

Menéndez, N., A.F. Braña, J.A. Salas, and C. Méndez. (2007). Involvement of a chromomycin ABC transporter system in secretion of a deacetylated precursor during chromomycin biosynthesis. Microbiology 153: 3061-3070.

Meneau, I., A.T. Coste, and D. Sanglard. (2016). Identification of Aspergillus fumigatus multidrug transporter genes and their potential involvement in antifungal resistance. Med Mycol 54: 616-627.

Mentewab, A. and C.N. Stewart, Jr. (2005). Overexpression of an Arabidopsis thaliana ABC transporter confers kanamycin resistance to transgenic plants. Nat Biotechnol 23: 1177-1180.

Metcalf, W.W. and R.S. Wolfe. (1998). Molecular genetic analysis of phosphite and hypophosphite oxidation by Pseudomonas stutzeri WM88. J. Bacteriol. 180: 5547-5558.

Meyer, D., S. Pajonk, C. Micali, R. O''Connell, and P. Schulze-Lefert. (2009). Extracellular transport and integration of plant secretory proteins into pathogen-induced cell wall compartments. Plant J. 57: 986-999.

Mi, W., Y. Li, S.H. Yoon, R.K. Ernst, T. Walz, and M. Liao. (2017). Structural basis of MsbA-mediated lipopolysaccharide transport. Nature 549: 233-237.

Michaelis, S. and J. Barrowman. (2012). Biogenesis of the Saccharomyces cerevisiae pheromone a-factor, from yeast mating to human disease. Microbiol. Mol. Biol. Rev. 76: 626-651.

Miethke, M., O. Klotz, U. Linne, J.J. May, C.L. Beckering, and M.A. Marahiel. (2006). Ferri-bacillibactin uptake and hydrolysis in Bacillus subtilis. Mol. Microbiol. 61: 1413-1427.

Miglionico, R., A. Gerbino, A. Ostuni, M.F. Armentano, M. Monné, M. Carmosino, and F. Bisaccia. (2016). New insights into the roles of the N-terminal region of the ABCC6 transporter. J. Bioenerg. Biomembr. [Epub: Ahead of Print]

Mikkat, S. and M. Hagemann. (2000). Molecular analysis of the ggtBCD gene cluster of Synechocystis sp. strain PCC6803 encoding subunits of an ABC transporter for osmoprotective compounds. Arch. Microbiol. 174: 273-282.

Mikolay, A. and D.H. Nies. (2009). The ABC-transporter AtmA is involved in nickel and cobalt resistance of Cupriavidus metallidurans strain CH34. Antonie Van Leeuwenhoek 96: 183-191.

Millan, C.R., M. Francis, N. Kumar Khandelwal, V.F. Thompson, T.M. Thaker, and T.M. Tomasiak. (2021). A conserved motif in intracellular loop 1 stabilizestheoutward-facing conformation of TmrAB. J. Mol. Biol. 166834. [Epub: Ahead of Print]

Miranda, E.R., E.A. Nam, A. Kuspa, and G. Shaulsky. (2015). The ABC transporter, AbcB3, mediates cAMP export in D. discoideum development. Dev Biol 397: 203-211.

Misawa, T., H. Hayashi, M. Makishima, Y. Sugiyama, and Y. Hashimoto. (2012). E297G mutated bile salt export pump (BSEP) function enhancers derived from GW4064: Structural development study and separation from farnesoid X receptor-agonistic activity. Bioorg Med Chem Lett 22: 3962-3966.

Mishima, Y., K. Momma, W. Hashimoto, B. Mikami, and K. Murata. (2001). Super-channel in bacteria: function and structure of the macromolecule import system mediated by a pit-dependent ABC transporter. FEMS Microbiol. Lett. 204: 215-221.

Mishra S., Verhalen B., Stein RA., Wen PC., Tajkhorshid E. and Mchaourab HS. (2014). Conformational dynamics of the nucleotide binding domains and the power stroke of a heterodimeric ABC transporter. Elife. 3:e02740.

Mitchell-White, J.I., T. Stockner, N. Holliday, S.J. Briddon, and I.D. Kerr. (2021). Analysis of Sequence Divergence in Mammalian ABCGs Predicts a Structural Network of Residues That Underlies Functional Divergence. Int J Mol Sci 22:.

Mitsuhashi, N., T. Miki, H. Senbongi, N. Yokoi, H. Yano, M. Miyazaki, N. Nakajima, T. Iwanaga, Y. Yokoyama, T. Shibata, and S. Seino. (2000). MTABC3, a novel mitochondrial ATP-binding cassette protein involved in iron homeostasis. J. Biol. Chem. 275: 17536-17540.

Mizutani, M., K. Mukaiyama, J. Xiao, M. Mori, R. Satou, S. Narita, S. Okuda, and H. Tokuda. (2013). Functional differentiation of structurally similar membrane subunits of the ABC transporter LolCDE complex. FEBS Lett. 587: 23-29.

Modali, S.D. and H.I. Zgurskaya. (2011). The periplasmic membrane proximal domain of MacA acts as a switch in stimulation of ATP hydrolysis by MacB transporter. Mol. Microbiol. 81: 937-951.

Mohammad, M.M., N. Tomita, M. Ohta, and L. Movileanu. (2016). The Transmembrane Domain of a Bicomponent ABC Transporter Exhibits Channel-forming Activity. ACS Chem Biol. [Epub: Ahead of Print]

Möhle, L., B. Schwarzová, M. Krohn, S.M. Stefan, and J. Pahnke. (2020). Using a qPCR device to screen for modulators of ABC transporter activity: A step-by-step protocol. J Pharmacol Toxicol Methods 104: 106882. [Epub: Ahead of Print]

Mohn, W.W., R. van der Geize, G.R. Stewart, S. Okamoto, J. Liu, L. Dijkhuizen, and L.D. Eltis. (2008). The actinobacterial mce4 locus encodes a steroid transporter. J. Biol. Chem. 283: 35368-35374.

Moisi, M., S. Lichtenegger, S. Tutz, A. Seper, S. Schild, and J. Reidl. (2013). Characterizing the hexose-6-phosphate transport system of Vibrio cholerae, a utilization system for carbon and phosphate sources. J. Bacteriol. 195: 1800-1808.

Moitra, K., L. Silverton, K. Limpert, K. Im, and M. Dean. (2011). Moving out: from sterol transport to drug resistance - the ABCG subfamily of efflux pumps. Drug Metabol Drug Interact 26: 105-111.

Mok S., Liong KY., Lim EH., Huang X., Zhu L., Preiser PR. and Bozdech Z. (2014). Structural polymorphism in the promoter of pfmrp2 confers Plasmodium falciparum tolerance to quinoline drugs. Mol Microbiol. 91(5):918-34.

Molday RS., Zhong M. and Quazi F. (2009). The role of the photoreceptor ABC transporter ABCA4 in lipid transport and Stargardt macular degeneration. Biochim Biophys Acta. 1791(7):573-83.

Molday, R.S. (2007). ATP-binding cassette transporter ABCA4: molecular properties and role in vision and macular degeneration. J. Bioenerg. Biomembr. 39: 507-517.

Mollapour, M., A. Shepherd, and P.W. Piper. (2008). Novel stress responses facilitate Saccharomyces cerevisiae growth in the presence of the monocarboxylate preservatives. Yeast 25: 169-177.

Momma, K., M. Okamoto, Y. Mishima, S. Mori, W. Hashimoto, and K. Murata. (2000). A novel bacterial ATP-binding cassette transporter system that allows uptake of macromolecules. J. Bacteriol. 182: 3998-4004.

Monod, M., M. Feuermann, K. Salamin, M. Fratti, M. Makino, M.M. Alshahni, K. Makimura, and T. Yamada. (2019). Trichophyton rubrum Azole Resistance Mediated by a New ABC Transporter, TruMDR3. Antimicrob. Agents Chemother. 63:.

Montañez, G.E., M.N. Neely, and Z. Eichenbaum. (2005). The streptococcal iron uptake (Siu) transporter is required for iron uptake and virulence in a zebrafish infection model. Microbiology 151: 3749-3757.

Monte, N., K.B.C.C. Pantoja, J.C.G. Rodrigues, D.C. de Carvalho, T.C.B. Azevedo, E.E.B. Pereira, P.P. de Assumpção, S.E.B. Dos Santos, M.R. Fernandes, and N.P.C. Dos Santos. (2021). Polymorphisms in the CYP2A6 and ABCC4 genes are associated with a protective effect on chronic myeloid leukemia in the Brazilian Amazon population. Mol Genet Genomic Med 9: e1694.

Moodley C., Reid SJ. and Abratt VR. (2015). Molecular characterisation of ABC-type multidrug efflux systems in Bifidobacterium longum. Anaerobe. 32:63-9.

Moosavi, F., T. Damghani, S. Ghazi, and S. Pirhadi. (2022). screening of c-Met tyrosine kinase inhibitors targeting nucleotide and drug-substrate binding sites of ABCB1 as potential MDR reversal agents. J Recept Signal Transduct Res 1-13. [Epub: Ahead of Print]

Mora Lagares, L. and M. Novič. (2022). Recent Advances on P-Glycoprotein (ABCB1) Transporter Modelling with In Silico Methods. Int J Mol Sci 23:.

Mora Lagares, L., N. Minovski, A.Y. Caballero Alfonso, E. Benfenati, S. Wellens, M. Culot, F. Gosselet, and M. Novič. (2020). Homology Modeling of the Human P-glycoprotein (ABCB1) and Insights into Ligand Binding through Molecular Docking Studies. Int J Mol Sci 21:.

Mora Lagares, L., N. Minovski, and M. Novič. (2019). Multiclass Classifier for P-Glycoprotein Substrates, Inhibitors, and Non-Active Compounds. Molecules 24:.

Moradi, M. and E. Tajkhorshid. (2013). Mechanistic picture for conformational transition of a membrane transporter at atomic resolution. Proc. Natl. Acad. Sci. USA 110: 18916-18921.

Moreno, M.J., H.A.L. Filipe, S.V.P. Cunha, C.V. Ramos, P.A.T. Martins, B. Abel, L.M.S. Loura, and S.V. Ambudkar. (2023). Interaction of a Homologous Series of Amphiphiles with P-glycoprotein in a Membrane Environment-Contributions of Polar and Non-Polar Interactions. Pharmaceutics 15:.

Moresco, M., L.N. Riccardi, F. Pizza, C. Zenesini, L. Caporali, G. Plazzi, and S. Pelotti. (2016). Pharmacogenetics and Treatment Response in Narcolepsy Type 1: Relevance of the Polymorphisms of the Drug Transporter Gene ABCB1. Clin Neuropharmacol 39: 18-23.

Morgan, J.L., J.F. Acheson, and J. Zimmer. (2017). Structure of a Type-1 Secretion System ABC Transporter. Structure 25: 522-529.

Moslavac, S., K. Nicolaisen, O. Mirus, F. Al Dehni, R. Pernil, E. Flores, I. Maldener, and E. Schleiff. (2007). A TolC-like protein is required for heterocyst development in Anabaena sp. strain PCC 7120. J. Bacteriol. 189: 7887-7895.

Moussatova, A., C. Kandt, M.L. O'Mara, and D.P. Tieleman. (2008). ATP-binding cassette transporters in Escherichia coli. Biochim. Biophys. Acta. 1778: 1757-1771.

Mukherjee, A., P.K. Padmanabhan, S. Singh, G. Roy, I. Girard, M. Chatterjee, M. Ouellette, and R. Madhubala. (2007). Role of ABC transporter MRPA, γ-glutamylcysteine synthetase and ornithine decarboxylase in natural antimony-resistant isolates of Leishmania donovani. J Antimicrob Chemother 59: 204-211.

Müller, S.I., M. Valdebenito, and K. Hantke. (2009). Salmochelin, the long-overlooked catecholate siderophore of Salmonella. Biometals 22: 691-695.

Münch, G., A. Bültmann, Z. Li, H.P. Holthoff, J. Ullrich, S. Wagner, and M. Ungerer. (2012). Overexpression of ABCG1 protein attenuates arteriosclerosis and endothelial dysfunction in atherosclerotic rabbits. Heart Int 7: e12.

Murakami, M., A. Sajid, S. Lusvarghi, S.R. Durell, B. Abel, S. Vahedi, J. Golin, and S.V. Ambudkar. (2023). Second-site suppressor mutations reveal connection between the drug-binding pocket and nucleotide-binding domain 1 of human P-glycoprotein (ABCB1). Drug Resist Updat 71: 101009. [Epub: Ahead of Print]

Murali, R., R.B. Gennis, and J. Hemp. (2021). Evolution of the cytochrome bd oxygen reductase superfamily and the function of CydAA'' in Archaea. ISME J 15: 3534-3548.

Murat, D., L. Goncalves, and E. Dassa. (2008). Deletion of the Escherichia coli uup gene encoding a protein of the ATP binding cassette superfamily affects bacterial competitiveness. Res. Microbiol. 159: 671-677.

Murata, K., S. Kawai, and W. Hashimoto. (2022). Bacteria with a mouth: Discovery and new insights into cell surface structure and macromolecule transport. Proc Jpn Acad Ser B Phys Biol Sci 98: 529-552.

Murata, K., S. Kawai, B. Mikami, and W. Hashimoto. (2008). Superchannel of bacteria: biological significance and new horizons. Biosci. Biotechnol. Biochem. 72: 265-277.

Naaz, S., N. Ahmad, M.R. Jameel, A.A. Al-Huqail, F. Khan, and M.I. Qureshi. (2023). Impact of Some Toxic Metals on Important ABC Transporters in Soybean ( L.). ACS Omega 8: 27597-27611.

Nabi, D., D. Bosi, N. Gupta, N. Thaker, R.A. Fissore, and L.M. Brayboy. (2022). Multidrug resistance transporter-1 dysfunction perturbs meiosis and Ca2+ homeostasis in oocytes. Reproduction. [Epub: Ahead of Print]

Nagao K., Kimura Y., Mastuo M. and Ueda K. (2010). Lipid outward translocation by ABC proteins. FEBS Lett. 584(13):2717-23.

Nagao, K., Y. Taguchi, M. Arioka, H. Kadokura, A. Takatsuki, K. Yoda, and M. Yamasaki. (1995). bfr1+, a novel gene of Schizosaccharomyces pombe which confers brefeldin A resistance, is structurally related to the ATP-binding cassette superfamily. J. Bacteriol. 177: 1536-1543.

Nagy R., Grob H., Weder B., Green P., Klein M., Frelet-Barrand A., Schjoerring JK., Brearley C. and Martinoia E. (2009). The Arabidopsis ATP-binding cassette protein AtMRP5/AtABCC5 is a high affinity inositol hexakisphosphate transporter involved in guard cell signaling and phytate storage. J Biol Chem. 284(48):33614-22.

Nagy, T., &.#.1.9.3.;. Tóth, &.#.1.9.3.;. Telbisz, B. Sarkadi, H. Tordai, A. Tordai, and T. Hegedűs. (2020). The transport pathway in the ABCG2 protein and its regulation revealed by molecular dynamics simulations. Cell Mol Life Sci. [Epub: Ahead of Print]

Nakamura, H., T. Hisano, M.M. Rahman, T. Tosha, M. Shirouzu, and Y. Shiro. (2022). Structural basis for heme detoxification by an ATP-binding cassette-type efflux pump in gram-positive pathogenic bacteria. Proc. Natl. Acad. Sci. USA 119: e2123385119.

Nakano, S., M. Fukaya, and S. Horinouchi. (2006). Putative ABC transporter responsible for acetic acid resistance in Acetobacter aceti. Appl. Environ. Microbiol. 72: 497-505.

Nakayama, H., K. Tanabe, M. Bard, W. Hodgson, S. Wu, D. Takemori, T. Aoyama, N.S. Kumaraswami, L. Metzler, Y. Takano, H. Chibana, and M. Niimi. (2007). The Candida glabrata putative sterol transporter gene CgAUS1 protects cells against azoles in the presence of serum. J Antimicrob Chemother 60: 1264-1272.

Namasivayam, V., K. Stefan, J. Pahnke, and S.M. Stefan. (2021). Binding mode analysis of ABCA7 for the prediction of novel Alzheimer''s disease therapeutics. Comput Struct Biotechnol J 19: 6490-6504.

Nan, J., Y. Zhou, C. Yang, E. Brostromer, O. Kristensen, and X.D. Su. (2009). Structure of a fatty-acid-binding protein from Bacillus subtilis determined by sulfur-SAD phasing using in-house chromium radiation. Acta Crystallogr D Biol Crystallogr 65: 440-448.

Nanavati, D.M., K. Thirangoon, and K.M. Noll. (2006). Several archaeal homologs of putative oligopeptide-binding proteins encoded by Thermotoga maritima bind sugars. Appl. Environ. Microbiol. 72: 1336-1345.

Nanavati, D.M., T.N. Nguyen, and K.M. Noll. (2005). Substrate specificities and expression patterns reflect the evolutionary divergence of maltose ABC transporters in Thermotoga maritima. J. Bacteriol. 187: 2002-2009.

Naoe, Y., N. Nakamura, A. Doi, M. Sawabe, H. Nakamura, Y. Shiro, and H. Sugimoto. (2016). Crystal structure of bacterial haem importer complex in the inward-facing conformation. Nat Commun 7: 13411.

Narayanan, S., Y.F. Fan, N.A. Gujarati, Q.X. Teng, J.Q. Wang, C.Y. Cai, Y. Yang, A.J. Chintalapati, Y. Lei, V.L. Korlipara, and Z.S. Chen. (2021). VKNG-1 Antagonizes ABCG2-Mediated Multidrug Resistance via p-AKT and Bcl-2 Pathway in Colon Cancer: In Vitro and In Vivo Study. Cancers (Basel) 13:.

Narita, S., K. Kanamaru, S. Matsuyama, and H. Tokuda. (2003). A mutation in the membrane subunit of an ABC transporter LolCDE complex causing outer membrane localization of lipoproteins against their inner membrane-specific signals. Mol. Microbiol. 49: 167-177.

Narita, S., K. Tanaka, S. Matsuyama, and H. Tokuda. (2002). Disruption of lolCDE, encoding an ATP-binding cassette transporter, is lethal for Escherichia coli and prevents release of lipoproteins from the inner membrane. J. Bacteriol. 184: 1417-1422.

Naseer, N., J.A. Shapiro, and M. Chander. (2014). RNA-Seq analysis reveals a six-gene SoxR regulon in Streptomyces coelicolor. PLoS One 9: e106181.

Nasyrova, R.F., N.A. Shnayder, S.M. Osipova, A.K. Khasanova, I.S. Efremov, M. Al-Zamil, M.M. Petrova, E.A. Narodova, N.P. Garganeeva, and G.A. Shipulin. (2023). Genetic Predictors of Antipsychotic Efflux Impairment via Blood-Brain Barrier: Role of Transport Proteins. Genes (Basel) 14:.

Navarro-Quiles, C., E. Mateo-Bonmatí, and J.L. Micol. (2018). ABCE Proteins: From Molecules to Development. Front Plant Sci 9: 1125.

Navis, A. and M. Bagnat. (2015). Loss of cftr function leads to pancreatic destruction in larval zebrafish. Dev Biol 399: 237-248.

Navis, A., L. Marjoram, and M. Bagnat. (2013). Cftr controls lumen expansion and function of Kupffer's vesicle in zebrafish. Development 140: 1703-1712.

Negri, A., M. Ferrari, R. Nodari, E. Coppa, V. Mastrantonio, S. Zanzani, D. Porretta, C. Bandi, S. Urbanelli, and S. Epis. (2019). Gene silencing through RNAi and antisense Vivo-Morpholino increases the efficacy of pyrethroids on larvae of Anopheles stephensi. Malar J 18: 294.

Nemchinova, M., G.K. Schuurman-Wolters, J.J. Whittaker, V. Arkhipova, S.J. Marrink, B. Poolman, and A. Guskov. (2024). Exploring the Ligand Binding and Conformational Dynamics of the Substrate-Binding Domain 1 of the ABC Transporter GlnPQ. J Phys Chem B 128: 7822-7832.

Nessa A., Aziz QH., Thomas AM., Harmer SC., Tinker A. and Hussain K. (2015). Molecular mechanisms of congenital hyperinsulinism due to autosomal dominant mutations in ABCC8. Hum Mol Genet. 24(18):5142-53.

Netz, D.J., H.G. Sahl, R. Marcelino, J. dos Santos Nascimento, S.S. de Oliveira, M.B. Soares, M. do Carmo de Freire Bastos, and R. Marcolino. (2001). Molecular characterisation of aureocin A70, a multi-peptide bacteriocin isolated from Staphylococcus aureus. J. Mol. Biol. 311: 939-949.

Neubauer, O., A. Alfandega, J. Schoknecht, U. Sternberg, A. Pohlmann, and T. Eitinger. (2009). Two essential arginine residues in the T components of energy-coupling factor transporters. J. Bacteriol. 191: 6482-6488.

Neubauer, O., C. Reiffler, L. Behrendt, and T. Eitinger. (2011). Interactions among the A and T units of an ECF-type biotin transporter analyzed by site-specific crosslinking. PLoS One 6: e29087.

Neumann, L. and R. Tampé. (1999). Kinetic analysis of peptide binding to the TAP transport complex: evidence for structural rearrangements induced by substrate binding. J. Mol. Biol. 294: 1203-1213.

Neupane, D.P., S. Kumar, and E.T. Yukl. (2018). Two ABC Transporters and a Periplasmic Metallochaperone Participate in Zinc Acquisition in Paracoccus denitrificans. Biochemistry. [Epub: Ahead of Print]

Neville, S.L., J. Sjöhamn, J.A. Watts, H. MacDermott-Opeskin, S.J. Fairweather, K. Ganio, A. Carey Hulyer, A.P. McGrath, A.J. Hayes, T.R. Malcolm, M.R. Davies, N. Nomura, S. Iwata, M.L. O''Mara, M.J. Maher, and C.A. McDevitt. (2021). The structural basis of bacterial manganese import. Sci Adv 7:.

Nguyen, P.T., J.Y. Lai, A.T. Lee, J.T. Kaiser, and D.C. Rees. (2018). Noncanonical role for the binding protein in substrate uptake by the MetNI methionine ATP Binding Cassette (ABC) transporter. Proc. Natl. Acad. Sci. USA 115: E10596-E10604.

Nguyet Nguyen, T.M., H. Park, T.T. Do, J.Y. Kwak, C.K. Lee, S.H. Lee, J.I. Park, S.Y. Yoon, H. Kim, J. Park, and J.T. Park. (2024). CE9A215 (inotodiol), a lanostane-type oxysterol, mitigates LPS-induced sepsis through multifaceted mechanisms. Eur J Pharmacol 980: 176836.

Nicolaou, S.A., A.G. Fast, E. Nakamaru-Ogiso, and E.T. Papoutsakis. (2013). Overexpression of fetA (ybbL) and fetB (ybbM), Encoding an Iron Exporter, Enhances Resistance to Oxidative Stress in Escherichia coli. Appl. Environ. Microbiol. 79: 7210-7219.

Nielsen, L.E., E.C. Snesrud, F. Onmus-Leone, Y.I. Kwak, R. Avilés, E.D. Steele, D.E. Sutter, P.E. Waterman, and E.P. Lesho. (2014). IS5 element integration, a novel mechanism for rapid in vivo emergence of tigecycline nonsusceptibility in Klebsiella pneumoniae. Antimicrob. Agents Chemother. 58: 6151-6156.

Nieves-Morión, M. and E. Flores. (2017). Multiple ABC glucoside transporters mediate sugar-stimulated growth in the heterocyst-forming cyanobacterium Anabaena sp. strain PCC 7120. Environ Microbiol Rep. [Epub: Ahead of Print]

Nigam, S.K. (2015). What do drug transporters really do? Nat Rev Drug Discov 14: 29-44.

Niimi K., Harding DR., Holmes AR., Lamping E., Niimi M., Tyndall JD., Cannon RD. and Monk BC. (2012). Specific interactions between the Candida albicans ABC transporter Cdr1p ectodomain and a D-octapeptide derivative inhibitor. Mol Microbiol. 85(4):747-67.

Niimi, M., K. Niimi, K. Tanabe, R.D. Cannon, and E. Lamping. (2021). Inhibitor Resistant Mutants Give Important Insights into ABC Transporter Cdr1 Substrate Specificity and Help Elucidate Efflux Pump Inhibition. Antimicrob. Agents Chemother. AAC0174821. [Epub: Ahead of Print]

Nim S., Rawal MK. and Prasad R. (2014). FK520 interacts with the discrete intrahelical amino acids of multidrug transporter Cdr1 protein and acts as antagonist to selectively chemosensitize azole-resistant clinical isolates of Candida albicans. FEMS Yeast Res. 14(4):624-32.

Nim, S., L.G. Lobato, A. Moreno, V. Chaptal, M.K. Rawal, P. Falson, and R. Prasad. (2016). Atomic modelling and systematic mutagenesis identify residues in multiple drug binding sites that are essential for drug resistance in the major Candida transporter Cdr1. Biochim. Biophys. Acta. 1858: 2858-2870. [Epub: Ahead of Print]

Ning Y., Dang H., Liu G., Xiong J., Yuan D., Feng L. and Miao W. (2015). ATP-binding cassette transporter enhances tolerance to DDT in Tetrahymena. Sci China Life Sci. 58(3):297-304.

Nishi, K., M. Yoshida, M. Nishimura, M. Nishikawa, M. Nishiyama, S. Horinouchi, and T. Beppu. (1992). A leptomycin B resistance gene of Schizosaccharomyces pombe encodes a protein similar to the mammalian P-glycoproteins. Mol. Microbiol. 6: 761-769.

Nishijyo, T., S.M. Park, C.D. Lu, Y. Itoh, and A.T. Abdelal. (1998). Molecular characterization and regulation of an operon encoding a system for transport of arginine and ornithine and the ArgR regulatory protein in Pseudomonas aeruginosa. J. Bacteriol. 180: 5559-5566.

Nishimura, T., Y. Takahashi, O. Yamaguchi, H. Suzuki, S. Maeda, and T. Omata. (2008). Mechanism of low CO2-induced activation of the cmp bicarbonate transporter operon by a LysR family protein in the cyanobacterium Synechococcus elongatus strain PCC 7942. Mol. Microbiol. 68: 98-109.

Niu, L., H. Li, Z. Song, B. Dong, H. Cao, T. Liu, T. Du, W. Yang, R. Amin, L. Wang, Q. Yang, D. Meng, and Y. Fu. (2021). The functional analysis of ABCG transporters in the adaptation of pigeon pea () to abiotic stresses. PeerJ 9: e10688.

Niu, W., X. Rong, Q. Zhao, X. Liu, L. Xu, S. Li, and X. Li. (2023). [Wine-processed enhances efficacy of aumolertinib against EGFRmutant non-small cell lung cancer xenografts in nude mouse brain]. Nan Fang Yi Ke Da Xue Xue Bao 43: 375-382.

Nodwell, J.R., K. McGovern, and R. Losick. (1996). An oligopeptide permease responsible for the import of an extracellular signal governing aerial mycelium formation in Streptomyces coelicolor. Mol. Microbiol. 22: 881-893.

Nöll, A., C. Thomas, V. Herbring, T. Zollmann, K. Barth, A.R. Mehdipour, T.M. Tomasiak, S. Brüchert, B. Joseph, R. Abele, V. Oliéric, M. Wang, K. Diederichs, G. Hummer, R.M. Stroud, K.M. Pos, and R. Tampé. (2017). Crystal structure and mechanistic basis of a functional homolog of the antigen transporter TAP. Proc. Natl. Acad. Sci. USA 114: E438-E447.

Norimatsu, Y., A. Ivetac, C. Alexander, J. Kirkham, N. O'Donnell, D.C. Dawson, and M.S. Sansom. (2012). Cystic fibrosis transmembrane conductance regulator: a molecular model defines the architecture of the anion conduction path and locates a "bottleneck" in the pore. Biochemistry 51: 2199-2212.

Novikova, M., A. Metlitskaya, K. Datsenko, T. Kazakov, A. Kazakov, B. Wanner, and K. Severinov. (2007). The Escherichia coli Yej transporter is required for the uptake of translation inhibitor microcin C. J. Bacteriol. 189: 8361-8365.

Novischi, S.Y.P., A. Karoly-Lakatos, K. Chok, C. Bonifer, J. Becker-Baldus, and C. Glaubitz. (2024). Probing the allosteric NBD-TMD crosstalk in the ABC transporter MsbA by solid-state NMR. Commun Biol 7: 43.

Nyathi, Y., C. De Marcos Lousa, C.W. van Roermund, R.J. Wanders, B. Johnson, S.A. Baldwin, F.L. Theodoulou, and A. Baker. (2010). The Arabidopsis peroxisomal ABC transporter, comatose, complements the Saccharomyces cerevisiae pxa1 pxa2Delta mutant for metabolism of long-chain fatty acids and exhibits fatty acyl-CoA-stimulated ATPase activity. J. Biol. Chem. 285: 29892-29902.

Nygaard, T.K., G.C. Blouin, M. Liu, M. Fukumura, J.S. Olson, M. Fabian, D.M. Dooley, and B. Lei. (2006). The mechanism of direct heme transfer from the streptococcal cell surface protein Shp to HtsA of the HtsABC transporter. J. Biol. Chem. 281: 20761-20771.

Obando S, T.A., M.M. Babykin, and V.V. Zinchenko. (2018). A Cluster of Five Genes Essential for the Utilization of Dihydroxamate Xenosiderophores in Synechocystis sp. PCC 6803. Curr. Microbiol. 75: 1165-1173.

Obis, D., A. Guillot, J.C. Gripon, P. Renault, A. Bolotin, and M.Y. Mistou. (1999). Genetic and biochemical characterization of a high-affinity betaine uptake system (BusA) in Lactococcus lactis reveals a new functional organization within bacterial ABC transporters. J. Bacteriol. 181: 6238-6246.

Oepen, K., V. Mater, and D. Schneider. (2023). Unfolding Individual Domains of BmrA, a Bacterial ABC Transporter Involved in Multidrug Resistance. Int J Mol Sci 24:.

Ogura, M., K. Tsukahara, K. Hayashi, and T. Tanaka. (2007). The Bacillus subtilis NatK-NatR two-component system regulates expression of the natAB operon encoding an ABC transporter for sodium ion extrusion. Microbiology (Reading) 153: 667-675.

Ohki, R., Giyanto, K. Tateno, W. Masuyama, S. Moriya, K. Kobayashi, and N. Ogasawara. (2003). The BceRS two-component regulatory system induces expression of the bacitracin transporter, BceAB, in Bacillus subtilis. Mol. Microbiol. 49: 1135-1144.

Ohki, R., K. Tateno, T. Takizawa, T. Aiso, and M. Murata. (2005). Transcriptional termination control of a novel ABC transporter gene involved in antibiotic resistance in Bacillus subtilis. J. Bacteriol. 187: 5946-5954.

Ohnuma, S., E. Chufan, K. Nandigama, L.M. Jenkins, S.R. Durell, E. Appella, Z.E. Sauna, and S.V. Ambudkar. (2011). Inhibition of multidrug resistance-linked P-glycoprotein (ABCB1) function by 5'-fluorosulfonylbenzoyl 5'-adenosine: evidence for an ATP analogue that interacts with both drug-substrate-and nucleotide-binding sites. Biochemistry 50: 3724-3735.

Okada, U., E. Yamashita, A. Neuberger, M. Morimoto, H.W. van Veen, and S. Murakami. (2017). Crystal structure of tripartite-type ABC transporter MacB from Acinetobacter baumannii. Nat Commun 8: 1336.

Okuda, K., S. Yanagihara, T. Sugayama, T. Zendo, J. Nakayama, and K. Sonomoto. (2010). Functional significance of the E loop, a novel motif conserved in the lantibiotic immunity ATP-binding cassette transport systems. J. Bacteriol. 192: 2801-2808.

Okuda, K., Y. Aso, J. Nakayama, and K. Sonomoto. (2008). Cooperative transport between NukFEG and NukH in immunity against the lantibiotic nukacin ISK-1 produced by Staphylococcus warneri ISK-1. J. Bacteriol. 190: 356-362.

Oldham, M.L. and J. Chen. (2011). Crystal structure of the maltose transporter in a pretranslocation intermediate state. Science 332: 1202-1205.

Oldham, M.L. and J. Chen. (2011). Snapshots of the maltose transporter during ATP hydrolysis. Proc. Natl. Acad. Sci. USA 108: 15152-15156.

Oldham, M.L., D. Khare, F.A. Quiocho, A.L. Davidson, and J. Chen. (2007). Crystal structure of a catalytic intermediate of the maltose transporter. Nature 450: 515-521.

Oldham, M.L., S. Chen, and J. Chen. (2013). Structural basis for substrate specificity in the Escherichia coli maltose transport system. Proc. Natl. Acad. Sci. USA 110: 18132-18137.

Oliveira, A.S., A.M. Baptista, and C.M. Soares. (2011). Conformational changes induced by ATP-hydrolysis in an ABC transporter: a molecular dynamics study of the Sav1866 exporter. Proteins 79: 1977-1990.

Olsen, J.A., A. Alam, J. Kowal, B. Stieger, and K.P. Locher. (2019). Structure of the human lipid exporter ABCB4 in a lipid environment. Nat Struct Mol Biol. [Epub: Ahead of Print]

Onnée, M., B. Duriez, S. Simon, C. Szczepaniak, A. Guguin, R. Epaud, S. Hamouda, P. Fanen, and A. de Becdelièvre. (2022). Instability of Mature ABCA3 Protein: Toward a New Classification of ABCA3 Mutations? Am J Respir Cell Mol Biol 67: 602-605.

Onnée, M., P. Fanen, I. Callebaut, and A. de Becdelièvre. (2021). Structure-Based Understanding of ABCA3 Variants. Int J Mol Sci 22:.

Oram, J.F., G. Wolfbauer, C. Tang, W.S. Davidson, and J.J. Albers. (2008). An amphipathic helical region of the N-terminal barrel of phospholipid transfer protein is critical for ABCA1-dependent cholesterol efflux. J. Biol. Chem. 283: 11541-11549.

Orlowski, S., C. Coméra, F. Tercé, and X. Collet. (2007). Lipid rafts: dream or reality for cholesterol transporters? Eur Biophys. J. 36: 869-885.

Ortega, &.#.1.9.3.;., M.A. Matilla, and T. Krell. (2022). The Repertoire of Solute-Binding Proteins of Model Bacteria Reveals Large Differences in Number, Type, and Ligand Range. Microbiol Spectr 10: e0205422.

Ortiz D., Gossack L., Quast U. and Bryan J. (2013). Reinterpreting the action of ATP analogs on K(ATP) channels. J Biol Chem. 288(26):18894-902.

Ortiz, D. and J. Bryan. (2015). Neonatal Diabetes and Congenital Hyperinsulinism Caused by Mutations in ABCC8/SUR1 are Associated with Altered and Opposite Affinities for ATP and ADP. Front Endocrinol (Lausanne) 6: 48.

Ortiz, D.F., L. Kreppel, D.M. Speiser, G. Scheel, G. McDonald, and D.W. Ow. (1992). Heavy metal tolerance in the fission yeast requires an ATP-binding cassette-type vacuolar membrane transporter. EMBO. J. 11: 3491-3499.

Ouattara, M., E.B. Cunha, X. Li, Y.S. Huang, D. Dixon, and Z. Eichenbaum. (2010). Shr of group A streptococcus is a new type of composite NEAT protein involved in sequestering haem from methaemoglobin. Mol. Microbiol. 78: 739-756.

Ousalem, F., S. Singh, O. Chesneau, J.F. Hunt, and G. Boël. (2019). ABC-F proteins in mRNA translation and antibiotic resistance. Res. Microbiol. 170: 435-447.

Ozguc Caliskan, B., K. Uslu, N. Sinim Kahraman, K. Erkilic, A. Oner, and M. Dundar. (2024). Beyond the phenotype: Exploring inherited retinal diseases with targeted next-generation sequencing in a Turkish cohort. Clin Genet 106: 258-266.

Ozvegy-Laczka, C., G. Köblös, B. Sarkadi, and A. Váradi. (2005). Single amino acid (482) variants of the ABCG2 multidrug transporter: major differences in transport capacity and substrate recognition. Biochim. Biophys. Acta. 1668: 53-63.

Padayatti, P.S., S.C. Lee, R.L. Stanfield, P.C. Wen, E. Tajkhorshid, I.A. Wilson, and Q. Zhang. (2019). Structural Insights into the Lipid A Transport Pathway in MsbA. Structure. [Epub: Ahead of Print]

Pagès, J.M., L. Amaral, and S. Fanning. (2011). An original deal for new molecule: reversal of efflux pump activity, a rational strategy to combat gram-negative resistant bacteria. Curr. Med. Chem. 18: 2969-2980.

Paik, S., A. Brown, C.L. Munro, C.N. Cornelissen, and T. Kitten. (2003). The sloABCR operon of Streptococcus mutans encodes an Mn and Fe transport system required for endocarditis virulence and its Mn-dependent repressor. J. Bacteriol. 185: 5967-5975.

Pakuła, K., C. Sequeiros-Borja, W. Biała-Leonhard, A. Pawela, J. Banasiak, A. Bailly, M. Radom, M. Geisler, J. Brezovsky, and M. Jasiński. (2023). Restriction of access to the central cavity is a major contributor to substrate selectivity in plant ABCG transporters. Cell Mol Life Sci 80: 105.

Palikhe, S., U. Uuganbayar, H.K.T. Trinh, G.Y. Ban, E.M. Yang, H.S. Park, and S.H. Kim. (2017). A Role of the ABCC4 Gene Polymorphism in Airway Inflammation of Asthmatics. Mediators Inflamm 2017: 3549375.

Pan, C., J. Weng, and W. Wang. (2016). ATP Hydrolysis Induced Conformational Changes in the Vitamin B12 Transporter BtuCD Revealed by MD Simulations. PLoS One 11: e0166980.

Pan, D., R. Oyama, T. Sato, T. Nakane, R. Mizunuma, K. Matsuoka, Y. Joti, K. Tono, E. Nango, S. Iwata, T. Nakatsu, and H. Kato. (2022). Crystal structure of CmABCB1 multi-drug exporter in lipidic mesophase revealed by LCP-SFX. IUCrJ 9: 134-145.

Pandey, V., V. Krishnan, N. Basak, A. Marathe, V. Thimmegowda, A. Dahuja, M. Jolly, and A. Sachdev. (2018). Molecular modeling andcharacterization ofABCC5: a phytate transporter and potential target for low-phytate crops. 3 Biotech 8: 54.

Pang, K., Y. Li, M. Liu, Z. Meng, and Y. Yu. (2013). Inventory and general analysis of the ATP-binding cassette (ABC) gene superfamily in maize (Zea mays L.). Gene 526: 411-428.

Panikashvili, D. and A. Aharoni. (2008). ABC-type transporters and cuticle assembly: Linking function to polarity in epidermis cells. Plant Signal Behav 3: 806-809.

Papadelli, M., A. Karsioti, R. Anastasiou, M. Georgalaki, and E. Tsakalidou. (2007). Characterization of the gene cluster involved in the biosynthesis of macedocin, the lantibiotic produced by Streptococcus macedonicus. FEMS Microbiol. Lett. 272: 75-82.

Papadopoulos, M. and F. Momburg. (2007). Multiple residues in the transmembrane helix and connecting peptide of mouse tapasin stabilize the transporter associated with the antigen-processing TAP2 subunit. J. Biol. Chem. 282: 9401-9410.

Park, H.S., S.K. Shin, Y.Y. Yang, H.J. Kwon, and J.W. Suh. (2005). Accumulation of S-adenosylmethionine induced oligopeptide transporters including BldK to regulate differentiation events in Streptomyces coelicolor M145. FEMS Microbiol. Lett. 249: 199-206.

Park, J.S., J.S. Ko, J.K. Seo, J.S. Moon, and S.S. Park. (2016). Clinical and ABCB11 profiles in Korean infants with progressive familial intrahepatic cholestasis. World J Gastroenterol 22: 4901-4907.

Park, J.T., D. Raychaudhuri, H. Li, S. Normark, and D. Mengin-Lecreulx. (1998). MppA, a periplasmic binding protein essential for import of the bacterial cell wall peptide L-alanyl-γ-D-glutamyl-meso-diaminopimelate. J. Bacteriol. 180: 1215-1223.

Parreira, B., J.C.R. Cardoso, R. Costa, A.R. Couto, J. Bruges-Armas, and D.M. Power. (2018). Persistence of the ABCC6 genes and the emergence of the bony skeleton in vertebrates. Sci Rep 8: 6027.

Patel, A., T.W. Li, N. Anreddy, D.S. Wang, K. Sodani, S. Gadhia, R. Kathawala, D.H. Yang, C. Cheng, and Z.S. Chen. (2017). Suppression of ABCG2 mediated MDR in vitro and in vivo by a novel inhibitor of ABCG2 drug transport. Pharmacol Res 121: 184-193.

Patzer, S.I. and K. Hantke. (1998). The ZnuABC high-affinity zinc uptake system and its regulator Zur in Escherichia coli. Mol. Microbiol. 28: 1199-1210.

Paul, S., S. Banerjee, and H.J. Vogel. (2016). Ligand binding specificity of the Escherichia coli periplasmic histidine binding protein, HisJ. Protein. Sci. [Epub: Ahead of Print]

Paulsen, I.T., A.M. Beness, and M.H. Saier, Jr. (1997). Computer-based analyses of the protein constituents of transport systems catalysing export of complex carbohydrates in bacteria. Microbiology 143(Pt8): 2685-2699.

Paumi, C.M., J. Menendez, A. Arnoldo, K. Engels, K.R. Iyer, S. Thaminy, O. Georgiev, Y. Barral, S. Michaelis, and I. Stagljar. (2007). Mapping protein-protein interactions for the yeast ABC transporter Ycf1p by integrated split-ubiquitin membrane yeast two-hybrid analysis. Mol. Cell 26: 15-25.

Paytubi, S., X. Wang, Y.W. Lam, L. Izquierdo, M.J. Hunter, E. Jan, H.S. Hundal, and C.G. Proud. (2009). ABC50 promotes translation initiation in mammalian cells. J. Biol. Chem. 284: 24061-24073.

Pei, J. and Q. Cong. (2022). Evolutionary origin and sequence signatures of the heterodimeric ABCG5/ABCG8 transporter. Protein. Sci. 31: e4297.

Peng, M., C. Zhang, Y.Y. Duan, H.B. Liu, X.Y. Peng, Q. Wei, Q.Y. Chen, H. Sang, and Q.T. Kong. (2023). Antifungal activity of the repurposed drug disulfiram against. Front Pharmacol 14: 1268649.

Pérez Carrillo, V.H., D. Rose-Sperling, M.A. Tran, C. Wiedemann, and U.A. Hellmich. (2022). Backbone NMR assignment of the nucleotide binding domain of the Bacillus subtilis ABC multidrug transporter BmrA in the post-hydrolysis state. Biomol NMR Assign. [Epub: Ahead of Print]

Perez, C., A.R. Mehdipour, G. Hummer, and K.P. Locher. (2019). Structure of Outward-Facing PglK and Molecular Dynamics of Lipid-Linked Oligosaccharide Recognition and Translocation. Structure 27: 669-678.e5.

Perez, C., S. Gerber, J. Boilevin, M. Bucher, T. Darbre, M. Aebi, J.L. Reymond, and K.P. Locher. (2015). Structure and mechanism of an active lipid-linked oligosaccharide flippase. Nature 524: 433-438.

Perez, D.R., L.A. Sklar, A. Chigaev, and K. Matlawska-Wasowska. (2020). Drug repurposing for targeting cyclic nucleotide transporters in acute leukemias - A missed opportunity. Semin Cancer Biol. [Epub: Ahead of Print]

Pérez-Victoria, J.M., F. Cortés-Selva, A. Parodi-Talice, B.I. Bavchvarov, F.J. Pérez-Victoria, F. Muñoz-Martínez, M. Maitrejean, M.P. Costi, D. Barron, A. Di Pietro, S. Castanys, and F. Gamarro. (2006). Combination of suboptimal doses of inhibitors targeting different domains of LtrMDR1 efficiently overcomes resistance of Leishmania spp. to Miltefosine by inhibiting drug efflux. Antimicrob. Agents Chemother. 50: 3102-3110.

Pernil, R., A. Herrero, and E. Flores. (2010). A TRAP transporter for pyruvate and other monocarboxylate 2-oxoacids in the cyanobacterium Anabaena sp. strain PCC 7120. J. Bacteriol. 192: 6089-6092.

Pernil, R., S. Picossi, V. Mariscal, A. Herrero, and E. Flores. (2008). ABC-type amino acid uptake transporters Bgt and N-II of Anabaena sp. strain PCC 7120 share an ATPase subunit and are expressed in vegetative cells and heterocysts. Mol. Microbiol. 67: 1067-1080.

Perria, C.L., V. Rajamanickam, P.E. Lapinski, and M. Raghavan. (2006). Catalytic site modifications of TAP1 and TAP2 and their functional consequences. J. Biol. Chem. 281: 39839-39851.

Phennicie, R.T., M.J. Sullivan, J.T. Singer, J.A. Yoder, and C.H. Kim. (2010). Specific resistance to Pseudomonas aeruginosa infection in zebrafish is mediated by the cystic fibrosis transmembrane conductance regulator. Infect. Immun. 78: 4542-4550.

Philpott, C.C., S. Leidgens, and A.G. Frey. (2012). Metabolic remodeling in iron-deficient fungi. Biochim. Biophys. Acta. 1823: 1509-1520.

Picossi, S., M.L. Montesinos, R. Pernil, C. Lichtlé, A. Herrero, and E. Flores. (2005). ABC-type neutral amino acid permease N-I is required for optimal diazotrophic growth and is repressed in the heterocysts of Anabaena sp. strain PCC 7120. Mol. Microbiol. 57: 1582-1592.

Pietz, H.L., A. Abbas, Z.L. Johnson, M.L. Oldham, H. Suga, and J. Chen. (2023). A macrocyclic peptide inhibitor traps MRP1 in a catalytically incompetent conformation. Proc. Natl. Acad. Sci. USA 120: e2220012120.

Pighin, J.A., H. Zheng, L.J. Balakshin, I.P. Goodman, T.L. Western, R. Jetter, L. Kunst, and A.L. Samuels. (2004). Plant cuticular lipid export requires an ABC transporter. Science 306: 702-704.

Pimpat, Y., N. Saralamba, U. Boonyuen, S. Pukrittayakamee, F. Nosten, F. Smithuis, N.P.J. Day, A.M. Dondorp, and M. Imwong. (2020). Genetic analysis of the orthologous crt and mdr1 genes in Plasmodium malariae from Thailand and Myanmar. Malar J 19: 315.

Pinkett, H.W., A.T. Lee, P. Lum, K.P. Locher, and D.C. Rees. (2007). An inward-facing conformation of a putative metal-chelate-type ABC transporter. Science 315: 373-377.

Pipatthana, M., P. Harnvoravongchai, P. Pongchaikul, S. Likhitrattanapisal, M. Phanchana, S. Chankhamhaengdecha, and T. Janvilisri. (2021). The repertoire of ABC proteins in. Comput Struct Biotechnol J 19: 2905-2920.

Pletzer, D., C. Lafon, Y. Braun, T. Köhler, M.G. Page, M. Mourez, and H. Weingart. (2014). High-throughput screening of dipeptide utilization mediated by the ABC transporter DppBCDF and its substrate-binding proteins DppA1-A5 in Pseudomonas aeruginosa. PLoS One 9: e111311.

Ploetz, E., G.K. Schuurman-Wolters, N. Zijlstra, A.W. Jager, D.A. Griffith, A. Guskov, G. Gouridis, B. Poolman, and T. Cordes. (2021). Structural and biophysical characterization of the tandem substrate-binding domains of the ABC importer GlnPQ. Open Biol 11: 200406.

Podlesek, Z., A. Comino, B. Herzog-Velikonja, D. Zgur-Bertok, R. Komel, and M. Grabnar. (1995). Bacillus licheniformis bacitracin-resistance ABC transporter: relationship to mammalian multidrug resistance. Mol. Microbiol. 16: 969-976.

Podolski-Renić, A., J. Dinić, T. Stanković, M. Jovanović, A. Ramović, A. Pustenko, R. Žalubovskis, and M. Pešić. (2019). Sulfocoumarins, specific carbonic anhydrase IX and XII inhibitors, interact with cancer multidrug resistant phenotype through pH regulation and reverse P-glycoprotein mediated resistance. Eur J Pharm Sci 138: 105012.

Poetz, O., T. Dieze, H.S. Hammer, F. Weiss, C. Sommersdorf, M.F. Templin, C. Esdar, A. Zimmermann, S. Stevanović, J. Bedke, A. Stenzl, and T.O. Joos. (2018). A Peptide-Based Sandwich Immunoassay for the Quantification of the Membrane Transporter Multidrug Resistance Protein 1. Anal Chem. [Epub: Ahead of Print]

Polgar, O., R.W. Robey, K. Morisaki, M. Dean, C. Michejda, Z.E. Sauna, S.V. Ambudkar, N. Tarasova, and S.E. Bates. (2004). Mutational analysis of ABCG2: role of the GXXXG motif. Biochemistry 43: 9448-9456.

Polyviou, D., A. Hitchcock, A.J. Baylay, C.M. Moore, and T.S. Bibby. (2015). Phosphite utilization by the globally important marine diazotroph Trichodesmium. Environ Microbiol Rep 7: 824-830.

Popella, P., S. Krauss, P. Ebner, M. Nega, J. Deibert, and F. Götz. (2016). VraH Is the Third Component of the Staphylococcus aureus VraDEH System Involved in Gallidermin and Daptomycin Resistance and Pathogenicity. Antimicrob. Agents Chemother. 60: 2391-2401.

Popovic, M., R. Zaja, J. Loncar, and T. Smital. (2010). A novel ABC transporter: the first insight into zebrafish (Danio rerio) ABCH1. Mar Environ Res 69Suppl: S11-13.

Posteraro, B., M. Sanguinetti, D. Sanglard, M. La Sorda, S. Boccia, L. Romano, G. Morace, and G. Fadda. (2003). Identification and characterization of a Cryptococcus neoformans ATP binding cassette (ABC) transporter-encoding gene, CnAFR1, involved in the resistance to fluconazole. Mol. Microbiol. 47: 357-371.

Potdukhe, R.M., P. Bedi, B.K. Sarangi, R.A. Pandey, and S.T. Thul. (2018). Root transcripts associated with arsenic accumulation in hyperaccumulator Pteris vittata. J Biosci 43: 105-115.

Pouliot, B., M. Jbel, A. Mercier, and S. Labbé. (2010). abc3+ encodes an iron-regulated vacuolar ABC-type transporter in Schizosaccharomyces pombe. Eukaryot. Cell. 9: 59-73.

Pozzi, R., M. Coles, D. Linke, A. Kulik, M. Nega, W. Wohlleben, and E. Stegmann. (2015). Distinct mechanisms contribute to immunity in the lantibiotic NAI-107 producer strain Microbispora ATCC PTA-5024. Environ Microbiol. [Epub: Ahead of Print]

Pratt, E.B. and S.L. Shyng. (2011). ATP activates ATP-sensitive potassium channels composed of mutant sulfonylurea receptor 1 and Kir6.2 with diminished PIP2 sensitivity. Channels (Austin) 5: 314-319.

Pratt, E.B., P. Tewson, C.E. Bruederle, W.R. Skach, and S.L. Shyng. (2011). N-terminal transmembrane domain of SUR1 controls gating of Kir6.2 by modulating channel sensitivity to PIP2. J Gen Physiol 137: 299-314.

Pratte, B.S. and T. Thiel. (2006). High-affinity vanadate transport system in the cyanobacterium Anabaena variabilis ATCC 29413. J. Bacteriol. 188: 464-468.

Prell, J., J.P. White, A. Bourdes, S. Bunnewell, R.J. Bongaerts, and P.S. Poole. (2009). Legumes regulate Rhizobium bacteroid development and persistence by the supply of branched-chain amino acids. Proc. Natl. Acad. Sci. USA 106: 12477-12482.

Prestin, K., S. Wolf, R. Feldtmann, J. Hussner, I. Geissler, C. Rimmbach, H.K. Kroemer, U. Zimmermann, and H.E. Meyer zu Schwabedissen. (2014). Transcriptional regulation of urate transportosome member SLC2A9 by nuclear receptor HNF4α. Am. J. Physiol. Renal Physiol 307: F1041-1051.

Prévéral, S., L. Gayet, C. Moldes, J. Hoffmann, S. Mounicou, A. Gruet, F. Reynaud, R. Lobinski, J.M. Verbavatz, A. Vavasseur, and C. Forestier. (2009). A common highly conserved cadmium detoxification mechanism from bacteria to humans: heavy metal tolerance conferred by the ATP-binding cassette (ABC) transporter SpHMT1 requires glutathione but not metal-chelating phytochelatin peptides. J. Biol. Chem. 284: 4936-4943.

Prieß, M., H. Göddeke, G. Groenhof, and L.V. Schäfer. (2018). Molecular Mechanism of ATP Hydrolysis in an ABC Transporter. ACS Cent Sci 4: 1334-1343.

Proctor, R.H., D.W. Brown, R.D. Plattner, and A.E. Desjardins. (2003). Co-expression of 15 contiguous genes delineates a fumonisin biosynthetic gene cluster in Gibberella moniliformis. Fungal Genet Biol 38: 237-249.

Proks, P., A.L. Arnold, J. Bruining, C. Girard, S.E. Flanagan, B. Larkin, K. Colclough, A.T. Hattersley, F.M. Ashcroft, and S. Ellard. (2006). A heterozygous activating mutation in the sulphonylurea receptor SUR1 (ABCC8) causes neonatal diabetes. Hum Mol Genet 15: 1793-1800.

Prosecka, J., A.V. Orlov, Y.S. Fantin, V.V. Zinchenko, M.M. Babykin, and M. Tichy. (2009). A novel ATP-binding cassette transporter is responsible for resistance to viologen herbicides in the cyanobacterium Synechocystis sp. PCC 6803. FEBS J. 276: 4001-4011.

Puri, N., M. Gaur, M. Sharma, S. Shukla, S.V. Ambudkar, and R. Prasad. (2009). The amino acid residues of transmembrane helix 5 of multidrug resistance protein CaCdr1p of Candida albicans are involved in substrate specificity and drug transport. Biochim. Biophys. Acta. 1788: 1752-1761.

Puris, E., L. Saveleva, V. Górová, P. Vartiainen, M. Kortelainen, H. Lamberg, O. Sippula, T. Malm, P.I. Jalava, S. Auriola, G. Fricker, and K.M. Kanninen. (2022). Air Pollution Exposure Increases ABCB1 and ASCT1 Transporter Levels in Mouse Cortex. Environ Toxicol Pharmacol 104003. [Epub: Ahead of Print]

Qi, W., J. Li, and J.A. Cowan. (2014). A structural model for glutathione-complexed iron-sulfur cluster as a substrate for ABCB7-type transporters. Chem Commun (Camb) 50: 3795-3798.

Qi, X., E. Wagenaar, W. Xu, K. Huang, and A.H. Schinkel. (2017). Ochratoxin A transport by the human breast cancer resistance protein (BCRP), multidrug resistance protein 2 (MRP2), and organic anion-transporting polypeptides 1A2, 1B1 and 2B1. Toxicol Appl Pharmacol 329: 18-25.

Qian, L., G. Wang, B. Li, H. Su, and L. Qin. (2024). Regulation of lipid metabolism by APOE4 in intrahepatic cholangiocarcinoma the enhancement of ABCA1 membrane expression. PeerJ 12: e16740.

Qiu, Y., H. Li, J. Xie, X. Qiao, and J. Wu. (2021). Identification of ABCC5 Among ATP-Binding Cassette Transporter Family as a New Biomarker for Hepatocellular Carcinoma Based on Bioinformatics Analysis. Int J Gen Med 14: 7235-7246.

Quazi, F. and R.S. Molday. (2011). Lipid transport by mammalian ABC proteins. Essays Biochem 50: 265-290.

Quazi, F., S. Lenevich, and R.S. Molday. (2012). ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat Commun 3: 925.

Quentin, Y. and G. Fichant. (2000). ABCdb: an ABC transporter database. J. Mol. Microbiol. Biotechnol. 2: 501-504.

Quintero, M.J., M.L. Montesinos, A. Herrero, and E. Flores. (2001). Identification of genes encoding amino acid permeases by inactivation of selected ORFs from the Synechocystis genomic sequence. Genome Res 11: 2034-2040.

Racolta, S., P.B. Juhl, D. Sirim, and J. Pleiss. (2012). The triterpene cyclase protein family: a systematic analysis. Proteins 80: 2009-2019.

Rafii, F. and M. Park. (2008). Detection and characterization of an ABC transporter in Clostridium hathewayi. Arch. Microbiol. 190: 417-426.

Raghuraman, G., P.E. Lapinski, and M. Raghavan. (2002). Tapasin interacts with the membrane-spanning domains of both TAP subunits and enhances the structural stability of TAP1 x TAP2 Complexes. J. Biol. Chem. 277: 41786-41794.

Rahman, H., M.J. Ware, A. Sajid, S. Lusvarghi, S.R. Durell, and S.V. Ambudkar. (2023). Residues from Homologous Transmembrane Helices 4 and 10 Are Critical for P-Glycoprotein (ABCB1)-Mediated Drug Transport. Cancers (Basel) 15:.

Rahman, S.J. and P. Kaur. (2018). Conformational changes in a multidrug resistance ABC transporter DrrAB: Fluorescence-based approaches to study substrate binding. Arch Biochem Biophys 658: 31-45. [Epub: Ahead of Print]

Raichaudhuri, A., M. Peng, V. Naponelli, S. Chen, R. Sánchez-Fernández, H. Gu, J.F. Gregory, 3rd, A.D. Hanson, and P.A. Rea. (2009). Plant Vacuolar ATP-binding Cassette Transporters That Translocate Folates and Antifolates in Vitro and Contribute to Antifolate Tolerance in Vivo. J. Biol. Chem. 284: 8449-8460.

Ramjeesingh, M., F. Ugwu, F.L. Stratford, L.J. Huan, C. Li, and C.E. Bear. (2008). The intact CFTR protein mediates ATPase rather than adenylate kinase activity. Biochem. J. 412: 315-321.

Ran, Y. and P.H. Thibodeau. (2016). Stabilization of Nucleotide-Binding Domain Dimers Rescues ABCC6 Mutants Associated with Pseudoxanthoma Elasticum. J. Biol. Chem. [Epub: Ahead of Print]

Randak, C.O. and M.J. Welsh. (2007). Role of CFTR's intrinsic adenylate kinase activity in gating of the Cl- channel. J. Bioenerg. Biomembr. 39: 473-479.

Raschka, S.L., A. Harris, B.F. Luisi, and L. Schmitt. (2022). Flipping and other astonishing transporter dance moves in fungal drug resistance. Bioessays e2200035. [Epub: Ahead of Print]

Rasmussen, M.R., K.L. Nielsen, M.R. Laursen, C.B. Nielsen, P. Svendsen, H. Dimke, E.I. Christensen, M. Johannsen, and S.K. Moestrup. (2016). Untargeted Metabolomics Analysis of ABCC6-Deficient Mice Discloses an Altered Metabolic Liver Profile. J Proteome Res 15: 4591-4600.

Rasmussen, M.R., M. Sommerlund, and S.K. Moestrup. (2013). Is classical pseudoxanthoma elasticum a consequence of hepatic ''intoxication'' due to ABCC6 substrate accumulation in the liver? Expert Rev Endocrinol Metab 8: 37-46.

Rawal MK., Khan MF., Kapoor K., Goyal N., Sen S., Saxena AK., Lynn AM., Tyndall JD., Monk BC., Cannon RD., Komath SS. and Prasad R. (2013). Insight into pleiotropic drug resistance ATP-binding cassette pump drug transport through mutagenesis of Cdr1p transmembrane domains. J Biol Chem. 288(34):24480-93.

Rayapuram, N., J. Hagenmuller, J.M. Grienenberger, P. Giegé, and G. Bonnard. (2007). AtCCMA interacts with AtCcmB to form a novel mitochondrial ABC transporter involved in cytochrome c maturation in Arabidopsis. J. Biol. Chem. 282: 21015-21023.

Rea, P.A., Z.S. Li, Y.P. Lu, Y.M. Drozdowicz, and E. Martinoia. (1998). FROM VACUOLAR GS-X PUMPS TO MULTISPECIFIC ABC TRANSPORTERS. Annu. Rev. Plant Physiol. Plant Mol. Biol. 49: 727-760.

Reboul, E. (2013). Absorption of vitamin A and carotenoids by the enterocyte: focus on transport proteins. Nutrients 5: 3563-3581.

Reid, G., P. Wielinga, N. Zelcer, I. van der Heijden, A. Kuil, M. de Haas, J. Wijnholds, and P. Borst. (2003). The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc. Natl. Acad. Sci. USA 100: 9244-9249.

Rempel, S., W.K. Stanek, and D.J. Slotboom. (2018). Energy-Coupling Factor-Type ATP-Binding Cassette Transporters. Annu. Rev. Biochem. [Epub: Ahead of Print]

Reuter, G., T. Janvilisri, H. Venter, S. Shahi, L. Balakrishnan, and H.W. van Veen. (2003). The ATP binding cassette multidrug transporter LmrA and lipid transporter MsbA have overlapping substrate specificities. J. Biol. Chem. 278: 35193-35198.

Revilla-Guarinos, A., S. Gebhard, C. Alcántara, A. Staron, T. Mascher, and M. Zúñiga. (2013). Characterization of a regulatory network of peptide antibiotic detoxification modules in Lactobacillus casei BL23. Appl. Environ. Microbiol. 79: 3160-3170.

Reznicek, J., M. Ceckova, Z. Ptackova, O. Martinec, L. Tupova, L. Cerveny, and F. Staud. (2017). MDR1 and BCRP transporter-mediated drug-drug interaction between rilpivirine and abacavir; effect on intestinal absorption. Antimicrob. Agents Chemother. [Epub: Ahead of Print]

Riaz, H., B. Zheng, Y. Zheng, Z. Liu, H.M. Gu, M. Imran, T. Yaqoob, M.A. Bhinder, D.W. Zhang, and M.Y. Zahoor. (2024). The spectrum of novel ABCB11 gene variations in children with progressive familial intrahepatic cholestasis type 2 in Pakistani cohorts. Sci Rep 14: 18876.

Ricardo, S. and R. Lehmann. (2009). An ABC transporter controls export of a Drosophila germ cell attractant. Science 323: 943-946.

Richard-Fogal, C. and R.G. Kranz. (2010). The CcmC:heme:CcmE complex in heme trafficking and cytochrome c biosynthesis. J. Mol. Biol. 401: 350-362.

Richard-Fogal, C.L., E.R. Frawley, and R.G. Kranz. (2008). Topology and function of CcmD in cytochrome c maturation. J. Bacteriol. 190: 3489-3493.

Richardson, J.S. and I.J. Oresnik. (2007). L-Rhamnose transport is sugar kinase (RhaK) dependent in Rhizobium leguminosarum bv. trifolii. J. Bacteriol. 189: 8437-8446.

Richardson, J.S., M.F. Hynes, and I.J. Oresnik. (2004). A genetic locus necessary for rhamnose uptake and catabolism in Rhizobium leguminosarum bv. trifolii. J. Bacteriol. 186: 8433-8442.

Richet, E., A.L. Davidson, and N. Joly. (2012). The ABC transporter MalFGK(2) sequesters the MalT transcription factor at the membrane in the absence of cognate substrate. Mol. Microbiol. 85: 632-647.

Riedel, S., B. Siemiatkowska, M. Watanabe, C.S. Müller, V. Schünemann, R. Hoefgen, and S. Leimkühler. (2019). The ABCB7-Like Transporter PexA in Is Involved in the Translocation of Reactive Sulfur Species. Front Microbiol 10: 406.

Riordan, J.R. (2008). CFTR function and prospects for therapy. Annu. Rev. Biochem. 77: 701-726.

Ríos Colombo, N.S., M.C. Chalón, F.G. Dupuy, C.F. Gonzalez, and A. Bellomio. (2019). The case for class II bacteriocins: a biophysical approach using "suicide probes" in receptor-free hosts to study their mechanism of action. Biochimie. [Epub: Ahead of Print]

Ripperger, A., A. Krischer, D. Robaa, W. Sippl, and R.A. Benndorf. (2018). Pharmacogenetic Aspects of the Interaction of AT1 Receptor Antagonists With ATP-Binding Cassette Transporter. Front Pharmacol 9: 463.

Rismondo, J. and L.M. Schulz. (2021). Not Just Transporters: Alternative Functions of ABC Transporters in and. Microorganisms 9:.

Ritter, C.A., G. Jedlitschky, H. Meyer zu Schwabedissen, M. Grube, K. Köck, and H.K. Kroemer. (2005). Cellular export of drugs and signaling molecules by the ATP-binding cassette transporters MRP4 (ABCC4) and MRP5 (ABCC5). Drug Metab Rev 37: 253-278.

Rius, M., J. Hummel-Eisenbeiss, and D. Keppler. (2008). ATP-dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J Pharmacol Exp Ther 324: 86-94.

Rivera-Lugo, R., S. Huang, F. Lee, R. Méheust, A.T. Iavarone, A.M. Sidebottom, E. Oldfield, D.A. Portnoy, and S.H. Light. (2023). Distinct Energy-Coupling Factor Transporter Subunits Enable Flavin Acquisition and Extracytosolic Trafficking for Extracellular Electron Transfer in Listeria monocytogenes. mBio 14: e0308522.

Rocafull, M.A., F.J. Romero, L.E. Thomas, and J.R. del Castillo. (2011). Isolation and cloning of the K+-independent, ouabain-insensitive Na+-ATPase. Biochim. Biophys. Acta. 1808: 1684-1700.

Rockwell, N.C., H. Wolfger, K. Kuchler, and J. Thorner. (2009). ABC transporter Pdr10 regulates the membrane microenvironment of Pdr12 in Saccharomyces cerevisiae. J. Membr. Biol. 229: 27-52.

Rodionov, D.A., A.G. Vitreschak, A.A. Mironov, and M.S. Gelfand. (2002). Comparative genomics of thiamin biosynthesis in procaryotes. New genes and regulatory mechanisms. J. Biol. Chem. 277: 48949-48959.

Rodionov, D.A., A.G. Vitreschak, A.A. Mironov, and M.S. Gelfand. (2003). Comparative genomics of the vitamin B12 metabolism and regulation in prokaryotes. J. Biol. Chem. 278: 41148-41159.

Rodionov, D.A., C. Yang, X. Li, I.A. Rodionova, Y. Wang, A.Y. Obraztsova, O.P. Zagnitko, R. Overbeek, M.F. Romine, S. Reed, J.K. Fredrickson, K.H. Nealson, and A.L. Osterman. (2010). Genomic encyclopedia of sugar utilization pathways in the Shewanella genus. BMC Genomics 11: 494.

Rodionov, D.A., M.S. Gelfand, and N. Hugouvieux-Cotte-Pattat. (2004). Comparative genomics of the KdgR regulon in Erwinia chrysanthemi 3937 and other γ-proteobacteria. Microbiology 150: 3571-3590.

Rodionov, D.A., P. Hebbeln, A. Eudes, J. ter Beek, I.A. Rodionova, G.B. Erkens, D.J. Slotboom, M.S. Gelfand, A.L. Osterman, A.D. Hanson, and T. Eitinger. (2009). A novel class of modular transporters for vitamins in prokaryotes. J. Bacteriol. 191: 42-51.

Rodionov, D.A., P. Hebbeln, M.S. Gelfand, and T. Eitinger. (2006). Comparative and functional genomic analysis of prokaryotic nickel and cobalt uptake transporters: evidence for a novel group of ATP-binding cassette transporters. J. Bacteriol. 188: 317-327.

Rodionova, I.A., C. Yang, X. Li, O.V. Kurnasov, A.A. Best, A.L. Osterman, and D.A. Rodionov. (2012). Diversity and versatility of the Thermotoga maritima sugar kinome. J. Bacteriol. 194: 5552-5563.

Rodionova, I.A., D.A. Scott, N.V. Grishin, A.L. Osterman, and D.A. Rodionov. (2012). Tagaturonate-fructuronate epimerase UxaE, a novel enzyme in the hexuronate catabolic network in Thermotoga maritima. Environ Microbiol 14: 2920-2934.

Rodionova, I.A., S.A. Leyn, M.D. Burkart, N. Boucher, K.M. Noll, A.L. Osterman, and D.A. Rodionov. (2013). Novel inositol catabolic pathway in Thermotoga maritima. Environ Microbiol 15: 2254-2266.

Rodionova, I.A., X. Li, A.E. Plymale, K. Motamedchaboki, A.E. Konopka, M.F. Romine, J.K. Fredrickson, A.L. Osterman, and D.A. Rodionov. (2015). Genomic distribution of B-vitamin auxotrophy and uptake transporters in environmental bacteria from the Chloroflexi phylum. Environ Microbiol Rep 7: 204-210.

Rodriguez, G.M. and I. Smith. (2006). Identification of an ABC transporter required for iron acquisition and virulence in Mycobacterium tuberculosis. J. Bacteriol. 188: 424-430.

Rosenberg, M.F., A.B. Kamis, R. Callaghan, C.F. Higgins, and R.C. Ford. (2003). Three-dimensional structures of the mammalian multidrug resistance P-glycoprotein demonstrate major conformational changes in the transmembrane domains upon nucleotide binding. J. Biol. Chem. 278: 8294-8299.

Rosenberg, M.F., L.P. O'Ryan, G. Hughes, Z. Zhao, L.A. Aleksandrov, J.R. Riordan, and R.C. Ford. (2011). The Cystic Fibrosis Transmembrane Conductance Regulator (CFTR): THREE-DIMENSIONAL STRUCTURE AND LOCALIZATION OF A CHANNEL GATE. J. Biol. Chem. 286: 42647-42654.

Rosenberg, M.F., Z. Bikadi, E. Hazai, T. Starborg, L. Kelley, N.E. Chayen, R.C. Ford, and Q. Mao. (2015). Three-dimensional structure of the human breast cancer resistance protein (BCRP/ABCG2) in an inward-facing conformation. Acta Crystallogr D Biol Crystallogr 71: 1725-1735.

Roston, R.L., J. Gao, M.W. Murcha, J. Whelan, and C. Benning. (2012). TGD1, -2, and -3 proteins involved in lipid trafficking form ATP-binding cassette (ABC) transporter with multiple substrate-binding proteins. J. Biol. Chem. 287: 21406-21415.

Roth, J.R., J.G. Lawrence, M. Rubenfield, S. Kieffer-Higgins, and G.M. Church. (1993). Characterization of the cobalamin (vitamin B12) biosynthetic genes of Salmonella typhimurium. J. Bacteriol. 175: 3303-3316.

Roxo-Rosa, M., R. Jacinto, P. Sampaio, and S.S. Lopes. (2015). The zebrafish Kupffer''s vesicle as a model system for the molecular mechanisms by which the lack of Polycystin-2 leads to stimulation of CFTR. Biol Open 4: 1356-1366.

Rudolf, M., R. Tampé, and B. Joseph. (2023). Time-Resolved Mn2+-NO and NO-NO Distance Measurements Reveal That Catalytic Asymmetry Regulates Alternating Access  in an ABC Transporter. Angew Chem Int Ed Engl e202307091. [Epub: Ahead of Print]

Rufer E., Kagebein D., Leonhardt RM. and Knittler MR. (2015). Hydrophobic Interactions Are Key To Drive the Association of Tapasin with Peptide Transporter Subunit TAP2. J Immunol. 195(11):5482-94.

Ruknudin, A., D.H. Schulze, S.K. Sullivan, W.J. Lederer, and P.A. Welling. (1998). Novel subunit composition of a renal epithelial KATP channel. J. Biol. Chem. 273: 14165-14171.

Russell, R.R., J. Aduse-Opoku, I.C. Sutcliffe, L. Tao, and J.J. Ferretti. (1992). A binding protein-dependent transport system in Streptococcus mutans responsible for multiple sugar metabolism. J. Biol. Chem. 267: 4631-4637.

Russo, D.M., A. Williams, A. Edwards, D.M. Posadas, C. Finnie, M. Dankert, J.A. Downie, and A. Zorreguieta. (2006). Proteins exported via the PrsD-PrsE type I secretion system and the acidic exopolysaccharide are involved in biofilm formation by Rhizobium leguminosarum. J. Bacteriol. 188: 4474-4486.

Russo, G., S. Capitanio, M. Trasoletti, C. Morabito, P. Korwin Krukowski, I. Visentin, A. Genre, A. Schubert, and F. Cardinale. (2023). Strigolactones promote the localisation of the ABA exporter ABCG25 at the plasma membrane in root epidermal cells of Arabidopsis thaliana. J Exp Bot. [Epub: Ahead of Print]

Rutledge, R.M., L. Esser, J. Ma, and D. Xia. (2011). Toward understanding the mechanism of action of the yeast multidrug resistance transporter Pdr5p: a molecular modeling study. J Struct Biol 173: 333-344.

Ryndak, M.B., S. Wang, I. Smith, and G.M. Rodriguez. (2010). The Mycobacterium tuberculosis high-affinity iron importer, IrtA, contains an FAD-binding domain. J. Bacteriol. 192: 861-869.

Sabrialabe, S., J.G. Yang, E. Yariv, N. Ben-Tal, and O. Lewinson. (2020). Substrate recognition and ATPase activity of the E. coli cysteine/cystine ABC transporter YecSC-FliY. J. Biol. Chem. [Epub: Ahead of Print]

Sachrajda, I. and M. Ratajewski. (2011). Mithramycin A suppresses expression of the human melanoma-associated gene ABCB8. Mol. Genet. Genomics 285: 57-65.

Said, A.M., Y.E. Mansour, R.R. Soliman, R. Islam, and S.S. Fatahala. (2024). Design, synthesis, molecular modeling, in vitro and in vivo biological evaluation of potent anthranilamide derivatives as dual P-glycoprotein and CYP3A4 inhibitors. Eur J Med Chem 273: 116492.

Saier, M.H., Jr. (1994). Computer-aided analyses of transport protein sequences: gleaning evidence concerning function, structure, biogenesis, and evolution. Microbiol Rev 58: 71-93.

Saier, M.H., Jr. (2000). A functional-phylogenetic classification system for transmembrane solute transporters. Microbiol. Mol. Biol. Rev. 64: 354-411.

Saito, A., T. Fujii, T. Shinya, N. Shibuya, A. Ando, and K. Miyashita. (2008). The msiK gene, encoding the ATP-hydrolysing component of N,N'-diacetylchitobiose ABC transporters, is essential for induction of chitinase production in Streptomyces coelicolor A3(2). Microbiology 154: 3358-3365.

Sajid, A., S. Lusvarghi, M. Murakami, E.E. Chufan, B. Abel, M.M. Gottesman, S.R. Durell, and S.V. Ambudkar. (2020). Reversing the direction of drug transport mediated by the human multidrug transporter P-glycoprotein. Proc. Natl. Acad. Sci. USA 117: 29609-29617.

Sakai, K., M. Akiyama, Y. Sugiyama-Nakagiri, J.R. McMillan, D. Sawamura, and H. Shimizu. (2007). Localization of ABCA12 from Golgi apparatus to lamellar granules in human upper epidermal keratinocytes. Exp Dermatol 16: 920-926.

Sakamoto, M., H. Suzuki, and K. Yura. (2019). Relationship between conformation shift and disease related variation sites in ATP-binding cassette transporter proteins. Biophys Physicobiol 16: 68-79.

Saken, E., A. Rakin, and J. Heesemann. (2000). Molecular characterization of a novel siderophore-independent iron transport system in Yersinia. Int. J. Med. Microbiol. 290: 51-60.

Salgado, L.T., L.S. Oliveira, J. Echevarria-Lima, V.M. Reis, D.B. Sudatti, F.L. Thompson, and R.C. Pereira. (2023). Role of ABC Proteins in Secondary Metabolism and Immune (=Defensive) Response in Seaweeds. Cells 12:.

Salmon, R.C., M.J. Cliff, J.B. Rafferty, and D.J. Kelly. (2013). The CouPSTU and TarPQM transporters in Rhodopseudomonas palustris: redundant, promiscuous uptake systems for lignin-derived aromatic substrates. PLoS One 8: e59844.

Samuels, V., A.E. Mulelu, H. Ndlovu, and M.J. Marakalala. (2024). Mycobacterial FtsEX-RipC interaction is required for normal growth and cell morphology in rifampicin and low ionic strength conditions. Microbiol Spectr e0251523. [Epub: Ahead of Print]

San Francisco, B. and R.G. Kranz. (2014). Interaction of holoCcmE with CcmF in heme trafficking and cytochrome c biosynthesis. J. Mol. Biol. 426: 570-585.

San Paolo, S., J. Huang, S.N. Cohen, and C.J. Thompson. (2006). rag genes: novel components of the RamR regulon that trigger morphological differentiation in Streptomyces coelicolor. Mol. Microbiol. 61: 1167-1186.

Sarin, J., S. Aggarwal, R. Chaba, G.C. Varshney, and P.K. Chakraborti. (2001). B-subunit of phosphate-specific transporter from Mycobacterium tuberculosis is a thermostable ATPase. J. Biol. Chem. 276: 44590-44597.

Sarwar, S., V.M. Morozov, M.A. Newcomb, B. Yan, J.O. Brant, R. Opavsky, O.A. Guryanova, and A.M. Ishov. (2024). Overcoming ABCB1 mediated multidrug resistance in castration resistant prostate cancer. Cell Death Dis 15: 558.

Sasikumar, A.N. and T.G. Kinzy. (2014). Mutations in the chromodomain-like insertion of translation elongation factor 3 compromise protein synthesis through reduced ATPase activity. J. Biol. Chem. 289: 4853-4860.

Sass, V., U. Pag, A. Tossi, G. Bierbaum, and H.G. Sahl. (2008). Mode of action of human β-defensin 3 against Staphylococcus aureus and transcriptional analysis of responses to defensin challenge. Int. J. Med. Microbiol. 298: 619-633.

Saum, R., A. Mingote, H. Santos, and V. Müller. (2009). Genetic analysis of the role of the ABC transporter Ota and Otb in glycine betaine transport in Methanosarcina mazei Gö1. Arch. Microbiol. 191: 291-301.

Saurin, W. and E. Dassa. (1994). Sequence relationships between integral inner membrane proteins of binding protein-dependent transport systems: evolution by recurrent gene duplications. Protein. Sci. 3: 325-344.

Saurin, W., M. Hofnung, and E. Dassa. (1999). Getting in or out: early segregation between importers and exporters in the evolution of ATP-binding cassette (ABC) transporters. J. Mol. Evol. 48: 22-41.

Savková, K., S. Huszár, P. Baráth, Z. Pakanová, S. Kozmon, M. Vancová, M. Tesařová, J. Blaško, M. Kaliňák, V. Singh, J. Korduláková, and K. Mikušová. (2021). An ABC transporter Wzm-Wzt catalyzes translocation of lipid-linked galactan across the plasma membrane in mycobacteria. Proc. Natl. Acad. Sci. USA 118:.

Schirner, K., L.K. Stone, and S. Walker. (2011). ABC transporters required for export of wall teichoic acids do not discriminate between different main chain polymers. ACS Chem Biol 6: 407-412.

Schleker, E.S.M., S. Buschmann, H. Xie, S. Welsch, H. Michel, and C. Reinhart. (2022). Structural and functional investigation of ABC transporter STE6-2p from reveals unexpected interaction with sterol molecules. Proc. Natl. Acad. Sci. USA 119: e2202822119.

Schlösser, A., J. Jantos, K. Hackmann, and H. Schrempf. (1999). Characterization of the binding protein-dependent cellobiose and cellotriose transport system of the cellulose degrader Streptomyces reticuli. Appl. Environ. Microbiol. 65: 2636-2643.

Schlösser, A., T. Kampers, and H. Schrempf. (1997). The Streptomyces ATP-binding component MsiK assists in cellobiose and maltose transport. J. Bacteriol. 179: 2092-2095.

Schmidt, R.S., J.P. Macêdo, M.E. Steinmann, A.G. Salgado, P. Bütikofer, E. Sigel, D. Rentsch, and P. Mäser. (2018). Transporters of Trypanosoma brucei-phylogeny, physiology, pharmacology. FEBS J. 285: 1012-1023.

Schmidt, S., K. Pflüger, S. Kögl, R. Spanheimer, and V. Müller. (2007). The salt-induced ABC transporter Ota of the methanogenic archaeon Methanosarcina mazei Gö1 is a glycine betaine transporter. FEMS Microbiol. Lett. 277: 44-49.

Schmitt, L. and R. Tampé. (2002). Structure and mechanism of ABC transporters. Curr. Opin. Struct. Biol. 12: 754-760.

Schneider, E. and S. Hunke. (1998). ATP-binding-cassette (ABC) transport systems: functional and structural aspects of the ATP-hydrolyzing subunits/domains. FEMS Microbiol. Rev. 22: 1-20.

Schneider, E., V. Eckey, D. Weidlich, N. Wiesemann, A. Vahedi-Faridi, P. Thaben, and W. Saenger. (2012). Receptor-transporter interactions of canonical ATP-binding cassette import systems in prokaryotes. Eur J. Cell Biol. 91: 311-317.

Schrodt, S., J. Koch, and R. Tampé. (2006). Membrane topology of the transporter associated with antigen processing (TAP1) within an assembled functional peptide-loading complex. J. Biol. Chem. 281: 6455-6462.

Schuetzer-Muehlbauer, M., B. Willinger, G. Krapf, S. Enzinger, E. Presterl, and K. Kuchler. (2003). The Candida albicans Cdr2p ATP-binding cassette (ABC) transporter confers resistance to caspofungin. Mol. Microbiol. 48: 225-235.

Schulz, H., R.A. Fabianek, E.C. Pellicioli, H. Hennecke, and L. Thöny-Meyer. (1999). Heme transfer to the heme chaperone CcmE during cytochrome c maturation requires the CcmC protein, which may function independently of the ABC-transporter CcmAB. Proc. Natl. Acad. Sci. USA 96: 6462-6467.

Schulze-Gahmen, U., J. Pelaschier, H. Yokota, R. Kim, and S.H. Kim. (2003). Crystal structure of a hypothetical protein, TM841 of Thermotoga maritima, reveals its function as a fatty acid-binding protein. Proteins 50: 526-530.

Schwarz, F.M., J.D. Kuhlmann, J. Kämpfer, A. Klimova, D.M. Klotz, L. Freitag, P. Herrmann, V. Zinnow, J. Smith, T. Scheller, W. Walther, P. Wimberger, and U. Stein. (2024). Induction of circulating ABCB1 transcripts under platinum-based chemotherapy indicates poor prognosis and a bone micrometastatic phenotype in ovarian cancer patients. Mol Cancer 23: 174.

Scortecci, J.F., F.A. Garces, J.K. Mahto, L.L. Molday, F. Van Petegem, and R.S. Molday. (2024). Structural and Functional Characterization of the Nucleotide-binding Domains of ABCA4 and their Role in Stargardt Disease. J. Biol. Chem. 107666. [Epub: Ahead of Print]

Scortecci, J.F., L.L. Molday, S.B. Curtis, F.A. Garces, P. Panwar, F. Van Petegem, and R.S. Molday. (2021). Cryo-EM structures of the ABCA4 importer reveal mechanisms underlying substrate binding and Stargardt disease. Nat Commun 12: 5902.

Seelheim P., Wullner A. and Galla HJ. (2013). Substrate translocation and stimulated ATP hydrolysis of human ABC transporter MRP3 show positive cooperativity and are half-coupled. Biophys Chem. 171:31-7.

Seelig, A. and X. Li-Blatter. (2023). P-glycoprotein (ABCB1) - weak dipolar interactions provide the key to understanding allocrite recognition, binding, and transport. Cancer Drug Resist 6: 1-29.

Segrest, J.P., C. Tang, H.D. Song, M.K. Jones, W.S. Davidson, S.G. Aller, and J.W. Heinecke. (2022). ABCA1 is an extracellular phospholipid translocase. Nat Commun 13: 4812.

Seidler, U., A.K. Singh, A. Cinar, M. Chen, J. Hillesheim, B. Hogema, and B. Riederer. (2009). The role of the NHERF family of PDZ scaffolding proteins in the regulation of salt and water transport. Ann. N.Y. Acad. Sci. 1165: 249-260.

Sekowska, A., S. Robin, J.J. Daudin, A. Hénaut, and A. Danchin. (2001). Extracting biological information from DNA arrays: an unexpected link between arginine and methionine metabolism in Bacillus subtilis. Genome Biol 2: RESEARCH0019.

Sellers, Z.M., A.P. Naren, Y. Xiang, and P.M. Best. (2012). MRP4 and CFTR in the regulation of cAMP and β-adrenergic contraction in cardiac myocytes. Eur J Pharmacol 681: 80-87.

Serra, G., L. Memo, P. Cavicchioli, M. Cutrone, M. Giuffrè, M.L. La Torre, I.A.M. Schierz, and G. Corsello. (2022). Novel mutations of the ABCA12, KRT1 and ST14 genes in three unrelated newborns showing congenital ichthyosis. Ital J Pediatr 48: 145.

Severinov, K. and S.K. Nair. (2012). Microcin C: biosynthesis and mechanisms of bacterial resistance. Future Microbiol 7: 281-289.

Shah AH., Banerjee A., Rawal MK., Saxena AK., Mondal AK. and Prasad R. (2015). ABC transporter Cdr1p harbors charged residues in the intracellular loop and nucleotide-binding domain critical for protein trafficking and drug resistance. FEMS Yeast Res. 15(5).

Shah, A.H., M.K. Rawal, S. Dhamgaye, S.S. Komath, A.K. Saxena, and R. Prasad. (2015). Mutational Analysis of Intracellular Loops Identify Cross Talk with Nucleotide Binding Domains of Yeast ABC Transporter Cdr1p. Sci Rep 5: 11211.

Shah, B., J. Gray, I. Abraham, and M. Chang. (2022). Pharmacy considerations: Use of anti-CD38 monoclonal antibodies in relapsed and/or refractory multiple myeloma. J Oncol Pharm Pract 10781552221107850. [Epub: Ahead of Print]

Shah, P., D.G. Romero, and E. Swiatlo. (2008). Role of polyamine transport in Streptococcus pneumoniae response to physiological stress and murine septicemia. Microb. Pathog. 45: 167-172.

Shah, P., M. Marquart, L.R. Quin, and E. Swiatlo. (2006). Cellular location of polyamine transport protein PotD in Streptococcus pneumoniae. FEMS Microbiol. Lett. 261: 235-237.

Shahpouri, P., H. Mehralitabar, M. Kheirabadi, and S. Kazemi Noureini. (2023). Potential suppression of multidrug-resistance-associated protein 1 by coumarin derivatives: an insight from molecular docking and MD simulation studies. J Biomol Struct Dyn 1-17. [Epub: Ahead of Print]

Shani, N., P.A. Watkins, and D. Valle. (1995). PXA1, a possible Saccharomyces cerevisiae ortholog of the human adrenoleukodystrophy gene. Proc. Natl. Acad. Sci. USA 92: 6012-6016.

Shao, H., D. James, R.J. Lamont, and D.R. Demuth. (2007). Differential interaction of Aggregatibacter (Actinobacillus) actinomycetemcomitans LsrB and RbsB proteins with autoinducer 2. J. Bacteriol. 189: 5559-5565.

Sharma, K.G., D.L. Mason, G. Liu, P.A. Rea, A.K. Bachhawat, and S. Michaelis. (2002). Localization, regulation, and substrate transport properties of Bpt1p, a Saccharomyces cerevisiae MRP-type ABC transporter. Eukaryot. Cell. 1: 391-400.

Sharma, S., R. Zhou, L. Wan, S. Feng, K. Song, C. Xu, Y. Li, and M. Liao. (2021). Mechanism of LolCDE as a molecular extruder of bacterial triacylated lipoproteins. Nat Commun 12: 4687.

Shavva, V.S., A.V. Babina, E.V. Nekrasova, A.V. Lisunov, E.B. Dizhe, G.N. Oleinikova, and S.V. Orlov. (2022). Insulin Downregulates the Expression of ATP-binding Cassette Transporter A-I in Human Hepatoma Cell Line HepG2 in a FOXO1 and LXR Dependent Manner. Cell Biochem Biophys. [Epub: Ahead of Print]

Shen, X.M., K. Ohno, S.M. Sine, and A.G. Engel. (2005). Subunit-specific contribution to agonist binding and channel gating revealed by inherited mutation in muscle acetylcholine receptor M3-M4 linker. Brain 128: 345-355.

Shi, H., R. Chen, M. Li, and J. Ge. (2024). Acute hepatotoxicity of intravenous amiodarone in a Becker muscular dystrophy patient with decompensated heart failing and ABCB4 gene mutation: as assessed for causality using the updated RUCAM. J Cardiothorac Surg 19: 464.

Shi, J., H. Wang, K. Schellin, B. Li, M. Faller, J.M. Stoop, R.B. Meeley, D.S. Ertl, J.P. Ranch, and K. Glassman. (2007). Embryo-specific silencing of a transporter reduces phytic acid content of maize and soybean seeds. Nat Biotechnol 25: 930-937.

Shi, Z., A.K. Tiwari, S. Shukla, R.W. Robey, S. Singh, I.W. Kim, S.E. Bates, X. Peng, I. Abraham, S.V. Ambudkar, T.T. Talele, L.W. Fu, and Z.S. Chen. (2011). Sildenafil reverses ABCB1- and ABCG2-mediated chemotherapeutic drug resistance. Cancer Res 71: 3029-3041.

Shibata, Y., M. Ojika, A. Sugiyama, K. Yazaki, D.A. Jones, K. Kawakita, and D. Takemoto. (2016). The Full-Size ABCG Transporters Nb-ABCG1 and Nb-ABCG2 Function in Pre- and Postinvasion Defense against Phytophthora infestans in Nicotiana benthamiana. Plant Cell 28: 1163-1181.

Shimizu, Y., Y. Ogawa, K. Sugiura, J. Takeda, K. Sakai-Sawada, T. Yanagi, A. Kon, D. Sawamura, H. Shimizu, and M. Akiyama. (2014). A Palindromic Motif in the -2084 to -2078 Upstream Region is Essential for ABCA12 Promoter Function in Cultured Human Keratinocytes. Sci Rep 4: 6737.

Shin, J.H., A.K. Singh, D.J. Cheon, and J.H. Roe. (2011). Activation of the SoxR regulon in Streptomyces coelicolor by the extracellular form of the pigmented antibiotic actinorhodin. J. Bacteriol. 193: 75-81.

Shin, S.K., H.S. Park, H.J. Kwon, H.J. Yoon, and J.W. Suh. (2007). Genetic characterization of two S-adenosylmethionine-induced ABC transporters reveals their roles in modulations of secondary metabolism and sporulation in Streptomyces coelicolor M145. J Microbiol Biotechnol 17: 1818-1825.

Shintre, C.A., A.C. Pike, Q. Li, J.I. Kim, A.J. Barr, S. Goubin, L. Shrestha, J. Yang, G. Berridge, J. Ross, P.J. Stansfeld, M.S. Sansom, A.M. Edwards, C. Bountra, B.D. Marsden, F. von Delft, A.N. Bullock, O. Gileadi, N.A. Burgess-Brown, and E.P. Carpenter. (2013). Structures of ABCB10, a human ATP-binding cassette transporter in apo- and nucleotide-bound states. Proc. Natl. Acad. Sci. USA 110: 9710-9715.

Shitan, N., I. Bazin, K. Dan, K. Obata, K. Kigawa, K. Ueda, F. Sato, C. Forestier, and K. Yazaki. (2003). Involvement of CjMDR1, a plant multidrug-resistance-type ATP-binding cassette protein, in alkaloid transport in Coptis japonica. Proc. Natl. Acad. Sci. USA 100: 751-756.

Shukla, S., S.V. Ambudkar, and R. Prasad. (2004). Substitution of threonine-1351 in the multidrug transporter Cdr1p of Candida albicans results in hypersusceptibility to antifungal agents and threonine-1351 is essential for synergic effects of calcineurin inhibitor FK520. J Antimicrob Chemother 54: 38-45.

Shukla, S., V. Rai, P. Saini, D. Banerjee, A.K. Menon, and R. Prasad. (2007). Candida drug resistance protein 1, a major multidrug ATP binding cassette transporter of Candida albicans, translocates fluorescent phospholipids in a reconstituted system. Biochemistry 46: 12081-12090.

Shulami, S., O. Gat, A.L. Sonenshein, and Y. Shoham. (1999). The glucuronic acid utilization gene cluster from Bacillus stearothermophilus T-6. J. Bacteriol. 181: 3695-3704.

Siche, S., O. Neubauer, P. Hebbeln, and T. Eitinger. (2010). A bipartite S unit of an ECF-type cobalt transporter. Res. Microbiol. 161: 824-829.

Sidler, M., P. Hassa, S. Hasan, C. Ringli, and R. Dudler. (1998). Involvement of an ABC transporter in a developmental pathway regulating hypocotyl cell elongation in the light. Plant Cell 10: 1623-1636.

Silva, Z., M.M. Sampaio, A. Henne, A. Böhm, R. Gutzat, W. Boos, M.S. da Costa, and H. Santos. (2005). The high-affinity maltose/trehalose ABC transporter in the extremely thermophilic bacterium Thermus thermophilus HB27 also recognizes sucrose and palatinose. J. Bacteriol. 187: 1210-1218.

Simcoe, M.J., G. Arno, P.G. Hysi, T. Ko, M. Michaelides, C.J. Hammond, P.J. Patel, O.A. Mahroo, A.R. Webster, and. (2023). An Analysis of the Effect of ABCA4 p.Asn1868Ile Genotypes on Retinal Structure in 26,558 Participants in the UK Biobank. Invest Ophthalmol Vis Sci 64: 31.

Singh, A., S.K. Patel, P. Kumar, K.C. Das, D. Verma, R. Sharma, T. Tripathi, R. Giri, N. Martins, and N. Garg. (2022). Quercetin acts as a P-gp modulator via impeding signal transduction from nucleotide-binding domain to transmembrane domain. J Biomol Struct Dyn 40: 4507-4515.

Singh, B. and K.H. Röhm. (2008). A new subfamily of bacterial glutamate/aspartate receptors. Biol Chem 389: 33-36.

Singh, B. and K.H. Röhm. (2008). Characterization of a Pseudomonas putida ABC transporter (AatJMQP) required for acidic amino acid uptake: biochemical properties and regulation by the Aau two-component system. Microbiology 154: 797-809.

Singh, H., S. Velamakanni, M.J. Deery, J. Howard, S.L. Wei, and H.W. van Veen. (2016). ATP-dependent substrate transport by the ABC transporter MsbA is proton-coupled. Nat Commun 7: 12387.

Singh, P., V.M. Katoch, K.K. Mohanty, and D.S. Chauhan. (2016). Analysis of expression profile of mce operon genes (mce1, mce2, mce3 operon) in different Mycobacterium tuberculosis isolates at different growth phases. Indian J Med Res 143: 487-494.

Situ, D., A. Haimeur, G. Conseil, K.E. Sparks, D. Zhang, R.G. Deeley, and S.P. Cole. (2004). Mutational analysis of ionizable residues proximal to the cytoplasmic interface of membrane spanning domain 3 of the multidrug resistance protein, MRP1 (ABCC1): glutamate 1204 is important for both the expression and catalytic activity of the transporter. J. Biol. Chem. 279: 38871-38880.

Sjöstedt, N., J.J. van den Heuvel, J.B. Koenderink, and H. Kidron. (2017). Transmembrane Domain Single-Nucleotide Polymorphisms Impair Expression and Transport Activity of ABC Transporter ABCG2. Pharm Res. [Epub: Ahead of Print]

Sleator, R.D., H.H. Wemekamp-Kamphuis, C.G. Gahan, T. Abee, and C. Hill. (2005). A PrfA-regulated bile exclusion system (BilE) is a novel virulence factor in Listeria monocytogenes. Mol. Microbiol. 55: 1183-1195.

Slotboom, D.J. (2014). Structural and mechanistic insights into prokaryotic energy-coupling factor transporters. Nat. Rev. Microbiol. 12: 79-87.

Smart, J.P., M.J. Cliff, and D.J. Kelly. (2009). A role for tungsten in the biology of Campylobacter jejuni: tungstate stimulates formate dehydrogenase activity and is transported via an ultra-high affinity ABC system distinct from the molybdate transporter. Mol. Microbiol. 74: 742-757.

Smith, A.J., A. van Helvoort, G. van Meer, K. Szabo, E. Welker, G. Szakacs, A. Varadi, B. Sarkadi, and P. Borst. (2000). MDR3 P-glycoprotein, a phosphatidylcholine translocase, transports several cytotoxic drugs and directly interacts with drugs as judged by interference with nucleotide trapping. J. Biol. Chem. 275: 23530-23539.

Smith, T.J., H. Sondermann, and G.A. O''Toole. (2018). Type 1 Does the Two-Step: Type 1 Secretion Substrates with a Functional Periplasmic Intermediate. J. Bacteriol. 200:.

Smriti, , P. Zou, and H.S. McHaourab. (2009). Mapping Daunorubicin-binding Sites in the ATP-binding Cassette Transporter MsbA Using Site-specific Quenching by Spin Labels. J. Biol. Chem. 284: 13904-13913.

Sodani K., Patel A., Anreddy N., Singh S., Yang DH., Kathawala RJ., Kumar P., Talele TT. and Chen ZS. (2014). Telatinib reverses chemotherapeutic multidrug resistance mediated by ABCG2 efflux transporter in vitro and in vivo. Biochem Pharmacol. 89(1):52-61.

Sodani, K., A.K. Tiwari, S. Singh, A. Patel, Z.J. Xiao, J.J. Chen, Y.L. Sun, T.T. Talele, and Z.S. Chen. (2012). GW583340 and GW2974, human EGFR and HER-2 inhibitors, reverse ABCG2- and ABCB1-mediated drug resistance. Biochem Pharmacol 83: 1613-1622.

Sofianos, G., E. Piombo, M. Dubey, M. Karlsson, G. Karaoglanidis, and G. Tzelepis. (2024). Transcriptomic and functional analyses on a Botrytis cinerea multidrug-resistant (MDR) strain provides new insights into the potential molecular mechanisms of MDR and fitness. Mol Plant Pathol 25: e70004.

Soichi, O., N. Masanori, T. Hideo, A. Kazunori, I. Nobuya, and K. Jun-ichi. (2007). Clinical significance of ABCA2'' a possible molecular marker for oligodendrogliomas. Neurosurgery 60: 707-14; discussion 714.

Solbiati, J.O., M. Ciaccio, R.N. Farías, J.E. González-Pastor, F. Moreno, and R.A. Salomón. (1999). Sequence analysis of the four plasmid genes required to produce the circular peptide antibiotic microcin J25. J. Bacteriol. 181: 2659-2662.

Song, G., S. Zhang, M. Tian, L. Zhang, R. Guo, W. Zhuo, and M. Yang. (2021). Molecular insights into the human ABCB6 transporter. Cell Discov 7: 55.

Soriano, E.V., K.R. Rajashankar, J.W. Hanes, S. Bale, T.P. Begley, and S.E. Ealick. (2008). Structural similarities between thiamin-binding protein and thiaminase-I suggest a common ancestor. Biochemistry 47: 1346-1357.

Sperandio, B., C. Gautier, S. McGovern, D.S. Ehrlich, P. Renault, I. Martin-Verstraete, and E. Guédon. (2007). Control of methionine synthesis and uptake by MetR and homocysteine in Streptococcus mutans. J. Bacteriol. 189: 7032-7044.

Spille, D.C., E.C. Bunk, C. Thomas, Z. Özdemir, A. Wagner, B.H. Akkurt, M. Mannil, W. Paulus, O.M. Grauer, W. Stummer, V. Senner, and B. Brokinkel. (2023). Protoporphyrin IX (PpIX) Fluorescence during Meningioma Surgery: Correlations with Histological Findings and Expression of Heme Pathway Molecules. Cancers (Basel) 15:.

Srikant, S., R. Gaudet, and A.W. Murray. (2020). Selecting for Altered Substrate Specificity Reveals the Evolutionary Flexibility of ATP-Binding Cassette Transporters. Curr. Biol. [Epub: Ahead of Print]

Srinivasan, V., A.J. Pierik, and R. Lill. (2014). Crystal structures of nucleotide-free and glutathione-bound mitochondrial ABC transporter Atm1. Science 343: 1137-1140.

Staron, P., K. Forchhammer, and I. Maldener. (2011). Novel ATP-driven pathway of glycolipid export involving TolC protein. J. Biol. Chem. 286: 38202-38210.

Stauff, D.L., D. Bagaley, V.J. Torres, R. Joyce, K.L. Anderson, L. Kuechenmeister, P.M. Dunman, and E.P. Skaar. (2008). Staphylococcus aureus HrtA is an ATPase required for protection against heme toxicity and prevention of a transcriptional heme stress response. J. Bacteriol. 190: 3588-3596.

Stefanato, F.L., E. Abou-Mansour, A. Buchala, M. Kretschmer, A. Mosbach, M. Hahn, C.G. Bochet, J.P. Métraux, and H.J. Schoonbeek. (2009). The ABC transporter BcatrB from Botrytis cinerea exports camalexin and is a virulence factor on Arabidopsis thaliana. Plant J. 58: 499-510.

Stein, M., J. Dittgen, C. Sánchez-Rodríguez, B.H. Hou, A. Molina, P. Schulze-Lefert, V. Lipka, and S. Somerville. (2006). Arabidopsis PEN3/PDR8, an ATP binding cassette transporter, contributes to nonhost resistance to inappropriate pathogens that enter by direct penetration. Plant Cell 18: 731-746.

Stepler, K.E., T.R. Gillyard, C.B. Reed, T.M. Avery, J.S. Davis, and R.A.S. Robinson. (2022). ABCA7, a Genetic Risk Factor Associated with Alzheimer''s Disease Risk in African Americans. J Alzheimers Dis. [Epub: Ahead of Print]

Stewart, J.B. and M.A. Hermodson. (2003). Topology of RbsC, the membrane component of the Escherichia coli ribose transporter. J. Bacteriol. 185: 5234-5239.

Stindt, J., P. Ellinger, K. Weissenberger, C. Dröge, D. Herebian, E. Mayatepek, B. Homey, S. Braun, J. Schulte am Esch, M. Horacek, A. Canbay, L. Schmitt, D. Häussinger, and R. Kubitz. (2013). A novel mutation within a transmembrane helix of the bile salt export pump (BSEP, ABCB11) with delayed development of cirrhosis. Liver Int 33: 1527-1535.

Stinson, M.W., M.A. Cohen, and J.M. Merrick. (1977). Purification and properties of the periplasmic glucose-binding protein of Pseudomonas aeruginosa. J. Bacteriol. 131: 672-681.

Stockner, T., R. Gradisch, and L. Schmitt. (2020). The role of the degenerate nucleotide binding site in type I ABC exporters. FEBS Lett. [Epub: Ahead of Print]

Strachan, L.R., T.J. Stevenson, B. Freshner, M.D. Keefe, D. Miranda Bowles, and J.L. Bonkowsky. (2017). A zebrafish model of X-linked adrenoleukodystrophy recapitulates key disease features and demonstrates a developmental requirement for abcd1 in oligodendrocyte patterning and myelination. Hum Mol Genet 26: 3600-3614.

Strouse, J.J., I. Ivnitski-Steele, A. Waller, S.M. Young, D. Perez, A.M. Evangelisti, O. Ursu, C.G. Bologa, M.B. Carter, V.M. Salas, G. Tegos, R.S. Larson, T.I. Oprea, B.S. Edwards, and L.A. Sklar. (2013). Fluorescent substrates for flow cytometric evaluation of efflux inhibition in ABCB1, ABCC1, and ABCG2 transporters. Anal Biochem 437: 77-87.

Stubbendieck, R.M. and P.D. Straight. (2017). Linearmycins activate a two-component signaling system involved in bacterial competition and biofilm morphology. J. Bacteriol. [Epub: Ahead of Print]

Stukkens, Y., A. Bultreys, S. Grec, T. Trombik, D. Vanham, and M. Boutry. (2005). NpPDR1, a pleiotropic drug resistance-type ATP-binding cassette transporter from Nicotiana plumbaginifolia, plays a major role in plant pathogen defense. Plant Physiol. 139: 341-352.

Stumpe, S. and E.P. Bakker. Requirement of a large K+-uptake capacity and of extracytoplasmic protease activity for protamine resistance of Escherichia coli. Arch. Microbiol. 167: 126-136.

Stumpp, T., S. Himbert, and J. Altenbuchner. (2005). Cloning of the netropsin resistance genes from Streptomyces flavopersicus NRRL 2820. J Basic Microbiol 45: 355-362.

Sugden, C., S. Ross, G. Bloomfield, A. Ivens, J. Skelton, A. Mueller-Taubenberger, and J.G. Williams. (2010). Two novel Src homology 2 domain proteins interact to regulate dictyostelium gene expression during growth and early development. J. Biol. Chem. 285: 22927-22935.

Sumitani, M., D.S. Yamamoto, J.M. Lee, and M. Hatakeyama. (2005). Isolation of white gene orthologue of the sawfly, Athalia rosae (Hymenoptera) and its functional analysis using RNA interference. Insect Biochem Mol Biol 35: 231-240.

Sun YL., Kathawala RJ., Singh S., Zheng K., Talele TT., Jiang WQ. and Chen ZS. (2012). Zafirlukast antagonizes ATP-binding cassette subfamily G member 2-mediated multidrug resistance. Anticancer Drugs. 23(8):865-73.

Sun, B.Z. and G.S. Sawicki. (2024). Advances in Care and Outcomes for Children with Cystic Fibrosis. Clin Chest Med 45: 625-637.

Sun, D. (2018). Pull in and Push Out: Mechanisms of Horizontal Gene Transfer in Bacteria. Front Microbiol 9: 2154.

Sun, H. (2012). Membrane receptors and transporters involved in the function and transport of vitamin A and its derivatives. Biochim. Biophys. Acta. 1821: 99-112.

Sun, N., Y.F. Xie, Y. Wu, N. Guo, D.H. Li, and J.S. Gao. (2021). Genome-wide identification of ABCC gene family and their expression analysis in pigment deposition of fiber in brown cotton (Gossypium hirsutum). PLoS One 16: e0246649.

Sun, S., Y. Gao, X. Yang, X. Yang, T. Hu, J. Liang, Z. Xiong, Y. Ran, P. Ren, F. Bai, L.W. Guddat, H. Yang, Z. Rao, and B. Zhang. (2023). Cryo-EM structures for the Mycobacterium tuberculosis iron-loaded siderophore transporter IrtAB. Protein Cell 14: 448-458.

Sun, X., R. Ge, D. Zhang, H. Sun, and Q.Y. He. (2010). Iron-containing lipoprotein SiaA in SiaABC, the primary heme transporter of Streptococcus pyogenes. J Biol Inorg Chem 15: 1265-1273.

Sun, Y., J. Wang, T. Long, X. Qi, L. Donnelly, N. Elghobashi-Meinhardt, L. Esparza, J.C. Cohen, X.S. Xie, H.H. Hobbs, and X. Li. (2021). Molecular basis of cholesterol efflux via ABCG subfamily transporters. Proc. Natl. Acad. Sci. USA 118:.

Sun, Z., Y. Chen, C. Yang, S. Yang, Y. Gu, and W. Jiang. (2015). A novel three-component system-based regulatory model for d-xylose sensing and transport in Clostridium beijerinckii. Mol. Microbiol. 95: 576-589.

Sunidhi, S., S. Sacher, Atul, P. Garg, and A. Ray. (2021). Elucidating the Structural Features of ABCA1 in its Heterogeneous Membrane Environment. Front Mol Biosci 8: 803078.

Suzuki, H., T. Koyanagi, S. Izuka, A. Onishi, and H. Kumagai. (2005). The yliA, -B, -C, and -D genes of Escherichia coli K-12 encode a novel glutathione importer with an ATP-binding cassette. J. Bacteriol. 187: 5861-5867.

Suzuki, R., J. Wada, T. Katayama, S. Fushinobu, T. Wakagi, H. Shoun, H. Sugimoto, A. Tanaka, H. Kumagai, H. Ashida, M. Kitaoka, and K. Yamamoto. (2008). Structural and thermodynamic analyses of solute-binding Protein from Bifidobacterium longum specific for core 1 disaccharide and lacto-N-biose I. J. Biol. Chem. 283: 13165-13173.

Suzuki, T., T. Murai, I. Fukuda, T. Tobe, M. Yoshikawa, and C. Sasakawa. (1994). Identification and characterization of a chromosomal virulence gene, vacJ, required for intercellular spreading of Shigella flexneri. Mol. Microbiol. 11: 31-41.

Swain, K., I. Casabon, L.D. Eltis, and W.W. Mohn. (2012). Two transporters essential for reassimilation of novel cholate metabolites by Rhodococcus jostii RHA1. J. Bacteriol. 194: 6720-6727.

Swapnil, P., M. Meena, and A.K. Rai. (2021). Molecular interaction of nitrate transporter proteins with recombinant glycinebetaine results in efficient nitrate uptake in the cyanobacterium Anabaena PCC 7120. PLoS One 16: e0257870.

Swartz, D.J., A. Singh, N. Sok, J.N. Thomas, J. Weber, and I.L. Urbatsch. (2020). Replacing the eleven native tryptophans by directed evolution produces an active P-glycoprotein with site-specific, non-conservative substitutions. Sci Rep 10: 3224.

Swoboda, J.G., T.C. Meredith, J. Campbell, S. Brown, T. Suzuki, T. Bollenbach, A.J. Malhowski, R. Kishony, M.S. Gilmore, and S. Walker. (2009). Discovery of a small molecule that blocks wall teichoic acid biosynthesis in Staphylococcus aureus. ACS Chem Biol 4: 875-883.

Szöllősi, D., D. Rose-Sperling, U.A. Hellmich, and T. Stockner. (2017). Comparison of mechanistic transport cycle models of ABC exporters. Biochim. Biophys. Acta. [Epub: Ahead of Print]

Szöllősi, D., P. Chiba, G. Szakacs, and T. Stockner. (2020). Conversion of chemical to mechanical energy by the nucleotide binding domains of ABCB1. Sci Rep 10: 2589.

Taank, Y., V. Randhawa, and N. Agnihotri. (2024). Ergosterol and its metabolites as agonists of Liver X receptor and their anticancer potential in colorectal cancer. J Steroid Biochem Mol Biol 243: 106572.

Taga, M.E., J.L. Semmelhack, and B.L. Bassler. (2001). The LuxS-dependent autoinducer AI-2 controls the expression of an ABC transporter that functions in AI-2 uptake in Salmonella typhimurium. Mol. Microbiol. 42: 777-793.

Taga, M.E., S.T. Miller, and B.L. Bassler. (2003). Lsr-mediated transport and processing of AI-2 in Salmonella typhimurium. Mol. Microbiol. 50: 1411-1427.

Tagirasa, R., K. Rana, H. Kaza, D. Parai, S. Damera, and S. Basu. (2020). Role of Multidrug Resistance Proteins in Nonresponders to Immunomodulatory Therapy for Noninfectious Uveitis. Transl Vis Sci Technol 9: 12.

Takeno, M., H. Taguchi, and T. Akamatsu. (2011). Essential involvement of the Bacillus subtilis ABC transporter, EcsB, in genetic transformation of purified DNA but not native DNA from protoplast lysates. J Biosci Bioeng 112: 209-214.

Takeuchi, M., T. Kegasa, A. Watanabe, M. Tamura, and Y. Tsutsumi. (2018). Expression analysis of transporter genes for screening candidate monolignol transporters using Arabidopsis thaliana cell suspensions during tracheary element differentiation. J Plant Res 131: 297-305.

Tam, R. and M.H. Saier, Jr. (1993). Structural, functional, and evolutionary relationships among extracellular solute-binding receptors of bacteria. Microbiol Rev 57: 320-346.

Tamura, K., H. Sugimoto, Y. Shiro, and Y. Sugita. (2019). Chemo-Mechanical Coupling in the Transport Cycle of a Heme ABC Transporter. J Phys Chem B. [Epub: Ahead of Print]

Tanabe, K., E. Lamping, M. Nagi, A. Okawada, A.R. Holmes, Y. Miyazaki, R.D. Cannon, B.C. Monk, and M. Niimi. (2011). Chimeras of Candida albicans Cdr1p and Cdr2p reveal features of pleiotropic drug resistance transporter structure and function. Mol. Microbiol. 82: 416-433.

Tanabe, K., M. Bonus, S. Tomiyama, K. Miyoshi, M. Nagi, K. Niimi, A. Chindamporn, H. Gohlke, L. Schmitt, R.D. Cannon, M. Niimi, and E. Lamping. (2018). FK506 resistance of Pdr5 and Cdr1 involves mutations in the transmembrane domains and extracellular loops. Antimicrob. Agents Chemother. [Epub: Ahead of Print]

Tanabe, T., T. Funahashi, H. Nakao, S. Miyoshi, S. Shinoda, and S. Yamamoto. (2003). Identification and characterization of genes required for biosynthesis and transport of the siderophore vibrioferrin in Vibrio parahaemolyticus. J. Bacteriol. 185: 6938-6949.

Tang, C. and J.F. Oram. (2009). The cell cholesterol exporter ABCA1 as a protector from cardiovascular disease and diabetes. Biochim. Biophys. Acta. 1791: 563-572.

Tang, Q., M. Sinclair, H. Hasdemir, R. Stein, E. Karakas, E. Tajkhorshid, and H. Mchaourab. (2023). Asymmetric conformations and lipid interactions shape the ATP-coupled cycle of a heterodimeric ABC transporter. bioRxiv.

Taniguchi, N. and H. Tokuda. (2008). Molecular events involved in a single cycle of ligand transfer from an ATP binding cassette transporter, LolCDE, to a molecular chaperone, LolA. J. Biol. Chem. 283: 8538-8544.

Tanjala, A.C., J.X. Jiang, P.D.W. Eckford, M. Ramjeesingh, C. Li, L.J. Huan, G. Langeveld, C. Townsend, D.V. Paone, J. Busch-Petersen, R. Pekhletski, L. Tang, V. Raju, S.M. Rowe, and C.E. Bear. (2024). Comparison of a novel potentiator of CFTR channel activity to ivacaftor in ameliorating mucostasis caused by cigarette smoke in primary human bronchial airway epithelial cells. Respir Res 25: 269.

Tanji, T., H. Shiraishi, K. Nishikori, R. Aoyama, K. Ohashi, M. Maeda, and A. Ohashi-Kobayashi. (2017). Molecular dissection of Caenorhabditis elegans ATP-binding cassette transporter protein HAF-4 to investigate its subcellular localization and dimerization. Biochem. Biophys. Res. Commun. [Epub: Ahead of Print]

Tanji, T., K. Nishikori, H. Shiraishi, M. Maeda, and A. Ohashi-Kobayashi. (2013). Co-operative function and mutual stabilization of the half ATP-binding cassette transporters HAF-4 and HAF-9 in Caenorhabditis elegans. Biochem. J. 452: 467-475.

Tanji, T., K. Nishikori, S. Haga, Y. Kanno, Y. Kobayashi, M. Takaya, K. Gengyo-Ando, S. Mitani, H. Shiraishi, and A. Ohashi-Kobayashi. (2016). Characterization of HAF-4- and HAF-9-localizing organelles as distinct organelles in Caenorhabditis elegans intestinal cells. BMC Cell Biol 17: 4.

Tarasova, N.I., R. Seth, S.G. Tarasov, T. Kosakowska-Cholody, C.A. Hrycyna, M.M. Gottesman, and C.J. Michejda. (2005). Transmembrane inhibitors of P-glycoprotein, an ABC transporter. J Med Chem 48: 3768-3775.

Tarling, E.J. and P.A. Edwards. (2011). ATP binding cassette transporter G1 (ABCG1) is an intracellular sterol transporter. Proc. Natl. Acad. Sci. USA 108: 19719-19724.

Tassis, K., R. Vietrov, M. de Koning, M. de Boer, G. Gouridis, and T. Cordes. (2020). Single-molecule studies of conformational states and dynamics in the ABC importer OpuA. FEBS Lett. [Epub: Ahead of Print]

Tauch, A., S. Krieft, A. Pühler, and J. Kalinowski. (1999). The tetAB genes of the Corynebacterium striatum R-plasmid pTP10 encode an ABC transporter and confer tetracycline, oxytetracycline and oxacillin resistance in Corynebacterium glutamicum. FEMS Microbiol. Lett. 173: 203-209.

Taylor, N.M.I., I. Manolaridis, S.M. Jackson, J. Kowal, H. Stahlberg, and K.P. Locher. (2017). Structure of the human multidrug transporter ABCG2. Nature 546: 504-509.

Tearle, R.G., J.M. Belote, M. McKeown, B.S. Baker, and A.J. Howells. (1989). Cloning and characterization of the scarlet gene of Drosophila melanogaster. Genetics 122: 595-606.

Teichmann, L., H. Kümmel, B. Warmbold, and E. Bremer. (2018). OpuF, a New Bacillus Compatible Solute ABC Transporter with a Substrate-Binding Protein Fused to the Transmembrane Domain. Appl. Environ. Microbiol. 84:.

Temesszentandrási-Ambrus, C., G. Nagy, A. Bui, and Z. Gáborik. (2023). A Unique In Vitro Assay to Investigate ABCB4 Transport Function. Int J Mol Sci 24:.

Teng, M.S., K.H. Yeh, L.A. Hsu, H.H. Chou, L.K. Er, S. Wu, and Y.L. Ko. (2023). Differential Effects of Gene Region Variants on Lipid Profile, Blood Pressure Status, and Gallstone Disease History in Taiwan. Genes (Basel) 14:.

ter Beek, J., R.H. Duurkens, G.B. Erkens, and D.J. Slotboom. (2011). Quaternary structure and functional unit of energy coupling factor (ECF)-type transporters. J. Biol. Chem. 286: 5471-5475.

Terao, R., B.S. Sohn, T. Yamamoto, T.J. Lee, J. Colasanti, C.W. Pfeifer, J.B. Lin, A. Santeford, S. Yamaguchi, M. Yoshida, and R.S. Apte. (2024). Cholesterol Accumulation Promotes Photoreceptor Senescence and Retinal Degeneration. Invest Ophthalmol Vis Sci 65: 29.

Terasaka, K., J.J. Blakeslee, B. Titapiwatanakun, W.A. Peer, A. Bandyopadhyay, S.N. Makam, O.R. Lee, E.L. Richards, A.S. Murphy, F. Sato, and K. Yazaki. (2005). PGP4, an ATP binding cassette P-glycoprotein, catalyzes auxin transport in Arabidopsis thaliana roots. Plant Cell 17: 2922-2939.

Terlizzi, V., E. Pesce, V. Capurro, V. Tomati, M. Lena, C. Pastorino, R. Bocciardi, F. Zara, C. Centrone, G. Taccetti, C. Castellani, and N. Pedemonte. (2023). Clinical Consequences and Functional Impact of the Rare S737F CFTR Variant and Its Responsiveness to CFTR Modulators. Int J Mol Sci 24:.

Teschner, J., N. Lachmann, J. Schulze, M. Geisler, K. Selbach, J. Santamaria-Araujo, J. Balk, R.R. Mendel, and F. Bittner. (2010). A novel role for Arabidopsis mitochondrial ABC transporter ATM3 in molybdenum cofactor biosynthesis. Plant Cell 22: 468-480.

Thöny-Meyer, L. (1997). Biogenesis of respiratory cytochromes in bacteria. Microbiol. Mol. Biol. Rev. 61: 337-376.

Thaker, T.M., S. Mishra, W. Zhou, M. Mohan, Q. Tang, J.D. Faraldo-Goméz, H.S. Mchaourab, and T.M. Tomasiak. (2021). Asymmetric drug binding in an ATP-loaded inward-facing state of an ABC transporter. Nat Chem Biol. [Epub: Ahead of Print]

Thangapandian, S., K. Kapoor, and E. Tajkhorshid. (2020). Probing cholesterol binding and translocation in P-glycoprotein. Biochim. Biophys. Acta. Biomembr 1862: 183090.

Thélot, F.A., W. Zhang, K. Song, C. Xu, J. Huang, and M. Liao. (2021). Distinct allosteric mechanisms of first-generation MsbA inhibitors. Science 374: 580-585.

Thomas, C., S.G. Aller, K. Beis, E.P. Carpenter, G. Chang, L. Chen, E. Dassa, M. Dean, F. Duong Van Hoa, D. Ekiert, R. Ford, R. Gaudet, X. Gong, I.B. Holland, Y. Huang, D.K. Kahne, H. Kato, V. Koronakis, C.M. Koth, Y. Lee, O. Lewinson, R. Lill, E. Martinoia, S. Murakami, H.W. Pinkett, B. Poolman, D. Rosenbaum, B. Sarkadi, L. Schmitt, E. Schneider, Y. Shi, S.L. Shyng, D.J. Slotboom, E. Tajkhorshid, D.P. Tieleman, K. Ueda, A. Váradi, P.C. Wen, N. Yan, P. Zhang, H. Zheng, J. Zimmer, and R. Tampé. (2020). Structural and functional diversity calls for a new classification of ABC transporters. FEBS Lett. 594: 3767-3775.

Thomas, G.H. (2010). Homes for the orphans: utilization of multiple substrate-binding proteins by ABC transporters. Mol. Microbiol. 75: 6-9.

Thompson, C.C., S.S. Nicod, D.S. Malcolm, S.S. Grieshaber, and R.A. Carabeo. (2012). Cleavage of a putative metal permease in Chlamydia trachomatis yields an iron-dependent transcriptional repressor. Proc. Natl. Acad. Sci. USA 109: 10546-10551.

Thompson, S.A., O.L. Shedd, K.C. Ray, M.H. Beins, J.P. Jorgensen, and M.J. Blaser. (1998). Campylobacter fetus surface layer proteins are transported by a type I secretion system. J. Bacteriol. 180: 6450-6458.

Thong, S., B. Ercan, F. Torta, Z.Y. Fong, H.Y. Wong, M.R. Wenk, and S.S. Chng. (2016). Defining key roles for auxiliary proteins in an ABC transporter that maintains bacterial outer membrane lipid asymmetry. Elife 5:.

Tian, M., F. Zhou, Z. Teng, C. Wang, X. Zhang, Y. Wang, and Y. Li. (2021). Curcumin ameliorates lipid metabolic disorder and cognitive dysfunction via the ABCA1 transmembrane transport system in APP/PS1 double transgenic mice. J Integr Neurosci 20: 895-903.

Tian, M., X. Zhang, L. Wang, and Y. Li. (2013). Curcumin induces ABCA1 expression and apolipoprotein A-I-mediated cholesterol transmembrane in the chronic cerebral hypoperfusion aging rats. Am J Chin Med 41: 1027-1042.

Tikhonova, E.B., V.K. Devroy, S.Y. Lau, and H.I. Zgurskaya. (2007). Reconstitution of the Escherichia coli macrolide transporter: the periplasmic membrane fusion protein MacA stimulates the ATPase activity of MacB. Mol. Microbiol. 63: 895-910.

Timachi, M.H., C.A. Hutter, M. Hohl, T. Assafa, S. Böhm, A. Mittal, M.A. Seeger, and E. Bordignon. (2017). Exploring conformational equilibria of a heterodimeric ABC transporter. Elife 6:.

Tjalsma, H., A. Bolhuis, J.D. Jongbloed, S. Bron, and J.M. van Dijl. (2000). Signal peptide-dependent protein transport in Bacillus subtilis: a genome-based survey of the secretome. Microbiol. Mol. Biol. Rev. 64: 515-547.

Togami, K., S. Chono, and K. Morimoto. (2012). Transport characteristics of clarithromycin, azithromycin and telithromycin, antibiotics applied for treatment of respiratory infections, in Calu-3 cell monolayers as model lung epithelial cells. Pharmazie 67: 389-393.

Tommasini, R., E. Vogt, M. Fromenteau, S. Hörtensteiner, P. Matile, N. Amrhein, and E. Martinoia. (1998). An ABC-transporter of Arabidopsis thaliana has both glutathione-conjugate and chlorophyll catabolite transport activity. Plant J. 13: 773-780.

Tong, A.Y., E.L. Tong, M.A. Hannani, S.N. Shaffer, D. Santiago, A.R. Ferré-D''Amaré, L.F.M. Passalacqua, and M.M. Abdelsayed. (2024). RNA thermometers are widespread upstream of ABC transporter genes in bacteria. J. Biol. Chem. 300: 107547. [Epub: Ahead of Print]

Torres, C., C. Galián, C. Freiberg, J.R. Fantino, and J.M. Jault. (2009). The YheI/YheH heterodimer from Bacillus subtilis is a multidrug ABC transporter. Biochim. Biophys. Acta. 1788: 615-622.

Torres, S.E., C.M. Gallagher, L. Plate, M. Gupta, C.R. Liem, X. Guo, R. Tian, R.M. Stroud, M. Kampmann, J.S. Weissman, and P. Walter. (2019). Ceapins block the unfolded protein response sensor ATF6α by inducing a neomorphic inter-organelle tether. Elife 8:.

Torres, Y.P., S.T. Granados, and R. Latorre. (2014). Pharmacological consequences of the coexpression of BK channel α and auxiliary β subunits. Front Physiol 5: 383.

Toussaint, F., B. Pierman, A. Bertin, D. Lévy, and M. Boutry. (2017). Purification and biochemical characterization of NpABCG5/NpPDR5, a plant pleiotropic drug resistance transporter expressed in Nicotiana tabacum BY-2 suspension cells. Biochem. J. 474: 1689-1703.

Tsujibo, H., M. Kosaka, S. Ikenishi, T. Sato, K. Miyamoto, and Y. Inamori. (2004). Molecular characterization of a high-affinity xylobiose transporter of Streptomyces thermoviolaceus OPC-520 and its transcriptional regulation. J. Bacteriol. 186: 1029-1037.

Tsuruoka, S., K. Ishibashi, H. Yamamoto, M. Wakaumi, M. Suzuki, G.J. Schwartz, M. Imai, and A. Fujimura. (2002). Functional analysis of ABCA8, a new drug transporter. Biochem. Biophys. Res. Commun. 298: 41-45.

Tsybovsky Y., Orban T., Molday RS., Taylor D. and Palczewski K. (2013). Molecular organization and ATP-induced conformational changes of ABCA4, the photoreceptor-specific ABC transporter. Structure. 21(5):854-60.

Tumulka, F., C. Roos, F. Löhr, C. Bock, F. Bernhard, V. Dötsch, and R. Abele. (2013). Conformational stabilization of the membrane embedded targeting domain of the lysosomal peptide transporter TAPL for solution NMR. J Biomol NMR 57: 141-154.

Turi, T.G. and J.K. Rose. (1995). Characterization of a novel Schizosaccharomyces pombe multidrug resistance transporter conferring brefeldin A resistance. Biochem. Biophys. Res. Commun. 213: 410-418.

Tutulan-Cunita, A.C., M. Mikoshi, M. Mizunuma, D. Hirata, and T. Miyakawa. (2005). Mutational analysis of the yeast multidrug resistance ABC transporter Pdr5p with altered drug specificity. Genes Cells 10: 409-420.

Udasin RG., Wen X., Bircsak KM., Aleksunes LM., Shakarjian MP., Kong AN., Heck DE., Laskin DL. and Laskin JD. (2016). Nrf2 Regulates the Sensitivity of Mouse Keratinocytes to Nitrogen Mustard via Multidrug Resistance-Associated Protein 1 (Mrp1). Toxicol Sci. 149(1):202-12.

Ueda, K., K. Oinuma, G. Ikeda, K. Hosono, Y. Ohnishi, S. Horinouchi, and T. Beppu. (2002). AmfS, an extracellular peptidic morphogen in Streptomyces griseus. J. Bacteriol. 184: 1488-1492.

Uehara, T. and J.T. Park. (2008). Growth of Escherichia coli: significance of peptidoglycan degradation during elongation and septation. J. Bacteriol. 190: 3914-3922.

Umashankar, B., L. Eliasson, C.Y. Ooi, K.W. Kim, J.A.M. Shaw, and S.A. Waters. (2024). Beyond insulin: Unraveling the complex interplay of ER stress, oxidative damage, and CFTR modulation in CFRD. J Cyst Fibros 23: 842-852.

Ungerer, J.L., B.S. Pratte, and T. Thiel. (2008). Regulation of fructose transport and its effect on fructose toxicity in Anabaena spp. J. Bacteriol. 190: 8115-8125.

Upadhyay, H.C., G.R. Dwivedi, S. Roy, A. Sharma, M.P. Darokar, and S.K. Srivastava. (2014). Phytol derivatives as drug resistance reversal agents. ChemMedChem 9: 1860-1868.

Vaccarin, C., G. Veit, T. Hegedus, O. Torres, A. Chilin, G.L. Lukacs, and G. Marzaro. (2024). Synthesis and Biological Evaluation of Pyrazole-Pyrimidones as a New Class of Correctors of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). J Med Chem 67: 13891-13908.

Vaché, C., O. Camares, F. De Graeve, B. Dastugue, A. Meiniel, C. Vaury, S. Pellier, E. Leoz-Garziandia, and M. Bamdad. (2006). Drosophila melanogaster p-glycoprotein: a membrane detoxification system toward polycyclic aromatic hydrocarbon pollutants. Environ Toxicol Chem 25: 572-580.

Vache, C., O. Camares, M.C. Cardoso-Ferreira, B. Dastugue, I. Creveaux, C. Vaury, and M. Bamdad. (2007). A potential genomic biomarker for the detection of polycyclic aromatic hydrocarbon pollutants: multidrug resistance gene 49 in Drosophila melanogaster. Environ Toxicol Chem 26: 1418-1424.

Vagts, J., K. Kalvelage, A. Weiten, R. Buschen, J. Gutsch, S. Scheve, L. Wöhlbrand, S. Diener, H. Wilkes, M. Winklhofer, and R. Rabus. (2021). Responsiveness of EbN1 to lignin-derived phenylpropanoids. Appl. Environ. Microbiol. [Epub: Ahead of Print]

Valladares, A., M.L. Montesinos, A. Herrero, and E. Flores. (2002). An ABC-type, high-affinity urea permease identified in cyanobacteria. Mol. Microbiol. 43: 703-715.

Van Bibber, M., C. Bradbeer, N. Clark, and J.R. Roth. (1999). A new class of cobalamin transport mutants (btuF) provides genetic evidence for a periplasmic binding protein in Salmonella typhimurium. J. Bacteriol. 181: 5539-5541.

van Bon BW., Gilissen C., Grange DK., Hennekam RC., Kayserili H., Engels H., Reutter H., Ostergaard JR., Morava E., Tsiakas K., Isidor B., Le Merrer M., Eser M., Wieskamp N., de Vries P., Steehouwer M., Veltman JA., Robertson SP., Brunner HG., de Vries BB. and Hoischen A. (2012). Cantu syndrome is caused by mutations in ABCC9. Am J Hum Genet. 90(6):1094-101.

van den Berg van Saparoea, H.B., J. Lubelski, R. van Merkerk, P.S. Mazurkiewicz, and A.J. Driessen. (2005). Proton motive force-dependent Hoechst 33342 transport by the ABC transporter LmrA of Lactococcus lactis. Biochemistry 44: 16931-16938.

van den Brûle, S., A. Müller, A.J. Fleming, and C.C. Smart. (2002). The ABC transporter SpTUR2 confers resistance to the antifungal diterpene sclareol. Plant J. 30: 649-662.

van der Does, C., C. Presenti, K. Schulze, S. Dinkelaker, and R. Tampé. (2006). Kinetics of the ATP hydrolysis cycle of the nucleotide-binding domain of Mdl1 studied by a novel site-specific labeling technique. J. Biol. Chem. 281: 5694-5701.

van der Heide, T. and B. Poolman. (2002). ABC transporters: one, two or four extracytoplasmic substrate-binding sites? EMBO Rep 3: 938-943.

van der Noord, V.E., W. van der Stel, G. Louwerens, D. Verhoeven, H.J. Kuiken, C. Lieftink, M. Grandits, G.F. Ecker, R.L. Beijersbergen, P. Bouwman, S.E. Le Dévédec, and B. van de Water. (2023). Systematic screening identifies ABCG2 as critical factor underlying synergy of kinase inhibitors with transcriptional CDK inhibitors. Breast Cancer Res 25: 51.

van der Ploeg, J.R., M.A. Weiss, E. Saller, H. Nashimoto, N. Saito, M.A. Kertesz, and T. Leisinger. (1996). Identification of sulfate starvation-regulated genes in Escherichia coli: a gene cluster involved in the utilization of taurine as a sulfur source. J. Bacteriol. 178: 5438-5446.

van Der Ploeg, J.R., R. Iwanicka-Nowicka, T. Bykowski, M.M. Hryniewicz, and T. Leisinger. (1999). The Escherichia coli ssuEADCB gene cluster is required for the utilization of sulfur from aliphatic sulfonates and is regulated by the transcriptional activator Cbl. J. Biol. Chem. 274: 29358-29365.

van Endert, P.M. (1999). Role of nucleotides and peptide substrate for stability and functional state of the human ABC family transporters associated with antigen processing. J. Biol. Chem. 274: 14632-14638.

van Gelder, M.A., Y. Li, D.P.A. Wander, I. Berlin, H.S. Overkleeft, S.Y. van der Zanden, and J.J.C. Neefjes. (2024). Novel -Dimethyl-idarubicin Analogues Are Effective Cytotoxic Agents for ABCB1-Overexpressing, Doxorubicin-Resistant Cells. J Med Chem 67: 13802-13812.

van Roermund, C.W., W.F. Visser, L. Ijlst, A. van Cruchten, M. Boek, W. Kulik, H.R. Waterham, and R.J. Wanders. (2008). The human peroxisomal ABC half transporter ALDP functions as a homodimer and accepts acyl-CoA esters. FASEB J. 22: 4201-4208.

van Roermund, C.W., W.F. Visser, L. Ijlst, H.R. Waterham, and R.J. Wanders. (2011). Differential substrate specificities of human ABCD1 and ABCD2 in peroxisomal fatty acid β-oxidation. Biochim. Biophys. Acta. 1811: 148-152.

van Veen, H.W. and W.N. Konings. (1998). The ABC family of multidrug transporters in microorganisms. Biochim. Biophys. Acta. 1365: 31-36.

van Veen, H.W., A. Margolles, M. Müller, C.F. Higgins, and W.N. Konings. (2000). The homodimeric ATP-binding cassette transporter LmrA mediates multidrug transport by an alternating two-site (two-cylinder engine) mechanism. EMBO. J. 19: 2503-2514.

van Wezel, G.P., J. White, M.J. Bibb, and P.W. Postma. (1997). The malEFG gene cluster of Streptomyces coelicolor A3(2): characterization, disruption and transcriptional analysis. Mol. Gen. Genet. 254: 604-608.

van Wonderen JH., McMahon RM., O'Mara ML., McDevitt CA., Thomson AJ., Kerr ID., MacMillan F. and Callaghan R. (2014). The central cavity of ABCB1 undergoes alternating access during ATP hydrolysis. FEBS J. 281(9):2190-201.

Vanakker, O.M., M.J. Hosen, and A.D. Paepe. (2013). The ABCC6 transporter: what lessons can be learnt from other ATP-binding cassette transporters? Front Genet 4: 203.

vanBerge-Henegouwen, G.P., N.G. Venneman, P. Portincasa, A. Kosters, K.J. van Erpecum, and A.K. Groen. (2004). Relevance of hereditary defects in lipid transport proteins for the pathogenesis of cholesterol gallstone disease. Scand J Gastroenterol Suppl 60-69.

Vanderpool, C.K. and S.K. Armstrong. (2004). Integration of environmental signals controls expression of Bordetella heme utilization genes. J. Bacteriol. 186: 938-948.

Vatamaniuk, O.K., E.A. Bucher, M.V. Sundaram, and P.A. Rea. (2005). CeHMT-1, a putative phytochelatin transporter, is required for cadmium tolerance in Caenorhabditis elegans. J. Biol. Chem. 280: 23684-23690.

Vats, P., Y.L. Shih, and L. Rothfield. (2009). Assembly of the MreB-associated cytoskeletal ring of Escherichia coli. Mol. Microbiol. 72: 170-182.

Velamakanni, S., Y. Yao, D.A. Gutmann, and H.W. van Veen. (2008). Multidrug transport by the ABC transporter Sav1866 from Staphylococcus aureus. Biochemistry 47: 9300-9308.

Vergauwen, B., J. Elegheert, A. Dansercoer, B. Devreese, and S.N. Savvides. (2010). Glutathione import in Haemophilus influenzae Rd is primed by the periplasmic heme-binding protein HbpA. Proc. Natl. Acad. Sci. USA 107: 13270-13275.

Verhalen, B., R. Dastvan, S. Thangapandian, Y. Peskova, H.A. Koteiche, R.K. Nakamoto, E. Tajkhorshid, and H.S. Mchaourab. (2017). Energy transduction and alternating access of the mammalian ABC transporter P-glycoprotein. Nature 543: 738-741.

Vernhet, L., N. Allain, C. Bardiau, J.P. Anger, and O. Fardel. (2000). Differential sensitivities of MRP1-overexpressing lung tumor cells to cytotoxic metals. Toxicology 142: 127-134.

Verweij, M.C., D. Koppers-Lalic, S. Loch, F. Klauschies, H. de la Salle, E. Quinten, P.J. Lehner, A. Mulder, M.R. Knittler, R. Tampé, J. Koch, M.E. Ressing, and E.J. Wiertz. (2008). The varicellovirus UL49.5 protein blocks the transporter associated with antigen processing (TAP) by inhibiting essential conformational transitions in the 6+6 transmembrane TAP core complex. J Immunol 181: 4894-4907.

Victor, A.K., T. Hedgecock, M. Donaldson, D. Johnson, C.M. Rand, D.E. Weese-Mayer, and L.T. Reiter. (2023). Analysis and comparisons of gene expression changes in patient- derived neurons from ROHHAD, CCHS, and PWS. Front Pediatr 11: 1090084.

Vigonsky, E., E. Ovcharenko, and O. Lewinson. (2013). Two molybdate/tungstate ABC transporters that interact very differently with their substrate binding proteins. Proc. Natl. Acad. Sci. USA 110: 5440-5445.

Villarreal, D.M., C.L. Phillips, A.M. Kelley, S. Villarreal, A. Villaloboz, P. Hernandez, J.S. Olson, and D.P. Henderson. (2008). Enhancement of recombinant hemoglobin production in Escherichia coli BL21(DE3) containing the Plesiomonas shigelloides heme transport system. Appl. Environ. Microbiol. 74: 5854-5856.

Vincent, P.A. and R.D. Morero. (2009). The structure and biological aspects of peptide antibiotic microcin J25. Curr. Med. Chem. 16: 538-549.

Vishwakarma, P., A. Banerjee, R. Pasrija, R. Prasad, and A.M. Lynn. (2019). The E-helix is a central core in a conserved helical bundle involved in nucleotide binding and transmembrane domain intercalation in the ABC transporter superfamily. Int J Biol Macromol 127: 95-106. [Epub: Ahead of Print]

Visser, W.F., C.W. van Roermund, L. Ijlst, H.R. Waterham, and R.J. Wanders. (2007). Metabolite transport across the peroxisomal membrane. Biochem. J. 401: 365-375.

Vitreschak, A.G., A.A. Mironov, V.A. Lyubetsky, and M.S. Gelfand. (2008). Comparative genomic analysis of T-box regulatory systems in bacteria. RNA 14: 717-735.

Vitreschak, A.G., D.A. Rodionov, A.A. Mironov, and M.S. Gelfand. (2002). Regulation of riboflavin biosynthesis and transport genes in bacteria by transcriptional and translational attenuation. Nucleic Acids Res 30: 3141-3151.

Viturro, E., C. Farke, H.H. Meyer, and C. Albrecht. (2006). Identification, sequence analysis and mRNA tissue distribution of the bovine sterol transporters ABCG5 and ABCG8. J Dairy Sci 89: 553-561.

Voith von Voithenberg, L., J. Park, R. Stübe, C. Lux, Y. Lee, and K. Philippar. (2019). A Novel Prokaryote-Type ECF/ABC Transporter Module in Chloroplast Metal Homeostasis. Front Plant Sci 10: 1264.

Voloshyna, I. and A.B. Reiss. (2011). The ABC transporters in lipid flux and atherosclerosis. Prog Lipid Res 50: 213-224.

Volpe, D.A. (2024). Knockout Transporter Cell Lines to Assess Substrate Potential Towards Efflux Transporters. AAPS J 26: 79.

Wada, J., R. Suzuki, S. Fushinobu, M. Kitaoka, T. Wakagi, H. Shoun, H. Ashida, H. Kumagai, T. Katayama, and K. Yamamoto. (2007). Purification, crystallization and preliminary X-ray analysis of the galacto-N-biose-/lacto-N-biose I-binding protein (GL-BP) of the ABC transporter from Bifidobacterium longum JCM1217. Acta Crystallogr Sect F Struct Biol Cryst Commun 63: 751-753.

Wadsack, D., H. Sattler, M.E. Kleber, K. Eirich, H. Scharnagl, G. Fauler, W. März, and T.B. Grammer. (2018). Recurrent tendosynovitis as a rare manifestation of a lipid disorder. J Clin Lipidol 13: 54-61.

Wakabayashi, K., H. Nakagawa, A. Tamura, S. Koshiba, K. Hoshijima, M. Komada, and T. Ishikawa. (2007). Intramolecular disulfide bond is a critical check point determining degradative fates of ATP-binding cassette (ABC) transporter ABCG2 protein. J. Biol. Chem. 282: 27841-27846.

Wakabayashi, Y., H. Kipp, and I.M. Arias. (2006). Transporters on demand: intracellular reservoirs and cycling of bile canalicular ABC transporters. J. Biol. Chem. 281: 27669-27673.

Walshaw, D.L. and P.S. Poole. (1996). The general L-amino acid permease of Rhizobium leguminosarum is an ABC uptake system that also influences efflux of solutes. Mol. Microbiol. 21: 1239-1252.

Walshaw, D.L., S. Lowthorpe, A. East, and P.S. Poole. (1997). Distribution of a sub-class of bacterial ABC polar amino acid transporter and identification of an N-terminal region involved in solute specificity. FEBS Lett. 414: 397-401.

Wan, Y., Z. Chen, Y. Wang, W. Zhao, Z. Pei, L. Pu, Y. Lv, J. Li, J. Li, and Y. Pei. (2023). A hyaluronic acid modified cuprous metal-organic complex for reversing multidrug resistance via redox dyshomeostasis. Carbohydr Polym 311: 120762.

Wang F., Li G., Gu HM. and Zhang DW. (2013). Characterization of the role of a highly conserved sequence in ATP binding cassette transporter G (ABCG) family in ABCG1 stability, oligomerization, and trafficking. Biochemistry. 52(52):9497-509.

Wang W. and Linsdell P. (2012). Conformational change opening the CFTR chloride channel pore coupled to ATP-dependent gating. Biochim Biophys Acta. 1818(3):851-60.

Wang, B., M. Dukarevich, E.I. Sun, M.R. Yen, and M.H. Saier, Jr. (2009). Membrane porters of ATP-binding cassette transport systems are polyphyletic. J. Membr. Biol. 231: 1-10.

Wang, C., C. Cao, N. Wang, X. Wang, X. Wang, and X.C. Zhang. (2020). Cryo-electron microscopy structure of human ABCB6 transporter. Protein. Sci. [Epub: Ahead of Print]

Wang, D., A. Tosevska, E.H. Heiß, A. Ladurner, C. Mölzer, M. Wallner, A. Bulmer, K.H. Wagner, V.M. Dirsch, and A.G. Atanasov. (2017). Bilirubin Decreases Macrophage Cholesterol Efflux and ATP-Binding Cassette Transporter A1 Protein Expression. J Am Heart Assoc 6:.

Wang, E.J. and W.W. Johnson. (2003). The farnesyl protein transferase inhibitor lonafarnib (SCH66336) is an inhibitor of multidrug resistance proteins 1 and 2. Chemotherapy 49: 303-308.

Wang, F., X. Xiao, A. Saito, and H. Schrempf. (2002). Streptomyces olivaceoviridis possesses a phosphotransferase system that mediates specific, phosphoenolpyruvate-dependent uptake of N-acetylglucosamine. Mol. Genet. Genomics 268: 344-351.

Wang, H., E.W. Lee, X. Cai, Z. Ni, L. Zhou, and Q. Mao. (2008). Membrane topology of the human breast cancer resistance protein (BCRP/ABCG2) determined by epitope insertion and immunofluorescence. Biochemistry 47: 13778-13787.

Wang, H., Y. Liang, Y. Yin, J. Zhang, W. Su, A.M. White, Bin Jiang, J. Xu, Y. Zhang, S. Stewart, X. Lu, and X. He. (2021). Carbon nano-onion-mediated dual targeting of P-selectin and P-glycoprotein to overcome cancer drug resistance. Nat Commun 12: 312.

Wang, J., F. Sun, D.W. Zhang, Y. Ma, F. Xu, J.D. Belani, J.C. Cohen, H.H. Hobbs, and X.S. Xie. (2006). Sterol transfer by ABCG5 and ABCG8: in vitro assay and reconstitution. J. Biol. Chem. 281: 27894-27904.

Wang, J., L.L. Zou, and A.X. Li. (2013). A novel iron transporter in Streptococcus iniae. J Fish Dis 36: 1007-1015.

Wang, J., N. Grishin, L. Kinch, J.C. Cohen, H.H. Hobbs, and X.S. Xie. (2011). Sequences in the nonconsensus nucleotide-binding domain of ABCG5/ABCG8 required for sterol transport. J. Biol. Chem. 286: 7308-7314.

Wang, J., X. Li, F.F. Wang, M.T. Cheng, Y.X. Mao, S.C. Fang, L. Wang, C.Z. Zhou, W.T. Hou, and Y. Chen. (2023). Placing steroid hormones within the human ABCC3 transporter reveals a compatible amphiphilic substrate-binding pocket. EMBO. J. e113415. [Epub: Ahead of Print]

Wang, L., X. Sun, J. He, and Z. Liu. (2022). Identification and Validation of Prognostic Related Hallmark ATP-Binding Cassette Transporters Associated With Immune Cell Infiltration Patterns in Thyroid Carcinoma. Front Oncol 12: 781686.

Wang, L.X., S. Tao, Y.C. Zhang, X.G. Pei, Y. Gao, X.Y. Song, Z.T. Yu, and C.F. Gao. (2021). Overexpression of ATP-binding cassette transporter Mdr49-like confers resistance to imidacloprid in the field populations of brown planthopper, Nilaparvata lugens. Pest Manag Sci. [Epub: Ahead of Print]

Wang, S., G. He, Y. Liu, Y. Wang, Y. Ma, C. Fu, H. Xu, R. Hu, and S. Li. (2024). A P1-like MYB transcription factor boosts biosynthesis and transport of C-glycosylated flavones in duckweed. Int J Biol Macromol 277: 134138.

Wang, T., G. Fu, X. Pan, J. Wu, X. Gong, J. Wang, and Y. Shi. (2013). Structure of a bacterial energy-coupling factor transporter. Nature 497: 272-276.

Wang, W. and P. Linsdell. (2012). Alternating access to the transmembrane domain of the ATP-binding cassette protein cystic fibrosis transmembrane conductance regulator (ABCC7). J. Biol. Chem. 287: 10156-10165.

Wang, Y., M.J. Tu, and A.M. Yu. (2024). Efflux ABC transporters in drug disposition and their posttranscriptional gene regulation by microRNAs. Front Pharmacol 15: 1423416.

Wang, Y., T.W. Loo, M.C. Bartlett, and D.M. Clarke. (2007). Correctors promote maturation of cystic fibrosis transmembrane conductance regulator (CFTR)-processing mutants by binding to the protein. J. Biol. Chem. 282: 33247-33251.

Wang, Y., X. Li, Y. Wang, S. Schwarz, J. Shen, and X. Xia. (2018). Intracellular Accumulation of Linezolid and Florfenicol in OptrA-Producing and. Molecules 23:.

Wang, Y., X. Zhang, Y. Yan, T. Niu, M. Zhang, C. Fan, W. Liang, Y. Shu, C. Guo, D. Guo, and Y. Bi. (2023). GmABCG5, an ATP-binding cassette G transporter gene, is involved in the iron deficiency response in soybean. Front Plant Sci 14: 1289801.

Wang, Z., P. Zeng, and B. Zhou. (2022). Identification and characterization of a heme exporter from the MRP family in Drosophila melanogaster. BMC Biol 20: 126.

Wang, Z., W. Hu, and H. Zheng. (2020). Pathogenic siderophore ABC importer YbtPQ adopts a surprising fold of exporter. Sci Adv 6: eaay7997.

Ward, A., C.L. Reyes, J. Yu, C.B. Roth, and G. Chang. (2007). Flexibility in the ABC transporter MsbA: Alternating access with a twist. Proc. Natl. Acad. Sci. USA 104: 19005-19010.

Ward, M.J., K.C. Mok, D.P. Astling, H. Lew, and D.R. Zusman. (1998). An ABC transporter plays a developmental aggregation role in Myxococcus xanthus. J. Bacteriol. 180: 5697-5703.

Ware, D., Y. Jiang, W. Lin, and E. Swiatlo. (2006). Involvement of potD in Streptococcus pneumoniae polyamine transport and pathogenesis. Infect. Immun. 74: 352-361.

Watanabe, S., Y. Oguchi, N. Yokota, and H. Tokuda. (2007). Large-scale preparation of the homogeneous LolA lipoprotein complex and efficient in vitro transfer of lipoproteins to the outer membrane in a LolB-dependent manner. Protein. Sci. 16: 2741-2749.

Watzlawick, H., K. Morabbi Heravi, and J. Altenbuchner. (2016). The role of the ganSPQAB operon in degradation of galactan by Bacillus subtilis. J. Bacteriol. [Epub: Ahead of Print]

Webb, A.J. and A.H. Hosie. (2006). A member of the second carbohydrate uptake subfamily of ATP-binding cassette transporters is responsible for ribonucleoside uptake in Streptococcus mutans. J. Bacteriol. 188: 8005-8012.

Webb, A.J., K.A. Homer, and A.H. Hosie. (2008). Two closely related ABC transporters in Streptococcus mutans are involved in disaccharide and/or oligosaccharide uptake. J. Bacteriol. 190: 168-178.

Webb, E., K. Claas, and D. Downs. (1998). thiBPQ encodes an ABC transporter required for transport of thiamine and thiamine pyrophosphate in Salmonella typhimurium. J. Biol. Chem. 273: 8946-8950.

Wei, X., Y. Guo, C. Shao, Z. Sun, D. Zhurina, D. Liu, W. Liu, D. Zou, Z. Jiang, X. Wang, J. Zhao, W. Shang, X. Li, X. Liao, L. Huang, C.U. Riedel, and J. Yuan. (2012). Fructose uptake in Bifidobacterium longum NCC2705 is mediated by an ATP-binding cassette transporter. J. Biol. Chem. 287: 357-367.

Wei, Y., A.A. Guffanti, and T.A. Krulwich. (1999). Sequence analysis and functional studies of a chromosomal region of alkaliphilic Bacillus firmus OF4 encoding an ABC-type transporter with similarity of sequence and Na+ exclusion capacity to the Bacillus subtilis NatAB transporter. Extremophiles 3: 113-120.

Weigl, K.E., G. Conseil, A.J. Rothnie, M. Arama, Y. Tsfadia, and S.P.C. Cole. (2018). An Outward-Facing Aromatic Amino Acid Is Crucial for Signaling between the Membrane-Spanning and Nucleotide-Binding Domains of Multidrug Resistance Protein 1 (MRP1; ABCC1). Mol Pharmacol 94: 1069-1078.

Wen, Y., R. Zhao, P. Gupta, Y. Fan, Y. Zhang, Z. Huang, X. Li, Y. Su, L. Liao, Y.A. Xie, D. Yang, Z.S. Chen, and G. Liang. (2019). The epigallocatechin gallate derivative Y reverses drug resistance mediated by the ABCB1 transporter both and. Acta Pharm Sin B 9: 316-323.

Weng, J., K. Fan, and W. Wang. (2012). The conformational transition pathways of ATP-binding cassette transporter BtuCD revealed by targeted molecular dynamics simulation. PLoS One 7: e30465.

Wescombe, P.A., M. Upton, P. Renault, R.E. Wirawan, D. Power, J.P. Burton, C.N. Chilcott, and J.R. Tagg. (2011). Salivaricin 9, a new lantibiotic produced by Streptococcus salivarius. Microbiology 157: 1290-1299.

Westover, D. and F. Li. (2015). New trends for overcoming ABCG2/BCRP-mediated resistance to cancer therapies. J Exp Clin Cancer Res 34: 159.

White, A.K. and W.W. Metcalf. (2004). Two C-P lyase operons in Pseudomonas stutzeri and their roles in the oxidation of phosphonates, phosphite, and hypophosphite. J. Bacteriol. 186: 4730-4739.

White, D.W., S.R. Elliott, E. Odean, L.T. Bemis, and A.D. Tischler. (2018). Pst/SenX3-RegX3 Regulates Membrane Vesicle Production Independently of ESX-5 Activity. MBio 9:.

White, J.P., J. Prell, V.K. Ramachandran, and P.S. Poole. (2009). Characterization of a {gamma}-aminobutyric acid transport system of Rhizobium leguminosarum bv. viciae 3841. J. Bacteriol. 191: 1547-1555.

Wilkens, S. (2015). Structure and mechanism of ABC transporters. F1000Prime Rep 7: 14.

Wille T., Wagner C., Mittelstadt W., Blank K., Sommer E., Malengo G., Dohler D., Lange A., Sourjik V., Hensel M. and Gerlach RG. (2014). SiiA and SiiB are novel type I secretion system subunits controlling SPI4-mediated adhesion of Salmonella enterica. Cell Microbiol. 16(2):161-78.

Willemsen, P.T., I. Vulto, M. Boxem, and J. de Graaff. (1997). Characterization of a periplasmic protein involved in iron utilization of Actinobacillus actinomycetemcomitans. J. Bacteriol. 179: 4949-4952.

Willis, L.B. and G.C. Walker. (1999). A novel Sinorhizobium meliloti operon encodes an α-glucosidase and a periplasmic-binding-protein-dependent transport system for α-glucosides. J. Bacteriol. 181: 4176-4184.

Windsor, B., S.J. Roux, and A. Lloyd. (2003). Multiherbicide tolerance conferred by AtPgp1 and apyrase overexpression in Arabidopsis thaliana. Nat Biotechnol 21: 428-433.

Winkler M., Kuhner P., Russ U., Ortiz D., Bryan J. and Quast U. (2012). Role of the amino-terminal transmembrane domain of sulfonylurea receptor SUR2B for coupling to K(IR)6.2, ligand binding, and oligomerization. Naunyn Schmiedebergs Arch Pharmacol. 385(3):287-98.

Wong, S.L., E. Kardia, A. Vijayan, B. Umashankar, E. Pandzic, L. Zhong, A. Jaffe, and S.A. Waters. (2023). Molecular and Functional Characteristics of Airway Epithelium under Chronic Hypoxia. Int J Mol Sci 24:.

Woodson, J.D., A.A. Reynolds, and J.C. Escalante-Semerena. (2005). ABC transporter for corrinoids in Halobacterium sp. strain NRC-1. J. Bacteriol. 187: 5901-5909.

Wooff, E., S.L. Michell, S.V. Gordon, M.A. Chambers, S. Bardarov, W.R. Jacobs, Jr, R.G. Hewinson, and P.R. Wheeler. (2002). Functional genomics reveals the sole sulphate transporter of the Mycobacterium tuberculosis complex and its relevance to the acquisition of sulphur in vivo. Mol. Microbiol. 43: 653-663.

Wu, C.P., M. Murakami, Y.C. Li, Y.H. Huang, Y.T. Chang, T.H. Hung, Y.S. Wu, and S.V. Ambudkar. (2023). Imperatorin Restores Chemosensitivity of Multidrug-Resistant Cancer Cells by Antagonizing ABCG2-Mediated Drug Transport. Pharmaceuticals (Basel) 16:.

Wu, C.P., S. Lusvarghi, S.H. Hsiao, T.C. Liu, Y.Q. Li, Y.H. Huang, T.H. Hung, and S.V. Ambudkar. (2020). Licochalcone A Selectively Resensitizes ABCG2-Overexpressing Multidrug-Resistant Cancer Cells to Chemotherapeutic Drugs. J Nat Prod. [Epub: Ahead of Print]

Wu, C.P., Y.Q. Li, T.H. Hung, Y.T. Chang, Y.H. Huang, and Y.S. Wu. (2021). Sophoraflavanone G Resensitizes ABCG2-Overexpressing Multidrug-Resistant Non-Small-Cell Lung Cancer Cells to Chemotherapeutic Drugs. J Nat Prod 84: 2544-2553.

Wu, D., A.R. Mehdipour, F. Finke, H.G. Goojani, R.R. Groh, T.N. Grund, T.M.B. Reichhart, R. Zimmermann, S. Welsch, D. Bald, M. Shepherd, G. Hummer, and S. Safarian. (2023). Dissecting the conformational complexity and mechanism of a bacterial heme transporter. Nat Chem Biol. [Epub: Ahead of Print]

Wu, J.X., D. Ding, and L. Chen. (2022). The Emerging Structural Pharmacology of ATP-Sensitive Potassium Channels. Mol Pharmacol 102: 234-239.

Wu, L.F. and M.A. Mandrand-Berthelot. (1995). A family of homologous substrate-binding proteins with a broad range of substrate specificity and dissimilar biological functions. Biochimie 77: 744-750.

Wu, S.S., J. Wu, Y.L. Cheng, and D. Kaiser. (1998). The pilH gene encodes an ABC transporter homologue required for type IV pilus biogenesis and social gliding motility in Myxococcus xanthus. Mol. Microbiol. 29: 1249-1261.

Wu, T.K., Y.K. Wang, Y.C. Chen, J.M. Feng, Y.H. Liu, and T.Y. Wang. (2007). Identification of a Vibrio furnissii oligopeptide permease and characterization of its in vitro hemolytic activity. J. Bacteriol. 189: 8215-8223.

Wu, Y., L. Chen, Z. Xie, C. Wang, J. Zhang, and X. Yan. (2022). Effects of ABCG1 knockout on proteomic composition of HDL in mice on a chow diet and a high-fat diet. Proteomics e2100028. [Epub: Ahead of Print]

Wunderlich, J. (2022). Updated List of Transport Proteins in. Front Cell Infect Microbiol 12: 926541.

Wuttge S., Bommer M., Jager F., Martins BM., Jacob S., Licht A., Scheffel F., Dobbek H. and Schneider E. (2012). Determinants of substrate specificity and biochemical properties of the sn-glycerol-3-phosphate ATP binding cassette transporter (UgpB-AEC2 ) of Escherichia coli. Mol Microbiol. 86(4):908-20.

Xavier, B.M., A.A. Zein, A. Venes, J. Wang, and J.Y. Lee. (2020). Transmembrane Polar Relay Drives the Allosteric Regulation for ABCG5/G8 Sterol Transporter. Int J Mol Sci 21:.

Xavier, B.M., W.J. Jennings, A.A. Zein, J. Wang, and J.Y. Lee. (2018). Structural snapshot of the cholesterol-transport ABC proteins. Biochem. Cell Biol. [Epub: Ahead of Print]

Xiao, K.R., C.Y. Wu, L. Yang, J. Wang, Q.S. Song, D. Stanley, S.J. Wei, and J.Y. Zhu. (2022). Comparative genomic analysis of ABC transporter genes in Tenebrio molitor and four other tenebrionid beetles (Coleoptera: Tenebrionidea). Arch Insect Biochem Physiol e21916. [Epub: Ahead of Print]

Xie, T., Z. Zhang, J. Yue, Q. Fang, and X. Gong. (2022). Cryo-EM structures of the human surfactant lipid transporter ABCA3. Sci Adv 8: eabn3727.

Xie, T., Z. Zhang, Q. Fang, B. Du, and X. Gong. (2021). Structural basis of substrate recognition and translocation by human ABCA4. Nat Commun 12: 3853.

Xie, Y., J. Ma, X. Qin, Q. Li, and J. Ju. (2017). Identification and utilization of two important transporters: SgvT1 and SgvT2, for griseoviridin and viridogrisein biosynthesis in Streptomyces griseoviridis. Microb Cell Fact 16: 177.

Xing, J., H. Mei, S. Huang, D. Zhang, and X. Pan. (2019). An Energetically Favorable Ligand Entrance Gate of a Multidrug Transporter Revealed by Partial Nudged Elastic Band Simulations. Comput Struct Biotechnol J 17: 319-323.

Xiong, C., L.N. Jia, W.X. Xiong, X.T. Wu, L.L. Xiong, T.H. Wang, D. Zhou, Z. Hong, Z. Liu, and L. Tang. (2023). Structural insights into substrate recognition and translocation of human peroxisomal ABC transporter ALDP. Signal Transduct Target Ther 8: 74.

Xiong, J., L. Feng, D. Yuan, C. Fu, and W. Miao. (2010). Genome-wide identification and evolution of ATP-binding cassette transporters in the ciliate Tetrahymena thermophila: A case of functional divergence in a multigene family. BMC Evol Biol 10: 330.

Xu, C., J. Fan, W. Riekhof, J.E. Froehlich, and C. Benning. (2003). A permease-like protein involved in ER to thylakoid lipid transfer in Arabidopsis. EMBO. J. 22: 2370-2379.

Xu, H.Q., M. Ma, Y.P. Ma, S.Y. Zhang, W.J. Li, D. Wei, and J.J. Wang. (2021). Identification and Expression Characterization of ATP-Binding Cassette (ABC) Transporter Genes in Melon Fly. Insects 12:.

Xu, J., H. Peng, Q. Chen, Y. Liu, Z. Dong, and J.T. Zhang. (2007). Oligomerization domain of the multidrug resistance-associated transporter ABCG2 and its dominant inhibitory activity. Cancer Res 67: 4373-4381.

Xu, J., Y. Liu, Y. Yang, S. Bates, and J.T. Zhang. (2004). Characterization of oligomeric human half-ABC transporter ATP-binding cassette G2. J. Biol. Chem. 279: 19781-19789.

Xu, K., M. Zhang, Q. Zhao, F. Yu, H. Guo, C. Wang, F. He, J. Ding, and P. Zhang. (2013). Crystal structure of a folate energy-coupling factor transporter from Lactobacillus brevis. Nature 497: 268-271.

Xu, R., L. Lin, Z. Jiao, R. Liang, Y. Guo, Y. Zhang, X. Shang, Y. Wang, X. Wang, L. Yao, S. Liu, X. Deng, J. Yuan, X.Z. Su, and J. Li. (2024). Deaggregation of mutant Plasmodium yoelii de-ubiquitinase UBP1 alters MDR1 localization to confer multidrug resistance. Nat Commun 15: 1774.

Xu, X.H., H.J. Zhao, Q.L. Liu, T. Frank, K.H. Engel, G. An, and Q.Y. Shu. (2009). Mutations of the multi-drug resistance-associated protein ABC transporter gene 5 result in reduction of phytic acid in rice seeds. Theor Appl Genet 119: 75-83.

Xu, Y., I. Jo, L. Wang, J. Chen, S. Fan, Y. Dong, C. Quan, and N.C. Ha. (2017). Hexameric assembly of membrane fusion protein YknX of the sporulation delaying efflux pump from Bacillus amyloliquefaciens. Biochem. Biophys. Res. Commun. 493: 152-157.

Xu, Y., S.H. Sim, K.H. Nam, X.L. Jin, H.M. Kim, K.Y. Hwang, K. Lee, and N.C. Ha. (2009). Crystal structure of the periplasmic region of MacB, a noncanonic ABC transporter. Biochemistry 48: 5218-5225.

Yadav, V., I. Molina, K. Ranathunge, I.Q. Castillo, S.J. Rothstein, and J.W. Reed. (2014). ABCG transporters are required for suberin and pollen wall extracellular barriers in Arabidopsis. Plant Cell 26: 3569-3588.

Yamada Y., Tikhonova EB. and Zgurskaya HI. (2012). YknWXYZ is an unusual four-component transporter with a role in protection against sporulation-delaying-protein-induced killing of Bacillus subtilis. J Bacteriol. 194(16):4386-94.

Yamada, S., N. Awano, K. Inubushi, E. Maeda, S. Nakamori, K. Nishino, A. Yamaguchi, and H. Takagi. (2006). Effect of drug transporter genes on cysteine export and overproduction in Escherichia coli. Appl. Environ. Microbiol. 72: 4735-4742.

Yamada, T., M. Miyashita, J. Kasahara, T. Tanaka, M. Hashimoto, and H. Yamamoto. (2018). The transmembrane segment of TagH is required for wall teichoic acid transport under heat stress in Bacillus subtilis. Microbiology 164: 935-945.

Yamanaka, H., H. Kobayashi, E. Takahashi, and K. Okamoto. (2008). MacAB is involved in the secretion of Escherichia coli heat-stable enterotoxin II. J. Bacteriol. 190: 7693-7698.

Yamashita, M., M. Shepherd, W.I. Booth, H. Xie, V. Postis, Y. Nyathi, S.B. Tzokov, R.K. Poole, S.A. Baldwin, and P.A. Bullough. (2014). Structure and Function of the Bacterial Heterodimeric ABC Transporter CydDC: STIMULATION OF ATPASE ACTIVITY BY THIOL AND HEME COMPOUNDS. J. Biol. Chem. 289: 23177-23188.

Yang K., Wang M. and Metcalf WW. (2009). Uptake of glycerol-2-phosphate via the ugp-encoded transporter in Escherichia coli K-12. J Bacteriol. 191(14):4667-70.

Yang, B., Y. Yan, D. Wang, Y. Zhang, J. Yin, and G. Zhu. (2023). On-target inhibition of Cryptosporidium parvum by nitazoxanide (NTZ) and paclitaxel (PTX) validated using a novel MDR1-transgenic host cell model and algorithms to quantify the effect on the parasite target. PLoS Negl Trop Dis 17: e0011217.

Yang, C.S., W.C. Huang, T.P. Ko, Y.C. Wang, A.H. Wang, and Y. Chen. (2020). Crystal structure of the N-terminal domain of TagH reveals a potential drug targeting site. Biochem. Biophys. Res. Commun. 536: 1-6. [Epub: Ahead of Print]

Yang, D.C., N.T. Peters, K.R. Parzych, T. Uehara, M. Markovski, and T.G. Bernhardt. (2011). An ATP-binding cassette transporter-like complex governs cell-wall hydrolysis at the bacterial cytokinetic ring. Proc. Natl. Acad. Sci. USA 108: E1052-1060.

Yang, H., C. Zhou, X.B. Yang, G.Y. Long, and D.C. Jin. (2019). Effects of Insecticide Stress on Expression of Transporter Gene in the Brown Planthopper,. Insects 10:.

Yang, H.B., W.T. Hou, M.T. Cheng, Y.L. Jiang, Y. Chen, and C.Z. Zhou. (2018). Structure of a MacAB-like efflux pump from Streptococcus pneumoniae. Nat Commun 9: 196.

Yang, L., Y. Jin, W. Huang, Q. Sun, F. Liu, and X. Huang. (2018). Full-length transcriptome sequences of ephemeral plant Arabidopsis pumila provides insight into gene expression dynamics during continuous salt stress. BMC Genomics 19: 717.

Yang, W., X. Liu, S. Yu, J. Liu, L. Jiang, X. Lu, Y. Liu, J. Zhang, X. Li, and S. Zhang. (2023). The maize ATP-binding cassette (ABC) transporter ZmMRPA6 confers cold and salt stress tolerance in plants. Plant Cell Rep 43: 13.

Yang, X., Z. Wu, X. Wang, C. Yang, H. Xu, and Y. Shen. (2009). Crystal structure of lipoprotein GNA1946 from Neisseria meningitidis. J Struct Biol 168: 437-443.

Yang, Y., N. Ji, C.Y. Cai, J.Q. Wang, Z.N. Lei, Q.X. Teng, Z.X. Wu, Q. Cui, Y. Pan, and Z.S. Chen. (2020). Modulating the function of ABCB1: in vitro and in vivo characterization of sitravatinib, a tyrosine kinase inhibitor. Cancer Commun (Lond). [Epub: Ahead of Print]

Ye, X.L., F.H. Yu, J. Zhou, C.N. Zhao, and J. Wu. (2024). [Clinical phenotype and genotype analysis of progressive familial intrahepatic cholestasis type 3 caused by novel ABCB4 gene mutation]. Zhonghua Er Ke Za Zhi 62: 649-654.

Yernaux, C., M. Fransen, C. Brees, S. Lorenzen, and P.A. Michels. (2006). Trypanosoma brucei glycosomal ABC transporters: identification and membrane targeting. Mol. Membr. Biol. 23: 157-172.

Yerushalmi, G.M., B. Shuraki, Y. Yung, E. Maman, M. Baum, J.D. Hennebold, E.Y. Adashi, and A. Hourvitz. (2023). ABCC4 is a PGE2 efflux transporter in the ovarian follicle: A mediator of ovulation and a potential non-hormonal contraceptive target. FASEB J. 37: e22858.

Ying, W., L. Liao, H. Wei, Y. Gao, X. Liu, and L. Sun. (2023). Structural basis for abscisic acid efflux mediated by ABCG25 in Arabidopsis thaliana. Nat Plants. [Epub: Ahead of Print]

Yoneyama F., Imura Y., Ohno K., Zendo T., Nakayama J., Matsuzaki K. and Sonomoto K. (2009). Peptide-lipid huge toroidal pore, a new antimicrobial mechanism mediated by a lactococcal bacteriocin, lacticin Q. Antimicrob Agents Chemother. 53(8):3211-7.

Yoshida, K.I., Y. Fujita, and S.D. Ehrlich. (2000). An operon for a putative ATP-binding cassette transport system involved in acetoin utilization of Bacillus subtilis. J. Bacteriol. 182: 5454-5461.

Yoshimura, K. and T. Kouyama. (2008). Structural role of bacterioruberin in the trimeric structure of archaerhodopsin-2. J. Mol. Biol. 375: 1267-1281.

Yoshiura, K., A. Kinoshita, T. Ishida, A. Ninokata, T. Ishikawa, T. Kaname, M. Bannai, K. Tokunaga, S. Sonoda, R. Komaki, M. Ihara, V.A. Saenko, G.K. Alipov, I. Sekine, K. Komatsu, H. Takahashi, M. Nakashima, N. Sosonkina, C.K. Mapendano, M. Ghadami, M. Nomura, D.S. Liang, N. Miwa, D.K. Kim, A. Garidkhuu, N. Natsume, T. Ohta, H. Tomita, A. Kaneko, M. Kikuchi, G. Russomando, K. Hirayama, M. Ishibashi, A. Takahashi, N. Saitou, J.C. Murray, S. Saito, Y. Nakamura, and N. Niikawa. (2006). A SNP in the ABCC11 gene is the determinant of human earwax type. Nat. Genet. 38: 324-330.

Yost, C.K., A.M. Rath, T.C. Noel, and M.F. Hynes. (2006). Characterization of genes involved in erythritol catabolism in Rhizobium leguminosarum bv. viciae. Microbiology 152: 2061-2074.

Young, J. and I.B. Holland. (1999). ABC transporters: bacterial exporters-revisited five years on. Biochim. Biophys. Acta. 1461: 177-200.

Young, L., K. Leonhard, T. Tatsuta, J. Trowsdale, and T. Langer. (2001). Role of the ABC transporter Mdl1 in peptide export from mitochondria. Science 291: 2135-2138.

Young, P.G., J. Levring, K. Fiedorczuk, S.C. Blanchard, and J. Chen. (2024). Structural basis for CFTR inhibition by CFTR-172. Proc. Natl. Acad. Sci. USA 121: e2316675121.

Younus, I., S. Kochkina, C.C. Choi, W. Sun, and R.C. Ford. (2022). ATP-Binding Cassette Transporters: Snap-on Complexes? Subcell Biochem 99: 35-82.

Yu J., Ge J., Heuveling J., Schneider E. and Yang M. (2015). Structural basis for substrate specificity of an amino acid ABC transporter. Proc Natl Acad Sci U S A. 112(16):5243-8.

Yu Y., Zhou M., Kirsch F., Xu C., Zhang L., Wang Y., Jiang Z., Wang N., Li J., Eitinger T. and Yang M. (2014). Planar substrate-binding site dictates the specificity of ECF-type nickel/cobalt transporters. Cell Res. 24(3):267-77.

Yu, J.Y., T. Chen, Z.H. Wang, J. Zheng, and T.S. Zeng. (2022). Diagnosis, treatment and genetic analysis of a case of skin hyperpigmentation as the only manifestation with X-linked adrenoleukodystrophy. Yi Chuan 44: 983-989.

Yuan, X., J. Shen, and H. Zeng. (2024). Artificial transmembrane potassium transporters: designs, functions, mechanisms and applications. Chem Commun (Camb) 60: 482-500.

Yuan, Y.R., S. Blecker, O. Martsinkevich, L. Millen, P.J. Thomas, and J.F. Hunt. (2001). The crystal structure of the MJ0796 ATP-binding cassette. Implications for the structural consequences of ATP hydrolysis in the active site of an ABC transporter. J. Biol. Chem. 276: 32313-32321.

Yum, S., Y. Xu, S. Piao, S.H. Sim, H.M. Kim, W.S. Jo, K.J. Kim, H.S. Kweon, M.H. Jeong, H. Jeon, K. Lee, and N.C. Ha. (2009). Crystal structure of the periplasmic component of a tripartite macrolide-specific efflux pump. J. Mol. Biol. 387: 1286-1297.

Zaremba-Niedzwiedzka, K., E.F. Caceres, J.H. Saw, D. Bäckström, L. Juzokaite, E. Vancaester, K.W. Seitz, K. Anantharaman, P. Starnawski, K.U. Kjeldsen, M.B. Stott, T. Nunoura, J.F. Banfield, A. Schramm, B.J. Baker, A. Spang, and T.J. Ettema. (2017). Asgard archaea illuminate the origin of eukaryotic cellular complexity. Nature 541: 353-358.

Zdravkovic, B., T.P. Zdravkovic, M. Zdravkovic, B. Strukelj, and P. Ferk. (2019). The influence of nano-TiO2 on metabolic activity, cytotoxicity and ABCB5 mRNA expression in WM-266-4 human metastatic melanoma cell line. J BUON 24: 338-346.

Zhang, D.W., G.A. Graf, R.D. Gerard, J.C. Cohen, and H.H. Hobbs. (2006). Functional asymmetry of nucleotide-binding domains in ABCG5 and ABCG8. J. Biol. Chem. 281: 4507-4516.

Zhang, D.W., K. Nunoya, M. Vasa, H.M. Gu, S.P. Cole, and R.G. Deeley. (2006). Mutational analysis of polar amino acid residues within predicted transmembrane helices 10 and 16 of multidrug resistance protein 1 (ABCC1): effect on substrate specificity. Drug Metab Dispos 34: 539-546.

Zhang, H., A. Patel, Y.J. Wang, Y.K. Zhang, R.J. Kathawala, L.H. Qiu, B.A. Patel, L.H. Huang, S. Shukla, D.H. Yang, S.V. Ambudkar, L.W. Fu, and Z.S. Chen. (2017). The BTK Inhibitor Ibrutinib (PCI-32765) Overcomes Paclitaxel Resistance in ABCB1- and ABCC10-Overexpressing Cells and Tumors. Mol Cancer Ther 16: 1021-1030.

Zhang, H., H. Xu, C.R. Ashby, Jr, Y.G. Assaraf, Z.S. Chen, and H.M. Liu. (2020). Chemical molecular-based approach to overcome multidrug resistance in cancer by targeting P-glycoprotein (P-gp). Med Res Rev. [Epub: Ahead of Print]

Zhang, H., J.P. Herman, H. Bolton, Jr, Z. Zhang, S. Clark, and L. Xun. (2007). Evidence that bacterial ABC-type transporter imports free EDTA for metabolism. J. Bacteriol. 189: 7991-7997.

Zhang, H., W. Zhang, S. Huang, P. Xu, Z. Cao, M. Chen, and X. Lin. (2022). The potential role of plasma membrane proteins in response to Zn stress in rice roots based on iTRAQ and PRM under low Cd condition. J Hazard Mater 429: 128324. [Epub: Ahead of Print]

Zhang, H.H., D.R. Blanco, M.M. Exner, E.S. Shang, C.I. Champion, M.L. Phillips, J.N. Miller, and M.A. Lovett. (1999). Renaturation of recombinant Treponema pallidum rare outer membrane protein 1 into a trimeric, hydrophobic, and porin-active conformation. J. Bacteriol. 181: 7168-7175.

Zhang, L. and T.F. Mah. (2008). Involvement of a novel efflux system in biofilm-specific resistance to antibiotics. J. Bacteriol. 190: 4447-4452.

Zhang, L., S. Zhang, W. Zou, Y. Hu, Y. Gao, J. Zhang, and J. Zheng. (2024). Maternal high-fat diet regulates offspring hepatic ABCG5 expression and cholesterol metabolism via the gut microbiota and its derived butyrate. Clin Sci (Lond) 138: 1039-1054.

Zhang, S., X. Lu, X. Fang, Z. Wang, S. Cheng, and J. Song. (2022). Cigarette smoke extract combined with LPS reduces ABCA3 expression in chronic pulmonary inflammation may be related to PPARγ/ P38 MAPK signaling pathway. Ecotoxicol Environ Saf 244: 114086.

Zhang, W., Z. Zhang, Y. Zhang, and A.P. Naren. (2017). CFTR-NHERF2-LPA₂ Complex in the Airway and Gut Epithelia. Int J Mol Sci 18:.

Zhang, W.K., D. Wang, Y. Duan, M.M. Loy, H.C. Chan, and P. Huang. (2010). Mechanosensitive gating of CFTR. Nat. Cell Biol. 12: 507-512.

Zhang, X., F. Qiu, J. Jiang, C. Gao, and Y. Tan. (2011). Intestinal absorption mechanisms of berberine, palmatine, jateorhizine, and coptisine: involvement of P-glycoprotein. Xenobiotica 41: 290-296.

Zhang, X., Y. Zhang, J. Liu, and H. Liu. (2013). PotD protein stimulates biofilm formation by Escherichia coli. Biotechnol Lett 35: 1099-1106.

Zhang, Y. and V.N. Gladyshev. (2008). Molybdoproteomes and evolution of molybdenum utilization. J. Mol. Biol. 379: 881-899.

Zhang, Y., I. Tatsuno, R. Okada, N. Hata, M. Matsumoto, M. Isaka, K. Isobe, and T. Hasegawa. (2016). Predominant role of msr(D) over mef(A) in macrolide resistance in Streptococcus pyogenes. Microbiology 162: 46-52.

Zhang, Y., S. Huang, W. Zhong, W. Chen, B. Yao, and X. Wang. (2021). 3D organoids derived from the small intestine: An emerging tool for drug transport research. Acta Pharm Sin B 11: 1697-1707.

Zhang, Y., W. Gong, Y. Wang, Y. Liu, and C. Li. (2018). Exploring movement and energy in human P-glycoprotein conformational rearrangement. J Biomol Struct Dyn 1-16. [Epub: Ahead of Print]

Zhang, Y., X. Niu, M. Shi, G. Pei, X. Zhang, L. Chen, and W. Zhang. (2015). Identification of a transporter Slr0982 involved in ethanol tolerance in cyanobacterium Synechocystis sp. PCC 6803. Front Microbiol 6: 487.

Zhang, Y.K., H. Zhang, G.N. Zhang, Y.J. Wang, R.J. Kathawala, R. Si, B.A. Patel, J. Xu, and Z.S. Chen. (2015). Semi-synthetic ocotillol analogues as selective ABCB1-mediated drug resistance reversal agents. Oncotarget 6: 24277-24290.

Zhang, Y.K., Y.J. Wang, Z.N. Lei, G.N. Zhang, X.Y. Zhang, D.S. Wang, S.B. Al-Rihani, S. Shukla, S.V. Ambudkar, A. Kaddoumi, Z. Shi, and Z.S. Chen. (2019). Regorafenib antagonizes BCRP-mediated multidrug resistance in colon cancer. Cancer Lett 442: 104-112.

Zhang, Z., J.N. Feige, A.B. Chang, I.J. Anderson, V.M. Brodianski, A.G. Vitreschak, M.S. Gelfand, and M.H. Saier, Jr. (2003). A transporter of Escherichia coli specific for L- and D-methionine is the prototype for a new family within the ABC superfamily. Arch. Microbiol. 180: 88-100.

Zhao QF., Yu JT., Tan MS. and Tan L. (2015). ABCA7 in Alzheimer's Disease. Mol Neurobiol. 51(3):1008-16.

Zhao, C., W. Haase, R. Tampé, and R. Abele. (2008). Peptide specificity and lipid activation of the lysosomal transport complex ABCB9 (TAPL). J. Biol. Chem. 283: 17083-17091.

Zhao, F., X. Lin, K. Cai, Y. Jiang, T. Ni, Y. Chen, J. Feng, S. Dang, C.Z. Zhou, and Q. Zeng. (2022). Biochemical and structural characterization of the cyanophage-encoded phosphate-binding protein: implications for enhanced phosphate uptake of infected cyanobacteria. Environ Microbiol. [Epub: Ahead of Print]

Zhao, H., J. Lee, and J. Chen. (2022). The hemolysin A secretion system is a multi-engine pump containing three ABC transporters. Cell 185: 3329-3340.e13.

Zhao, Q., C. Wang, C. Wang, H. Guo, Z. Bao, M. Zhang, and P. Zhang. (2015). Structures of FolT in substrate-bound and substrate-released conformations reveal a gating mechanism for ECF transporters. Nat Commun 6: 7661.

Zhao, R.Q., Y. Wen, P. Gupta, Z.N. Lei, C.Y. Cai, G. Liang, D.H. Yang, Z.S. Chen, and Y.A. Xie. (2018). Y, an Epigallocatechin Gallate Derivative, Reverses ABCG2-Mediated Mitoxantrone Resistance. Front Pharmacol 9: 1545.

Zhao, Z.J., X.Y. Gao, J.C. Zeng, S.L. Zhang, X.M. Meng, Y.J. Shen, and X.H. Sheng. (2020). Theoretical Insights into the Cotransport Mechanism of GSH with Anticancer Drugs by MRP1. J Phys Chem B 124: 9803-9811.

Zheng, S., J.I. Nagao, M. Nishie, T. Zendo, and K. Sonomoto. (2017). ATPase activity regulation by leader peptide processing of ABC transporter maturation and secretion protein, NukT, for lantibiotic nukacin ISK-1. Appl. Microbiol. Biotechnol. [Epub: Ahead of Print]

Zheng, W.H., &.#.1.9.7.;. Västermark, M.A. Shlykov, V. Reddy, E.I. Sun, and M.H. Saier, Jr. (2013). Evolutionary relationships of ATP-Binding Cassette (ABC) uptake porters. BMC Microbiol 13: 98.

Zhong, X. and P.C. Tai. (1998). When an ATPase is not an ATPase: at low temperatures the C-terminal domain of the ABC transporter CvaB is a GTPase. J. Bacteriol. 180: 1347-1353.

Zhou, C., H. Shi, M. Zhang, L. Zhou, L. Xiao, S. Feng, W. Im, M. Zhou, X. Zhang, and Y. Huang. (2021). Structural insight into phospholipid transport by the MlaFEBD complex from P. aeruginosa. J. Mol. Biol. 166986. [Epub: Ahead of Print]

Zhou, T., W. Niu, Z. Yuan, S. Guo, Y. Song, C. Di, X. Xu, X. Tan, and L. Yang. (2018). ABCA1 Is Coordinated with ABCB1 in the Arsenic-Resistance of Human Cells. Appl Biochem Biotechnol. [Epub: Ahead of Print]

Zhu, W., J.E. Arceneaux, M.L. Beggs, B.R. Byers, K.D. Eisenach, and M.D. Lundrigan. (1998). Exochelin genes in Mycobacterium smegmatis: identification of an ABC transporter and two non-ribosomal peptide synthetase genes. Mol. Microbiol. 29: 629-639.

Zhu, Y., G.H. Lu, Z.W. Bian, F.Y. Wu, Y.J. Pang, X.M. Wang, R.W. Yang, C.Y. Tang, J.L. Qi, and Y.H. Yang. (2017). Involvement of LeMDR, an ATP-binding cassette protein gene, in shikonin transport and biosynthesis in Lithospermum erythrorhizon. BMC Plant Biol 17: 198.

Zhu, Y., S.J. Chu, Y.L. Luo, J.Y. Fu, C.Y. Tang, G.H. Lu, Y.J. Pang, X.M. Wang, R.W. Yang, J.L. Qi, and Y.H. Yang. (2017). Involvement of LeMRP, an ATP-binding cassette transporter, in shikonin transport and biosynthesis in Lithospermum erythrorhizon. Plant Biol (Stuttg). [Epub: Ahead of Print]

Zhu, Y., W. Qiu, Y. Li, J. Tan, X. Han, L. Wu, Y. Jiang, Z. Deng, C. Wu, and R. Zhuo. (2021). Quantitative proteome analysis reveals changes of membrane transport proteins in Sedum plumbizincicola under cadmium stress. Chemosphere 287: 132302. [Epub: Ahead of Print]

Zhu, Y., X. Xing, F. Wang, L. Chen, C. Zhong, X. Lu, Z. Yu, Y. Yang, Y. Yao, Q. Song, S. Han, Z. Liu, and P. Zhang. (2024). The ATP-bound inward-open conformation of ABCC4 reveals asymmetric ATP binding for substrate transport. FEBS Lett. 598: 1967-1980.

Ziegler, J., S. Schmidt, N. Strehmel, D. Scheel, and S. Abel. (2017). Arabidopsis Transporter ABCG37/PDR9 contributes primarily highly oxygenated Coumarins to Root Exudation. Sci Rep 7: 3704.

Zoghbi ME. and Altenberg GA. (2014). ATP binding to two sites is necessary for dimerization of nucleotide-binding domains of ABC proteins. Biochem Biophys Res Commun. 443(1):97-102.

Zolnerciks, J.K., B.G. Akkaya, M. Snippe, P. Chiba, A. Seelig, and K.J. Linton. (2014). The Q loops of the human multidrug resistance transporter ABCB1 are necessary to couple drug binding to the ATP catalytic cycle. FASEB J. 28: 4335-4346.

Zolnerciks, J.K., E.J. Andress, M. Nicolaou, and K.J. Linton. (2011). Structure of ABC transporters. Essays Biochem 50: 43-61.

Zou, L., J. Pottel, N. Khuri, H.X. Ngo, Z. Ni, E. Tsakalozou, M.S. Warren, Y. Huang, B.K. Shoichet, and K.M. Giacomini. (2020). Interactions of Oral Molecular Excipients with Breast Cancer Resistance Protein, BCRP. Mol Pharm. [Epub: Ahead of Print]

Zou, Y., W. Gao, H. Jin, C. Mao, Y. Zhang, X. Wang, D. Mei, and L. Zhao. (2023). Cellular Uptake and Transport Mechanism of 6-Mercaptopurine Nanomedicines for Enhanced Oral Bioavailability. Int J Nanomedicine 18: 79-94.

Zuber, R., M. Norum, Y. Wang, K. Oehl, N. Gehring, D. Accardi, S. Bartozsewski, J. Berger, M. Flötenmeyer, and B. Moussian. (2018). The ABC transporter Snu and the extracellular protein Snsl cooperate in the formation of the lipid-based inward and outward barrier in the skin of Drosophila. Eur J. Cell Biol. 97: 90-101.

Zutz, A., J. Hoffmann, U.A. Hellmich, C. Glaubitz, B. Ludwig, B. Brutschy, and R. Tampé. (2011). Asymmetric ATP hydrolysis cycle of the heterodimeric multidrug ABC transport complex TmrAB from Thermus thermophilus. J. Biol. Chem. 286: 7104-7115.



3.A.1.1 The Carbohydrate Uptake Transporter-1 (CUT1) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.1.1

Maltooligosaccharide porter. The 3-D structure has been reported by Oldham et al. (2007). An altering access mechanism has been suggested for the maltose transporter resulting from rigid-body rotations (Khare et al., 2009). The maltose-binding protein is open in the catalytic transition state for ATP hydrolysis during maltose transport (Austermuhle et al. 2004). Bordignon et al. (2010) and Schneider et al. (2012) reviewed the extensive knowledge available on MalEFGK2, its mode of action and its regulatory interactions.  The transporter sequesters the MalT transcriptional activator at the cytoplasmic surface of the membrane in the absence of the transport substrate (Richet et al. 2012).  The crystal structures of the transporter complex MBP-MalFGK2 bound with large malto-oligosaccharide in two different conformational states have also been determined. In the pretranslocation structure, Oldham et al. 2013 found that the transmembrane subunit MalG forms two hydrogen bonds with malto-oligosaccharide at the reducing end. In the outward-facing conformation, the transmrembrane subunit MalF binds three glucosyl units from the nonreducing end. These structural features explain why large modified malto-oligosaccharides are not transported by MalFGK2 despite their high binding affinity to MBP. In the transport cycle, substrate is channeled from MBP into the transmembrane pathway with a polarity such that both MBP and MalFGK2 contribute to the overall substrate selectivity of the system (Oldham et al. 2013).  Stabilization of the semi-open MalK2 conformation by maltose-bound MBP is key to the coupling of maltose transport to ATP hydrolysis in vivo, because it facilitates the progression of the MalK dimer from the open to the semi-open conformation, from which it can proceed to hydrolyze ATP (Alvarez et al. 2015). Both the binding of MalE to the periplasmic side of the transmembrane complex and binding of ATP to MalK2 are necessary to facilitate the conformational change from the inward-facing state to the occluded state, in which MalK2 is completely dimerized (Hsu et al. 2017). An integrated transport mechanism of the maltose ABC importer has been proposed (Mächtel et al. 2019).

Proteobacteria

MalEFGK of E. coli
MalE (receptor [R])
MalF (membrane [M])
MalG (membrane [M])
MalK (cytoplasmic [C])

 
3.A.1.1.10

Alginate (MW 27,000 Da) (and Alginate oligosaccharides) uptake porter. ABC transport system, AlgQ1AlgM1AlgM2(AlgS)2: AlgS, 363 aas, BAB03314; AlgQ1, 502 aas, 3VLW_A;  AlgM1, 324 aas, BAB03315.1; AlgM2, 293 aas, BAB03316.1. Sphingomonas species A1 is a 'pit-forming' bacterium that directly incorporates alginate into its cytoplasm through a pit-dependent transport system, termed a 'superchannel' (Murata et al., 2008). The pit is a novel organ acquired through the fluidity and reconstitution of cell surface molecules, and through cooperation with the transport machinery in the cells. It confers upon bacterial cells a more efficient way to secure and assimilate macromolecules (Murata et al., 2008).  The substrate-transport characteristics and quaternary structure of AlgM1M2SS with AlgQ1 have been determined (Maruyama et al. 2015). The addition of poly- or oligoalginate enhanced the ATPase activity of reconstituted AlgM1M2SS coupled with one of the periplasmic solute-binding proteins, AlgQ1 or AlgQ2. External fluorescence-labeled oligoalginates were specifically imported into AlgM1M2SS-containing proteoliposomes in the presence of AlgQ2, ATP, and Mg2+. The crystal structure of AlgQ2-bound AlgM1M2SS adopts an inward-facing conformation. The interaction between AlgQ2 and AlgM1M2SS induces the formation of an alginate-binding tunnel-like structure accessible to solvent. The translocation route inside the transmembrane domains contains charged residues suitable for the import of acidic saccharides (Maruyama et al. 2015). 

This bacterium  is a Gram-negative rod, containing glycosphingolipids in the cell envelope, and is named Sphingomonas sp. strain A1 (Murata et al. 2022). The pit was dynamic, with repetitive opening and closing during growth on alginate, and directly included alginate concentrated around the pit, particularly by flagellins, alginate-binding proteins, localized on the cell surface. Alginate incorporated into the periplasm was subsequently transferred to the cytoplasm by cooperative interactions of periplasmic solute-binding proteins and an ATP-binding cassette transporter in the cytoplasmic membrane. The mechanisms of assembly, functions, and interactions between the above-mentioned molecules were clarified using structural biology. The pit was transplanted into other strains of sphingomonads, and the pitted recombinant cells were effectively applied to the production of bioethanol, bioremediation for dioxin removal (Murata et al. 2022). The outer membranes of Sphingomonas strains contain GSL and is different from that of other Gram-negative bacteria, which contain LPSs in their cell envelope. Because of this property, the cell surface of Sphingomonas strains is more hydrophobic than that of other Gram-negative bacteria and shows high affinity toward hydrophobic chemicals such as dioxin and polypropylene glycol.

Strain A1 cells use polyuronates (alginate and pectin) and their depolymerization products as carbon sources for growth. Glucose and pyruvate can be utilized as carbon sources, but far less efficiently than polyuronates. Strain A1 cells grew well on alginate and oligoalginates with different M/G ratios at pH 6–7, 30 ℃ in aerobic conditions, with a doubling time of approximately 25 min. However, unlike almost all of the alginate-degrading bacteria analyzed to date, the cells of strain A1 contained most of their alginate lyases in the cytoplasm. This means that alginate in the medium has to enter the cells in order to make contact with alginate lyases (Murata et al. 2022).

The morphological characteristics of the cell surface were examined with cells of strain A1 grown in the presence or absence of alginate. The following morphological observations were made (Murata et al. 2022): (i) cells grown on alginate were of two types that always coexisted in the medium: cells with or without a pit, and this feature was not observed in the absence of alginate. (ii) The surface of cells grown in the absence of alginate showed a pleat-like structure without a pit. (iii) Cells grown in the presence of alginate produced pits on their cell surface, and The pits contained even globular particles, some of which were insoluble forms (granules) of alginate. (iv) When the alginate-grown cells were treated with ruthenium red, an agent used to stain mucopolysaccharides, the pit periphery was strongly and specifically stained, suggesting that alginate was concentrated in the pit. (v) The thin section of cells grown on alginate showed a specific region where the cell surface sunk into the cells but no such structures were observed in cells grown in the absence of alginate. (vi) The average pit size was 0.02–0.1 µm in diameter (Murata et al. 2022). Thus, (a) the pit is formed only in the presence of alginate, (b) the pit functions as a concentrator of alginate, and (c) strain A1 cells have a pit-dependent alginate assimilation system, which differs from the alginate import and degradation pathway of other alginate-degrading microbes.

There are six protein constituents in the ABC transporter: AlgQ1, Q2, M1 M2 and S (AlgS is present with two copies where Q1 and 2 are periplasmic binding proteins, M1 and M2 comprise the integral membrane transport channel, and S is the dimeric ATPase. Alginate accumulated in the pit is delivered into the periplasm and then transported to the cytoplasm by this ABC transporter. Alginate is finally degraded into constituent monosaccharides by alginate lyases present in the cytoplasm. The gene cluster encoding these proteins are AlgO (regulatory protein)-AlgS-AlgM1-AlgM2-AlgQ1-AlgQ2. There are 8 cell surface proteins, p1 - p8.  P1 - p4 are TonB-dependent outer membrane transporters; p5 and p6 are flagellin-like proteins with alterred central domains of ~150 aas and high affinity for alginate (Kd = 10-9), and p7 and p8 are periplasmic alginate binding proteins (Murata et al. 2022).

Proteobacteria

AlgSM1M2Q1 of Sphingomonas sp.A1
AlgS (C)
AlgM1 (M)
AlgM2 (M)
AlgQ1 (R)
AlgQ2 (R)

 
3.A.1.1.11

Saturated and unsaturated oligogalacturonide transporter, TogMNAB (transports di- to tetrasaccharide pectin degradation products which consist of D-galacuronate, sometimes with 4-deoxy-L-threo-5-hexosulose uronate at the reducing end of the oligosaccharide) (Hugouvieux-Cotte-Pattat et al. 2001). Regulated by pectin utilization regulator KdgR (Rodionov et al. 2004)

Proteobacteria

Oligogalacturonide transporter TogMNAB of Erwinia chrysanthemi
TogM (M)
TogN (M)
TogA (C)
TogB (R)

 
3.A.1.1.12Palatinose (isomaltulose; 6-O-α-D-glucopyranosyl-D-fructose) uptake porterProteobacteriaPalEFGK of Erwinia rhapontici
PalE (R)
PalF (M)
PalG (M)
PalK (C)
 
3.A.1.1.13Glucose, mannose, galactose porterCrenarchaeotaGlcSTUV of Sulfolobus solfataricus
GlcS (R)
GlcT (M)
GlcU (M)
GlcV (C)
 
3.A.1.1.14Arabinose, fructose, xylose porterCrenarchaeotaAraSTUV of Sulfolobus solfataricus
AraS (R)
AraT (M)
AraU (M)
AraV (C)
 
3.A.1.1.15Trehalose porterCrenarchaeotaTreSTUV of Sulfolobus solfataricus
TreS (R)
TreT (M)
TreU (M)
TreV (C)
 
3.A.1.1.16Maltooligosaccharide porter (Maltose is not a substrate, but maltotriose is.)EuryarchaeotaPF1933, 1936, 1937, 1938 of Pyrococcus furiosus
PF1938 (R)
PF1937 (M)
PF1936 (M)
PF1933 (C)
 
3.A.1.1.17Trehalose/maltose/sucrose porter (trehalose inducible)ProteobacteriaThuEFGK of Sinorhizobium meliloti
ThuE (R)
ThuF (M)
ThuG (M)
ThuK (C)
 
3.A.1.1.18N-Acetylglucosamine/N,N'-diacetyl chitobiose porter (NgcK (C) not identified)ActinobacteriaNgcEFG of Streptomyces olivaceoviridis
NgcE (R)
NgcF (M)
NgcG (M)
 
3.A.1.1.19Platinose (isomaltulose) (6-O-α-D-glucopyranosyl-D-fructofuranose) porterProteobacteriaPalEFGK of Agrobacterium tumefaciens
PalE (R)
PalF (M)
PalG (M)
PalK (C)
 
3.A.1.1.2

The galactooligosaccharide (transports the di, tri and tetrasaccharides) uptake porter GanOPQ (GanSPQ; MalEFG) functioning with the energizing ATPase MsmX (see 3.A.1.1.26).  These oligosaccharides are derived from galactans and arabinogalactans, compenents of pectins in plant cell walls (Watzlawick et al. 2016).

Bacteria

GanOPG of Bacillus subtilis
YufK, GanO or GanS (R) (O07009)
YufL or GanP (M) (O32261)
YufM or GanQ (M) (O07011)

MsmX (C) (see 3.A.1.1.26)

 
3.A.1.1.20The fructooligosaccharide porter, MsmEFGK (Barrangou et al., 2003) BacteriaMsmEFGK of Lactobacillus acidophilus
MsmE (R) AAO21856
MsmF (M) AAO21857
MsmG (M) AAO21858
MsmK (C) AAO21860
 
3.A.1.1.21The xylobiose porter; BxlEFG(K) (Tsujibo et al., 2004)BacteriaBxlEFGK of Streptomyces thermoviolaceus
BxlE (R) CAB88161
BxlF (M) CAB88162
BxlG (M) CAB88163
BxlK (C) Unknown
 
3.A.1.1.22

The maltose, maltotriose, mannotetraose (MalE1)/maltose, maltotriose, trehalose (MalE2) porter (Nanavati et al., 2005). For MalG1 (823aas) and MalG2 (833aas), the C-terminal transmembrane domain with 6 putative TMSs is preceded by a single N-terminal TMS and a large (600 residue) hydrophilic region showing sequence similarity to MLP1 and 2 (9.A.14; e-12 & e-7) as well as other proteins.

Thermotogae

MalE1E2FGK of Thermotoga maritima
MalE1 (R) (binds maltose, maltotriose and mannotetraose) (AAD36279)
MalE2 (R) (binds maltose, maltotriose and trehalose) (AAD36901)
MalF1 (M) (AAD36278)
MalG1 (M) (AAD36277)
[MalG2 (M) (AAD36899]
MalK (C) (AAD36351)

 
3.A.1.1.23

The cellobiose/cellotriose (and possibly higher cellooligosaccharides), CebEFGMsiK [MsiK functions to energize several ABC transporters including those for maltose/maltotriose and trehalose] (Schlösser et al., 1997, Schlösser et al., 1999)

Bacteria

CebEFGMsiK of Streptomyces reticuli
CebE (R) (CAB46342)
CebF (M) (CAB46343)
CebG (M) (CAB46344)
MsiK (CAA70125)

 
3.A.1.1.24The glucose/mannose porter TTC0326-8 plus MalK1 (ABC protein, shared with 3.A.1.1.25) (Chevance et al., 2006).Bacteria

TTC0326-8 MalK1 of Thermus thermophilus
TTC0326 (M) - Q72KX4
TTC0327 (M) - Q72KX3
TTC0328 (R) - Q72KX2
MalK1 or TTC0211 (C) - Q72L52

 
3.A.1.1.25

The trehalose/maltose/sucrose/palatinose porter (TTC1627-9) plus MalK1 (ABC protein, shared with 3.A.1.1.24) (Silva et al. 2005; Chevance et al., 2006). The receptor (TTC1627) binds disaccharide alpha-glycosides, namely trehalose (alpha-1,1), sucrose (alpha-1,2), maltose (alpha-1,4), palatinose (alpha-1,6) and glucose.  The structures have been solved to a resolution range of 1.6-2.0 Å (Chandravanshi et al. 2019).

D

Bacteria

TTC1627-9 + MalK1 of Thermus thermophilus
TTC1627 (R) (Q72H68)
TTC1628 (M) (Q72H67)
TTC1629 (M) (Q72H66)
MalK1 (TTC0211) (C) (Q72L52)

 
3.A.1.1.26

The maltose porter, MdxEFG and MsmX (Ferreira and Sá-Nogueira, 2010). The MsmX ATPase can function with other receptor-dependent ABC transporters (TC# 3.A.1.1.2 and 3.A.1.1.34). The crystal structure of MsmX provides a framework to understand the molecular basis of the broader specificity of interaction with the transmembrane subunits of these systems (Leisico et al. 2020).

Bacteria

The maltose porter of Bacillus subtilis, MalEFG/MsmX.
MalE (R) - O06989
MalF (M) - O06990
MalG (M) - O06991
MsmX (C) - P94360

 
3.A.1.1.27

Maltose/Maltotriose/maltodextrin (up to 7 glucose units) transporters MalXFGK (MsmK (3.A.1.1.28) can probably substitute for MalK; Webb et al., 2008).

Bacteria

MalXFGK of Streptococcus mutans:
MalX (R) (Q8DT28)
MalF (M) (Q8DT27)
MalG (M) (Q8DT26)
MalK (C) (Q8DT25)

 
3.A.1.1.28

The raffinose/stachyose transporter, MsmEFGK (MalK (3.A.1.1.27) can probably substitute for MsmK; Webb et al., 2008). This system may also transport melibiose, isomaltotriose and sucrose as well as isomaltosaccharides (Russell et al. 1992).

Bacteria

MsmEFGK of Streptococcus mutans:
MsmE (R) (Q00749)
MsmF (M) (Q00750)
MsmG (M) (Q00751)
MsmK (C) (Q00752)

 
3.A.1.1.29Aldouronate transporter, LplA,B,C (Chow et al., 2007)BacteriaLplABC of Paenibacillus sp. JDR-2:
LplA (R)(A9QDR6)
LplB (M)(A9QDR7)
YtcP (M)(A9QDR8)
LplC - not identified
 
3.A.1.1.3

Glycerol-phosphate porter. Transports both glycerol-3-P and glycerol-3-P diesters including glycerophosphocholine but not glycerol-2-P (Yang et al. 2009; Wuttge et al. 2012).  UgpB (the receptor) binds glycerol 3-P with high affinity, but not glycerol 2-P (Wuttge et al. 2012).

Proteobacteria

UgpABCE of E. coli
UgpB (R)
UgpA (M)
UgpE (M)
UgpC (C)

 
3.A.1.1.30

Glucose porter, GtsABCD (del Castillo et al., 2008).  The orthologue of GtsA (receptor) in P. aeruginosa (64% identical to the P. putida GtsA has been biochemically characterized (Stinson et al. 1977) and corresponds to the sequence with UniProt acc# Q9HZ48 (Friedhelm Pfeiffer, personal communication).

Bacteria

The glucose porter of Pseudomonas putida, GtsABCD:
GtsA (R) (Q88P38)
GtsB (M) (Q88P37)
GtsC (M) (Q88P36)
GtsD (C) (Q88P35)

 
3.A.1.1.31

The trehalose-recycling ABC transporter, LpqY-SugA-SugB-SugC (essential for virulence) (Kalscheuer et al., 2010). It is probably involved in the recycling of extracellular or cell wall trehalose released from trehalose-containing molecules (De la Torre et al. 2021).

Bacteria

LpqY-SugA-SugB-SugC of Mycobacterium tuberculosis
LpqY (R) (Q7D8J9)
SugA (M) (O50452)
SugB (M) (O50453)
SugC (C) (O50454)

 

 
3.A.1.1.32The glucosylglycerol uptake transporter (functions in osmoprotection and also transports sucrose and trehalose (Mikkat and Hagemann, 2000) (most similar to 3.A.1.1.8).BacteriaGgtABCD of Synechocystis sp. strain PCC6803
GgtA (C) (Q55035)
GgtB (R) (Q55471)
GtC (M) (Q55472)
GgTD (M) (Q55473)
 
3.A.1.1.33

The N,N'-diacetylchitobiose uptake transporter, DasABC/MsiK (MsiK energizes several ABC transporters (see 3.A.1.1.23)) (Saito et al., 2008).

Bacteria

DasABC MsiK of Streptomyces coelicolor
DasA (R) (Q8KN19)
DasB (M) (Q8KN18)
DasC (M) (Q8KN17)
MsiK (C) (Q9L0Q1)

 
3.A.1.1.34

The arabinosaccharide transporter AraNPQMsmX. Transports α-1,5-arabinooligosaccharides, at least up to four L-arabinosyl units; the key transporter for α-1,5-arabinotriose and α-1,5-arabinotetraose, but not for α-1,5-arabinobiose which is transported by AraE. MsmX is also used by the MdxEFG-MsmX system (3.A.1.1.36) (Ferreira and Sá-Nogueira, 2010). Involved in the uptake of pectin oligosaccharides with either MsmX or YurJ as the ATPase (Ferreira et al. 2017).

Bacteria

AraNPQ-MsmX of Bacillus subtilis 
AraN (R) (P94528) 
AraP (M) (P94529)
AraQ (M) (P94530)
MsmX (C) (P94360) 

 
3.A.1.1.35

Glycerol uptake porter, GlpSTPQV (Ding et al., 2012).

  α-proteobacteria

GlpSTPQV of Rhizobium leguminosarum 
GlpS (C) (G3LHY8)
GlpT (C) (G3LHY9)
GlpP (M) (G3LHZ0)
GlpQ (M) (G3LHZ1)
GlpV (R) (G3LHZ3) 

 
3.A.1.1.36

Putative transport system

Actinobacteria



Q93J94 (R)
Q93J93 (M)
Q93J92 (M)
Q9L0Q1 (C?)

 
3.A.1.1.37

Predicted arabinoside porter. Regulated by arabinose-responsive regulator AraR (Rodionova et al. 2012).

Thermotogae

AraEFG of Thermotoga maritima
AraE (R) (TM0277) -
AraF (M) (TM0278) Q9WYB4
AraG (M) (TM0279) Q9WYB5

 
3.A.1.1.38

Inositol phosphate porter (Rodionova et al. 2013). Binds inositol phosphate with low Kd and inositol with a lower affinity.

Thermotogae

InoEFGK of Thermotoga maritima
InoE (R) TM0418 (Q9WYP9)
InoF (M) TM0419 (Q9WYQ0)
InoG (M) TM0420 (Q9WYQ1)
InoK (C) TM0421 (Q9WYQ2)

 
3.A.1.1.39

Alpha-1,4-digalacturonate porter (Nanavati et al., 2006). Regulated by pectin utilization regulon UxaR (Rodionova et al. 2012).

Thermotogae

AguEFG of Thermotoga maritima
AguE (R) (TM0432) (Q9WYR3)
AguF (M) (TM0431) (Q9WYR2)
AguG (M) (TM0430) (Q9WYR1)

 
3.A.1.1.4Lactose porterProteobacteriaLacEFGK of Agrobacterium radiobacter
LacE (R)
LacF (M)
LacG (M)
LacK (C)
 
3.A.1.1.40

Predicted chitobiose porter. Regulated by chitobiose-responsive regulator ChiR (Kazanov et al., 2012).

Thermotogae

ChiEFG of Thermotoga maritima
ChiE (R) (TM0810) (Q9WZR7)
ChiF (M) (TM0811) (Q9WZR8)
ChiG (C) (TM0812) (Q9WZR9)

 
3.A.1.1.41

Trehalose porter. Also binds sucrose (Boucher and Noll, 2011). Induced by glucose and trehalose. Directly regulated by trehalose-responsive regulator TreR (Kazanov et al., 2012).

Thermotogae

TreG (M) (ThemaDRAFT_1378) G4FGN6

TreF (M) (ThemaDRAFT_1379) G4FGN7

TreE (R) (ThemaDRAFT_1380) G4FGN8

 
3.A.1.1.42

α-glucoside uptake permease, Agl3E/Agl3F/Agl3G. Plays a role in normal morphogenesis and antibiotic production. Strongly induced by trehalose and melibiose, and weakly induced by lactose and glycerol but not glucose (Hillerich and Westpheling 2006).The operon is controlled by a GntR homologue, Agl3R, and downstream of the gntR gene is a gene encoding an extracellular carbohydrase.

Actinobacteria

Agl2E/3F/3G of Streptomyces coelicolor
Agl3E (R); 425aas (Q9FBS5)
Agl3F (M) 6TMSs; 310aas (Q9FBS6)
Agl3G (M) 7TMSs; 303aas (Q9FBS7)
(ABC protein (C) not identified) 

 
3.A.1.1.43

Agl3E, Agl3F and Agl3G ABC porter. Induced by trehalose and melibiose using a GntR-like transcription factor (Hillerich and Westpheling 2006).  The ATPase subunit, Agl3K, may be the MsiK (Sco4240; see 3.A.1.1.33) protein (Saito et al. 2008).

Actinobacteria

Agl3EFG (Sco7167-5) of Streptomyces coelicolor.
Agl3E (R)
Agl3F (M)
Agl3G (M)
Agl3K (unknown)  

 
3.A.1.1.44

MalEFG (K unknown), involved in maltose and maltodextrin uptake (van Wezel et al. 1997).  The MalK protein may be the MsiK (Sco4240; Q9L0Q1; see 3.A.1.1.33) protein.

Actinobacteria

MalEFG (Sco2231-29) of Streptomyces coelicolor.
MalE (R)
MalF (M)
MalG (M)
 

 
3.A.1.1.45

Maltose transporter, MusEFGKI.  All five genes have been reported to be essential for uptake activity (Henrich et al. 2013).  The MusI gene product is of 215 aas with 5 TMSs and comprises the founding member of a distinct family of poorly characterized protein in TC family 9.B.28. 

Actinobacteria

MusEFGKI of Corynebacterium glutamicum

 
3.A.1.1.46

Probable glucoside uptake porter, YcjNOPV.  Encoded in an operon or gene cluster with a glucosyl hydrolase and two oxidoreductases (Moussatova et al. 2008).

Proteobacteria

YcjNOPV of E. coli
YcjN (R) (430 aas)
YcjO (M) (293 aas)
YcjP (M) (280 aas)
YcjV (C) (360 aas)

 
3.A.1.1.47

ABC-type fucose uptake porter FucABCD.  The ATPase subunit, FucD, has not been identified (Manzoor I., Shafeeq S., Afzal M. and Kuipers OP, JMMB, in press, 2015).

Firmicutes

FucABCD of Streptococcus pneumoniae
FucA, (R)
FucB, (M)
FucC, (M)

 
3.A.1.1.48

The lacto-N-biose I (LNB; Gal β-1,3-GlcNAc)/galacto-N-biose (GNB; Gal β-1,3-GalNAc) transporter.  The solute-binding protein crystallizes only in the presence of LNB or GNB, and it was therefore named GNB/LNB-binding protein (GL-BP) (Wada et al. 2007; Suzuki et al. 2008; Asakuma et al. 2011). Isothermal titration calorimetry measurements revealed that GL-BP specifically binds LNB and GNB with K(d) values of 0.087 and 0.010 μM, respectively, and the binding process is enthalpy-driven. The crystal structures of GL-BP complexed with LNB, GNB, and lacto-N-tetraose (Galbeta1-3GlcNAcbeta1-3Galbeta1-4Glc) were determined.  The MalF and MalG membrahe proteins are encoded adjacent to the gene for GL-BP, but the ATPase was not identified.

Actinobacteria

The LNB/GNB uptake transporter of Bifidobacterium longum
MalE homologue
MalF homologue
MalG homologue
MalK homologue, not identified.

 
3.A.1.1.49

The polyol (mannitol, glucitol (sorbitol), arabitol (arabinitol; lyxitol)) uptake porter, MtlEFGK (Brünker et al. 1998).

MtlEFGK of Pseudomonas fluorescens
MtlE, R, 436 aas
MtlF, M, 296 aas
MtlG, M, 276 aas
MtlK, C, 367 aas

 
3.A.1.1.5Hexitol (glucitol; mannitol) porterProteobacteriaSmoEFGK of Rhodobacter sphaeroides
SmoE (R)
SmoF (M)
SmoG (M)
SmoK (C)
 
3.A.1.1.50

Probable glycerophosphocholine (GPC) uptake porter (Chandravanshi et al. 2016). The system may include a receptor and three membrane proteins (of 378 aas and 6 TMSs, 299 aas and 7 TMSs, and 113 aas and 3 TMSs (?). The ATPase has not been identified.

GPC uptake porter of Thermus thermophilus

 
3.A.1.1.51

Maltose - maltoheptaose transporter, MalEFGK.  MalEF is a R-M fusion protein with the MalE domain N-terminal and the MalF domain C-terminal. The protein, of 733 aas, has 8 TMSs, one N-terminal to MalE (a signal sequence for export of the MalE domain to the periplasm), an extra TMS at the N-terminus to bring the N-terminus to the periplasmic side of the inner membrane, and then the usual 6 TMSs observed for many ABC membrane proteins.  MalG (M, 272 aas, 6 TMSs) and MalK (C, 374 aas) are of normal size and composition. While MalE of E. coli was able to additionally increase ATPase activity of MalFGK2 in vitro, the isolated MalE domain of B. bacteriovorus failed to stimulate the E. coli system (Licht et al. 2018). The adjacent genes are an α-amylase (Q6MNM3) and a glucokinase (Q6MNM4).

MalEF/MalG/MalK of Bdellovibrio bacteriovorus
MalEF, R-M, 733 aas, 8 TMSs (Q6MNM0)
MalG, M, 272 aas, 6 TMSs (Q6MNM1)
MalK, C, 374 aas, (Q6MNM2)

 
3.A.1.1.52

Sugar (sucrose, maltose, glucose, fructose, esculin (coumarin β-glucoside)) uptake system possibly consisting of 5 or 6 proteins (see below) (Nieves-Morión and Flores 2017). These proteins are all implicated in sugar uptake, but they may include components of multiple transporters. The system may also regulate formation of septal nanopores (Flores et al. 2018).

Sugar uptake porter of Nostoc (Anabaena) strain PCC7120
GlsR, MalE-like, All1916, 418 aas and 1 N-terminal TMS (R) (Q8YVQ8)
GlsQ, MalF-like, Alr2532, 301 aas and 6 TMSs (M) (Q8YU29)
GlsP, MalG-like, All0261, 276 aas and 6 TMSs (M) (Q8Z042)
GlsC, MalK-like, Alr4781, 432 aas and 0 TMSs (C) (Q8YMZ3)
GlsD, MalK-like, All1823, 366 aas and 0 TMSs (C) (Q8YVZ3)

 
3.A.1.1.53

Oligosaccharide transporter RafEFGK. RafE, the binding protein, has be extensively characterized.  It binds α-(1,6)-linked glucosides and galactosides of varying size, linkage, and monosaccharide composition with preference for the trisaccharides raffinose and panose. This preference is reflected in the α-(1,6)-galactoside uptake profile of the bacterium. Structures of RafE (BlG16BP) in complex with raffinose and panose revealed the basis for the ligand binding plasticity, which recognizes the non-reducing α-(1,6)-diglycosidic linkages in its ligands (Ejby et al. 2016). RafK has not be identified experimentally, but it may be NCIB protein acc# WP_022543180.1, ATP binding protein, annotated as UgpC, and this protein has been enterred into TCDB as RafK. Sugar binding substrates of RafE include: raffinose (highest affinity), panose, melibiose, stachyose, verbascose, isomaltose, isomaltotriose, isomaltotetraose, isomaltopentaose, isomaltohexaose, and isomaltoheptaose (Ejby et al. 2016).

RafEFGK of Bifidobacterium animalis
RafE, (R) D3R799; 439 aas and 1 N-terminal TMS
RafF, (M) D3R798;  330 aas and 6 TMSs
RafG, (M) D3R797; 301 aas and 6 TM
RafK (C) WP_022543180.1, 377 aas and 0 TMSs.

 
3.A.1.1.54

Putative ABC sugar uptake porter with 4 constituents which unlike other members of this subfamily, has two large membrane proteins of 16 - 18 TMSs.

ABC porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)
OLS24721 R, 427 aas
OLS20027, M, 641 aas and 16 putative TMSs
OLS20028, M, 779 aas and 18 putative TMSs
OLS20029, C, 379 aa

 
3.A.1.1.55

Four comoponent ABC uptake porter, possibly transporting mannosyl glycerate.  The four components of this system and a potential mannosyl glycerate hydrolase are encoded within a single operon.

Putative mannosy glycerate transporter
R, 428 aas and 1 N-terminla TMS, D7BAR7
M, 281 aas and 6 TMSs, D7BAR6
M, 266 aas and 6 TMSs, D7BAR5
C, ATPase, cytoplasmic, D7BAR4

 
3.A.1.1.56

Uptake transport system for L-arabinose and D-xylose, XacGHIJK (Johnsen et al. 2019). The system has a receptor, two 6 TMS membrane proteins and two ATPases.  xacGHIJK is upregulated by growth in the presence of either D-xylose or L-arabinose, mediated by the transcriptional regulator, XacR, the general regulator of xac catabolic genes (Johnsen et al. 2019).

XacGHIJK of Halofax volcanii

 
3.A.1.1.57

ABC uptake porter consisting of AbnE (R), AbnF (M) and AbnJ (M).  The ATPase (C) has not been identified and does not appear to be encoded within the same gene cluster. This gene cluster also encodes an extracellular arabinanase, an intracellular arabinofuranosidase, and many other enzymes of arabinose/pentose metabolism as well as a sensor kinase/response regulator and another ABC transporter, probably specific for arabinose (TC# 3.A.1.2.32).

AbnEF(G?)J of Geobacillus stearothermophilus (Bacillus stearothermophilus)
AbnE, R, B3EYM9
AbnF, M, B3EYN0
AbnG, C, ?
AbnJ, M, B3EYN1

 

 
3.A.1.1.58

Four component putative ABC transporter specific for N-acetylglucosamine.

Putative N-acetylglucosamine transporter of Rhizobium meliloti (strain 1021) (Ensifer meliloti) (Sinorhizobium meliloti)
ATPase (C) of 352 aas
Membrane protein (M) of 279 aas and 6 TMSs
Membrane protein (M) of 313 aas and 6 TMSs
Binding receptor of 419 aas and 1 N-terminal TMS

 
3.A.1.1.59

Probable ABC transport system for the uptake of oligosaccharides (including a tetrasaccharide) of glucose, galactose and N-acetylglucosamine. The two membrane proteins are of 320 and 317 aas, both with 6 TMSs, and an extracytoplasmic binding receptor; the ATPase has not been identified.

ABC transporter of Bifidobacterium longum subsp. infantis

 
3.A.1.1.6Cyclodextrin porterProteobacteriaCymDEFG of Klebsiella oxytoca
CymE (R)
CymF (M)
CymG (M)
CymD (C)
 
3.A.1.1.60

UspABC putative sugar uptake transporter, that probably imports peptidoglycan precursors (Karlikowska et al. 2021).

UspABC of Mycolicibacterium smegmatis (Mycobacterium smegmatis)
UspA, M, I7GC87, 290 aas and 6 TMSs
UspB, C, I7G593, 275 aas
UapC or MdxE, R, I7FQ33, 430 aas and 1 TMS

 
3.A.1.1.61

ABC sugar uptake porter with four protein components, 1 C, 2 M, and 1 R. This system has been found only in pathogenic Mycobacterium species (De la Torre et al. 2021).

Rv2038c - 0Rv2041c of Mycobacterium tuberculosis
Rv2038c, O53482, C, 357 aas
Rv2039c, O53483, M, 280 aas and 6 TMSs
Rv2080c, O53484, M, 300 aas and 6 TMSs
Rv2081c, O53485, R, 439 aas and 1 TMS

 
3.A.1.1.62

MdxEFG/MsmX maltose/cyclodextrin uptake system of the ABC-type. This system transports (takes up) maltose and malto-cyclodextrins.  The binding specificity of MdxE and its role in the cyclodextrin import in Thermoanaerobacterales has been determined (Aranda-Caraballo et al. 2023).

MdxEFG/MsmX of Thermoanaerobacterium xylanolyticum
MdxE, 420 aas and 1 N-terminal TMS
(F6BI02)
MdxF, 306 aas and 6 TMSs (F6BI03)
MdxG, 293 aas and 6 TMSs (F6BI04)
MsmX, 371 aas and 0 TMSs (F6BHH9)

 
3.A.1.1.7Maltose/trehalose porterEuryarchaeotaMalEFGK of Thermococcus litoralis
MalE (R)
MalF (M)
MalG (M)
MalK (C) (not sequenced)
 
3.A.1.1.8Sucrose/maltose/trehalose porter (sucrose-inducible)ProteobacteriaAglEFGK of Sinorhizobium meliloti
AglE (R)
AglF (M)
AglG (M)
AglK (C)
 
3.A.1.1.9

The oligosaccharide (glucuronate-linked to a xylo-oligosaccharide) ABC uptake porter, GuoEFGK in AguEFGK. GuoE binds with high affinity a four sugar aldotetrouronic
acid [2-O-α-(4-O-methyl-α-D-glucuronosyl)-xylotriose] (Shulami et al., 1999; S.Shulami, personal communication)

Bacteria

GuoEFGK of Geobacillus stearothermophilus
AguE or GuoE (R) (C9RT46)
AguF or GuoF (M) (Q09LY7)
AguG or GuoG (M) (Q09LY6)
AguK or GuoK (C) (not identified)

 


3.A.1.10 The Ferric Iron Uptake Transporter (FeT) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.10.1

Ferric iron (Fe3+) porter, SfuABC (Angerer et al. 1990). These iron uptake systems have been reviewed (Angerer et al. 1992). Delepelaire 2019 reviewed structural and functional aspects of iron ABC transporters with emphasis on their substrate binding proteins.

Proteobacteria

SfuABC of Serratia marcescens
SfuA (R)
SfuB (M)
SfuC (C)

 
3.A.1.10.2Ferric iron (Fe3+) porterCyanobacteriaFut A1A2BC of SynechocystisPCC6803
FutA1 (R)
FutA2 (R)
FutB (M)
FutC (C)
 
3.A.1.10.3

Ferric iron (Fe3+) porter, FbpABC or HitABC (selective for trivalent cations, Fe3+, Ga3+ and Al3+) (Anderson et al., 2004)

Proteobacteria

FbpABC (HitABC) of Haemophilus influenzae
FbpA (R) (AAC21773)
FbpB (M) (AAC21774)
FbpC (C) (AAC21775)

 
3.A.1.10.4

The Fe-hydroxamate-type siderophore uptake porter (transports Fe+3 bound to ferrioxamine, ferrichrome or pyoverdine siderophores) (Vajrala et al., 2010).

Bacteria

NitABC of Nitrosomonas europaea
NitA (R) (Q82VN7)
NitB (M) (Q82VN6)
NitC (C) (Q82VN5)

 
3.A.1.10.5

Siderophore-independent iron uptake system, AfuABC (Saken et al. 2000).

Proteobacteria

AfuABC of Yersinia enterocolitica
AfuA (R)
AfuB (M)
AfuC (C)

 
3.A.1.10.6

Fe3+ uptake porter consisting of 3 subunits, R, 330 aas, M, 516 aas and 12 TMSs, and C, 350 aas (Mandal et al. 2019).

Fe3+ uptake porter of Thermus thermophilus with 3 subunits.

 


3.A.1.101 The Capsular Polysaccharide Exporter (CPSE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.101.1Capsular polysaccharide exporterGram-negative bacteriaKpsMT of E. coli KpsM
KpsM (M) - (P24584)
KpsT (C) - (P24586)
 
3.A.1.101.2Vi polysaccharide exporter, VexBC (Hashimoto et al, 1993).Gram-negative bacteriaVexBC of Salmonella typhi
VexB (M) - (P43109)
VexC (C) - (P43110)
 
3.A.1.101.3

Capsular polysialate exporter, CtrC/D (functions with 1.B.18.2.3 (OMA) and 1.B.4.2.1 (MPA2)) (Larue et al., 2011).

Bacteria

CtrABCD of Neisseria meningitidis
CtrC (M) (B3FHE1)
CtrD (C) (B3FHE0) 

 


3.A.1.102 The Lipooligosaccharide Exporter (LOSE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.102.1Lipooligosaccharide exporter (nodulation proteins, NodIJ)Gram-negative bacteriaNodIJ of Rhizobium galegae
NodJ (M)
NodI (C)
 
3.A.1.102.2

NodIJ nodulation factor transporter.  Involved in lipo-chitooligosaccharide secretion (Fernández-López et al. 1996).

NodIJ of Azorhizobium caulinodans
NodI, C, Q07756
NodJ, M, Q07757

 


3.A.1.103 The Lipopolysaccharide Exporter (LPSE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.103.1Lipopolysaccharide exporterGram-negative bacteriaRfbAB of Klebsiella pneumoniae
RfbA (M)
RfbB (C)
 
3.A.1.103.10

Wzm (M; 280 aas; 6 or 7 TMSs in a 2 + 3 + 1 or 2 TMS arrangement)-Wzt (C) ABC lipid-linked galactan precursor exporter. Deletion of the encoding genes results in severe morphological changes and the accumulation of an aberrantly long galactan precursor. A model for coupled synthesis and export of the galactanpolymer precursor in mycobacteria has been proposed (Savková et al. 2021). The two encoding genes are separated by a gene that encodes a galactofuranosyltransferase, GlfT1.

Wzm-Wzt of Mycobacterium tuberculosis
Wzm, M, P72049 (Rv3783)
Wzt, C, P72047 (Rv3781)

 
3.A.1.103.11

ABC exporter for lipid-linked O-antigen lipopolysaccharide: Wzm (membane constituent; 261 aas with 5 or 6 TMSs) + Wzt (ATPase constituent; 431 aas with 0 TMSs).  This system transports the lipid-linked LPS from the cytoplasm to the periplasm of the bacterial cell (Kelly et al. 2024).

ABC transporter of Klebsiella pneumoniae

 
3.A.1.103.2

Heteropolysaccharide O-antigen exporter, Wzm/Wzt (Feng et al., 2004). The C-terminal cytoplasmic domain of Wzt (an IgG-like β-sandwich) determines the specificity of the transporter for either O8 or O9a O-PS (Cuthbertson et al., 2007). The transporter structure reveals a continuous transmembrane channel in a nucleotide-free state (Caffalette et al. 2019). Upon ATP binding, large structural changes within the nucleotide-binding and transmembrane regions push conserved hydrophobic residues at the substrate entry site towards the periplasm and provide a model for polysaccharide translocation. With ATP bound, the transporter forms a large transmembrane channel with openings toward the membrane and periplasm. The channel's periplasmic exit is sealed by detergent molecules that block solvent permeation. Molecular dynamics simulation data suggest that, in a biological membrane, lipid molecules occupy this periplasmic exit and prevent water flux in the transporter's resting state (Caffalette et al. 2019).

Gram-negative bacteria

Wzm/Wzt of E. coli
Wzm (M) (AAS99164)
Wzt (C) (AAS99165)

 
3.A.1.103.3

ABC transporter required for O-antigen biosynthesis and multicellular development, RfbAB (Guo et al. 1996). Functions with the RfbC glycosyl transferase (TC#4.D.1.3.4). 

Proteobacteria

RfbAB of Myxococcus xanthus 
RfbA (M) 260aas (Q50862)
RfbB (C) 437aas (Q50863) 

 
3.A.1.103.4

RfbAB lipopolysaccharide exporter (Guo et al. 1996).

Proteobacteria

RfbAB of Myxococcus xanthus.
RfbA (MXAN_4623) (M)
RfbB (MXAN_4622) (C) 

 
3.A.1.103.5

ABC transporter mediating ethanol tolerance, Slr0977 (M)/Slr0982 (C) (Zhang et al. 2015).  Present in a gene cluster with (lipo)polysaccharide biosynthetic enzymes, so could be a cell surface carbohydrate export system.

Cyanobacteria

Ethanol tolerance transporter of Synechocystis sp. (strain PCC 6803 / Kazusa)

 
3.A.1.103.6

Two component lipopolysaccharide exporter, Wzm/Wzt.  Wzm is the membrane component (265 aas with 6 TMSs) which forms a ring-like large ion conductance channel. The ATPase, Wzt, functions both as the energizer and regulator (Mohammad et al. 2016).

Wzm/Wzt of Pseudomonas aeruginosa

 
3.A.1.103.7

ABC-type polysaccharide/polyol phosphate export systems, permease componentof 262 aas and 6 or 7 TM

Transporter of Acidovorax sp. MR-S7

 
3.A.1.103.8

ABC transporter of 258 aas and 6 TMSs.

ABC transporter of Moranbacteria bacterium

 
3.A.1.103.9

ABC O-antigen lipopolysaccharide/polysaccharide export transporter, Wzm/Wzt of 253 aas and 6 TMSs (Wzm; also called AbcT3) and 396 aas and 0 TMSs (Wzt). The crystal structure is available (PDB 6AN7) (Bi et al. 2018) for the Wzm-Wzt homologue from Aquifex aeolicus in an open conformation. The transporter forms a transmembrane channel that is sufficiently wide to accommodate a linear polysaccharide. Its nucleotide-binding domain and a periplasmic extension form 'gate helices' at the cytosolic and periplasmic membrane interfaces that probably serve as substrate entry and exit points (Bi et al. 2018). O antigen structures are serotype specific and form extended cell surface barriers endowing many pathogens with survival benefits. In the ABC transporter-dependent biosynthesis pathway, O antigens are assembled on the cytosolic side of the inner membrane on a lipid anchor and reoriented to the periplasmic leaflet by WzmWzt for ligation to the core lipopolysaccharides. This process depends on the chemical modification of the O antigen's nonreducing terminus, sensed by WzmWzt via a carbohydrate-binding domain (CBD) that extends its nucleotide-binding domain (NBD). Caffalette and Zimmer 2021 provided the cryoEM structure of this full-length WzmWzt transporter bound to ATP in a lipid environment, revealing a highly asymmetric transporter organization. The CBDs dimerize and associate with only one NBD. Conserved loops at the CBD dimer interface straddle a conserved peripheral NBD helix. The CBD dimer is oriented perpendicularly to the NBDs and its putative ligand-binding sites face the transporter to likely modulate ATPase activity upon O antigen binding. A closed WzmWzt conformation in which an aromatic belt near the periplasmic channel exit seals the transporter in a resting, ATP-bound state. The sealed transmembrane channel is asymmetric, with one open and one closed cytosolic and periplasmic portal (Caffalette and Zimmer 2021). 

 

Wzm/Wzt of Aquifex aeolicus

 


3.A.1.104 The Teichoic Acid Exporter (TAE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.104.1

Teichoic acid exporter, TagGH.  It is present in a large complex with the teichoic acid precursor synthetic enzymes (Formstone et al. 2008).  The substrate may be the diphospholipid-linked disaccharide portion of the teichoic acid precursor (Schirner et al. 2011).  3-d structural studies have been reported (Ko et al. 2016) showing that TagG and TagH are localized on the cytoplasmic membrane in a patch, and the TMS of TagH is important for normal transport activity (Yamada et al. 2018). The crystal structure of the N-terminal domain of TagH reveals a potential drug targeting site (Yang et al. 2020). The ATPase activity of TagH-N was inhibited by clodronate, a bisphosphonate, in a non-competitive manner, consistent with the proposed wall teichoic acid-binding site for drug targeting (Yang et al. 2020).

Gram-positive bacteria

TagGH of Bacillus subtilis
TagG (M)
TagH (C)

 
3.A.1.104.2

The teichoic acid precursor exporter, TarGH. May be specific for the diphospholipid linked disaccharide portion of the teichoic acid precursor (Schirner et al. 2011). TarG is the target of a small antimicrobial inhibitor of S. aureus growth (Swoboda et al. 2009). TarGH is a WTA transporter and has been purified and reconstituted in proteoliposomes (Matano et al. 2017). They showed that a new compound series inhibits TarH-catalyzed ATP hydrolysis even though the binding site maps to TarG, near the opposite side of the membrane. These are the first ABC transporter inhibitors to block ATPase activity by binding to the transmembrane domain.

Firmicutes

TarGH of Staphylococcus aureus 
TarG (M) (D1GQ18)
TarH (C) (D1GQ17) 

 


3.A.1.105 The Drug Exporter-1 (DrugE1) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.105.1

Daunorubicin, doxorubicin etc. (multidrug resistance) exporter, DrrAB.  DrrB binds drugs with variable affinities and contains multiple drug binding sites. The two asymmetric nucleotide binding sites in DrrA have strikingly different binding affinities. Long-range conformational changes occur between DrrA and DrrB. The transduction pathway from the nucleotide-binding DrrA subunit to the substrate binding DrrB subunit includes the Q-loop and CREEM motifs in DrrA and the EAA-like motif in DrrB (Rahman and Kaur 2018).

Gram-positive bacteria

DrrAB of Streptomyces peucetius
DrrA (C), 330 aas
DrrB (M), 283 aas and 6 TM

 
3.A.1.105.10

AbcG homologue, ABCH1 of 705 aas and 6 TMSs in a C-M arrangement.  May be involved in steroid or drug efflux (Popovic et al. 2010).  Of the vertbrates, it may be restricted to fish. 

Animals

AbcH1 (C-M) of Danio rerio

 
3.A.1.105.11

ABC-2 transporter probably specific for a lantibiotic.  The genes for this system are adjacent to an S2P-M50 peptidase (G0Q3D2), probably involved in pro-lantibiotic processing, as well as a lantibiotic biosynthetic enzyme (G0Q3D1) and a lantibiotic dehydratase (G0Q3D0). 

Actinobacteria

ABC-2/ATPase of Streptomyces griseus
ABC-2 (M) (G0Q3D4)
ATPase (C) (G0Q3D3)

 
3.A.1.105.12

Three component ABC-2 transporter: (1) the membrane (M) subunit with a C-terminal CBS domain, (2) an ABC ATPase subunit and (3) an M50 peptidase (Zn2+-metalopeptidase) of 392 aas and 6TMSs. they may export a bacteriocin, and the protease cleaves off the signal peptide during export. The three encoding genes are in a single gene cluster.

Archaea

ABC transporter
ABC2 (M) (F8D412)
ABC ATPase (C) (F8D413)
M50 peptidese,  (F8D414)

 
3.A.1.105.13

SclAB (Sco4359-60) (Gominet et al. 2011).

Actinobacteria

SclAB of Streptomyces coelicolor.
SclA (C)
SclB (M)

 
3.A.1.105.14

RagAB, involved in both aerial hyphae formation and sporulation (San Paolo et al. 2006).

Actinobacteria

RagAB of Streptomyces coelicolor.
RagA: Sco4075 (C)
RagB: Sco4074 (M) 

 
3.A.1.105.15

Putative drug exporter, YbhFGRS (Moussatova et al. 2008).

Proteobacteria

YbhFGRS of E. coli
YbhF, (C) (578 aas)
YbhG, (MFP) (332 aas)
YbhR, (M) (368 aas)
YbhS, (M) ((377 aas)

 

 
3.A.1.105.16

Putative ABC export system (MDR?), RbbA/YhhJ/YhiI (All three genes are in a single operon; this system may comprise a single ABC exporter with MFP; substrate unknown (Moussatova et al. 2008 and unpublished observations).

Proteobacteria


RbbA/YhhJ/YhiI of E. coli
RbbA (C-M; 911 aas; C8TJS4)
YhhJ (M; 374 aas; P0AGH1)
YhiI (MFP; 355 aas; P37626)

 
3.A.1.105.17

The putative polyketide drug exporter, YadGH.  May also transport phospholipids, participating in phospholipid trafficking together with the Mla complex. It interacts with MlaABCDEF (TC# 3.A.1.27.3) to preserve outer membrane asymmetry (Malinverni and Silhavy 2009; Babu et al. 2018).

Proteobacteria

YadGH of E. coli
YadG (C; 308 aas)
YadH (M, 256 aas)

 
3.A.1.105.19

Poorly characterized ABC exporter involved in bacterial competitiveness and bioflim morphology, YfiLMN (Stubbendieck and Straight 2017).

YfiLMN of Bacillus subtilis
YfiL (C) 311 aas, 0 TMSs
YfiM (M) 296 aas, 6 TMSs
YfiN (N) 385 aas, 6 TMSs

 
3.A.1.105.2Oleandomycin (drug resistance) exporterGram-positive bacteriaOleC4-OleC5 of Streptomyces antibioticus
OleC4 (C)
OleC5 (M)
 
3.A.1.105.20

Putative 5 component ABC exporter with two membrane constituents, two cytoplasmic ATPases, and one membrane fusion protein (truncated at the N-terminus, probably because of an incorrect initiation codon assignment).

5-component ABC exporter of Bdellovibrio bacteriovorus
Q6MLX4 (M)
Q6MLX5 (M)
Q6MLX6 (C)
Q6MLX7 (C)
Q6MLX8 (MFP)

 
3.A.1.105.21

Uncharacterized ABC transporter with two components, a transmembrane protein with 6 TMSs and an ATPase. The substrate in unknown.

ABC system of Candidatus Saccharibacteria bacterium

 
3.A.1.105.22

Uncharacterized protein pair of a presumed ABC transporter. One is of 280 aas and 7 putative TMSs; the other is of 275 aas and 6 putative TMSs. The genes encoding these two proteins map adjacent to each other.  The ATPase has not been identified.

UP of Candidatus Eisenbacteria bacterium RBG_16_71_46 (subsurface metagenome)

 
3.A.1.105.23

Putative ABC exporter with two consitutents, M is of 256 aas and 6 TMSs; C is of 317 aas.  The genes encoding these two proteins are adjacent to each other.

ABC exporter of Armatimonadetes bacterium (groundwater metagenome)

 
3.A.1.105.24

Putative ABC exporter with two membrane constituents encoded by adjacent genes.  The ATPase does not map adjacent to these genes and has not been identified.

Putative ABC exporter of Methanomassiliicoccus sp.

 
3.A.1.105.25

ATP-binding cassette transporter subfamily Gof 687 aas and 7 TMSs in a 1 + 6 TMS arrangement. 13 ABCG genes were identified in N. lugens, and expression levels of these ABCG transporter genes after treatment with thiamethoxam, abamectin, and cyantraniliprole has been examined.  Some increase in amounts while others do not (Yang et al. 2019).

ABCG of Nilaparvata lugens (Brown plant leafhopper)

 
3.A.1.105.26

Putative ABC transporter with two membrane proteins, both with 6 TMSs, one with them in a 2 + 2 + 2 TMS arrangement, the other in a 2 + 3 + 1 TMS arrangement. The two genes encoding these proteins are next to each other on the chromosome.  The ATPase is fused to the first of these two membrane protein domains (acc # C7QI22). These two genes, presumable encoding an ABC exporter, are adjacent to lantibiotic biosynthesis genes. Therefore their function may be to export a lantibiotic. The N-terminal hydrophilic domain of C7QI22 is an S2P-M50-like peptidase (TC# 9.B.149) that may process the pro-lantibiotic during export.

ABC membrane transport proteins of Catenulispora acidiphila

 
3.A.1.105.27

Putative ABC exporter with two protein components, the first, a large protein with an N-terminal membrane domain with 6 TMSs in a 2 + 2 + 2 TMS arrangement, and a C-terminal ABC-type ATPase domain. The second protein is a smaller protein with only a membrane domain with 6 TMSs in a 2 + 3 + 1 TMS arrangement.  The two genes encoding these proteins are adjacent to each other, and are adjacent to a lantibiotic dehydrophenase gene. They may therefore export the newly synthesized lantibiotic.

ABC exporter of Catenulispora acidiphila

 
3.A.1.105.28

ABC-like transporter with 4 components, two integrals membrane ABC proteins (O26020 and O26021, 376 and 365 aas, respectively) , an MFP protein (OP94851; 329 aas) and a TolC-like protein (O026022; 510 aas). This system appears to play a role in flagellar stability and bacterial motility (Gibson et al. 2022).

4 component ABC-like transporter

 
3.A.1.105.3

The 4A-4E-O-dideacetyl-chromomycin A3 (biosynthetic precursor of chromomycin) exporter (may also export chromomycin and mithramycin (Menendez et al., 2007).

Gram-positive Bacteria

CmrAB of Streptomyces greseus
CmrA(C) (Q70J75)
CmrB(M) (Q70J76)

 
3.A.1.105.4

The pyoluteorin (a chlorinated polyketide) efflux pump, PltHIJKN (Brodhagen et al. 2005; Huang et al. 2006).

γ-Proteobacteria

PltHIJKN of Pseudomonas sp. M18:
PltH (336aas; MFP) - (Q4VWD0)
PltI (589aas; C-C) - (Q4VWC9)
PltJ (377aas; M; COG0842; similar to 9.B.74.2 (ABC-2)) - (Q4VWC8)
PltK (372aas; M; The C-terminal hydrophobic half has 5TMSs and is most similar to PltJ, and then to 9.B.74.2, but it is also homologous to 3.A.1.105.2 and 3.A.1.102.1) - (Q4VWC7)
PltN (480aas; OMF) - (Q4VWC6)

 
3.A.1.105.5

AbcG homologue, Snustorr, sioform A, Snu, of 808 aas and 6 TMSs in a 1 + 5 TMS arrangement at the C-terminal part of the protein.  The N-terminal domain is the ATPase domain.  The protein therefore has a C-M domain arrangement.  Lipids in extracellular matrices (ECM) contribute to barrier function and stability of epithelial tissues such as the pulmonary alveoli and the skin. In insects, skin waterproofness depends on the outermost layer of the extracellular cuticle envelope that contains cuticulin, an unidentified water-repellent complex molecule composed of proteins, lipids and catecholamines. Based on live-imaging analyses of fruit fly larvae, Zuber et al. 2018 found that initially, envelope units are assembled within putative vesicles harbouring the ABC transporter Snu and the extracellular protein Snsl. In a second step, the content of these vesicles is distributed to cuticular lipid-transporting nanotubes named pore canals and to the cuticle surface, dependent on Snu function. The surface of snu and snsl mutant larvae is depleted of lipids and cuticulin. Consequently, these animals suffer uncontrolled water loss and penetration of xenobiotics. The data allude to a two-step model of envelope (i.e. barrier) formation. The proposed mechanism in principle parallels the events occurring during differentiation of the lipid-based ECM by keratinocytes in the vertebrate skin, suggesting establishment of analogous mechanisms of skin barrier formation in vertebrates and invertebrates (Zuber et al. 2018).

Animals

AbcG homologue, Snu, of Drosophila melanogaster

 
3.A.1.105.6

The ABC-2-like transporter

Bacteria

ABC-2-like transporter of Dehalococcoides ethenogenes
ABC2 protein (M) (Q3Z8A7)
ATPase (C) (Q3Z8A8)

 
3.A.1.105.7

Putative ABC2 tranport system, SagGHI; may export streptolysin S.

Firmicutes

Putative Streptolysin ABC2 tranport system, SagGHI.
SagG (C) (Q9A0K0)
SagH (M) (Q9A0J9)
SagI (M) (Q9A0J8)

 
3.A.1.105.8

ABC-2 transporter.  The two genes encoding this system are adjacent to one encoding an squalene-hopene cyclase that coverts squalene to hopene.  The substrate could therefore be hopene or a hydrocarbon triterpene derivative of it (Racolta et al. 2012).

Planctomycetes

ABC2 membrane protein (Q7UE57) and ATPase (Q7UE58) of Rhodopirellula baltica

 
3.A.1.105.9

ABC2 membrane proteins (J7ZHK9 and J8A8S6) with ATPase (J8ABC0) transporter

Firmicutes

ABC2 transporter of Bacillus cereus

 


3.A.1.106 The Lipid Exporter (LipidE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.106.1

Phospholipid, LPS, lipid A and drug exporter, MsbA, which flips the substrate from the inner leaflet of the cytoplasmic membrane to the outer leaflet (Eckford and Sharom, 2010). MsbA also confers drug resistance to azidopine, daunomycin, vinblastine, Hoechst 33342 and ethidium (Reuter et al., 2003). Four x-ray structures, trapped in different conformations, two with and two without nucleotide, have been solved (Ward et al., 2007). They suggest an alternating accessibility mode of transport with major conformational changes.  The mechanism and conformational transitions have been discussed (Moradi and Tajkhorshid 2013).  MsbA is energized both by ATP hydrolysis and the H+ electrochemical gradient (Singh et al. 2016). Mi et al. 2017 used single-particle cryo-electron microscopy to elucidate the structures of lipid-nanodisc-embedded MsbA in three functional states. The 4.2 Å-resolution structure of the transmembrane domains of nucleotide-free MsbA revealed that LPS binds deeply inside MsbA at the height of the periplasmic leaflet. Two sub-nanometre-resolution structures of MsbA with ADP-vanadate and ADP revealed a closed and an inward-facing conformation, respectively. A  2.9 A resolution structure of MsbA in complex with G907, a selective small-molecule antagonist with bactericidal activity, revealed an unanticipated mechanism of ABC transporter inhibition. G907 traps MsbA in an inward-facing, lipopolysaccharide-bound conformation by wedging into an architecturally conserved transmembrane pocket. A second allosteric mechanism of antagonism occurs through structural and functional uncoupling of the nucleotide-binding domains (Ho et al. 2018). Coupled ATPase-adenylate kinase activity in ABC transporters including MsbA has been demonstrated (Kaur et al. 2016). Close-proximity effects and structural coupling of the transmembrane domains with the NBDs has been suggested (Josts et al. 2019). Two first-generation inhibitors of MsbA, TBT1 and G247, induce opposite effects on ATP hydrolysis. Using single-particle cryo-electron microscopy and functional assays, TBT1 and G247 were found to bind adjacent yet separate pockets in the MsbA transmembrane domains (Thélot et al. 2021). MsbA adopts the wide inward-open conformation in E. coli cells (Galazzo et al. 2022).  Solid-state NMR spectroscopy rvealed that substantial chemical shift changes within both CH1 and CH2 occur upon substrate binding, in the ATP hydrolysis transition state, and upon inhibitor binding. CH2 is domain-swapped within the MsbA structure, and substrate binding induces a larger response in CH2 compared to CH1. These data show that CH1 and CH2 undergo structural changes as part of the TMD-NBD cross-talk (Novischi et al. 2024).

Gram-negative bacteria

MsbA (M-C) of E. coli

 
3.A.1.106.10

Involved in the export of a molecule required for the autochemotactic process. AbcA integrated permease/ATPase (M-C) protein, MXAN_1286 (Ward et al. 1998). 

Proteobacteria

MXAN_1286 (M-C) of Myxococcus xanthus.

 
3.A.1.106.11

HlyA/MsbA family transporter of 595 aas.  The gene for this protein is adjacent to and probably in the same operon as that encoding 3.A.1.106.12.  They both have 6 TMSs, so they may together comprise a single heterodimeric system. 

Cyanobacteria

ABC exporter of Gloeobacter violaceus

 
3.A.1.106.12

HlyA/MsbA family transporter of 577 aas.  The gene encoding this protein is adjacent to and in the same operon with that encoding 3.A.1.106.11.  They both have 6 TMSs, so they may together comprise a single heterodimeric system. 

Cyanobacteria

ABC exporter of Gloeobacter violaceus

 
3.A.1.106.13

Multidrug resistance-like ABC exporter, MdlAB; exports peptides of 6 - 21 aas (Moussatova et al. 2008).

Proteobacteria

MdlAB of E. coli
MdlA (M-C; 590 aas)
MdlB (M-C; 593 aas)

 
3.A.1.106.14

Lipid A exporter homologue of 593 aas and 6 TMSs (N-terminal with a C-terminal ATPase domain.  Essential for acid, salt and thermal tolerance (Matsuhashi et al. 2015).

Exporter of Synechocystis sp. PCC6803

 
3.A.1.106.15

Lipid flippase, PglK or WlaB, of 564 aas and 6 N-terminal TMSs with a C-terminal ATPase domain.  Mediates the ATP-dependent translocation of an undecaprenylpyrophosphate-linked heptasaccharide intermediate (LLO) across the cell membrane, an essential step during the N-linked protein glycosylation pathway. Transport across the membrane is effected via ATP-driven conformation changes. Most likely, only the polar and charged part of the glycolipid enter the substrate-binding cavity, and the lipid tail remains exposed to the membrane lipids during the transmembrane flipping process (Alaimo et al. 2006; Kelly et al. 2006; Perez et al. 2015). PglK may employ a "substrate-hunting" mechanism to locally increase the LLO concentration and facilitate its jump into the translocation pathway, for which sugars from the LLO head group are essential; the release of LLO to the outside occurs before ATP hydrolysis and is followed by the closing of the periplasmic cavity of PglK (Perez et al. 2019).

PglK (M-C) of Campylobacter jejuni

 
3.A.1.106.16

Probable integral membrane protein NMA1777 with 6 TMSs in a 2 + 2 + 2 arrangement, ; function and ATPase unknown.

UP of Klebsiella pneumoniae

 
3.A.1.106.17

ABC1 transporter

transporter of Acidobacterium capsulatum

 
3.A.1.106.18

Peptide and multidrug resistance porter of the ABC superfamily, TmrAB. TmrA (Q72J05; 600 aas with 6 N-terminal TMSs) and TmrB (Q72J04; 578 aas with 6 N-terminal TMSs) comprise this heterodimeric transporter, both proteins of the M-C structure.  The system has been found to export the dye, hoechst 33342, and to be inhibited by verapamil (Zutz et al. 2011). The subnanometre-resolution structure of detergent-solubilized TmrAB in a nucleotide-free, inward-facing conformation by single-particle electron cryomicroscopy has been solved (Kim et al. 2015). A cavity in the transmembrane domain is accessible laterally from the cytoplasmic side of the membrane as well as from the cytoplasm, indicating that the transporter lies in an inward-facing open conformation. The two nucleotide-binding domains remain in contact via their carboxy-terminal helices. Comparison between this structure and those of other ABC transporters suggests a possible trajectory of conformational changes that involves a sliding and rotating motion between the two nucleotide-binding domains during the transition from the inward-facing to outward-facing conformations (Kim et al. 2015). A subset of annular lipids is normally invariant in composition, with negatively charged lipids binding tightly to TmrAB, suggesting that this exporter may be involved in glycolipid translocation (Bechara et al. 2015). Coupled ATPase-adenylate kinase activity in ABC transporters including TmrAB has been demonstrated (Kaur et al. 2016). A 2.7-Å X-ray structure of TmrAB has been determined. It not only shares structural homology with the antigen translocation complex TAP, but is also able to restore antigen processing in human TAP-deficient cells. TmrAB exhibits a broad peptide specificity and can concentrate substrates several thousandfold, using only one single active ATP-binding site. It adopts an asymmetric inward-facing state, and the C-terminal helices, arranged in a zipper-like fashion, play a role in guiding the conformational changes associated with substrate transport (Nöll et al. 2017). Conformational coupling and trans-inhibition have been characterized (Barth et al. 2018), and a  conserved motif in intracellular loop 1 stabilizes the outward-facing conformation of TmrAB (Millan et al. 2021). A strong entropy-enthalpy compensation mechanism enables the closure of the nucleotide-binding domains (NBDs) over a wide temperature range. This is mechanically coupled with an outward opening of the transmembrane domains (TMDs) accompanied by an entropy gain (Barth et al. 2020).  TmrAB undergoes a reversible transition in the presence of ATP with a significantly faster forward transition. The impaired degenerate NBS stably binds Mn2+-ATP, and Mn2+ is preferentially released at the active consensus NBS (Rudolf et al. 2023). ATP hydrolysis at the consensus NBS considerably accelerates the reverse transition. Both NBSs fully open during each conformational cycle, and the degenerate NBS may regulate the kinetics of this process (Rudolf et al. 2023).

TmrAB of Thermus thermophilus

 
3.A.1.106.19

ABC exporter.  It has been suggested that it might be a glycolate exporter (Braakman et al. 2017). However it's closest hit in TCDB (31% identity in the transmembrane domain) has TC# 3.A.1.106.18, which is probably a peptide/multidrug (and possibly glycolipid) exporter with broad substrate specificity. 

ABC exporter of Prochlorococcus marinus

 
3.A.1.106.2

The homodimeric Sav1866 multidrug exporter (transports doxorubicin, verapamil, ethidium, tetraphenylphosphonium, vinblastine and the fluorescent dye, Hoechst 33342; 3-D structure known at 3 Å resolution; Dawson and Locher, 2006; Velamakanni et al., 2008) The empty site opens by rotation of the nucleotide-binding domain whereas the ATP-bound site remains occluded (Jones and George, 2011). Conformational changes induced by ATP-binding and hydrolysis have been proposed (Becker et al. 2010; Oliveira et al., 2011). The alternating access mechanism and the flippase activity of this ABC exporter has been shown to be lipid-dependent (Immadisetty et al. 2019).

Gram-positive Bacteria

Sav1866 of Staphylococcus aureus (M-C) 2HYDA/2HYDB (578 aas)

 
3.A.1.106.20

MsbA of 582 aas and 6 TMSs in an M-C arrangement.  The X-ray structure at 2.8 Å resolution in an inward-facing conformation after cocrystallization with lipid A and using a stabilizing facial amphiphile has been reported (Padayatti et al. 2019). The structure displays a large amplitude opening in the transmembrane portal, which is likely to be required for lipid A to pass from its site of synthesis into the protein-enclosed transport pathway. Putative lipid A density is observed further inside the transmembrane cavity, consistent with a trap and flip model. Additional electron density attributed to lipid A is observed near an outer surface cleft at the periplasmic ends of the transmembrane helices (Padayatti et al. 2019). This protein is 96% identical to the E. coli ortholog, TC# 3.A.1.106.1.

MsbA of Salmonella enterica

 
3.A.1.106.21

Quiinol:oxygen oxidoreductzase; thiol reductant ABC exporter subunit CydC, of 583 aas and 6 N-terminal TMSs in a 2 + 2 + 2 TMS arrangement plus a hydrophilic C-terninal half (Murali et al. 2021).

CydC of Methanothrix soehngenii

 
3.A.1.106.3

The dimeric multidrug resistance exporter, ABC1/2 (exports the peptide antimicrobials, nisin and polymyxin; (Margolles et al., 2006) (both ABC1 and ABC2 also show striking similarity to family 3.A.1.117).

Gram-positive Bacteria

ABC1/2 of Brevibacterium longum:
ABC-1 (M-C) (ZP_00121338)
ABC-2 (M-C) (ZP_00121339)

 
3.A.1.106.4The duplicated ABC transporter, CgR_1214 (1247 aas; MC(poorly conserved) MC(well conserved))BacteriaCgR_1214 of Corynebacterium glutamicum (MCMC) (A4QD95)
 
3.A.1.106.5The heterodimeric multidrug efflux pump, SmdAB (exports norfloxacin, tetracycline, 4',6-diamidino-2-phenylindole (DAPI), and Hoechst 33342) (Matsuo et al., 2008).BacteriaSmdAB of Serratia marcescens:
SmdA (M-C) (A7VN01)
SmdB (M-C) (A7VN02)
 
3.A.1.106.6Multidrug efflux pump, Rv0194 (exports & causes resistance to ampicillin, streptomycin and chloramphenicol by 32- to 64-fold and to vancomycin and tetracycline by 4- to 8-fold (Danilchanka et al., 2008)).BacteriaRv0194 of Mycobacterium tuberculosis (MCMC) (O53645)
 
3.A.1.106.7

The Salmochelin/Enterobactin secretory exporter, IroC (Crouch et al., 2008; Müller et al. 2009).

Bacteria

IroC of Salmonella enterica (MCMC) (Q8RMB7)

 
3.A.1.106.8

The heterodimeric BmrC/BmrD (YheHI) MDR transporter.  Transports a wide range of structurally unrelated drugs including doxorubicin, mitoxantrone, ethidium, and hoechst 33342 (Torres et al., 2009). It activates the sensor kinase, KinA, during sporulation initiation (Fukushima et al. 2006). Large scale purification has been achieved (Galián et al. 2011).  It has been reconstituted in giant unilamellar vesicles (Dezi et al. 2013).  It exhibits an asymmetric configuration of catalytically inequivalent nucleotide binding sites. The two-state transition of the TMS domains, from an inward- to an outward-facing conformation, may be driven exclusively by ATP hydrolysis (Mishra et al. 2014). A novel intermediate of BmrCD, a heterodimeric multidrug ABC exporter from Bacillus subtilis. has been identified (Thaker et al. 2021). In the cryo-EM structure, ATP-bound BmrCD adopts an inward-facing architecture featuring two molecules of the substrate Hoechst-33342 in an asymmetric head-to-tail arrangement. Deletion of the extracellular domain capping the substrate-binding chamber or mutation of Hoechst-coordinating residues abrogates cooperative stimulation of ATP hydrolysis. These findings support a mechanistic role for symmetry mismatch between the nucleotide binding and the transmembrane domains in the conformational cycle of ABC transporters (Thaker et al. 2021). Lipid interactions with BmrCD modulate the energy landscape, suggesting a distinct transport model that highlights the role of asymmetric conformations in the ATP-coupled cycle with implications to the mechanism of ABC transporters in general (Tang et al. 2023).

Bacteria

BmrC/BmrD (YheHI) of Bacillus subtilis
YheH (M-C) (O07549)
YheI (M-C) (O07550)

 
3.A.1.106.9

SoxR regulon single protein ABC exporter, Sco7008, containing an N-terminal membrane domain and a C-terminal ATPase domain (Shin et al. 2011). SoxR responds to extracellular redox-active compounds.  Thus, it is induced in stationary phase during the production of the benzochromanequinone blue-pigmented antibiotic, actinorhodin (Naseer et al. 2014). Possibly an actinorhodin exporter.

Actinobacteria

Sco7008 (M-C) of Streptomyces coelicolor.

 


3.A.1.107 The Putative Heme Exporter (HemeE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.107.1Putative heme exporter, CcmABC=CycVWZ (Note: CcmC may function independently of CcmAB) (Feissner et al., 2006; Christensen et al., 2007)Gram-negative bacteriaCycVWZ of Bradyrhizobium japonicum
CycV (C)
CycW (M)
CycZ (M)
 
3.A.1.107.2The mitochondrial ABC transporter involved in cytochrome c maturation, CcmA/CcmB. (Note: CcmA is nuclearly encoded while CcmB is mitochondrially encoded) (Rayapuram et al., 2007) Plant Mitochondria

CcmA/CcmB of Arabidopsis thaliana
CcmA (C) (Q9C8T1)
CcmB (M) (P93280)

 
3.A.1.107.3

CcmABCD exporter; CcmD (69aas, 1TMS) is required for the release of CcmE (which binds heme in the periplasm) from CcmABC. CcmC (9.B.14.2.3) is required for the transfer of heme to CcmE in the periplasm (Richard-Fogal et al., 2008) In the presence of heme, CcmC and CcmE form a stable complex (Richard-Fogal & Kranz, 2010) as do CcmE and CcmF (San Francisco and Kranz 2014). The cytochrome c maturation system I, consisting of eight membrane/periplasmic proteins (CcmABCDEFGH), results in the attachment of heme to cysteine residues of cytochrome c proteins. Since all c-type cytochromes are periplasmic, heme is first transported to a periplasmic heme chaperone, CcmE. A large membrane complex, CcmABCD has been proposed to carry out this transport and linkage to CcmE. Li et al. 2022 described high resolution cryo-EM structures of CcmABCD in an unbound form, in complex with inhibitor AMP-PNP, and in complex with ATP and heme. The ATP-binding site in CcmA and the heme-binding site in CcmC were identified. They proposed a model of heme trafficking, heme transfer to CcmE, and ATP-dependent release of holoCcmE from CcmABCD. CcmABCD represents an ABC transporter complex using the energy of ATP hydrolysis for the transfer of heme from one binding partner (CcmC, see TC# 9.B.14.2.3) to another (CcmE) (Li et al. 2022). It appers that CcmC is in a complex with CcmABD but is not part of the ABC transporter. The same may be true of CcmD (see description above).

Proteobacteria

CcmABCD of E. coli
CcmA (C) (Q8XE58)
CcmB (M; 7 TMSs) (P0ABM0)
CcmC (M; 6 TMSs) (P0ABM1 = P0ABM3) (listed under TC# 9.B.14.2.3 not here)
CcmD (M; 1 TMS) (P0ABM7)

 
3.A.1.107.4

Cytochrome c maturation system (heme exporter?), CcmA/B

γ-Proteobacteria

CcmAB of Pseudomonas virdiflava
CcmA (C) (K6BJ24)
CcmB (M) (K6BIH6)

 
3.A.1.107.5

CcmB of  353 aas with 9 TMSs. It may act with TC# 9.B.14.1.21, involved in heme insertioin into cytochrome c (Gupta et al. 2022).

CcmB of Methanosarcina acetivorans

 


3.A.1.108 The β-Glucan Exporter (GlucanE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.108.1β-Glucan exporterGram-negative bacteriaNdvA (M-C) of Rhizobium meliloti
 


3.A.1.109 The Protein-1 Exporter (Prot1E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.109.1

α-Hemolysin exporter. HlyB has an (inactive?) N-terminal C39 peptidase-like domain (Lecher et al., 2011).  It is essential for secretion and interacts with the unfolded HlyA, thereby protecting it from cytoplasmic degradation (Lecher et al. 2012). Type 1 secretion systems (T1SSs)  extruding protein substrates following synthesis of the entire polypeptide. The E. coli hemolysin A secretion system has three membrane proteins - an inner membrane ABC transporter HlyB, an adaptor protein HlyD TC# 8.A.1.3.1), and an outer membrane porin TolC (TC# 1.B.17.1.1). All are required for secretion. Cryo-EM structures determined in two conformations revealed that the inner membrane complex is a hetero-dodecameric assembly comprising three HlyB homodimers and six HlyD subunits. Oligomerization of HlyB and HlyD is essential for protein secretion, and polypeptides translocate through a canonical ABC transporter pathway in HlyB (Zhao et al. 2022).

Gram-negative bacteria

HlyB (M-C) of E. coli

 
3.A.1.109.2

Cyclolysin exporter, CyaB (Glaser et al., 1988) (Possesses an N-terminal lysosomal sorting signal within the amino-terminal transmembrane domain; Kamakura et al., 2008).

Gram-negative bacteria

CyaB (M-C) of Bordetella pertussis

 
3.A.1.109.3LapA adhesin protein exporter, LapB (Hinsa et al., 2003)BacteriaLapB of Pseudomonas putida
LapB (MC) (AAN65800)
 
3.A.1.109.4

The biofilm inducible ABC drug (tobramycin, gentamycin, and ciprofloxacin) resistance pump, PA1875-PA1877 (Zhang and Mah, 2008).  It is specifically induced and is most active when  growing in a biofilm.

Proteobacteria

PA1875-PA1877 of Pseudomonas aeruginosa
PA1875 (OMF; 425 aas) (Q9I2M2)
PA1876 (ABC; M-C; 723 aas) (Q9I2M1)
PA1877 (MFP; 395 aas) (Q9I2M0)

 
3.A.1.109.5

Probable giant non-fimbrial adhesin, SiiE, exporter, SiiFDC.  SiiF interacts with SiiAB (TC# 1.A.30.4.1) which probably forms a proton channel homologous to that of MotAB (TC# 1.A.30.1.1) and facilitates energization of SiiE export using the pmf (Wille et al. 2013).

Proteobacteria

SiiFDC of Salmonella enterica
SiiF (M-C; 688 aas; E1WEV2)
SiiD (MFP; 425 aas; E1WEV0)
SiiC (OMF; 439 aas; E1WEU9)

 
3.A.1.109.6

Probable 2646 aa extracellular adhesin (acc# C6BWI7) ABC exporter of 715 aas.  Functions as a type I protein secretion system together with an MFP and an OMF which all are encoded within a single operon together with the adhesin and SiiAB homologues as for TC# 3.A.1.109.5.

Proteobacteria

ABC/MFP/OMF type I protein secretion system of Desulfovibrio salexigens
ABC protein (M-C; 715 aas; C6BWI0)
MFP protein (430 aas; C6BWj0)
OMF protein (513 aas; C6BWI6)

 
3.A.1.109.8

Leukotoxin export protein of 707 aas, LtxB (has a fused M-C structure with 6 TMSs) (Guthmiller et al. 1995). Functions with the MFP, LtxD (TC# 8.A.1.3.4) and the TolC-like protein, TdeA (TC# 1.B.17.3.11).

Leukotoxin exporter of Aggregatibacter (Actinobacillus; Haemophilus) actinomycetemcomitans

 


3.A.1.11 The Polyamine/Opine/Phosphonate Uptake Transporter (POPT) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.11.1

Polyamine (putrescine/spermidine) uptake porter.  Plays a role in biofilm formation (Zhang et al. 2013).  Spermidine-preferring (Igarashi and Kashiwagi 1996).

Proteobacteria

PotABCD of E. coli
PotA (C)
PotB (M)
PotC (M)
PotD (R)

 
3.A.1.11.10

The 3-component polyamine uptake transporter, PotABD. Transports homospermidine and possibly other polyamines. Inactivation of the potADB gene cluster (potADB) disrupted diazotrophic growth, clearly suggesting the importance of polyamine homeostasis in Anabaena. (Burnat et al. 2018). 

PotABD of Anabaena variabilis

 
3.A.1.11.2

Putrescine porter (Igarashi and Kashiwagi 1996).

Proteobacteria

PotGHIF of E. coli
PotG (C)
PotH (M)
PotI (M)
PotF (R)

 
3.A.1.11.3Mannopine porterProteobacteriaMotABCD of Agrobacterium tumefaciens plasmid pTi15955
MotA (R)
MotB (C)
MotC (M)
MotD (M)
 
3.A.1.11.4Chrysopine porterProteobacteriaChtGHIJK of Agrobacterium tumefaciens
ChtG (C)
ChtH (R)
ChtI (R)
ChtJ (M)
ChtK (M)
 
3.A.1.11.52-aminoethyl phosphonate porterProteobacteriaPhnSTUV of Salmonella typhimurium
PhnS (R)
PhnT (C)
PhnU (M)
PhnV (M)
 
3.A.1.11.6The γ-aminobutyrate (GABA) uptake system, GtsABCD (White et al., 2009).

Bacteria

GtsABCD of Rhizobium leguminosarum
GtsA (R) (Q1M7Q4)
GtsB (M) (Q1M7Q3)
GtsC (M) (Q1M7Q2)
GtsD (C) (Q1M7Q1)

 
3.A.1.11.7

The spermidine/putrescine uptake porter, PotABCD (Shah et al. 2008; Shah et al. 2006; Ware et al. 2006).

Firmictues

PotABCD of Streptococcus pneumoniae
PotA (C) 385 aas
PotB (M) 275 aas (also called PotH)
PotC (M) 257 aas
PotD (R) 356 aas

 
3.A.1.11.8

The spermine/spermidine uptake porter, PotABCD.

Firmicutes

PotABCD of Staphylococcus aureus
PotA (C)
PotB (M)
PotC (M)
PotD (R)

 

 
3.A.1.11.9

Putative polyamine (spermidine/putrescine) uptake porter, YdcSTUV (Moussatova et al. 2008). May also be involved in the uptake of double stranded DNA (Sun 2018).

Proteobacteria

YdcSTUV of E. coli
YdcS (R; 381 aas)
YdcT (C; 337 aas)
YdcU (M; 313 aas)
YdcV (M; 264 aas)

 


3.A.1.110 The Protein-2 Exporter (Prot2E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.110.10The AlgE-type Mannuronan C-5-Epimerase exporter, EexD (PrtD) (Gimmestad et al., 2006).Bacteria

EexD of Azotobacter vinelandii (C1DS84)

 
3.A.1.110.11

Secretion system for metalloprotease, PrtA, PrtDEF (Akatsuka et al. 1997). (PrtF=1.B.17.1.2)

Gram-negative bacteria

PrtDEF of Erwinia chysanthemi 
PrtD (M-C) (P23596)
PrtE (MFP) (P23597) 

 
3.A.1.110.12

Thermostable lipase, TliA (Q9ZG91; 476 aas with a C-terminal region that shows similarity to members of the RTX toxin family (1.C.11)) exporter, TliDEF.  The wild type transporter has a temperature sensitive defect which can be corrected by a single mutation in TliD (Eom et al. 2016).

TliDEF of Pseudomonas fluorescens
TliD, 578 aas (M-C) and 6 N-terminal TMSs
TliE, 433 aas (MFP)
TliF, 481 aas (OMF)

 
3.A.1.110.13

Protein export system, PrtD of 564 aas and 6 TMSs. The 3.15 Å structure has been solved (Morgan et al. 2017).  The structure suggests a substrate entry window just above the transporter's nucleotide binding domains. Highly kinked transmembrane helices, which frame a narrow channel, not observed in canonical peptide transporters, are likely involved in substrate translocation. The PrtD structure supports a polypeptide transport mechanism distinct from alternating access (Morgan et al. 2017).

PrtD of Aquifex aeolicus

 
3.A.1.110.3The multiple protein exporter, PrsD/PrsE (exports EPS glycanases, PlyA and PlyB, as well as Rhizobium adhering proteins) (Russo et al., 2006). 12 substrates have been identified; PrsDE provide the major route of protein export in R. leguminosarum (Krehenbrink and Downie, 2008).Gram-negative bacteriaPrsD/PrsE of Rhizobium leguminosarum
PrsD(M-C) (O05693)
PrsE(MFP) (O05694)
 
3.A.1.110.4Alkaline protease exporterGram-negative bacteriaAprD (M-C) of Pseudomonas aeruginosa
 
3.A.1.110.5S-layer protein exporterGram-negative bacteriaRsaD (M-C) of Caulobacter crescentus
 
3.A.1.110.6

Exporter for lipase LipA, protease PrtA and S-layer protein SlaA, LipBCD (Akatsuka et al. 1997).   LipABC is also called PrtDEF.

Gram-negative bacteria

LipBCD of Serratia marcescens
LipB (M-C) (Q54456)
LipC (MFP) (Q54457)
LipD (OMF) (O87950)

 
3.A.1.110.7

Exporter for heme-binding protein, HasA and metaloprotease, PrtA.  Functions as a complex spanning the two membranes of the cell envelope: HasDEF (HasD = ABC protein; HasE = the MFP; HasF = the OMF (see 2.A.6.2.31 for HasF) (Akatsuka et al. 1997).

Gram-negative bacteria

HasDEF of Serratia marcescens
HasD (M-C) (Q53368)
HasE (MFP) (Q57387)
HasF (OMF) (Q54452) 

 

 
3.A.1.110.8Surface layer protein exporterGram-negative bacteriaSapD (M-C) of Campylobacter fetus
 
3.A.1.110.9Exporter of HasA lipase, and alkaline proteaseGram-negative bacteriaHasD (M-C) of Pseudomonas fluorescens
 


3.A.1.111 The Peptide-1 Exporter (Pep1E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.111.1

Hemolysin/bacteriocin (cytolysin) exporter with associated proteolytic activity

Gram-positive bacteria

CylT (M-C) (CylB) of Enterococcus faecalis

 
3.A.1.111.2Subtilin (toxic peptide) exporterGram-positive bacteriaSpaB (M-C) of Bacillus subtilis
 
3.A.1.111.3Nisin exporterGram-positive bacteriaNisT (M-C) of Lactococcus lactis
 
3.A.1.111.4Bacteriocin immunity protein, SmbG (198 aas; 6TMSs in a 2+2+2 arrangement. (Exports bacteriocins and causes resistance to antibiotics such as tetracycline, penicillin and triclosan). Upregulated by exposure to antibiotics (Matsumoto-Nakano and Kuramitsu, 2006)Gram-positive bacteriaSmbG (M-C) of Streptococcus mutans (Q5TLL2)
 
3.A.1.111.5The lacticin Q exporter, LcnDR3 (Yoneyama et al., 2009).

Gram-positive bacteria

LcnDR3 (M-C) of Lactococcus lactis (P37608)

 
3.A.1.111.6

Salivericin 9 exporter, SivT (692 aas; 6 TMSs) (Wescombe et al., 2011)

Firmicutes

SivT of Strepococcus salivarius (F8LI02)

 
3.A.1.111.7

Nukacin ISK-1 bacteriocin exporter, NukT of 694 aas and 6 TMSs.  The protease domain is N-terminal, the membrane domain is central, and the ATPase domain in C-terminal. NukT and its peptidase-inactive mutant have been expressed, purified, and reconstituted into liposomes for analysis of their peptidase and ATPase activities. The ATPase activity of the NBD (C) region is required for the cysteine-type peptidase activity, and leader peptide cleavage enhances the ATPase activity (Zheng et al. 2017).

NukT of Staphylococcus warneri (P-M-C)

 
3.A.1.111.8

Uncharacterized ABC export system of 608 aas and 6 N-terminal TMSs in a 2 + 2 + 2 TMS arrangement followed by the ATPase domain (M-C).  It is adjacent to a 10  protein where the TMSs are in a 5 + 5 TMS arrangement.  Possibly this latter protein is a chaparone protein for proper insertion and folding of the transporter (see TC# 9.B.29.2.17 whick seems to be a chaparone protein for insertion and folding of ABC transporter with TC# 3.A.1.122.2. 

ABC exporter of Lachnospiraceae bacterium

 


3.A.1.112 The Peptide-2 Exporter (Pep2E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.112.1

Competence factor (CSF; a heptadecapeptide) exporter of 717 aas.  The transporter is fused to an N-terminal peptidase  domain and functions with an MFP  accessory protein, ComB (TC# 8.A.1.4.2) (Ishii et al. 2006). 

Gram-positive bacteria

ComA (peptidase-M-C) of Streptococcus pneumoniae (functions with MFP accessory protein, ComB)

 
3.A.1.112.10

Bacteriocin exporter of 721 aas and 7 TMSs. Residues 10 - 134: peptidase with N-terminal TMS; residues 167 - 446: TM domain; residues 480 - 715: ATPase.

Peptide exporter of Bacteroides salanitronis

 
3.A.1.112.11

Enterocin CRL35 exporter, MunB, of 674 aas and 6 TMSs in an M-C domain arrangement.  The specific receptor for Enterocin CRL35 (MunA; TC# 1.C.24.1.15) acts as a docking molecule, not a structural part of the pore, but the bacteriocin must be anchored to the membrane (Ríos Colombo et al. 2019).

Enterocin CRL35 exporter of Enterococcus mundtii

 
3.A.1.112.12

Colicin V exporter. The ATPase is a GTPase (Zhong and Tai 1998; ).

Enteric bacteria

CvaB (M-C) of E. coli

 
3.A.1.112.13

Microcin E492 exporter, MceFGH (MceF has 5 - 7 TMSs and is most likely a CAAX amino terminal protease that might function in the processing of microcin E492; MceG has a short hydrophilic N-terminus, a centra 6 TMS ABC domain, and a C-terminal ABC ATPase domain; MceH has 1 N-terminal TMS) (Bieler et al., 2006; Lagos et al., 1999)

Proteobacteria

MceGH of Klebsiella pneumoniae
MceG (C-M-C) (Q93GK5)
MceH (MFP) (Q93GK4)

 
3.A.1.112.2Pediocin PA-1 exporterGram-positive bacteriaPedD (M-C) of Pediococcus acidilactici
 
3.A.1.112.3

Bacteriocin (lactococcin) exporter. 

Gram-positive bacteria

LcnC (M-C) of Lactococcus lactis (functions with putative MFP accessory protein LcnD)

 
3.A.1.112.4Sublancin exporter, SunTGram-positive bacteriaSunT (M-C) of Bacillus subtilis
 
3.A.1.112.5

Exporter of the BlpC peptide pheromone (B5E242) and several bacteriocins, BlpAB (Kochan and Dawid 2013).

Firmicute

BlpAB of Streptococcus pneumoniae
BlpA (M-C) (B3E244)
BlpB (MFP) (B3E242)

 
3.A.1.112.6

Putative ABC transporter (6 TMSs)

Bacteria

ABC Transporter of Ureaplasma parvum (Q9PPY0)

 
3.A.1.112.8

Mesenterici Y105 (bacteriocin) ABC exporter and porcessing protease, MesD(E) of 722 aas and 6 TMSs (MesD) (Fremaux et al. 1995). MesDE can transport and catalyze maturation of the two pre-bacteriocins, mesentericin Y105 and B105 (Aucher et al. 2004).  Hydrophobic conserved amino acyl residues and the C-terminal GG doublet of the leader peptide of pre-mesentericin Y105 are critical for optimal secretion (Aucher et al. 2005).  MesE has TC# 8.A.1.4.1.

Firmicutes

MesDE of Leuconostoc mesenteroides

 
3.A.1.112.9

ABC bacteriocin exporter with two peptidase domains, Pcat1. 3-D structures are known (4S0F, 6V9Z, 4RY2). The pathway for peptide export consists of an large α-helical barrel for small folded peptides.  ATP binding alternates access to the transmembrane pathway and reglates protease activity (Lin et al. 2015). Subunit asymmetry of the M3-M4 loops contribute to optimizing AChR activation through allosteric links to the channel and the agonist binding site (Shen et al. 2005). Structures were more recently determined in the absence and presence of ATP, revealing how ATP binding regulates the protease activity and access to the translocation pathway. Two substrate CtAs, 90-residue polypeptides, are bound via their N-terminal leader peptides, but only one is positioned for cleavage and translocation. The structures were determined in the absence and presence of ATP, revealing how ATP binding regulates the protease activity and access to the translocation pathway. It seems that substrate cleavage, ATP hydrolysis, and substrate translocation are coordinated in a transport cycle (Kieuvongngam et al. 2020). The N-terminal C39 peptidase (PEP) domain of PCAT1 processes its natural substrate, CtA, by cleaving a conserved -GG- motif to separate the cargo from the leader peptide prior to secretion. The ATP-mediated association between PEP and the transmembrane domains of PCAT1 offers a putative mechanism to optimize peptide cleavage by regulating the width and flexibility of the enzyme active site (Bhattacharya and Palillo 2021). Structures of the peptidase-containing ABC transporter PCAT1 under equilibrium and nonequilibrium conditions have been solved (Kieuvongngam and Chen 2022).

Pcat1 of Ruminiclostridium thermocellus (Clostridium thermocellum; Hungateiclostridium thermocellum)

 


3.A.1.113 The Peptide-3 Exporter (Pep3E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.113.1Modified cyclic peptide (syringomycin) exporter, SyrD Gram-negative bacteriaSyrD (M-C) of Pseudomonas syringae
 
3.A.1.113.2Pyoverdin (siderophore) exporterGram-negative bacteriaPvdE (M-C) of Pseudomonas aeruginosa
 
3.A.1.113.3

The multidrug/microcin J25 (MccJ25; 21 aa cyclic peptide antibiotic; the precursor peptide is McjA) exporter, YojI (Delgado et al., 2005). TolC is also required for export; Vincent and Morero, 2009). This system exports many phytol derivatives (Upadhyay et al. 2014).  Also exports L-cysteine (Yamada et al., 2006).  This is one of two microcin J25 exporters, the other being McjD (TC# 3.A.1.118.1).

Gram-negative bacteria

YojI of E. coli (P33941)

 


3.A.1.114 The Probable Glycolipid Exporter (DevE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.114.1

Glycolipid exporter (under nitrogen control in heterocysts), DevABC-HgdD (Moslavac et al., 2007). Heterocyst envelope glycolipids (HGLs) function as an O2 diffusion barrier, being deposited over the heterocyst outer membrane, surrounded by an outermost heterocyst polysaccharide envelope. DevBCA and TolC form an ATP-driven efflux pump required for the export of HGLs across the Gram-negative cell wall (Staron et al., 2011). DevB, the MFP, must be hexameric to create a functional export complex.  This system is under NtcA and nitrogen control and is required for heterocyst development (Fiedler et al. 2001).

Cyanobacteria

DevABC-HgdD of Anabaena variabilis (sp. strain PCC7120)
DevA (C)
DevB (MFP)
DevC (M)
HgdD (TolC like)

 


3.A.1.115 The Na+ Exporter (NatE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.115.1

Na+ efflux pump NatAB. It is induced by ethanol andby protonophores (Cheng et al. 1997). A similar system was found in the alkaliphilic Bacillus firmus OF4 species (Wei et al. 1999). The Bacillus subtilis NatK-NatR two-component system regulates expression of the natAB operon (Ogura et al. 2007).

Gram-positive bacteria

NatAB of Bacillus subtilis
NatA (M)
NatB (C)

 
3.A.1.115.2

Putative Na extrusion pump, NatAB.  NatB has an N-terminal NatB domain (residues 1 - 375) as well as a C-terminal CAAX protease domain (9.B.2; residues 380 - 650).

Planctomycetes

NatAB of Rhodopirellula baltica

 
3.A.1.115.3

ABC transporter of unknown function

ABC transporter
AKM79972, (M)
AKM79973, (C)

 
3.A.1.115.4

Probable two component Na+ efflux system, NatAB, where NatB is a 228 aa ATPase and NatA is a 416 aa membrane protein with 6 TMSs in a 1 + 5 TMS arrangement.

NatAB of Evansella cellulosilytica (Bacillus cellulosilyticus)

 


3.A.1.116 The Microcin B17 Exporter (McbE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.116.1Microcin B17 exporterEnteric bacteriaMcbEF of E. coli
McbE (M)
McbF (C)
 


3.A.1.117 The Drug Exporter-2 (DrugE2) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.117.1

The multidrug exporter, LmrA (can also substitute for MsbA [TC #3.A.1.106.1] to export lipid A; Reuter et al., 2003).  Structural models have been presented (Ecker et al. 2004; Federici et al. 2007). Hoechst 33342 is a substrate (van den Berg and van Saparoea et al. 2005). Coupled ATPase-adenylate kinase activity in ABC transporters including LmrA has been demonstrated (Kaur et al. 2016). This efflux porter mediates efflux of hydrophobic cationic substrates including antibiotics. TMS 3 of one monomer probably contacts TMS 5 or TMS 6 of the opposite monomer where substrate-binding occurs at the monomer/monomer interface (Ecker et al. 2004).

Gram-positive bacteria

LmrA (M-C) of Lactococcus lactis

 
3.A.1.117.2

Hop resistance protein, HorA. Reconstitution in phosphatidyl ethanolamine bilayers resulted in normal activity, but reconstitution in phosphatidyl choline resulted in uncoupling of ATP hydrolysis from transport and a change in the orientations of the TMSs (Gustot et al. 2010).

Gram-positive bacteria

HorA (M-C) of Lactobacillus brevis

 
3.A.1.117.3

Multidrug resistance homodimeric efflux pump, BmrA (YvcC) of 589 aas (Dalmas et al. 2005).  The low resolution cryo-electron microscopy reconstitution suggests large conformational changes occur during it's catalytic cycle (Fribourg et al. 2014). Backbone NMR assignments of the nucleotide binding domain of BmrA in the post-hydrolysis state have been determined (Pérez Carrillo et al. 2022). The protein is homodimeric, and it's unfolding and themodynamic stability have been studied (Oepen et al. 2023).

Firmictues

BmrA of Bacillus subtilis

 


3.A.1.118 The Microcin J25 Exporter (McjD) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.118.1

The cyclic peptide antibiotic, microcin J25 (MccJ25; the precursor peptide is JcjA) exporter, the self immunity protein, McjD. TolC is also required for export; Vincent and Morero, 2009.  The 3-d structure has been determined to 2.7Å resolution in an outward occluded state (Choudhury et al. 2014).  Binding and efflux as well as stimulation of the ATPase activity upon binding of MccJ25 have been demonstrated (Choudhury et al. 2014).  This is one of two MCCJ25 exporters, the other being YojI (TC# 3.A.1.113.3).  The large conformational changes in some crystal structures may not be necessary even for a large substrate like MccJ25 (Gu et al. 2015).

Gram-negative bacteria

McjD (M-C) of E. coli

 


3.A.1.119 The Drug/Siderophore Exporter-3 (DrugE3) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.119.15-Hydroxystreptomycin and other streptomycin-like aminoglycoside exporter, StrVWGram-positive bacteriaStrVW of Streptomyces glaucescens
StrV (M-C)
StrW (M-C)
 
3.A.1.119.2Tetracycline/oxytetracycline/oxacillin exporter, TetABGram-positive bacteriaTetAB (StrAB) of Corynebacterium striatum
TetA (M-C)
TetB (M-C)
 
3.A.1.119.3

Exochelin exporter, ExiT (Zhu et al. 1998).

Gram-positive bacteria

ExiT of Mycobacterium smegmatis
(MC-M-C)

 
3.A.1.119.4

Putative coelichelin (hydroxamate siderophore) exporter, Sco0493; the gene is in a gene cluster encoding the recognized coelichelin uptake system (TC# 3.A.1.14.12) as well as coelichelin biosynthetic enzymes (Barona-Gómez et al. 2006).  Sco0493 may function together with Sco0540 which is another putative ABC exporter of similar equence (see TC# 3.A.1.119.5).  However, alternatively, these two genes may encode two distinct transport systems.

Actinobacteria

Putative coelichelin exporter, Sco0493, of Streptomyces coelicolor (M-C)

 
3.A.1.119.5

Putative coelichelin (hydroxamate siderophore) exporter, Sco0493; the gene is in a gene cluster encoding the recognized coelichelin uptake system (TC# 3.A.1.14.12) as well as coelichelin biosynthetic enzymes (Barona-Gómez et al. 2006).  Sco0493  (see TC# 3.A.1.119.4) may function together with Sco0540, both of which are putative ABC exporters of similar sequence. Alternatively, these two genes may encode two distinct transport systems.

Actinobacteria

Sco0540 of Streptomyces coelicolor (M-C)

 


3.A.1.12 The Quaternary Amine Uptake Transporter (QAT) Family (Similar to 3.A.1.16 and 3.A.1.17)


Examples:

TC#NameOrganismal TypeExample
3.A.1.12.1

Glycine betaine/proline porter, ProU or ProVWX (also transports proline betaine, carnitine, dimethyl proline, homobetaine, γ-butyrobetaine and choline with low affinity).  Contributes to the regulation of cell volume is response to osmolarity.  A reconsituted system shows osmotic strength-gating (Gul and Poolman 2012).

Proteobacteria

ProVWX of E. coli
ProW (M)
ProX (R)
ProV (C)

 
3.A.1.12.10The OpuC transporter selective for glycine betaine > choline, acetylcholine, carnitine and proline betaine (contains tandem cystathionine-β-synthase (CBS) domains in the ABC component of OpuC that are required for osmoregulatory function (Chen and Beattie, 2007)). ProteobacteriaOpuCA, CB, CC of Pseudomonas syringae
OpuCC (R) (Q87WH3)
OpuCB (M) (Q87WH4)
OpuCA (C) (Q87WH5)
 
3.A.1.12.11The glycine betaine uptake porter, GbpABCD (Saum et al., 2009).

Archaea

GbpABCD of Methanosarcina mazei
GbpA (R) (Q8Q040)
GbpB (M) (Q8Q043)
GbpC (M) (Q9Q042)
GbpD (C) (Q8Q041)

 
3.A.1.12.12

The CbcWV/CbcX (choline)/CaiX (carnitine)/BetX (betaine) transporter with 3 binding receptors for distinct quaternary ammonium compounds. Only the ligand-bound receptor binds to the transporter with high affinity (Chen et al., 2010; Thomas et al., 2010).

Bacteria

CbcWV/CbcX/CaiX/BetX of Pseudomonas aeruginosa
CbcW (M) (Q9HTI7)
CbcV (C) (Q9HTI8)
CbcX (R) (Q9HTI6)
CaiX (R) (Q9HTH6)
BetX (R) (Q9HZ04)

 
3.A.1.12.13

High affinity (2mμM) choline uptake porter. The choline binding receptor exhibits a venus fly trap mechanism of substrate binding. (ChoX binds acetyl choline and betaine with low affinity (80μM and 470μM, respectively) (Aktas et al., 2011) (most similar to 3.A.1.12.7)

Bacteria

ChoVWX of Agrobacterium tumefaciens 
ChoX (R) (Q7CXG0)
ChoW (M) (Q7CXG1)
ChoV (C) (A9CI32)

 
3.A.1.12.14

OsmU (OsmVWXY) transporter for glycine betaine and choline-O-sulfate uptake. Induced by osmotic stress (0.3M NaCl) (Frossard et al., 2012). Also called OpuCA/CB1/CB2/CC.

Proteobacteria

OsmU or OsmVWXY of Salmonella enterica 
OsmV (STM1491) (C) (Q8ZPK4)
OsmW (STM1492) (M) (Q8ZPK3)
OsmX (STM1493) (R) (Q8ZPK2)
OsmY (STM1494) (M) (Q8ZPK1) 

 
3.A.1.12.15

Putative osmoprotectant (glycine/betaine/choline) uptake transporter, YehWXYZ.  Induced by osmotic stress and growth into the stationary phase; under RpoS (σS) control (Ibanez-Ruiz et al. 2000; Checroun and Gutierrez 2004).  YehZ is also called OsmF.

Proteobacteria

YehWXYZ of E. coli
YehW (M) 243 aas
YehX (C) 308 aas
YehY (M) 385 aas
YehZ or OsmF (R) 305 aas

 
3.A.1.12.16

Glycine betaine/carnitine/choline/proline transporter, OpuABC. It is not a dominant proline transporter which in S. aureus are, however, PutP and ProT (Lehman et al. 2023). The sequence of OpuB, the membrane component of the system, is not included here. The 3-d structure of OpuC (or a part of it) has been solved (5IIP_A-D).

.

OpuABC of Staphylococcus aureus

 
3.A.1.12.17

OpuFB of 505 aas with 6 or 7 N-terminal TMSs in a 3 + 3 or 4 TMS arrangement.  It is an osmoprotectant ABC transporter/substrate-binding subunit OpuFB.  The B. subtilis ortholog a compatible solute ABC transporter with a substrate-binding protein fused to the transmembrane domain (Teichmann et al. 2018).  There are five transport systems (OpuA, OpuB, OpuC, OpuD, and OpuE) for compatible solutes in B. subtilis (Teichmann et al. 2018). The new system is called OpuF (OpuFA-OpuFB). OpuF is not present in B. subtilis but is widely distributed in members of the genus Bacillus. OpuF mediates the import of glycine betaine, proline betaine, homobetaine, and the marine osmolyte dimethylsulfoniopropionate (DMSP).  It has an aromatic cage, a structural feature commonly present in ligand-binding sites of compatible solute importers (Teichmann et al. 2018).

OpuFB of Bacillus safensis

 
3.A.1.12.2

Glycine betaine OpuAA/AB/AC porter (also transports dimethylsulfonioacetate and dimethylsulfoniopropionate).  The system has been reconstituted in nanodiscs and shows substrate-dependent ionic stringth-gated gating and energy coupling dependent on anionic lipids (Karasawa et al. 2013).

Firmicutes

OpuAA, AB, AC of Bacillus subtilis
OpuAA (C)
OpuAB (M)
OpuAC (R)

 
3.A.1.12.3Choline porterFirmicutesOpuBA, BB, BC, BD of Bacillus subtilis
OpuBA (C)
OpuBB (M)
OpuBC (R)
OpuBD (M)
 
3.A.1.12.4Uptake system for choline, L-carnitine, D-carnitine, glycine betaine, proline betaine, crotonobetaine, γ-butyrobetaine, dimethylsulfonioacetate, dimethylsulfoniopropionate, ectoine and choline-O-sulfateFirmicutesOpuCA, CB, CC, CD of Bacillus subtilis
OpuCA (C)
OpuCB (M)
OpuCC (R)
OpuCD (M)
 
3.A.1.12.5

Uptake system for glycine-betaine (high affinity) and proline (low affinity) (OpuAA-OpuABC) or BusAA-ABC of Lactococcus lactis). BusAA, the ATPase subunit, has a C-terminal tandem cystathionine β-synthase (CBS) domain which is the cytoplasmic K+ sensor for osmotic stress (osmotic strength)while the BusABC subunit has the membrane and receptor domains fused to each other (Biemans-Oldehinkel et al., 2006; Mahmood et al., 2006; Gul et al. 2012). An N-terminal amphipathic α-helix of OpuA is necessary for high activity but is not critical for biogenesis or the ionic regulation of transport (Gul et al., 2012). ATP and glycine betaine dependences of conformational changes have been examined (Tassis et al. 2020).

Firmicutes

BusAA-AB of Lactococcus lactis
BusAA (C-CBS)
BusAB (M-R)

 
3.A.1.12.6Uptake system for hisitidine, proline, proline-betaine and glycine-betaineProteobacteriaHutXWV of Sinorhizobium meliloti
HutX (R)
HutW (M)
HutV (C)
 
3.A.1.12.7High affinity (3 μM) choline-specific uptake system (Dupont et al., 2004)ProteobacteriaChoXWV of Sinorhizobium meliloti
ChoX (R) (AAM00244)
ChoW (M) (AAM00245)
ChoV (C) (AAM00246)
 
3.A.1.12.8A proline/glycine betaine uptake system. Also reported to be a bile exclusion system that exports oxgall and other bile compounds, BilEA/EB or OpuBA/BB (required for normal virulence) (R.D. Sleator et al., 2005). BacteriaOpuBA/BB or BilEA/EB of Listeria monocytogenes
OpuBA (C) (Q93A35)
OpuBB (M-R) (Q93A34)
 
3.A.1.12.9The salt-induced glycine betaine OtaABC transporter (Schmidt et al., 2007)ArchaeaOtaABC of Methanosarcina mazei Go1
OtaA (C) Q8U4S5
OtaB (M) Q8U4S4
OtaC (R) Q8U4S3
 


3.A.1.120 The (Putative) Drug Resistance ATPase-1 (Drug RA1) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.120.1Macrolide ATPase (membrane constituent unknown) Gram-positive bacteriaSrmB (C-C) of Streptomyces ambofaciens
 
3.A.1.120.2Tylosin ATPase (membrane constituent unknown) Gram-positive bacteriaTlrC (C-C) of Streptomyces fradiae
 
3.A.1.120.3Oleandomycin resistance ATPase (membrane constituent unknown)Gram-positive bacteriaOleB (C-C) of Streptomyces antibioticus
 
3.A.1.120.4Carbomycin resistance ATPase (membrane constituent unknown)Gram-positive bacteriaCarbomycin, CarA (C-C), protein of Streptomyces thermotolerans
 
3.A.1.120.5The acetate resistance ABC acetate exporter (Nankano et al., 2006)Gram-negative bacteriaAatA (C-C) of Acetobacter aceti (BAE71146)
 
3.A.1.120.6

The Uup protein (required for bacterial competitiveness (Murat et al., 2008); 39% identical to 3.A.1.120.5).

Gram-negative bacteria

Uup of E. coli (P43672)

 
3.A.1.120.7

ABC transporter, SgvT2 (ATP-hydrolyzing subunit of 551 aas. Functions to export griseoviridin and viridogrisein (etamycin) (Xie et al. 2017). However, it may also function as an ATP-binding cassette domain of elongation factor 3, interacting with the ribosome which stimulates its ATPase activity (Sasikumar and Kinzy 2014).

SgvT2 of Streptomyces griseoviridis

 
3.A.1.120.8

 ABC protein of 558 aas and 0 TMSs, Rv2477c.  It is a translation factor that gates the progression of the 70S ribosomal initiation complex (IC, containing tRNA (fMet) in the P site) into the translation elongation cycle by using a mechanism sensitive to the ATP/ADP ratio. Binds to the 70S ribosome E site where it modulates the state of the translating ribosome during subunit translocation. It is an ABC-F subfamily protein, members of which are implicated in diverse cellular processes such as translation, antibiotic resistance, cell growth and nutrient sensing. Daniel et al. 2018 showed that Rv2477c displays strong ATPase activity (Vmax = 45 nmol/mg/min; Km = 90 muM) that is sensitive to orthovanadate. The ATPase activity was maximal in the presence of Mn2+ at pH 5.2. The protein hydrolyzed GTP, TTP and CTPas well as ATP but at lower rates. Glutamate to glutamine substitutions of amino acid residues 185 and 468 in the two Walker B motifs severely inhibited its ATPase activity. The antibiotics, tetracycline and erythromycin, which target protein translation, were able to inhibit the ATPase activity. Daniel et al. 2018 postulated that Rv2477c is involved in mycobacterial protein translation and in resistance to tetracyclines and macrolides.

v2477c of Mycobacterium tuberculosis

 


3.A.1.121 The (Putative) Drug Resistance ATPase-2 (Drug RA2) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.121.1Erythromycin ATPase (membrane constituent unknown) Gram-positive bacteriaMsrA (C-C) of Staphylococcus epidermidis
 
3.A.1.121.10

ABCF1 (out of 5 isoforms) of 595 aas and 0 TMSs. Functions as a ribosome regulator.

ABCF1 of Arabidopsis thaliana (Mouse-ear cress)

 
3.A.1.121.11

ATP-binding cassette sub-family F member 2, ABCF2 of 623 aas and 0 TMSs.  Its function is unknown, but it is probably not a transporter (Sakamoto et al. 2019).

ABCF2 of Homo sapiens

 
3.A.1.121.2Pristinamycin resistance protein, VgaGGram-positive bacteriaVgaB (C-C) of Staphylococcus aureus
 
3.A.1.121.3Antibiotic (virginiamycin and lincomycin) resistance protein, VmlRGram-positive bacteriaVmlR (C-C) of Bacillus subtilis (P39115)
 
3.A.1.121.5

ABC-type streptogammin A resistance exporter, VgaA of 522 aas and 0 TMSs (C-C arrangement).  Inhibited by pristinamycin IIA (Jacquet et al. 2008). A transport function is not known.

VgaA of Staphylococcus aureus

 
3.A.1.121.6

MsrD of 487 aas and 0 TMSs. Involved in macrolide resistance (Zhang et al. 2016). Two ATPase domains are present in tandem. A membrane constituent is not known. Iannelli et al. 2018 suggested that MefA (TC# 2.a.1.21.1) can function with MsrD, and therefore that this MFS exporter can function as an ABC drug exporter.  However, the data presented seem inconsistent with this suggestion. Nevertheless, the two genes encoding these two proteins are adjacent to each other, suggesting that they may somehow function together (Iannelli et al. 2018).

MsrD of Streptococcus pyogenes (C-C)

 
3.A.1.121.7

Putative ABC protein of 684 aas and 0 TMSs, ATCF1.  Found to be essential for bloodstream-form Trypanosoma brucei through a genome-wide RNAi screen (Schmidt et al. 2018).

ABCF1 of Trypanosoma brucei

 
3.A.1.121.8

ATP-binding cassette subfamily F member 1, ABCF1 or ABC50, of 845 aas and 0 TMSs. There is no transmembrane protein associated with ABCF1, and this protein does not function in transport.  It is required for efficient Cap- and IRES-mediated mRNA translational initiation, not in ribosome biogenesis (Paytubi et al. 2009). ABCF1 regulates dsDNA-induced immune responses in human airway epithelial cells (Cao et al. 2020).

ABCF1 of Homo sapiens

 
3.A.1.121.9

ABCF3 of 709 aas and 0 TMSs. It is not a transporter, but is a translational regulator that also promotes apoptosis (Hirose and Horvitz 2014). It has an antiviral effect against flaviviruses (Sakamoto et al. 2019).

ABCF3 of Homo sapiens

 


3.A.1.122 The Macrolide Exporter (MacB) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.122.1

Macrolide (14- and 15- but not 16-membered lactone macrolides including erythromycin) exporter, MacAB (formerly YbjYZ). Both MacA and MacB are required for activity (Tikhonova et al., 2007). MacAB functions with TolC to export multiple drugs and heat-stable enterotoxin II (enterotoxin STII) (Yamanaka et al., 2008). The crystal structure of MacA is available (Yum et al., 2009). MacB is a dimer whose ATPase activity and macrolide-binding capacity are regulated by the membrane fusion protein MacA (Lin et al., 2009). Xu et al. (2009) have reported the crystal structure of the periplasmic region of MacB which they claim resembles the periplasmic domain of RND-type transporters such as AcrB (TC# 2.A.6.2.2). Also exports L-cysteine (Yamada et al., 2006). The periplasmic membrane proximal domain of MacA acts as a switch in stimulation of ATP hydrolysis by the MacB transporter (Modali and Zgurskaya, 2011). Fitzpatrick et al. 2017 presented an electron cryo-microscopy structure of the tripartite assembly (MacAB-TolC) at near-atomic resolution. A hexamer of the periplasmic protein MacA bridges a TolC trimer in the outer membrane to a MacB dimer in the inner membrane, generating a quaternary structure with a central channel for substrate translocation. A gating ring found in MacA may act as a one-way valve in substrate transport. The MacB structure features an atypical transmembrane domain with a closely packed dimer interface and a periplasmic opening that is the likely portal for substrate entry from the periplasm, with subsequent displacement through an allosteric transport mechanism (Fitzpatrick et al. 2017). The structure of ATP-bound MacB has been solved, revealing precise molecular details of its mechanism (Crow et al. 2017). MacB has a fold that is different from other structurally characterized ABC transporters and uses a unique molecular mechanism termed mechanotransmission. Unlike other bacterial ABC transporters, MacB does not transport substrates across the inner membrane in which it is based, but instead couples cytoplasmic ATP hydrolysis with transmembrane conformational changes that are used to perform work in the extra-cytoplasmic space. In the MacAB-TolC tripartite pump, mechanotransmission drives efflux of antibiotics and export of a protein toxin from the periplasmic space via the TolC exit duct. Homologous tripartite systems from pathogenic bacteria similarly export protein-like signaling molecules, virulence factors and siderophores (Greene et al. 2018).

Gram-negative bacteria

MacAB of E. coli:
MacA(MFP) (P75830)
MacB(C-M) (P75831)

 
3.A.1.122.10

Duf214 (423aas) ABC3 membrane protein with ABC-type ATPase (232 aas).  Sandwiched inbetween the genes encoding these two proteins is a large protein of 869 aas with 2 TMSs, N- and C-terminal. Some homologues are annotated as "S-layer domain protein".  It may be an ABC auxiliary protein.  Most members occur in archaea, but distant homologues are also found in bacteria.

Archaea

Duf214/ABC system of Sulfurisphaera tokodaii (Sulfolobus tokodaii):
Duf214 protein (M) (Q973J4)
ATPase (C) (Q973J6)
Putative auxiliary protein (Q973J5)

 
3.A.1.122.11

The hemin resistance transporter, HrtAB. Expression is activated by hemin or hemoglobin via the ChrAS transmembrane sensor kinase/response regulator system (Bibb and Schmitt 2010). HrtBA extracts heme from the membrane and expells it.  HrtBA consists of two cytoplasmic HrtA ATPase subunits and two transmembrane HrtB permease subunits. A heme-binding site is formed in the HrtB dimer and is laterally accessible to heme in the outer leaflet of the membrane. The heme-binding site captures heme from the membrane using a glutamate residue of either subunit as an axial ligand and sequesters the heme within the rearranged transmembrane helix bundle. By ATP-driven HrtA dimerization, the heme-binding site is squeezed to extrude the bound heme (Nakamura et al. 2022).

Bacteria

HrtAB of Corynebacterium diphtheriae
HrtA (C) (H2GZC3)
HrtB (M) (H2GZC4) 

 
3.A.1.122.12

Arthrofactin efflux pump, ArfDE (Balibar et al. 2005).

γ-Proteobacteria

ArfDE of Pseudomonas sp. MIS38
ArfD (MFP) (Q84BQ3)
ArfE (ABC) (A0ZUB1)

 
3.A.1.122.13Putative ABC3-type antimicrobial peptide transporter, fused ATPase-porter protein, U-ABC3-1b (667aas; 4TMSs:1+3)BacteriaU-ABC3-1b of Lactobacillus brevis (CM) (Q03RZ6)
 
3.A.1.122.14

ABC transporter of unknown function, but aspects of its structure and mechanism of action are known (Yuan et al. 2001; Zoghbi and Altenberg 2013).  Nucleotide-binding domain dimerization occurs as a result of binding to the natural nucleotide triphosphates, ATP, GTP, CTP and UTP, as well as the analog ATP-gamma-S. All the natural nucleotide triphosphates are hydrolyzed at similar rates, whereas ATP-gamma-S is not hydrolyzed. The non-hydrolyzable ATP analog AMP-PNP, frequently assumed to produce the nucleotide-bound conformation, failed to elicit nucleotide-binding domain dimerization (Fendley et al. 2016).

Archaea

ABC transporter of Methanocaldococcus jannaschii (Methanococcus jannaschii)
Membrane protein, MJ0797 (M) (Q58207)
ATPase, MJ0796 (C) (Q58206)

 
3.A.1.122.15

Putative heavy metal ion exporter, YbbAB (Moussatova et al. 2008).

Proteobacteria

YbbAB of E. coli
YbbA (C; 228 aas)
YbbB (M; 804 aas)

 
3.A.1.122.16

Putative macrolide-specific efflux system, MacAB

MacAB of Bifidobacterium longum

 
3.A.1.122.17

LolC/E family lipoprotein releasing system, transmembrane protein of 639 aas and 4 TMSs

LolC/E family lipoprotein releasing system, transmembrane protein of Candidatus Saccharibacteria bacterium

 
3.A.1.122.18

MacAB-TolC MDR effllux porter. Exports macrolide antibiotics, virulence factors, peptides and cell envelope precursors. The 3-d crystal structure of MacB has been solved at 3.4 Å resolution (Okada et al. 2017). MacB forms a dimer in which each protomer contains a nucleotide-binding domain and four TMSs that protrude in the periplasm into a binding domain for interaction with the membrane fusion protein MacA. It has unique structural features (Okada et al. 2017).

MacAB of Acinetobacter baumannii
MacA, Q2FD52, 445 aas and 1 TMS
MacB, N9J6M5, 664 aas and 4 TMSs

 
3.A.1.122.19

ABC3-type efflux porter, YtrEF, encoded within an operon, ytrABCDEF, apparently encoding two ABC exporters, one, YtrBCD, with TC# 3.A.1.153.1, and the other, this one. The operon is induced in early stationary phase under the control of YtrA, a GntR-type HTH transcriptional regulator, probably a repressor (Yoshida et al. 2000). These authors suggest this operon may be involve in acetoin secretion and/or reutilization.

YtrEF of Bacillus subtilis
YtrE, C, 231 aas; O34392
YtrF, M, 436 aas; O35005

 
3.A.1.122.2

The SpdC antimicrobial peptide resistance efflux pump, YknXYZ (Butcher and Helmann, 2006).  YknW (TC# 9.B.29.2.17), a 5 TMS protein, interacts directly with YknXYZ and is essential for facilitation of its assembly, thus serving as an integral membrane chaparone  (Yamada et al., 2012). The MFP YknX requires the ATP-binding cassette (ABC) transporter YknYZ and the Yip1 family protein YknW to form a functional complex. YknX (MFP) is hexameric (Xu et al. 2017).

Bacteria

YknXYZ of Bacillus subtilis
YknX (MFP) (O31710)
YknY (C) (O31711)
YknZ (M) (O31712)

 
3.A.1.122.20

MacAB-MFP complex of 3 subunits involved in the resistance of antibiotics and antimicrobial peptides. Yang et al. 2018 reported the crystal structures of Spr0694-0695 (MacAB) at 3.3 Å and Spr0693 (MFP; TC# 8.A.1) at 3.0 Å resolution, revealing a MacAB-like efflux pump. The dimeric MacAB adopts a non-canonical fold, the transmembrane domain of which consists of a dimer with eight tightly packed TMSs (4 per subunit) with an extracellular domain between the first and second helices, whereas Spr0693 (the MFP) forms a nanotube channel docked onto the ABC transporter. Structural analyses, combined with ATPase activity and antimicrobial susceptibility assays, enabled the proposal of a putative substrate-entrance tunnel with lateral access controlled by a guard helix (Yang et al. 2018).

MacAB-MFP of Streptococcus pneumoniae
MacA, Spr0694, 233 aas (C)
MacB, Spr0695, 419 aas (M)
MFP, Spr0693, 399 aas, (MFP)

 
3.A.1.122.21

ABC transport system with a type 3 ABC membrane protein (386 aas and 4 TMSs; B8GHI1) and an ABC ATPase (234 aas; B8GHI2).  The encoding genes are adjacent to those encoding a putative transport system with TC# 9.B.29.2.7.

ABC transporter of Methanocorpusculum labreanum

 
3.A.1.122.22

Uncharacterized ABC exporter of two subunits, a 4 TMS membrane subunit of 177 aas, and an ATPase of 229 aas

UP ABC exporter
(M) 177 aas and 4 TMSs, KKK40843
(C)  229 aas and ) TMSs, KKK48044

 
3.A.1.122.23

Uncharacterized ABC exporter

Uncharacterized ABC exporter of Candidatus Thorarchaeota
(M) 173 aas and 4 TMSs, RDE13437
(C) 225 aas, RDE13438

 
3.A.1.122.24

Uncharacterized ABC exporter

ABC exporter of Candidatus Odinarchaeota
(M) 166 aas and 4 TMSs, OLS17116
(C)  232 aas, OLS17115

 
3.A.1.122.25

3-component ABC3-type transporter with two 4 TMS membrane proteins and one ATPase, all encoded within a single operon with the three genes next to each other.

ABC exporter of Corallococcus coralloides

 
3.A.1.122.26

Uncharacterized two comoponent ABC3-type efflux transporter of 805 aas and 8 TMSs in a 1 + 3 + 1 +3 TMS arrangement. The ATPase is a distinct protein of 250 aas.

Uncharacterized ATP-energized exporter of Candidatus Heimdallarchaeota
ABC3-type membrane protein of 805 aas and 8 TMSs (M)
ATPase of 250 aas (C).

 
3.A.1.122.27

Putative ABC3-type transporter with an ATPase and a possible auxiliary protein encoded by a gene sandwiched in between the membrane protein and the ATPase. Some homologues of the auxiliary protein are annotated as S-layer domain proteins. This system resembles 3.A.1.122.10 which also has such an auxiliary protein.

ABC3-type transporter
E6NBB1 (M), 413 aas with 4 TMSs in a 1 + 3 TMS arrangement
E6NBB0 (C), 236 aas
E6N374 (Putative auxiliary protein), 597 aas and 1 TMS at the C-terminus

 
3.A.1.122.28

ABC3 exporter including a membrane protein of 392 aas and 4 TMSs in a 1 + 3 TMS arrangement and a putative auxiliary transport protein of 944 aas and 1 C-terminal TMS. It is annotated as an S-layer domain protein. While these two recognized proteins are encoded by adjacent genes, an ATPase was not encoded nearby, and it has not been identified.

Putative incomplete ABC3 exporter of Ignicoccus hospitalis

 
3.A.1.122.29

ABC3-type exporter with 3 components, the permease of 457 aas with 4 TMSs, an ATPase of 236 aas, and a putative auxiliary protein of 805 aas and 2 TMSs, N-and C-terminal. The permease subunit is annotated as an ABC-type lipoprotein release transport system, and the auxiliary protein is a COG1361 protein.

ABC3 porter of Anaerobacterium chartisolvens

 
3.A.1.122.3The enterocin AS-48 exporter, As-48FGHGram-positive bacteriaAs-48FGH on plasmid pMBL of Enterococcus faecalis:
As-48F (MFP) (Q7AUQ4)
As-48H (M) (Q8RKC0)
As-48G (C) (Q8RKC1)
 
3.A.1.122.30

ABC3 exporter with three constituents, the 4 TMS membrane protein of 529 aas, the ATPase of 251 aas, and an auxiliary protein of 431 aas and 2 TMSs, N-terminal and C-terminal.

ABC3 exporter of Bifidobacterium longum subsp. infantis

 
3.A.1.122.31

Uncharacterized ABC porter with a single membrane protein of 224 aas and 4 TMSs, plus two ATPases, of 237 and 243 aas, respectively.

ABC porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)
Porter, M, OLS20810
ATPase, C, OLS20809
ATPase, C, OLS20808

 
3.A.1.122.32

Putative 3-component ABC exporter with two uncharacterized homologous membrane proteins of 930 aas and 1090 aas, both with 10 TMSs in a 1 + (3 +2 +1) +3 TMS arrangement, plus an ATPase.

Actinobacteria

Uncharacterized  ABC exporter of Cellulomonas flavigena

 
3.A.1.122.33

Putative 3 component ABC transporter with two membrane proteins of 907 and 1113 aas plus an ATPase of 319 aas.  Both membrane proteins have 10 TMSs in a 1 + (3 + 2 + 1) + 3 TMS arrangement.

ABC exporter of Streptomyces coelicolor

 
3.A.1.122.34

Uncharacterized putative ABC exporter of 4 components, all encoded by adjacent genes: one membrane protein, two ATPases and one membrane fusion protein (MFP).

ABC exporter of Paenibacillus mucilaginosus

 
3.A.1.122.35

The MacAB drug exporter.  MacB is an ABC transporter that collaborates with the MacA adaptor protein (a membrane fusion protein, MFP) and the TolC exit duct to drive efflux of antibiotics and enterotoxin STII out of the bacterial cell. Crow et al. 2017 presented the structure of ATP-bound MacB and reveal precise molecular details of its mechanism. The MacB transmembrane domain lacks a central cavity through which substrates could be passed, but instead conveys conformational changes from one side of the membrane to the other, a process termed mechanotransmission. Comparison of ATP-bound and nucleotide-free states revealed how reversible dimerization of the nucleotide binding domains drives opening and closing of the MacB periplasmic domains via concerted movements of the second transmembrane segment and the major coupling helix. They proposed that the assembled tripartite pump acts as a molecular bellows to propel substrates through the TolC exit duct, driven by MacB mechanotransmission. Homologs of MacB that do not form tripartite pumps, but share structural features underpinning mechanotransmission, include the LolCDE lipoprotein trafficking complex and FtsEX cell division signaling protein. The MacB architecture serves as a blueprint for understanding the structure and mechanism of an entire ABC transporter superfamily and the many diverse functions it supports (Crow et al. 2017). The crystal structure of MacA has been solved (Yum et al. 2009).

MacAB of Aggregatibacter actinomycetemcomitans (Actinobacillus actinomycetemcomitans)

 
3.A.1.122.36

ABC-type antimicrobial peptide transport system with two components, one having a domain structure of C-M and 653 aas with 4 TMSs, and the other being a membrane fusion protein (see TC# 8.A.1) (Cho and Kang 2012). A mutant showed significant reduction in secretion of syringomycin (74%) and syringopeptin (71%), as compared to the parental strain (Cho and Kang 2012). The PseEF efflux system has a role in secretion of syringomycin and syringopeptin, and is required for full virulence in P. syringae pv. syringae.

PseEF of Pseudomonas syringae

 
3.A.1.122.37

ABC transporter permease of 831 aas and 8 TMSs in a 1 + 3 + 1 + 3 TMS arrangement.

ABC permease of Gemmatimonadota bacterium (marine sediment metagenome)

 
3.A.1.122.4Probable Heme exporter, HrtAB (Stauff et al., 2008)BacteriaHrtAB of Staphylococcus aureus:
HrtA (C) (Q7A3X3)
HrtB (M) (Q7A7X2)
 
3.A.1.122.5ABC transporter of unknown function (DUF214 protein) (4TMSs)/ABC protein [Msed1528/Msed1530] ArchaeaMsed1528/Msed1530 of Metallosphaera sedula (M) (A4YGY2)
 
3.A.1.122.6ABC transporter of unknown function (DUF214 protein) (4TMSs)/ABC protein [MA2839/MA2840]ArchaeaMA2839/MA2840 of Methanosarcina acetivorans
MA2839 (M) (Q8TM31)
MA2840 (C) (Q8TM30)
 
3.A.1.122.7ABC transporter of unknown function (Duf214 protein (409aas; 4TMSs:1+3)/ABC protein)ArchaeaDuf214 protein/ ABC protein of Methanococcus voltae:
Duf214 protein (M) (A8TDX0)
ABC protein (C) (A8TDW7)
 
3.A.1.122.8Putative ABC3 permease, PC1,2,3.BacteriaPC1,2,3 of Treponema denticola:
PC1 (C) - Q73MJ2
PC2 (M) - Q73MJ3
PC3 (M) - Q73MJ4
 
3.A.1.122.9Duf214 protein (405aas)/ ABC proteinArchaeaDuf214/ABC system of Caldivirga maquilingensis:
Duf214 protein (M) (A8M8Z1)
 


3.A.1.123 The Peptide-4 Exporter (Pep4E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.123.1

Pep5 lantibiotic exporter, PepT

Gram-positive bacteria

PepT (M-C) of Staphylococcus epidermidis

 
3.A.1.123.2Aureocin A70 multipeptide bacteriocin (AurA, AurB, AurC, AurD) exporter, AurTGram-positive bacteriaAurT (M-C) of Staphylococcus aureus
 
3.A.1.123.3The one component lantibiotic exporter, GdmT (Sibbald et al., 2006) Gram-positive bacteriumGdmT (M-C) of Staphylococcus gallinarum (A3QNP2)
 


3.A.1.124 The 3-component Peptide-5 Exporter (Pep5E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.124.1

The 3-component nisin immunity exporter, NisFEG. Contains an essential E-loop (Okuda et al., 2010).

Gram-positive bacteria

NisFEG of Lactococcus lactis
NisF (C)
NisE (M)
NisG (M)

 
3.A.1.124.2The 3-component subtilin immunity exporter, SpaEFGGram-positive bacteriaSpaEFG of Bacillus subtilis
SpaE (M)
SpaF (C)
SpaG (M)
 
3.A.1.124.3The lantibiotic Nukacin ISK-1 (TC# 1.C.21.1.5)/NukH (BAD01013; 92aas) exporter, NukEFG (Okuda et al., 2008)Gram-positive bacteriaNukEFG of Staphylococcus warneri
NukE (M) (Q75V14)
NukF (C) (Q75V15)
NukG (M) (Q75V13)
 
3.A.1.124.4The macedocin exporter, McdEFG (Papadelli et al., 2007) Gram-positive bacteriaMcdEFG of Streptococcus macedonicus
McdE (M; 254 aas) (A6MER6)
McdG (M; 245 aas) (A6MER7)
McdF (C; 304 aas) (A6MER5)
 
3.A.1.124.5The salivaricin exporter, SboEFG (Hyink et al., 2007)Gram-positive bacteriaSboEFG of Streptococcus salivarius
SboE (M; 249 aas) (Q09IH9)
SboF (C; 303 aas) (Q09II0)
SboG (M; 242 aas) (Q09IH8)
 
3.A.1.124.6

CprABC antimicrobial peptide resistance ABC exporter.  Exports both mammalian and bacterial toxic peptides (McBride and Sonenshein 2011).

Firmicutes

CprABC of Clostridium difficile
CprA (C, 235 aas)
CprB (M, 238 aas, 6 TMSs)
CprC (M, 252 aas, 6 TMSs)

 


3.A.1.125 The Lipoprotein Translocase (LPT) Family (This TC subfamily overlaps with TC# 3.A.1.122)


Examples:

TC#NameOrganismal TypeExample
3.A.1.125.1

Lipoprotein translocation system (translocates lipoproteins from the inner membrane to periplasmic chaperone, LolA, which transfers the lipoproteins to an outer membrane receptor, LolB, which anchors the lipoprotein to the outer membrane of the Gram-negative bacterial cell envelope) (see 1.B.46; Narita et al., 2003; Ito et al., 2006; Watanabe et al., 2007). The structure of ligand-bound LolCDE has been solved (Ito et al., 2006). LolC and LolE each have 4 TMSs (1+3). Unlike most ATP binding cassette transporters mediating the transmembrane flux of substrates, the LolCDE complex catalyzes the extrusion of lipoproteins anchored to the outer leaflet of the inner membrane. The LolCDE complex is unusual in that it can be purified as a liganded form, which is an intermediate of the lipoprotein release reaction (Taniguchi and Tokuda, 2008). LolCDE has been reconstituted from separated subunits (Kanamaru et al., 2007).  LolE binds the outer membrane lipoprotein, PAL (Mizutani et al. 2013). The mechanism of LolCDE as a molecular extruder of bacterial triacylated lipoproteins has been reported (Sharma et al. 2021) who determined the cryo-EM structures of nanodisc-embedded LolCDE in the nucleotide-free and nucleotide-bound states at 3.8-Å and 3.5-Å resolution, respectively. The structural analyses, together with biochemical and mutagenesis studies, uncovered how LolCDE recognizes its substrate by interacting with the lipid and N-terminal peptide moieties of the lipoprotein, and identify the amide-linked acyl chain as the key element for LolCDE interaction. Upon nucleotide binding, the transmembrane helices and the periplasmic domains of LolCDE undergo large-scale, asymmetric movements, resulting in extrusion of the captured lipoprotein. Comparison of LolCDE and MacB revealed the conserved mechanism of type VII ABC transporters and emphasized the unique properties of LolCDE as a molecule extruder of triacylated lipoproteins (Sharma et al. 2021).

Gram-negative bacteria

LolCDE of E. coli
LolC (M)
LolD (C)
LolE (M)

 
3.A.1.125.2Putative lipoprotein LolCDE homologue LolCE (10TMSs:1+6+3)/LolDBacteriaLolCE/LolD of Mycobacterium tuberculosis
LolCE (M) (Q7D911)
LolD (C) (O53899)
 
3.A.1.125.3Duf214 protein (843aas; 10TMSs:1+6+3)BacteriaDuf214 protein/ ABC protein of Frankia sp. CcI3:
Duf214 protein (M) - Q2J9P4
[LolD/FtsE/SalX]-type ABC protein (C) - Q2J9P5
 
3.A.1.125.5

Uncharacterized ABC transporter with two consituents, a 4 TMS (in a 1 + 3 TMS arrangement) membrane (M) protein and an ATPase (C). 

Uncharacterized ABC transporter of Opitutus terrae
M: B1ZMT9
C: B1ZMU0

 
3.A.1.125.6

Putative ABC transporter, LolCDE, with three components, similar to (but substantially different from) LolC, LolD and LolE of E. coli. The three genes encoding these proteins are adjectent to each other on the bacteria chromosome, but there is no direct experimental evidence that they function together as lipoprotein exporters.

LolCDE of Candidatus Heimdallarchaeota archaeon LC_3
LolC, 690 aas and 4 TMSs in a 1 + 3 TMS arrangement (MC)
LolD, 236 aas with 2 or 3 TMSs followed by a hydrophilic C-terminal domain (MC)
LolE, 1106 aas and 12 TMSs in a 1 + 3 + 4 +1 + 3 TMS arrangement (MM)

 
3.A.1.125.7

ABC-type transport system, possibly involved in lipoprotein release. There are two and possibly three protein constituents. First, there is a permease component of 1004 aas with 13 TMSs in a 2 (N-terminal) + 3 +4 + 1 + 3 TMS arrangement (I2F7A7)).  This protein is unusual because it has 2 N-terminal TMSs rather than the usual 1, and between the two 1 + 3 TMS repeat units, there are 4 TMSs rather than the more usual 0 or 2 TMSs. Second, there is an ABC-type ATPase (I2F7A6) and third, there is a possible auxiliary subunit of 183 aas and 4 TMSs in a 2 + 2 TMS arrangement that appears to be two repeated 2 TMS sequences, one near the N-terminus and one near the C-terminus; I2F7A5).  Adjacent to these genes is one annotated as a "PRC barrel protein" (with a single N-terminal TMS and is a conserved, ubiquitous, chromo domain, shared by photosynthetic reaction center subunits and proteins of RNA metabolism (Anantharaman and Aravind 2002); I2F7A4) of unknown function, and adjacent to that gene are two genes encoding putative chromate resistance efflux (transport) protein, ChrA (I2F7A3 and I2F7A1; see TC family 2.A.51).

ABC type 3 transporter of Mesotoga prima MesG1 Ag.4.2

 

 
3.A.1.125.8

FtsX-type ABC transporter of 902 aas and 10 TMSs in a 1 + 3 + 2 + 1 + 3 TMS arrangement.

ABC transporter of Vescimonas coprocola

 
3.A.1.125.9

ABC transporter of 1253 aas and 9 TMSs in a 1 + 3 +2 + 2 + 1 TMS arrangement.

ABC transporter of Exiguobacterium undae

 


3.A.1.126 The β-Exotoxin I Exporter (βETE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.126.1Exporter of β-exotoxin I, BerABBacteriaβ-exotoxin exporter, BerAB, of Bacillus thuringiensis
BerA (C)
BerB (M)
 
3.A.1.126.2

Putative ABC transporter with a 6 TMS membrane protein and an ATPase of the ABC-type encoded by the adjacent gene.

Bacteria

Putative ABC transporter of Arthrobacter (Paenarthrobacter) aurescens (A1R938)

 
3.A.1.126.3

Putative exporter of polyketide antibiotic-like protein (~12 TMSs) with an ABC ATPase encoded by the adjacent gene.

Actinobacteria

Putative exporter of Amycolicicoccus (Hoyosella) subflavus (F6EHL8)

 
3.A.1.126.4

6TMS putative ABC transporter protein with an ABC-type ATPase encoded by the adjacent gene.  This memebrane protein also maps adjacent to protein fragments that show similarity to ABC transport proteins as well as a protease (9.B.218.1.4; D4TYE3).

Bacteria

Putative ABC transporter system of Actinomyces odontolyticus (D4TYE0)

 
3.A.1.126.5

Polyether inonophore exporter, NarAB:  NarA, ATPase (C) of 293 aas and NarB, membrane protein (M) of 537 aas and 12 TMSs. The polyether ionophores, narasin, salinomycin, and maduramicin, but not monensin, are actively exported (A-O Naemi et al., NarAB Is an ABC-type Transporter That Confers Resistance to the Polyether Ionophores Narasin, Salinomycin, and Maduramicin, but Not Monensin, 2020 Front Microbiol.).

NarAB of Enterococcus faecium

 
3.A.1.126.6

Cereulide (cyclic depsipeptide) K+ ionophore exporter, CerCD. CerC is 291 aas without TMSs, while CerD is 268 aas with 5 or 6 TMSs (Yuan et al. 2024).

CerCD of the Bacillus cereus group

 


3.A.1.127 The AmfS Peptide Exporter (AmfS-E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.127.1Exporter of AmfS extracellular peptidic morphogen (Chater and Horinouchi, 2003; Ueda et al., 2002)BacteriaAmfS exporter, AmfAB of Streptomyces griseus
AmfA (MC) (BAA33537)
AmfB (MC) (BBA33538)
 


3.A.1.128 The SkfA Peptide Exporter (SkfA-E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.128.1

Exporter of SkfA processed peptide (spO31422), SkfEF (González-Pastor et al., 2003)

Bacteria

SkfEF (YbdAB) of Bacillus subtilis
SkfE (C) O31427
SkfF (M-M) O31438

 
3.A.1.128.10

Apparent two component ABC transporter, probably an exporter of the sporulation killing factor, SkfB. The membrane component has 12 TMSs, and therefore is probably either due to an intragenic duplication of the usual 6 TMS domain protein or a fusion of two such proteins that are more usual for this subfamily of ABC exporters.  This system acts in conjunction with a CAAX protease (EMI14127; TC# 9.B.2.13.1) that presumably processes SkfB to the mature form. The two genes of this ABC system are preceded by a gene coding for a 174 aa proteins possibly involved in SkfB synthesis and/or export.

ABC SkfB exporter of Bacillus stratosphericus

 
3.A.1.128.11

ABC exporter, possibly for the sporulation killer factor SkfB.

ABC exporter of Parageobacillus thermoglucosidasius

 
3.A.1.128.2Putative ABC exporter, Teth 514-0346 & 0347

Bacteria

Teth 514-0346 & 0347 of Thermoanaerobacter sp. x514:
Teth514-0346 (C) (B0K2P2)
Teth514-0347 (M-M) (B0K2P3)

 
3.A.1.128.3Putative ABC exporter, CLK2533/CLK2534

Bacteria

CLK2533/CLK2534 of Clostridium botulinum
CLK2533 (M-M) (B1L0U0)
CLK2534 (C) (B1L0U1)

 
3.A.1.128.4Putative ABC exporter Tiet1371/1372

Bacteria

Tiet1371/72 of Thermotoga lettingae
Tiet1371 (M-M) (A8F6Z4)
Tiet1372 (C) (A8F6Z5)

 
3.A.1.128.5

Putative ABC transporter.  The genes encoding this system map adjacent to a beta-lactamase (A9BGZ6) gene and one encoding a C4 anaerobic dicarboxylate carrier (A9BGZ7).

Thermatogae

Putative ABC transporter of Petrotoga mobilis

 
3.A.1.128.6

Putative ABC exporter

Euryarchaea

ABC exporter of Pyrococcus horikoshii
Membrane protein (M) (O58947)
ATPase (C) O58948)

 
3.A.1.128.7

Uncharacterized ABC permease, TA0065/TA0066

Euryarchaea

UP of Thermoplasma acidophilum
TA0065 (M-M; permease; 515 aas, 12 TMSs)
TA0066 (C; ATPase)

 
3.A.1.128.8

ABC transporter encoded by two adjacent genes, a membrane protein and an ABC ATPase.

ABC transporter
(M) KXH73395
(C)  KXH73394

 
3.A.1.128.9

Three component ABC transport system of unknown function.

ABC porter of Paenibacillus larvae subsp. pulvifaciens

 


3.A.1.129 The CydDC Cysteine Exporter (CydDC-E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.129.1

Heme transporter (previously proposed to be a thiol (cysteine/glutathione) exporter, CydDC; CydC is also called MdrH (periplasmic cysteine is required for cytochrome bd assembly) (Cruz-Ramos et al., 2004).  The purified asymmetric heterodimer exhibits low ATPase activity which is activated by both thiols and heme (e.g., heme b) (Yamashita et al. 2014).  Bacterial redox homoeostasis during nitrosative stress is influenced by CydDC.  Periplasmic low molecular weight thiols restore haem incorporation into a cytochrome complex (Holyoake et al. 2016). Iron-bound cyclic tetrapyrroles (hemes) are redox-active cofactors in bioenergetic enzymes. Wu et al. 2023 used cellular, biochemical, structural and computational methods to characterize CydDC which is a heme transporter required for functional maturation of cytochrome bd. The conformational landscape of CydDC during substrate binding and occlusion was revealed. Heme binds laterally from the membrane space to the transmembrane region of CydDC, enabled by a highly asymmetrical inward-facing CydDC conformation. During the binding process, heme propionates interact with positively charged residues on the surface and later in the substrate-binding pocket of the transporter, causing the heme orientation to rotate 180° (Wu et al. 2023).

Bacteria

CydDC of E. coli
CydD (M-C) (P29018)
CydC (M-C) (P23886)

 


3.A.1.13 The Vitamin B12 Uptake Transporter (B12T) Family (Similar to 3.A.1.14)


Examples:

TC#NameOrganismal TypeExample
3.A.1.13.1

Vitamin B12 porter. The 3-D structure of BtuCDF has been solved to 2.6 Å (Hvorup et al., 2007). The conformational transition pathways of BtuCD has been revealed by targeted molecular dynamics simulations (Weng et al., 2012). Asymmetric states of BtuCD are not discriminated by its cognate substrate binding protein BtuF (Korkhov et al., 2012).  ATP hydrolysis occurs at the nucleotide-binding domain (NBD) dimer interface, whereas substrate translocation takes place at the translocation pathway between the TM subunits, which is more than 30 angstroms away from the NBD dimer interface.  Hydrolysis of ATP appears to facilitate substrate translocation by opening the cytoplasmic end of translocation pathway (Pan et al. 2016). The molecular mechanism of ATP hydrolysis by BtuCD-F may proceeds in a stepwise manner (Prieß et al. 2018). First, nucleophilic attack of an activated lytic water molecule at the ATP gamma-phosphate yields ADP + HPO42-. A conserved glutamate located close to the gamma-phosphate transiently accepts a proton acting as a catalytic base. In the second step, the proton transfers back from the catalytic base to the gamma-phosphate, yielding ADP + H2PO4-. These two reaction steps are followed by rearrangements of the hydrogen bond network and the coordination of the Mg2+ ion. The overall free energy change of the reaction is close to zero, suggesting that ATP binding is essential for tight dimerization of the nucleotide-binding domains and the transition of the transmembrane domains from inward- to outward-facing. ATP hydrolysis resets the conformational cycle (Prieß et al. 2018).

Proteobacteria

BtuCDF of E. coli
BtuC (M)
BtuD (C)
BtuF (R)

 
3.A.1.13.2

Putative cobalamin (vitamin B12) uptake porter, BtuFCD (Rodionova et al. 2015).

BtuFCD of Chloroflexus aurantiacus
BtuF (R; 1 TMS)
BtuC (M; 9 TMSs)
BtuD (C; 0 TMSs)

 


3.A.1.130 The Multidrug/Hemolysin Exporter (MHE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.130.1The multidrug/hemolysin exporter, CylA/B (note: CylK (AAF01071) may influence its activity)(Gottschalk et al., 2006)BacteriaCylA/B of Streptococcus agalactiae
CylA (C) (Q9X432)
CylB (M) (Q9X433)
 
3.A.1.130.2

ABC export system, possibly an MDR pump, consisting of two proteins, and membrane protein of 293 aas and 6 TMSs, and an ATPase of 301 aas.

ABC exporter of Acidipropionibacterium virtanenii

 
3.A.1.130.3

ABC exporter with two constituents, a membrane protein of 263 aas and 6 TMSs, and an ATPase of 310 aas.

ABC exporter of Lactobacillus hokkaidonensis

 


3.A.1.131 The Bacitracin Resistance (Bcr) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.131.1

The 2 or 3 component bacitracin-resistance efflex pump, BcrAB or BcrABC (Podlesek et al., 1995; Bernard et al., 2003) (BcrA is most similar to SpaF (3.A.1.124.2), but BcrB (5-6 TMSs) is only distantly related to other ABC2-type membrane proteins (Wang et al., 2009). BcrC is not sufficiently similar to detect similarity in BLAST searches. BcrC (5TMSs) belongs to the PAP2 phosphatase superfamily and may not be a contituent of the BcrAB transporter. Transcription is regulated by BcrR, a one-component transmembrane signal transduction system (Darnell et al. 2019).

Bacteria

BcrABC of Bacillus licheniformis
BcrA (C) - (P42332)
BcrB (M) - (P42333)

 
3.A.1.131.2

Lantibiotic immunity system, LanEF. Contains an essential E-loop, a variant of the Q-loop, well conserved in nucleotide binding domains of lantibiotic exporters (Okuda et al., 2010).

Gram-positive bacteria

LanEF of Bacillus licheniformis
LanE (M) (Q65DD3)
LanF (C) (Q65DD1)

 
3.A.1.131.3

Transporter homologue, Tiet1372

Bacteria

Tiet1372 of Thermotoga lettingae (A8F6Z5)

 


3.A.1.132 The Gliding Motility ABC Transporter (Gld) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.132.1

The GldAFG putative ABC transporter required for ratchet-type gliding motility; may function in secretion of a macromolecule such as an exopolysaccharide. (Agarwal et al., 1997; Hunnicutt et al., 2002; McBride and Zhu 2013). Soluble GldG homologues (no TMSs) are found in eukaryotes (e.g. intraflagellar protein transporter, IPT52 of Chlamydomonas reinhardtii; XP_001692161)

Bacteria

GldAFG of Flavobacterium johnsoniae:
GldA (C; 298 aas) - (O30489)
GldF (M; 241 aas; 6TMSs (2+2+2) - (Q93LN1)
GldG (M-periplasm; putative auxillary subunit with 2TMSs at the N and C-termini; 561 aas)- (Q93LN0).

 
3.A.1.132.10

Putative ABC exporter of unknown function, Gll1303/Gll1302, with two probable subunits of 477 and 494 aas with 6 TMSs each at their N-termini (M) and ATPase domains (C) in the C-termini.

Cyanobacteria

Gll1303/Gll1304 putative ABC exporter of Gloeobacter violaceus
Gll1303, (M)
Gll1302, (M)

 
3.A.1.132.11

Putative ABC exporter with two membrane proteins of 478 and 417 aas and 6 TMSs respectively, and one ATPase.  The encoding genes are adjacent to a TonB-dependent OMR with possible specificity for a siderophore.  Thus, this ABC exporter could transport a siderophore.

Proteobacteria

Uncharacterized ABC exporter of Saccharophagus degradans
Sde_3610 (C), 249 aas (Q21EL4)
Sde_3609 (M), 478 aas and 6 TMSs (Q21EL5)
Sde_3608 (M), 417 aas and 6 TMSs (Q21EL6)

 
3.A.1.132.12

ABC exporter necessary for social motility, pilus assembly and pilus subunit (PilA) export, PilGHI. Mutants show elevated sporulation rates and abnormal development (Wu et al. 1998).

Proteobacteria

PilHI of Myxococcus xanthus
PilH (C) ABC protein (O30385)
PilT (M) 6 TMS membrane protein of 255aas (O30386) 

 
3.A.1.132.13

ABC transporter permease, membrane subunit of 736 aas and 7 N-terminal TMSs in a 2 + 3 + 2 TMS arrangement and a large hydrophilic C-terminal domain, similar to those in TC family 9.B.359, but with poor sequence similarity with these proteins.

ABC membrane subunit from Mahella australiensis

 
3.A.1.132.14

Putative ABC exporter with three components, two 6 TMS proteins and one ATPase, all encoded by genes that are adjacent to each other. The first of these (G2SET3) is a MusI homologue of 229 aas and 6 TMSs.

Bacteroidetes

MusI homologue of Rhodothermus marinus

 
3.A.1.132.2The NosDFY Copper ABC transporter (Chan et al., 1997)BacteriaNosDFY of Sinorhizobium meliloti
NosD (R; periplasmic copper binding receptor)(Q52899)
NosF (C; like GldA) (Q52900)
NosY (M; like GldF) (O07330)
 
3.A.1.132.3

The uncharacterized ABC transporter with GldF-GldG homologues fused.  The adjacent gene encodes the ATPase, GldA, and the next gene encodes an auxiliary protein of the MPA1-C family (TC# 8.A.3).

Bacteria

GldAFG homologues of Magnetococcus sp. MC-1
GldFG (M-Aux; 964 aas) (A0L4K8)
GldA (C; 399 aas) (A0L4L0)

 
3.A.1.132.4The uncharacterized ABC transporter with GldF-GldG homologues fusedBacteriaGldAFG homologues of Hahella chejuensis
GldF-G (M-Aux; 978 aas) (Q2SDB0)
GldA (C; 315 aas) (Q2SDB1)
 
3.A.1.132.5

Putative ABC2 transporter: Membrane protein of 274aas and 6 TMSs; Cytoplasmic ATPase of 302aas.

Proteobacteria

Putative ABC2 transporter of Shewanella pealeana
(M) (A8GZV3)
(C) (A8GZV2) 

 
3.A.1.132.6

Putative ABC2 transporter: Membrane protein of 274aas and 6 TMSs; Cytoplasmic ATPase of 302aas.

Firmicutes

Putative ABC-2 transporter of Streptococcus pyogenes 
(M) (Q99ZC7)
(C) (Q99ZC8) 

 
3.A.1.132.7

Putative ABC membrane protein with 12 TMSs. (ATPase subunit unknown, and not encoded by an adjacent gene).

Planctomycetes

ABC membrane protein of Rhodopirellula baltica

 
3.A.1.132.8

ABC transporter, annotated as involved in multi copper protein maturation

Archaea

ABC exporter of Methanocella conradii
permease (M) (H8I780)
ATPase (C) (H8I779)

 
3.A.1.132.9

Putative ABC exporter, Odosp_3144/Odosp_3145. Odosp_3144 is a 6 TMS ABC2 membrane protein (N-terminal 250 aas) fused to an auxiliary protein with one N- and one C-terminal TMS, homologous to GldG of Cytophaga johnsonae (3.A.1.132.1).

Bacteroidetes

Putative ABC transporter of Odoribacter splanchnicus 
Odosp_3144 (M) (761 aas; 7 TMSs) (F9Z892)
Odosp_3145 (C) (306 aas) (F9Z893) 

 


3.A.1.133 The Peptide-6 Exporter (Pep6E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.133.1The modified YydF* peptide exporter, YydIJ (Butcher et al., 2007)BacteriaYydIJ of Bacillus subtilis:
YydI (C) (Q45593)
YydJ (M) (Q45592)
 
3.A.1.133.2A 6TMS homologue of YydJ (ORF1) of 280aasBacteriaORF1 of Flavobacteria bacterium BBFL7 (Q26C21)
 


3.A.1.134 The Peptide-7 Exporter (Pep7E) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.134.1The lantibiotic, salivericin A exporter, SalXYGram-positive bacteriaSalXY of Streptococcus salivarius
SalX (C)
SalY (M)
 
3.A.1.134.10

Peptide exporter, YsaB (667 aas and 10 TMSs)/YsaC (257 aas).  Probably exports lantibiotic antibiotics (Draper et al. 2015).

Firmicutes

YsaBC of Lactococcus lactis
YsaB (M)
YsaC (C)

 
3.A.1.134.11

Lantibiotic detoxification ABC transporter, VraD (252 aas)/VraE (626 aas; 10 TMSs)/VraH ( (Draper et al. 2015).  Upregulated in response to exposure to beta-defensin 3 (Sass et al. 2008).  Exports antimicrobial peptides such as nisin, bacitracin, daptomycin and gallidermin. Expression of vraH in the absence of vraDE is sufficient to mediate low-level resistance, but VraDEH is required to  confer high-level resistance against daptomycin and gallidermin. (Popella et al. 2016).

Firmicutes

VraDE of Staphylococcus aureus

VraD (Q9RL74)
VraE (Q9KWJ6)
VraH (T1YED1)

 
3.A.1.134.12

ABC multidrug resistance efflux pump, AnrAB.  Exports nisin, gallidermin, bacitracin and β-lactam antibiotics  (Collins et al. 2010).

Firmicutes

AnrAB of Listeria monocytogenes
AnrA (C)
AnrB (M; 642 aas and 10 TMSs)

 
3.A.1.134.13

Putative ABC exporter with three constituent proteins, two membrane proteins with a probable 10 TMS topology in a 1 (N-terminal) + 6 (middle) + 3 (C-terminal) TMS arrangement, and one ATPase

ABC transporteer of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 
3.A.1.134.14

Putative ABC3-type porter with a membrane protein and an ATPase encoded by adjacent genes, but next to the genes that encoded by the systems in TCDB under TC#s 3.A.1.207.7 and 8.

ABC3-type porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 
3.A.1.134.2The bacitracin-resistance (putative bacitracin exporter), MbrAB. Participate with BreSR to control its own gene expression (Bernard et al., 2007).Gram-positive bacteriaMbrAB of Streptococcus mutans
MbrA (C)
MbrB (M)
 
3.A.1.134.3

The putative bacitracin exporter, BceAB (BarAB; YtsCD) (Bernard et al., 2003; Ohki et al., 2003).  Functions in both signaling to the two component system, BceRS, and in export of the antimicrobial peptide (Dintner et al. 2014).  BceB interacts directly with BceS, and BceB binds bacitracin (Dintner et al. 2014).  Specific regions and residues are involved in signalling or transport (Kallenberg et al. 2013).  More recent studies suggest that BceAB may cause bacitracin resistance by transferring undecaprenyl pyrophosphate from the exteral to the internal leaflet of the inner membrane where it can't bind bacitracin and other lantibiotics that use Lipid II as a receptor (Draper et al. 2015). It may cause drug detoxification by targeet protection (Rismondo and Schulz 2021).

Firmicutes

BceAB (YtsCD) of Bacillus subtilis
BceA (C) CAB15016
BceB (M) CAB15015

 
3.A.1.134.4

The bacitracin/vancoresmycin (a tetramic acid antibiotic) resistance exporter (Becker et al. 2009) (most like 3.A.1.134.2)

Firmicutes

SPR0812/SPR0813 of Streptococcus pnenmoiae
SPR0812 (C) (Q8DQ77)
SPR0813 (M) (Q8DQ76)

 
3.A.1.134.5

The MDR exporter, YvcRS. Possibly linked to regulation by a sensor kinase/response regulator system (YvcQP) (Joseph et al., 2002; Bernard et al., 2007).

Bacteria

YvcRS of Bacillus subtilis
YvcR (C) (O06980)
YvcR (M) (O06981)

 
3.A.1.134.6

The cationic peptide/MDR exporter, YxdLM. Possibly linked to a sensor kinase/reponse regulator system (YxdJK) (Joseph et al., 2002; Bernard et al., 2007).

Bacteria

YxdLM of Bacillus subtilis
YxdL (C) (P42423)
YxdM (M) (P42424)

 
3.A.1.134.7

The VraFG ABC transporter interacts with GraXSR [GraX, Q7A2W7; GraS, A6QEW9; GraR, A6QEW8] to form a five- or six-component system required for cationic antimicrobial peptide sensing and resistance (Falord et al., 2012).  VraX has been termed a two component system connector and may not be a component of the transporter. VraFG may be a sensor rather than the transporter for the substrate peptide. The actual transporter regulated by VraFGX may have TC# 4.H.1.1.1 (MprF) (Falord et al. 2012). The extracellular loop of the membrane permease VraG interacts with GraS to sense cationic antimicrobial peptides in Staphylococcus aureus (Cho et al. 2021).

Bacteria

VraFG/GraXSR of Staphylococcus aureus 
VraF (A6QEX0)
VraG (A6QEX1)
VraX (Q7A2W7)

 
3.A.1.134.8

Antimicrobial peptide exporter, ABC12 or YvoST (Revilla-Guarinos et al. 2013).

Firmicutes

YvoST of Lactobacillus casei

 
3.A.1.134.9

Two component toxic peptide exporter with a membrane subunit of 663 aas and 10 TMSs and an ATPase of 256 aas, ABC09 (Revilla-Guarinos et al. 2013).

Firmicutes

ABC09 of Lactobacillus casei

 


3.A.1.135 The Drug Exporter-4 (DrugE4) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.135.1

The heterodimeric multidrug exporter, YdaG/YbdA  (Both proteins are ABC half transporters; only the heterodimer is active; ethidium, daunomycin and BCECF-AM are substrates; Lubelski et al., 2004) These proteins have been renamed LmrC and LmrD (Lubelski et al., 2006)

Gram-positive bacteriaYdaG/YdbA of Lactococcus lactis YdaG (M-C) (AAK04408)
YdbA (M-C) (AAK04409)
 
3.A.1.135.10

Uncharacterized two component ABC transporter, both of M-C domain order with 6 N-terminal TMSs. The genes encoding these two proteins are adjacent to  a putative ABC transporter of TC# 9.B.29.2.13.

UPs of Caldicellulosiruptor saccharolyticus

 
3.A.1.135.11

ABC transporter with two components, each with 6 N-terminal TMSs + a C-terminal ATPase

ABC exporter of Methanobrevibacter sp.

 
3.A.1.135.12

Two component ABC exporter, both subunits with 611 and 600 aas with 6 N-terminal TMSs in a 2 + 2 + 2 arrangement.  The C-terminal domain is the ATPase

ABC exporter of Lokiarchaota

 
3.A.1.135.13

Two component netropsin resistance ABC netropsin exporter, NetP1/NetP2 (Stumpp et al. 2005).

Netropsin resistance efflux porter of Streptomyces netropsis (Streptoverticillium netropsis)
NetP1 of 618 aas and 6 TMSs (M-C) (Q66LJ1)
NetP2 of 635 aas and 6 TMSs (M-C) (Q66LJ0)

 
3.A.1.135.2The heterodimeric putative multidrug exporter, RscA/RscB; probably orthologous to YdaG/YbdA (TC #3.A.1.117.4) [Transcription is activated by stress conditions (heat, acid) and repressed by a 2-component system, CovRS (Dalton et al., 2006)]Gram-positive bacteriaRscAB of Streptococcus pyogenes RscA (M-C) (568 aas) (Q9A1K5)
RscB (M-C) (594 aas) (Q9A1K4)
 
3.A.1.135.3

Narrow spectrum fluoroquinolone (ciprofloxacin and norfloxacin) efflux pump, SatAB (Escudero et al. 2011).

Firmicutes

SatAB of Streptococcus suis
SatA, 568 aas (M-C) (G9CHY8)
SatB, 594 aas, (M-C) (G9CHY9)

 
3.A.1.135.4

Multidrug resistance ABC exporter, PatAB (PatA, 564 aas; PatB, 588 aas) (Bidossi et al. 2012). Drug-dependent inhibition of nucleotide hydrolysis by PatAB has been demonstrated (Guffick et al. 2022). Ethidium-like inhibition was observed with propidium, novobiocin and coumermycin A1, which all inhibit nucleotide hydrolysis by a non-competitive mechanism. This fact casts light on potential mechanisms by which drugs can regulate nucleotide hydrolysis by PatAB, which might involve a novel drug binding site near the nucleotide-binding domains (Guffick et al. 2022).

Firmicutes

PatAB of Streptococcus pneumoniae
PatA (M-C)
PatB (M-C)

 
3.A.1.135.5

The hetrodimeric ABC transporter, TM287/TM288.  The 2.9-Å crystal structure has been solved in the inward-facing state. The two nucleotide binding domains (NBDs) remain in contact through an interface involving conserved motifs that connect the two ATP hydrolysis sites.  AMP-PNP binds to a degenerate catalytic site which deviates from the consensus sequence in the same positions as the eukaryotic homologs, CFTR (TC# 3.A.1.202.1) and TAP1-TAP2 (TC# 3.A.1.209.1) (Hohl et al. 2012).  The structural basis for allosteric crosstalk (positive cooperativity) between the two ATP binding sites has been studied (Hohl et al. 2014).  The two NBDs exhibit unexpected differences and flexibility (Bukowska et al. 2015). It exports daunomycin and the nonfluorescent 2,7-bis(carboxyethyl)-5(6)-carboxyfluorescein-acetoxymethylester (BCECF-AM) (Hohl et al. 2012). Timachi et al. 2017 observed hydrolysis-independent closure of the NBD dimer, further stabilized as the consensus site nucleotide is committed to hydrolysis.

Thermatogae

TM287/TM288 of Thermatoga maritima

 
3.A.1.135.6

Two component multidrug efflux pump with the 6 TMS membrane domain preceding the ATPase domain in both proteins.  Confers resistance to erythromycin and tetracycline and catalyzes export of Hoechst 33342 (Moodley et al. 2014).  Expression is induced by the presence of erythromycin.

Actinobacteria

MDR pump of Bifidobacterium longum

 
3.A.1.135.7

Multidrug exporter, EfrAB.  Confers resistance to many structurally unrelated antimicrobial agents, such as norfloxacin, ciprofloxacin, doxycycline, acriflavine, 4,6-diamidino-2-phenylindole, tetraphenylphosphonium chloride, daunorubicin, and doxorubicin (Lee et al. 2003).  Induced by half minimal inhibitory concentrations (MIC) of gentamicin, streptomycin and chloramphenicol which are also exporter (Lavilla Lerma et al. 2014).  In some strains, this system may not be the primary drug exporter (Hürlimann et al. 2016).

EfrAB of Enterococcus faecalis
EfrA (MC), 567 aas and 6 TMSs
EfrB (MC), 589 aas and 6 TMSs

 
3.A.1.135.8

Multidrug efflux pump, EfrCD.  Exports daunorubicin, doxorubicin, ethidium and Hoechst 33342.  Mediates efflux of fluorescent substrates and confers resistance towards multiple dyes and drugs including fluoroquinolones (Hürlimann et al. 2016).

EfrCD of Enterococcus faecalis
EfrC, MC, 571 aas and 6 TMSs
EfrD, MC, 589 aas and 6 TMSs

 
3.A.1.135.9

Multidrug exporter, EfrEF.  Mediates efflux of fluorescent substrates and confers resistance towards multiple dyes and drugs including fluoroquinolones (Hürlimann et al. 2016).

EfrEF of Enterococcus faecalis
EfrE, MC, 575 aas and 6 TMSs
EfrF, MC, 592 aas and 6 TMSs

 


3.A.1.136 The Uncharacterized ABC-3-type (U-ABC3-1) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.136.1

Putative ABC3 permease complex U-ABC3-1a (403 aas; 4 TMSs in a 1 + 3 TMS arrangement) + an ATPase of 230 aas. The two genes encoding these two proteins are separated by a single gene encoding a putative FMN binding domain-containing protein of 149 aas.

Bacteria

U-ABC3-1a of Treponema denticola (M) (Q73MJ0)

 
3.A.1.136.2

ABC-type antimicrobial peptide transporter with a membrane protein of 421 aas and 4 TMSs (Q6MNW8) and an ATPase of encoded by a gene adjacent to the membrane protein (Q6MNW9). Adjacent to these two genes is one annotated as an iron-regluated protein, FrpA (Q6MNW7).

ABC transporter of Bdellovibrio bacteriovorus

 


3.A.1.137 The Uncharacterized ABC-3-type (U-ABC3-2) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.137.1Putative ABC-3-type permease complex, ABC3-2aArchaeaABC3-2a of Pyrobaculum calidifontis:
ABC3-2a (M) (A3MWP2)
ABC3-2a (C) (A3MWP1)
 
3.A.1.137.2

ABC-type antimicrobial peptide transporter of 786 aas and 8 TMSs

ABC transporter of Bdellovibrio bacteriovorus

 


3.A.1.138 The Unknown ABC-2-type (ABC2-1) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.138.1Unknown ABC-2 transporter complex-1, U-ABC2-TC-1ArchaeaU-ABC2-TC-1 of Picrophilus torridus:
U-ABC2-TC-1a (M) (Q6KYW9)
U-ABC2-TC-1a (C) (Q6KYW8)
 


3.A.1.139 The UDP-Glucose/Iron Exporter (U-GlcE) Family (UPF0014 Family)


Examples:

TC#NameOrganismal TypeExample
3.A.1.139.1

UDP-glucose exporter, STAR1/STAR2 (sensitive to aluminum rhizotoxicity) (Probable Type I topology) (Huang et al. 2009).

Plants

STAR1/STAR2 of Oryza sativa
STAR1 (C) (Q5Z8H2)
STAR2 (M) (Q5W7C1)

 
3.A.1.139.2

The FetA (YbbL)/FetB (YbbM) iron exporter (SwissProt family UDF0014; 6 or 7 putative TMSs).  Expression enhances resistance to oxidative stress (Nicolaou et al. 2013).

Bacteria

FetA/B of E. coli
FetA (C) (P77279)
FetB (M) (P77307)

 
3.A.1.139.3

The uncharacterized ABC exporter, U-ABC-M/C

Bacteria

U-ABCC/U-ABC-M of Spirochaeta africana
U-ABC-C (C) (H9UM45)
U-ABC-M (M) (H9UM46)

 
3.A.1.139.4

Plasma membrane ABC exporter, sensitive to aluminum rhizotoxicity 1/2, STAR1/STAR2 (Larsen et al., 2005). Induced in response to aluminum exposure. The system is also called the ALS3/STAR1 ABC transporter that may transport an Al3+/Malate complex (). External and internal aluminum resistance by ALS3-dependent STAR1-mediated promotion of STOP1 degradation has been demonstrated (Fan et al. 2024).

Plants

STAR1/2 of Arabidopsis thaliana 
STAR1 (C) (Q9C9W0)
STAR2 (M) (Q9ZUT3) 

 
3.A.1.139.5

Uncharacterized protein of 318 aas and 7 TMSs in a 5 + 2 TMS arrangement.  An ATPase for this putative porter has not been identified.

UP of Tetrapyrgos nigripes

 


3.A.1.14 The Iron Chelate Uptake Transporter (FeCT) Family (Similar to 3.A.1.13 and 3.A.1.15)


Examples:

TC#NameOrganismal TypeExample
3.A.1.14.1

Iron (Fe3+) or ferric-dicitrate porter, FecBCDE (Braun and Herrmann, 2007). The Mycobacterium tuberculosis (Mtb) ortholog has two substrate binding proteins, FecB and FecB2 (de Miranda et al. 2023).  The crystallographic structures of Mtb FecB and FecB2 were determined to 2.0 Å and 2.2 Å resolution, respectively, and they show distinct ligand binding pockets. In vitro ligand binding experiments for FecB and FecB2 were performed with heme and bacterial siderophores from Mtb and other species, revealing that both FecB and FecB2 bind heme, while only FecB binds the Mtb sideophore ferric-carboxymycobactin (Fe-cMB). Subsequent structure-guided mutagenesis of FecB identified a single glutamate residue-Glu339-that significantly contributes to Fe-cMB binding (de Miranda et al. 2023).

Proteobacteria

FecBCDE of E. coli
FecB (R)
FecC (M)
FecD (M)
FecE (C)

 
3.A.1.14.10

The heme porter, Shp/SiaABC (HtsABC). Shp is a cell surface heme binding protein that transfers the heme directly to HstA (Nygaard et al., 2006). The crystal structure of the heme binding domain of Shp has been solved (Aranda et al., 2007). HtsABC is required for the uptake of staphyloferrin A (Beasley et al. 2009). The Shp cell surface heme receptor feeds iron-heme to the transporter in preparation for uptake (Sun et al. 2010; Ouattara et al., 2010). 

Bacteria

Shp/HtsABC of Streptococcus pyogenes
Shp (R1) (291 aas; Q1J548)
HtsA (R2) (294 aas; Q99YA2)
HtsB (M) (340 aas; Q99YA3)
HtsC (C) (278 aas; Q99YA4)

 
3.A.1.14.11

The molybdate/tungstate ABC transporter, MolABC.  For MolC; HI1470(C)/MolB; HI1471(M), the 3D structure is known at 2.4 Å resolution; Pinkett et al., 2007).  MolA binds to MolBC with low affinity (50 - 100μM), forming a transient complex that is stabilitzed by ligand binding (Vigonsky et al. 2013).

Proteobacteria

MolABC of Haemophilus influenzae
MolC; HI1470 (C) (Q57399)
MolB; HI1471 (M; 10 TMSs; type II fold) (Q57130)
MolA; HI1472 (R) (E3GUW2)

 
3.A.1.14.12

Desferrioxamine B uptake porter, DesABC (Barona-Gomez et al., 2006)

Bacteria

DesABC of Streptomyces coelicolor
DesA (R) (1 TMS) (Sco7499; Q9L178)
DesB (M-M) (18 TMSs; 9 9 TMSs) (Sco7498; Q9L179)
DesC (C) (0 TMSs) (Sco7400; Q9L177)

 
3.A.1.14.13

Ferric iron-coelichelin uptake porter, CchCDEF (Barona-Gomez et al., 2006).

Actinobacteria

CchCDEF of Streptomyces coelicolor
CchC (M) (Sco0497) (Q9RK09)
CchD (M) (Sco0496) (Q9RK10)
CchE (C) (Sco0495) (Q9RK11)
CchF (R) (Sco0494) (Q9RK12)

 
3.A.1.14.14

The Fe3+ /Fe3+ferrichrome/Fe3+heme uptake porter; SiuABDG (FtsABCD) (Montañez et al., 2005; Hanks et al. 2005; Li et al. 2013).  A similar system has been characterized in S. iniae (Wang et al. 2013).

Bacteria

SiuABDG (FtsABCD) of Streptococcus pyogenes
SiuA; FtsA (C) (Q9A197)
SiuD; FtsB (R) (Q9A199)
SiuB; FtsC (M) (Q9A198)
SiuG; FtsD (M) (Q06A41) 

 
3.A.1.14.15

Uptake transporter for the catecholic trilactone (2, 3-dihydroxybenzoate-glycine-threonine)3 siderophore bacillibactin (for ferric iron scavenging), FeuABC (Gaballa and Helmann, 2007; Miethke et al., 2006).

Bacteria

FeuABC of Bacillus subtilis
FeuA (R) (P40409)
FeuB (M) (P40410)
FeuC (M) (P40411)

 
3.A.1.14.16

The heme-specific uptake porter, HemTUV (Létoffé et al., 2008).

Bacteria

HemTUV of Serratia proteamaculans
HemT (R) - (A8GDS8)
HemU (M) - (A8GDS7)
HemV (C) - (A8GDS6)

 
3.A.1.14.17Heme acquisition ABC uptake transporter, IsdDEF (Liu et al., 2008)FirmicutesIsdDEF of Staphylococcus aureus
IsdD (?) (358aas, 2TMSs) (Q5HGV2)
IsdE (R) (295aas, 1TMS) (Q7A652)
IsdF (M) (273aas; 8TMSs) (Q7A651)
 
3.A.1.14.18

The heme uptake porter, ShuTUV (Burkhard and Wilks, 2008). Transports a single heme per reaction cycle (Mattle et al., 2010). (3-d structure of ShuT is known (2RG7).

Bacteria

ShuTUV of Shigella dysenteriae
ShuT(R) (Q32AX9)
ShuU(M) (Q32AY2)
ShuV(C) (Q32AY3)

 
3.A.1.14.19Heme uptake porter, HugBCD (Villarreal et al., 2008); also called HmuTUV.

Bacteria

HugBCD of Plesiomonas shigelloides
HugB (R) (Q93SS3)
HugC (M) (Q93SS2)
HugD (C) (Q93SS1)

 
3.A.1.14.2

Iron (Fe3+)-enterobactin porter

Proteobacteria

FepBCDG of E. coli
FepB (R) (C8U2V6)
FepC (C) (P23878)
FepD (M) (P23876)
FepG (M) (P23877)

 
3.A.1.14.20

Heme-iron (hemin) utilization transporter BhuTUV ( Brickman et al., 2006; Vanderpool and Armstrong, 2004).  The crystal structures of BhuUV with or without the periplasmic haem-binding protein BhuT have been solved (Naoe et al. 2016). The TMSs show an inward-facing conformation, in which the cytoplasmic gate of the haem translocation pathway is completely open. Since this conformation is found in both the haem- and nucleotide-free form, the structure of BhuUV-T provides the post-translocation state and the missing piece in the transport cycle of type II importers.

Gram-negative bacteria

BhuTUV of Bordetella pertussis
BhuT (R) (Q7VSQ6)
BhuU (M) (Q7W024)
BhuV (C) (Q7W025)

 
3.A.1.14.21

The heme uptake porter, PhuTUV (transports one heme per reaction cycle) (Mattle et al., 2010).

Proteobacteria

PhuTUV of Pseudomonas aeruginosa
PhuT (R) (Q9HV90)
PhuU (M) (O68878)
PhuV (C) (O68877)

 
3.A.1.14.22

The putative ferric iron-desferrioxamine E uptake porter, DesEFGH.  The DesE binding receptor has been characterized (Barona-Gómez et al. 2006).  The remaining three (desFGH) genes cluster together without a gene encoding a receptor (R).  They are believed to function with DesE based on sequence similarity and phylogenetic analyses (Getsin et al., 2013).

Actinobacteria

DesEFGH of Streptomyces coelicolor
DesE (Sco2780) (R) (349 aas; 1 TMS) (Q9L074)
DesF (Sco1785) (C) (301 aas; 0 TMSs) (Q9S215)
DesG (Sco1786) (M) (375 aas; 9 TMSs) (Q9S214)
DesH (Sco1787) (M) (345 aas; 9 TMSs) (Q9S213)

 
3.A.1.14.23

Two components of a vitamin B12 (cobalamin) uptake porter, BtuCD.  BtuAB must exist but have not been identified (Deutschbauer et al. 2011). 

BtuCD of Shewanella oneidensistu
BtuC (M) of 380 aas
BtuD (C) of 314 aas

 
3.A.1.14.24

FecB1CDE iron siderophore uptake transporter. Transports iron chelated dihydroxamate xenosiderophores, either ferric schizokinen (FeSK) or a ferric siderophore of the filamentous cyanobacterium Anabaena variabilis ATCC 29413 (a schizokinen derivative, SAV), as the sole source of iron in a TonB-dependent manner (Obando S et al. 2018). The gene schT encodes the TonB-dependent outer membrane transporter (TC# 1.B.14.9.6).

FecB1CDE of Synechocystis sp. PCC 6803
FecB1 (R), 315 aas, P72593
FecC (M), 343 aas, 9 TMSs
FecD (M), 349 aas, 9 TMSs
FecE (C), 268 aa

 
3.A.1.14.25

Heme uptake porter with three subunits (Mandal et al. 2019).

Heme porter of Thermus thermophilus

 
3.A.1.14.26

Cyanocobalamin uptake porter with 3 components, R, M and C (Mandal et al. 2019).

Cyanocobalamin porter of Thermus thermophilus

 
3.A.1.14.27

Heme transporter with three components, HmuU (M), HmuV (C) and HmuT (R).  Chemo-mechanical coupling in the transport cycle has been proposed with outward open, inward open and occluded states (Tamura et al. 2019).

HmuUVT of Burkholderia cenocepacia (Burkholderia cepaci)
HmuU, B4EKB4 (M)
HmuV, B4EKB5 (C)
HmuT, B4EKB3 (R)

 
3.A.1.14.28

Iron-siderophore (staphylobactin, made by S. aureus) uptake system, SirABCD (A = periplasmic receptor (R); B and C = membrane proteins with 9 or 10 TMSs (M), and D (IsdC) is likely to be the periplasmic auxiliary protein with 2 TMSs, N- and C-terminal (Dale et al. 2004). This last protein is also given the TC# 9.A.39.1.2 as a member of its own family. The ATPase subunit is not known, but it could be the protein with genbank acc# WP_001080807.1.

SirABCD of Staphylococcus aureus

 
3.A.1.14.3

Iron (Fe3+)-hydroxamate (ferrichrome, coprogen, aerobactin, ferrioxamine B, schizakinen, rhodotorulic acid) porter, albomycin porter.  A FtsZ inhibitor can utilize siderophore-ferric iron uptake transporter systems, FepA, CirA (outer membrane transporters) and FhuBC (inner membrane transporter) for activity against Gram-negative bacterial pathogens (Bryan et al. 2024).

Proteobacteria

FhuBCD of E. coli
FhuB (M-M; 20 TMSs; 10+10)
FhuC (C)
FhuD (R)

 
3.A.1.14.4Iron-chrysobactine porterProteobacteriaCbrABCD of Erwinia chrysanthemi
CbrA (R)
CbrB (M)
CbrC (M)
CbrD (C)
 
3.A.1.14.5

Heme (hemin) uptake porter. The receptor, HmuT, binds two parallel stacked heme molecules, and two are transported per reaction cycle (Mattle et al., 2010).

Proteobacteria

HmuTUV of Yersinia pestis
HmuT (R) (Q56991)
HmuU (M) (Q56992)
HmuV (C) (Q56993)

 
3.A.1.14.6The iron-vibriobactin/enterobactin uptake porterProteobacteriaViuPDGC of Vibrio cholerae
ViuP (R)
ViuD (M)
ViuG (M)
ViuC (C)
 
3.A.1.14.7

Iron (Fe3+)-hydroxamate porter (transports Fe3+-ferrichrome and Fe3+-ferrioxamine B with FhuD1, and these compounds plus aerobactin and coprogen with FhuD2).  FhuB may function with FhuG (A6QEV8) together with FhuD2 to form a ferrichrome transporter where FhuB and FhuG have conserved arginine residues (R71 and R61, respectively) that form essential salt bridges with FhuD2 (Vinés et al. 2013).

Firmicutes

FhuBCD1D2 of Staphylococcus aureus
FhuB (M)
FhuC (C)
FhuD1 (R)
FhuD2 (R)

 
3.A.1.14.8The iron-vibrioferrin uptake porter (Tanabe et al., 2003) ProteobacteriaPvuBCDE of Vibrio parahaemolyticus
PvuB (R) (BAC16540)
PvuC (M) (BAC16541)
PvuD (M) (BAC16542)
PvuE (C) (BAC16543)
 
3.A.1.14.9The Corrinoid porter, BtuCDE (Woodson et al., 2005)ArchaeaBtuCDE of Halobacterium sp. strain NRC-1
BtuC (M) (AAG19698)
BtuD (C) (NP_444218)
BtuE (R) (AAG19697)
 


3.A.1.140 The FtsX/FtsE Septation (FtsX/FtsE) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.140.1

The FtsX/FtsE ABC transporter (Arends et al., 2009) (FtsX is of the type III topology). FtsEX directly recruits EnvC to the septum via an interaction between EnvC and a periplasmic loop of FtsX. FtsEX variants predicted to be ATPase defective still recruit EnvC to the septum but fail to promote cell separation. Amidase activation via EnvC in the periplasm is regulated by conformational changes in the FtsEX complex mediated by ATP hydrolysis in the cytoplasm. Since FtsE has been reported to interact with FtsZ, amidase activity may be coupled with the contraction of the FtsZ cytoskeletal ring (Yang et al., 2011).

Bacteria

FtsX/FtsE of E. coli
FtsX (M) (P0AC31)
FtsE (C) (P0A9R7)

 
3.A.1.140.2

The cell division ABC system, FtsX/FtsE

FstE/X of Caldanaerobacter subterraneus subsp. tengcongensis (Thermoanaerobacter tengcongensis)

 
3.A.1.140.3

Cell division ABC system, FtsXE.

FtsXE of Nostoc punctiforme
FtsX (M), 300 aas, 4 TMSs
FtsE (C), 248 aas

 
3.A.1.140.4

Cell division ABC system, FtsXE of 300 aas and 4 TMSs, and 229 aas and 0 TMSs, respectively.

FtsXE of Actinokineospora spheciospongiae
FtsX, (M), 300 aas and 4 TMSs
FtsE, (C), 229 aas and 0 TMSs

 
3.A.1.140.5

Cell division ABC system, FtsXE.

FtsXE of Candidatus Nitrosopumilus salaria
FtsX, (M), 301 aas, 4 TMSs
FtsE, (C), 222 aas, 0 TMSs

 
3.A.1.140.6

Cell division system, FtsXE.  The FtsEX:PcsB complex forms a molecular machine that carries out peptidoglycan (PG) hydrolysis during normal cell division. FtsEX transduces signals from the cell division apparatus to stimulate PG hydrolysis by PcsB, an amidase, which interacts with extracellular domains of FtsX (Bajaj et al. 2016).

FtsXE of Streptococcus pneumoniae
FtsX, (M), 308 aas and 4 TMSs
FtsE, (C), 226 aas and 0 TMSs

 
3.A.1.140.7

FtsEX, ABC transporter involved in cell division FtsE (229 aas, 0 TMSs) is the ATPase subunit while FtsX (298 aas) is the transmembrane protein with 4 TMSs in a 1 + 3 TMS arrangement.  It interacts with RipC, a periplasmic hydrolase of 381 aas and 1 N-terminal TMS (Samuels et al. 2024).

FtsEX of Mycobacterium smegmatis

 


3.A.1.141 The Ethyl Viologen Exporter (EVE) Family (DUF990 Family)


Examples:

TC#NameOrganismal TypeExample
3.A.1.141.1

The ethyl (methyl; benzyl) viologen export pump, EvrABC (EvrB and EvrC of 6 TMSs are members of the large DUF990 superfamily (Prosecka et al., 2009); They appear to be of the ABC-2 topological type).

Bacteria

EvrABC of Synechocystis sp. PCC6803
P73329 slr1910, ABC protein (EvrA)
P74256 slr1174, membrane protein (EvrB)
P74757 slr0610, membrane protein (EvrC)

 
3.A.1.141.2

ABC transporter of unknown specificity, AbcABC

Bacteria

AbcABC of Thermoanaerobacter tengcongensis
AbcA (M) (Q8R6Q6)
AbcB (M) (Q8R6Q5)
AbcC (C) (Q8R6Q4)

 


3.A.1.142 The Glycolipid Flippase (G.L.Flippase) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.142.1

Glycolipid translocase (flippase) Spr1816/Spr1817 (R.Hakenbeck, personal communication)

Firmicutes

Glycolipid flippase, Spr1816/Spr1817, of Streptococcus pneumoniae
Spr1816 (M) (Q8DNC0)
Spr1817 (C) (Q8DNB9)

 
3.A.1.142.2

ABC exporter, YvfS/YvfR of 284 and 287 aas, respectively

YvfSR of Bdellovibrio bacteriovorus
YvfS (M)
YvfR (C)

 
3.A.1.142.3

Uncharacterized ABC2 exporter consisting of a 6 TMS membrane protein of 254 aas and an ATPase, encoded by the gene adjacent to the 6 TMS membrane protein.

ABC exporter of Mobiluncus curtisii (Falcivibrio vaginalis)

 


3.A.1.143 The Exoprotein Secretion System (EcsAB(C))


Examples:

TC#NameOrganismal TypeExample
3.A.1.143.1

The exoprotein (including α-amylase) secretion system, EcsAB(C) (Leskelä et al., 1999). Also may play roles in sporulation, competence (Leskelä et al., 1996) and transformation using purified DNA (Takeno et al., 2011). An involvement of EcsC in transport is not established, but it is similar in sequence to the C-terminus of the P-type ATPase, 3.A.3.31.2.

Bacteria

EcsAB(C) of Bacillus subtilis 
EcsA (C) (P55339)
EcsB (M) (P55340)
EcsC (M) (P55341) 

 
3.A.1.143.2

YthQ (386aas; 8-9 TMSs)/YthP (ATPase; 0 TMSs)

Bacteria

YthPQ (EscAB) of Bacillus amyloliquefaciens
EscA (YthP) (G0IP52)
EscB (YthQ) (G0IP51)

 
3.A.1.143.3

ABC exporter with two components, EcsA, a membrane protein of 430 aas and 10 TMSs in a 2 + 2 + 2 + 2 + 2 TMS arrangement (EOP55101) and EcsB, an ABC-type ATPase of 241 aas (EOP55100).

ABC exporter of Bacillus cereus

 
3.A.1.143.4

ABC-type exoprotein exporter with three componenets, a membrane constituent of 434 aas and 10 TMSs in a 2 + 2 + 2 + 2 + 2 arrangement (AGA59062), an ATPase of 278 aas (AGA59063) and a membrane protease of 289 aas and 2 or 3 N-terminal TMSs (TC# 8.A.21.2.6; AGA59064). The presence of the latter protein encoded adjacent to the transport system suggests that the substrate of the ABC exporter may be a protein that is processed by this protease.

ABC exporter of Thermobacillus composti KWC4

 
3.A.1.143.5

ABC exporter with two components, a membrane protein of 399 aas and 10 TMSs (PFJ32189) and an ATPase of 236 aas (PFJ32188). The gene adjacent to the two genes encoding this system is annotated as a FtsX cell division protein of 195 aas.

ABC exporter of Bacillus anthracis

 


3.A.1.144 Functionally Uncharacterized ABC2-1 (ABC2-1) Family

 


Examples:

TC#NameOrganismal TypeExample
3.A.1.144.1

Functionally uncharacterized ABC2 transporter #1.  This system is encoded by two genes that overlap and are therefore probably translationally coupled; they are in the same operon with the genes for 2.A.1.144.2.

Archaea

ABC2 transporter #1 of Methanocella arvoryzae 
ABC2-1 (M) (Q0W8T3)
ABC2-1 (C) (Q0W8T4) 

 
3.A.1.144.2

Functionally uncharacterized ABC2 transporter #2.  This system is encoded by two genes that overlap and are therefore probably translationally coupled; they are in the same operon with the genes for 2.A.1.144.1.

Archaea

ABC2 transporter #2 of Methanocella arvoryzae
ABC2-2 (M) (Q0W8T6)
ABC2-2 (C) (Q0W8T7) 

 
3.A.1.144.3

Functionally uncharacterized ABC2 transporter #3.

%u03B4-Proteobacteria

ABC2 transporter of Myxococcus xanthus
ABC2-3 (M) (Q1D0V0)
ABC2-3 (C) (Q1D0V1) 

 
3.A.1.144.4

Functionally uncharacterized ABC2 transporter #4 of 751 aas with 18 putative TMSs.  The first 6 TMSs are duplicated to give the N-terminal 12 TMSs.  The last 6 TMSs are non-homologous and are of the DUF95 family (TC #9.B.98) which may consist of ABC exporter auxiliary subunits/domains.

Chloroflexi

ABC2 transporter of Oscillochloris trichoides 
ABC2 (M) (E1IBA3)
ABC2 (C) (E1IBA4) 

 


3.A.1.145 Peptidase Fused Functionally Uncharacterized ABC2-2 (ABC2-2) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.145.1

ABC2 transporter domain fused to an aminopeptidase N domain (Peptidase M1 family) of 1200 aas with 13 putative N-terminal TMSs.

δ-proteobacteria

ABC2 protein of Myxococcus xanthus

 
3.A.1.145.2

Putative ABC2 permease of 529 aas and 12 TMSs, Glr0437.

Cyanobacteria

Glr0437 of Gloeobacter violaceus

 
3.A.1.145.3

ABC2 fusion protein of 1194 aas and 13 putative TMSs.  Annotated as ABC transporter involved in multi-copper enzyme maturation; permease component.

Bacteroidetes

ABC2 protein of Cecembia lonarensis

 
3.A.1.145.4

Putative ABC2 protein of 537 aas and 14 putative TMSs

Archaea

ABC2 permease of Methanocella paludicola

 
3.A.1.145.5

Uncharacterized ABC membrane transport protein of 222 aas and 6 TMSs.

UP of Candidatus Wolfebacteria bacterium

 


3.A.1.146 The actinorhodin (ACT) and undecylprodigiosin (RED) exporter (ARE) family


Examples:

TC#NameOrganismal TypeExample
3.A.1.146.1

The probable actinorhodin (ACT) and undecylprodigiosin (RED) exporter (Lee et al. 2012), AreABCD (Sco3956-9).

Actinobacteria

 

AreABCD (Sco3956-9) of Streptomyces coelicolor
AreA (C) (Sco3956)
AreB (M) (Sco3957)
AreC (C) (Sco3958)
AreD (M) (Sco3959)

 
3.A.1.146.2

Putative ABC exporter, Isop2111-Isop2114

Planctomycetes

Isop2111-Isop2114 of Isophaera pallida
Isop2111 (C) (332 aas) (E8R490)
Isop2112 (M) (359 aas; 6 TMSs) (E8R491)
Isop2113 (C) (340 aas) (E8R492)
Isop2114 (M) (298 aas; 7 TMSs) (E8R493) 

 
3.A.1.146.3

Putative four component ABC exporter with two membrane proteins and two ABC ATPases.

Putative ABC porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 


3.A.1.147 Functionally Uncharacterized ABC2-3 (ABC2-3) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.147.1

Putative two component ABC exporter with a membrane protein of 573 aas and 12 TMSs and an ATPase encoded adjacent to the membrane protein and also adjacent to a gene encoding an adenine glycosylase, probably within a single operon.

Gemmatimonadetes

ABC exporter of Gemmatimonas aurantiaca
Membrane protein (M) (C1A6K7)
ATPase (C) (C1A6K8)

 
3.A.1.147.10

Uncharacterized protein of 627 aas and 12 TMSs

Firmicutes

UP of Desulfosporosinus meridiei

 
3.A.1.147.2

Putative 2-component sporulation-related ABC exporter.  The genes encoding this system are adjacent to a spore germination receptor (TC# 2.A.3.9.5) and a putative signalling molecule transporter (2.A.86.1.11).

Firmicutes

Putative 3-component ABC exporter of Paenibacillus mucilaginosus
Protein of 572 aas and 12 putative TMSs (M) (F8FLY8)
ATPase protein of 243 aas (C) (F8FLY7)

 
3.A.1.147.3

Putative two component ABC exporter with the membrane protein having 623 aas and 12 TMSs.

Planctomycetes

ABC exporter of Isosphaera pallida
Membrane protein (M) (E8R692)
ATPase (C) (E8R694)

 
3.A.1.147.4

Putative two component ABC exporter with a membrane protein of 537 aas and 12 TMSs.

Firmicutes

ABC exporter of Ruminococcus torques
Membrane protein (M) (D4M3V3)
ATPase (C) (D4M3V2)

 
3.A.1.147.5

Putative 2 component ABC exporter with a membrane protein of 569 aas and 12 TMSs.

Firmicutes

Putative exporter of Natranaerobius thermophilus
Membrane protein (M) (B2A6N2)
ATPase (C) (B2A6N1)

 
3.A.1.147.6

Putative two component ABC exporter. There are 78 ABC systems including 28 importers and 50 exporters. Based on NBD sequence similarity, ABC transporters in C. difficile were classified into 12 sub-families according to their substrates (Pipatthana et al. 2021). All ABC exporters, accounting for 64% of the total ABC systems, may be involved in antibiotic resistance.

Firmicutes

Putative ABC exporter of Clostridium difficile
Membrane protein (M) (C9XJW9)
ATPase (C) (C9XJX0)

 
3.A.1.147.7

Putative ABC transporter with a membrane protein of 582 aas and 11 TMSs.

Firmicutes

ABC transporter of Thermaerobacter marianensis
Membrane protein (M) (E6SIR8)
ATPase (C) (E6SIR7)

 
3.A.1.147.8

Putative ABC exporter with a membrane protein of 544 aas and 12 TMSs

Firmicutes

ABC exporter of Streptococcus pneumoniae
Membrane protein (M) (B8ZKM8)
ATPase (C) (B8ZKM9)

 
3.A.1.147.9

Putative ABC exporter

Euryarchaea

ABC exporter of Methanocella conradii
Membrane protein (M) (H8I7G4)
ATPase (C) (H8I7G5)

 


3.A.1.148 Functionally Uncharacterized ABC2-4 (ABC2-4) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.148.1

ABC lantibiotic NAI-107 immunity exporter, MlbYZ (Pozzi et al. 2015).

Actinobacteria

MlbYZ of Microbispora sp. ATCC PTA-5024
MlbY (258 aas, 6 TMSs; M)
MlbZ (300 aas; C)

 
3.A.1.148.2

ABC transport system, PspY (264 aas)/PspZ (301 aas)

Actinobacteria

PspYZ of Planomonospora alba
PspY (M; 264 aas)
PspZ (C; 301 aas)

 
3.A.1.148.3

Uncharacterized ABC transporter

Chloroflexi

Uncharacterized ABC transporter of Ktedonobacter racemifer

 
3.A.1.148.4

Uncharacterized ABC transporter, AbcYZ [Y (D2BBE4) = M with 6 TMSs; Z (D2BBE3)= C.]

Actinobacteria

AbcYZ of Streptosporangium roseum

 


3.A.1.149 Functionally Uncharacterized ABC2-5 (ABC2-5) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.149.1

ABC immunity system, TrnFG, protecting the bacteria from the bacteriocin, thuricin CD. TrnF is of 213 aas and 6 TMSs while TrnG is of 285 aas and 0 TMSs.  A 79 aa protein, TrnI with 2 TMSs, also provides immunity against thuricin CD, but the mechanism is unknown (Mathur et al. 2014). These proteins incoded in the thuricin operon.

TrnFG of Bacillus thuringiensis

 
3.A.1.149.2

Uncharacterized two component ABC-2 transporter.

UP of Clostridium intestinale
U2PSG5, M, 216 aas, 6 TMSs in a 2 + 4 arrangement
U2NJR5, C, ATPase of 290 aas

 
3.A.1.149.3

Putative 2 component ABC exporter.

Putative ABC exporter
S7U3S6, M, 215 aas, 6 TMSs in a 2 + 4 arrangement
S7U8X0, C, 285 aas, ATPase, ABC-2

 


3.A.1.15 The Manganese/Zinc/Iron Chelate Uptake Transporter (MZT) Family (Similar to 3.A.1.12, 3.A.1.14 and 3.A.1.16)


Examples:

TC#NameOrganismal TypeExample
3.A.1.15.1Manganese (Mn2+) porterCyanobacteriaMntABC of Synechocystis 6803
MntA (C)
MntB (M)
MntC (R)
 
3.A.1.15.10The Mn2+/Zn2+ transporter MntABC (KB of Mn2+ and Zn2+ is 0.1μM which bind with equal affinity to the same site (Lim et al., 2008)BacteriaMntABC of Neisseria meningitidis:
MntA (C) (A1IQK5)
MntB (M) (A1IQK4)
MntC (R) (Q5FA63)
 
3.A.1.15.11

The zinc uptake porter, YcdHI-YceA; AdcA/AdcC/AdcB (Gaballa et al., 2002).

Firmicutes

YcdHI-YceA of Bacillus subtilis
AdcA (YcdH) (R) (O34966)
AdcC (YcdI) (C) (O34946)
AdcB (YceA) (M) (O34610)

 
3.A.1.15.12

Metal ion (probably iron) uptake permease , YtgABC-RD. The third gene in the ytg operon is fused, the N-terminal membrane domain being fused to the C-terminal transcriptional regulator homologous to the diphtheria toxin repressor, DtxR. These two domains may be proteolitically processed yielding the two active proteins (Thompson et al. 2012). 

Chlamydiae

YtgABC-RD of Chlamydia trachomatis 
YtgA (R) (O9S529)
YtgB (C) (084071)
YtgC-R (M-R) (084072)
YtgD (M) (084073) 

 
3.A.1.15.13

The ZnuA18/ZnuA08/ZnuB/ZnuC zinc (Zn2+) uptake system (Hudek et al. 2013).  ZnuB (M) and ZnuC (C) can function with either of two zinc ion receptors, ZnuA18 (R) which is encoded in the znuACB operon, and ZnuA08 (R) which is encoded elsewhere on the chromosome.  ZnuA18 is more efficient that ZnuA08 in promoting uptake (Hudek et al. 2013).

Cyanobacteria

Zn2+ uptake system of Nostoc punctiforme
ZnuA18 (R) (B2IWS9)
ZnuA08 (R) (B2J0B7)
ZnuB (M) B2IWT1)
ZnuC (C) (B2IWT0)

 
3.A.1.15.14

High affinity Mn2+ uptake complex, PsaABC (Lisher et al. 2013). The crystal structure of the manganese transporter PsaBC from Streptococcus pneumoniae has been solved in an open-inward conformation (Neville et al. 2021). The type II transporter has a tightly closed transmembrane channel due to "extracellular gating" residues that prevent water permeation and ion reflux. Below these residues, the channel contains a metal coordination site, which is essential for manganese translocation. Mutagenesis of the extracellular gate perturbs manganese uptake, while coordination site mutagenesis abolishes import. These structural features are well conserved in metal-specific ABC transporters and are represented throughout the kingdoms of life. Collectively, these results define the structure of PsaBC and reveal the features required for divalent cation transport (Neville et al. 2021).

Firmicutes

PsaABC of Streptococcus pneumoniae
PsaA (R; 309 aas)
PsaB (C; 240 aas)
PsaC (M; 282 aas)

 
3.A.1.15.15

High affinity Mn2+ uptake complex, MntABC.  The 3-d structure of MntC has been  solved to 2.2Å resolution (Gribenko et al. 2013).

Firmictues

MntABC of Staphylococcus aureus 
MntA of 247 aas (C)
MntB of 278 aas (M)
MntC of 309 aas (R)

 
3.A.1.15.16

ZnuABC Zinc/Manganese/iron uptake porter

ZnuABC of Leptospira sp.
ZnuA (R) 345 aas
ZnuB (M) 275 aas
ZnuC C) 210 aas

 
3.A.1.15.17

ZnuABC Zinc/Manganese/Iron uptake porter

 

ZnuABC of Bdellovibrio bacteriovorus
ZnuA (R)
ZnuB (M)
ZnuC (C)

 
3.A.1.15.18

ABC high affinity Zinc (Zn2+) uptake porter, ZnuABC.  The similar system from Y. pestis has been characterized (Bobrov et al. 2014; Neupane et al. 2018). ZnuA (R) of that systems can bind up to 5 zinc ions with high affinity.

ZnuABC of Yersinia pseudotuberculosis
ZnuA, 318 aas, Q66AT6
ZnuB, 261 aas, Q66AT8
ZnuC, 253 aas, Q66AT7

 
3.A.1.15.19

Zinc ion ABC uptake system, AztABCD, where AztD is a periplasmic chaparone protein that feeds Zn2+ into AztC, the periplasmic receptor/binding protein for the transporter (Neupane et al. 2018).

AztABCD of Paracoccus denitrificans
AztA, 309 aas, R, (A1B2F3)
AztB, 288 aas, 9 TMSs, M, (A1B2F2)
AztC. 263 aas, C, (A1B2F1)
AztD, 408 aas, Periplasmic chaparone (A1B2F4)

 
3.A.1.15.2Manganese (Mn2+) and zinc (Zn2+) porterFirmicutesScaABC of Streptococcus gordonii
ScaA (R)
ScaB (M)
ScaC (C)
 
3.A.1.15.20

ABC uptake transporter specific for Mn2+ and Fe2+, SloABC (Paik et al. 2003). The system is repressed by Mn2+ together with the SloR repressor, encoded by a gene downstream of sloABC.

SloABC of Streptococcus mutans

 
3.A.1.15.21

Mn2+ ABC uptake system, MntABC. Titratable transmembrane residues and a hydrophobic plug are essential for manganese import via the Bacillus anthracis ABC transporter MntBC-A (Kuznetsova et al. 2021). Zinc is a high-affinity inhibitor. The transmembrane metal permeation pathway is lined with six titratable residues that can coordinate the positively charged metal, and mutagenesis studies showed that they are essential for manganese transport. Modelling suggested that access to these titratable residues is blocked by a ladder of hydrophobic residues, and ATP-driven conformational changes open and close this hydrophobic seal to permit metal binding and release. The conservation of this arrangement of titratable and hydrophobic residues among ABC transporters of transition metals suggests a common mechanism (Kuznetsova et al. 2021).

MntABC of Bacillus cereus
MntA, R, 311 aas, 1 N-terminal TMSs, Q4V0W6
MntB, C, 249 aas, 0 TMSs, Q4V0W4
MntC, M, 288 aas, 7 TMSs, Q4V0W5

 
3.A.1.15.3

Zinc (Zn2+) porter, AdcABC/AII

Firmicutes

AdcABC of Streptococcus pneumoniae
AdcA (R)
AdcB (M)
AdcC (C)
AdcAII (Lmb) (R)

 
3.A.1.15.4Iron and manganese porterProteobacteriaYfeABCD of Yersinia pestis
YfeA (R)
YfeB (C)
YfeC (M)
YfeD (M)
 
3.A.1.15.5

Zinc (Zn2+) porter of E. coli, ZnuABC.  Required for Zn2+ homeostasis and virulence in the close E. coli relative, Salmonella enterica (Ammendola et al., 2007).

Proteobacteria

ZnuABC (YebLMI) of E. coli
ZnuA (R)
ZnuC (C)
ZnuB (M)

 
3.A.1.15.6Iron (Fe2+)/Zinc (Zn2+)/Copper (Cu2+) porterFirmicutesMtsABC of Streptococcus pyogenes
MtsA (R)
MtsB (C)
MtsC (M)
 
3.A.1.15.7

Manganese (Mn2+) (Km=0.1 μM) and iron (Fe2+) (5 μM) porter (inhibited by Cd2+ > Co2+ > Ni2+, Cu2+) (most similar to YfeABCD of Yersinia pestis (TC #3.A.1.15.4)). Important for virulence in Salmonella (Karlinsey et al., 2010).

Proteobacteria

SitABCD of Salmonella typhimurium
SitA (R)
SitB (C)
SitC (M)
SitD (M)

 
3.A.1.15.8

Manganese (Mn2+), zinc (Zn2+) and possibly iron (Fe2+) uptake porter, TroABCD (Hazlett et al., 2003).  Transcription of the operon is controlled by the Mn2+-activated (not Zn2+- or Fe2+-activated) repressor, TroR (153 aas, acc# F7IW50;) TroR contains a metal-binding domain homologous to the YtgC-R protein (3.A.1.15.12) which has the membrane domain of this ABC transporter (N-terminus) fused to the repressor domain (C-terminus) (Liu et al. 2013).  TroA (Tromp1), the periplasmic metal binding protein, was originally reported to be an outer membrane porin (Zhang et al. 1999), but this proved to be incorrect.

Spirochaetes

TroABCD of Treponema pallidum
TroA (R) P96116
TroB (C) P96117
TroC (M) P96118
TroD (M) P96119

 
3.A.1.15.9Manganese (Mn2+) and Iron (Fe2+) porter, SitABCD (Davies and Walker, 2007)BacteriaSit ABCD of Sinorhizobium meliloti
SitA (R) - (Q92LL5)
SitB (M) - (Q92LL4)
SitC (C) - (Q92LL3)
SitD (M) - (Q92LL2)
 


3.A.1.150 Functionally Uncharacterized ABC2-6 (ABC2-6) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.150.1

Putative ABC transporter consisting of an ATPase and three membrane proteins having 4, 10 and 2 TMSs, respectively.  The structure of the ATPase is similar to those of ABC transorteers, and expression is down regulated in response to cold shock (Gerwe et al. 2007).

Putative ABC transporter of Pyrococcus furiosus

 
3.A.1.150.2

Putative ABC transporter consisting of an ATPase and 3 membrane proteins having 4, 10 and 2 TMSs.

Putative ABC transporter of Pyrococcus furiosus

 


3.A.1.151 Functionally Uncharacterized ABC2-7 (ABC2-7) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.151.1

3-component putative ABC transporter with two membrane proteins and an ATPase. These three genes are adjacent to a gene encoding a DegV domain-containing protein, a fatty acid binding domain, also found in PTS mannose EIIA proteins (TC# 4.A.6) and dihydrolyacetone kinases (Schulze-Gahmen et al. 2003; Kinch et al. 2005; Nan et al. 2009).

Putative ABC transporter of Halothermothrix orenii

 
3.A.1.151.2

Putative 3-compenent ABC transporter consisting of two membrane proteins and a cytoplasmic ATPase.  Adjacent to genes coding for a MoaJ/NirJ iron-sulfur nitrite-like oxidoreductase and an antilisterial bacteriocin biosynthetic enzyme, AlbA (B5YBB2 and 3, respectively).  The system could be a bacteriocin exporter.

ABC transporter of Dictyoglomus thermophilum
B5YBA9, M, 186 aas and 6 TMSs (may be N-terminally truncated)
B5YBB0, M, 223 aas and 6 TMSs (both in a 2 + 4 arrangement)
BSYBB1, C, 239 aas, ATPase

 


3.A.1.152 The lipopolysaccharide export (LptBFG) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.152.1

LPS export system, LptF (M), LptG (M) and LptB (C).  This system is also listed in TCDB under TC#1.B.42.1.2 as part of a multicomponent system.  The entire system is described in detail there. LptB2FG extracts LPSs from the IM and transports them to the outer membrane. Luo et al. 2017 reported the crystal structure of nucleotide-free LptB2FG from P. aeruginosa. It shows that LPS transport proteins LptF and LptG each contain a TM domain (TMD), a periplasmic beta-jellyroll-like domain and a coupling helix that interacts with LptB on the cytoplasmic side. The LptF and LptG TMDs form a large outward-facing V-shaped cavity in the IM. Mutational analyses suggested that LPS may enter the central cavity laterally, via the interface of the TMD domains of LptF and LptG, and is expelled into the beta-jellyroll-like domains upon ATP binding and hydrolysis by LptB. These studies suggest a mechanism for LPS extraction by LptB2FG that is distinct from those of classical ABC transporters that transport substrates across the IM (Luo et al. 2017). LptB2FG extracts LPS from the periplasmic face of the IM through a pair of lateral gates and then powers transperiplasmic transport to the OM through a slide formed by either of the periplasmic domains of LptF or LptG, LptC, LptA and the N-terminal domain of LptD. The structural and functional studies of the seven lipopolysaccharide transport proteins provide a platform to explore the unusual mechanisms of LPS extraction, transport and insertion from the inner membrane to the outer membrane (Dong et al. 2017). LptB2 binds novobiocin which stimulates its export activity and renders the membrane more impermeable to novobiocin (May et al. 2017).

LptFGB2 of Pseudomonas aeruginosa
LptF, M, Q9HXH4, 375 aas, 6 TMSs
LptG, M, Q9HXH5, 354 aas, 6 TMSs
LptB, C, Q9HVV6, 241 aas, ATPase

 
3.A.1.152.10

Probable LptF/G or YjgP/Q membrane proteins of an ABC exporter, possibly specific for a lipopolysaccharide.  Adjacent genes code for proteases/peptidases.  An ATPase that energizes this system has not been identified. The two membrane proteins are of 360 and 380 aas with 6 TMSs in a 3 + 3 TMS arrangement, separated by a large hydrophilic loop. Members of this family often work together with outer membrane proteins of TC family 1.B.42, and some of these outer membrane proteins are fused to one of the subunites of these systems.

LptF/G of Candidatus Pelagibacter sp. (marine metagenome)

 
3.A.1.152.2

Putative ABC exporter of the YjgP/Q (LptFG) family.  The membrane protein has 772 aas and 12 TMSs in a (3 + 3)2 duplicated topology.  The gene adjacent to this membrane protein gene encodes an ABC1 ATPase of 583 aas and 6 N-terminal TMSs with a C-terminal ATPase domain. Most ATPases of family 3.A.1.152 are of the ABC2-type. Thus, it is unlikely that this protein serves to energized the YjgP/Q-dependent transport process. This protein is in TCDB with TC# 3.A.1.106.17.

Putative ABC transporter of Acidobacterium ailaaui

 
3.A.1.152.3

Uncharacterized ABC system of the YjgP/Q family; the two membrane proteins are encoded by adjacent genes, but the gene for the ATPase was not found.  However, a soluble OstA homologue (Q5SL97) of 824 aas is encoded adjacent to the two membrane protein-encoding genes.

UP of Thermus thermophilus

 
3.A.1.152.4

Uncharacterized YjgP/YjgQ family homologue of 441 aas and 6 TMSs. No other YjgP homologue and no ATPase is encoded adjacent to the gene encoding this protein.

UP of Chlorobium phaeovibrioides (Prosthecochloris vibrioformis)

 
3.A.1.152.5

Uncharacterized YjgP/Q homologue of 266 aas and 6 TMSs. No ATPase or another YjgP homologue is encoded by a gene adjacent to this one.

YjgP homologue of Leptonema illini

 
3.A.1.152.6

YjgP/Q homologue of 584 aas an 8 TMSs in a 2 + 3 +3 arrangement.

YjgP homologue of Bizionia argentinensis

 
3.A.1.152.7

YjgP/Q family protein of 392 aas and 6 TMSs

YjgP homologue of Gimesia maris

 
3.A.1.152.8

Uncharacterized YgjP homologue of 585 aas and 6 TMSs; the central hydrophilic domain is 350 aas long, about twice that of many of the homologues.  It might be duplicated.

YgjP homologue of Niabella soli

 
3.A.1.152.9

Lipopolysaccharide transporter that exports LPS from the external surface of the cytoplasmic membrane to the outer membrane, LptB2FG. The 134-kDa protein complex is unique among ABC transporters because it extracts lipopolysaccharide from the external leaflet of the inner membrane and propels it along a filament that extends across the periplasm to directly deliver lipopolysaccharide into the external leaflet of the outer membrane. Dong et al. 2017 reported the crystal structure of this transporter in which both LptF and LptG are composed of a beta-jellyroll-like periplasmic domain and six TMSs. LptF and LptG together form a central cavity containing highly conserved hydrophobic residues. Structural and functional studies suggest that LptB2FG uses an alternating lateral access mechanism to extract lipopolysaccharide and traffic it along the hydrophobic cavity toward the transporter's periplasmic domains. The structure has been presented by Dong et al. 2017. LPS transport involves long-ranging bi- directional coupling across cellular compartments between cytoplasmic LptB and periplasmic regions of the Lpt transporter (Lundstedt et al. 2020). LPS transport from the inner membrane (IM) to the OM is achieved by seven lipopolysaccharide transport proteins (LptA-G). LptB2FG, a type VI ABC transporter, forms a stable complex with LptC, extracts LPS from the IM and powers LPS transport to the OM. Luo et al. 2021 reported the cryo-EM structures of LptB2FG and LptB2FGC from Klebsiella pneumoniae in complex with LPS. The LptB2FG-LPS structure provides detailed interactions between LPS and the transporter, while the LptB2FGC-LPS structure may represent an intermediate state in which the transmembrane helix of LptC has not been fully inserted into the transmembrane domains of LptB2FG (Luo et al. 2021).

 

LptB2FG of Klebsiella pneumoniae
LptB, 241 aas; 0 TMSs, A6TEM0
LptF, 365 aas, 6 TMSs, A6THI3
LptG, 360 aas, 6 TMSs, A6THI4

 


3.A.1.153 The Functionally Uncharacterized ABC-X (ABC-X) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.153.1

ABC transporter complex YtrBCD that may play a role in acetoin utilization during stationary phase and sporulation (Yoshida et al. 2000). Expression is induced early in the stationary phase. The six ytr genes form a single operon, transcribed from a promoter present upstream of ytrA. YtrA, which possesses a helix-turn-helix motif of the GntR family, may be a repressor that regulates its own transcription as well as the whole operon. Inactivation of the operon led to a decrease in the maximal cell yield and less-efficient sporulation. B. subtilis produces acetoin as an external carbon storage compound and then reuses it later during stationary phase and sporulation. Possibly the Ytr porter plays a role (Yoshida et al. 2000). The YtrEF system, believed to be a distinct ABC efflux system (M. Saier, unpublished results), can be found under TC# 3.A.1.122.19.

YtrBCD of Bacillus subtilis
YtrB, 292 aas (C)
YtrC, 328 aas (M)
YtrD, 325 aas (M)

 
3.A.1.153.2

Putative ABC acetoin exporter, ABC-2-like protein (M) plus ATPase (C).

ABC porter of Bacillus thuringiensis
ABC2-2-like protein of 375 aas and 8 TMSs (A0RI83)
BAC-type ATPase of 298 aas (A0RI82)

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.154.1

Uncharacterized protein

Actinobacteria

Uncharacterized protein of Streptomyces coelicolor (Q9K3K9)  

 
3.A.1.154.2

Uncharacterized protein of 316 aas and 6 TMSs.

UP of Hoyosella subflava (Amycolicicoccus subflavus)

 
3.A.1.154.3

Uncharacterized protein of 353 aas and 5 or 6 TMSs.

UP of Gordonia alkanivorans

 
3.A.1.154.4

Uncharacterized protein of 12 TMSs in a 1 + 5 + 1 +5 arrangement.  TMSs 5 and 6 as well as 11 and 12 are separated by about 30 - 50 residues.

UP of Mycobacterium abscessus

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.155.1

The phage infection protein of 901 aas, PIP (Geller et al. 1993). The PIP family (3.A.1.155) includes large proteins with 1 N-terminal hydrophobic TMS, a hydrophilic domain of variable length, and 5 C-terminal putative TMSs. The functionally characterized protein from Lactococcus lactis is of 901 aas (Geller et al., 1993). Homologues obtained with one PSI-BLAST iteration include members of the MmpL family of the RND superfamily (e.g., a Bacillus protein, gi#89208076; 1038 aas). With poorer scores, a protein annotated as an ABC-2-like sequence (gi#89200681; 392 aas with 1 TMS followed by a 150 residue hydrophilic domain followed by a C-terminal 5 putative TMSs) was retrieved. Another protein annotated as ABC-2 was smaller with 6 putative TMSs in a 2 + 3 + 1 arrangement (gi#57234453; 241 aas). The hydrophilic domain in these proteins may show sequence similarity with the large periplasmic hydrophilic domains of RND porters (2.A.6.1 - 9).

Bacteria

PIP of Lactococcus lactis (P49022)

 
3.A.1.155.2The putative ABC-2-like protein of 678 aas (topology-like PIP) BacteriaABC-2-like protein of Arthrobacter sp. (gi#116669229)
 
3.A.1.155.3

Uncharacterized protein YhgE (ORFB)

Bacilli

YhgE of Bacillus subtilis

 
3.A.1.155.4

X(3)LX(2)G heptad repeat protein of 779 aas

Firmicutes

Heptad repeat protein of Lachnospiraceae bacterium 2_1_46FAA

 
3.A.1.155.5

Uncharacterized YhgE/Pip domain-containing protein of 432 aas and 6 TMSs.

UP of Streptomyces himastatinicus

 
3.A.1.155.6

Uncharacterized protein of 499 aas and 7 putative TMSs in a 1 + 5 + 1 TMS arrangement.  This protein may interrelate 9.B.74 and 2.A.6.10 (subfamily) which may NOT belong to the RND superfamily.

UP of Dietzia alimentaria

 
3.A.1.155.7

YhfE/Pip domain protein of 740 aas and 6 or 7 TMSs in a 1 + 5 or 6  arrangement.

YhfE protein of Gulosibacter molinativorax

 
3.A.1.155.8The ABC-2-like protein of 392 aas

Bacteria

ABC-2-like protein of Bacillus cereus (A7GKA4)

 
3.A.1.155.9

Uncharacterized protein of 397 aas and  6 TMSs in a 1 + 5 TMS arrangement

UP of Bacillus cereus

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.156.1

ABC transporter permease

ABC permease of Rubeoparvulum massiliense
(M) 232 aas and 6 TMSs (WP_048600991.1)
(C) 244 aas (WP_048600992.1)

 
3.A.1.156.2

ABC transporter with a membrane protein of 219 aas and 6 TMSs and an ATP-binding protein, YxlF, of 316 aas.

ABC-2 transporter of Lokiarchaeum sp.
(M), 219 aas and 6 TMSs
(C), 316 aa

 
3.A.1.156.3

ABC2 transporter of unknown substrate specificity with two membrane constituents and one ATPase.

Tricomponent ABC exporter of Bacillus licheniformis
M1, 170 aas and 5 TMSs, ARC67021
M2, 212 aas and 5 TMSs, ARC67023
C, ATPase, YxlF, 307 aas, ARC67024

 
3.A.1.156.4

Two component ABC transporter with one M subunit and one C subunit.

ABC transporter of Clostridioides difficile

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.157.1

Putative ABC3 porter with a 10 TMS membrane protein and an ATPase.  This system is encoded by genes that are adjacent to those encoding 3.A.1.207.7 and 3.A.1.207.9. It is not known if these three systems are distinct or function somehow together.

ABC3 porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 
3.A.1.157.1

Putative ABC3 porter with a 10 TMS membrane protein and an ATPase. This system is encoded by genes that are adjacent to those encoding 3.A.1.207.7 and 3.A.1.207.9. It is not known if these three systems are distinct or function somehow together.

ABC3 porter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 
3.A.1.157.2

Putative ABC3-type porter of 956 aas with 10 TMSs in a 1 + 3 + 2 + 1 + 3 TMS arrangement. Two ABC-type ATPases are encoded by genes adjacent to the membrane protein, and on the other side is encoded a membrane protein of 509 aas with two TMSs, one N-termnal, and one C-terminal. This last protein is included here because if could be an auxilliary protein of the ABC exporter.

ABC3-type exporter of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 
3.A.1.157.3

Putative ABC3 transporter of two constituents

ABC3 porter of Candidatus Heimdallarchaeota archaeon

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.158.1

Uncharacterized ABC-type exporter possibly with 4 protein components, all of which are encoded by genes that map adjacent to each other, YybMLKJ.  The proteins of this system are distantly related to members of ABC families 3.A.1.124, 132, 146 and 157. 

YybJKLM of Bacillus subtilis
YybJ, C, 218 and 0 TMSs
YybK, M, 251 aas and 6 TMSs
YybL, M, 236 aas and 6 TMSs
YybM, M, 251 aas and 6 TMSs

 
3.A.1.158.2

Putative ABC-type multidrug transport system with 3 protein components.

BAC porter with three components
ABC-A, M, 264 aas and 6 TMSs
ABC-B, M, 287 aas and 6 TMSs
ABC-C, C, 220 aas and 0 TMSs

 
3.A.1.158.3

ABC exporter of three protein components

ABC exporter of Clostridium beijerinckii (Clostridium acetobutylicum)
ABC-A, M, 6 TMSs, A6LQ39
ABC-B, M, 6 TMSs, A6LQ40
ABC-C, C, 0 TMSs, A6LQ41

 
3.A.1.158.4

Putative 4 component ABC system with 3 membrane proteins, all with 6 TMSs, and one ATPase, all encoded by genes that are adjacent to each other.

Putative 4 component ABC exporter of Lachnospiraceae bacterium (gut metagenome)

HCO29007, M, 6 TMSs
HCO29006, M, 6 TMSs
HCO29005, M, 6 TMSs
HCO29004, C, ATPase, 0 TMSs

 
3.A.1.158.6

ABC-2 type transport system with three 6 TMS membrane proteins (M) and one ATP-binding protein (C).

ABC2 system of Murimonas intestini
PWJ78148, M
PWJ78149, M
PWJ78150, M
PWJ78151, C

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.159.1

Uncharacterized ABC transport protein of 541 aas and 12 TMSs in a 6 + 6 TMS arrangement, where each 6 TMS unit exhibits a 2 + 2 + 2 TMS arrangement.  The gene adjacent to the transporter gene encodes the ATPase.

UP ABC transporter of Isoptericola variabilis

 

 
3.A.1.159.2

Putative 12 TMS ABC permease of 534 aas, HalU (Besse et al. 2015). The gene adjacent to the membrane protein gene is the putative ATPase gene.

HalU of Halalkalicoccus jeotgali

 
3.A.1.159.3

Putative two compoonent ABC permease, with a membrane protein of 12 TMSs and an ATPase encoded by the adjacent gene.

ABC permease of Actinoplanes friuliensis

 
3.A.1.159.4

Putative two component ABC permease of 510 aas for the membrane protein and 12 TMSs.

ABC permease of Halobacterium salinarum (Halobacterium halobium)

 
3.A.1.159.5

Putative ABC membrane transport protein of 525 aas and 14 TMSs in a 2 + 2 + 2 + 1 or 2 + 2 + 2 TMS arrangement. It is homologous to other proteins annotated as ABC transporters and hypothetical proteins.

PT of Subtercola boreus

 
3.A.1.159.6

PAM68 family protein of 524 aas and 14 TMSs in a 2 +2 + 2 + 2 + 2 + 2 + 2 TMS arrangement. As for 3.A.1.159.5, The two central TMSs are suspect.

PAM68 protein of Cryobacterium sp.

 


3.A.1.16 The Nitrate/Nitrite/Cyanate Uptake Transporter (NitT) Family (Similar to 3.A.1.12 and 3.A.1.17)


Examples:

TC#NameOrganismal TypeExample
3.A.1.16.1

Four component nitrate/nitrite porter (Kikuchi et al. 1996).  It's synthesis occurs in response to nitrite, not nitrate in a nitrate reductase mutant (Kikuchi et al. 1996).

Cyanobacteria

NrtABCD of Synechococcus sp. (PCC 7942)
NrtA (R)
NrtB (M)
NrtC (C)
NrtD (C)

 
3.A.1.16.2Bispecific cyanate/nitrite transporter (functions in both cyanate and nitrite assimilation; Maeda and Omata, 2009).
Cyanobacteria

CynABD of Synechococcus PCC7942
CynA (R)
CynB (M)
CynD (C)

 
3.A.1.16.3Bicarbonate porter (activated by low [CO2] mediated by CmpR; (Nishimura et al., 2008))CyanobacteriaCmpABCD of Synechococcus sp.
CmpA (R)
CmpB (M)
CmpC (C)
CmpD (C)
 
3.A.1.16.4

Nitrate uptake system, NrtABCD (Frías et al., 1997). Molecular interactions within the nitrate uptake system of Anabaena PCC7120 have been reported (but are a bit confusing) (Swapnil et al. 2021).

Cyanobacteria

NrtABCD of Anabaena (Nostoc) sp. PCC 7120
NrtA (R) (Q44292)
NrtB (M) (Q8YRV7)
NrtC (C-R) (Q8YRV8)
NrtD (C) (Q8YZ25)

 
Examples:

TC#NameOrganismal TypeExample
Examples:

TC#NameOrganismal TypeExample


3.A.1.17 The Taurine Uptake Transporter (TauT) Family (Similar to 3.A.1.12 and 3.A.1.16)


Examples:

TC#NameOrganismal TypeExample
3.A.1.17.1Taurine (2-aminoethane sulfonate) porterProteobacteriaTauABC of E. coli
TauA (R)
TauB (C)
TauC (M)
 
3.A.1.17.10

Aliphatic sulfonate (alkanesulfonate) import permease, SsuABC (YcbOEM) and is regluated by the transcriptional activator, Cbl (van Der Ploeg et al. 1999; Eichhorn and Leisinger 2001).

Proteobacteria

SsuABC of E. coli
SsuA (YcbO), (R), 319 aas
SsuB (YcbE), (C), 255 aas
SsuC (YcbM), (M), 263 aa

 

 
3.A.1.17.11

Putative ABC transporter specific for riboflavin, RibXYZ. RibY is called "NMT1/THI5 like domain protein" (Anderson et al. 2015).

Riboflavin transporter, RibXYZ, of Thermobaculum terrenum
RibY, 1 N-terminal TMS; R (D1CEG8)
RibX, 7 TMSs, M (D1CEG9)
RibZ, unknown

 
3.A.1.17.12

Sulfonate and sulfonate ester uptake transporter, SsuABC (Koch et al. 2005).

SsuABC of Corynebacterium glutamicum
SsuA (R)
SsuB (C)
SsuC (M)

 
3.A.1.17.13

Putative thiamine (vitamin B1)-specific transporter, ThiXYZ (Rodionova et al. 2015).

ThiXYZ of Chloroflexus aurantiacus
ThiX, (M, 5 TMSs) (A9WDS0)
ThiY, (R, 1 TMS) (A9WDR9)
ThiZ, (C, 0 TMSs) (A9WDR8)

 
3.A.1.17.14

Riboflavin uptake porter, RibXY (RibX, 168 aas and 6 TMSs; RibY, 351 aas) (Gutiérrez-Preciado et al. 2015).

RibXY of Chloroflexus aurantiacus

 
3.A.1.17.2Aromatic sulfonate porterProteobacteriaSsuABC of Pseudomonas putida
SsuA (R)
SsuB (C)
SsuC (M)
 
3.A.1.17.3

Putative hydroxymethylpyrimidine transport system, ThiXYZ (Rodionov et al., 2002). Regulated by TPP (thiamin) riboswitch. Potentially takes up a pyrimidine moiety of thiamin.

Bacteria

ThiXYZ of Haemophilus influenzae
ThiZ (C) (P44656)
ThiX (M) (Q57306)
ThiY (R) (P44658)

 
3.A.1.17.4

The taurine uptake system, TauABC (Krejcík et al., 2008).

Proteobacteria

TauABC of Neptuniibacter caesariensis
TauA (R) (Q2BM68)
TauB (C) (Q2BM69)
TauC (M) (Q2BM70)

 
3.A.1.17.5The phthalate uptake system, OphFGH (Chang et al. 2009).

Bacteria

OphFGH of Burkholderia capacia
OphF (R) (C0LZR7)
OphG (M) (C0LZR8)
OphH (C) (C0LZR9)

 
3.A.1.17.6

Putative hydroxymethylpyrimidine transport system, ThiXYZ (Rodionov et al., 2002). Regulated by TPP (thiamin) riboswitch. Potentially takes up a pyrimidine moiety of thiamin. ThiY is homologous to the yeast THI5 HMP-P synthase (P43534) (Bale et al., 2010).

Actinobacteria, Proteobacteria

ThiXYZ of Pasteurella multocida
ThiX (M) (Q9CLG9)
ThiY (R) (Q9CLH1)
ThiZ (C) (Q9CLG8)

 
3.A.1.17.7

Putative riboflavin transport system, RibXY. Regulated by an FMN riboswitch (Vitreschak et al. 2002).

Chloroflexi

RibXY of Roseiflexus castenholzii
RibX (M) (A7NLS3)
RibY (R) (A7NLS2)

 
3.A.1.17.8

Putative thiamine transport system, ThiXYZ (Rodionov et al., 2002). Regulated by TPP (thiamin) riboswitch.

Chloroflexi

ThiXYZ of Roseiflexus castenholzi

ThiX (M) (A7NH43)

ThiY (R) (A7NH44)

ThiZ (C) (A7NH45)

 
3.A.1.17.9

Uncharacterized membrane protein of 733 aas and 12 TMSs. The other constituents of the system have not been identified.

Rhodophyta

UP of Chondrus crispus

 


3.A.1.18 The Cobalamin Precursor/Cobalt (CPC) Family

The putative cobalamin precursor/cobalt (CPC) transporter family includes proteins of about 190 aas with 4-6 TMSs. These proteins are encoded in operons that are subject to regulation by vitamin B12 (Rodionov et al. 2003). These and other ECF ABC families (3.A.1.18, 23, 25, 26, 28, 31 and 32) have been reviewed (Rempel et al. 2018).


Examples:

TC#NameOrganismal TypeExample
3.A.1.18.1

Putative ECF transporter, EcfSTA; regulated by a cobalamin riboswitch.

Bacteria

EcfSTA of Roseifluxes sp. RS-1
EcfS (S) (A5UXW2)
EcfT (T) (A5UXW1)
EcfA (A) (A5UXW0)

 
3.A.1.18.2

Putative Co2+ ECF transporter, EcfSTA

Bacteria

EcfSTA of Gloeobacter violaceus
EcfS (S) (Q7NIY0)
EcfT (T) (Q7NIX9)
EcfA (A) (Q7NIX8)

 
3.A.1.18.3

Putative Co2+ ECF transporter, EcfSTA

Bacteria

EcfSTA of Syntrophobotulus glycolicus
EcfS (S) (F0SWZ4)
EcfT (T) (F0SWZ5)
EcfA (A) (F0SWZ6)

 


3.A.1.19 The Thiamin Uptake Transporter (ThiT) Family (Most similar to 3.A.1.10, 3.A.1.6 and 3.A.1.8 in that order)


Examples:

TC#NameOrganismal TypeExample
3.A.1.19.1Thiamin, thiamin monophosphate and thiamin pyrophosphate porter. The 2.25 Å structure of ThiB (TbpA) has been solved (Soriano et al., 2008). ProteobacteriaThiBPQ of Salmonella typhimurium (functionally characterized and partially sequenced) and E. coli (fully sequenced but not functionally characterized)
ThiB; TbpA (R)
ThiP; YabK (M)
ThiQ; YabJ (C)
 
3.A.1.19.2

The thiamine pyrophosphate (TPP) uptake porter (Bian et al., 2011).

Bacteria

TPP transporter of Treponena denticola TDE0143/TDE0144/TDE0145
TDE0143 (R) (Q73RE6)
TDE0144 (M) (Q73RE5)
TDE0145 (C) (Q73RE4)

 
3.A.1.19.3

ABC transporter of unknown function. The three genes encoding this system are adjacent to a gene homologous to a mycothiol maleylpyruvate isomerase.

Actinobacteria

ABC transporter of Streptomyces hygroscopicus
Periplasmic binding protein (R) (H2JXL4)
Permease (M) (H2JXL5)
ATPase (C) (H2JXL6)

 
3.A.1.19.4

The putative sulfate/thiosulfate transporter, YnjBCD. YnjB has 12 TMSs. The three genes encoding this system are adjacent to one encoding a thiosulfate:sulfur transferase or a rhodanese (B7L6N1).  Also considered to be a thiamine transporter (Moussatova et al. 2008).

γ-Proteobacteria

YnjBCD of E. coli
YnjB (possible receptor, R) (B7L6M8)
YnjC (M) (B7L6M9)
YnjD (C) (B7L6N0)

 
3.A.1.19.5

Putative ABC transporter, WtpB1/C1: molybdate/tungstate transport system.

Deinococcus-Thermus

ABC transporter of Deinococcus deserti
Permesae (M) (C1CWI2)
ATPase (C) (C1CWI3)
Possible periplasmic receptor (R) (C1CWI4)

 
3.A.1.19.6

Probable 4 component ABC transporter with two ATPase of 387 and 368 aas, respectively, both annotated as MalK, one membrane protein that maps together with the two ATPases and is annotated CysW, and one receptor that maps separately for the other three and is designated MalE. It is not established that this repector maps with the other three constituents, but this has been inferred by the similarities of the two ATPases to MalK. 

Putative 4 component ABC uptake porter of unknown specificity, CysW/MalK/MalK/MalE of Candidatus Heimdallarchaeota archaeon LC_3 (marine sediment metagenome)

 


3.A.1.2 The Carbohydrate Uptake Transporter-2 (CUT2) Family


Examples:

TC#NameOrganismal TypeExample
3.A.1.2.1

Ribose porter.  RbsA has two ATPase domains fused together; RbsB is the substrate receptor; RbsC has 10 TMSs with N- and C-termini in the cytoplasm and forms a dimer (Stewart and Hermodson, 2003).  ABC importers can be divided into two classes. Type I importers follow an alternating access mechanism driven by the presence of the substrate. Type II importers accept substrates in a nucleotide-free state, with hydrolysis driving an inward-facing conformation.  RbsABC2 seems to share functional traits with both type I and type II importers, as well as possessing unique features, and employs a distinct mechanism relative to other ABC transporters (Clifton et al. 2014).

Proteobacteria

RbsABC of E. coli
RbsA (C)
RbsB (R)
RbsC (M)

 
3.A.1.2.10The purine nucleoside permease (probably transports guanosine, adenosine, 2'-deoxyguanosine, inosine and xanthosine with decreasing affinity in this order) (Deka et al., 2006)SpirochaetesPnrA-E of Treponema pallidum
PnrA (R) (TmpC; Tp0319) (P29724)
PnrB (?51 aas; 1 TMS; Tp0320) (O83340)
PnrC (C) (533 aas; duplicated; Tp0321) (NP_218761)
PnrD (M) (400 aas; 10 TMSs; Tp0322) (NP_218762)
PnrE (M) (316 aas; 10 TMSs; Tp0323) (NP_218763)
 
3.A.1.2.11

The erythritol permease, EryEFG (Geddes et al., 2010) (probably orthologous to 3.A.1.2.16)

Bacteria

EryEFG of Sinorhizobium meliloti
EryE (C) (CAC48737)
EryF (M) (CAC48738)
EryG (R) (CAC48735)

 
3.A.1.2.12The (deoxy)ribonucleoside permease; probably takes up all deoxy- and ribonucleosides (cytidine, uridine, adenosine and toxic analogues, fluorocytidine and fluorouridine tested), but not ribose or nucleobases (Webb and Hosie, 2006)BacteriaRnsABCD of Streptococcus mutans
RnsA (R) (AAN58814)
RnsB (C) (AAN58813)
RnsC (M) (AAN58812)
RnsD (M) (AAN58811)
 
3.A.1.2.13

The probable autoinducer-2 (AI-2;, a furanosyl borate diester: 3aS,6S,6aR)-2,2,6,6a-tetrahydroxy-3a-methyltetrahydrofuro[3,2-d][1,3,2]dioxaborolan-2-uide) uptake porter (Shao et al., 2007) (50-70% identical to RbsABC of E. coli; TC# 3.A.1.2.1)

Bacteria

RbsABC of Aggregatibacter actinomycetemcomitans (Actinobacillus succinogens)
RbsA (C) (A6VKS8)
RbsB (R) (A6VKT0)
RbsC (M) (A6VKS9)

 
3.A.1.2.14

Putative L-arabinose porter (Rodionov et al. 2010).

Proteobacteria

AraUVWZ of Shewanella oneidensis
AraU (R) (Q0HIQ8)
AraV (C-C) (Q0HIQ7)
AraW (M; 10 TMSs) (Q0HIQ6)
AraZ (M; 9 TMSs) (Q0HIQ5)

 
3.A.1.2.15

The putative xylitol uptake porter, XltABC (Rodionov et al., 2010)

Proteobacteria

XltABC of Shewanella pealeana
XltA (C) (A8H4W7)
XltB (M; 9 TMSs) (A8H4W6)
XltC (R) (A8H4W5)

 
3.A.1.2.16

The erythritol uptake permease, EryEFG (Yost et al., 2006) (probably orthologous to 3.A.1.2.11)

Bacteria

EryEFG of Rhizobium leguminosarum
EryE (C) (Q1M4Q7)
EryF (M) (Q1M4Q8)
EryG (R) (Q1M4Q9)

 
3.A.1.2.17

General nucleoside uptake porter, NupABC/BmpA (transports all common nucleosides as well as 5-fluorocytidine, inosine, deoxyuridine and xanthosine) (Martinussen et al., 2010) (Most similar to 3.A.1.2.12). NupA is 506aas with two ABC (C) domains. NupB has 8 predicted TMSs, NupC has 9 or 10 predicted TMSs in a 4 + 1 (or 2) + 4 arrangement.

Bacteria

NupABC/BmpA of Lactococcus lactis
BmpA (R) (D2BKA1)
NupA (C) (A2RKA7)
NupB (M) (A2RKA6)
NupC (M) (A2RKA5)

 
3.A.1.2.18

Xylose porter (Nanavati et al. 2006). Regulated by xylose-responsive regulator XylR (Kazanov et al., 2012).

Thermotogae

XylFEK of Thermotoga maritima
XylF (M) (TM0112) (Q9WXW7)
XylE (R) (TM0114) (Q9WXW9)
XylK (C) (TM0115) (Q9WXX0)

 
3.A.1.2.19

D-ribose porter (Nanavati et al., 2006). Induced by ribose (Conners et al., 2005).

Thermotogae

RbsABC of Thermotoga maritima
RbsA (C) (TM0956) (Q9X051)
RbsB (R) (TM0958) (Q9X053)
RbsC (M) (TM0955) (Q9X050)

 

 
3.A.1.2.2

Arabinose porter (Horazdovsky and Hogg 1989).

Proteobacteria

AraFGH of E. coli
AraF (R)
AraG (C)
AraH (M)

 
3.A.1.2.20

Glucose porter. Also bind xylose (Boucher and Noll 2011). Induced by glucose (Frock et al. 2012). Directly regulated by glucose-responsive regulator GluR (Kazanov et al., 2012).

Thermotogae

GluEFK of Thermotoga maritima
GluE (

 

 

GluE (R) (ThemaDRAFT_1377) (G4FGN5)
GluF (M) (ThemaDRAFT_1376) (G4FGN4); 9 TMSs
GluK (C) (ThemaDRAFT_1375) (G4FGN3)

 

 
3.A.1.2.21

The myoinositol (high affinity)/ D-ribose (low affinity) transporter IatP/IatA/IbpA. The structure of IbpA with myoinositol bound has been solved (Herrou and Crosson 2013).

α-Proteobacteria

IatP/IatA/IbpA of Caulobacter crescentus
IatP (M) (B8H230)
IatA (C) (B8H229)
IbpA (R) (B8H228)

 
3.A.1.2.22

ABC sugar transporter that plays a role in the probiotic benefits through acetate production (Fukuda et al. 2012).

Actinobacteria

Sugar transporter of Bifidobacterium longum
BL1694, 385 aas (R) (Q8G3R1)
BL1695, 517 aas (C) (Q8G3R0)
BL1696, 405 aas (M) (Q8G3Q9)

 
3.A.1.2.23

ABC sugar transporter, FruEFGK, important for the probiotic effect of Bifidobacterium longum and involved in producing acetate (Fukuda et al. 2012).  The system is specific for fructose (highest affinity) ribose and xylose.  All three sugars induce the system (Wei et al. 2012). 

Actinobacteria

Sugar transporter of Bifidobacterium longum
BL0033 of 327 aas (R) (Q8G848)
BL0034 of 513 aas (C) (Q8G847)
BL0035 of 356 aas (M) (Q8G846)
BL0036 of 340 aas (M) (Q8G845)

 
3.A.1.2.24

XylFGH downstream of characterized transcriptional regulator, ROK7B7 (Sco6008); XylF (Sco6009); XylG (Sco6010); XylH (Sco6011)) (Świątek et al. 2013).

Actinobacteria

 

XylFGH of Streptomyces coelicolor 
XylF (R)
XylG (C)
XylH (M; 12 TMSs) 

 
3.A.1.2.25

Putative sugar uptake porter, YtfQRT/YjfF (Moussatova et al. 2008).

Proteobacteria

YtfQRT/YjfF of E. coli
YtfQ (R)
YtfR (C)
YtfT (M)
YjfF (M)

 
3.A.1.2.26

Xylose transporter, XylFGH (XylF (R), 359 aas; XylG (C), 525 aas; XylH (M), 389 aas.  Controlled by a 3 component sensor kinase/response regulator system (XylFII, sensor, A6LW07; LytS, SK, A6LW08; YesN, RR, A6LW09) (Sun et al. 2015). The XylFII-LytS complex provides the molecular basis for D-xylose utilization and metabolic modification (Li et al. 2017).

Firmicutes

XylFGH of Clostridium beijerinckii
XylF (R)
XylG (C)
XylH (M; 12 TMSs)

 
3.A.1.2.27

Sugar (pentose?) transport system, YphDEF

YphDEF of E. coli
YphD (M) 332 aas, 10 TMSs
YphE (C) 503 aas
YphF (R) 327 aas

 
3.A.1.2.28

Riboflavin uptake ABC transporter, RfuABCD.  The periplasmic binding protein (RfuA) has been crystallized at 1.3 Å resolution with riboflavin bound (Deka et al. 2013). Similar systems are found in other spirochetes such as Treponema denticola, and Borrelia burgdorferi (Deka et al. 2013).

RfuABCD of Treponema pallidum
RfuA, R, 343 aas and 1 N-terminal TMS
RfuB, C, 586 aas and 0 TMSs
RfuC, M, 377 aas and 9 or 10 TMSs
RfuD, M, 313 aas and 9 TMSs (may be N-terminally truncated)

 
3.A.1.2.29

High affinity fructose uptake porter, FrtABC, Km (fructose) = ~100μM; expression of the frtABC operon is regulated by the product of the upstream gene, frtR, FrtR, a LacI/GalR-type repressor that allows activation in the presence of fructose (Ungerer et al. 2008). When FruR is eliminated, the cells become hypersensitive to fructose, and the level of fruABC expression is much higher than in the presence of wild type cells grown on fructose (Ungerer et al. 2008).

FrtABC of Anabaena (Nostoc) variabilis
FrtA, Ava2171, Q3MB45, 341 aas with 1 N-terminal TMS (R)
FrtB, Ava2172, Q3MB44, 517 aas and 0 TMSs (C)
FrtC, Ava2173, Q3MB43, 332 aas and 8 TMSs (M)

 
3.A.1.2.3Galactose/glucose (methyl galactoside) porterProteobacteriaMglABC of E. coli
MglA (C)
MglB (R)
MglC (M)
 
3.A.1.2.30

3-component ABC-type putative general nucleoside uptake porter consisting of a receptor, a putative lipoprotein with two N- and C-terminal TMSs (R; 405 aas), an integral membrane protein of about 20 TMSs in a 1 + 4 (tight) + 4 (loose) +2 +1 + 4 (tight) +4 (loose) TMS arrangement (M; 864 aas), and a cytoplasmic ATPase (C; 563 aas).  It appears that the membrane protein contains a 9 (or 10) TMS repeat unit, and that there are two extra TMSs separating the two repeat units.  These are homologous to the two membrane constituents of TC# 3.A.1.2.17.

ABC uptake porter of Candidatus Heimdallarchaeota
OLS24537, R
OLS24538, C
OLS24539, M

 
3.A.1.2.31

Putative purine porter with 4 components (Chandravanshi et al. 2019).

Putative purine porter of Thermus thermophilus
R, 379 aas and 1 TMS (Q5SIR3)
M, 277 aas and 9 TMSs
(Q5SIR2)
M, 349 aas and 8 - 10 TMSs
(Q5SIR1)
C, 489 aas and 0 TMSs
(Q5SIR0)

 
3.A.1.2.32

ABC-type putative arabinose transport system, AraEGHP. The genes encoding this system are found within a large gene cluster including many arabinose/pentose metabolic enzymes, a sensor kinase/response regulator pair, and an ABC uptake system specific for araino-oligosaccharides of 2 to 8 sugar units (TC# 3.A.1.1.57) (Lansky et al. 2020).

AraEGHP of Geobacillus stearothermophilus (Bacillus stearothermophilus)
AraE, R, B3EYL8
AraG, C, ATPase, B3EYM1
AraH, M, B3EYM2
AraP, R, B3EYL5

 
3.A.1.2.33

XylFGH of T. ethanolicus (Erbeznik et al. 2004).

XylFGH of Thermoanaerobacter ethanolicus (Clostridium thermohydrosulfuricum), a thermophilic, anaerobic, ethanol-producing eubacterium
XylF (R), 366 aas
XylG (C),507 aas
XylH (M)388 aas and 12 TMSs.

 
3.A.1.2.4Xylose porterProteobacteriaXylFGH of E. coli
XylF (R)
XylG (C)
XylH (M)
 
3.A.1.2.5Multiple sugar (arabinose, xylose, galactose, glucose, fucose) putative porterProteobacteriaChvE, GguAB of Agrobacterium tumefaciens
ChvE (R)
GguA (C)
GguB (M)
 
3.A.1.2.6

D-allose porter.  The structure of AlsB has been solved at 1.8 Å resolution (Chaudhuri et al. 1999). Ten residues from both the domains form 14 hydrogen bonds with the sugar. 6-Deoxy-allose, 3-deoxy-glucose and ribose bind with reduced affinity so AlbP can function as a low affinity transporter for D-ribose (Chaudhuri et al. 1999).

Proteobacteria

AlsABC of E. coli
AlsB (R)
AlsA (C)
AlsC (M)

 
3.A.1.2.7Fructose/mannose/ribose porterProteobacteriaFrcABC of Sinorhizobium meliloti
FrcA (C)
FrcB (R)
FrcC (M)
 
3.A.1.2.8

Autoinducer-2 (AI-2, a furanosyl borate diester: (3aS,6S,6aR)-2,2,6,6a-tetrahydroxy-3a-methyltetrahydrofuro[3,2-d][1,3,2]dioxaborolan-2-uide) uptake porter (Taga et al., 2001, 2003)

Proteobacteria

LsrACDB of E. coli
LsrB (R) AAC74589
LsrA (C) AAC74586
LsrC (M) AAC74587
LsrD (M) AAC74588

 
3.A.1.2.9Rhamnose porter (Richardson et al., 2004) (Transport activity is dependent on rhamnokinase (RhaK; AAQ92412) activity (Richardson and Oresnik, 2007) This could be an example of group translocation!)ProteobacteriaRhaSTP of Rhizobium leguminosarum bv. trifolii
RhaS (R) AAQ92407
RhaT (C) AAQ92408
RhaP (M) AAQ92409
 


3.A.1.20 The Brachyspira Iron Transporter (BIT) Family (Most similar to 3.A.1.6, 3.A.1.8 and 3.A.1.11)


Examples:

TC#NameOrganismal TypeExample
3.A.1.20.1

The iron transporter, BitABCDEF (Dugourd et al. 1999).

Spirochaetes

BitABCDEF of Brachyspira (Serpulina) hyodysenteriae
BitA (R)
BitB (R)
BitC (R)
BitD (C)
BitE (M)
BitF (M)

 
3.A.1.20.2

Hexose-phosphate transporter.  Transports glucose-6-phosphate (Km = 0.3 υM) and fructose-6-phosphate (1.3 υM).  Sugar phsophates can be used as both carbon and phosphate sources (Moisi et al. 2013).

Proteobacteria

Sugar phosphate uptake permease, FbpABC of Vibrio cholerae
FbpA 344 aas (R) (Q9KLQ7)
FbpB 700 aas (M) (Q9KLQ6)
FbpC 351 aas (C) (Q9KLQ5)

 
3.A.1.20.3

Iron (Fe3+) uptake porter, AfuABC (FbpABC) (Chin et al. 1996). AfuA has been characterized (Willemsen et al. 1997).

Proteobacteria

AfuABC (FbpABC) of Actinobacillus pleuropneumoniae
AfuA (R)
AfuB (M)
AfuC (C)

 

 
3.A.1.20.4

Putative glycerol phosphodiester uptake transporter.  The three genes encoding this system are in an operon with a gene encoding a glycerophosphodiester phosphodiesterase, providing the evidence that this transporter might function to take up such substrates.

Putative glycerol phosphodiester uptake porter of Bdellovibrio exovorus

A11Q_2445 (R), 344 aas and 1 TMS
A11Q_2446 (M), 541 aas and 12 TMSs
A11Q_2447 (C), 245 aas and 0 TM

 

 
3.A.1.20.5

Possibly a Mg2+-citrate uptake porter with three components, R, M and C, as suggested by Mandal et al. 2019.  However, this system appears more likely to be a ferric iron uptake system, based on sequence similarity studies (see other members of TC sub-family 3.A.1.20).

Fe3+ or Mg2+-citrate porter of Thermus thermophilus

 
Examples:

TC#NameOrganismal TypeExample
3.A.1.200.51

ABCC4 efflux transporter exports C-glycosylated flavones (CGFs), which are the main flavonoids in duckweed (Lemna turionifera), known for their diverse pharmacological activities and nutritional values (Wang et al. 2024). The protein is of 1496 aas with 15 or 16 TMSs in a 5 or 6 + 6 + 6 TMS arrangement. The LtP1L transcription factor directly binds to a novel AC-like cis-element in the promoter of a tonoplast-localized ATP-binding cassette (ABC) transporter LtABCC4 and activated its expression. The preference of LtABCC4 for isoorientin over orientin during vacuolar transport was evidenced by the significant reduction of isoorientin compared to orientin in the Ltabcc4crispr lines (Wang et al. 2024).

ABCC4 of Lemna turionifera (duckweed)

 


3.A.1.201 The Multidrug Resistance Exporter (MDR) Family (ABCB)


Examples:

TC#NameOrganismal TypeExample
3.A.1.201.1

Broad specificity multidrug resistance (MDR1; MDR-1; Pgp; P-gp; ABCB1; P-glycoprotein) efflux pump. It exports organic cations and amphiphilic compounds of unrelated chemical structure.  These include: antibiotics, anti-viral agents, cancer chemotheraputic agents, hypertensives, depressants, histamines, emetics, and the protease inhibitor, lopinavir. Pgp also exports immunosuppressants, detergents, long-chain fatty acids, HIV protease inhibitors, synthetic tetramethylrosamine analogues, calcein M, etc.); it is also a peptide efflux pump, and peptide inhibitors have been designed (Tarasova et al. 2005). It is also a phospholipid (e.g., phosphatidyl serine), cholesterol and sterol flippase. It binds and probably transports inhibitors and agonists of SUR (3.A.1.208.4) (Bessadok et al., 2011). Modulatory effects of inhibitory amlodipine and tamoxifen on P-glycoprotein efflux activities have been studied (Darvari and Boroujerdi 2004).  It is found in many tissues (intestine, kideny, blood brain barrier, liver, etc. (Wang et al. 2024). The 3-d structure has been determined (Aller et al., 2009). It can pump from the cytoplasmic leaflet to either the outer leaflet or the outer medium (Katzir et al., 2010). The inhibitor, 5''-fluorosulfonylbenzoyl 5''-adenosine, an ATP analogue, interacts with both drug-substrate- and nucleotide-binding sites (Ohnuma et al., 2011). Inhibited by sildenafil (Shi et al., 2011), verapamil, indomethacin, probenecid, cetirizine (He et al. 2010), and lapatinib derivatives (Sodani et al., 2012), several of which are also substrates. HG-829 is a potent non-competitive inhibitor (Caceres et al., 2012).  Berberine, palmatine, jateorhizine, cetirizine and coptisine are all P-gp substrates, and cyclosporin A and verapamil are potent inhibitors (He et al. 2010; Zhang et al., 2011).  Transports clarithromycin (CAM), a macrolide antibiotic used to treat lung infections, more effectively than azithromycin (AZM) or telithromycin (TEL) (Togami et al. 2012).  Nucleotides, lipids and drugs bind synergistically to the pump (Marcoux et al. 2013).  Fluorescent substrates have been identified (Strouse et al. 2013).  The central cavity undergoes alternating access during ATP hydrolysis (van Wonderen et al. 2014).  Structural data suggest that signals are transduced through intracellular loops of the TMSs that slot into grooves on the NBDs. The Q loops at the base of these grooves are required to couple drug binding to the ATP catalytic cycle of drug export (Zolnerciks et al. 2014). Ocotillol analogues are strong competitive inhibitors (Zhang et al. 2015).  Durmus et al. 2015 have reviewed PGP transport of cancer chemotheraputic agents.  ABCB1 variants modulate therapeutic responses to modafinil and may partly explain pharmacoresistance in Narcolepse type 1 (NT1) patients (Moresco et al. 2016).  Many inhibitors have been identified (Hemmer et al. 2015).  The open-and-close motion of the protein alters the surface topology of P-gp within the drug-binding pocket, explaining its polyspecificity (Esser et al. 2016). The ATP- and substrate-coupled conformational cycle of the mouse Pgp transporter have been defined, showing that the energy released by ATP hydrolysis is harnessed in the NBDs in a two-stroke cycle (Verhalen et al. 2017).  Rilpivirine inhibits MDR1- and BCRP-mediated efflux of abacavir and increases its transmembrane transport (Reznicek et al. 2017).  It transports Huerzine A in the brain, a drug that is used for the treatment of Alzheimer's disease (Li et al. 2017). AbcB1 acts in concert with ABCA1, ABCG2 and ABCG4 to efflux amyloid-β peptide (Aβ) from the brain across the blood-brain barrier (BBB) (Kuai et al. 2018).The structure has been determined with the ABCB1 inhibitor, zosuquidar, bound.  This structure reveals the transporter in an occluded conformation with a central, enclosed, inhibitor-binding pocket lined by residues from all TMSs. The pocket spans almost the entire width of the lipid membrane and is occupied exclusively by two closely interacting zosuquidar molecules (Alam et al. 2018).  Iit is also inhibited by dacomitinib (Fan et al. 2018). Moreover, Kim and Chen 2018 presented the structure of human P-glycoprotein in the outward-facing conformation, determined by cryo-electron microscopy at 3.4-Å resolution. The two nucleotide-binding domains form a closed dimer occluding two ATP molecules. The drug-binding cavity observed in the inward-facing structures is reorientated toward the extracellular space and is compressed to preclude substrate binding. This observation indicates that ATP binding, not hydrolysis, promotes substrate release (Kim and Chen 2018). P-gp also transports opioid peptides (Ganapathy and Miyauchi 2005). MDR1 has been quantified in primary human renal cell carcinoma cells and corresponding normal tissue, and down-regulation or expression loss was documented in tumor tissues, corroborating its importance in drug resistance and efficacy (Poetz et al. 2018). Regarding the conformational transitions, first the transition is driven by the NBDs, then transmitted to the cytoplasmic parts of TMSs, and finally to the periplasmic parts. The trajectories show that besides the translational motions, the NBDs undergo a rotation movement (Zhang et al. 2018). Isoxanthohumol is a substrate and competitive inhibitor which reverses ABCB1-mediated doxorubicin resistance (Liu et al. 2017). Tariquidar is a potent inhibitor, even when taken orally (Matzneller et al. 2018). Combined oral administration of the ovarian hormones, ethinyl estradiol and progesterone, significantly lowered both MDR-1 mRNA and MDR-1 protein in the ovary (Brayboy et al. 2018). Its expression in immune cells plays a protective role from xenobiotics and toxins (Bossennec et al. 2018). Oxypeucedanin reverses P-gp-mediated drug transport by inhibition of P-gp activity and P-gp protein expression as well as downregulation of P-gp mRNA levels (Dong et al. 2018). Alam et al. 2019 determined the 3.5-Å cryo-EM structure of substrate-bound human ABCB1 reconstituted in lipidic nanodiscs, revealing a single molecule of the chemotherapeutic compound paclitaxel (Taxol) bound in a central, occluded pocket. A second structure of inhibited, human-mouse chimeric ABCB1 revealed two molecules of zosuquidar occupying the same drug-binding pocket. Minor structural differences between substrate- and inhibitor-bound ABCB1 sites are amplified toward the nucleotide-binding domains (NBDs), revealing how the plasticity of the drug-binding site controls the dynamics of the ATP-hydrolyzing NBDs. Ordered cholesterol and phospholipid molecules suggest how the membrane modulates the conformational changes associated with drug binding and transport (Alam et al. 2019). The TMS4/6 cleft may be an energetically favorable entrance gate for ligand entry into the binding pocket of P-gp (Xing et al. 2019). The epigallocatechin gallate derivative Y6 reverses drug resistance mediated by ABCB1 (Wen et al. 2019). Substrate-induced acceleration of ATP hydrolysis correlates with stabilization of a high-energy, post-ATP hydrolysis state characterized by structurally asymmetric nucleotide-binding sites, but this state is destabilized in the substrate-free cycle and by high-affinity inhibitors in favor of structurally symmetric nucleotide binding sites (Dastvan et al. 2019). It transports temozolomide (TMZ) which is used as a treatment of glioblasomas (Malmström et al. 2019). Unconventional cholesterol translocation on the surface of Pgp provides a secondary transport model for the known flippase activity of ABC exporters of cholesterol (Thangapandian et al. 2020). An in silico multiclass classification model capable of predicting the probability of a compound to interact with P-gp has been developed using a counter-propagation artificial neural network (CP ANN) based on a set of 2D molecular descriptors, as well as an extensive dataset of 2512 compounds (1178 P-gp inhibitors, 477 P-gp substrates and 857 P-gp non-active compounds) (Mora Lagares et al. 2019).  Jervine is a natural teratogenic compound isolated from Veratrum californicumLiu et al. 2019 showed that jervine sensitizes the anti-proliferation effect of doxorubicin (DOX) and that the synergistic mechanism was related to the intracellular accumulation of DOX via modulating ABCB1 transport. Jervine did not affect the expression of ABCB1 in mRNA or protein levels. However, jervine increased the ATPase activity of ABCB1 and probably served as a substrate of ABCB1. Jervine binds to a closed ABCB1 conformation and blocks drug entrance to the central binding site at the transmembrane domain (Liu et al. 2019). 6-Triazolyl-substituted sulfocoumarins inhibit P-gp (Podolski-Renić et al. 2019). ATP binding causes the conformational change to the outward-facing state, and ATP hydrolysis and subsequent release of γ-phosphate from both NBDs allow the outward-facing state to return to the original inward-facing state (Futamata et al. 2020). Replacing the eleven native tryptophans by directed evolution produces an active P-glycoprotein with site-specific, non-conservative substitutions (Swartz et al. 2020). ABCB1 polymorphisms alter P-gp-mediated drug (sunitinib) sensitivities. Homology modeling provided insight into ligand binding through molecular docking studies (Mora Lagares et al. 2020). Sitravatinib  reverses MDR mediated by ABCB1 and partially antagonized ABCC10-mediated MDR (Yang et al. 2020). Apiole from parsley blocks the active P-gp site, with strong binding energy, which, in turn, inhibits doxorubicin and vincristine efflux, increasing the antiproliferative response of these chemotherapeutic agents (Afonso de Lima et al. 2020). The mechanisms of action of synthetic, potent, small molecule P-gp inhibitors have been reviewed (Zhang et al. 2020). ATP binding to the open NBDs and ATP hydrolysis in the closed NBD dimer represent two steps of energy input, each leading to the formation of a high energy state. Relaxation from these high energy states occurs through conformational changes that push ABCB1 through the transport cycle (Szöllősi et al. 2020). 14 conserved residues (seven in both TMsSs 6 and 12) were substituted with alanine and generated a mutant termed 14A (Sajid et al. 2020). Although the 14A mutant lost the ability to pump most of the substrates tested out of cancer cells, it was able to import four substrates, including rhodamine 123 (Rh123) and the taxol derivative flutax-1. Similar to the efflux function of wild-type P-gp, uptake was ATP hydrolysis-, substrate concentration-, and time-dependent. Further mutagenesis identified residues in both TMSs 6 and 12 that synergistically form a switch in the central region of the two helices that governs whether a given substrate is pumped out of or into the cell (Sajid et al. 2020). Helix repacking may be the basis for P-glycoprotein promiscuity (Bonito et al. 2020). The use of carbon nano-onion-mediated dual targeting of P-selectin and P-glycoprotein has been shown to overcome cancer drug resistance (Wang et al. 2021). A sequentially responsive Nnanosystem breaches cascaded bio-barriers and suppresses P-Glycoprotein function for reversing cancer drug resistance (Liu et al. 2020). Lys-268 and the cytoplasmic end of TMS5 may comprise a drug binding site (Demmer et al. 2021). MDR1 protein (ABC-C1) is overexpressed in Giardia intestinalis following incubation with the drugs, albendazole and nitazoxanide (Ángeles-Arvizu et al. 2021). The human P-gp is inhibited by benzophenone sulfonamide derivatives (Farman et al. 2020) and androstano-arylpyrimidines (Gopisetty et al. 2021), and possibly by tepoxalin (McQuerry et al. 2021). The inward facing state of P-glycoprotein in a lipid membrane has been confirmed (Carey Hulyer et al. 2020). For transmembrane pharmaceutical drug transport, non-specific trans-phospholipid bilayer transport may be negligible (Kell 2021). Glabratephrin reverses doxorubicin resistance in triple negative breast cancer by inhibiting P-glycoprotein (Abd-Ellatef et al. 2021). In silico screening of c-Met tyrosine kinase inhibitors targeting nucleotide and drug-substrate binding sites of ABCB1 are potential MDR reversal agents (Moosavi et al. 2022). Quercetin acts as a P-gp modulator via impeding signal transduction from the nucleotide-binding domain to the transmembrane domain (Singh et al. 2022). MDR1 promotes intrinsic and acquired resistance to PROTACs in cancer cells and exports the antiseizure drug, levetiracetam (Behmard et al. 2022). Air pollution exposure increases ABCB1 and ASCT1 transporter levels in mouse cortex (Puris et al. 2022). MDR-1 dysfunction perturbs meiosis and Ca2+ homeostasis in oocytes (Nabi et al. 2022). The P-glycoprotein (ABCB1) transporter has been modelled with in silico methods (Mora Lagares and Novič 2022). A new ABCB1 inhibitor enhances the anticancer effect of doxorubicin in models of non-small cell lung cancer (NSCLC) (Adorni et al. 2023). A homologous series of amphiphiles interact with P-glycoprotein in a membrane environment, and the contributions of polar and non-polar interactions has been estimated (Moreno et al. 2023). TRIP6 transcription is regulated primarily by the cyclic AMP response element (CRE) in hypomethylated proximal promoters in both taxane-sensitive and taxane-resistant MCF-7 cells. In taxane-resistant MCF-7 sublines, TRIP6 co-amplifies with the neighboring ABCB1 gene (Daniel et al. 2023). Inhibition of Cryptosporidium parvum by nitazoxanide (NTZ) and paclitaxel (PTX) has been validated (Yang et al. 2023). New inhibitors of ABCB1 have been identified (Cheema et al. 2023). Inhibitors of MDR pumps (MDR1, MRP1/2 and BCRP) have been described (Kaproń et al. 2023). A hyaluronic acid modified cuprous metal-organic complex reverses multidrug resistance via redox dyshomeostasis (Wan et al. 2023; Duan et al. 2023). The high sensitivity of the steady-state ATP hydrolysis rate to the nature and number of dipolar interactions, as well as to the dielectric constant of the membrane, points to a flopping process, which occurs to a large extent at the membrane-transporter interface (Seelig and Li-Blatter 2023). ABCB1, NCF4, and GSTP1 polymorphisms predicted lower hematological toxicity during induction, while ABCB1 and CRBN polymorphisms predicted lower risk of grade >/=3 infections (Ferrero et al. 2023). Wine-processed Chuanxiong Rhizoma enhances the efficacy of aumolertinib against EGFR mutant non-small cell lung cancer xenografts in nude mouse brain (Niu et al. 2023). The efflux of anti-psychotics through the blood-brain barrier (BBB) via this system has been demonstrated (Nasyrova et al. 2023). Residues from homologous TMSs 4 and 10 are critical for P-glycoprotein (ABCB1)-mediated drug transport (Rahman et al. 2023). Emamectin B1a, Emamectin B1b, Vincristine, Vinblastine, and Vindesine are promising ABCB1 inhibitors that can reverse MDR (Ibrahim et al. 2023). Other substrates and inhibitors from Anemarrhenae rhizoma have been identified (Dai et al. 2022).  Peptides and their analogs can cross the BBB by transmembrane diffusion, saturable transport, and adsorptive transcytosis (Banks 2023). Saturable transport systems are adaptable to physiologic changes and can be altered by disease states. In particular, transport across the BBB of insulin and of pituitary adenylate cyclase activating polypeptide (PACAP) illustrate many of the concepts regarding peptide transport across the BBB (Banks 2023).  Second-site suppressor mutations reveal connections between the drug-binding pocket and the nucleotide-binding domain 1 of human P-glycoprotein (ABCB1) (Murakami et al. 2023).  Betulin derivatives are multidrug reversal agents targeting P-glycoprotein (Laiolo et al. 2024). Deaggregation of mutant Plasmodium yoelii de-ubiquitinase UBP1 alters MDR1 localization to confer multidrug resistance (Xu et al. 2024). One can overcome ABCB1-mediated multidrug resistance in castration resistant prostate cancer cases (Sarwar et al. 2024). Pyridoquinoxaline-based P-gp inhibitors are coadjutant against Multi Drug Resistance in cancer (Ibba et al. 2024). Lansoprazole (LPZ) reverses multidrug resistance in cancer through impeding ABCB1 and ABCG2 transporter-mediated chemotherapeutic drug efflux and lysosomal sequestration (Ji et al. 2024). N,N-dimethyl-idarubicin analogues are effective cytotoxic agents for ABCB1-overexpressing, doxorubicin-resistant cells (van Gelder et al. 2024).  Anthranilamide derivatives are dual P-glycoprotein and CYP3A4 (see TC# 9.B.208) inhibitors (Said et al. 2024).  FRα and multiple transporters such as PCFT, RFC, OAT4, and OATPs are likely involved in the uptake of methotraxate (MTX), whereas MDR1 and BCRP are implicated in the efflux of MTX from choriocarcinoma cells (Bai et al. 2024). Sofosbuvir (SOF) is a P-glycoprotein (P-gp) substrate, and carvedilol (CAR) is an inhibitor of P-gp (Fahmy et al. 2024). The effect of ABCB1 polymorphisms on the accumulation of bictegravir has been studied (De Greef et al. 2024). Two other substrates of P-gp are digoxin and paclitaxel (Volpe 2024). ABCB1 transcripts are readily traceable in the liquid-biopsy of ovarian cancer patients (Schwarz et al. 2024). Quinolinone-pyrimidine hybrids are reversal agents of multidrug resistance mediated by P-gp (Laiolo et al. 2021).

Animals, fungi, bacteria

MDR1 of Homo sapiens

 
3.A.1.201.10

Mdr1; resistance to Cilofungin and other drugs (Lamping et al., 2010)

Fungi

Mdr1 (MCMC) of Aspergillus fumigatus (B0Y3B6)

 
3.A.1.201.11

Mdr1 azole resistance efflux pump (Lamping et al., 2010).  Antifungal activity of the repurposed drug disulfiram against Cryptococcus neoformans has been studied (Peng et al. 2023).

Fungi

Mdr1 (MCMC) of Cryptococcus (Filobasidiella) neoformans (O43140)

 
3.A.1.201.12

California mussel ABCB/MDR multixenobiotic resistance efflux pump (Luckenbach and Epel, 2008).

Animals

ABCB/MDR transporter of Mytilus californianus (MCMC) (B2WTH9)

 
3.A.1.201.13

Plasma membrane AbcB5, of 812 aas and 6 TMSs, mediates resistance of tumor cells to doxorubicin and other drugs including taxanes and anthracyclines (Kawanobe et al. 2012) by catalyzing efflux of these drugs (Sakamoto et al. 2019).  Expression in metastatic melanoma cells is affected by nano-TiO2 exposure, which as a sunscreen ingredient, may play a role in metastatic melanoma progression (Zdravkovic et al. 2019).

Animals

ABCB5 of Homo sapiens (Q2M3G0)

 
3.A.1.201.14

P-glycoprotein-1 MDR exporter.  Transports multiple drugs, cancer chemotherapy agents, cancer unrelated compounds and many xenobiotics including ivermectin (Ardelli 2013).  The crystal structure at 3.4 A resolution is available (Jin et al. 2012).  It has 4,000x higher affinity for actinomycin D in the membrane bilayers than in detergent.  A "ball and socket joint" and salt bridges similar to ABC importers suggested that both types of systems, importers and exporters, use the same mechanism to interconnect ATP hydrolysis with transport and achieve alternating access of the substrate binding site to the two sides of the membrane. 

Animals

P-glycoprotein-1 of Caenorhabditis elegans

 
3.A.1.201.15

MDR efflux pump, ABCB1a.  Exports canonical MDR susbtrates such as calcein-AM, bodipy-verapamil, bodipy-vinblastine and mitoxantrone (Gokirmak et al. 2012).

Animals

ABCB1a of Stronglycentrotus purpuratus

 
3.A.1.201.16

MDR efflux pump, ABCB4a.  Exports canonical MDR susbtrates such as calcein-AM, bodipy-verapamil, bodipy-vinblastine and mitoxantrone (Gokirmak et al. 2012).

Animals

ABCB4a of Stronglycentrotus purpuratus

 
3.A.1.201.17

Mitochondrial ABCB10 or ABC-me transporter. It is a mitochondrial inner membrane erythroid transporter involved in heme biosynthesis. ABCB10 possesses an unusually long 105-amino acid mitochondrial targeting presequence, the central subdomain of which (aas 36-70) is sufficient for mitochondrial import (Graf et al. 2004) and is essential for erythropoiesis and protection of mitochondria against oxidative stress.  The 3-d structures of several conformations are available (3ZDQ; Shintre et al. 2013; Sakamoto et al. 2019). Sitravatinib  reverses MDR mediated by ABCB1 and partially antagonized ABCC10-mediated MDR (Yang et al. 2020).

Animals

ABCB10 of Homo sapiens

 
3.A.1.201.18

Leptomycin B resistance protein 1, Pmd1, of 1362 aas and 13 predicted TMSs (Nishi et al. 1992). This protein is similar in sequence to Ste6-2b  of Pichia pastoralis the structure of which has been solved by cryoEM. It transports rodamines 6G and 123, Dvarapamil, flconazole, and itraconazole, and it's ATPase activity is inhibited by terbinafine, niftifine, ketoconazole and asmorilfine, It also appears to interact with sterols (Schleker et al. 2022).

Yeast

Pmd1 of Schizosaccharomyces pombe

 
3.A.1.201.19

Mitochondrial iron/sulfur complex transporter, AbcB13 of 663 aas (Xiong et al. 2010).

Alveolata (Ciliates)

AbcB13 (M-C) of Tetrahymena thermophila

 
3.A.1.201.2

Bile salt export pump, BSEP, ABCB11 or SPGP in the canalicular membrane of liver cells, is associated with progressive familial intrahepatic cholestasis-2 and benign recurrent intrahepatic cholestasis (Kagawa et al., 2008; Stindt et al. 2013; Park et al. 2016). It exports unconjugaged bile salts and glycine conjugates > taurine conjugates as well as the statin, pravastatin (Nigam 2015). BSEP mediates biliary excretion of bile acids from hepatocytes. Compounds based on GW4064 (Q96RI1), a representative farnesoid X receptor (RXR) agonist, enhance E297G BSEP transport activity (Misawa et al., 2012). Rescue of bile acid transport by ABCB11 variants by CFTR potentiators has been extensively documented as a possible treatment for progressive familial intrahepatic cholestasis-2 (Mareux et al. 2022).  BSEP is expressed in hepatocytes and extrudes bile salts into the canaliculi of the liver. BSEP dysfunction, caused by mutations or induced by drugs is frequently associated with severe cholestatic liver disease. Liu et al. 2023 reported the cryo-EM structure of glibenclamide-bound human BSEP in nanodiscs, revealing the basis of small-molecule inhibition. Glibenclamide binds the apex of a central binding pocket between the transmembrane domains, preventing BSEP from undergoing conformational changes, and thus rationalizing the reduced uptake of bile salts. Two high-resolution structures of BSEP trapped in distinct nucleotide-bound states by using a catalytically inactivated BSEP variant (BSEP(E1244Q)) to visualize a pre-hydrolysis state, and wild-type BSEP trapped by vanadate to visualize a post-hydrolysis state. These studies provide structural and functional insight into the mechanism of bile salt extrusion and into small-molecule inhibition of BSEP, which may rationalize drug-induced liver toxicity (Liu et al. 2023). ABCB11 gene variations in children with progressive familial intrahepatic cholestasis type 2 have been identified (Riaz et al. 2024).  Estradiol 17β-d-glucuronide (E217G) induces cholestasis by triggering endocytosis and further intracellular retention of the canalicular transporters Bsep and Mrp2, in a cPKC- and PI3K-dependent manner, respectively. Pregnancy-induced cholestasis has been associated with an E217G cholestatic effect, and is routinely treated with ursodeoxycholic acid (UDCA). TUDC restores function and localization of Bsep/Mrp2 impaired by E217G, by preventing both cPKC and PI3K/Akt activation in a protein-phosphatase-independent manner (Medeot et al. 2024).

Animals

BSEP of Homo sapiens

 
3.A.1.201.20

12 TMS multidrug resistance transprter of 1318 aas, AbcB15 (Xiong et al. 2010) is the probable exporter of dichlorodiphenyltrichloroethane (DDT). Expression is induced by treatment with DDT, and this transporter appears to be responsible for DDT tolerance by pumping it out of the cell (Ning et al. 2014).

Alveolata (Ciliates)

AbcB15 (M-C-M-C) of Tetrahymena thermophila

 
3.A.1.201.21

Half sized ABCB1 drug (verapamil; rhodamine 6G) exporter of specificity similar to that of P-glycoprotein (3.A.1.201.1).  The 3-d structures of the unbound (2.6 Å) and the allosteric inhibitor-bound (2.4 Å) forms have been determined (Kodan et al. 2014).  The outward opening motion is required for ATP hydrolysis. Kodan et al. 2019 have reported a pair of structures of this homodimeric P-glycoprotein: an outward-facing conformational state with bound nucleotide, and an inward-facing apo state, at resolutions of 1.9 Å and 3.0 Å, respectively. Features that can be clearly visualized include ATP binding with octahedral coordination of Mg2+; an inner chamber that significantly changes in volume with the aid of tight connections among TMSs 1, 3, and 6; a glutamate-arginine interaction that stabilizes the outward-facing conformation; and extensive interactions between TMS1 and TMS3, a property that distinguishes multidrug transporters from floppases (Kodan et al. 2019). The crystal structure of the CmABCB1 G132V mutant, which favors the outward-facing state, reveals the mechanism of the pivotal joint between TMS1 and TMS3 (Matsuoka et al. 2021). The crystal structure of this CmABCB1 multi-drug exporter in lipidic mesophase has been revealed by LCP-SFX, suggesting flexibility of the substrate exit region of the protein (Pan et al. 2022).  Structure-based alteration of tryptophan residues of CmABCB1 allows assessment of substrate binding using fluorescence spectroscopy (Inoue et al. 2022).

Rhodophyta (Algae)

ABCB1 of Cyanidioschyzon merolae

 
3.A.1.201.22

Mitochondrial ATP-binding cassette 1, ABCB8.  Mediates doxorubicin resistance in melanoma cells (Elliott and Al-Hajj 2009). It is regulated by the Sp1 transcription factor and down regulated by mthramycin A which blocks Sp1 binding to the DNA (Sachrajda and Ratajewski 2011). It is also regulated by neuropilin-1, NRP1 (TC# 8.A.47.1.5) (Issitt et al. 2018). The cryo-EM structure of ABCB8 bound to AMPPNP in the inward-facing conformation was solved with a resolution of 4.1 Å. hABCB8 shows an open-inward conformation when ATP is bound, and cholesterol molecules were identified in the transmembrane domain of hABCB8 (Li et al. 2021).

Animals

ABCB8 of Homo sapiens

 
3.A.1.201.23

The cyclic AMP efflux pump of 1432 aas, ABCB3 (Miranda et al. 2015).

Slime molds

ABCB3 of Dictyostelium discoideum

 
3.A.1.201.24

Multidrug exporter, MDR49 or Pgp of 1302 aas and 12 TMSs.  Exports many drugs as well as pollutants such as polycyclic aromatic hydrocarbons (PAHs) which are major sources of air, water and soil pollution.  MDR49 is expressed at all developmental stages of the life cycle and in many tissues (Vache et al. 2007). It is essential for early development, probably because Drosophila germ cell migration depends on lipid-modified peptides that are secreted by MDR49 (Ricardo and Lehmann 2009).

MDR49 of Drosophila melanogaster (Fruit fly)

 
3.A.1.201.25

MDR transporter, Crmdr1 of 1266 aas and 12 TMSs.  Crmdr1 is constitutively expressed in the root, stem and leaf with lower expression in leaf. It has two transmembrane domains (TMDs) and two nucleotide binding domains (NBDs) arranging in "TMD1-NBD1-TMD2-NBD2" direction (Jin et al. 2007).

 

Crmdr1 of Catharanthus roseus (Madagascar periwinkle) (Vinca rosea)

 
3.A.1.201.26

ABC multidrug exporter, MDR1 of 1341 aas, 12 TMSs and two ATPase domains in an MCMC arrangement.  Miltefosine (hexadecylphosphocholine), the first orally available drug available to treat leishmaniasis, is pumped out of the parasite by MDR1, a P-glycoprotein-like transporter. Overexpression of LtrMDR1 increases miltefosine efflux, leading to a decrease in drug accumulation in the parasites and resistance (Pérez-Victoria et al. 2006).

MDR1 of Leishmania major

 
3.A.1.201.27

Multidrug resistance exporter of 1331 aas and 12 TMSs, TratrD or MDR2. Almost identical throughout must of its length to F2PRR1 from T equinum of 1235 aas and 12 TMSs (Martins et al. 2016). Displays increased levels of transcription of the TruMDR2 gene when mycelia were exposed to acriflavine, benomyl, ethidium bromide, ketoconazole, chloramphenicol, griseofulvin, fluconazole, imazalil, itraconazole, methotrexate, 4-nitroquinoline N-oxide (4NQO) or tioconazole. Disruption of the TruMDR2 gene rendered the mutant more sensitive to terbinafine, 4NQO and ethidium bromide than the control strain, suggesting that this transporter plays a role in modulating drug susceptibility in T. rubrum (Fachin et al. 2006).

TratrD or MDR2 of Trichophyton rubrum (Athlete's foot fungus) (Epidermophyton rubrum)

 
3.A.1.201.28

MDR1 alkaloid/multiple drug efflux transporter of 1292 aas and 12 TMSs (Shitan et al. 2003). 

CjMDR1 of Coptis japonica (Japanese goldthread)

 
3.A.1.201.29

ABC transporter B family member 11 isoform X1 or ABCB11 of 1303 aas and 12 TMSs. Functions to export shikonin (Zhu et al. 2017). Shikonin is a naphthoquinone secondary metabolite with  medicinal value, found in Lithospermum erythrorhizon.

ABCB11 of Jatropha curcas (Barbados nut) (closely related to Lithospermum erythrorhizon)

 
3.A.1.201.3

Short chain fatty acid phosphatidylcholine translocase (phospholipid flippase), MDR3; AbcB4; Pgy3.  ABCB4 regulates the secretion into bile of phosphatidylcholine (PC), while ABCG5/G8 is active in the excretion of cholesterol and sterols into bile. It is associated with progressive familial intrahepatic cholestasis type 3 (PFIC3) (Degiorgio et al. 2007) and progressive intrafamilial hepatic disease (Quazi and Molday, 2011)). ABCB4 exhibits narrow drug specificity relative to MDR1 but exports digoxin, paclitaxel, vinblastin and bile acids. It regulates phosphatidylcholine secretion into bile and its translocation across the plasma membrane in hepatocytes (Voloshyna and Reiss, 2011; Kluth et al. 2014) and functions as a floppase (Sakamoto et al. 2019). The cryo-EM structure trapped in an ATP-bound state at a resolution of 3.2 Å has been described (Olsen et al. 2019). The nucleotide binding domains form a closed conformation containing two bound ATP molecules, but only one of the ATPase sites contains bound Mg2+. The transmembrane domains adopt a collapsed conformation at the level of the lipid bilayer, but a large, hydrophilic and fully occluded cavity at the level of the cytoplasmic membrane boundary, with no ligand bound, is present. This indicates a state following substrate release but prior to ATP hydrolysis. These results rationalize disease-causing mutations in human ABCB4 and suggest an 'alternating access' mechanism of lipid extrusion, distinct from the 'credit card swipe' model of other lipid transporters (Olsen et al. 2019). An in vitro assay to investigate ABCB4 transport function has been developed (Temesszentandrási-Ambrus et al. 2023). ABCB4 is located at the canalicular membrane of hepatocytes and is responsible for the secretion of phosphatidylcholine into bile (Lakli et al. 2024). Genetic variations of this transporter are correlated with rare cholestatic liver diseases, the most severe being progressive familial intrahepatic cholestasis type 3 (PFIC3). New small molecular correctors for have been identified to correct traffic-defective ABCB4 variants (Lakli et al. 2024). Progressive familial intrahepatic cholestasis type 3 is caused by ABCB4 gene mutations (Ye et al. 2024).

Animals

MDR3 of Homo sapiens

 
3.A.1.201.30

ABCB10 transporter of 655 aas and 6 TMSs.  It functions in resistance to acaricides (Koh-Tan et al. 2016). Cardiomyocyte-specific deletion of AbcB10 causes cardiac dysfunction via lysosomal-mediated ferroptosis (Do et al. 2024).  AbcB10 knockout cardiomyocytes exhibit increased ROS production, iron accumulation, and lysosomal hypertrophy (Do et al. 2024).

ABCB10 of Rhipicephalus microplus (Cattle tick) (Boophilus microplus)

 
3.A.1.201.31

Permeability glycoprotein, P-Glycoprotein 65, P-GP65, MDR65 of 1302 aas and 12 TMSs. a detoxification efflux pump transporting various lipophilic xenobiotics out of the cells. Exports Polycyclic aromatic hydrocarbons (PAHs), ubiquitous environmental contaminants (Vaché et al. 2006). When flies are exposed to benzo[a]pyrene or to ambient air polluted by higher or lower PAH concentrations, P-gp expression was induced (Vaché et al. 2006).

PGP65 of Drosophila melanogaster (Fruit fly)

 
3.A.1.201.33

ABCB14 of 1247 aas and an MCMC domain arrangement. Transports malate and auxins (Lefèvre and Boutry 2018).

ABCB14 of Arabidopsis thaliana (Mouse-ear cress)

 
3.A.1.201.34

ABCB15 of 1240 aas with a domain order of MCMC.

ABCB15 of Arabidopsis thaliana (Mouse-ear cress)

 
3.A.1.201.35

P-glycoprotein, Pgp, ABCB1, of 1241 aas and 12 TMSs with a domain order MCMC.  Exports geraniol and other monoterpenes. Demissie et al. 2018 reported two structures of this homodimeric P-glycoprotein: an outward-facing conformational state with bound nucleotide and an inward-facing apo state, at resolutions of 1.9 Å and 3.0 Å, respectively. Features that could be clearly visualized include ATP binding with octahedral coordination of Mg2+; an inner chamber that significantly changes in volume with the aid of tight connections among transmembrane helices (TMSs) 1, 3, and 6; a glutamate-arginine interaction that stabilizes the outward-facing conformation; and extensive interactions between TMS1 and TMS3, a property that distinguishes multidrug transporters from floppases. These structural elements were proposed to participate in the mechanism of the transporter (Demissie et al. 2018).

ABCB1 of Lavandula angustifolia

 
3.A.1.201.36

ABCB21 of 1296 aas and a domain order of MCMC. Transports auxins (Lefèvre and Boutry 2018).

ABCB21 of Arabidopsis thaliana (Mouse-ear cress)

 
3.A.1.201.37

ABC protei, MDR1 of 1288 aas with 12 TMSs and a domain order of MCMC. Transports alkaloids (Lefèvre and Boutry 2018).

MDR1 of Coptis japonica

 
3.A.1.201.38

Fusion protein with a complete ABC transporter (domain order MCMC) followed by a complete probable phosphate uptake transpoter, a member of the DASS family (TC# 2.A.47).  Many fusion proteins of this type are present in the NCBI protein database.

Fusion protein of Pochonia chlamydosporia

 
3.A.1.201.39

Multidrug resistance-1, Mdr1, of 1464 aas and 12 TMSs in a 6 +6 TMS arrangement (domain order: M-C-M-C).  Confers resistance to chloroquine (CQ) and primaquine (PQ), but mutations decrease resistance (Kittichai et al. 2018).

Mdr-1 of Plasmodium vivax

 
3.A.1.201.4

The multidrug resistance/chloroquine resistance protein, PfMdr1 (ABCB1, Pgh1).  PfMdr1 is the central system in P. falciparum artemisinin therapy regimen resistance (Gil and Krishna 2017).  PfMDR1 is inhibited by 4 nM actelion (ACT)-213615 and actelion (ACT)-451840 (Brunner et al. 2012, Brunner et al. 2013, Krause et al. 2016),

Protozoa

Pfmdr1 of Plasmodium falciparum (P13568)

 
3.A.1.201.40

Serine protease/ABC transporter B family protein TagA of 1752 aas and 9 putative TMSs with one N-terminal TMS followed by a large hydrophilic region that may correspond to the protease domain, followed by 8 putative TMSs in a 2 + 2 + 2 + 2 TMS arrangement and the ATPase domain. It is required for general cell fate determination at the onset of development andis required for the specification of an initial population of prespore cells in which tagA is expressed. It is also required for normal SDF-2 signaling during spore encapsulation (Good et al. 2003; Cabral et al. 2006).

TagA of Dictyostelium discoideum (Slime mold)

 
3.A.1.201.41

ABC type B transporter of 1207 aas and 12 TMSs. ATP-dependent transporter genes are associated with cystic development (Bai et al. 2020).

ABC transporter of Echinococcus granulosus (tape worm)

 
3.A.1.201.42

Multidrug resistance protein 1, MDR1, of 1475 aas and 12 TMSs in an M-C-M-C arrangement (Pimpat et al. 2020).

MDR1 of Plasmodium malariae

 
3.A.1.201.43

Probable MDR efflux pump of 1451 aas and 12 TMSs in a MCMC domain arrangement where each M domain has 6 TMSs.

MDR exporter of Marchantia polymorpha (liverwort)

 
3.A.1.201.44

ATP-binding cassette transporter ABCB1, P-glycoprotein, MDR1, of 1339 aas and 12 TMSs. P-glycoprotein inhibitors differently affect Toxoplasma gondii, Neospora caninum and Besnoitia besnoiti proliferation (Larrazabal et al. 2021). These organisms are all obligatory intracellular protozoan parasites, and of them, tariquidar treatment affected proliferation only of B. besnoiti (Larrazabal et al. 2021).

MDR1 of Besnoitia besnoiti

 
3.A.1.201.45

ABCB28 homolog of 732 aas and 6 N-terminal TMSs plus a C-terminal ATPase. It may play a role in the Cd2+ stress response, possibly by pumping Cd2+ out of the cell (Zhu et al. 2021).

ABC homolog (M-C) of Digitaria exilis

 
3.A.1.201.46

Multidrug resistance protein homolog 49 of 542 aas and 6 or 7 N-terminal  TMSs (M-C). The sequence may be incomplete. Over-expression of the Mdr49-like transporter in the brown planthopper, Nilaparvata lugens, confers resistance to imidacloprid (Wang et al. 2021).

Mdr49-like of Eumeta japonica, the brown planthopper

 
3.A.1.201.47

Active peptide/heavy metal cation exporter, MDR4 or ABCB4, of 1365 aas and 7 TMSs in a 1 + 2 + 2 + 2 TMS arrangement, followed by a large hydrophilic ATPase domain; it probably has an M-C domain order (Wunderlich 2022).

MDR4 of Plasmodium falciparum

 
3.A.1.201.48

Putative solute exporter of 925 aas and 7 TMSs in a 1 + 2 + 2 + 2 TMS arrangement.

Solute exporter of Plasmodium falciparum

 
3.A.1.201.49

MDR7, ABCB7 active peptide exporter of 1049 aas and 6 TMSs (Wunderlich 2022).

MDR7 of Plasmodium falciparum

 
3.A.1.201.5

Auxin efflux pump Pgp1 (MDR1; ABCB1) (Carraro et al. 2012). Regulated by Twd1, an FK506-binding protein immunophilin prolyl/peptidyl isomerase; 8.A.11.1.1 (Bouchard et al., 2006).  Involved in light-dependent hypocotyl elongation (Sidler et al. 1998). The combination of ibrutinib and paclitaxel can effectively antagonize ABCB1- or ABCC10-mediated paclitaxel resistance (Zhang et al. 2017). Pgp1 also confers herbicide tolerance to cycloheximide, toxic leves of the plant hormone N6-[2-isopentyl]adenine (2iP) and multiple herbicides (Windsor et al. 2003). It is up-regulated under salt stress conditions (Yang et al. 2018).

Plants

Pgp1 of Arabidopsis thaliana (Q9ZR72)

 
3.A.1.201.50

The human ABCB5 exists in two forms (812 aas and 1257 aas). The latter full length protein confers resistance to taxanes and anthracyclines (Kawanobe et al., 2012). Resistance and transport were demonstrated for paclitaxel and docetaxel as well. It is present in a number of stem cells that acts as a regulator of cellular differentiation. It is able to mediate efflux from cells of the rhodamine dye and of the therapeutic drug doxorubicin (Frank et al. 2005; Huang et al. 2004). It is specifically present in limbal stem cells, where it plays a key role in corneal development and repair (Frank et al. 2003).

ABCB5 of Homo sapiens

 
3.A.1.201.51

Multidrug resistance protein 1, ABCB1, of 495 aas and 6 TMSs in a 2 + 2 + 2 TMS arrangement. This system confers triclabendazole resistance in Fasciola hepatica and shows dominant inheritance (Beesley et al. 2023).

ABCB1 of Fasciola hepatica

 
3.A.1.201.52

ABCB4 of 1275 aas and probably 12 TMSs in a 6 + 6 TMS arrangement.  Human ABCB1 and zebrafish (Danio rerio) AbcB4 are functionally homologous multixenobiotic/multidrug (MXR/MDR) efflux transporters that confer the efflux of a broad range of diverse chemical compounds from the cell. These two transporters have different temperature dependencies as expected since Homo is warm blooded while Danio is cold blooded (Luckenbach and Burkhardt-Medicke 2024).

ABCB4 of  Danio rerio (Zebrafish) (Brachydanio rerio)

 
3.A.1.201.6

Auxin efflux pump Pgp19 (MDR11; ABCB19; ABCB21) (regulated by Twd1, an FK506-binding protein immunophilin prolyl/peptidyl isomerase (TC# 8.A.11.1.1) (Bouchard et al., 2006).

Plants

Pgp19 of Arabidopsis thaliana (Q9LJX2)

 
3.A.1.201.7

Auxin efflux pump Pgp4; AbcB4; MDR4; PGP4 (Lefèvre and Boutry 2018) of 1286 aas and 12 TMSs in a MCMC domain arrangement.  Functions in the basipetal redirection of auxin from the root tip. Strongly expressed in root cap and epidermal cells (Terasaka et al., 2005).  Contributes to the basipetal transport in hypocotyls and root tips by establishing an auxin uptake sink in the root cap. Confers sensitivity to 1-N-naphthylphthalamic acid (NPA). Regulates root elongation, the initiation of lateral roots and the development of root hairs. Can transport IAA, indole-3-propionic acid, NPA syringic acid, vanillic acid and some auxin metabolites, but not 2,4-D and 1-naphthaleneacetic acid (Terasaka et al., 2005). Pgps and PINs (TC# 2.A.69) function in coordinated but independent auxin transport but also function interactively in a tissue-specific manner (Blakeslee et al. 2007). Found in the plasma membranes of root hair cells (Cho et al. 2012).  ABCB4 gene mutations may be involved in amiodarone-induced hepatotoxicity (Shi et al. 2024).

Plants

Pgp4 of Arabidopsis thaliana (MCMC) O80725

 
3.A.1.201.8The aluminum chelate (aluminum sensitivity (ALS1)) protein; expressed in root vacuoles half-type ABC transporter (not induced by aluminum; Larsen et al., 2007).PlantsALS1 (M-C) of Arabidopsis thaliana (Q0WML0)
 
3.A.1.201.9Marine skate liver bile salt exporter, BSEP (1348 aas) (transports taurocholine in an ATP-dependent fashion (Cai et al., 2001)) (Most similar to 3.A.1.201.2)AnimalsBSEP of Raja erinacea (MC MC) (Q90Z35)
 


3.A.1.202 The Cystic Fibrosis Transmembrane Conductance Exporter (CFTR) Family (ABCC)


Examples:

TC#NameOrganismal TypeExample
3.A.1.202.1

Cystic fibrosis transmembrane conductance regulator (CFTR) (also called ABCC7); cyclic AMP-dependent chloride channel; also catalyzes nucleotide (ATP-ADP)-dependent glutathione and glutathione-conjugate flux (Kogan et al., 2003) (may also activate inward rectifying K+ channels). The underlying mechanism by which ATP hydrolysis controls channel opening is described by Gadsby et al., 2006. The most common cause of cystic fibrosis (CF) is defective folding of a cystic fibrosis transmembrane conductance regulator (CFTR) mutant lacking Phe508 (DeltaF508) (Riordan, 2008). The DeltaF508 protein appears to be trapped in a prefolded state with incomplete packing of the transmembrane segments, a defect that can be repaired by direct interaction with correctors such as corr-4a, VRT-325, and VRT-532 (Wang et al., 2007). CFTR interacts directly with MRP4 (3.A.1.208.7) to control Cl- secretion (Li et al., 2007). It has intrinsic adenylate kinase activity that may be of functional importance (Randak and Welsh, 2007). The intact CFTR protein mediates ATPase rather than adenylate kinase activity (Ramjeesingh et al., 2008). Regulated by Na+/H+ exchange regulatory cofactors (NHERF; O14745; TC #8.A.24.1.1) (Seidler et al., 2009). Regulated by protein kinase A and C phosphorylation (Csanády et al., 2010). It is also activated by membrane stretch induced by negative pressures (Zhang et al., 2010). TMS6 plays roles in gating and permeation (Bai et al., 2010; 2011). The 3-D structure revealed the probable location of the channel gate (Rosenberg et al., 2011). Conformational changes opening the CFTR chloride channel pore, coupled to ATP-dependent gating, have been studied (Wang and Linsdell, 2012). Alternating access to the transmembrane domain of CFTR has been demonstrated (Wang and Linsdell, 2012). MRP4 and CFTR function in the regulation of cAMP and beta-adrenergic contraction in cardiac myocytes (Sellers et al., 2012). An asymmetric hourglass, comprising a shallow outward-facing vestibule that tapers toward a narrow "bottleneck" linking the outer vestibule to a large inner cavity extending toward the cytoplasmic extent of the lipid bilayer has been proposed (Norimatsu et al., 2012). Small molecule CFTR potentiators and correctors that overcome the efects of deleterious mutations have been identified (Kym et al. 2018).  The intracellular processing, trafficking, apical membrane localization, and channel function of CFTR are regulated by dynamic protein-protein interactions in a complex network. Zhang et al. 2017 reviewed the macromolecular complex of CFTR, Na⁺/H⁺ exchanger regulatory factor 2 (NHERF2; TC# 8.A.24.1.2), and lysophosphatidic acids (LPA) receptor 2 (LPA2; see TC# 9.A.14.2.5) at the apical plasma membrane of airway and gut epithelial cells.  The structure, gating and regulation of the CFTR anion channel has been reviewed (Csanády et al. 2019). Mutants impairing ion conductance giving rise to CF, are partially corrected using the drug ivacaftor, and the structure of CFTR bound to this drug, which keeps the channel open has been solved by cryoEM (Liu et al. 2019). The drug binds to a site with a hinge involved in channel gating. CFTR modulators reduce agonist-induced platelet activation and function; modulators, such as ivacaftor, present a promising therapeutic strategy for thrombocytopathies, including severe COVID-19 (Asmus et al. 2023). Chronic hypoxia reduces the activities of epithelial sodium and CFTR ion channels (Wong et al. 2023). CFTR function on ex vivo nasal epithelial cell models has been evaluated (Terlizzi et al. 2023). The therapeutic potential of phytochemicals for cystic fibrosis has been considered, and curcumin, genistein, and resveratrol have been shown to be effective.  These compounds  have beneficial effects on transporter function, transmembrane conductivity, and overall channel activity (Baharara et al. 2023). VX-661 and VX-445 exert effects on the plasma membrane expression of clinical CFTR variants (McKee et al. 2023). The Cl--transporting proteins CFTR, SLC26A9 (TC# 2.A.53.2.15), and anoctamins (ANO1; ANO6) (TC#s 1.A.17.1.1 and 1.4) appear to have more in common than initially suspected, as they all participate in the pathogenic process and clinical outcomes of airway and renal diseases in humans. Kunzelmann et al. 2023 reviewed electrolyte transport in the airways and kidneys, and the role of CFTR, SLC26A9, and the anoctamins ANO1 and ANO6. Emphasis was placed on cystic fibrosis and asthma, as well as renal alkalosis and polycystic kidney disease. They summarize evidence indicating that CFTR is the only relevant secretory Cl- channel in airways under basal (nonstimulated) conditions and after stimulation by secretagogues. The expressions of ANO1 and ANO6 are important for the correct expression and function of CFTR. The Cl- transporter, SLC26A9, expressed in the airways, may have a reabsorptive rather than a Cl--secretory function. In the renal collecting ducts, bicarbonate secretion occurs through the synergistic action of CFTR and the Cl-/HCO3- transporter SLC26A4 (pendrin; TC# 2.A.53.2.17), which is probably supported by ANO1. In autosomal dominant polycystic kidney disease (ADPKD), the secretory function of CFTR in renal cyst formation may have been overestimated, whereas ANO1 and ANO6 have been shown to be crucial in ADPKD and therefore represent new pharmacological targets for the treatment of polycystic kidney disease (Kunzelmann et al. 2023).  AlphaMissense pathogenicity predictions have been made against cystic fibrosis variants (McDonald et al. 2024). The selectivity filter is accessible from the cytosol through a large inner vestibule and opens to the extracellular solvent through a narrow portal. The identification of a chloride-binding site at the intra- and extracellular bridging point leads to a complete conductance path that permits dehydrated chloride ions to traverse the lipid bilayer (Levring and Chen 2024). The structural basis for CFTR inhibition by CFTRinh-172 has been presented (Young et al. 2024). Fat malabsorption in cystic fibrosis pathophysiology of cystic fibrosis in the gastrointestinal tract may play a role in disease symptoms (McDonald et al. 2024).  Tricyclic pyrrolo-quinazolines interact with CFTR as a novel class of CFTR correctors suitable for combinatorial pharmacological treatments for the basic defect in CF (Barreca et al. 2024). Care for children with CF has been reviewed (Sun and Sawicki 2024).  Cystic Fibrosis causing mutations in the gene CFTR, reduce the activity of the CFTR channel protein and leads to mucus aggregation, airway obstruction and poor lung function. A role for CFTR in the pathogenesis of other muco-obstructive airway diseases such as Chronic Obstructive Pulmonary Disease (COPD) is known. The CFTR modulatory compound, Ivacaftor (VX-770), potentiates channel activity of CFTR and certain CF-causing mutations and has been shown to ameliorate mucus obstruction and improve lung function in people harbouring these CF-causing mutations. SK-POT1 is another compound that can also be used to intervene in the treatment of COPD (Tanjala et al. 2024).  CF-related diabetes (CFRD) is a prevalent comorbidity in people with Cystic Fibrosis (CF), significantly impacting morbidity and mortality rates. Umashankar et al. 2024 evaluated the current understanding of CFRD molecular mechanisms, including the role of CFTR protein, oxidative stress, the unfolded protein response (UPR) and intracellular communication. CFRD manifests from a complex interplay between exocrine pancreatic damage and intrinsic endocrine dysfunction, further complicated by the deleterious effects of misfolded CFTR protein on insulin secretion and action. Studies indicate that ER stress and subsequent UPR activation play critical roles in both exocrine and endocrine pancreatic cell dysfunction, contributing to β-cell loss and insulin insufficiency. Additionally, oxidative stress and altered calcium flux, exacerbated by CFTR dysfunction, impair β-cell survival and function, highlighting the significance of antioxidant pathways in CFRD pathogenesis. Emerging evidence underscores the importance of exosomal microRNAs (miRNAs) in mediating inflammatory and stress responses, offering novel insights into CFRD's molecular landscape. Despite insulin therapy remaining the cornerstone of CFRD management, the variability in response to CFTR modulators underscores the need for personalized treatment approaches (Umashankar et al. 2024).Pyrazole-pyrimidones comprise a new class of correctors of CFTR (Vaccarin et al. 2024).  Rectal organoid morphology analysis (ROMA) provides a novel physiological assay for diagnostic classification in cystic fibrosis (Cuyx et al. 2024).  6,9-dihydro-5H-pyrrolo[3,2-h]quinazolines is a new class of F508del-CFTR correctors for the treatment of cystic fibrosis (Barreca et al. 2024).  CFTR inhibitors display antiviral activity against Herpes Simplex Virus and can effectively suppress HSV-1 and HSV-2 infections, revealing a previously unknown role of CFTR inhibitors in HSV infection (Jiang et al. 2024).

 

Animals

CFTR of Homo sapiens

 
3.A.1.202.2

CFTR, an epithelial ion channel, plays a role in the regulation of epithelial ion and water transport and fluid homeostasis (Bagnat et al. 2010; Navis et al. 2013; Navis and Bagnat 2015). It mediates the transport of chloride ions across the cell membrane. Channel activity is coupled to ATP hydrolysis. The ion channel is also permeable to HCO3-; selectivity depends on the extracellular chloride concentration. CFTR exerts its function in part by modulating the activity of other ion channels and transporters, and it contributes to the regulation of the pH and the ion content of the epithelial fluid layer. It is required for normal fluid homeostasis in the gut (Bagnat et al. 2010) and for normal volume expansion of Kupffer's vesicle during embryonic development as well as for normal establishment of left-right body patterning (Navis et al. 2013; Roxo-Rosa et al. 2015). It is also required for normal resistance to infection by Pseudomonas aeruginosa (Phennicie et al. 2010).

CFTR of Danio rerio (Zebrafish) (Brachydanio rerio)

 


3.A.1.203 The Peroxysomal Fatty Acyl CoA Transporter (P-FAT) Family (ABCD)


Examples:

TC#NameOrganismal TypeExample
3.A.1.203.1

Peroxisomal long chain fatty acyl (LCFA; especially branched chain fatty acids) transporter of 659 aas; associated with Zellweger Syndrome, ABCD3, PMP70, PXMP1.  Can form heterodimers with ABCD1/ALD and ABCD2/ALDR, but the transporter is perdominantly a homodimer (Hillebrand et al. 2007). Dimerization is necessary to form an active transporter. It interacts with PEX19.  abcd3-knockout mice accumulate bile acid precursors suggesting that Abcd3 imports these compounds as CoA derivatives into peroxisomes (Visser et al. 2007).  These mutants also accumulate pristanic acid suggesting that Abcd3 also imports branched chain substrates into peroxisomes (Sakamoto et al. 2019). The unfolded protein response (UPR) detects and restores deficits in the endoplasmic reticulum (ER) protein folding capacity (Torres et al. 2019). Ceapins are aromatic compounds that specifically inhibit the UPR sensor ATF6alpha, an ER-tethered transcription factor, by retaining it at the ER. Ceapin's function is dependent on ABCD3. ABCD3 physically associates with ER-resident ATF6alpha in cells and in vitro in a Ceapin-dependent manner. Ceapins induce the neomorphic association of ER and peroxisomes by directly tethering the cytosolic domain of ATF6alpha to ABCD3's transmembrane regions without inhibiting or depending on ABCD3 transport activity (Torres et al. 2019).  Ceapins act through ABCD3 which binds to ATF6α. causing the ER to be tethered to the peroxysome, preventing ATF6α from carrying out its function as the unfolded protein response sensor (Torres et al. 2019). A CCG expansion in ABCD3 causes oculopharyngodistal myopathy in individuals of European ancestry (Cortese et al. 2024).

Animals

PMP70 of Homo sapiens

 
3.A.1.203.10

Long chain fatty acid transporter consisting of a heterodimer of AbcD1 (719 aas) and AbcD2 (694 aas) (Xiong et al. 2010).

Alveolata (ciliates)

AbcD1/AbcD2 of Tetrahymena thermophila

 
3.A.1.203.11

Putative fatty acid exporter; homodimer (Moussatova et al. 2008).

Proteobacteria

YddA (M-C) of E. coli; 561 aas

 
3.A.1.203.12

ABC transporter, BclA, of 586 aas and 6 TMSs in a 2 + 2 + 2 arrangement in the N-terminus and the ABC domain in the C-terminus.  It is a peptide transprter required for bacteroid differentiation.  It catalyzes import of peptides called nodule-specific cysteine-rich (NCR) peptides in the symbiotic nodule cells which house the bacteroids. NCR peptides are related to antimicrobial peptides of innate immunity, but they induce the endosymbionts into a differentiated, enlarged, and polyploid state (Guefrachi et al. 2015). BclA is required for the formation of differentiated and functional bacteroids in the nodules of the NCR peptide-producing Aeschynomene legumes. BclA catalyzes import of NCR peptides and provides protection against the antimicrobial activity of these peptides. Moreover, BclA can complement the role of the related BacA transporter of Sinorhizobium meliloti, which has a similar symbiotic function in the interaction with Medicago legumes (Guefrachi et al. 2015).

BclA of Bradyrhizobium sp. ORS 285

 
3.A.1.203.13

Glycosomal ABC transporter of 683 aas and 6 N-terminal TMSs followed by the ATPase domain. Insertion into the glycosomal membrane is facilitated by the chaparone/receptor, Pex19 (Yernaux et al. 2006).

Glycosomal ABC half transporter of Trypanosoma brucei

 
3.A.1.203.14

Glycosomal ABC transporter of 641 aas and 6 N-terminal TMSs followed by the ATPase domain. Insertion into the glycosomal membrane is facilitated by the chaparone/receptor, Pex19 (Yernaux et al. 2006).

ABC half transporter of Trypanosoma brucei

 
3.A.1.203.15

ABCD1 transporter of 766 aas and 6 TMSs. Similar to human patients with X-linked adrenoleukodystrophy (ALD), zebrafish abcd1 mutants have elevated very long chain fatty acid levels, and CNS development was disrupted, with hypomyelination in the spinal cord, abnormal patterning, decreased numbers of oligodendrocytes, and impaired motor function followed by increased cell death (Strachan et al. 2017). Expression of human ABCD1 in zebrafish oligodendrocytes rescued apoptosis in the abcd1 mutant (Strachan et al. 2017).

ABCD1 of Danio rerio (Zebrafish) (Brachydanio rerio)

 
3.A.1.203.3

The peroxysomal long chain fatty acid (LCFA) half transporter, ABCD1 (ALD, ALDP, the X-linked adrenoleukodystrophy (X-ALD or ALDP) protein) (functions as a homodimer and accepts acyl-CoA esters (van Roermund et al. 2008)). It transports C24:0 and C26:0 fatty acids and their CoA-derivatives as substrates (van Roermund et al., 2011; Jia et al. 2022).  ABCD1 deficiency or mutation is associated with plasma and tissue elevation of C24:0 and C26:0 accompanied by demyelination and inflamation (Baarine et al. 2012).  X-ALD is a recessive neurodegenerative disorder that affects the brain's white matter and is associated with adrenal insufficiency. It is characterized by abnormal function of peroxisomes, which leads to an accumulation of very long-chain fatty acids (VLCFA) in plasma and tissues, especially in the cortex of the adrenal glands and the white matter of the central nervous system, causing demyelinating disease and adrenocortical insufficiency (Addison's disease or X-linked adrenoleukodystrophy (X-ALD) (Kallabi et al. 2013; Andreoletti et al. 2017) The system forms heterodimers with PMP70 (ABCD3; TC#3.A.1.203.1) (Hillebrand et al. 2007). X-ALD, the most common peroxisomal disorder, results from mutations in ABCD1 (ALDP) (Margoni et al. 2017). The structure and function of the ABCD1 variant database have been described (Mallack et al. 2022). This peroxisomal very long chain fatty acid (VLCFA) transporter is central to fatty acid catabolism and lipid biosynthesis. Its dysfunction underlies toxic cytosolic accumulation of VLCFAs, progressive demyelination, and neurological impairment including X-ALD. Le et al. 2022 presented cryo-EM structures of ABCD1 in phospholipid nanodiscs in a nucleotide bound conformation open to the peroxisomal lumen and an inward facing conformation open to the cytosol at up to 3.5 Å resolution, revealing details of its transmembrane cavity and ATP- dependent conformational spectrum. They identified features distinguishing ABCD1 from its closest homologs and showed that coenzyme A (CoA) esters of VLCFAs modulate ABCD1 activity in a species dependent manner. A transport mechanism was suggested in which the CoA moieties of VLCFA-CoAs enter the hydrophilic transmembrane domain while the acyl chains extend out into the surrounding membrane bilayer. The structures help rationalize disease causing mutations (Le et al. 2022). Three cryogenic EM structures of ABCD1: the apo-form, substrate- and ATP-bound forms have been solved (Chen et al. 2022). Distinct from what was seen in the previously reported ABC transporters, the two symmetric molecules of behenoyl coenzyme A (C22:0-CoA) cooperatively bind to the transmembrane domains (TMDs). For each C22:0-CoA, the hydrophilic 3'-phospho-ADP moiety of the CoA portion inserts into one TMD, with the succeeding pantothenate and cysteamine moiety crossing the inter-domain cavity, whereas the hydrophobic fatty acyl chain extends to the opposite TMD. Structural analysis combined with biochemical assays illustrated snapshots of the ABCD1-mediated substrate transport cycle (Chen et al. 2022). Jia et al. 2022 reported the cryo-EM structure of human ALDP at 3.4 Å resolution. ALDP exhibits a cytosolic-facing conformation. Compared to other lipid ATP-binding cassette transporters, ALDP has two substrate binding cavities formed within the transmembrane domains. Such structural organization may be suitable for the coordination of VLCFAs. X-ALD is caused by a mutation in the ABCD1 gene, encoding a peroxisomal protein, which has various clinical manifestations and a rapid progression from initial symptoms to fatal inflammatory demyelination (Yu et al. 2022). Structural insights into substrate recognition and translocation of human peroxisomal ABC transporter, ALDP, have appeared (Xiong et al. 2023). An innovative tree-based method for sampling molecular conformations allows prediction of conformations (Haschka et al. 2024).

Animals

LCFA transporter ABCD1 of Homo sapiens

 
3.A.1.203.4

The BacA (Rv1819c) porter (selective for the uptake of bleomycin and antimicrobial peptides) (essential for maintenance of extended chronic infection) (Domenech et al., 2009).

Actinobacteria

BacA of Mycobacterium tuberculosis (M-C) (Q50614)

 
3.A.1.203.5

Peroxisomal importer, Comatose, of substrates for β-oxidation (transports fatty acids and precursors 2,4-dichlorophenoxybutyric acid (2,4-DB) and indole butyric acid (IBA) (Dietrich et al., 2009; Visser et al. 2007). The peroxisomal fatty acyl-CoA transporter, Comatose (CTS, ABCD1, ABCD1, ABCC1, PED3, Pxa1; 1337aas) (Nyathi et al., 2010) determines germination potential and fertility and is essential for acetate metabolism (Linka and Esser 2012).  It associates with long chain fatty acyl-CoA synthetases (LACS6 (Q8LPS1) and LACS7 (Q8LKS5) and has intrinsic acyl CoA thiesterase activity (De Marcos Lousa et al. 2013). It has been proposed that it transports and hydrolyzes acyl-CoA esters, releasing a non-esterified fatty acid into the peroxisomal matrix which then needs to be re-activated by peroxisomal LACS6 or LACS7 (Visser et al. 2007). Mutagenesis of three residues in TMS 9 differentially affected the ATPase and thioesterase activities (Carrier et al. 2019).

Plants

Comatose of Arabidopsis thaliana (Q94FB9)

 
3.A.1.203.6