TCDB is operated by the Saier Lab Bioinformatics Group

2.A.1 The Major Facilitator Superfamily (MFS)

The MFS is a very old, large and diverse superfamily that includes millions of sequenced members. They catalyze uniport, solute:cation (H+, but seldom Na+) symport and/or solute:H+ or solute:solute antiport. Most are of 400-600 amino acyl residues in length and possess either 12, 14, or occasionally, 24 transmembrane α-helical spanners (TMSs). The mechanistic principles applicable to all MFS carriers have been summarized by Law et al (2008), while Zhang et al. 2015 considered the interaction between protonation and the negative-inside membrane potential. Functional roles of the conserved sequence motifs were also discussed in the context of the 3D structures. Transporters can be modified posttranslationally by phsophorylation, ubiquitination, glycosylation and/or palmitoylation (Czuba et al. 2018). The mammalian proteins have been analyzed from phylogenetic standpoints, revealing three major groups with differing conserved residues (Jia et al. 2019). Proteins of different topologies were also identified. Hu et al. 2020 reviewed the main substrates and functions of SLCs that are expressed in the brain, with an emphasis on selected SLCs that are important physiologically, pathologically, and pharmacologically in the blood-brain barrier, astrocytes, and neurons. The glucose (hexose) transporters, GLUTs, TC# 2.A.1.1, have been reviewed (Holman 2020; Ismail and Tanasova 2022). Single-molecule FRET studies of membrane transport proteins has revealed their conformational flexibilities (Löw et al. 2021). All MFS porters have the MFS fold (Ferrada and Superti-Furga 2022). Anagnostakis et al. 2023 discuss the key structural and functional characteristics of SLC family members involved in glioma pathogenesis, and active ion transmembrane transporter activities were overrepresented in patients with Alzheimer's disease (D'Angiolini et al. 2023). Similarly, Parkinson's disease involves activation of ion transporters via the Leucine Rich Repeat Kinase 2 (LRRK2) in miroglia of mice (Nazish et al. 2023).  Differentially expressed SLC transcripts in phagocytes (macrophages, dendritic cells, and neutrophils) have been identified, compared to adaptive immune cells; new potential immune cell markers based on SLC expressionhave been revealed; and user-friendly online tools for researchers to explore SLC genes of interesthave been created (Aaes et al. 2023). The motions of transport systems have been documented (Loland et al. 2023). 

.The 24 TMS MFS permease, NarK, of Paracoccus pantotrophus has two 12 TMS domains, NarK1 and NarK2, both of which are required for normal nitrate uptake. NarK1 catalyzes NO3-:H+ symport, dependent on the pmf, while NarK2 catalyzes NO3-:NO2- antiport, independently of the pmf (Wood et al., 2002). Thus, the protein is a fusion protein of two homologous but distinct MFS permeases.  Various mechanisms for importing sugars needed to create cellular homeostasis and survival in several kinds of cells have been reviewed (Carbó and Rodríguez 2023).

MFS permeases exhibit specificity for sugars, polyols, drugs, neurotransmitters, Krebs cycle metabolites, phosphorylated glycolytic intermediates, amino acids, peptides, osmolites, siderophores (efflux), iron-siderophores (uptake), nucleosides, organic anions, inorganic anions, etc. They are found ubiquitously in all three kingdoms of living organisms. Among the yeast MFS MDR transporters, the DHA2 family (TC 2.A.1.3) is more variable between species than the DHA1 family (TC 2.A.1.2) (Gbelska et al. 2006). Genome-wide analysis of 40 Major Facilitator Superfamily members and their expression in response of poplar to Fusarium oxysporum infection has been published (Diao et al. 2021). Nano-targeted delivery systems have been created to overcome the main drawbacks of conventional drug treatment, including insufficient stability and solubility, lack of transmembrane transport, short circulation time, and undesirable toxic effects. These systems have been reviewed (Cheng et al. 2023).  A new method allows modeling of multiple conformational states of SLC proteins generally (Swapna et al. 2024).

One member of the DHA2 family with 14 TMSs, the TetL Me2+· tetracycline:H+ antiporter of B. subtilis (TC #2.A.1.3.16), which also exhibits monovalent ion antiport activity, can be converted to a monovalent cation (Na+, K+, H+) antiporter with no tetracycline transport activity by deletion of TMSs 7 and 8, the two central and extra TMSs (Jin et al., 2001). Genome analyses of MFS permeases have been published (Lorca et al., 2007).

A 6.5 Å resolution structure for the MFS permease, OxlT (TC #2.A.1.11.1) was obtained in early studies (Heymann et al., 2001; Hirai et al., 2002) which shows the positions of the transmembrane α-helices but does not allow assignment of the TMS # to these helices. Molecular modeling (Hirai et al., 2003) led to the suggestion that the 12 TMS protein arose from a 3 TMS element by two successive duplication events. The same suggestion resulted from sequence comparisons showing that the primordial 3 TMS element may have arisen from a VIC family (TC #1.A.1) 2 TMS channel-forming unit Hvorup & Saier, (2002).This conclusion has been extensively confirmed in several more recent studies.

The high-resolution 3-dimensional structures (3.3 and 3.5 Å resolution) of the glycerol-3-P:P antiporter (GlpT; TC #2.A.1.4.3) and the lactose:H+ symporter (LacY; TC #2.A.1.5.1), respectively (Huang et al., 2003 and Abramson et al., 2003, respectively; see also Locher et al., 2003; Guan et al., 2007) have been determined. These structures reveal the 2-fold symmetry expected, based on sequence similarity of the two halves. The substrate pathway is predicted to exist between the two halves of the permeases using an alternating access mechanism with a single substrate binding site (Huang et al., 2003). This mechanism is termed a 'rocker switch' type of movement. Several putative mechanisms involving MFS and other types of transporters underly etridiazole (EDZ)-induced developmental deformities in Zebrafish (Vasamsetti et al. 2023).

As suggested above, MFS symporters and antiporters are believed to operate via a single binding site, alternating-access mechanism that involves a rocker-switch type movement of the two halves of the protein (Law et al., 2008). In the sn-glycerol-3-phosphate transporter (GlpT) from Escherichia coli, the substrate-binding site is formed by several charged residues and a histidine that can be protonated. Salt-bridge formation and breakage are involved in the conformational changes of the protein during transport (Law et al., 2008).

Vesicular glutamate transporters (VGLUTs [2.A.1.14.13, 2.A.1.14.16, etc.]) are responsible for the vesicular storage of L-glutamate and play an essential role in glutamatergic signal transmission in the central nervous system. VGLUT2 facilitates L-glutamate uptake in a membrane potential (ΔΨ)-dependent fashion. Uptake exhibited an absolute requirement for ~4 mM Cl- and was sensitive to Evans blue, but was insensitive to D,L-aspartate. VGLUT2s with mutations in the transmembrane-located residues Arg184, His128, and Glu191 showed a dramatic loss in L-glutamate transport activity. VGLUT2 appears to possess two intrinsic transport machineries that are independent of each other: a ΔΨ-dependent L-glutamate uptake and a Na+-dependent Pi uptake (Juge et al., 2006). VGLUTs (TC# 2.A.1.14) and EAATs (TC# 2.A.23.2) may be potential targets in the treatment of Parkinson's Disease (PD). VGLUTs and EAATs can be used as clinical drug targets to achieve better efficacy (Li et al. 2021).

Trichoderma spp. are avirulent, fungal, opportunistic plant symbionts present in nearly all climatic soils. These Trichoderma strains produce secondary metabolites that are potent bio-control agents against microbial pathogens and also can be plant growth promoters. The MFS includes a large proportion of efflux-pumps which are linked with membrane transport of these secondary metabolites in T. reesei (Chaudhary et al. 2016). Many of these export drugs such an anticancer agents and antibiotics.  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).

The generalized transport reactions catalyzed by MFS porters are:

(1) Uniport: S (out) ⇌ S (in)

(2) Symport: S (out) + [H+ (or Na+)] (out) ⇌ S (in) + [H+ (or Na+)] (in)

(3) Antiport: S1 (out) + S2 (in) ⇌ S1 (in) + S2 (out) (S1 may be H+ or a solute)

This family belongs to the: Major Facilitator (MFS) Superfamily.

References associated with 2.A.1 family:

Aaes, T.L., J. Burgoa Cardás, and K.S. Ravichandran. (2023). Defining solute carrier transporter signatures of murine immune cell subsets. Front Immunol 14: 1276196. 38077407
Abbas, A., J.E. McGuire, D. Crowley, C. Baysse, M. Dow, and F. O'Gara. (2004). The putative permease PhlE of Pseudomonas fluorescens F113 has a role in 2,4-diacetylphloroglucinol resistance and in general stress tolerance. Microbiology 150: 2443-2450. 15256586
Abdel-Motaal, H., L. Meng, Z. Zhang, A.H. Abdelazez, L. Shao, T. Xu, F. Meng, S. Abozaed, R. Zhang, and J. Jiang. (2018). An Uncharacterized Major Facilitator Superfamily Transporter From Exhibits Dual Functions as a Na(Li, K)/H Antiporter and a Multidrug Efflux Pump. Front Microbiol 9: 1601. 30061877
Abdulhussein AA. and Wallace HM. (2014). Polyamines and membrane transporters. Amino Acids. 46(3):655-60. 23851697
Abplanalp, J., E. Laczko, N.J. Philp, J. Neidhardt, J. Zuercher, P. Braun, D.F. Schorderet, F.L. Munier, F. Verrey, W. Berger, S.M. Camargo, and B. Kloeckener-Gruissem. (2013). The cataract and glucosuria associated monocarboxylate transporter MCT12 is a new creatine transporter. Hum Mol Genet 22: 3218-3226. 23578822
Abramson, J., I. Smirnova, V. Kasho, G. Verner, H.R. Kaback, and S. Iwata. (2003). Structure and mechanism of the lactose permease of Escherichia coli. Science 301: 610-615. 12893935
AbuAli, G. and S. Grimm. (2014). Isolation and Characterization of the Anticancer Gene Organic Cation Transporter Like-3 (ORCTL3). Adv Exp Med Biol 818: 213-227. 25001539
Adeva-Andany, M.M., I. Calvo-Castro, C. Fernández-Fernández, C. Donapetry-García, and A.M. Pedre-Piñeiro. (2017). Significance of l-carnitine for human health. IUBMB Life. [Epub: Ahead of Print] 28653367
Adler, J. and E. Bibi. (2004). Determinants of substrate recognition by the Escherichia coli multidrug transporter MdfA identified on both sides of the membrane. J. Biol. Chem. 279: 8957-8965. 14688269
Adler, J. and E. Bibi. (2005). Promiscuity in the geometry of electrostatic interactions between the Escherichia coli multidrug resistance transporter MdfA and cationic substrates. J. Biol. Chem. 280: 2721-2729. 15557318
Agbani, E.O., L. Skeith, and A. Lee. (2023). Preeclampsia: Platelet procoagulant membrane dynamics and critical biomarkers. Res Pract Thromb Haemost 7: 100075. 36923708
Ahmadi, F., S.N. Akmar Abdullah, S. Kadkhodaei, S.M. Ijab, L. Hamzah, M.A. Aziz, Z.A. Rahman, and S.S. Rabiah Syed Alwee. (2018). Functional characterization of the gene promoter for an Elaeis guineensis phosphate starvation-inducible, high affinity phosphate transporter in both homologous and heterologous model systems. Plant Physiol. Biochem 127: 320-335. [Epub: Ahead of Print] 29653435
Ai, P., S. Sun, J. Zhao, X. Fan, W. Xin, Q. Guo, L. Yu, Q. Shen, P. Wu, A.J. Miller, and G. Xu. (2009). Two rice phosphate transporters, OsPht1;2 and OsPht1;6, have different functions and kinetic properties in uptake and translocation. Plant J. 57: 798-809. 18980647
Al-Azab, M., E. Idiiatullina, Z. Liu, M. Lin, K. Hrovat-Schaale, H. Xian, J. Zhu, M. Yang, B. Lu, Z. Zhao, Y. Liu, J. Chang, X. Li, C. Guo, Y. Liu, Q. Wu, J. Chen, C. Lan, P. Zeng, J. Cui, X. Gao, W. Zhou, Y. Zhang, Y. Zhang, and S.L. Masters. (2024). Genetic variants in UNC93B1 predispose to childhood-onset systemic lupus erythematosus. Nat Immunol 25: 969-980. 38831104
Albrecht, S., W. Hartmann, F. Houshdaran, A. Koch, B. Gärtner, D. Prawitt, B.U. Zabel, P. Russo, D. Von Schweinitz, and T. Pietsch. (2004). Allelic loss but absence of mutations in the polyspecific transporter gene BWR1A on 11p15.5 in hepatoblastoma. Int J Cancer 111: 627-632. 15239143
Aldahmesh, M.A., A.O. Khan, J.Y. Mohamed, H. Hijazi, M. Al-Owain, A. Alswaid, and F.S. Alkuraya. (2012). Genomic analysis of pediatric cataract in Saudi Arabia reveals novel candidate disease genes. Genet Med 14: 955-962. 22935719
Alexander, N.J., S.P. McCormick, and T.M. Hohn. (1999). TRI12, a trichothecene efflux pump from Fusarium sporotrichioides: gene isolation and expression in yeast. Mol. Gen. Genet. 261: 977-984. 10485289
Alfonso, A., K. Grundahl, J.R. McManus, J.M. Asbury, and J.B. Rand. (1994). Alternative splicing leads to two cholinergic proteins in Caenorhabditis elegans. J. Mol. Biol. 241: 627-630. 8057385
Ali, R.S., M.F. Dick, S. Muhammad, D. Sarver, L. Hou, G.W. Wong, and K.C. Welch, Jr. (2020). Glucose transporter expression and regulation following a fast in the ruby-throated hummingbird,. J Exp Biol 223:. 32895327
Allard, K.A., V. K. Viswanathan, and N.P. Cianciotto. (2006). lbtA and lbtB Are Required for Production of the Legionella pneumophila Siderophore Legiobactin. J. Bacteriol. 188: 1351-1363. 16452417
Almagro, G., A.M. Viale, M. Montero, F.J. Muñoz, E. Baroja-Fernández, H. Mori, and J. Pozueta-Romero. (2018). A cAMP/CRP-controlled mechanism for the incorporation of extracellular ADP-glucose in Escherichia coli involving NupC and NupG nucleoside transporters. Sci Rep 8: 15509. 30341391
Almaguer, C., E. Fisher, and J. Patton-Vogt. (2006). Posttranscriptional regulation of Git1p, the glycerophosphoinositol/glycerophosphocholine transporter of Saccharomyces cerevisiae. Curr. Genet. 50: 367-375. 16924500
Alomari, D.Z., A.M. Alqudah, K. Pillen, N. von Wirén, and M.S. Röder. (2021). A Major Facilitator Superfamily Transporter is a putative candidate gene for nutrient mineral (Ca, K, Mg, Mn, P and S) accumulation in bread wheat grains. J Exp Bot. [Epub: Ahead of Print] 34145452
Aluri, S. and M. Büttner. (2007). Identification and functional expression of the Arabidopsis thaliana vacuolar glucose transporter 1 and its role in seed germination and flowering. Proc. Natl. Acad. Sci. U.S.A. 104: 2537-2542. 17284600
Aluri, S., R. Zhao, A. Fiser, and I.D. Goldman. (2017). Residues in the eighth transmembrane domain of the proton-coupled folate transporter (SLC46A1) play an important role in defining the aqueous translocation pathway and in folate substrate binding. Biochim. Biophys. Acta. [Epub: Ahead of Print] 28802835
Aluri, S., R. Zhao, C. Lubout, S.M.I. Goorden, A. Fiser, and I.D. Goldman. (2018). Hereditary folate malabsorption due to a mutation in the external gate of the proton-coupled folate transporter SLC46A1. Blood Adv 2: 61-68. 29344585
Aluri, S., R. Zhao, K. Lin, D.S. Shin, A. Fiser, and I.D. Goldman. (2019). Substitutions that lock and unlock the proton-coupled folate transporter (PCFT-SLC46A1) in an inward-open conformation. J. Biol. Chem. [Epub: Ahead of Print] 30858177
Alvarez, F.J., and J.B. Konopka. (2007). Identification of an N-acetylglucosamine transporter that mediates hyphal induction in Candida albicans. Mol. Biol. Cell. 18: 965-975. 17192409
Alves, R., M. Sousa-Silva, D. Vieira, P. Soares, Y. Chebaro, M.C. Lorenz, M. Casal, I. Soares-Silva, and S. Paiva. (2020). Carboxylic Acid Transporters in Pathogenesis. mBio 11:. 32398310
Alves, R., S. Mota, S. Silva, C. F Rodrigues, A.J. P Brown, M. Henriques, M. Casal, and S. Paiva. (2017). The carboxylic acid transporters Jen1 and Jen2 affect the architecture and fluconazole susceptibility of Candida albicans biofilm in the presence of lactate. Biofouling 33: 943-954. 29094611
Amahong, K., M. Yan, J. Li, N. Yang, H. Liu, X. Bi, D.A. Vuitton, R. Lin, and G. Lü. (2021). EgGLUT1 Is Crucial for the Viability of Metacestode: A New Therapeutic Target? Front Cell Infect Microbiol 11: 747739. 34858873
Ambrose, K.D., R. Nisbet, and D.S. Stephens. (2005). Macrolide efflux in Streptococcus pneumoniae is mediated by a dual efflux pump (mel and mef) and is erythromycin inducible. Antimicrob. Agents Chemother. 49: 4203-4209. 16189099
Amilhon, B., E. Lepicard, T. Renoir, R. Mongeau, D. Popa, O. Poirel, S. Miot, C. Gras, A.M. Gardier, J. Gallego, M. Hamon, L. Lanfumey, B. Gasnier, B. Giros, and S. El Mestikawy. (2010). VGLUT3 (vesicular glutamate transporter type 3) contribution to the regulation of serotonergic transmission and anxiety. J. Neurosci. 30: 2198-2210. 20147547
Amorese, A.J., E.C. Minchew, M.D. Tarpey, A.T. Readyoff, N.C. Williamson, C.A. Schmidt, S.L. McMillin, E.J. Goldberg, Z.S. Terwilliger, Q.A. Spangenburg, C.A. Witczak, J.J. Brault, E.D. Abel, J.M. McClung, K.H. Fisher-Wellman, and E.E. Spangenburg. (2023). Hypoxia Resistance Is an Inherent Phenotype of the Mouse Flexor Digitorum Brevis Skeletal Muscle. Function (Oxf) 4: zqad012. 37168496
Anagnostakis, F., M. Kokkorakis, M. Markouli, and C. Piperi. (2023). Impact of Solute Carrier Transporters in Glioma Pathology: A Comprehensive Review. Int J Mol Sci 24:. 37298344
Anderson, P.M., Y.C. Sung, and J.A. Fuchs. (1990). The cyanase operon and cyanate metabolism. FEMS Microbiol. Rev. 7: 247-252. 2094285
Anzai, N. and H. Endou. (2011). Urate transporters: an evolving field. Semin Nephrol 31: 400-409. 22000646
Anzai, N., H. Miyazaki, R. Noshiro, S. Khamdang, A. Chairoungdua, J.-J. Shin, A. Enomoto, S. Sakamoto, T. Hirata, K. Tomita, Y. Kanai, and H. Endou. (2004). The multivalent PDZ domain-containing protein PDZK1 regulates transport activity of renal urate-anion exchanger URAT1 via its C terminus. J. Biol. Chem. 279: 45942-45950. 15304510
Anzai, N., K. Ichida, P. Jutabha, T. Kimura, E. Babu, C.J. Jin, S. Srivastava, K. Kitamura, I. Hisatome, H. Endou, and H. Sakurai. (2008). Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. J. Biol. Chem. 283: 26834-26838. 18701466
AONO, K. (1962). [Studies on protein in gallstone]. Fukuoka Igaku Zasshi 53: 897-926. 14013424
Aouameur, R., S. Da Cal, P. Bissonnette, M.J. Coady, and J.Y. Lapointe. (2007). SMIT2 mediates all myo-inositol uptake in apical membranes of rat small intestine. Am. J. Physiol. Gastrointest. Liver. Physiol. 293(6):G1300-G1307.
Aouida, M., R. Poulin, and D. Ramotar. (2010). The human carnitine transporter SLC22A16 mediates high affinity uptake of the anticancer polyamine analogue bleomycin-A5. J. Biol. Chem. 285: 6275-6284. 20037140
Araki N., Suzuki T., Miyauchi K., Kasai D., Masai E. and Fukuda M. (201). Identification and characterization of uptake systems for glucose and fructose in Rhodococcus jostii RHA1. J Mol Microbiol Biotechnol. 20(3):125-36. 21464575
Arjona, F.J., E. de Vrieze, T.J. Visser, G. Flik, and P.H. Klaren. (2011). Identification and functional characterization of zebrafish solute carrier Slc16a2 (Mct8) as a thyroid hormone membrane transporter. Endocrinology 152: 5065-5073. 21952246
Arponen, O., P. Wodtke, F.A. Gallagher, and R. Woitek. (2023). Hyperpolarised C-MRI using C-pyruvate in breast cancer: A review. Eur J Radiol 167: 111058. [Epub: Ahead of Print] 37666071
Babu, E., Y. Kanai, A. Chairoungdua, D.K. Kim, Y. Iribe, S. Tangtrongsup, P. Jutabha, Y. Li, N. Ahmed, S. Sakamoto, N. Anzai, S. Nagamori, and H. Endou. (2003). Identification of a novel system L amino acid transporter structurally distinct from heterodimeric amino acid transporters. J. Biol. Chem. 278: 43838-43845. 12930836
Bacher, P., S. Giersiefer, M. Bach, C. Fork, E. Schömig, and D. Gründemann. (2009). Substrate discrimination by ergothioneine transporter SLC22A4 and carnitine transporter SLC22A5: gain-of-function by interchange of selected amino acids. Biochim. Biophys. Acta. 1788: 2594-2602. 19814996
Bagchi, S., E. Perland, K. Hosseini, J. Lundgren, N. Al-Walai, S. Kheder, and R. Fredriksson. (2020). Probable role for major facilitator superfamily domain containing 6 (MFSD6) in the brain during variable energy consumption. Int J. Neurosci. 130: 476-489. 31906755
Bahn, A., Y. Hagos, S. Reuter, D. Balen, H. Brzica, W. Krick, B.C. Burckhardt, I. Sabolic, and G. Burckhardt. (2008). Identification of a New Urate and High Affinity Nicotinate Transporter, hOAT10 (SLC22A13). J. Biol. Chem. 283: 16332-16341. 18411268
Bahrenberg, T., E.H. Yardeni, A. Feintuch, E. Bibi, and D. Goldfarb. (2021). Substrate binding in the multidrug transporter MdfA in detergent solution and in lipid nanodiscs. Biophys. J. [Epub: Ahead of Print] 33771471
Bai J., Mosley L. and Fralick JA. (2010). Evidence that the C-terminus of OprM is involved in the assembly of the VceAB-OprM efflux pump. FEBS Lett. 584(8):1493-7. 20206171
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. 38823222
Bailey, T.L., A. Nieto, and P.H. McDonald. (2019). A Nonradioactive High-Throughput Screening-Compatible Cell-Based Assay to Identify Inhibitors of the Monocarboxylate Transporter Protein 1. Assay Drug Dev Technol 17: 275-284. 31532712
Bajraktari-Sylejmani, G., C. Bay, L. Gebauer, J. Burhenne, J. Weiss, and M. Sauter. (2024). A Highly Sensitive UPLC-MS/MS Method for the Quantification of the Organic Cation Transporters'' Mediated Metformin Uptake and Its Inhibition in Cells. Molecules 29:. 39519803
Baker, J., S.H. Wright, and F. Tama. (2012). Simulations of substrate transport in the multidrug transporter EmrD. Proteins 80: 1620-1632. 22434745
Baldwin, S.A. (1993). Mammalian passive glucose transporters: members of an ubiquitous family of active and passive transport proteins. Biochim. Biophys. Acta 1154: 17-49. 8507645
Balmaceda-Aguilera C., Martos-Sitcha JA., Mancera JM. and Martinez-Rodriguez G. (2012). Cloning and expression pattern of facilitative glucose transporter 1 (GLUT1) in gilthead sea bream Sparus aurata in response to salinity acclimation. Comp Biochem Physiol A Mol Integr Physiol. 163(1):38-46. 22580053
Baltazar, F., C. Pinheiro, F. Morais-Santos, J. Azevedo-Silva, O. Queirós, A. Preto, and M. Casal. (2014). Monocarboxylate transporters as targets and mediators in cancer therapy response. Histol Histopathol 29: 1511-1524. 24921258
Bannam, T.L., P.A. Johanesen, C.L. Salvado, S.J. Pidot, K.A. Farrow, and J.I. Rood. (2004). The Clostridium perfringens TetA(P) efflux protein contains a functional variant of the Motif A region found in major facilitator superfamily transport proteins. Microbiology 150: 127-134. 14702405
Bansal, A., D. Mallik, D. Kar, and A.S. Ghosh. (2016). Identification of a multidrug efflux pump in Mycobacterium smegmatis. FEMS Microbiol. Lett. [Epub: Ahead of Print] 27190152
Bapna, A., L. Federici, H. Venter, S. Velamakanni, B. Luisi, T.P. Fan, and H.W. van Veen. (2007). Two proton translocation pathways in a secondary active multidrug transporter. J. Mol. Microbiol. Biotechnol. 12: 197-209. 17587868
Baranova, N. and H. Nikaido. (2002). The baeSR two-component regulatory system activates transcription of the yegMNOB (mdtABCD) transporter gene cluster in Escherichia coli and increases its resistance to novobiocin and deoxycholate. J. Bacteriol. 184: 4168-4176. 12107134
Bardaweel, S. and A. Issa. (2022). Exploring the Role of Sodium-Glucose Cotransporter as a New Target for Cancer Therapy. J Pharm Pharm Sci 25: 253-265. 35977549
Barton, A., S. Eyre, J. Bowes, P. Ho, S. John, and J. Worthington. (2005). Investigation of the SLC22A4 gene (associated with rheumatoid arthritis in a Japanese population) in a United Kingdom population of rheumatoid arthritis patients. Arthritis Rheum 52: 752-758. 15751072
Baruffini, E., P. Goffrini, C. Donnini, and T. Lodi. (2006). Galactose transport in Kluyveromyces lactis: major role of the glucose permease Hgt1. FEMS Yeast Research DOI: 10.1111/j.1364.2006.00107.x. 17156020
Basak, D., D. Gamez, and S. Deb. (2023). SGLT2 Inhibitors as Potential Anticancer Agents. Biomedicines 11:. 37509506
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. 20348384
Batool, H., B. Zubaida, M.A. Hashmi, and M. Naeem. (2019). Genetic testing of two Pakistani patients affected with rare autosomal recessive Fanconi-Bickel syndrome and identification of a novel SLC2A2 splice site variant. J Pediatr Endocrinol Metab. [Epub: Ahead of Print] 31473689
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. 19400778
Beaudoin, J., R. Ioannoni, L. López-Maury, J. Bähler, S. Ait-Mohand, B. Guérin, S.C. Dodani, C.J. Chang, and S. Labbé. (2011). Mfc1 is a novel forespore membrane copper transporter in meiotic and sporulating cells. J. Biol. Chem. 286: 34356-34372. 21828039
Becker, H.M. and J.W. Deitmer. (2008). Nonenzymatic proton handling by carbonic anhydrase II during H+-lactate cotransport via monocarboxylate transporter 1. J. Biol. Chem. 283: 21655-21667. 18539591
Becker, H.M., D. Hirnet, C. Fecher-Trost, D. Sultemeyer, and J.W. Deitmer. (2005). Transport activity of MCT1 expressed in Xenopus oocytes is increased by interaction with carbonic anhydrase. J. Biol. Chem. 280: 39882-39889. 16174776
Becker, H.M., M. Klier, and J.W. Deitmer. (2010). Nonenzymatic augmentation of lactate transport via monocarboxylate transporter isoform 4 by carbonic anhydrase II. J. Membr. Biol. 234: 125-135. 20300744
Becker, P.C., M. Güth-Steffens, K. Lazarow, N. Sonntag, D. Braun, I. Masfaka, K. Renko, L. Schomburg, J. Köhrle, J.P. von Kries, U. Schweizer, G. Krause, and J. Protze. (2024). Identification of Human TRIAC Transmembrane Transporters. Thyroid 34: 920-930. 38801167
Beckham, K.S., J.A. Potter, and S.E. Unkles. (2010). Formate-nitrite transporters: optimisation of expression, purification and analysis of prokaryotic and eukaryotic representatives. Protein Expr Purif 71: 184-189. 20006708
Beenken, A., T. Shen, G. Jin, A. Ghotra, K. Xu, K. Nesanir, R.E. Sturley, S. Vijayakumar, A. Kahn, A. Levitman, J. Stauber, E.Y. Chavez, S.Y. Robbins-Juarez, L. Hao, T.B. Field, H. Erdjument-Bromage, T.A. Neubert, L. Shapiro, A. Qiu, and J. Barasch. (2024). Spns1 is an iron transporter essential for megalin-dependent endocytosis. Am. J. Physiol. Renal Physiol. [Epub: Ahead of Print] 39265081
Behr, S., L. Fried, and K. Jung. (2014). Identification of a novel nutrient-sensing histidine kinase/response regulator network in Escherichia coli. J. Bacteriol. 196: 2023-2029. 24659770
Bellocchio, E.E., R.J. Reimer, R.T. Fremeau, Jr., and R.H. Edwards. (2000). Uptake of glutamate into synaptic vesicles by an inorganic phosphate transporter. Science 289: 957-960. 10938000
Benko, Z., C. Fenyvesvolgyi, M. Pesti, and M. Sipiczki. (2004). The transcription factor Pap1/Caf3 plays a central role in the determination of caffeine resistance in Schizosaccharomyces pombe. Mol. Genet. Genomics 271: 161-170. 14758541
Bennett, K.M., J. Liu, C. Hoelting, and J. Stoll. (2011). Expression and analysis of two novel rat organic cation transporter homologs, SLC22A17 and SLC22A23. Mol. Cell Biochem 352: 143-154. 21359964
Berger, C. and D. Zdzieblo. (2020). Glucose transporters in pancreatic islets. Pflugers Arch. [Epub: Ahead of Print] 32394191
Bernal J., Guadano-Ferraz A. and Morte B. (2015). Thyroid hormone transporters--functions and clinical implications. Nat Rev Endocrinol. 11(7):406-17. 25942657
Besse A., Peduzzi J., Rebuffat S. and Carre-Mlouka A. (2015). Antimicrobial peptides and proteins in the face of extremes: Lessons from archaeocins. Biochimie. 118:344-55. 26092421
Beuming, T. and H. Weinstein. (2005). Modeling membrane proteins based on low-resolution electron microscopy maps: a template for the TM domains of the oxalate transporter OxlT. Protein Eng Des Sel 18: 119-125. 15820982
Beylerli, O., G. Sufianova, A. Shumadalova, D. Zhang, and I. Gareev. (2022). MicroRNAs-mediated regulation of glucose transporter (GLUT) expression in glioblastoma. Noncoding RNA Res 7: 205-211. 36157351
Bhaskar, B.V., T.M. Babu, N.V. Reddy, and W. Rajendra. (2016). Homology modeling, molecular dynamics, and virtual screening of NorA efflux pump inhibitors of Staphylococcus aureus. Drug Des Devel Ther 10: 3237-3252. 27757014
Bianco, M.V., F.C. Blanco, B. Imperiale, M.A. Forrellad, R.V. Rocha, L.I. Klepp, A.A. Cataldi, N. Morcillo, and F. Bigi. (2011). Role of P27 -P55 operon from Mycobacterium tuberculosis in the resistance to toxic compounds. BMC Infect Dis 11: 195. 21762531
Bleuel, C., Grosse, C., Taudte, N., Scherer, J., Wesenberg, D., Krauss, G.J., Nies, D.H., and Grass, G. (2005). TolC is involved in enterobactin efflux across the outer membrane of Escherichia coli. J. Bacteriol. 187: 6701-6707. 16166532
Bley, C., M. van der Linden, and R.R. Reinert. (2011). mef(A) is the predominant macrolide resistance determinant in Streptococcus pneumoniae and Streptococcus pyogenes in Germany. Int J Antimicrob Agents 37: 425-431. 21419605
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. 24979062
Boccone, L., S. Mariotti, V. Dessì, D. Pruna, A. Meloni, and G. Loudianos. (2010). Allan-Herndon-Dudley syndrome (AHDS) caused by a novel SLC16A2 gene mutation showing severe neurologic features and unexpectedly low TRH-stimulated serum TSH. Eur J Med Genet 53: 392-395. 20713192
Bodoy, S., L. Martin, A. Zorzano, M. Palacín, R. Estévez, and J. Bertran. (2005). Identification of LAT4, a novel amino acid transporter with system L activity. J. Biol. Chem. 280: 12002-12011. 15659399
Bohn, C. and P. Bouloc. (1998). The Escherichia coli cmlA gene encodes the multidrug efflux pump Cmr/MdfA and is responsible for isopropyl-β-D-thiogalactopyranoside exclusion and spectinomycin sensitivity. J. Bacteriol. 180: 6072-6075. 9811673
Boles, E. and C.P. Hollenberg. (1997). The molecular genetics of hexose transport in yeasts. FEMS Microbiol. Rev. 21: 85-111. 9299703
Boonyakanog, A., N. Charoenlap, S. Chattrakarn, P. Vattanaviboon, and S. Mongkolsuk. (2022). Contribution of Stenotrophomonas maltophilia MfsC transporter to protection against diamide and the regulation of its expression by the diamide responsive repressor DitR. PLoS One 17: e0272388. 35913917
Boorer, K.J., D.D. Loo, and E.M. Wright. (1994). Steady-state and presteady-state kinetics of the H+/hexose cotransporter (STP1) from Arabidopsis thaliana expressed in Xenopus oocytes. J. Biol. Chem. 269: 20417-20424. 8051137
Boot, A., J. Oosting, S. Doorn, S. Ouahoud, M. Ventayol Garcia, D. Ruano, H. Morreau, and T. van Wezel. (2019). Allelic Switching of , , and during Colorectal Cancer Tumorigenesis. Int J Genomics 2019: 1287671. 31093489
Bossuyt, X., M. Müller, B. Hagenbuch, and P.J. Meier. (1996). Polyspecific drug and steroid clearance by an organic anion transporter of mammalian liver. J. Pharmacol. Exper. Therapeutics 276: 891-896. 8786566
Bost, S., F. Silva, and D. Belin. (1999). Transcriptional activation of ydeA, which encodes a member of the major facilitator superfamily, interferes with arabinose accumulation and induction of the Escherichia coli arabinose PBAD promoter. J. Bacteriol. 181: 2185-2191. 10094697
Bozdag, G.O., I. Uluisik, G.S. Gulculer, H.C. Karakaya, and A. Koc. (2011). Roles of ATR1 paralogs YMR279c and YOR378w in boron stress tolerance. Biochem. Biophys. Res. Commun. 409: 748-751. 21621519
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. 28348231
Braibant, M., J. Chevalier, E. Chaslus-Dancla, J.M. Pagès, and A. Cloeckaert. (2005). Structural and functional study of the phenicol-specific efflux pump FloR belonging to the major facilitator superfamily. Antimicrob. Agents Chemother. 49: 2965-2971. 15980376
Brandenstein, L., M. Schweizer, J. Sedlacik, J. Fiehler, and S. Storch. (2015). Lysosomal dysfunction and impaired autophagy in a novel mouse model deficient for the lysosomal membrane protein Cln7. Hum Mol Genet. [Epub: Ahead of Print] 26681805
Brickman, T.J. and S.K. Armstrong. (2005). Bordetella AlcS transporter functions in alcaligin siderophore export and is central to inducer sensing in positive regulation of alcaligin system gene expression. J. Bacteriol. 187: 3650-3661. 15901687
Brignone, M.S., A. Lanciotti, S. Camerini, C. De Nuccio, T.C. Petrucci, S. Visentin, and E. Ambrosini. (2015). MLC1 protein: a likely link between leukodystrophies and brain channelopathies. Front Cell Neurosci 9: 66. 25883547
Brouns, I., I. Pintelon, J. Van Genechten, I. De Proost, J.P. Timmermans, and D. Adriaensen. (2004). Vesicular glutamate transporter 2 is expressed in different nerve fibre populations that selectively contact pulmonary neuroepithelial bodies. Histochem Cell Biol 121: 1-12. 14677070
Brown, E., S.P. Rajeev, D.J. Cuthbertson, and J.P.H. Wilding. (2019). A review of the mechanism of action, metabolic profile and haemodynamic effects of sodium-glucose co-transporter-2 inhibitors. Diabetes Obes Metab 21Suppl2: 9-18. 31081592
Brown, M.G., E.H. Mitchell, and D.L. Balkwill. (2008). Tet 42, a novel tetracycline resistance determinant isolated from deep terrestrial subsurface bacteria. Antimicrob. Agents Chemother. 52: 4518-4521. 18809935
Bukhari, F.J., H. Moradi, P. Gollapudi, H. Ju Kim, N.D. Vaziri, and H.M. Said. (2011). Effect of chronic kidney disease on the expression of thiamin and folic acid transporters. Nephrol Dial Transplant 26: 2137-2144. 21149507
Buyse, M., S.V. Sitaraman, X. Liu, A. Bado, and D. Merlin. (2002). Luminal leptin enhances CD147/MCT-1-mediated uptake of butyrate in the human intestinal cell line Caco2-BBE. J. Biol. Chem. 277: 28182-28190. 12034734
Cabedo Martinez, A.I., K. Weinhäupl, W.K. Lee, N.A. Wolff, B. Storch, S. Żerko, R. Konrat, W. Koźmiński, K. Breuker, F. Thévenod, and N. Coudevylle. (2016). Biochemical and Structural Characterization of the Interaction between the Siderocalin NGAL/LCN2 (Neutrophil Gelatinase-associated Lipocalin/Lipocalin 2) and the N-terminal Domain of Its Endocytic Receptor SLC22A17. J. Biol. Chem. 291: 2917-2930. 26635366
Cabrito, T.R., M.C. Teixeira, A.A. Duarte, P. Duque, and I. Sá-Correia. (2009). Heterologous expression of a Tpo1 homolog from Arabidopsis thaliana confers resistance to the herbicide 2,4-D and other chemical stresses in yeast. Appl. Microbiol. Biotechnol. 84: 927-936. 19440702
Cai, H., M. Hauser, F. Naider, and J.M. Becker. (2007). Differential regulation and substrate preferences in two peptide transporters of Saccharomyces cerevisiae. Eukaryot. Cell. 6: 1805-1813. 17693598
Calabrese, D., J. Bille, and D. Sanglard. (2000). A novel multidrug efflux transporter gene of the major facilitator superfamily from Candida albicans (FLU1) conferring resistance to fluconazole. Microbiology (Reading) 146(Pt11): 2743-2754. 11065353
Calderon-Rivera, A., S. Loya-Lopez, K. Gomez, and R. Khanna. (2022). Plant and fungi derived analgesic natural products targeting voltage-gated sodium and calcium channels. Channels (Austin) 16: 198-215. 36017978
Callewaert, B.L., A. Willaert, W.S. Kerstjens-Frederikse, J. De Backer, K. Devriendt, B. Albrecht, M.A. Ramos-Arroyo, M. Doco-Fenzy, R.C. Hennekam, R.E. Pyeritz, O.N. Krogmann, G. Gillessen-kaesbach, E.L. Wakeling, S. Nik-zainal, C. Francannet, P. Mauran, C. Booth, M. Barrow, R. Dekens, B.L. Loeys, P.J. Coucke, and A.M. De Paepe. (2008). Arterial tortuosity syndrome: clinical and molecular findings in 12 newly identified families. Hum Mutat 29(1): 150-158. 17935213
Campbell, C.L., C.J. Lehmann, S.S. Gill, W.A. Dunn, A.A. James, and B.D. Foy. (2011). A role for endosomal proteins in alphavirus dissemination in mosquitoes. Insect Mol Biol 20: 429-436. 21496127
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. 9580031
Cantón, R., A. Mazzariol, M.I. Morosini, F. Baquero, and G. Cornaglia. (2005). Telithromycin activity is reduced by efflux in Streptococcus pyogenes. J Antimicrob Chemother 55: 489-495. 15705635
Cao, C., E.K. Pressman, E.M. Cooper, R. Guillet, M. Westerman, and K.O. O'Brien. (2014). Placental heme receptor LRP1 correlates with the heme exporter FLVCR1 and neonatal iron status. Reproduction 148: 295-302. 24947444
Carayannopoulos, M.O., A. Schlein, A. Wyman, M. Chi, C. Keembiyehetty, and K.H. Moley. (2004). GLUT9 is differentially expressed and targeted in the preimplantation embryo. Endocrinology 145: 1435-1443. 14657010
Carayannopoulos, M.O., M.M. Chi, Y. Cui, J.M. Pingsterhaus, R.A. McKnight, M. Mueckler, S.U. Devaskar, and K.H. Moley. (2000). GLUT8 is a glucose transporter responsible for insulin-stimulated glucose uptake in the blastocyst. Proc. Natl. Acad. Sci. USA 97: 7313-7318. 10860996
Carbó, R. and E. Rodríguez. (2023). Relevance of Sugar Transport across the Cell Membrane. Int J Mol Sci 24:. 37047055
Carolé, S., S. Pichoff, and J.P. Bouché. (1999). Escherichia coli gene ydeA encodes a major facilitator pump which exports L-arabinose and isopropyl-β-D-thiogalactopyranoside. J. Bacteriol. 181: 5123-5125. 10438792
Carter, E.L., L. Jager, L. Gardner, C.C. Hall, S. Willis, and J.M. Green. (2007). Escherichia coli abg genes enable uptake and cleavage of the folate catabolite p-aminobenzoyl-glutamate. J. Bacteriol. 189: 3329-3334. 17307853
Cartier, E.A., L.A. Parra, T.B. Baust, M. Quiroz, G. Salazar, V. Faundez, L. Egaña, and G.E. Torres. (2010). A biochemical and functional protein complex involving dopamine synthesis and transport into synaptic vesicles. J. Biol. Chem. 285: 1957-1966. 19903816
Cecchetto, G., S. Amillis, G. Diallinas, C. Scazzocchio, and C. Drevet. (2004). The AzgA purine transporter of Aspergillus nidulans: characterization of a protein belonging to a new phylogenetic cluster. J. Biol. Chem. 279: 3132-3141. 14597637
Cesareo, R., M. Iozzino, D. Alva, C. Napolitano, B. De Rosa, S. Contini, L. Mallardo, A. Lauria, G. Reda, and A. Orsini. (2007). Evidence based medicine and effective interventions of pharmacological therapy for the prevention of osteoporotic fractures. Minerva Endocrinol 32: 275-295. 18091664
Chahboune, A., M. Decaffmeyer, R. Brasseur, and B. Joris. (2005). Membrane topology of the Escherichia coli AmpG permease required for recycling of cell wall anhydromuropeptides and AmpC β-lactamase induction. Antimicrob. Agents Chemother. 49: 1145-1149. 15728916
Chahine, S. and M.J. O'Donnell. (2010). Effects of acute or chronic exposure to dietary organic anions on secretion of methotrexate and salicylate by Malpighian tubules of Drosophila melanogaster larvae. Arch Insect Biochem Physiol 73: 128-147. 20077573
Chahine, S., A. Campos, and M.J. O''Donnell. (2012). Genetic knockdown of a single organic anion transporter alters the expression of functionally related genes in Malpighian tubules of Drosophila melanogaster. J Exp Biol 215: 2601-2610. 22786636
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. 22972675
Chaillou, S., Y.C. Bor, C.A. Batt, P.W. Postma, and P.H. Pouwels. (1998). Molecular cloning and functional expression in Lactobacillus plantarum 80 of xylT, encoding the D-xylose-H+ symporter of Lactobacillus brevis. Appl. Environ. Microbiol. 64: 4720-4728. 9835554
Chamarthy, S. and J.R. Mekala. (2023). Functional importance of glucose transporters and chromatin epigenetic factors in Glioblastoma Multiforme (GBM): possible therapeutics. Metab Brain Dis 38: 1441-1469. 37093461
Chang, H.-K. and G.J. Zylstra. (1999). Characterization of the phthalate permease OphD from Burkholderia cepacia ATCC 17616. J. Bacteriol. 181: 6197-6199. 10498738
Chang, J.C., R.V. Wilkening, K.M. Rahbari, and M.J. Federle. (2022). Quorum Sensing Regulation of a Major Facilitator Superfamily Transporter Affects Multiple Streptococcal Virulence Factors. J. Bacteriol. e0017622. [Epub: Ahead of Print] 35938850
Chang, R., J. Eriksen, and R.H. Edwards. (2018). The dual role of chloride in synaptic vesicle glutamate transport. Elife 7:. 30040066
Chang, Y.C., M.J. Tsai, Y.W. Huang, T.C. Chung, and T.C. Yang. (2011). SmQnrR, a DeoR-type transcriptional regulator, negatively regulates the expression of Smqnr and SmtcrA in Stenotrophomonas maltophilia. J Antimicrob Chemother 66: 1024-1028. 21393212
Chardwiriyapreecha, S., M. Shimazu, T. Morita, T. Sekito, K. Akiyama, K. Takegawa, and Y. Kakinuma. (2008). Identification of the fnx1(+) and fnx2(+) genes for vacuolar amino acid transporters in Schizosaccharomyces pombe. FEBS Lett. 582: 2225-2230. 18503766
Chasseigneaux, S., V. Cochois-Guégan, L. Lecorgne, M. Lochus, S. Nicolic, C. Blugeon, L. Jourdren, D. Gomez-Zepeda, S. Tenzer, S. Sanquer, V. Nivet-Antoine, M.C. Menet, J.L. Laplanche, X. Declèves, S. Cisternino, and B. Saubaméa. (2024). Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation. Fluids Barriers CNS 21: 33. 38589879
Chaudhary, N., I. Kumari, P. Sandhu, M. Ahmed, and Y. Akhter. (2016). Proteome scale census of major facilitator superfamily transporters in Trichoderma reesei using protein sequence and structure based classification enhanced ranking. Gene. [Epub: Ahead of Print] 27041239
Chaudhary, N., P. Sandhu, M. Ahmed, and Y. Akhter. (2016). Structural basis of transport function in major facilitator superfamily protein from Trichoderma harzianum. Int J Biol Macromol. [Epub: Ahead of Print] 27816530
Chen, D.E., S. Podell, J.D. Sauer, M.S. Swanson, and M.H. Saier. (2008). The phagosomal nutrient transporter (Pht) family. Microbiology. 154: 42-53. 18174124
Chen, J., M. Yoshinaga, L.D. Garbinski, and B.P. Rosen. (2016). Synergistic interaction of glyceraldehydes-3-phosphate dehydrogenase and ArsJ, a novel organoarsenical efflux permease, confers arsenate resistance. Mol. Microbiol. 100: 945-953. 26991003
Chen, L., P. Jia, Y. Liu, R. Wang, Z. Yin, D. Hu, H. Ning, and Y. Ge. (2023). Fluoride exposure disrupts the cytoskeletal arrangement and ATP synthesis of HT-22 cell by activating the RhoA/ROCK signaling pathway. Ecotoxicol Environ Saf 254: 114718. 36950989
Chen, S.Y., C.J. Pan, K. Nandigama, B.C. Mansfield, S.V. Ambudkar, and J.Y. Chou. (2008). The glucose-6-phosphate transporter is a phosphate-linked antiporter deficient in glycogen storage disease type Ib and Ic. FASEB J. 22: 2206-2213. 18337460
Chen, W.J., S.Y. Huang, Y.W. Chen, Y.F. Liu, and R.S. Huang. (2023). Dietary Folate Deficiency Promotes Lactate Metabolic Disorders to Sensitize Lung Cancer Metastasis through MTOR-Signaling-Mediated Druggable Oncotargets. Nutrients 15:. 36986244
Chenaux, G., L. Matt, T.C. Hill, I. Kaur, X.B. Liu, L.M. Kirk, D.J. Speca, S.A. McMahon, K. Zito, J.W. Hell, and E. Díaz. (2016). Loss of SynDIG1 Reduces Excitatory Synapse Maturation But Not Formation In Vivo. eNeuro 3:. 27800545
Cheng, L.C., S. Baboo, C. Lindsay, L. Brusman, S. Martinez-Bartolomé, O. Tapia, X. Zhang, J.R. Yates, 3rd, and L. Gerace. (2019). Identification of new transmembrane proteins concentrated at the nuclear envelope using organellar proteomics of mesenchymal cells. Nucleus 10: 126-143. 31142202
Cheng, Q. and J.T. Park. (2002). Substrate specificity of the AmpG permease required for recycling of cell wall anhydro-muropeptides. J. Bacteriol. 184: 6434-6436. 12426329
Cheng, X., Q. Xie, and Y. Sun. (2023). Advances in nanomaterial-based targeted drug delivery systems. Front Bioeng Biotechnol 11: 1177151. 37122851
Chiabrando, D., L. Scietti, A.G. Prajica, F. Bertino, E. Tolosano, and F. Magnani. (2020). Expression and purification of the heme exporter FLVCR1a. Protein Expr Purif 172: 105637. [Epub: Ahead of Print] 32278001
Cho, Y.-H., E.-J. Kim, H.-J. Chung, J.-H. Choi, K.F. Chater, B.-E. Ahn, J.-H. Shin, and Y.-H. Roe. (2003). The pqrAB operon is responsible for paraquat resistance in Streptomyces coelicolor. J. Bacteriol. 185: 6756-6763. 14617639
Choquer, M., M.H. Lee, H.J. Bau, and K.R. Chung. (2007). Deletion of a MFS transporter-like gene in Cercospora nicotianae reduces cercosporin toxin accumulation and fungal virulence. FEBS Lett. 581: 489-494. 17250832
Chou, J.Y. and B.C. Mansfield. (2014). The SLC37 family of sugar-phosphate/phosphate exchangers. Curr Top Membr 73: 357-382. 24745989
Choudhary, A., H. Purohit, and P.S. Phale. (2017). Benzoate transport in Pseudomonas putida CSV86. FEMS Microbiol. Lett. 364:. 28591829
Christensen, M., T. Borza, G. Dandanell, A.-M. Gilles, O. Barzu, R.A. Kelln, and J. Neuhard. (2003). Regulation of expression of the 2-deoxy-D-ribose utilization regulon, deoQKPX, from Salmonella enterica serovar typhimurium. J. Bacteriol. 185: 6042-6050. 14526015
Clark, T.J., C. Momany, and E.L. Neidle. (2002). The benPK operon, proposed to play a role in transport, is part of a regulon for benzoate catabolism in Acinetobacter sp. strain ADP1. Microbiology 148: 1213-1223. 11932465
Clegg, S., F. Yu, L. Griffiths, and J.A. Cole. (2002). The roles of the polytopic membrane proteins NarK, NarU and NirC in Escherichia coli K-12: two nitrate and three nitrite transporters. Mol. Microbiol. 44: 143-155. 11967075
Clegg, S.J., W. Jia, and J.A. Cole. (2006). Role of the Escherichia coli nitrate transport protein, NarU, in survival during severe nutrient starvation and slow growth. Microbiology 152: 2091-2100. 16804183
Cliburn, R.A., A.R. Dunn, K.A. Stout, C.A. Hoffman, K.M. Lohr, A.I. Bernstein, E.J. Winokur, J. Burkett, Y. Shmitz, W.M. Caudle, and G.W. Miller. (2016). Immunochemical localization of vesicular monoamine transporter 2 (VMAT2) in mouse brain. J Chem Neuroanat. [Epub: Ahead of Print] 27836486
Colangeli, R., D. Helb, C. Vilchèze, M.H. Hazbón, C.G. Lee, H. Safi, B. Sayers, I. Sardone, M.B. Jones, R.D. Fleischmann, S.N. Peterson, W.R. Jacobs, Jr, and D. Alland. (2007). Transcriptional regulation of multi-drug tolerance and antibiotic-induced responses by the histone-like protein Lsr2 in M. tuberculosis. PLoS Pathog 3: e87. 17590082
Collier, L.S., N.N. Nichols, and E.L. Neidle. (1997). benK encodes a hydrophobic permease-like protein involved in benzoate degradation by Acinetobacter sp. strain ADP1. J. Bacteriol. 179: 5943-5946. 9294456
Condemine, G. (2000). Characterization of SotA and SotB, two Erwinia chrysanthemi proteins which modify isopropyl-β-D-thiogalactopyranoside and lactose induction of the Escherichia coli lac promoter. J. Bacteriol. 182: 1340-1345. 10671456
Cooper, G.J., B. Leighton, A.C. Willis, and A.J. Day. (1989). The amylin superfamily: a novel grouping of biologically active polypeptides related to the insulin A-chain. Prog Growth Factor Res 1: 99-105. 2491258
Costa, C., C. Pires, T.R. Cabrito, A. Renaudin, M. Ohno, H. Chibana, I. Sá-Correia, and M.C. Teixeira. (2013). Candida glabrata drug:H+ antiporter CgQdr2 confers imidazole drug resistance, being activated by transcription factor CgPdr1. Antimicrob. Agents Chemother. 57: 3159-3167. 23629708
Costa, C., J. Ribeiro, I.M. Miranda, A. Silva-Dias, M. Cavalheiro, S. Costa-de-Oliveira, A.G. Rodrigues, and M.C. Teixeira. (2016). Clotrimazole Drug Resistance in Candida glabrata Clinical Isolates Correlates with Increased Expression of the Drug:H+ Antiporters CgAqr1, CgTpo1_1, CgTpo3, and CgQdr2. Front Microbiol 7: 526. 27148215
Coucke, P.J., A. Willaert, M.W. Wessels, B. Callewaert, N. Zoppi, J. De Backer, J.E. Fox, G.M. Mancini, M. Kambouris, R. Gardella, F. Facchetti, P.J. Willems, R. Forsyth, H.C. Dietz, S. Barlati, M. Colombi, B. Loeys, and A. De Paepe. (2006). Mutations in the facilitative glucose transporter GLUT10 alter angiogenesis and cause arterial tortuosity syndrome. Nat. Genet. 38: 452-457. 16550171
Courville, P., M. Quick, and R.J. Reimer. (2010). Structure-function studies of the SLC17 transporter sialin identify crucial residues and substrate-induced conformational changes. J. Biol. Chem. 285: 19316-19323. 20424173
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. 18194158
Cui, G., M.K. Konciute, L. Ling, L. Esau, J.B. Raina, B. Han, O.R. Salazar, J.S. Presnell, N. Rädecker, H. Zhong, J. Menzies, P.A. Cleves, Y.J. Liew, C.J. Krediet, V. Sawiccy, M.J. Cziesielski, P. Guagliardo, J. Bougoure, M. Pernice, H. Hirt, C.R. Voolstra, V.M. Weis, J.R. Pringle, and M. Aranda. (2023). Molecular insights into the Darwin paradox of coral reefs from the sea anemone Aiptasia. Sci Adv 9: eadf7108. 36921053
Cui, H., L. Li, W. Wang, J. Shen, Z. Yue, X. Zheng, X. Zuo, B. Liang, M. Gao, X. Fan, X. Yin, C. Shen, C. Yang, C. Zhang, X. Zhang, Y. Sheng, J. Gao, Z. Zhu, D. Lin, A. Zhang, Z. Wang, S. Liu, L. Sun, S. Yang, Y. Cui, and X. Zhang. (2014). Exome sequencing identifies SLC17A9 pathogenic gene in two Chinese pedigrees with disseminated superficial actinic porokeratosis. J Med Genet 51: 699-704. 25180256
Cui, S., H. Xia, T. Chen, Y. Gu, X. Lv, Y. Liu, J. Li, G. Du, and L. Liu. (2020). Cell Membrane and Electron Transfer Engineering for Improved Synthesis of Menaquinone-7 in Bacillus subtilis. iScience 23: 100918. [Epub: Ahead of Print] 32109677
Cuív, P.O., P. Clarke, D. Lynch, and M. O'Connell. (2004). Identification of rhtX and fptX, novel genes encoding proteins that show homology and function in the utilization of the siderophores rhizobactin 1021 by Sinorhizobium meliloti and pyochelin by Pseudomonas aeruginosa, respectively. J. Bacteriol. 186: 2996-3005. 15126460
Cunningham P. and Naftalin RJ. (2014). Reptation-induced coalescence of tunnels and cavities in Escherichia Coli XylE transporter conformers accounts for facilitated diffusion. J Membr Biol. 247(11):1161-79. 25163893
Cunningham, P., Afzal-Ahmed, I., and Naftalin, R.J. (2006). Docking studies show that D-glucose and quercetin slide through the transporter GLUT1. J. Biol. Chem. 281: 5797-5803. 16407180
Cura, A.J. and A. Carruthers. (2010). Acute modulation of sugar transport in brain capillary endothelial cell cultures during activation of the metabolic stress pathway. J. Biol. Chem. 285: 15430-15439. 20231288
Cushion, M.T., M.S. Collins, T. Sesterhenn, A. Porollo, A.K. Vadukoot, and E.J. Merino. (2016). Functional Characterization of Pneumocystis carinii Inositol Transporter 1. MBio 7:. 27965450
Czech, L., C. Gertzen, S.H.J. Smits, and E. Bremer. (2022). Guilty by association: Importers, exporters and MscS-type mechanosensitive channels encoded in biosynthetic gene clusters for the stress-protectant ectoine. Environ Microbiol 24: 5306-5331. 36104950
Czeredys, M., L. Samluk, K. Michalec, K. Tułodziecka, K. Skowronek, and K.A. Nałęcz. (2013). Caveolin-1 - a novel interacting partner of organic cation/carnitine transporter (octn2): effect of protein kinase C on this interaction in rat astrocytes. PLoS One 8: e82105. 24349196
Czuba, L.C., K.M. Hillgren, and P.W. Swaan. (2018). Post-translational modifications of transporters. Pharmacol Ther 192: 88-99. 29966598
D''Angiolini, S., M.S. Basile, E. Mazzon, and A. Gugliandolo. (2023). In Silico Analysis Reveals the Modulation of Ion Transmembrane Transporters in the Cerebellum of Alzheimer''s Disease Patients. Int J Mol Sci 24:. 37762226
D'Argenio, D.A., A. Segura, W.M. Coco, P.V. Bünz, and L.N. Ornston. (1999). The physiological contribution of Acinetobacter PcaK, a transport system that acts upon protocatechuate, can be masked by the overlapping specificity of VanK. J. Bacteriol. 181: 3505-3515. 10348864
da Cunha, A.C., L.S. Gomes, F. Godoy-Santos, F. Faria-Oliveira, J.A. Teixeira, G.M.S. Sampaio, M.J.M. Trópia, I.M. Castro, C. Lucas, and R.L. Brandão. (2019). High-affinity transport, cyanide-resistant respiration, and ethanol production under aerobiosis underlying efficient high glycerol consumption by Wickerhamomyces anomalus. J Ind Microbiol Biotechnol 46: 709-723. 30680472
Dagher, F., A. Nickzad, J. Zheng, M. Hoffmann, and E. Déziel. (2021). Characterization of the biocontrol activity of three bacterial isolates against the phytopathogen Erwinia amylovora. Microbiologyopen 10: e1202. 34180603
Dahlin, A., E. Geier, S.L. Stocker, C.D. Cropp, E. Grigorenko, M. Bloomer, J. Siegenthaler, L. Xu, A.S. Basile, D.D. Tang-Liu, and K.M. Giacomini. (2013). Gene expression profiling of transporters in the solute carrier and ATP-binding cassette superfamilies in human eye substructures. Mol Pharm 10: 650-663. 23268600
Dakal, T.C., R. Kumar, and D. Ramotar. (2017). Structural modeling of human organic cation transporters. Comput Biol Chem 68: 153-163. [Epub: Ahead of Print] 28343125
Dang, S., L. Sun, Y. Huang, F. Lu, Y. Liu, H. Gong, J. Wang, and N. Yan. (2010). Structure of a fucose transporter in an outward-open conformation. Nature 467: 734-738. 20877283
Darley, P.I., J.A. Hellstern, J.I. Medina-Bellver, S. Marqués, B. Schink, and B. Philipp. (2007). Heterologous expression and identification of the genes involved in anaerobic degradation of 1,3-dihydroxybenzene (resorcinol) in Azoarcus anaerobius. J. Bacteriol. 189: 3824-3833. 17369298
Dashtbani-Roozbehani, A., M. Chitsaz, and M.H. Brown. (2023). The role of TMS 12 in the staphylococcal multidrug efflux protein QacA. J Antimicrob Chemother. [Epub: Ahead of Print] 37100459
Date, S.S., C.Y. Chen, Y. Chen, and M. Jansen. (2016). Experimentally optimized threading structures of the proton-coupled folate transporter. FEBS Open Bio 6: 216-230. 27047750
David, C., C.A. Arango-Franco, M. Badonyi, J. Fouchet, G.I. Rice, B. Didry-Barca, L. Maisonneuve, L. Seabra, R. Kechiche, C. Masson, A. Cobat, L. Abel, E. Talouarn, V. Béziat, C. Deswarte, K. Livingstone, C. Paul, G. Malik, A. Ross, J. Adam, J. Walsh, S. Kumar, D. Bonnet, C. Bodemer, B. Bader-Meunier, J.A. Marsh, J.L. Casanova, Y.J. Crow, B. Manoury, M.L. Frémond, J. Bohlen, and A. Lepelley. (2024). Gain-of-function human UNC93B1 variants cause systemic lupus erythematosus and chilblain lupus. J Exp Med 221:. 38869500
de Ramón-Carbonell, M., M. López-Pérez, L. González-Candelas, and P. Sánchez-Torres. (2019). Transporter Contributes to Fungicide Resistance and Fungal Virulence during Citrus Fruit Infection. J Fungi (Basel) 5:. 31635246
De Zutter JK., Levine KB., Deng D. and Carruthers A. (2013). Sequence determinants of GLUT1 oligomerization: analysis by homology-scanning mutagenesis. J Biol Chem. 288(28):20734-44. 23720776
Deng, D., P. Sun, C. Yan, M. Ke, X. Jiang, L. Xiong, W. Ren, K. Hirata, M. Yamamoto, S. Fan, and N. Yan. (2015). Molecular basis of ligand recognition and transport by glucose transporters. Nature 526: 391-396. 26176916
Deng, Z., W. Peng, Z. Lu, and M. Fu. (2021). [Cloning and expression pattern of phosphate transporter 1;1 cDNA sequence from Spirodela polyrrhiza]. Sheng Wu Gong Cheng Xue Bao 37: 2474-2482. 34327912
Di Daniel, E., M.H. Mok, E. Mead, C. Mutinelli, E. Zambello, L.L. Caberlotto, T.J. Pell, C.J. Langmead, A.J. Shah, G. Duddy, J.N. Kew, and P.R. Maycox. (2009). Evaluation of expression and function of the H+/myo-inositol transporter HMIT. BMC Cell Biol 10: 54. 19607714
Diallinas, G. (2017). Transceptors as a functional link of transporters and receptors. Microb Cell 4: 69-73. 28357392
Diao, J., S. Li, L. Ma, P. Zhang, J. Bai, J. Wang, X. Ma, and W. Ma. (2021). Genome-Wide Analysis of Major Facilitator Superfamily and Its Expression in Response of Poplar to. Front Genet 12: 769888. 34745233
Díaz, E., A. Ferrández, and J.L. García. (1998). Characterization of the hca cluster encoding the dioxygenolytic pathway for initial catabolism of 3-phenylpropionic acid in Escherichia coli K-12. J. Bacteriol. 180: 2915-2923. 9603882
Dietvorst, J., J. Londesborough, and H.Y. Steensma. (2005). Maltotriose utilization in lager yeast strains: MTT1 encodes a maltotriose transporter. Yeast 22: 775-788. 16088872
Dietvorst, J., K. Karhumaa, M.C. Kielland-Brandt, and A. Brandt. (2010). Amino acid residues involved in ligand preference of the Snf3 transporter-like sensor in Saccharomyces cerevisiae. Yeast 27: 131-138. 20014043
Diezemann, A. and E. Boles. (2003). Functional characterization of the Frt1 sugar transporter and of fructose uptake in Kluyveromyces lactis. Curr. Genet. 43: 281-288. 12677461
Divito, C.B., K. Steece-Collier, D.T. Case, S.P. Williams, J.A. Stancati, L. Zhi, M.E. Rubio, C.E. Sortwell, T.J. Collier, D. Sulzer, R.H. Edwards, H. Zhang, and R.P. Seal. (2015). Loss of VGLUT3 Produces Circadian-Dependent Hyperdopaminergia and Ameliorates Motor Dysfunction and l-Dopa-Mediated Dyskinesias in a Model of Parkinson''s Disease. J. Neurosci. 35: 14983-14999. 26558771
Dmitrieva, N., S. Gholami, C. Alleva, P. Carloni, M. Alfonso-Prieto, and C. Fahlke. (2024). Transport mechanism of DgoT, a bacterial homolog of SLC17 organic anion transporters. EMBO. J. [Epub: Ahead of Print] 39455803
Do, H.Q. and M. Jansen. (2022). Cell-Free Expression of Proton-Coupled Folate Transporter in the Presence of Nanodiscs. Methods Mol Biol 2507: 425-444. 35773596
Do, H.Q., C.M. Bassil, E.I. Andersen, and M. Jansen. (2021). Impact of nanodisc lipid composition on cell-free expression of proton-coupled folate transporter. PLoS One 16: e0253184. 34793461
Doege, H., A. Schürmann, G. Bahrenberg, A. Brauers, and H.G. Joost. (2000). GLUT8, a novel member of the sugar transport facilitator family with glucose transport activity. J. Biol. Chem. 275: 16275-16280. 10821868
Domingos, L.T.S., D.C. de Moraes, M.F.C. Santos, J.A.R. Curvelo, B. Bayona-Pacheco, E.A. Marquez, A.W.B. Martinez, R.G.S. Berlinck, and A. Ferreira-Pereira. (2024). Batzelladine D, a Marine Natural Product, Reverses the Fluconazole Resistance Phenotype Mediated by Transmembrane Transporters in and Interferes with Its Biofilm: An In Vitro and In Silico Study. Mar Drugs 22:. 39590782
Doran, J., C. Walters, V. Kyle, P. Wooding, R. Hammett-Burke, and W.H. Colledge. (2016). Mfsd14a (Hiat1) gene disruption causes globozoospermia and infertility in male mice. Reproduction 152: 91-99. 27107036
Dowhan, W. and M. Bogdanov. (2011). Lipid-protein interactions as determinants of membrane protein structure and function. Biochem Soc Trans 39: 767-774. 21599647
Du, X., J. Li, M. Li, X. Yang, Z. Qi, B. Xu, W. Liu, Z. Xu, and Y. Deng. (2020). Research progress on the role of type I vesicular glutamate transporter (VGLUT1) in nervous system diseases. Cell Biosci 10: 26. 32158532
Duan, P., J. Wu, and G. You. (2011). Mutational analysis of the role of GXXXG motif in the function of human organic anion transporter 1 (hOAT1). Int J Biochem Mol Biol 2: 1-7. 21340049
Dube, F., A. Hinas, N. Delhomme, M. Åbrink, S. Svärd, and E. Tydén. (2023). Transcriptomics of ivermectin response in Caenorhabditis elegans: Integrating abamectin quantitative trait loci and comparison to the Ivermectin-exposed DA1316 strain. PLoS One 18: e0285262. 37141255
Duffy, S.P., J. Shing, P. Saraon, L.C. Berger, M.V. Eiden, A. Wilde, and C.S. Tailor. (2010). The Fowler syndrome-associated protein FLVCR2 is an importer of heme. Mol. Cell Biol. 30: 5318-5324. 20823265
Dumitrescu, A.M., X.H. Liao, T.B. Best, K. Brockmann, and S. Refetoff. (2004). A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. Am J Hum Genet 74: 168-175. 14661163
Durães, F., M. Pinto, and E. Sousa. (2018). Medicinal Chemistry Updates on Bacterial Efflux Pump Modulators. Curr. Med. Chem. 25: 6030-6069. 29424299
Eames, M. and T. Kortemme. (2012). Cost-benefit tradeoffs in engineered lac operons. Science 336: 911-915. 22605776
Ebert, K., M. Ewers, I. Bisha, S. Sander, T. Rasputniac, H. Daniel, I. Antes, and H. Witt. (2017). Identification of essential amino acids for glucose transporter 5 (GLUT5)- mediated fructose transport. J. Biol. Chem. [Epub: Ahead of Print] 29259131
Egenberger B., Gorboulev V., Keller T., Gorbunov D., Gottlieb N., Geiger D., Mueller TD. and Koepsell H. (2012). A substrate binding hinge domain is critical for transport-related structural changes of organic cation transporter 1. J Biol Chem. 287(37):31561-73. 22810231
Ekici, S., H. Yang, H.G. Koch, and F. Daldal. (2012). Novel transporter required for biogenesis of cbb3-type cytochrome c oxidase in Rhodobacter capsulatus. MBio 3:. 22294680
Elkins, C.A. and D.C. Savage. (1998). Identification of genes encoding conjugated bile salt hydrolase and transport in Lactobacillus johnsonii 100-100. J. Bacteriol. 180: 4344-4349. 9721268
Elkins, C.A. and D.C. Savage. (2003). CbsT2 from Lactobacillus johnsonii 100-100 is a transport protein of the major facilitator superfamily that facilitates bile acid antiport. J. Mol. Microbiol. Biotechnol. 6: 76-87. 15044826
Elkins, C.A. and L.B. Mullis. (2006). Mammalian steroid hormones are substrates for the major RND- and MFS-type tripartite multidrug efflux pumps of Escherichia coli. J. Bacteriol. 188: 1191-1195. 16428427
Elkins, C.A., S.A. Moser, and D.C. Savage. (2001). Genes encoding bile salt hydrolases and conjugated bile salt transporters in Lactobacillus johnsonii 100-100 and other Lactobacillus species. Microbiology (Reading) 147: 3403-3412. 11739773
Ellis, D.A., V. Mustonen, M. Rodríguez-López, C. Rallis, M. Malecki, D.C. Jeffares, and J. Bähler. (2018). Uncovering Natural Longevity Alleles from Intercrossed Pools of Aging Fission Yeast Cells. Genetics. [Epub: Ahead of Print] 30072377
Enomoto, A., H. Kumura, A. Chairoungdua, Y. Shigeta, P. Jutabha, S.H. Cha, M. Hosoyamada, M. Takeda, T. Sekine, T. Igarashi, H. Matsuo, Y. Kikuchi, T. Oda, K. Ichida, T. Hosoya, K. Shimokata, T. Niwa, Y. Kanai, and H. Endou. (2002). Molecular identification of a renal urate-anion exchanger that regulates blood urate levels. Nature 417: 447-450. 12024214
Enomoto, A., M.F. Wempe, H. Tsuchida, H.J. Shin, S.H. Cha, N. Anzai, A. Goto, A. Sakamoto, T. Niwa, Y. Kanai, M.W. Anders, and H. Endou. (2002). Molecular identification of a novel carnitine transporter specific to human testis. Insights into the mechanism of carnitine recognition. J. Biol. Chem. 277: 36262-36271. 12089149
Eraly, S.A., B.A. Hamilton, and S.K. Nigam. (2003). Organic anion and cation transporters occur in pairs of similar and similarly expressed genes. Biochem. Biophys. Res. Commun. 300: 333-342. 12504088
Eraly, S.A., J.C. Monte, and S.K. Nigam. (2004). Novel slc22 transporter homologs in fly, worm, and human clarify the phylogeny of organic anion and cation transporters. Physiol Genomics 18: 12-24. 15054140
Eraly, S.A., K.T. Bush, R.V. Sampogna, V. Bhatnagar, and S.K. Nigam. (2003). The molecular biology of organic anion transporters: from DNA to FDA? Mol. Pharmacol. in press. 12920404
Eraly, S.A., V. Vallon, T. Rieg, J.A. Gangoiti, W.R. Wikoff, G. Siuzdak, B.A. Barshop, and S.K. Nigam. (2008). Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiol Genomics 33: 180-192. 18270321
Erb, R.W., K.N. Timmis, and D.H. Pieper DH. (1998). Characterization of a gene cluster from Ralstonia eutropha JMP134 encoding metabolism of 4-methylmuconolactone. Gene. 206: 53-62. 9461415
Erickson, J.D., M.K. Schafer, T.I. Bonner, L.E. Eiden, and E. Weihe. (1996). Distinct pharmacological properties and distribution in neurons and endocrine cells of two isoforms of the human vesicular monoamine transporter. Proc. Natl. Acad. Sci. USA 93: 5166-5171. 8643547
Ernst, C.M., P. Staubitz, N.N. Mishra, S.J. Yang, G. Hornig, H. Kalbacher, A.S. Bayer, D. Kraus, and A. Peschel. (2009). The bacterial defensin resistance protein MprF consists of separable domains for lipid lysinylation and antimicrobial peptide repulsion. PLoS Pathog 5: e1000660. 19915718
Ernst, C.M., S. Kuhn, C.J. Slavetinsky, B. Krismer, S. Heilbronner, C. Gekeler, D. Kraus, S. Wagner, and A. Peschel. (2015). The lipid-modifying multiple Peptide resistance factor is an oligomer consisting of distinct interacting synthase and flippase subunits. MBio 6:. 25626904
Erro, R., K.P. Bhatia, A.J. Espay, and P. Striano. (2017). The epileptic and nonepileptic spectrum of paroxysmal dyskinesias: Channelopathies, synaptopathies, and transportopathies. Mov Disord. [Epub: Ahead of Print] 28090678
Essand, M., S. Vikman, J. Grawé, L. Gedda, C. Hellberg, K. Oberg, T.H. Totterman, and V. Giandomenico. (2005). Identification and characterization of a novel splicing variant of vesicular monoamine transporter 1. J Mol Endocrinol 35: 489-501. 16326835
Evers, M.E., H. Trip, M.A. van den Berg, R.A. Bovenberg, and A.J. Driessen. (2004). Compartmentalization and transport in β-lactam antibiotics biosynthesis. Adv Biochem Eng Biotechnol 88: 111-135. 15719554
Fàbrega, A., R.G. Martin, J.L. Rosner, M.M. Tavio, and J. Vila. (2010). Constitutive SoxS expression in a fluoroquinolone-resistant strain with a truncated SoxR protein and identification of a new member of the marA-soxS-rob regulon, mdtG. Antimicrob. Agents Chemother. 54: 1218-1225. 20008776
Fan, Y., H. Wang, Z. Yu, Z. Liang, Y. Li, and G. You. (2022). Inhibition of proteasome, but not lysosome, upregulates organic anion transporter 3 in vitro and in vivo. Biochem Pharmacol 208: 115387. [Epub: Ahead of Print] 36549459
Fang H., Wu Y., Guo J., Rong J., Ma L., Zhao Z., Zuo D. and Peng S. (2012). T-2 toxin induces apoptosis in differentiated murine embryonic stem cells through reactive oxygen species-mediated mitochondrial pathway. Apoptosis. 17(8):895-907. 22614820
Fang, L., J. Hou, Y. Cao, J.J. Shan, and J. Zhao. (2020). Spinster homolog 2 in cancers, its functions and mechanisms. Cell Signal 109821. [Epub: Ahead of Print] 33144184
Fang, X., Y. Liu, W. Xiao, N. Zhao, C. Zhu, D. Yu, and Y. Zhao. (2021). Prognostic SLC family genes promote cell proliferation, migration, and invasion in hepatocellular carcinoma. Acta Biochim Biophys Sin (Shanghai) 53: 1065-1075. 34128989
Farrow, M.F. and E.J. Rubin. (2008). Function of a mycobacterial major facilitator superfamily pump requires a membrane-associated lipoprotein. J. Bacteriol. 190: 1783-1791. 18156250
Farsi, Z., J. Preobraschenski, G. van den Bogaart, D. Riedel, R. Jahn, and A. Woehler. (2016). Single-vesicle imaging reveals different transport mechanisms between glutamatergic and GABAergic vesicles. Science 351: 981-984. 26912364
Farwick, A., S. Bruder, V. Schadeweg, M. Oreb, and E. Boles. (2014). Engineering of yeast hexose transporters to transport D-xylose without inhibition by D-glucose. Proc. Natl. Acad. Sci. USA 111: 5159-5164. 24706835
Fei, H., T. Karnezis, R.J. Reimer, and D.E. Krantz. (2007). Membrane topology of the Drosophila vesicular glutamate transporter. J Neurochem 101: 1662-1671. 17394549
Fernandez-Aguado M., Martin JF., Rodriguez-Castro R., Garcia-Estrada C., Albillos SM., Teijeira F. and Ullan RV. (2014). New insights into the isopenicillin N transport in Penicillium chrysogenum. Metab Eng. 22:89-103. 24480587
Fernández-Aguado, M., R.V. Ullán, F. Teijeira, R. Rodríguez-Castro, and J.F. Martín. (2013). The transport of phenylacetic acid across the peroxisomal membrane is mediated by the PaaT protein in Penicillium chrysogenum. Appl. Microbiol. Biotechnol. 97: 3073-3084. 23053082
Ferrada, E. and G. Superti-Furga. (2022). A structure and evolutionary-based classification of solute carriers. iScience 25: 105096. 36164651
Ferreira, C., F. van Voorst, A. Martins, L. Neves, R. Oliveira, M.C. Kielland-Brandt, C. Lucas, and A. Brandt. (2005). A member of the sugar transporter family, Stl1p is the glycerol/H+ symporter in Saccharomyces cerevisiae. Mol. Biol. Cell 16: 2068-2076. 15703210
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. 20693325
Filippo, C.A., O. Ardon, and N. Longo. (2011). Glycosylation of the OCTN2 carnitine transporter: study of natural mutations identified in patients with primary carnitine deficiency. Biochim. Biophys. Acta. 1812: 312-320. 21126579
Fiorentino, G., R. Ronca, R. Cannio, M. Rossi, and S. Bartolucci. (2007). MarR-like transcriptional regulator involved in detoxification of aromatic compounds in Sulfolobus solfataricus. J. Bacteriol. 189: 7351-7360. 17675388
Fisel P., Stuhler V., Bedke J., Winter S., Rausch S., Hennenlotter J., Nies AT., Stenzl A., Scharpf M., Fend F., Kruck S., Schwab M. and Schaeffeler E. (2015). MCT4 surpasses the prognostic relevance of the ancillary protein CD147 in clear cell renal cell carcinoma. Oncotarget. 6(31):30615-27. 26384346
Floyd, J.L., K.P. Smith, S.H. Kumar, J.T. Floyd, and M.F. Varela. (2010). LmrS is a multidrug efflux pump of the major facilitator superfamily from Staphylococcus aureus. Antimicrob. Agents Chemother. 54: 5406-5412. 20855745
Fluman, N., D. Cohen-Karni, T. Weiss, and E. Bibi. (2009). A promiscuous conformational switch in the secondary multidrug transporter MdfA. J. Biol. Chem. 284: 32296-32304. 19808670
Fomenko, D.E., A.Z. Metlitskaya, J. Péduzzi, C. Goulard, G.S. Katrukha, L.V. Gening, S. Rebuffat, and I.A. Khmel. (2003). Microcin C51 plasmid genes: possible source of horizontal gene transfer. Antimicrob. Agents Chemother. 47: 2868-2874. 12936987
Fonseca, M.V., J.D. Sauer, S. Crepin, B. Byrne, and M.S. Swanson. (2014). The phtC-phtD locus equips Legionella pneumophila for thymidine salvage and replication in macrophages. Infect. Immun. 82: 720-730. 24478086
Foong, W.E., H.K. Tam, J.J. Crames, B. Averhoff, and K.M. Pos. (2019). The chloramphenicol/H+ antiporter CraA of Acinetobacter baumannii AYE reveals a broad substrate specificity. J Antimicrob Chemother 74: 1192-1201. 30783664
Forment, J.V., M. Flipphi, D. Ramón, L. Ventura, and A.P. Maccabe. (2006). Identification of the mstE gene encoding a glucose-inducible, low affinity glucose transporter in Aspergillus nidulans. J. Biol. Chem. 281: 8339-8346. 16418173
Fox, V., F. Santoro, G. Pozzi, and F. Iannelli. (2021). Predicted transmembrane proteins with homology to Mef(A) are not responsible for complementing mef(A) deletion in the mef(A)-msr(D) macrolide efflux system in Streptococcus pneumoniae. BMC Res Notes 14: 432. 34823574
Franza, T., B. Mahé, and D. Expert. (2005). Erwinia chrysanthemi requires a second iron transport route dependent of the siderophore achromobactin for extracellular growth and plant infection. Mol. Microbiol. 55: 261-275. 15612933
Fredriksson, R., S. Sreedharan, K. Nordenankar, J. Alsiö, F.A. Lindberg, A. Hutchinson, A. Eriksson, S. Roshanbin, D.M. Ciuculete, A. Klockars, A. Todkar, M.G. Hägglund, S.V. Hellsten, V. Hindlycke, &.#.1.9.7.;. Västermark, G. Shevchenko, G. Olivo, C. K, K. Kullander, A. Moazzami, J. Bergquist, P.K. Olszewski, and H.B. Schiöth. (2019). The polyamine transporter Slc18b1(VPAT) is important for both short and long time memory and for regulation of polyamine content in the brain. PLoS Genet 15: e1008455. [Epub: Ahead of Print] 31800589
Friesema, E.C.H., S. Ganguly, A. Abdalla, J.E. Manning Fox, A.P. Halestrap, and T.J. Visser. (2003). Identification of monocarboxylate transporter 8 as a specific thyroid hormone transporter. J. Biol. Chem. 278: 40128-40135. 12871948
Frohlich, K.M. and J.P. Audia. (2013). Dual mechanisms of metabolite acquisition by the obligate intracytosolic pathogen Rickettsia prowazekii reveal novel aspects of triose phosphate transport. J. Bacteriol. 195: 3752-3760. 23772074
Fu, D. and P.C. Maloney. (1998). Structure-function relationships in OxlT, the oxalate/formate transporter of Oxalobacter formigenes. Topological features of transmembrane helix 11 as visualized by site-directed fluorescent labeling. J. Biol. Chem. 273: 17962-17967. 9651403
Fu, D., R.I. Sarker, K. Abe, E. Bolton, and P.C. Maloney. (2001). Structure/function relationships in OxlT, the oxalate-formate transporter of oxalobacter formigenes. Assignment of transmembrane helix 11 to the translocation pathway. J. Biol. Chem. 276: 8753-8760. 11113128
Fujisaki, S., S. Ohnuma, T. Horiuchi, I. Takahashi, S. Tsukui, Y. Nishimura, T. Nishino, M. Kitabatake, and H. Inokuchi. (1996). Cloning of a gene from Escherichia coli that confers resistance to fosmidomycin as a consequence of amplification. Gene 175: 83-87. 8917080
Fukuda, M., H. Takeda, H.E. Kato, S. Doki, K. Ito, A.D. Maturana, R. Ishitani, and O. Nureki. (2015). Structural basis for dynamic mechanism of nitrate/nitrite antiport by NarK. Nat Commun 6: 7097. 25959928
Fukuhara, S., S. Simmons, S. Kawamura, A. Inoue, Y. Orba, T. Tokudome, Y. Sunden, Y. Arai, K. Moriwaki, J. Ishida, A. Uemura, H. Kiyonari, T. Abe, A. Fukamizu, M. Hirashima, H. Sawa, J. Aoki, M. Ishii, and N. Mochizuki. (2012). The sphingosine-1-phosphate transporter Spns2 expressed on endothelial cells regulates lymphocyte trafficking in mice. J Clin Invest 122: 1416-1426. 22406534
Fukui, K., K. Nanatani, M. Nakayama, Y. Hara, M. Tokura, and K. Abe. (2019). Corynebacterium glutamicum CgynfM encodes a dicarboxylate transporter applicable to succinate production. J Biosci Bioeng 127: 465-471. 30392965
Furrer, J.L., D.N. Sanders, I.G. Hook-Barnard, and M.A. McIntosh. (2002). Export of the siderophore enterobactin in Escherichia coli: involvement of a 43 kDa membrane exporter. Mol. Microbiol. 44: 1225-1234. 12068807
Gaballa, A., M. Cao, and J.D. Helmann. (2003). Two MerR homologues that affect copper induction of the Bacillus subtilis copZA operon. Microbiology 149: 3413-3421. 14663075
Galazka, J.M., C. Tian, W.T. Beeson, B. Martinez, N.L. Glass, and J.H. Cate. (2010). Cellodextrin transport in yeast for improved biofuel production. Science 330: 84-86. 20829451
Galochkina, T., M. Ng Fuk Chong, L. Challali, S. Abbar, and C. Etchebest. (2019). New insights into GluT1 mechanics during glucose transfer. Sci Rep 9: 998. 30700737
Ganapathy, M.E., W. Huang, D.P. Rajan, A.L. Carter, M. Sugawara, K. Iseki, F.H. Leibach, and V. Ganapathy. (2000). β-lactam antibiotics as substrates for OCTN2, an organic cation/carnitine transporter. J. Biol. Chem. 275: 1699-1707. 10636865
Gao, H., J. Liang, J. Duan, L. Chen, H. Li, T. Zhen, F. Zhang, Y. Dong, H. Shi, and A. Han. (2021). A Prognosis Marker SLC2A3 Correlates With EMT and Immune Signature in Colorectal Cancer. Front Oncol 11: 638099. 34211835
Gao, W., C. Li, F. Wang, Y. Yang, L. Zhang, Z. Wang, X. Chen, M. Tan, G. Cao, and G. Zong. (2023). An efflux pump in genomic island GI-M202a mediates the transfer of polymyxin B resistance in Pandoraea pnomenusa M202. Int Microbiol 1-14. [Epub: Ahead of Print] 37316617
Gao, Z., L. Maurousset, R. Lemoine, S.D. Yoo, S. van Nocker, and W. Loescher. (2003). Cloning, expression, and characterization of sorbitol transporters from developing sour cherry fruit and leaf sink tissues. Plant Physiol. 131: 1566-1575. 12692316
Garcia, D.L. and J.P. Dillard. (2008). Mutations in ampG or ampD affect peptidoglycan fragment release from Neisseria gonorrhoeae. J. Bacteriol. 190: 3799-3807. 18390650
Garcia-Dominguez, M., L. Lopez-Maury, F.J. Florencio, and J.C. Reyes. (2000). A gene cluster involved in metal homeostasis in the cyanobacterium Synechocystis sp. strain PCC 6803. J. Bacteriol. 182: 1507-1514. 10692354
Garrido, D., J. Busscher, and A.J. van Tunen. (2006). Promoter activity of a putative pollen monosaccharide transporter in Petunia hybrida and characterisation of a transposon insertion mutant. Protoplasma 228: 3-11. 16937049
Gbelska, Y., J.J. Krijger, and K.D. Breunig. (2006). Evolution of gene families: the multidrug resistance transporter genes in five related yeast species. FEMS Yeast Res 6: 345-355. 16630275
Geistlinger, K., J.D.R. Schmidt, and E. Beitz. (2023). Human monocarboxylate transporters accept and relay protons via the bound substrate for selectivity and activity at physiological pH. PNAS Nexus 2: pgad007. 36874278
George, R.L., X. Wu, W. Huang, Y.J. Fei, F.H. Leibach, and V. Ganapathy. (1999). Molecular cloning and functional characterization of a polyspecific organic anion transporter from Caenorhabditis elegans. J Pharmacol Exp Ther 291: 596-603. 10525077
Gerin, I., M. Veiga-da-Cunha, Y. Achouri, J.F. Collet, and E.V. Schaftingen. (1999). Sequence of a putative glucose 6-phosphate translocase, mutated in glycogen storage disease type Ib. Fed. Euro. Biochem. 419: 235-238. 9428641
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. 27230378
Girardi, E., A. César-Razquin, S. Lindinger, K. Papakostas, J. Konecka, J. Hemmerich, S. Kickinger, F. Kartnig, B. Gürtl, K. Klavins, V. Sedlyarov, A. Ingles-Prieto, G. Fiume, A. Koren, C.H. Lardeau, R. Kumaran Kandasamy, S. Kubicek, G.F. Ecker, and G. Superti-Furga. (2020). A widespread role for SLC transmembrane transporters in resistance to cytotoxic drugs. Nat Chem Biol 16: 469-478. 32152546
Girgis, J., D. Yang, I. Chakroun, Y. Liu, and A. Blais. (2021). Six1 promotes skeletal muscle thyroid hormone response through regulation of the MCT10 transporter. Skelet Muscle 11: 26. 34809717
Giri, D., V. Govindaraj, S. Kumar, H. Ungati, and G. Mugesh. (2024). A Highly Selective Fluorescent Probe for Monitoring the Thyroid Hormone Transporter Activity in Mammalian Cells. Chemistry 30: e202401719. 38995511
Girin T., El-Kafafi el-S., Widiez T., Erban A., Hubberten HM., Kopka J., Hoefgen R., Gojon A. and Lepetit M. (2010). Identification of Arabidopsis mutants impaired in the systemic regulation of root nitrate uptake by the nitrogen status of the plant. Plant Physiol. 153(3):1250-60. 20448103
Giugno, A., E. Falcone, F. Fortunato, I. Sammarra, R. Procopio, M. Gagliardi, A. Bauleo, L. de Stefano, I. Martino, and A. Gambardella. (2024). Glucose transporter-1 deficiency syndrome with extreme phenotypic variability in a five-generation family carrying a novel SLC2A1 variant. Eur J Neurol 31: e16325. 38803061
Goddard, A.D., J.W. Moir, D.J. Richardson, and S.J. Ferguson. (2008). Interdependence of two NarK domains in a fused nitrate/nitrite transporter. Mol. Microbiol. 70: 667-681. 18823285
Goffeau, A., J. Park, I.T. Paulsen, J.-L. Jonniaux, T. Dinh, P. Mordant, and M.H. Saier, Jr. (1997). Multidrug resistant transport proteins in yeast. Complete inventory and phylogenetic characterization of yeast open reading frames within the major facilitator superfamily. Yeast 13: 43-54. 9046086
Gomez, M. and S.M. Cutting. (1997). Identification of a new sigmaB-controlled gene, csbX, in Bacillus subtilis. Gene 188: 29-33. 9099855
Gong, J., C.M. Hutter, P.A. Newcomb, C.M. Ulrich, S.A. Bien, P.T. Campbell, J.A. Baron, S.I. Berndt, S. Bezieau, H. Brenner, G. Casey, A.T. Chan, J. Chang-Claude, M. Du, D. Duggan, J.C. Figueiredo, S. Gallinger, E.L. Giovannucci, R.W. Haile, T.A. Harrison, R.B. Hayes, M. Hoffmeister, J.L. Hopper, T.J. Hudson, J. Jeon, M.A. Jenkins, J. Kocarnik, S. Küry, L. Le Marchand, Y. Lin, N.M. Lindor, R. Nishihara, S. Ogino, J.D. Potter, A. Rudolph, R.E. Schoen, P. Schrotz-King, D. Seminara, M.L. Slattery, S.N. Thibodeau, M. Thornquist, R. Toth, R. Wallace, E. White, S. Jiao, M. Lemire, L. Hsu, U. Peters, and. (2016). Genome-Wide Interaction Analyses between Genetic Variants and Alcohol Consumption and Smoking for Risk of Colorectal Cancer. PLoS Genet 12: e1006296. 27723779
Gonzalez, E., E. Flier, D. Molle, D. Accili, and T.E. McGraw. (2011). Hyperinsulinemia leads to uncoupled insulin regulation of the GLUT4 glucose transporter and the FoxO1 transcription factor. Proc. Natl. Acad. Sci. USA 108: 10162-10167. 21646544
González-Pasayo, R. and E. Martínez-Romero. (2000). Multiresistance genes of Rhizobium etli CFN42. Mol. Plant-Microbe Interact. 13: 572-577. 10796024
Gonzalez-Resines, S., P.J. Quinn, R.J. Naftalin, and C. Domene. (2021). Multiple Interactions of Glucose with the Extra-Membranous Loops of GLUT1 Aid Transport. J Chem Inf Model 61: 3559-3570. 34260246
Gora, N., L.J. Weselinski, V.V. Begoyan, A. Cooper, J.Y. Choe, and M. Tanasova. (2023). Discrimination of GLUTs by Fructose Isomers Enables Simultaneous Screening of GLUT5 and GLUT2 Activity in Live Cells. ACS Chem Biol 18: 1089-1100. 37116192
Gorboulev, V., N. Shatskaya, C. Volk, and H. Koepsell. (2005). Subtype-specific affinity for corticosterone of rat organic cation transporters rOCT1 and rOCT2 depends on three amino acids within the substrate binding region. Mol Pharmacol 67: 1612-1619. 15662045
Gründemann, D., J. Babin-Ebell, F. Martel, N. Ording, A. Schmidt, and E. Schömig. (1997). Primary structure and functional expression of the apical organic cation transporter from kidney epithelial LLC-PK1 cells. J. Biol. Chem. 272: 10408-10413. 9099681
Granados, J.C., A. Richelle, J.M. Gutierrez, P. Zhang, X. Zhang, V. Bhatnagar, N.E. Lewis, and S.K. Nigam. (2021). Coordinate regulation of systemic and kidney tryptophan metabolism by the drug transporters OAT1 and OAT3. J. Biol. Chem. 296: 100575. 33757768
Gras, C., E. Herzog, G.C. Bellenchi, V. Bernard, P. Ravassard, M. Pohl, B. Gasnier, B. Giros, and S. El Mestikawy. (2002). A third vesicular glutamate transporter expressed by cholinergic and serotoninergic neurons. J. Neurosci. 22: 5442-5451. 12097496
Green, A.L., E.J. Anderson, and R.J. Brooker. (2000). A revised model for the structure and function of the lactose permease. J. Biol. Chem. 275: 23240-23246. 10807929
Griffith, J.K., D.H. Cuellar, C.A. Fordyce, K.G. Hutchings, and A.A. Mondragon. (1995). Structure and function of the class C tetracycline/H+ antiporter: three independent groups of phenotypes are conferred by TetA (C). Mol. Membr. Biol. 11: 271-277. 7711837
Griffith, J.K., M.E. Baker, D.A. Rouch, M.G.P. Page, R.A. Skurray, I.T. Paulsen, K.F. Chater, S.A. Baldwin, and P.J.F. Henderson. (1992). Membrane transport proteins: implications of sequence comparisons. Curr. Opin. Cell Biol. 4: 684-695. 1419050
Grijota-Martínez, C., S. Bárez-López, D. Gómez-Andrés, and A. Guadaño-Ferraz. (2020). MCT8 Deficiency: The Road to Therapies for a Rare Disease. Front Neurosci 14: 380. 32410949
Groeneweg S., Friesema EC., Kersseboom S., Klootwijk W., Visser WE., Peeters RP. and Visser TJ. (2014). The role of Arg445 and Asp498 in the human thyroid hormone transporter MCT8. Endocrinology. 155(2):618-26. 24265446
Groeneweg, S., A. van den Berge, E.C. Lima de Souza, M.E. Meima, R.P. Peeters, and W.E. Visser. (2020). Insights Into the Mechanism of MCT8 Oligomerization. J Endocr Soc 4: bvaa080. 32724870
Gronskiy, S.V., N.P. Zakataeva, M.V. Vitushkina, L.R. Ptitsyn, I.B. Altman, A.E. Novikova, and V.A. Livshits. (2005). The yicM (nepI) gene of Escherichia coli encodes a major facilitator superfamily protein involved in efflux of purine ribonucleosides. FEMS Microbiol. Lett. 250: 39-47. 16040204
Großhennig, S., S.R. Schmidl, G. Schmeisky, J. Busse, and J. Stülke. (2013). Implication of glycerol and phospholipid transporters in Mycoplasma pneumoniae growth and virulence. Infect. Immun. 81: 896-904. 23297388
Grudzinska Pechhacker, M.K., G. Yoon, L.N. Hazrati, J. Maynes, H. MacDonald, E. Tavares, A. Vincent, and E. Heon. (2020). FLVCR1-related disease as a rare cause of retinitis pigmentosa and hereditary sensory autonomic neuropathy. Eur J Med Genet 63: 104037. 32822874
Gründling, A. and O. Schneewind. (2007). Genes required for glycolipid synthesis and lipoteichoic acid anchoring in Staphylococcus aureus. J. Bacteriol. 189: 2521-2530. 17209021
Guan, L., O. Mirza, G. Verner, S. Iwata, and H.R. Kaback. (2007). Structural determination of wild-type lactose permease. Proc. Natl. Acad. Sci. USA 104: 15294-15298. 17881559
Guardia, C.M., X.F. Tan, T. Lian, M.S. Rana, W. Zhou, E.T. Christenson, A.J. Lowry, J.D. Faraldo-Gómez, J.S. Bonifacino, J. Jiang, and A. Banerjee. (2020). Structure of Human ATG9A, the Only Transmembrane Protein of the Core Autophagy Machinery. Cell Rep 31: 107837. 32610138
Guillén-Yunta, M., V. Valcárcel-Hernández, &.#.1.9.3.;. García-Aldea, G. Soria, J.M. García-Verdugo, A. Montero-Pedrazuela, and A. Guadaño-Ferraz. (2023). Neurovascular unit disruption and blood-brain barrier leakage in MCT8 deficiency. Fluids Barriers CNS 20: 79. 37924081
Guo, H., T. Huang, J. Zhao, H. Chen, and G. Chen. (2018). Fungi short-chain carboxylate transporter: shift from microbe hereditary functional component to metabolic engineering target. Appl. Microbiol. Biotechnol. 102: 4653-4662. 29679102
Guo, Y., Z. Ran, Y. Zhang, Z. Song, L. Wang, L. Yao, M. Zhang, J. Xin, and X. Mao. (2020). Marein ameliorates diabetic nephropathy by inhibiting renal sodium glucose transporter 2 and activating the AMPK signaling pathway in db/db mice and high glucose-treated HK-2 cells. Biomed Pharmacother 131: 110684. 33152903
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. 25938806
Guzzo, J. and M.S. Dubow. (2000). A novel selenite- and tellurite-inducible gene in Escherichia coli. Appl. Environ. Microbiol. 66: 4972-4978. 11055951
Gyimesi, G. and M.A. Hediger. (2022). Systematic in silico discovery of novel solute carrier-like proteins from proteomes. PLoS One 17: e0271062. 35901096
Hadzi-Petrushev, N., R. Stojchevski, A. Jakimovska, M. Stamenkovska, S. Josifovska, A. Stamatoski, I. Sazdova, R. Sopi, A. Kamkin, H. Gagov, M. Mladenov, and D. Avtanski. (2024). GLUT5-overexpression-related tumorigenic implications. Mol Med 30: 114. 39107723
Halestrap, A.P. (2013). Monocarboxylic acid transport. Compr Physiol 3: 1611-1643. 24265240
Hamacher, T., J. Becker, M. Gárdonyi, B. Hahn-Hägerdal, and E. Boles. (2002). Characterization of the xylose-transporting properties of yeast hexose transporters and their influence on xylose utilization. Microbiology 148: 2783-2788. 12213924
Hampe, I.A.I., J. Friedman, M. Edgerton, and J. Morschhäuser. (2017). An acquired mechanism of antifungal drug resistance simultaneously enables Candida albicans to escape from intrinsic host defenses. PLoS Pathog 13: e1006655. 28953977
Hannes, F., P. Hammond, O. Quarrell, J.P. Fryns, K. Devriendt, and J.R. Vermeesch. (2012). A microdeletion proximal of the critical deletion region is associated with mild Wolf-Hirschhorn syndrome. Am J Med Genet A 158A: 996-1004. 22438245
Hanschmann EM., Godoy JR., Berndt C., Hudemann C. and Lillig CH. (2013). Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid Redox Signal. 19(13):1539-605. 23397885
Hansen, M.R., J. Tranekjaer Jørgensen, and G. Dandanell. (2006). Xanthosine utilization in Salmonella enterica serovar Typhimurium is recovered by a single aspartate-to-glycine substitution in xanthosine phosphorylase. J. Bacteriol. 188: 4153-4157. 16707709
Harb, J.F., C.L. Christensen, S.H. Kan, A.K. Rha, P. Andrade-Heckman, L. Pollard, R. Steet, J.Y. Huang, and R.Y. Wang. (2023). Base editing corrects the common Salla disease c.115C>T variant. Mol Ther Nucleic Acids 34: 102022. 37727271
Hariharan, P., D. Balasubramaniam, A. Peterkofsky, H.R. Kaback, and L. Guan. (2015). Thermodynamic mechanism for inhibition of lactose permease by the phosphotransferase protein IIAGlc. Proc. Natl. Acad. Sci. USA 112: 2407-2412. 25675534
Harris, K.G. and C.B. Coyne. (2015). Unc93b Induces Apoptotic Cell Death and Is Cleaved by Host and Enteroviral Proteases. PLoS One 10: e0141383. 26509685
Harvat, E.M., Y.M. Zhang, C.V. Tran, Z. Zhang, M.W. Frank, C.O. Rock, and M.H. Saier, Jr. (2005). Lysophospholipid flipping across the Escherichia coli inner membrane catalyzed by a transporter (LplT) belonging to the major facilitator superfamily. J. Biol. Chem. 280: 12028-12034. 15661733
Hassan, K.A., A.J. Brzoska, N.L. Wilson, B.A. Eijkelkamp, M.H. Brown, and I.T. Paulsen. (2011). Roles of DHA2 family transporters in drug resistance and iron homeostasis in Acinetobacter spp. J. Mol. Microbiol. Biotechnol. 20: 116-124. 21430390
Hassan, K.A., R.A. Skurray, and M.H. Brown. (2007). Transmembrane helix 12 of the Staphylococcus aureus multidrug transporter QacA lines the bivalent cationic drug binding pocket. J. Bacteriol. 189: 9131-9134. 17951386
Havukainen, S., J. Pujol-Giménez, M. Valkonen, M.A. Hediger, and C.P. Landowski. (2021). Functional characterization of a highly specific L-arabinose transporter from Trichoderma reesei. Microb Cell Fact 20: 177. 34496831
Havukainen, S., M. Valkonen, K. Koivuranta, and C.P. Landowski. (2020). Studies on sugar transporter CRT1 reveal new characteristics that are critical for cellulase induction in. Biotechnol Biofuels 13: 158. 32944074
Hawkins, A.C. and C.S. Harwood. (2002). Chemotaxis of Ralstonia eutropha JMP134(pJP4) to the herbicide 2,4-dichlorophenoxyacetate. Appl. Environ. Microbiol. 68: 968-972. 11823246
Hayashi, M., K. Tabata, M. Yagasaki, and Y. Yonetani. (2010). Effect of multidrug-efflux transporter genes on dipeptide resistance and overproduction in Escherichia coli. FEMS Microbiol. Lett. 304: 12-19. 20067529
Hayashi, T., Y. Tanaka, N. Sakai, U. Okada, M. Yao, N. Watanabe, T. Tamura, and I. Tanaka. (2013). SCO4008, a putative TetR transcriptional repressor from streptomyces coelicolor A3(2), regulates transcription of sco4007 by multidrug recognition. J. Mol. Biol. 425: 3289-3300. 23831227
He, E., W. Quan, J. Luo, C. Liu, W. Zheng, and Q. Shen. (2023). Absorption and Transport Mechanism of Red Meat-Derived -glycolylneuraminic Acid and Its Damage to Intestinal Barrier Function through the NF-κB Signaling Pathway. Toxins (Basel) 15:. 36828446
He, J., G. Liu, F. Kong, Q. Tan, Z. Wang, M. Yang, Y. He, X. Jia, C. Yan, C. Wang, and H. Qian. (2024). Structural basis for the transport and substrate selection of human urate transporter 1. Cell Rep 43: 114628. 39146184
He, W., M. Jiang, Y. Li, and X. Ge. (2024). Identification of the Major Facilitator Superfamily Efflux Pump KpsrMFS in That Is Down-Regulated in the Presence of Multi-Stress Factors. Int J Mol Sci 25:. 38338743
Heiland, S., N. Radovanovic, M. Höfer, J. Winderickx, and H. Lichtenberg. (2000). Multiple hexose transporters of Schizosaccharomyces pombe. J. Bacteriol. 182: 2153-2162. 10735857
Hellborg, L., M. Woolfit, M. Arthursson-Hellborg, and J. Piskur. (2008). Complex evolution of the DAL5 transporter family. BMC Genomics 9: 164. 18405355
Henderson, P.J. and R.A. Giddens. (1977). 2-Deoxy-D-galactose, a substrate for the galactose-transport system of Escherichia coli. Biochem. J. 168: 15-22. 23115
Henderson, P.J., R.A. Giddens, and M.C. Jones-Mortimer. (1977). Transport of galactose, glucose and their molecular analogues by Escherichia coli K12. Biochem. J. 162: 309-320. 15558
Hernández-Montalvo, V., F. Valle, F. Bolivar, and G. Gosset. (2001). Characterization of sugar mixtures utilization by an Escherichia coli mutant devoid of the phosphotransferase system. Appl. Microbiol. Biotechnol. 57: 186-191. 11693918
Hernández-Serda, M.A., A.Y. Alarcón-López, V.H. Vázquez-Valadez, P. Briseño-Lugo, P.A. Martínez-Soriano, V. Leguízamo, N. Torres, R. González-Terán, L.A. Cárdenas-Granados, F. Sánchez Muñoz, E. Rodríguez, C. Lerma, A.M. Zúñiga Muñoz, E. Ángeles, and R. Carbó. (2024). Hypoxic Cardioprotection by New Antihypertensive Compounds in High Salt-Diet Hypertensive Rats: Glucose Transport Participation and Its Possible Pathway. Int J Mol Sci 25:. 39201496
Hertz, L. and G.A. Dienel. (2013). Lactate transport and transporters: general principles and functional roles in brain cells. J. Neurosci. Res 79: 11-18. 15586354
Heuel, H., A. Shakeri-Garakani, S. Turgut, and J.W. Lengeler. (1998). Genes for D-arabinitol and ribitol catabolism from Klebsiella pneumoniae. Microbiology 144(Pt6): 1631-1639. 9639934
Heuel, H., S. Turgut, K. Schmid, and J.W. Lengeler. (1997). Substrate recognition domains as revealed by active hybrids between the D-arabinitol and ribitol transporters from Klebsiella pneumoniae. J. Bacteriol. 179: 6014-6019. 9324246
Heymann, J.A.W., R. Sarker, T. Hirai, D. Shi, J.L.S. Milne, P.C. Maloney, and S. Subramaniam. (2001). Projection structure and molecular architecture of OxlT, a bacterial membrane transporter. EMBO J. 20: 4408-4413. 11500368
Hiasa, M., T. Miyaji, Y. Haruna, T. Takeuchi, Y. Harada, S. Moriyama, A. Yamamoto, H. Omote, and Y. Moriyama. (2014). Identification of a mammalian vesicular polyamine transporter. Sci Rep 4: 6836. 25355561
Higgins, C.F. (2007). Multiple molecular mechanisms for multidrug resistance transporters. Nature 446: 749-757. 17429392
Higuchi, K. (2024). [Analysis of Endogenous Metabolite Transporters for the Development of Novel Pharmaceutical Therapies and Drug Targets]. Yakugaku Zasshi 144: 1051-1054. 39617467
Hiller, E., V. Hörz, and R. Sting. (2024). Corynebacterium pseudotuberculosis: Whole genome sequencing reveals unforeseen and relevant genetic diversity in this pathogen. PLoS One 19: e0309282. 39186721
Hirabayashi, Y., K.H. Nomura, and K. Nomura. (2013). The acetyl-CoA transporter family SLC33. Mol Aspects Med 34: 586-589. 23506891
Hirai, T., J.A.W. Heymann, D. Shi, R. Sarker, P.C. Maloney, and S. Subramaniam. (2002). Three-dimensional structure of a bacterial oxalate transporter. Nature Struct. Biol. 9: 597-600. 12118242
Hirai, T., J.A.W. Heymann, P.C. Maloney, and S. Subramaniam. (2003). A structural model for 12-helix transporters belonging to the major facilitator superfamily. J. Bacteriol. 185: 1712-1718. 12591890
Hisano, Y., N. Kobayashi, A. Kawahara, A. Yamaguchi, and T. Nishi. (2011). The sphingosine 1-phosphate transporter, SPNS2, functions as a transporter of the phosphorylated form of the immunomodulating agent FTY720. J. Biol. Chem. 286: 1758-1766. 21084291
Hoen, E., F.M. Goossens, K. Falize, S. Mayerl, A.H. van der Spek, and A. Boelen. (2024). The Differential Effect of a Shortage of Thyroid Hormone Compared with Knockout of Thyroid Hormone Transporters Mct8 and Mct10 on Murine Macrophage Polarization. Int J Mol Sci 25:. 38396788
Hoischen, C., J. Levin, S. Pitaknarongphorn, J. Reizer, and M.H. Saier, Jr. (1996). Involvement of the central loop of the lactose permease of Escherichia coli in its allosteric regulation by the glucose-specific enzyme IIA of the phosphoenolpyruvate-dependent phosphotransferase system. J. Bacteriol. 178: 6082-6086. 8830713
Hollands, K., C.M. Baron, K.J. Gibson, K.J. Kelly, E.A. Krasley, L.A. Laffend, R.M. Lauchli, L.A. Maggio-Hall, M.J. Nelson, J.C. Prasad, Y. Ren, B.A. Rice, G.H. Rice, and S.C. Rothman. (2019). Engineering two species of yeast as cell factories for 2''-fucosyllactose. Metab Eng 52: 232-242. 30557615
Holman, G.D. (2020). Structure, function and regulation of mammalian glucose transporters of the SLC2 family. Pflugers Arch. [Epub: Ahead of Print] 32591905
Honerlagen, H., H. Reyer, M. Oster, S. Ponsuksili, N. Trakooljul, B. Kuhla, N. Reinsch, and K. Wimmers. (2021). Identification of Genomic Regions Influencing N-Metabolism and N-Excretion in Lactating Holstein- Friesians. Front Genet 12: 699550. 34335696
Hopkins AP., Hawkhead JA. and Thomas GH. (2013). Transport and catabolism of the sialic acids N-glycolylneuraminic acid and 3-keto-3-deoxy-D-glycero-D-galactonononic acid by Escherichia coli K-12. FEMS Microbiol Lett. 347(1):14-22. 23848303
Horiba, N., S. Masuda, A. Takeuchi, D. Takeuchi, M. Okuda, and K. Inui. (2003). Cloning and characterization of a novel Na+-dependent glucose transporter (NaGLT1) in rat kidney. J. Biol. Chem. 278: 14669-14676. 12590146
Horváth, H.R., C.L. Fazekas, D. Balázsfi, S.K. Jain, J. Haller, and D. Zelena. (2018). Contribution of Vesicular Glutamate Transporters to Stress Response and Related Psychopathologies: Studies in VGluT3 Knockout Mice. Cell Mol Neurobiol 38: 37-52. 28776199
Hoshino, H. (2012). Cellular Factors Involved in HTLV-1 Entry and Pathogenicit. Front Microbiol 3: 222. 22737146
Hosoyamada, M., K. Ichida, A. Enomoto, T. Hosoya, and H. Endou. (2004). Function and localization of urate transporter 1 in mouse kidney. J Am Soc Nephrol 15: 261-268. 14747372
Hosoyamada, M., T. Sekine, Y. Kanai, and H. Endou. (1999). Molecular cloning and functional expression of a multispecific organic anion transporter from human kidney. Am. J. Physiol. 276: F122-128. 9887087
Hosseini Bereshneh, A. and M. Garshasbi. (2018). Novel in-frame deletion in MFSD8 gene revealed by trio whole exome sequencing in an Iranian affected with neuronal ceroid lipofuscinosis type 7: a case report. J Med Case Rep 12: 281. 30249282
Hou, X., Y. Wang, Y. Yang, and Z. Xiao. (2023). Discovery of Novel Biphenyl Carboxylic Acid Derivatives as Potent URAT1 Inhibitors. Molecules 28:. 37959834
Hu, B., H.K. Akula, D. Noh, Y.F. Mui, M. Slifstein, R. Parsey, and W. Qu. (2023). An improved synthesis of [ F]VAT and its precursor. J Labelled Comp Radiopharm 66: 384-392. 37615234
Hu, C., L. Tao, X. Cao, and L. Chen. (2020). The solute carrier transporters and the brain: Physiological and pharmacological implications. Asian J Pharm Sci 15: 131-144. 32373195
Hu, X., X. Peng, Y. Zhang, S. Fan, X. Liu, Y. Song, S. Ren, L. Chen, Y. Chen, R. Wang, J. Peng, X. Shen, and Y. Chen. (2024). Shikonin reverses cancer-associated fibroblast-induced gemcitabine resistance in pancreatic cancer cells by suppressing monocarboxylate transporter 4-mediated reverse Warburg effect. Phytomedicine 123: 155214. 38134861
Hu, Y., V. Fernández, and L. Ma. (2014). Nitrate transporters in leaves and their potential roles in foliar uptake of nitrogen dioxide. Front Plant Sci 5: 360. 25126090
Hu, Y.W., L. Xiao, L. Zheng, and Q. Wang. (2017). Synaptic vesicle 2C and its synaptic-related function. Clin Chim Acta 472: 112-117. 28774501
Huang, B., Z. Lin, Z. Chen, J. Chen, B. Shi, J. Jia, Y. Li, Y. Pan, Y. Liang, and Z. Cai. (2023). Strain differences in the drug transport capacity of intestinal glucose transporters in Sprague-Dawley versus Wistar rats, C57BL/6J versus Kunming mice. Int J Pharm 640: 123000. 37254285
Huang, H.T., J.H. Leu, P.Y. Huang, and L.L. Chen. (2012). A putative cell surface receptor for white spot syndrome virus is a member of a transporter superfamily. PLoS One 7: e33216. 22427993
Huang, J., P.W. O''Toole, W. Shen, H. Amrine-Madsen, X. Jiang, N. Lobo, L.M. Palmer, L. Voelker, F. Fan, M.N. Gwynn, and D. McDevitt. (2004). Novel chromosomally encoded multidrug efflux transporter MdeA in Staphylococcus aureus. Antimicrob. Agents Chemother. 48: 909-917. 14982783
Huang, P., H. Åbacka, D. Varela, R. Venskutonytė, L. Happonen, J.S. Bogan, P. Gourdon, M.R. Amiry-Moghaddam, I. André, and K. Lindkvist-Petersson. (2023). The intracellular helical bundle of human glucose transporter GLUT4 is important for complex formation with ASPL. FEBS Open Bio. [Epub: Ahead of Print] 37731227
Huang, X., H. Li, N. Shenkar, and A. Zhan. (2023). Multidimensional plasticity jointly contributes to rapid acclimation to environmental challenges during biological invasions. RNA. [Epub: Ahead of Print] 36810233
Huang, Y., M.J. Lemieux, J. Song, M. Auer, and D.N. Wang. (2003). Structure and mechanism of the glycerol-3-phosphate transporter from Escherichia coli. Science 301: 616-620. 12893936
Huber, R.J., J. Gray, and W.D. Kim. (2023). Loss of mfsd8 alters the secretome during Dictyostelium aggregation. Eur J. Cell Biol. 102: 151361. [Epub: Ahead of Print] 37742391
Huber, R.J., S. Mathavarajah, and S.Q. Yap. (2020). Mfsd8 localizes to endocytic compartments and influences the secretion of Cln5 and cathepsin D in Dictyostelium. Cell Signal 70: 109572. 32087303
Hung, H.C., L.C. Li, J.H. Guh, F.L. Kung, and L.C. Hsu. (2022). Discovery of New Glucose Uptake Inhibitors as Potential Anticancer Agents by Non-Radioactive Cell-Based Assays. Molecules 27:. 36432207
Huntley, S.A., D. Krofchick, and M. Silverman. (2004). Position 170 of Rabbit Na+/glucose cotransporter (rSGLT1) lies in the Na+ pathway; modulation of polarity/charge at this site regulates charge transfer and carrier turnover. Biophys. J. 87: 295-310. 15240465
Hvorup, R.N. and M.H. Saier, Jr. (2002). Sequence similarity between the channel-forming domains of voltage-gated ion channel proteins and the C-terminal domains of secondary carriers of the major facilitator superfamily. Microbiology 148: 3760-3762. 12480878
Iannaccone Farkašová, S., P. Vasovčák, D. Sopková, M. Pisarčíková, M. Švajdler, L. Fröhlichová, L. Mistríková, and D. Farkaš. (2019). Neuron.al ceroid lipofuscinosis with cardiac involvement. Cesk Patol 55: 176-181. 31726844
Iannelli, F., F. Santoro, M. Santagati, J.D. Docquier, E. Lazzeri, G. Pastore, M. Cassone, M.R. Oggioni, G.M. Rossolini, S. Stefani, and G. Pozzi. (2018). Type M Resistance to Macrolides Is Due to a Two-Gene Efflux Transport System of the ATP-Binding Cassette (ABC) Superfamily. Front Microbiol 9: 1670. 30108557
Igarashi, K. and K. Kashiwagi. (2010). Characteristics of cellular polyamine transport in prokaryotes and eukaryotes. Plant Physiol. Biochem 48: 506-512. 20159658
Ikeda, M., Y. Mizuno, S. Awane, M. Hayashi, S. Mitsuhashi, and S. Takeno. (2011). Identification and application of a different glucose uptake system that functions as an alternative to the phosphotransferase system in Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 90: 1443-1451. 21452034
Imai, H., K. Yoshimura, Y. Miyamoto, K. Sasa, M. Sugano, M. Chatani, M. Takami, M. Yamamoto, and R. Kamijo. (2019). Roles of monocarboxylate transporter subtypes in promotion and suppression of osteoclast differentiation and survival on bone. Sci Rep 9: 15608. 31666601
Inaoka, T., K. Takahashi, M. Ohnishi-Kameyama, M. Yoshida,and K. Ochi. (2003). Guanine nucleotides guanosine 5'-diphosphate 3'-diphosphate and GTP co-operatively regulate the production of an antibiotic bacilysin in Bacillus subtilis. J. Biol. Chem. 278: 2169-2176. 12372825
Ingoglia F., Visigalli R., Rotoli BM., Barilli A., Riccardi B., Puccini P. and Dall'Asta V. (2016). Functional activity of L-carnitine transporters in human airway epithelial cells. Biochim Biophys Acta. 1858(2):210-9. 26607009
Inoue, K., Y. Nakai, S. Ueda, S. Kamigaso, K.Y. Ohta, M. Hatakeyama, Y. Hayashi, M. Otagiri, and H. Yuasa. (2008). Functional characterization of PCFT/HCP1 as the molecular entity of the carrier-mediated intestinal folate transport system in the rat model. Am. J. Physiol. Gastrointest. Liver Physiol. 294: G660-668. 18174275
Iserovich, P., D. Wang, L. Ma, H. Yang, F.A. Zuniga, J.M. Pascual, K. Kuang, D.C. De Vivo, and J. Fischbarg. (2002). Changes in glucose transport and water permeability resulting from the T310I pathogenic mutation in Glut1 are consistent with two transport channels per monomer. J. Biol. Chem. 277: 30991-30997. 12032147
Ishida, H., J. Asami, Z. Zhang, T. Nishizawa, H. Shigematsu, U. Ohto, and T. Shimizu. (2021). Cryo-EM structures of Toll-like receptors in complex with UNC93B1. Nat Struct Mol Biol 28: 173-180. 33432245
Ismail, A. and M. Tanasova. (2022). Importance of GLUT Transporters in Disease Diagnosis and Treatment. Int J Mol Sci 23:. 35955833
Isnardi, I., Y.S. Ng, I. Srdanovic, R. Motaghedi, S. Rudchenko, H. von Bernuth, S.Y. Zhang, A. Puel, E. Jouanguy, C. Picard, B.Z. Garty, Y. Camcioglu, R. Doffinger, D. Kumararatne, G. Davies, J.I. Gallin, S. Haraguchi, N.K. Day, J.L. Casanova, and E. Meffre. (2008). IRAK-4- and MyD88-dependent pathways are essential for the removal of developing autoreactive B cells in humans. Immunity 29: 746-757. 19006693
Iwabuchi, T., and S. Harayama. (1997). Biochemical and genetic characterization of 2-carboxybenzaldehyde dehydrogenase, an enzyme involved in phenanthrene degradation by Nocardioides sp. strain KP7. J. Bacteriol. 179: 6488-6494. 9335300
Iwai, Y., S. Kamatani, S. Moriyama, and H. Omote. (2019). Function of essential chloride and arginine residue in nucleotide binding to vesicular nucleotide transporter. J Biochem. [Epub: Ahead of Print] 30649354
Iwayama, H., T. Tanaka, K. Aoyama, M. Moroto, S. Adachi, Y. Fujisawa, H. Matsuura, K. Takano, H. Mizuno, and A. Okumura. (2021). Regional Difference in Myelination in Monocarboxylate Transporter 8 Deficiency: Case Reports and Literature Review of Cases in Japan. Front Neurol 12: 657820. 34335438
Izbicka, E. and R.T. Streeper. (2023). Mitigation of Insulin Resistance by Natural Products from a New Class of Molecules, Membrane-Active Immunomodulators. Pharmaceuticals (Basel) 16:. 37513825
Jacobs, C., L. Huang, E. Bartowsky, S. Normark, and J.T. Park. (1994). Bacterial cell wall recycling provides cystolic muropeptides as effectors for β-lactamase induction. EMBO J. 13: 4684-4694. 7925310
Janaszkiewicz, A., &.#.1.9.3.;. Tóth, Q. Faucher, H. Arnion, N. Védrenne, C. Barin-Le Guellec, P. Marquet, and F. Di Meo. (2023). Substrate binding and lipid-mediated allostery in the human organic anion transporter 1 at the atomic-scale. Biomed Pharmacother 160: 114342. [Epub: Ahead of Print] 36739760
Jansen, J., E.C. Friesema, M.H. Kester, C.E. Schwartz, and T.J. Visser. (2008). Genotype-phenotype relationship in patients with mutations in thyroid hormone transporter MCT8. Endocrinology 149(5): 2184-2190. 18187543
Jayaram, D.T., P. Sivaram, P. Biswas, Y. Dai, E.A. Sweeny, and D.J. Stuehr. (2024). Heme allocation in eukaryotic cells relies on mitochondrial heme export through FLVCR1b to cytosolic GAPDH. Res Sq. 38746106
Jeanguenin, L., A. Lara-Núñez, D.A. Rodionov, A.L. Osterman, N.Y. Komarova, D. Rentsch, J.F. Gregory, 3rd, and A.D. Hanson. (2012). Comparative genomics and functional analysis of the NiaP family uncover nicotinate transporters from bacteria, plants, and mammals. Funct Integr Genomics 12: 25-34. 21953179
Jensen, L.T., M. Ajua-Alemanji, and V. Cizewski Culotta. (2003). The Saccharomyces cerevisiae high affinity phosphate transporter encoded by PHO84 also functions in manganese homeostasis. J. Biol. Chem. 278: 42036-42040. 12923174
Jia, B., P. Yuan, W.J. Lan, Y.H. Xuan, and C.O. Jeon. (2019). New insight into the classification and evolution of glucose transporters in the Metazoa. FASEB J. fj201802617R. [Epub: Ahead of Print] 30888203
Jia, R., C. Martens, M. Shekhar, S. Pant, G.A. Pellowe, A.M. Lau, H.E. Findlay, N.J. Harris, E. Tajkhorshid, P.J. Booth, and A. Politis. (2020). Hydrogen-deuterium exchange mass spectrometry captures distinct dynamics upon substrate and inhibitor binding to a transporter. Nat Commun 11: 6162. 33268777
Jia, W., N. Tovell, S. Clegg, M. Trimmer, and J. Cole. (2009). A single channel for nitrate uptake, nitrite export and nitrite uptake by Escherichia coli NarU and a role for NirC in nitrite export and uptake. Biochem. J. 417: 297-304. 18691156
Jiang, D., Y. Zhao, J. Fan, X. Liu, Y. Wu, W. Feng, and X.C. Zhang. (2014). Atomic resolution structure of the E. coli YajR transporter YAM domain. Biochem. Biophys. Res. Commun. 450: 929-935. 24952155
Jiang, D., Y. Zhao, X. Wang, J. Fan, J. Heng, X. Liu, W. Feng, X. Kang, B. Huang, J. Liu, and X.C. Zhang. (2013). Structure of the YajR transporter suggests a transport mechanism based on the conserved motif A. Proc. Natl. Acad. Sci. USA 110: 14664-14669. 23950222
Jiang, H., J. Chen, X. Du, D. Feng, Y. Zhang, J. Qi, Y. He, Z. An, Y. Lu, C. Ge, and Y. Wang. (2024). Unveiling Synergistic Potency: Exploring Butyrolactone I to Enhance Gentamicin Efficacy against Methicillin-Resistant (MRSA) Strain USA300. ACS Infect Dis 10: 196-214. 38127778
Jin, J., A.A. Guffanti, C. Beck, and T.A. Krulwich. (2001). Twelve-transmembrane-segment (TMS) version (*TMS VII-VIII) of the 14-TMS Tel(L) antibiotic resistance protein retains monovalent cation transport modes but lacks tetracycline efflux capacity. J. Bacteriol. 183: 2667-2671. 11274128
Jin, J., A.A. Guffanti, D.H. Bechhofer, and T.A. Krulwich. (2002). Tet(L) and Tet(K) tetracycline-divalent metal/H+ antiporters: characterization of multiple catalytic modes and a mutagenesis approach to differences in their efflux substrate and coupling ion preferences. J. Bacteriol. 184: 4722-4732. 12169596
Jin, Q., L. Agrawal, Z. VanHorn-Ali, and G. Alkhatib. (2006). Infection of CD4+ T lymphocytes by the human T cell leukemia virus type 1 is mediated by the glucose transporter GLUT-1: evidence using antibodies specific to the receptor''s large extracellular domain. Virology 349: 184-196. 16519917
Johannes, J., D. Braun, A. Kinne, D. Rathmann, J. Köhrle, and U. Schweizer. (2016). Few amino acid exchanges expand the substrate spectrum of monocarboxylate transporter 10. Mol Endocrinol me20161037. [Epub: Ahead of Print] 27244477
Johannes, J., R. Jayarama-Naidu, F. Meyer, E. Katrin Wirth, U. Schweizer, L. Schomburg, J. Köhrle, and K. Renko. (2016). Silychristin, a flavonolignan derived from the milk thistle is a potent inhibitor of the thyroid hormone transporter MCT8. Endocrinology en20151933. [Epub: Ahead of Print] 26910310
Johansen, L.E., P. Nygaard, C. Lassen, Y. Agerso, and H.H. Saxild. (2003). Definition of a second Bacillus subtilis pur regulon comprising the pur and xpt-pbuX operons plus pbuG, nupG (yxjA), and pbuE (ydhL). J. Bacteriol. 185: 5200-5209. 12923093
Johansson, L. and G. Lidén. (2006). Transcriptome analysis of a shikimic acid producing strain of Escherichia coli W3110 grown under carbon- and phosphate-limited conditions. J Biotechnol 126: 528-545. 16828913
Johnston, M. (1999). Feasting, fasting and fermenting, glucose sensing in yeast and other cells. Trends Genet. 15: 29-33. 10087931
Jolfayi, A.G., N. Naderi, S. Ghasemi, A. Salmanipour, S. Adimi, M. Maleki, and S. Kalayinia. (2024). A novel pathogenic variant in the carnitine transporter gene, SLC22A5, in association with metabolic carnitine deficiency and cardiomyopathy features. BMC Cardiovasc Disord 24: 1. 38166572
Jones, R.S. and M.E. Morris. (2016). Monocarboxylate Transporters: Therapeutic Targets and Prognostic Factors in Disease. Clin Pharmacol Ther 100: 454-463. 27351344
Joost, H.-G. and B. Thorens. (2001). The extended GLUT-family of sugar/polyol transport facilitators: nomenclature, sequence characteristics, and potential function of its novel members. Mol. Membr. Biol. 18: 247-256. 11780753
Juge, N., Y. Yoshida, S. Yatsushiro, H. Omote, and Y. Moriyama. (2006). Vesicular glutamate transporter contains two independent transport machineries. J. Biol. Chem. 281: 39499-39506. 17046815
Jung, S.K., R. Morimoto, M. Otsuka, and H. Omote. (2006). Transmembrane topology of vesicular glutamate transporter 2. Biol Pharm Bull 29: 547-549. 16508164
Jutabha, P., Y. Kanai, M. Hosoyamada, A. Chairoungdua, D.K. Kim, Y. Iribe, E. Babu, J.Y. Kim, N. Anzai, V. Chatsudthipong, and H. Endou. (2003). Identification of a novel voltage-driven organic anion tansporter present at apical membrane of renal proximal tubule. J. Biol. Chem. 278: 27930-27938. 12740363
Jørgensen T.R., P.A. vanKuyk, B.R. Poulsen, G.J. Ruijter, J. Visser, J.J. Iversen. (2007). Glucose uptake and growth of glucose-limited chemostat cultures of Aspergillus niger and a disruptant lacking MstA, a high-affinity glucose transporter. Microbiology. 153: 1963-1973. 17526853
Kaback, H.R. (2015). A chemiosmotic mechanism of symport. Proc. Natl. Acad. Sci. USA 112: 1259-1264. 25568085
Kaback, H.R. and J. Wu. (1997). From membrane to molecule to the third amino acid from the left with a membrane transport protein. Quart. Rev. Biophys. 30: 333-364. 9634651
Kalaba, P., K. Pacher, P.J. Neill, V. Dragacevic, M. Zehl, J. Wackerlig, M. Kirchhofer, S.B. Sartori, H. Gstach, S. Kouhnavardi, A. Fabisikova, M. Pillwein, F. Monje-Quiroga, K. Ebner, A. Prado-Roller, N. Singewald, E. Urban, T. Langer, C. Pifl, J. Lubec, J.J. Leban, and G. Lubec. (2023). Chirality Matters: Fine-Tuning of Novel Monoamine Reuptake Inhibitors Selectivity through Manipulation of Stereochemistry. Biomolecules 13:. 37759815
Kalashnikova, E., R.A. Lorca, I. Kaur, G.A. Barisone, B. Li, T. Ishimaru, J.S. Trimmer, D.P. Mohapatra, and E. Díaz. (2010). SynDIG1: an activity-regulated, AMPA- receptor-interacting transmembrane protein that regulates excitatory synapse development. Neuron. 65: 80-93. 20152115
Kaler, G., D.M. Truong, A. Khandelwal, M. Nagle, S.A. Eraly, P.W. Swaan, and S.K. Nigam. (2007). Structural variation governs substrate specificity for organic anion transporter (OAT) homologs. Potential remote sensing by OAT family members. J. Biol. Chem. 282: 23841-23853. 17553798
Kaler, G., D.M. Truong, D.E. Sweeney, D.W. Logan, M. Nagle, W. Wu, S.A. Eraly, and S.K. Nigam. (2006). Olfactory mucosa-expressed organic anion transporter, Oat6, manifests high affinity interactions with odorant organic anions. Biochem. Biophys. Res. Commun. 351: 872-876. 17094945
Kanamori, A., J. Nakayama, M.N. Fukuda, W.B. Stallcup, K. Sasaki, M. Fukuda, and Y. Hirabayashi. (1997). Expression cloning and characterization of a cDNA encoding a novel membrane protein required for the formation of O-acetylated ganglioside: a putative acetyl-CoA transporter. Proc. Natl. Acad. Sci. USA 94: 2897-2902. 9096318
Kanamori, Y., A. Saito, Y. Hagiwara-Komoda, D. Tanaka, K. Mitsumasu, S. Kikuta, M. Watanabe, R. Cornette, T. Kikawada, and T. Okuda. (2010). The trehalose transporter 1 gene sequence is conserved in insects and encodes proteins with different kinetic properties involved in trehalose import into peripheral tissues. Insect Biochem Mol Biol 40: 30-37. 20035867
Kanbay, M., M.C. Bulbul, S. Copur, B. Afsar, A.A. Sag, D. Siriopol, M. Kuwabara, S. Badarau, A. Covic, and A. Ortiz. (2020). Therapeutic implications of shared mechanisms in non-alcoholic fatty liver disease and chronic kidney disease. J Nephrol. [Epub: Ahead of Print] 32440840
Karimian, M. and L.N. Ornston. (1981). Participation of the β-ketoadipate transport system in chemotaxis. J Gen Microbiol 124: 25-28. 7320700
Kasahara, T. and M. Kasahara. (2010). Identification of a key residue determining substrate affinity in the yeast glucose transporter Hxt7: a two-dimensional comprehensive study. J. Biol. Chem. 285: 26263-26268. 20525688
Kasahara, T., K. Shimogawara, and M. Kasahara. (2011). Crucial effects of amino acid side chain length in transmembrane segment 5 on substrate affinity in yeast glucose transporter Hxt7. Biochemistry 50: 8674-8681. 21892826
Kasahara, T., M. Maeda, M. Ishiguro, and M. Kasahara. (2007). Identification by comprehensive chimeric analysis of a key residue responsible for high affinity glucose transport by yeast HXT2. J. Biol. Chem. 282: 13146-13150. 17369259
Kasho, V.N., I.N. Smirnova, and H.R. Kaback. (2006). Sequence alignment and homology threading reveals prokaryotic and eukaryotic proteins similar to lactose permease. J. Mol. Biol. 358: 1060-1070. 16574153
Kathare, P.K., S. Dharmasiri, E.D. Vincill, P. Routray, I. Ahmad, D.M. Roberts, and N. Dharmasiri. (2019). Arabidopsis PIC30 encodes a Major Facilitator Superfamily (MFS) transporter responsible for the uptake of picolinate herbicides. Plant J. [Epub: Ahead of Print] 31710151
Kaur, I., V. Yarov-Yarovoy, L.M. Kirk, K.E. Plambeck, E.V. Barragan, E.S. Ontiveros, and E. Díaz. (2016). Activity-Dependent Palmitoylation Controls SynDIG1 Stability, Localization, and Function. J. Neurosci. 36: 7562-7568. 27445135
Kaur, J. and A.K. Bachhawat. (2007). Yct1p, a novel, high-affinity, cysteine-specific transporter from the yeast Saccharomyces cerevisiae. Genetics 176: 877-890. 17435223
Kawachi, H., G.D. Han, N. Miyauchi, T. Hashimoto, K. Suzuki, and F. Shimizu. (2009). Therapeutic targets in the podocyte: findings in anti-slit diaphragm antibody-induced nephropathy. J Nephrol 22: 450-456. 19662599
Kawahara, A., T. Nishi, Y. Hisano, H. Fukui, A. Yamaguchi, and N. Mochizuki. (2009). The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science 323: 524-527. 19074308
Kawano-Kawada M., Pongcharoen P., Kawahara R., Yasuda M., Yamasaki T., Akiyama K., Sekito T. and Kakinuma Y. (2016). Vba4p, a vacuolar membrane protein, is involved in the drug resistance and vacuolar morphology of Saccharomyces cerevisiae. Biosci Biotechnol Biochem. 80(2):279-87. 26325352
Kaya A., Karakaya HC., Fomenko DE., Gladyshev VN. and Koc A. (2009). Identification of a novel system for boron transport: Atr1 is a main boron exporter in yeast. Mol Cell Biol. 29(13):3665-74. 19414602
Kayingo, G., A. Martins, R. Andrie, L. Neves, C. Lucas, and B. Wong. (2009). A permease encoded by STL1 is required for active glycerol uptake by Candida albicans. Microbiology 155: 1547-1557. 19383674
Kaynar, K., B. Güvercin, M. Şahin, N. Turan, and F. Açíkyürek. (2022). A novel mutation in a patient with familial renal hypouricemia type 2. Nefrologia (Engl Ed) 42: 347-350. 36210123
Kazakov, T., A. Metlitskaya, and K. Severinov. (2007). Amino acid residues required for maturation, cell uptake, and processing of translation inhibitor microcin C. J. Bacteriol. 189: 2114-2118. 17158672
Keates, R.A., D.E. Culham, Y.I. Vernikovska, A.J. Zuiani, J.M. Boggs, and J.M. Wood. (2010). Transmembrane helix I and periplasmic loop 1 of Escherichia coli ProP are involved in osmosensing and osmoprotectant transport. Biochemistry 49: 8847-8856. 20828170
Keel, S.B., R.T. Doty, Z. Yang, J.G. Quigley, J. Chen, S. Knoblaugh, P.D. Kingsley, I. De Domenico, M.B. Vaughn, J. Kaplan, J. Palis, and J.L. Abkowitz (2008). A heme export protein is required for red blood cell differentiation and iron homeostasis. Science 319: 825-828. 18258918
Kekuda, R., P.D. Prasad, X. Wu, H. Wang, Y.-J. Fei, F.H. Leibach, and V. Ganapathy. (1998). Cloning and functional characterization of a potential-sensitive polyspecific organic cation transporter (OCT3) most abundantly expressed in placenta. J. Biol. Chem. 273: 15971-15979. 9632645
Kemp, B.J., D.L. Church, J. Hatzold, B. Conradt, and E.J. Lambie. (2009). Gem-1 encodes an SLC16 monocarboxylate transporter-related protein that functions in parallel to the gon-2 TRPM channel during gonad development in Caenorhabditis elegans. Genetics 181: 581-591. 19087963
Kenny, T.C., A. Khan, Y. Son, L. Yue, S. Heissel, A. Sharma, H.A. Pasolli, Y. Liu, E.R. Gamazon, H. Alwaseem, R.K. Hite, and K. Birsoy. (2023). Integrative genetic analysis identifies FLVCR1 as a plasma-membrane choline transporter in mammals. Cell Metab. [Epub: Ahead of Print] 37100056
Khalfaoui-Hassani, B., A.F. Verissimo, H.G. Koch, and F. Daldal. (2016). Uncovering the Transmembrane Metal Binding Site of the Novel Bacterial Major Facilitator Superfamily-Type Copper Importer CcoA. MBio 7:. 26787831
Khalfaoui-Hassani, B., P.I. Trasnea, S. Steimle, H.G. Koch, and F. Daldal. (2021). Cysteine Mutants of the Major Facilitator Superfamily-Type Transporter CcoA Provide Insight into Copper Import. mBio e0156721. [Epub: Ahead of Print] 34281385
Khan, A., J. Cheng, A. Kitashova, L. Fürtauer, T. Nägele, C. Picco, J. Scholz-Starke, I. Keller, H.E. Neuhaus, and B. Pommerrenig. (2023). Vacuolar sugar transporter EARLY RESPONSE TO DEHYDRATION 6-LIKE4 affects fructose signaling and plant growth. Plant Physiol. [Epub: Ahead of Print] 37427783
Khan, A.A. and J.G. Quigley. (2011). Control of intracellular heme levels: heme transporters and heme oxygenases. Biochim. Biophys. Acta. 1813: 668-682. 21238504
Khan, A.A. and J.G. Quigley. (2018). Heme and FLVCR-related transporter families SLC48 and SLC49. Mol Aspects Med 34: 669-682. 23506900
Khan, A.A., T. Hanada, M. Mohseni, J.J. Jeong, L. Zeng, M. Gaetani, D. Li, B.C. Reed, D.W. Speicher, and A.H. Chishti. (2008). Dematin and adducin provide a novel link between the spectrin cytoskeleton and human erythrocyte membrane by directly interacting with glucose transporter-1. J. Biol. Chem. 283: 14600-14609. 18347014
Khare, P., A. Mulakaluri, and S.M. Parsons. (2010). Search for the acetylcholine and vesamicol binding sites in vesicular acetylcholine transporter: the region around the lumenal end of the transport channel. J Neurochem 115: 984-993. 20831599
Khlebnikov, A., K.A. Datsenko, T. Skaug, B.L. Wanner, and J.D. Keasling. (2001). Homogeneous expression of the P(BAD) promoter in Escherichia coli by constitutive expression of the low-affinity high-capacity AraE transporter. Microbiology 147: 3241-3247. 11739756
Khries, M., A. Lim, D. Mitra, M. Anderson, J. Bengtsson, A. Bowron, E. Harris, J. Blickwedel, K. Wood, and A.P. Basu. (2023). Broadening the Spectrum of Phenotype: Primary Carnitine Deficiency Presenting with Focal Myoclonus. Child Neurol Open 10: 2329048X231184183. 37475835
Kikawada, T., A. Saito, Y. Kanamori, Y. Nakahara, K. Iwata, D. Tanaka, M. Watanabe, and T. Okuda. (2007). Trehalose transporter 1, a facilitated and high-capacity trehalose transporter, allows exogenous trehalose uptake into cells. Proc. Natl. Acad. Sci. USA 104: 11585-11590. 17606922
Kim JY. and Kandror KV. (2012). The first luminal loop confers insulin responsiveness to glucose transporter 4. Mol Biol Cell. 23(5):910-7. 22262463
Kim M.G., F.A. Flomerfelt, K.N. Lee, C. Chen, R.H. Schwartz. (2000). A putative 12 transmembrane domain cotransporter expressed in thymic cortical epithelial cells. J. Immunol. 164:3185-3192. 10706709
Kim, D.K., K.H. Kim, E.J. Cho, S.J. Joo, J.M. Chung, B.Y. Son, J.H. Yum, Y.M. Kim, H.J. Kwon, B.W. Kim, T.H. Kim, and E.W. Lee. (2013). Gene cloning and characterization of MdeA, a novel multidrug efflux pump in Streptococcus mutans. J Microbiol Biotechnol 23: 430-435. 23462018
Kim, H.J., H. Jeong, and S.J. Lee. (2020). Short-Term Adaptation Modulates Anaerobic Metabolic Flux to Succinate by Activating ExuT, a Novel D-Glucose Transporter in. Front Microbiol 11: 27. 32038601
Kim, H.K., I. Lee, H. Bang, H.C. Kim, W.Y. Lee, S.H. Yun, J. Lee, S.J. Lee, Y.S. Park, K.M. Kim, and W.K. Kang. (2018). MCT4 Expression Is a Potential Therapeutic Target in Colorectal Cancer with Peritoneal Carcinomatosis. Mol Cancer Ther 17: 838-848. 29483215
Kim, J., S. Park, S. Kim, S. Ryu, H. Hwang, S. Cho, Y. Han, J. Kim, Y. Park, E.K. Lee, and M. Lee. (2024). Enhancing the anticancer effect of androgen deprivation therapy by monocarboxylate transporter 1 inhibitor in prostate cancer cells. Prostate 84: 814-822. 38558458
Kim, J.H., L. Mailloux, D. Bloor, B. Maddox, and J. Humble. (2023). The role of salt bridge networks in the stability of the yeast hexose transporter 1. Biochim. Biophys. Acta. Gen Subj 1867: 130490. [Epub: Ahead of Print] 37844739
Kim, J.H., L. Mailloux, D. Bloor, H. Tae, H. Nguyen, M. McDowell, J. Padilla, and A. DeWaard. (2024). Multiple roles for the cytoplasmic C-terminal domains of the yeast cell surface receptors Rgt2 and Snf3 in glucose sensing and signaling. Sci Rep 14: 4055. 38374219
Kim, O.B., H. Richter, T. Zaunmüller, S. Graf, and G. Unden. (2011). Role of secondary transporters and phosphotransferase systems in glucose transport by Oenococcus oeni. J. Bacteriol. 193: 6902-6911. 22020640
Kim, T.W., D.H. Pyo, E. Ko, N.H. Yun, S.J. Song, S.M. Choi, H.K. Hong, S.H. Kim, Y.L. Choi, J. Lee, W.Y. Lee, and Y.B. Cho. (2022). Expression of SLC22A18 regulates oxaliplatin resistance by modulating the ERK pathway in colorectal cancer. Am J Cancer Res 12: 1393-1408. 35411243
Kimura, Y., S. Ishida, H. Matoba, and N. Okahisa. (2004). A Myxococcus xanthus rppA-mmrA double mutant exhibits reduced uptake of amino acids and tolerance of some antimicrobials. FEMS Microbiol. Lett. 238: 145-150. 15336415
King, S.C. and T.H. Wilson. (1990). Identification of valine 177 as a mutation altering specificity for transport of sugars by the Escherichia coli lactose carrier. Enhanced specificity for sucrose and maltose. J. Biol. Chem. 265: 9638-9644. 2190983
Kinne, A., G. Kleinau, C.S. Hoefig, A. Grüters, J. Köhrle, G. Krause, and U. Schweizer. (2010). Essential molecular determinants for thyroid hormone transport and first structural implications for monocarboxylate transporter 8. J. Biol. Chem. 285: 28054-28063. 20628049
Kinne, A., R. Schülein, and G. Krause. (2011). Primary and secondary thyroid hormone transporters. Thyroid Res 4Suppl1: S7. 21835054
Kitagawa, W., S. Takami, K. Miyauchi, E. Masai, Y. Kamagata, J.M. Tiedje, and M. Fukuda. (2002). Novel 2,4-dichlorophenoxyacetic acid degradation genes from oligotrophic Bradyrhizobium sp. strain HW13 isolated from a pristine environment. J. Bacteriol. 184: 509-518. 11751829
Kitamura, Y., M. Kandeel, E. Oba, C. Iwai, K. Iritani, N. Nagaya, R. Namura, H. Katagiri, H. Ueda, and Y. Kitade. (2023). A Diversifiable Synthetic Platform for the Discovery of New Carbasugar SGLT2 Inhibitors Using Azide-Alkyne Click Chemistry. Chem Pharm Bull (Tokyo) 71: 240-249. 36858530
Klepek, Y.S., D. Geiger, R. Stadler, F. Klebl, L. Landouar-Arsivaud, R. Lemoine, R. Hedrich, and N. Sauer. (2005). Arabidopsis POLYOL TRANSPORTER5, a new member of the monosaccharide transporter-like superfamily, mediates H+-Symport of numerous substrates, including myo-inositol, glycerol, and ribose. Plant Cell 17: 204-218. 15598803
Kodippili, G.C., K. Giger, K.S. Putt, and P.S. Low. (2020). DARC, Glycophorin A, Band 3, and GLUT1 Diffusion in Erythrocytes: Insights into Membrane Complexes. Biophys. J. 119: 1749-1759. 33069269
Koepsell, H. (1998). Organic cation transporters in intestine, kidney, liver, and brain. Annu. Rev. Physiol. 60: 243-266. 9558463
Koepsell, H. (2013). The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 34: 413-435. 23506881
Koepsell, H., V. Gorboulev, and P. Arndt. (1999). Molecular pharmacology of organic cation transporters in kidney. J. Membr. Biol. 167: 103-117. 9916142
Kohyama, N., H. Shiokawa, M. Ohbayashi, Y. Kobayashi, and T. Yamamoto. (2013). Characterization of monocarboxylate transporter 6: expression in human intestine and transport of the antidiabetic drug nateglinide. Drug Metab Dispos 41: 1883-1887. 23935065
Koita, K. and C.V. Rao. (2012). Identification and analysis of the putative pentose sugar efflux transporters in Escherichia coli. PLoS One 7: e43700. 22952739
Koleske, M.L., G. McInnes, J.E.H. Brown, N. Thomas, K. Hutchinson, M.Y. Chin, A. Koehl, M.R. Arkin, A. Schlessinger, R.C. Gallagher, Y.S. Song, R.B. Altman, and K.M. Giacomini. (2022). Functional genomics of OCTN2 variants informs protein-specific variant effect predictor for Carnitine Transporter Deficiency. Proc. Natl. Acad. Sci. USA 119: e2210247119. 36343260
Kong, K.F., A. Aguila, L. Schneper, and K. Mathee. (2010). Pseudomonas aeruginosa β-lactamase induction requires two permeases, AmpG and AmpP. BMC Microbiol 10: 328. 21192796
Kramer, W., H.J. Burger, W.J. Arion, D. Corsiero, F. Girbig, C. Weyland, H. Hemmerle, S. Petry, P. Habermann, and A. Herling. (1999). Identification of protein components of the microsomal glucose 6-phosphate transporter by photoaffinity labelling. J. Biochem. 339: 629-638. 10215602
Krause, G. and K.M. Hinz. (2019). Molecular Mechanisms of Thyroid Hormone Transport by l-Type Amino Acid Transporter. Exp Clin Endocrinol Diabetes. [Epub: Ahead of Print] 31739345
Kravtsova, O., V. Levchenko, C.A. Klemens, T. Rieg, R. Liu, and A. Staruschenko. (2023). Effect of SGLT2 inhibition on salt-induced hypertension in female Dahl SS rats. Sci Rep 13: 19231. 37932290
Krings, E., K. Krumbach, B. Bathe, R. Kelle, V.F. Wendisch, H. Sahm, and L. Eggeling. (2006). Characterization of myo-inositol utilization by Corynebacterium glutamicum: the stimulon, identification of transporters, and influence on L-lysine formation. J. Bacteriol. 188: 8054-8061. 16997948
Kristoficova, I., C. Vilhena, S. Behr, and K. Jung. (2017). BtsT - a novel and specific pyruvate/H+ symporter in Escherichia coli. J. Bacteriol. [Epub: Ahead of Print] 29061664
Kroeger, J.K., K. Hassan, A. Vörös, R. Simm, M. Saidijam, K.E. Bettaney, A. Bechthold, I.T. Paulsen, P.J. Henderson, and A.B. Kolstø. (2015). Bacillus cereus efflux protein BC3310 - a multidrug transporter of the unknown major facilitator family, UMF-2. Front Microbiol 6: 1063. 26528249
Kröger, C., J. Stolz, and T.M. Fuchs. (2010). myo-Inositol transport by Salmonella enterica serovar Typhimurium. Microbiology 156: 128-138. 19833776
Kuczak, M., W. Cieślik, R. Musioł, and A. Mrozek-Wilczkiewicz. (2024). 4-Furanylvinylquinoline derivative as a new scaffold for the design of oxidative stress initiator and glucose transporter inhibitor drugs. Sci Rep 14: 28454. 39557921
Kuehne, A., S. Floerl, and Y. Hagos. (2022). Investigations with Drugs and Pesticides Revealed New Species- and Substrate-Dependent Inhibition by Elacridar and Imazalil in and Organic Cation Transporter OCT2. Int J Mol Sci 23:. 36555439
Kulakova, A.N., L.A. Kulakov, N.V. Akulenko, V.N. Ksenzenko, J.T. Hamilton, and J.P. Quinn. (2001). Structural and functional analysis of the phosphonoacetate hydrolase (phnA) gene region in Pseudomonas fluorescens 23F. J. Bacteriol. 183: 3268-3275. 11344133
Kumar, A., N. Sandhu, P. Kumar, G. Pruthi, J. Singh, S. Kaur, and P. Chhuneja. (2022). Genome-wide identification and in silico analysis of NPF, NRT2, CLC and SLAC1/SLAH nitrate transporters in hexaploid wheat (Triticum aestivum). Sci Rep 12: 11227. 35781289
Kumar, H., J.S. Finer-Moore, X. Jiang, I. Smirnova, V. Kasho, E. Pardon, J. Steyaert, H.R. Kaback, and R.M. Stroud. (2018). Crystal Structure of a ligand-bound LacY-Nanobody Complex. Proc. Natl. Acad. Sci. USA 115: 8769-8774. 30108145
Kumar, H., V. Kasho, I. Smirnova, J.S. Finer-Moore, H.R. Kaback, and R.M. Stroud. (2014). Structure of sugar-bound LacY. Proc. Natl. Acad. Sci. USA 111: 1784-1788. 24453216
Kumar, S., A. Athreya, A. Gulati, R.M. Nair, I. Mahendran, R. Ranjan, and A. Penmatsa. (2021). Structural basis of inhibition of a transporter from Staphylococcus aureus, NorC, through a single-domain camelid antibody. Commun Biol 4: 836. 34226658
Kusuhara, H., T. Sekine, N. Utsunomiya-Tate, M. Tsuda, R. Kojima, S.H. Cha, Y. Sugiyama, Y. Kanai, and H. Endou. (1999). Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain. J. Biol. Chem. 274: 13675-13680. 10224140
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:. 36012135
Lan, Q., Z. Zhao, H. Liao, F. Zheng, Y. Chen, T. Wu, Y. Tian, and J. Pang. (2022). Mutation in Transmembrane Domain 8 of Human Urate Transporter 1 Disrupts Uric Acid Recognition and Transport. ACS Omega 7: 34621-34631. 36188325
Langford, C.K., M.P. Kavanaugh, P.E. Stenberg, M.E. Drew, W. Zhang, and S.M. Landfear. (1995). Functional expression and subcellular localization of a high-Kmhexose transporter from Leishmania donovani. Biochemistry 34: 11814-11821. 7547915
Latunde-Dada, G.O., R.J. Simpson, and A.T. McKie. (2006). Recent advances in mammalian haem transport. Trends Biochem. Sci. 31: 182-188. 16487711
Laurent M., B. Aude and R. Cornelia. (2007). Ferripyochelin uptake genes are involved in pyochelin-mediated signaling in Pseudomonas aeruginosa. Microbiology 153: 1508-1518.
Law, C.J., P.C. Maloney, and D.N. Wang. (2008). Ins and outs of major facilitator superfamily antiporters. Annu. Rev. Microbiol. 62: 289-305. 18537473
Law, C.J., Q. Yang, C. Soudant, P.C. Maloney, and D.N. Wang. (2007). Kinetic evidence is consistent with the rocker-switch mechanism of membrane transport by GlpT. Biochemistry. 46: 12190-12197. 17915951
Lawal, H.O. and D.E. Krantz. (2018). SLC18: Vesicular neurotransmitter transporters for monoamines and acetylcholine. Mol Aspects Med 34: 360-372. 23506877
Lazard, M., S. Blanquet, P. Fisicaro, G. Labarraque, and P. Plateau. (2010). Uptake of selenite by Saccharomyces cerevisiae involves the high and low affinity orthophosphate transporters. J. Biol. Chem. 285: 32029-32037. 20688911
Leach, L.H. and T.A. Lewis. (2006). Identification and characterization of Pseudomonas membrane transporters necessary for utilization of the siderophore pyridine-2,6-bis(thiocarboxylic acid) (PDTC). Microbiology 152: 3157-3166. 17005994
Leal-Cardoso, J.H., F.W. Ferreira-da-Silva, A.N. Coelho-de-Souza, and K.S. da Silva-Alves. (2023). Diabetes-induced electrophysiological alterations on neurosomes in ganglia of peripheral nervous system. Biophys Rev 15: 625-638. 37681090
Leandro, M.J., H. Sychrová, C. Prista, and M.C. Loureiro-Dias. (2011). The osmotolerant fructophilic yeast Zygosaccharomyces rouxii employs two plasma-membrane fructose uptake systems belonging to a new family of yeast sugar transporters. Microbiology 157: 601-608. 21051487
Leandro, M.J., I. Spencer-Martins, and P. Gonçalves. (2008). The expression in Saccharomyces cerevisiae of a glucose/xylose symporter from Candida intermedia is affected by the presence of a glucose/xylose facilitator. Microbiology 154: 1646-1655. 18524919
Leandro, M.J., P. Gonçalves, and I. Spencer-Martins. (2006). Two glucose/xylose transporter genes from the yeast Candida intermedia: first molecular characterization of a yeast xylose-H+ symporter. Biochem. J. 395: 543-549. 16402921
Lechermeier, C.G., F. Zimmer, T.M. Lüffe, K.P. Lesch, M. Romanos, C. Lillesaar, and C. Drepper. (2019). Transcript Analysis of Zebrafish GLUT3 Genes, and , Define Overlapping as Well as Distinct Expression Domains in the Zebrafish () Central Nervous System. Front Mol Neurosci 12: 199. 31507372
Lee, E.-H., C. Rouquette-Loughlin, J.P. Folster, and W.M. Shafer. (2003). FarR regulates the farAB-encoded efflux pump of Neisseria gonorrhoeae via an MtrR regulatory mechanism. J. Bacteriol. 185: 7145-7152. 14645274
Lee, E.E., J. Ma, A. Sacharidou, W. Mi, V.K. Salato, N. Nguyen, Y. Jiang, J.M. Pascual, P.E. North, P.W. Shaul, M. Mettlen, and R.C. Wang. (2015). A Protein Kinase C Phosphorylation Motif in GLUT1 Affects Glucose Transport and is Mutated in GLUT1 Deficiency Syndrome. Mol. Cell 58: 845-853. 25982116
Lee, E.H., S.A. Hill, R. Napier, and W.M. Shafer. (2006). Integration Host Factor is required for FarR repression of the farAB-encoded efflux pump of Neisseria gonorrhoeae. Mol Microbiol. 60: 1381-1400. 16796676
Lee, H., H.J. Kim, D.K. Choi, E.N. Ko, J.H. Choi, Y. Seo, S. Lee, S.H. Kim, S. Jung, M. Kim, D. Kang, C.Y. Im, G.H. Bae, S.C. Jung, and O.B. Kwon. (2023). Dopaminergic cell protection and alleviation of neuropsychiatric disease symptoms by VMAT2 expression through the class I HDAC inhibitor TC-H 106. Pharmacol Res Perspect 11: e01135. 37740715
Lee, L.F., Y.J. Chen, R. Kirby, C. Chen, and C.W. Chen. (2007). A multidrug efflux system is involved in colony growth in Streptomyces lividans. Microbiology. 153: 924-934. 17379703
Lee, T., Y.K. Han, K.H. Kim, S.H. Yun, and Y.W. Lee. (2002). Tri13 and Tri7 determine deoxynivalenol- and nivalenol-producing chemotypes of Gibberella zeae. Appl. Environ. Microbiol. 68: 2148-2154. 11976083
Leeb, T., F. Leuthard, V. Jagannathan, S. Kiener, A. Letko, P. Roosje, M.M. Welle, K.L. Gailbreath, A. Cannon, M. Linek, F. Banovic, T. Olivry, S.D. White, K. Batcher, D. Bannasch, K.M. Minor, J.R. Mickelson, M.K. Hytönen, H. Lohi, E.A. Mauldin, and M.L. Casal. (2020). A Missense Variant Affecting the C-Terminal Tail of UNC93B1 in Dogs with Exfoliative Cutaneous Lupus Erythematosus (ECLE). Genes (Basel) 11:. 32028618
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. 36652494
Lei, M., Q. Cheng, Y. Zhao, T. Liu, X. Wang, Y. Deng, J. Yang, and Z. Zhang. (2012). [Expression and Its Clinical Significance of SLC22A18 in Non-small Cell Lung Cancer]. Zhongguo Fei Ai Za Zhi 15: 17-20. 22237119
Lekholm, E., E. Perland, M.M. Eriksson, S.V. Hellsten, F.A. Lindberg, J. Rostami, and R. Fredriksson. (2017). Putative Membrane-Bound Transporters MFSD14A and MFSD14B Are Neuron.al and Affected by Nutrient Availability. Front Mol Neurosci 10: 11. 28179877
Lelandais-Brière, C., M. Jovanovic, G.A. Torres, Y. Perrin, R. Lemoine, F. Corre-Menguy, and C. Hartmann. (2007). Disruption of AtOCT1, an organic cation transporter gene, affects root development and carnitine-related responses in Arabidopsis. Plant J. 51: 154-164. 17521409
Lemieux, M.J. (2007). Eukaryotic major facilitator superfamily transporter modeling based on the prokaryotic GlpT crystal structure. Mol. Membr. Biol. 24: 333-341. 17710637
Lemieux, M.J., Y. Huang, and d.a.N. Wang. (2005). Crystal structure and mechanism of GlpT, the glycerol-3-phosphate transporter from E. coli. J Electron Microsc (Tokyo) 54Suppl1: i43-46. 16157640
Lesuisse, E., M. Simon-Casteras, and P. Labbe. (1998). Siderophore-mediated iron uptake in Saccharomyces cerevisiae: the SIT1 gene encodes a ferrioxamine B permease that belongs to the major facilitator superfamily. Microbiology 144: 3455-3462. 9884238
Lewinson O., J. Adler, N. Sigal, E. Bibi. (2006). Promiscuity in multidrug recognition and transport: the bacterial MFS Mdr transporters. Mol. Microbiol. 61: 277-284. 16856936
Lewinson, O., and E. Bibi. (2001). Evidence for simultaneous binding of dissimilar substrates by the Escherichia coli multidrug transporter MdfA. Biochem. 40: 12612-12618. 11601985
Lewis, J.A., A.R. Horswill, B.E. Schwem, and J.C. Escalante-Semerena. (2004). The tricarballylate utilization (tcuRABC) genes of Salmonella enterica serovar Typhimurium LT2. J. Bacteriol. 186: 1629-1637. 14996793
Leyn, S.A., F. Gao, C. Yang, and D.A. Rodionov. (2012). N-acetylgalactosamine utilization pathway and regulon in proteobacteria: genomic reconstruction and experimental characterization in Shewanella. J. Biol. Chem. 287: 28047-28056. 22711537
Li H., Fan R., Li L., Wei B., Li G., Gu L., Wang X. and Zhang X. (2014). Identification and characterization of a novel copper transporter gene family TaCT1 in common wheat. Plant Cell Environ. 37(7):1561-73. 24372025
Li, D., Y. Ge, N. Wang, Y. Shi, G. Guo, Q. Zou, and Q. Liu. (2023). Identification and Characterization of a Novel Major Facilitator Superfamily Efflux Pump, SA09310, Mediating Tetracycline Resistance in Staphylococcus aureus. Antimicrob. Agents Chemother. e0169622. [Epub: Ahead of Print] 36951566
Li, F., J. Eriksen, J. Finer-Moore, R. Chang, P. Nguyen, A. Bowen, A. Myasnikov, Z. Yu, D. Bulkley, Y. Cheng, R.H. Edwards, and R.M. Stroud. (2020). Ion transport and regulation in a synaptic vesicle glutamate transporter. Science 368: 893-897. 32439795
Li, F., J. Eriksen, J. Finer-Moore, R.M. Stroud, and R.H. Edwards. (2022). Diversity of function and mechanism in a family of organic anion transporters. Curr. Opin. Struct. Biol. 75: 102399. 35660266
Li, H., C. Yang, J. Zhang, W. Zhong, L. Zhu, and Y. Chen. (2020). Identification of potential key mRNAs and LncRNAs for psoriasis by bioinformatic analysis using weighted gene co-expression network analysis. Mol. Genet. Genomics 295: 741-749. 32125527
Li, P., J. Ying, G. Yang, A. Li, J. Wang, J. Lu, J. Wang, T. Xu, H. Yi, K. Li, S. Jin, Q. Bao, and K. Zhang. (2016). Structure-Function Analysis of the Transmembrane Protein AmpG from Pseudomonas aeruginosa. PLoS One 11: e0168060. 27959942
Li, Q., X. Qin, X. Kou, J. Li, Z. Li, and C. Chen. (2022). Anagliptin promotes apoptosis in mouse colon carcinoma cells via MCT-4/lactate-mediated intracellular acidosis. Exp Ther Med 23: 282. 35317435
Li, R., R. Kumar, S. Tati, S. Puri, and M. Edgerton. (2013). Candida albicans flu1-mediated efflux of salivary histatin 5 reduces its cytosolic concentration and fungicidal activity. Antimicrob. Agents Chemother. 57: 1832-1839. 23380720
Li, S., J. Liu, Z. Li, L. Wang, W. Gao, Z. Zhang, and C. Guo. (2020). Sodium-dependent glucose transporter 1 and glucose transporter 2 mediate intestinal transport of quercetrin in Caco-2 cells. Food Nutr Res 64:. 32612490
Li, X., C. Brunner, Y. Wu, O. Leka, G. Schneider, and R.A. Kammerer. (2020). Structural insights into the interaction of botulinum neurotoxin a with its neuronal receptor SV2C. Toxicon 175: 36-43. 31783045
Li, X., H. Yang, D. Zhang, X. Li, H. Yu, and Z. Shen. (2015). Overexpression of specific proton motive force-dependent transporters facilitate the export of surfactin in Bacillus subtilis. J Ind Microbiol Biotechnol 42: 93-103. 25366377
Li, X., Q. Li, X. Jiang, S. Song, W. Zou, Q. Yang, S. Liu, S. Chen, and C. Wang. (2024). Inhibition of SGLT2 protects podocytes in diabetic kidney disease by rebalancing mitochondria-associated endoplasmic reticulum membranes. Cell Commun Signal 22: 534. 39511548
Li, X., W. Wang, J. Yan, and F. Zeng. (2021). Glutamic Acid Transporters: Targets for Neuroprotective Therapies in Parkinson''s Disease. Front Neurosci 15: 678154. 34220434
Li, X., Y. Yang, C. Zhan, Z. Zhang, X. Liu, H. Liu, and Z. Bai. (2017). Transcriptional analysis of impacts of glycerol transporter 1 on methanol and glycerol metabolism in Pichia pastoris. FEMS Yeast Res. [Epub: Ahead of Print] 29092019
Li, X.T., L.C. Thomason, J.A. Sawitzke, N. Costantino, and D.L. Court. (2013). Positive and negative selection using the tetA-sacB cassette: recombineering and P1 transduction in Escherichia coli. Nucleic Acids Res 41: e204. 24203710
Li, X.Z., L. Zhang, and H. Nikaido. (2004). Efflux pump-mediated intrinsic drug resistance in Mycobacterium smegmatis. Antimicrob. Agents Chemother. 48: 2415-2423. 15215089
Li, Y., Y. Ge, M. Zhao, F. Ding, X. Wang, Z. Shi, X. Ge, X. Wang, and X. Qian. (2023). HSP90B1-mediated plasma membrane localization of GLUT1 promotes radioresistance of glioblastomas. J Biomed Res 326-339. [Epub: Ahead of Print] 37750323
Li, Z. (2019). Further insights into cardiovascular outcomes in diabetic and non-diabetic states: inhibition of sodium-glucose co-transports. Cardiovasc Endocrinol Metab 8: 90-95. 31942549
Li, Z., Q. Wu, S. Sun, J. Wu, J. Li, Y. Zhang, C. Wang, J. Yuan, and S. Sun. (2018). Monocarboxylate transporters in breast cancer and adipose tissue are novel biomarkers and potential therapeutic targets. Biochem. Biophys. Res. Commun. 501: 962-967. 29775610
Lian, J., Y. Li, M. HamediRad, and H. Zhao. (2014). Directed evolution of a cellodextrin transporter for improved biofuel production under anaerobic conditions in Saccharomyces cerevisiae. Biotechnol Bioeng 111: 1521-1531. 24519319
Liang, H., A. Mokrani, H. Chisomo-Kasiya, O.M. Wilson-Arop, H. Mi, K. Ji, X. Ge, and M. Ren. (2018). Molecular characterization and identification of facilitative glucose transporter 2 (GLUT2) and its expression and of the related glycometabolism enzymes in response to different starch levels in blunt snout bream (Megalobrama amblycephala). Fish Physiol Biochem. [Epub: Ahead of Print] 29560575
Liang, H., S. Maulu, K. Ji, X. Ge, M. Ren, and H. Mi. (2020). Functional Characterization of Facilitative Glucose Transporter 4 With a Delay Responding to Plasma Glucose Level in Blunt Snout Bream (). Front Physiol 11: 582785. 33178047
Liang, X., F. Yan, Y. Gao, M. Xiong, H. Wang, K. Onxayvieng, R. Tang, L. Li, X. Zhang, W. Chi, M. Piria, M.M. Fuka, A. Gavrilović, and D. Li. (2020). Sugar transporter genes in grass carp (Ctenopharyngodon idellus): molecular cloning, characterization, and expression in response to different stocking densities. Fish Physiol Biochem. [Epub: Ahead of Print] 32062828
Lin, H.C., P.L. Yu, L.H. Chen, H.C. Tsai, and K.R. Chung. (2018). A Major Facilitator Superfamily Transporter Regulated by the Stress-Responsive Transcription Factor Yap1 Is Required for Resistance to Fungicides, Xenobiotics, and Oxidants and Full Virulence in. Front Microbiol 9: 2229. 30279684
Lin, M.W., C.I. Chen, T.T. Cheng, C.C. Huang, J.W. Tsai, G.M. Feng, T.Z. Hwang, and C.F. Lam. (2021). Prolonged preoperative fasting induces postoperative insulin resistance by ER-stress mediated Glut4 down-regulation in skeletal muscles. Int J Med Sci 18: 1189-1197. 33526980
Lin, Y., M. Bogdanov, S. Tong, Z. Guan, and L. Zheng. (2016). Substrate Selectivity of Lysophospholipid Transporter LplT Involved in Membrane Phospholipid Remodeling in Escherichia coli. J. Biol. Chem. 291: 2136-2149. 26613781
Lin, Y., R.N.V.K. Deepak, J.Z. Zheng, H. Fan, and L. Zheng. (2018). A dual substrate-accessing mechanism of a major facilitator superfamily protein facilitates lysophospholipid flipping across the cell membrane. J. Biol. Chem. [Epub: Ahead of Print] 30373772
Lindquist, S., K. Weston-Hafer, H. Schmidt, C. Pul, G. Korfmann, J. Erickson, C. Sanders, H.H. Martin, and S. Normark. (1993). AmpG, a single transducer in chromosomal β-lactamase induction. Mol. Microbiol. 9: 703-715. 8231804
Liu, B., S. Zhao, X. Wu, X. Wang, Y. Nan, D. Wang, and Q. Chen. (2017). Identification and characterization of phosphate transporter genes in potato. J Biotechnol 264: 17-28. 29055693
Liu, C., J. Su, G.K. Stephen, H. Wang, A. Song, F. Chen, Y. Zhu, S. Chen, and J. Jiang. (2018). Overexpression of Phosphate Transporter Gene Facilitated Pi Uptake and Alternated the Metabolic Profiles of Chrysanthemum Under Phosphate Deficiency. Front Plant Sci 9: 686. 30079072
Liu, H., Y. Wang, C. Wu, S. Schwarz, Z. Shen, B. Jeon, S. Ding, Q. Zhang, and J. Shen. (2012). A novel phenicol exporter gene, fexB, found in enterococci of animal origin. J Antimicrob Chemother 67: 322-325. 22096043
Liu, J., L. Yang, M. Luan, Y. Wang, C. Zhang, B. Zhang, J. Shi, F.G. Zhao, W. Lan, and S. Luan. (2015). A vacuolar phosphate transporter essential for phosphate homeostasis in Arabidopsis. Proc. Natl. Acad. Sci. USA 112: E6571-6578. 26554016
Liu, J., W.K. Versaw, N. Pumplin, S.K. Gomez, L.A. Blaylock, and M.J. Harrison. (2008). Closely related members of the Medicago truncatula PHT1 phosphate transporter gene family encode phosphate transporters with distinct biochemical activities. J. Biol. Chem. 283: 24673-24681. 18596039
Liu, J.Y., P.F. Miller, J. Willard, and E.R. Olson. (1999). Functional and biochemical characterization of Escherichia coli sugar efflux transporters. J. Biol. Chem. 274: 22977-22984. 10438463
Liu, J.Y., P.F. Miller, M. Gosink, and E.R. Olson. (1999). The identification of a new family of sugar efflux pumps in Escherichia coli. Mol. Microbiol. 31: 1845-1851. 10209755
Liu, K.-H. and Y.-F. Tsay. (2003). Switching between the two action modes of the dual-affinity nitrate transporter CHL1 by phosphorylation. EMBO J. 22: 1005-1013. 12606566
Liu, M., J. Liu, and W.M. Wang. (2012). Isolation and functional analysis of Thmfs1, the first major facilitator superfamily transporter from the biocontrol fungus Trichoderma harzianum. Biotechnol Lett 34: 1857-1862. 22661043
Liu, N., Q. Wang, C. He, and B. An. (2021). CgMFS1, a Major Facilitator Superfamily Transporter, Is Required for Sugar Transport, Oxidative Stress Resistance, and Pathogenicity of from. Curr Issues Mol Biol 43: 1548-1557. 34698108
Liu, Q., H. Dang, Z. Chen, J. Wu, Y. Chen, S. Chen, and L. Luo. (2018). Genome-Wide Identification, Expression, and Functional Analysis of the Sugar Transporter Gene Family in Cassava (Manihot esculenta). Int J Mol Sci 19:. 29587418
Liu, Y., D. Peter, A. Roghani, S. Schuldiner, G.G. Prive, D. Eisenberg, N. Brecha, and R.H. Edwards. (1992). A cDNA that suppresses MPP+ toxicity encodes a vesicular amine transporter. Cell 70: 539-551. 1505023
Liu, Y., F. Liu, K. Iqbal, I. Grundke-Iqbal, and C.X. Gong. (2008). Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease. FEBS Lett. 582: 359-364. 18174027
Liu, Y., P. Dodds, G. Emilion, A.J. Mungall, I. Dunham, S. Beck, R.S. Wells, F.M. Charnock, and T.S. Ganesan. (2002). The human homologue of unc-93 maps to chromosome 6q27 - characterisation and analysis in sporadic epithelial ovarian cancer. BMC Genet 3: 20. 12381271
Liu, Z., E. Boles, and B.P. Rosen. (2004). Arsenic trioxide uptake by hexose permeases in Saccharomyces cerevisiae. J. Biol. Chem. 279: 17312-17318. 14966117
Lo, C.H. and J. Zeng. (2023). Defective lysosomal acidification: a new prognostic marker and therapeutic target for neurodegenerative diseases. Transl Neurodegener 12: 29. 37287072
Locher, K.P., R.B. Bass, and D.C. Rees. (2003). Breaching the barrier. Science 301: 603-604. 12893929
Lodder-Gadaczek, J., V. Gieselmann, and M. Eckhardt. (2013). Vesicular uptake of N-acetylaspartylglutamate is catalysed by sialin (SLC17A5). Biochem. J. 454: 31-38. 23889254
Lofthouse EM., Brooks S., Cleal JK., Hanson MA., Poore KR., O'Kelly IM. and Lewis RM. (2015). Glutamate cycling may drive organic anion transport on the basal membrane of human placental syncytiotrophoblast. J Physiol. 593(20):4549-59. 26277985
Loland, C.J., P. Wellendorph, S.F. Pedersen, and U. Gether. (2023). Transmembrane transporter proteins: Capturing transport in motion. Basic Clin Pharmacol Toxicol. [Epub: Ahead of Print] 37945540
Long, W., R. Panigrahi, P. Panwar, K. Wong, D. O Neill, X.Z. Chen, M.J. Lemieux, and C.I. Cheeseman. (2017). Identification of Key Residues for Urate Specific Transport in Human Glucose Transporter 9 (hSLC2A9). Sci Rep 7: 41167. 28117388
López, D.M., L. Kählau, K.E.J. Jungnickel, C. Löw, and M. Damme. (2020). Characterization of the complex of the lysosomal membrane transporter MFSD1 and its accessory subunit GLMP. FASEB J. [Epub: Ahead of Print] 32959924
López-Errasquín, E., M.T. González-Jaén, C. Callejas, and C. Vázquez. (2006). A novel MFS transporter encoding gene in Fusarium verticillioides probably involved in iron-siderophore transport. Mycol Res 110: 1102-1110. 16938445
López-Igual, R., S. Lechno-Yossef, Q. Fan, A. Herrero, E. Flores, and C.P. Wolk. (2012). A major facilitator superfamily protein, HepP, is involved in formation of the heterocyst envelope polysaccharide in the cyanobacterium Anabaena sp. strain PCC 7120. J. Bacteriol. 194: 4677-4687. 22753066
Lorca, G.L., R.D. Barabote, V. Zlotopolski, C. Tran, B. Winnen, R.N. Hvorup, A.J. Stonestrom, E. Nguyen, L.W. Huang, D.S. Kim, and M.H. Saier, Jr. (2007). Transport capabilities of eleven gram-positive bacteria: comparative genomic analyses. Biochim. Biophys. Acta. 1768: 1342-1366. 17490609
Löscher, W., M. Gillard, Z.A. Sands, R.M. Kaminski, and H. Klitgaard. (2016). Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Beyond. CNS Drugs 30: 1055-1077. 27752944
Löw, C., K. Bartels, T. Lasitza-Male, and H. Hofmann. (2021). Single-Molecule FRET of Membrane Transport Proteins. Chembiochem. [Epub: Ahead of Print] 33945656
Lubitz, D., J.M. Jorge, F. Pérez-García, H. Taniguchi, and V.F. Wendisch. (2016). Roles of export genes cgmA and lysE for the production of L-arginine and L-citrulline by Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 100: 8465-8474. 27350619
Luo, Y., Z. Akhatayeva, C. Mao, F. Jiang, Z. Guo, H. Xu, and X. Lan. (2023). The ovine gene: mRNA expression, InDel mutations, and growth trait associations. Front Vet Sci 10: 1134903. 37138914
Ma, Y., J. Patel, and R.J. Parry. (2000). A novel valanimycin-resistance determinant (vlmF) from Streptomyces viridifaciens MG456-hF10. Microbiology 146(Pt2): 345-352. 10708373
Ma, Y., X. Chen, T. Diao, Y. Leng, X. Lai, and X. Wei. (2022). The Effect of Ferulic Acid-Grafted Chitosan (FA-g-CS) on the Transmembrane Transport of Anthocyanins by and. Foods 11:. 37431047
MacMillan, S.V., D.A. Alexander, D.E. Culham, H.J. Kunte, E.V. Marshall, D. Rochon, and J.M. Wood. (1999). The ion coupling and organic substrate specificities of osmoregulatory transporter ProP in Escherichia coli. Biochim. Biophys. Acta 1420: 30-44. 10446288
Madej, M.G., S.N. Soro, and H.R. Kaback. (2012). Apo-intermediate in the transport cycle of lactose permease (LacY). Proc. Natl. Acad. Sci. USA 109: E2970-2978. 23012238
Magalhães, F., V. Vidgren, L. Ruohonen, and B. Gibson. (2016). Maltose and maltotriose utilisation by group I strains of the hybrid lager yeast Saccharomyces pastorianus. FEMS Yeast Res 16:. 27364826
Mahesan, A., G. Kamila, P. Choudhary, P. Jauhari, B. Chakrabarty, A. Kumar, and S. Gulati. (2023). Novel Gene Mutation: A Rare Case of Delayed Myelination with Dysthyroidism,v Allan-Herndon-Dudley Syndrome. Neurol India 71: 1282-1283. 38174482
Mahrhold, S., A. Rummel, H. Bigalke, B. Davletov, and T. Binz. (2006). The synaptic vesicle protein 2C mediates the uptake of botulinum neurotoxin A into phrenic nerves. FEBS Lett. 580: 2011-2014. 16545378
Majumder, P., S. Ahmed, P. Ahuja, A. Athreya, R. Ranjan, and A. Penmatsa. (2023). Cryo-EM structure of antibacterial efflux transporter QacA from Staphylococcus aureus reveals a novel extracellular loop with allosteric role. EMBO. J. e113418. [Epub: Ahead of Print] 37458117
Majumder, P., S. Khare, A. Athreya, N. Hussain, A. Gulati, and A. Penmatsa. (2019). Dissection of Protonation Sites for Antibacterial Recognition and Transport in QacA, a Multi-Drug Efflux Transporter. J. Mol. Biol. [Epub: Ahead of Print] 30910733
Makuc, J., S. Paiva, M. Schauen, R. Krämer, B. André, M. Casal, C. Leão, and E. Boles. (2001). The putative monocarboxylate permeases of the yeast Saccharomyces cerevisiae do not transport monocarboxylic acids across the plasma membrane. Yeast 18: 1131-1143. 11536335
Mallik, D., S. Pal, and A.S. Ghosh. (2018). Involvement of AmpG in mediating a dynamic relationship between serine β-lactamase induction and biofilm-forming ability of Escherichia coli. FEMS Microbiol. Lett. 365:. 29566229
Mallonee, D.H. and P.B. Hylemon. (1996). Sequencing and expression of a gene encoding a bile acid transporter from Eubacterium sp. strain VPI 12708. J. Bacteriol. 178: 7053-7058. 8955384
Mandal A., Kumar A., Singh A., Lynn AM., Kapoor K. and Prasad R. (2012). A key structural domain of the Candida albicans Mdr1 protein. Biochem J. 445(3):313-22. 22587419
Mandal, A.K. and D.B. Mount. (2019). Interaction Between ITM2B and GLUT9 Links Urate Transport to Neurodegenerative Disorders. Front Physiol 10: 1323. 31695625
Manel, N., F.J. Kim, S. Kinet, N. Taylor, M. Sitbon, and J.-L. Battini. (2003). The ubiquitous glucose transporter GLUT-1 is a receptor for HTLV. Cell 115: 449-459. 14622599
Manolescu, A., A.M. Salas-Burgos, J. Fischbarg, and C.I. Cheeseman. (2005). Identification of a hydrophobic residue as a key determinant of fructose transport by the facilitative hexose transporter SLC2A7 (GLUT7). J. Biol. Chem. 280: 42978-42983. 16186102
Mansfield, T.A., N.P. Schultes, and G.S. Mourad. (2009). AtAzg1 and AtAzg2 comprise a novel family of purine transporters in Arabidopsis. FEBS Lett. 583: 481-486. 19121308
Marchi, E., T. Lodi, and C. Donnini. (2007). KNQ1, a Kluyveromyces lactis gene encoding a transmembrane protein, may be involved in iron homeostasis. FEMS Yeast Res 7: 715-721. 17428309
Mardones, L., V. Ormazabal, X. Romo, C. Jaña, P. Binder, E. Peña, M. Vergara, and F.A. Zúñiga. (2011). The glucose transporter-2 (GLUT2) is a low affinity dehydroascorbic acid transporter. Biochem. Biophys. Res. Commun. 410: 7-12. 21621509
Marger, M.D. and M.H. Saier, Jr. (1993). A major superfamily of transmembrane facilitators that catalyse uniport, symport and antiport. Trends Biochem. Sci. 18: 13-20. 8438231
Mariotta L., Ramadan T., Singer D., Guetg A., Herzog B., Stoeger C., Palacin M., Lahoutte T., Camargo SM. and Verrey F. (2012). T-type amino acid transporter TAT1 (Slc16a10) is essential for extracellular aromatic amino acid homeostasis control. J Physiol. 590(Pt 24):6413-24. 23045339
Marklevitz, J. and L.K. Harris. (2016). Prediction driven functional annotation of hypothetical proteins in the major facilitator superfamily of S. aureus NCTC 8325. Bioinformation 12: 254-262. 28197063
Martín-Venegas, R., M.J. Rodríguez-Lagunas, P.A. Geraert, and R. Ferrer. (2007). Monocarboxylate transporter 1 mediates DL-2-Hydroxy-(4-methylthio)butanoic acid transport across the apical membrane of Caco-2 cell monolayers. J. Nutr. 137: 49-54. 17182800
Martin, V.J. and W.W. Mohn. (2000). Genetic investigation of the catabolic pathway for degradation of abietane diterpenoids by Pseudomonas abietaniphila BKME-9. J. Bacteriol. 182: 3784-3793. 10850995
Martinez-Garriga, B., T. Vinuesa, J. Hernandez-Borrell, and M. Viñas. (2007). The contribution of efflux pumps to quinolone resistance in Streptococcus pneumoniae clinical isolates. Int. J. Med. Microbiol. 297: 187-195. 17350332
Martinez-Jéhanne, V., L. du Merle, C. Bernier-Fébreau, C. Usein, A. Gassama-Sow, A.A. Wane, M. Gouali, M. Damian, A. Aïdara-Kane, Y. Germani, A. Fontanet, B. Coddeville, Y. Guérardel, and C. Le Bouguénec. (2009). Role of deoxyribose catabolism in colonization of the murine intestine by pathogenic Escherichia coli strains. Infect. Immun. 77: 1442-1450. 19168744
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. 20595258
Maruyama, S.Y., M. Ito, Y. Ikami, Y. Okitsu, C. Ito, K. Toshimori, W. Fujii, and K. Yogo. (2016). A critical role of solute carrier 22a14 in sperm motility and male fertility in mice. Sci Rep 6: 36468. 27811987
Massa López, D., M. Thelen, F. Stahl, C. Thiel, A. Linhorst, M. Sylvester, I. Hermanns-Borgmeyer, R. Lüllmann-Rauch, W. Eskild, P. Saftig, and M. Damme. (2019). The lysosomal transporter MFSD1 is essential for liver homeostasis and critically depends on its accessory subunit GLMP. Elife 8:. 31661432
Masureel M., Martens C., Stein RA., Mishra S., Ruysschaert JM., Mchaourab HS. and Govaerts C. (2014). Protonation drives the conformational switch in the multidrug transporter LmrP. Nat Chem Biol. 10(2):149-55. 24316739
Matherly LH., Wilson MR. and Hou Z. (2014). The major facilitative folate transporters solute carrier 19A1 and solute carrier 46A1: biology and role in antifolate chemotherapy of cancer. Drug Metab Dispos. 42(4):632-49. 24396145
Matherly, L.H., M. Schneider, A. Gangjee, and Z. Hou. (2022). Biology and therapeutic applications of the proton-coupled folate transporter. Expert Opin Drug Metab Toxicol 1-12. [Epub: Ahead of Print] 36239195
Mathews, E.A., G.P. Mullen, J. Hodgkin, J.S. Duerr, and J.B. Rand. (2012). Genetic interactions between UNC-17/VAChT and a novel transmembrane protein in Caenorhabditis elegans. Genetics 192: 1315-1325. 23051648
Mathiesen, B.K., L.M. Miyakoshi, C.R. Cederroth, E. Tserga, C. Versteegh, P.A.R. Bork, N.L. Hauglund, R.S. Gomolka, Y. Mori, N.K. Edvall, S. Rouse, K. Møllgård, J.R. Holt, M. Nedergaard, and B. Canlon. (2023). Delivery of gene therapy through a cerebrospinal fluid conduit to rescue hearing in adult mice. Sci Transl Med 15: eabq3916. 37379370
Maulén, N.P., E.A. Henríquez, S. Kempe, J.G. Cárcamo, A. Schmid-Kotsas, M. Bachem, A. Grünert, M.E. Bustamante, F. Nualart, and J.C. Vera. (2003). Up-regulation and polarized expression of the sodium-ascorbic acid transporter SVCT1 in post-confluent differentiated CaCo-2 cells. J. Biol. Chem. 278: 9035-9041. 12381735
Mauri, A., A. Duse, G. Palm, R. Previtali, S.M. Bova, S. Olivotto, S. Benedetti, F. Coscia, P. Veggiotti, and C. Cereda. (2022). Molecular Genetics of GLUT1DS Italian Pediatric Cohort: 10 Novel Disease-Related Variants and Structural Analysis. Int J Mol Sci 23:. 36362347
Maurya, P., M. Kumar, R. Jain, H.T. Abdulhameed Almuqdadi, H. Singh, A. Gupta, C. Arenz, N.A. Gaur, and S. Singh. (2024). Expression of major facilitator superfamily protein in transporters - Δ identifies a drug transporter. Future Microbiol 1-15. [Epub: Ahead of Print] 39235058
Mayerl, S., M. Schmidt, D. Doycheva, V.M. Darras, S.S. Hüttner, A. Boelen, T.J. Visser, C. Kaether, H. Heuer, and J. von Maltzahn. (2018). Thyroid Hormone Transporters MCT8 and OATP1C1 Control Skeletal Muscle Regeneration. Stem Cell Reports. [Epub: Ahead of Print] 29706500
McClung, D.J., A. Calixto, M.N. Mosera, R. Kumar, E.L. Neidle, and K.T. Elliott. (2016). Novel heterologous bacterial system reveals enhanced susceptibility to DNA damage mediated by yqgF, a nearly ubiquitous and often essential gene. Microbiology 162: 1808-1821. 27527105
McCulloch, K.A., Y.B. Qi, S. Takayanagi-Kiya, Y. Jin, and S.J. Cherra, 3rd. (2017). Novel Mutations in Synaptic Transmission Genes Suppress Neuron.al Hyperexcitation in. G3 (Bethesda) 7: 2055-2063. 28468816
McGee, S.L. and M. Hargreaves. (2006). Exercise and skeletal muscle glucose transporter 4 expression: molecular mechanisms. Clin Exp Pharmacol Physiol 33: 395-399. 16620308
Meier, A., H. Erler, and E. Beitz. (2018). Targeting Channels and Transporters in Protozoan Parasite Infections. Front Chem 6: 88. 29637069
Meixner, E., U. Goldmann, V. Sedlyarov, S. Scorzoni, M. Rebsamen, E. Girardi, and G. Superti-Furga. (2020). A substrate-based ontology for human solute carriers. Mol Syst Biol 16: e9652. 32697042
Meyer, K., M. Kirchner, B. Uyar, J.Y. Cheng, G. Russo, L.R. Hernandez-Miranda, A. Szymborska, H. Zauber, I.M. Rudolph, T.E. Willnow, A. Akalin, V. Haucke, H. Gerhardt, C. Birchmeier, R. Kühn, M. Krauss, S. Diecke, J.M. Pascual, and M. Selbach. (2018). Mutations in Disordered Regions Can Cause Disease by Creating Dileucine Motifs. Cell. [Epub: Ahead of Print] 30197081
Meyer, M.J., A. Tuerkova, S. Römer, C. Wenzel, T. Seitz, J. Gaedcke, S. Oswald, J. Brockmöller, B. Zdrazil, and M.V. Tzvetkov. (2020). Drug Metab Dispos. [Epub: Ahead of Print] 33037045
Meyer, M.J., P.C.F. Schreier, M. Basaran, S. Vlasova, T. Seitz, J. Brockmöller, B. Zdrazil, and M.V. Tzvetkov. (2022). Amino acids in transmembrane helix 1 confer major functional differences between human and mouse orthologs of the polyspecific membrane transporter OCT1. J. Biol. Chem. 101974. [Epub: Ahead of Print] 35469921
Meyer-Tönnies, M.J. and M.V. Tzvetkov. (2023). The end of the beginning in understanding SLC22 polyspecificity. Trends Pharmacol Sci 44: 397-399. 37117104
Michel, L., A. Bachelard, and C. Reimmann. (2007). Ferripyochelin uptake genes are involved in pyochelin-mediated signalling in Pseudomonas aeruginosa. Microbiology 153: 1508-1518. 17464065
Miethke, M., S. Schmidt, and M.A. Marahiel. (2008). The major facilitator superfamily-type transporter YmfE and the multidrug-efflux activator Mta mediate bacillibactin secretion in Bacillus subtilis. J. Bacteriol. 190: 5143-5152. 18502870
Miller, A.J., X. Fan, M. Orsel, S.J. Smith, and D.M. Wells. (2007). Nitrate transport and signalling. J Exp Bot 58: 2297-306. 17519352
Miura, D., N. Anzai, P. Jutabha, S. Chanluang, X. He, T. Fukutomi, and H. Endou. (2011). Human urate transporter 1 (hURAT1) mediates the transport of orotate. J. Physiol. Sci 61: 253-257. 21350910
Miyaji, T., T. Kuromori, Y. Takeuchi, N. Yamaji, K. Yokosho, A. Shimazawa, E. Sugimoto, H. Omote, J.F. Ma, K. Shinozaki, and Y. Moriyama. (2015). AtPHT4;4 is a chloroplast-localized ascorbate transporter in Arabidopsis. Nat Commun 6: 5928. 25557369
Monte, J.C., M.A. Nagle, S.A. Eraly, and S.K. Nigam. (2004). Identification of a novel murine organic anion transporter family member, OAT6, expressed in olfactory mucosa. Biochem. Biophys. Res. Commun. 323: 429-436. 15369770
Montel-Hagen, A., S. Kinet, N. Manel, C. Mongellaz, R. Prohaska, J.L. Battini, J. Delaunay, M. Sitbon, and N. Taylor. (2008). Erythrocyte Glut1 triggers dehydroascorbic acid uptake in mammals unable to synthesize vitamin C. Cell 132: 1039-1048. 18358815
Moore, R.E., Y. Kim, and C.C. Philpott. (2003). The mechanism of ferrichrome transport through Arn1p and its metabolism in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA 100: 5664-5669. 12721368
Mor, A.L., T.W. Kaminski, M. Karbowska, and D. Pawlak. (2018). New insight into organic anion transporters from the perspective of potentially important interactions and drugs toxicity. J. Physiol. Pharmacol 69:. 30149367
Moraes, T.F. and R.A. Reithmeier. (2012). Membrane transport metabolons. Biochim. Biophys. Acta. 1818: 2687-2706. 22705263
Moraleda-Muñoz, A., J. Pérez, A.L. Extremera, and J. Muñoz-Dorado. (2010). Differential regulation of six heavy metal efflux systems in the response of Myxococcus xanthus to copper. Appl. Environ. Microbiol. 76: 6069-6076. 20562277
Morin, P., C. Sagné, and B. Gasnier. (2004). Functional characterization of wild-type and mutant human sialin. EMBO. J. 23: 4560-4570. 15510212
Morishita, M. and J. Engebrecht. (2008). Sorting signals within the Saccharomyces cerevisiae sporulation-specific dityrosine transporter, Dtr1p, C terminus promote Golgi-to-prospore membrane transport. Eukaryot. Cell. 7: 1674-1684. 18676951
Morrice, N., S. Vainio, K. Mikkola, L. van Aalten, J.R. Gallagher, M.L.J. Ashford, A.D. McNeilly, R.J. McCrimmon, A. Grosfeld, P. Serradas, J. Koffert, E.R. Pearson, P. Nuutila, and C. Sutherland. (2023). Metformin increases the uptake of glucose into the gut from the circulation in high-fat diet-fed male mice, which is enhanced by a reduction in whole-body Slc2a2 expression. Mol Metab 77: 101807. [Epub: Ahead of Print] 37717665
Moschen I., Broer A., Galic S., Lang F. and Broer S. (2012). Significance of short chain fatty acid transport by members of the monocarboxylate transporter family (MCT). Neurochem Res. 37(11):2562-8. 22878645
Mueckler, M. and C. Makepeace. (2004). Analysis of transmembrane segment 8 of the GLUT1 glucose transporter by cysteine-scanning mutagenesis and substituted cysteine accessibility. J. Biol. Chem. 279: 10494-10499. 14688257
Mueckler, M. and C. Makepeace. (2009). Model of the exofacial substrate-binding site and helical folding of the human Glut1 glucose transporter based on scanning mutagenesis. Biochemistry 48: 5934-5942. 19449892
Murphy, P.J., S.P. Trenz, W. Grzemski, F.J. De Bruijn, and J. Schell. (1993). The Rhizobium meliloti rhizopine mos locus is a mosaic structure facilitating its symbiotic regulation. J. Bacteriol. 175: 5193-5204. 8349559
Naem, A.A.A., M.N. Al-Terehi, F.A. Ghafil, F.S. Ataya, G.E. Batiha, A. Alexiou, M. Papadakis, N.N. Welson, and N.R. Hadi. (2024). The Influence of OCT3 and MATE2 Genetic Polymorphisms in Poor Response to Metformin in Type 2 Diabetes Mellitus. Endocrinol Diabetes Metab 7: e486. 39086121
Naftalin, R.J. (2008). Osmotic water transport with glucose in GLUT2 and SGLT. Biophys. J. 94: 3912-3923. 18234816
Nagakubo, S., K. Nishino, T. Hirata, and A. Yamaguchi. (2002). The putative response regulator BaeR stimulates multidrug resistance of Escherichia coli via a novel multidrug exporter system, MdtABC. J. Bacteriol. 184: 4161-4167. 12107133
Nagarathinam, K., F. Jaenecke, Y. Nakada-Nakura, Y. Hotta, K. Liu, S. Iwata, M.T. Stubbs, N. Nomura, and M. Tanabe. (2017). The multidrug-resistance transporter MdfA from Escherichia coli: crystallization and X-ray diffraction analysis. Acta Crystallogr F Struct Biol Commun 73: 423-430. 28695852
Naidu, V., B. Shah, C. Maher, I.T. Paulsen, and K.A. Hassan. (2023). AadT, a new weapon in fight against antibiotics. Microbiology (Reading) 169:. 37252766
Nair, A.V., H. Singh, S. Raturi, A. Neuberger, Z. Tong, N. Ding, K. Agboh, and H.W. van Veen. (2016). Relocation of active site carboxylates in major facilitator superfamily multidrug transporter LmrP reveals plasticity in proton interactions. Sci Rep 6: 38052. 27917857
Nakano, Y., K. Fujitani, J. Kurihara, J. Ragan, K. Usui-Aoki, L. Shimoda, T. Lukacsovich, K. Suzuki, M. Sezaki, Y. Sano, R. Ueda, W. Awano, M. Kaneda, M. Umeda, and D. Yamamoto. (2001). Mutations in the novel membrane protein spinster interfere with programmed cell death and cause neural degeneration in Drosophila melanogaster. Mol. Cell. Biol. 21: 3775-3788. 11340170
Nakata, T., T. Matsui, K. Kobayashi, Y. Kobayashi, and N. Anzai. (2013). Organic cation transporter 2 (SLC22A2), a low-affinity and high-capacity choline transporter, is preferentially enriched on synaptic vesicles in cholinergic neurons. Neuroscience 252: 212-221. 23958595
Nakayama, Y., N. Mukai, G. Kreitzer, P. Patwari, and J. Yoshioka. (2022). Interaction of ARRDC4 With GLUT1 Mediates Metabolic Stress in the Ischemic Heart. Circ Res 131: 510-527. 35950500
Nancolas, B., R.B. Sessions, and A.P. Halestrap. (2015). Identification of key binding site residues of MCT1 for AR-C155858 reveals the molecular basis of its isoform selectivity. Biochem. J. 466: 177-188. 25437897
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. 25162599
Naula, C.M., F.M. Logan, P.E. Wong, M.P. Barrett, and R.J. Burchmore. (2010). A glucose transporter can mediate ribose uptake: definition of residues that confer substrate specificity in a sugar transporter. J. Biol. Chem. 285: 29721-29728. 20601430
Nazish, I., A. Mamais, A. Mallach, C. Bettencourt, A. Kaganovich, T. Warner, J. Hardy, P.A. Lewis, J. Pocock, M.R. Cookson, and R. Bandopadhyay. (2023). Differential LRRK2 signalling and gene expression in WT-LRRK2 and G2019S-LRRK2 mouse microglia treated with zymosan and MLi2. bioRxiv. 37745519
Nałęcz, K.A. and M.J. Nałęcz. (2017). [Carnitine - mitochondria and beyond]. Postepy Biochem 62: 85-93. 28132459
Nele Bourgeois, M.A., S.L. Van Herck, P. Vancamp, J. Delbaere, C. Zevenbergen, S. Kersseboom, V.M. Darras, and T.J. Visser. (2016). CHARACTERIZATION OF CHICKEN THYROID HORMONE TRANSPORTERS. Endocrinology en20152025. [Epub: Ahead of Print] 27070099
Nemoz, G., J. Robert-Baudouy, and F. Stoeber. (1976). Physiological and genetic regulation of the aldohexuronate transport system in Escherichia coli. J. Bacteriol. 127: 706-718. 783117
Nesper, J., A. Brosig, P. Ringler, G.J. Patel, S.A. Müller, J.H. Kleinschmidt, W. Boos, K. Diederichs, and W. Welte. (2008). Omp85(Tt) from Thermus thermophilus HB27: an ancestral type of the Omp85 protein family. J. Bacteriol. 190: 4568-4575. 18456816
Ng, T.S., S.Y. Chew, P. Rangasamy, M.N. Mohd Desa, D. Sandai, P.P. Chong, and L.T. Than. (2015). SNF3 as High Affinity Glucose Sensor and Its Function in Supporting the Viability of Candida glabrata under Glucose-Limited Environment. Front Microbiol 6: 1334. 26648919
Nguyen, A.Q., J. Schneider, and V.F. Wendisch. (2015). Elimination of polyamine N-acetylation and regulatory engineering improved putrescine production by Corynebacterium glutamicum. J Biotechnol 201: 75-85. 25449016
Nichols, N.N. and C.S. Harwood. (1997). PcaK, a high-affinity permease for the aromatic compounds 4-hydroxybenzoate and protocatechuate from Pseudomonas putida. J. Bacteriol. 179: 5056-5061. 9260946
Nigam, S.K. (2015). What do drug transporters really do? Nat Rev Drug Discov 14: 29-44. 25475361
Nijnik, A., S. Clare, C. Hale, J. Chen, C. Raisen, L. Mottram, M. Lucas, J. Estabel, E. Ryder, H. Adissu, , N.C. Adams, R. Ramirez-Solis, J.K. White, K.P. Steel, G. Dougan, and R.E. Hancock. (2012). The role of sphingosine-1-phosphate transporter Spns2 in immune system function. J Immunol 189: 102-111. 22664872
Nilsson, D., M. Pettersson, P. Gustavsson, A. Förster, W. Hofmeister, J. Wincent, V. Zachariadis, B.M. Anderlid, A. Nordgren, O. Mäkitie, V. Wirta, M. Käller, F. Vezzi, J.R. Lupski, M. Nordenskjöld, E.S. Lundberg, C.M.B. Carvalho, and A. Lindstrand. (2017). Whole-Genome Sequencing of Cytogenetically Balanced Chromosome Translocations Identifies Potentially Pathological Gene Disruptions and Highlights the Importance of Microhomology in the Mechanism of Formation. Hum Mutat 38: 180-192. 27862604
Nishino, K. and A. Yamaguchi. (2001). Analysis of a complete library of putative drug transporter genes in Escherichia coli. J. Bacteriol. 183: 5803-5812. 11566977
Niu, C., G.A. Payne, and C.P. Woloshuk. (2016). Involvement of FST1 from Fusarium verticillioides in virulence and transport of inositol. Mol Plant Pathol. [Epub: Ahead of Print] 27195938
Niu, Y., R. Liu, C. Guan, Y. Zhang, Z. Chen, S. Hoerer, H. Nar, and L. Chen. (2021). Structural basis of inhibition of the human SGLT2-MAP17 glucose transporter. Nature. [Epub: Ahead of Print] 34880493
Nobre, A., C. Lucas, and C. Leão. (1999). Transport and utilization of hexoses and pentoses in the halotolerant yeast Debaryomyces hansenii. Appl. Environ. Microbiol. 65: 3594-3598. 10427054
Nomura N., Verdon G., Kang HJ., Shimamura T., Nomura Y., Sonoda Y., Hussien SA., Qureshi AA., Coincon M., Sato Y., Abe H., Nakada-Nakura Y., Hino T., Arakawa T., Kusano-Arai O., Iwanari H., Murata T., Kobayashi T., Hamakubo T., Kasahara M., Iwata S. and Drew D. (2015). Structure and mechanism of the mammalian fructose transporter GLUT5. Nature. 526(7573):397-401. 26416735
Norholm, M.H., H.H. Nour-Eldin, P. Brodersen, J. Mundy, and B.A. Halkier. (2006). Expression of the Arabidopsis high-affinity hexose transporter STP13 correlates with programmed cell death. FEBS Lett. 17: 2381-2387. 16616142
Normant, V., A. Brault, M. Avino, T. Mourer, T. Vahsen, J. Beaudoin, and S. Labbé. (2020). Hemeprotein Tpx1 interacts with cell-surface heme transporter Str3 in Schizosaccharomyces pombe. Mol. Microbiol. [Epub: Ahead of Print] 33140466
Notariale, R., E. Längst, P. Perrone, D. Crettaz, M. Prudent, and C. Manna. (2022). Effect of Mercury on Membrane Proteins, Anionic Transport and Cell Morphology in Human Erythrocytes. Cell Physiol Biochem 56: 500-513. 36126286
Nunvar, J., A.M. Hogan, S. Buroni, S. Savina, V. Makarov, S.T. Cardona, and P. Drevinek. (2019). The Effect of 2-Thiocyanatopyridine Derivative 11026103 on : Resistance Mechanisms and Systemic Impact. Antibiotics (Basel) 8:. 31546596
Nygaard, P. and H.H. Saxild. (2005). The purine efflux pump PbuE in Bacillus subtilis modulates expression of the PurR and G-box (XptR) regulons by adjusting the purine base pool size. J. Bacteriol. 187: 791-794. 15629952
Nørholm, M.H. and G. Dandanell. (2001). Specificity and topology of the Escherichia coli xanthosine permease, a representative of the NHS subfamily of the major facilitator superfamily. J. Bacteriol. 183: 4900-4904. 11466294
Ogasawara, H., S. Ohe, and A. Ishihama. (2015). Role of transcription factor NimR (YeaM) in sensitivity control of Escherichia coli to 2-nitroimidazole. FEMS Microbiol. Lett. 362: 1-8. 25790494
Ohki, R. and K. Tateno. (2004). Increased stability of bmr3 mRNA results in a multidrug-resistant phenotype in Bacillus subtilis. J. Bacteriol. 186: 7450-7455. 15489457
Ohki, R. and M. Murata. (1997). bmr3, a third multidrug transporter gene of Bacillus subtilis. J. Bacteriol. 179: 1423-1427. 9023234
Ojeda, A.M., N.G. Kolmakova, and S.M. Parsons. (2004). Acetylcholine binding site in the vesicular acetylcholine transporter. Biochemistry 43: 11163-11174. 15366926
Oku, Y., K. Kurokawa, N. Ichihashi, and K. Sekimizu. (2004). Characterization of the Staphylococcus aureus mprF gene, involved in lysinylation of phosphatidylglycerol. Microbiology 150: 45-51. 14702396
Oliveira, I.H.R., H.C.P. Figueiredo, C.P. Rezende, T. Verano-Braga, M.N. Melo-Braga, J.L. Reis Cunha, and H.M. de Andrade. (2020). Assessing the composition of the plasma membrane of Leishmania (Leishmania) infantum and L. (L.) amazonensis using label-free proteomics. Exp Parasitol 218: 107964. [Epub: Ahead of Print] 32822697
Oliveira-Tintino, C.D.M., D.F. Muniz, C.R.D.S. Barbosa, R.L.S. Pereira, I.M. Begnini, R.A. Rebelo, L.E.D. Silva, S.L. Mireski, M.C. Nasato, M.I.L. Krautler, P.S. Pereira, J.G.M.D. Costa, F.F.G. Rodrigues, A.M.R. Teixeira, J. Ribeiro-Filho, S.R. Tintino, I.R.A. de Menezes, H.D.M. Coutinho, and T.G.D. Silva. (2021). The 1,8-naphthyridines sulfonamides are NorA efflux pump inhibitors. J Glob Antimicrob Resist 24: 233-240. 33385589
Omote, H., T. Miyaji, N. Juge, and Y. Moriyama. (2011). Vesicular neurotransmitter transporter: bioenergetics and regulation of glutamate transport. Biochemistry 50: 5558-5565. 21612282
Ostash, I., B. Ostash, A. Luzhetskyy, A. Bechthold, S. Walker, and V. Fedorenko. (2008). Coordination of export and glycosylation of landomycins in Streptomyces cyanogenus S136. FEMS Microbiol. Lett. 285: 195-202. 18537830
Ottosson, F., F. Russo, A. Abrahamsson, N. MacSween, J. Courraud, K. Skogstrand, O. Melander, U. Ericson, M. Orho-Melander, A.S. Cohen, J. Grove, P.B. Mortensen, D.M. Hougaard, and M. Ernst. (2024). Unraveling the metabolomic architecture of autism in a large Danish population-based cohort. BMC Med 22: 302. 39026322
Ovens, M.J., C. Manoharan, M.C. Wilson, C.M. Murray, and A.P. Halestrap. (2010). The inhibition of monocarboxylate transporter 2 (MCT2) by AR-C155858 is modulated by the associated ancillary protein. Biochem. J. 431: 217-225. 20695846
Ozcan, S. and M. Johnston. (1999). Function and regulation of yeast hexose transporters. Microbiol. Mol. Biol. Rev. 63: 554-569. 10477308
Ozturk, T.N., C. Coumoundouros, D.E. Culham, and J.M. Wood. (2023). Structural Determinants and Functional Significance of Dimerization for Osmosensing Transporter ProP in. Biochemistry 62: 118-133. 36516499
Ozturk, T.N., D.E. Culham, L. Tempelhagen, J.M. Wood, and G. Lamoureux. (2020). Salt-Dependent Interactions between the C-Terminal Domain of Osmoregulatory Transporter ProP of and the Lipid Membrane. J Phys Chem B 124: 8209-8220. 32838524
Périchon, B., P. Bogaerts, T. Lambert, L. Frangeul, P. Courvalin, and M. Galimand. (2008). Sequence of conjugative plasmid pIP1206 mediating resistance to aminoglycosides by 16S rRNA methylation and to hydrophilic fluoroquinolones by efflux. Antimicrob. Agents Chemother. 52: 2581-2592. 18458128
Périchon, B., P. Courvalin, and M. Galimand. (2007). Transferable resistance to aminoglycosides by methylation of G1405 in 16S rRNA and to hydrophilic fluoroquinolones by QepA-mediated efflux in Escherichia coli. Antimicrob. Agents Chemother. 51: 2464-2469. 17470656
Packer, M., C.S. Wilcox, and J.M. Testani. (2023). Critical Analysis of the Effects of SGLT2 Inhibitors on Renal Tubular Sodium, Water and Chloride Homeostasis and Their Role in Influencing Heart Failure Outcomes. Circulation 148: 354-372. 37486998
Pamp, S.J., M. Gjermansen, H.K. Johansen, and T. Tolker-Nielsen. (2008). Tolerance to the antimicrobial peptide colistin in Pseudomonas aeruginosa biofilms is linked to metabolically active cells, and depends on the pmr and mexAB-oprM genes. Mol. Microbiol. 68: 223-240. 18312276
Pan PY., Marrs J. and Ryan TA. (2015). Vesicular glutamate transporter 1 orchestrates recruitment of other synaptic vesicle cargo proteins during synaptic vesicle recycling. J Biol Chem. 290(37):22593-601. 26224632
Pan, C.J., S.Y. Chen, H.S. Jun, S.R. Lin, B.C. Mansfield, and J.Y. Chou. (2011). SLC37A1 and SLC37A2 are phosphate-linked, glucose-6-phosphate antiporters. PLoS One 6: e23157. 21949678
Pan, W., R.S. Godoy, D.P. Cook, A.L. Scott, C.A. Nurse, and M.G. Jonz. (2022). Single-cell transcriptomic analysis of neuroepithelial cells and other cell types of the gills of zebrafish (Danio rerio) exposed to hypoxia. Sci Rep 12: 10144. 35710785
Panja, A.S., A. Sarkar, R. Biswas, B. Bandyopadhyay, and R. Bandopadhyay. (2019). Modification of drug-binding proteins associated with the efflux pump in MDR-MTB in course of evolution: an unraveled clue based on in silico approach. J Antibiot (Tokyo) 72: 282-290. 30799437
Pao, S.S., I.T. Paulsen, and M.H. Saier, Jr. (1998). The major facilitator superfamily. Microbiol. Mol. Biol. Rev. 62: 1-32. 9529885
Paolini, R., C. Moore, T. Matthyssen, N. Cirillo, M. McCullough, C.S. Farah, H. Botha, T. Yap, and A. Celentano. (2022). Transcriptional regulation of glucose transporters in human oral squamous cell carcinoma cells. J Oral Pathol Med 51: 679-683. 35920070
Papakonstantinou, E., D. Vlachakis, T. Thireou, P.G. Vlachoyiannopoulos, and E. Eliopoulos. (2021). A Holistic Evolutionary and 3D Pharmacophore Modelling Study Provides Insights into the Metabolism, Function, and Substrate Selectivity of the Human Monocarboxylate Transporter 4 (hMCT4). Int J Mol Sci 22:. 33805725
Parche, S., M. Beleut, E. Rezzonico, D. Jacobs, F. Arigoni, F. Titgemeyer, and I. Jankovic. (2006). Lactose-over-glucose preference in Bifidobacterium longum NCC2705: glcP, encoding a glucose transporter, is subject to lactose repression. J. Bacteriol. 188: 1260-1265. 16452407
Park, J.S., S.J. Lee, H.G. Rhie, and H.S. Lee. (2008). Characterization of a chromosomal nickel resistance determinant from Klebsiella oxytoca CCUG 15788. J Microbiol Biotechnol 18: 1040-1043. 18600044
Park, J.T. and T. Uehara. (2008). How bacteria consume their own exoskeletons (turnover and recycling of cell wall peptidoglycan). Microbiol. Mol. Biol. Rev. 72: 211-27, table of contents. 18535144
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. 9495761
Park, S.J., C.P. Smith, R.R. Wilbur, C.P. Cain, S.R. Kallu, S. Valasapalli, A. Sahoo, M.R. Guda, A.J. Tsung, and K.K. Velpula. (2018). An overview of MCT1 and MCT4 in GBM: small molecule transporters with large implications. Am J Cancer Res 8: 1967-1976. 30416849
Park, T.J., J. Reznick, B.L. Peterson, G. Blass, D. Omerbašić, N.C. Bennett, P.H.J.L. Kuich, C. Zasada, B.M. Browe, W. Hamann, D.T. Applegate, M.H. Radke, T. Kosten, H. Lutermann, V. Gavaghan, O. Eigenbrod, V. Bégay, V.G. Amoroso, V. Govind, R.D. Minshall, E.S.J. Smith, J. Larson, M. Gotthardt, S. Kempa, and G.R. Lewin. (2017). Fructose-driven glycolysis supports anoxia resistance in the naked mole-rat. Science 356: 307-311. 28428423
Pascual, J.M., D. Wang, R. Yang, L. Shi, H. Yang, and D.C. De Vivo. (2008). Structural Signatures and Membrane Helix 4 in GLUT1: INFERENCES FROM HUMAN BLOOD-BRAIN GLUCOSE TRANSPORT MUTANTS. J. Biol. Chem. 283: 16732-16742. 18387950
Patching, S.G., G. Psakis, S.A. Baldwin, J. Baldwin, P.J. Henderson, and D.A. Middleton. (2008). Relative substrate affinities of wild-type and mutant forms of the Escherichia coli sugar transporter GalP determined by solid-state NMR. Mol. Membr. Biol. 25: 474-484. 18798051
Patching, S.G., S.A. Baldwin, A.D. Baldwin, J.D. Young, M.P. Gallagher, P.J. Henderson, and R.B. Herbert. (2005). The nucleoside transport proteins, NupC and NupG, from Escherichia coli: specific structural motifs necessary for the binding of ligands. Org Biomol Chem 3: 462-470. 15678184
Patel, N., H. Alkuraya, S.S. Alzahrani, S.R. Nowailaty, M.Z. Seidahmed, A. Alhemidan, T. Ben-Omran, N.G. Ghazi, A. Al-Aqeel, M. Al-Owain, H.I. Alzaidan, E. Faqeih, W. Kurdi, Z. Rahbeeni, N. Ibrahim, F. Abdulwahab, M. Hashem, R. Shaheen, M. Abouelhoda, D. Monies, A.O. Khan, M.A. Aldahmesh, and F.S. Alkuraya. (2018). Mutations in known disease genes account for the majority of autosomal recessive retinal dystrophies. Clin Genet 94: 554-563. 30054919
Patrick PS., Lyons SK., Rodrigues TB. and Brindle KM. (2014). Oatp1 enhances bioluminescence by acting as a plasma membrane transporter for D-luciferin. Mol Imaging Biol. 16(5):626-34. 24798747
Paul, S., K.O. Alegre, S.R. Holdsworth, M. Rice, J.A. Brown, P. McVeigh, S.M. Kelly, and C.J. Law. (2014). A single-component multidrug transporter of the major facilitator superfamily is part of a network that protects Escherichia coli from bile salt stress. Mol. Microbiol. 92: 872-884. 24684269
Paulsen, I.T., M.H. Brown, and R.A. Skurray. (1996). Proton-dependent multidrug efflux systems. Microbiol. Rev. 60: 575-608. 8987357
Paulsen, I.T., S. Chauvaux, P. Choi, and M.H. Saier, Jr. (1998). Characterization of glucose-specific catabolite repression-resistant mutants of Bacillus subtilis: identification of a novel hexose:H+ symporter. J. Bacteriol. 180: 498-504. 9457850
Paulsen, P.A., T.F. Custódio, and B.P. Pedersen. (2019). Crystal structure of the plant symporter STP10 illuminates sugar uptake mechanism in monosaccharide transporter superfamily. Nat Commun 10: 407. 30679446
Pavón, L.R., F. Lundh, B. Lundin, A. Mishra, B.L. Persson, and C. Spetea. (2008). Arabidopsis ANTR1 is a thylakoid Na+-dependent phosphate transporter: functional characterization in Escherichia coli. J. Biol. Chem. 283: 13520-13527. 18353780
Pearson, S.M., A.G. Griffiths, P. Maclean, A.C. Larking, S.W. Hong, R. Jauregui, P. Miller, C.M. McKenzie, P.J. Lockhart, J.A. Tate, J.L. Ford, and M.J. Faville. (2022). Outlier analyses and genome-wide association study identify and as candidate genes for foliar water-soluble carbohydrate accumulation in. Front Plant Sci 13: 1095359. 36699852
Pedersen, B.P., H. Kumar, A.B. Waight, A.J. Risenmay, Z. Roe-Zurz, B.H. Chau, A. Schlessinger, M. Bonomi, W. Harries, A. Sali, A.K. Johri, and R.M. Stroud. (2013). Crystal structure of a eukaryotic phosphate transporter. Nature 496: 533-536. 23542591
Peekhaus, N. and T. Conway. (1998). What’s for dinner?: Entner-Doudoroff metabolism in Escherichia coli. J. Bacteriol 180: 3495-3502. 9657988
Pegg, A.E. and A.J. Michael. (2010). Spermine synthase. Cell Mol Life Sci 67: 113-121. 19859664
Pelis, R.M., X. Zhang, Y. Dangprapai, and S.H. Wright. (2006). Cysteine accessibility in the hydrophilic cleft of human organic cation transporter 2. J. Biol. Chem. 281: 35272-35280. 16990275
Pelka, K., D. Bertheloot, E. Reimer, K. Phulphagar, S.V. Schmidt, A. Christ, R. Stahl, N. Watson, K. Miyake, N. Hacohen, A. Haas, M.M. Brinkmann, A. Marshak-Rothstein, F. Meissner, and E. Latz. (2018). The Chaperone UNC93B1 Regulates Toll-like Receptor Stability Independently of Endosomal TLR Transport. Immunity 48: 911-922.e7. 29768176
Pelka, K., K. Phulphagar, J. Zimmermann, R. Stahl, J.L. Schmid-Burgk, T. Schmidt, J.H. Spille, L.I. Labzin, S. Agrawal, E.R. Kandimalla, J.L. Casanova, V. Hornung, A. Marshak-Rothstein, S. Höning, and E. Latz. (2014). Cutting edge: the UNC93B1 tyrosine-based motif regulates trafficking and TLR responses via separate mechanisms. J Immunol 193: 3257-3261. 25187660
Pelletier, B., J. Beaudoin, C.C. Philpott, and S. Labbé. (2003). Fep1 represses expression of the fission yeast Schizosaccharomyces pombe siderophore-iron transport system. Nucleic Acids Res 31: 4332-4344. 12888492
Pellizzaro A., Clochard T., Planchet E., Limami AM. and Morere-Le Paven MC. (2015). Identification and molecular characterization of Medicago truncatula NRT2 and NAR2 families. Physiol Plant. 154(2):256-69. 25430977
Pendse, P.Y., B.R. Brooks, and J.B. Klauda. (2010). Probing the periplasmic-open state of lactose permease in response to sugar binding and proton translocation. J. Mol. Biol. 404: 506-521. 20875429
Peng, Y., S. Kumar, R.L. Hernandez, S.E. Jones, K.M. Cadle, K.P. Smith, and M.F. Varela. (2009). Evidence for the transport of maltose by the sucrose permease, CscB, of Escherichia coli. J. Membr. Biol. 228: 79-88. 19294451
Perdomo-Ramirez, A., E. Cordoba-Lanus, C.J. Trujillo-Frias, C. Gonzalez-Navasa, E. Ramos-Trujillo, M.I. Luis-Yanes, V. Garcia-Nieto, F. Claverie-Martin, and. (2023). Pathogenic Variants of (URAT1) and (GLUT9) in Spanish Patients with Renal Hypouricemia: Founder Effect of Variant c.374C>T; p.(T125M). Int J Mol Sci 24:. 37176161
Pereira, C.A. and A.M. Silber. (2012). On the evolution of hexose transporters in kinetoplastid potozoans. PLoS One 7: e36303. 22567148
Perland, E., E. Lekholm, M.M. Eriksson, S. Bagchi, V. Arapi, and R. Fredriksson. (2016). The Putative SLC Transporters Mfsd5 and Mfsd11 Are Abundantly Expressed in the Mouse Brain and Have a Potential Role in Energy Homeostasis. PLoS One 11: e0156912. 27272503
Perland, E., S. Bagchi, A. Klaesson, and R. Fredriksson. (2017). Characteristics of 29 novel atypical solute carriers of major facilitator superfamily type: evolutionary conservation, predicted structure and neuronal co-expression. Open Biol 7:. 28878041
Perland, E., S.V. Hellsten, E. Lekholm, M.M. Eriksson, V. Arapi, and R. Fredriksson. (2016). The Novel Membrane-Bound Proteins MFSD1 and MFSD3 are Putative SLC Transporters Affected by Altered Nutrient Intake. J Mol Neurosci. [Epub: Ahead of Print] 27981419
Perland, E., S.V. Hellsten, N. Schweizer, V. Arapi, F. Rezayee, M. Bushra, and R. Fredriksson. (2017). Structural prediction of two novel human atypical SLC transporters, MFSD4A and MFSD9, and their neuroanatomical distribution in mice. PLoS One 12: e0186325. 29049335
Perry, J.L., N. Dembla-Rajpal, L.A. Hall, and J.B. Pritchard. (2006). A three-dimensional model of human organic anion transporter 1: aromatic amino acids required for substrate transport. J. Biol. Chem. 281: 38071-38079. 17038320
Persson, B.L., A. Berhe, U. Fristedt, P. Martinez, J. Pattison, J. Petersson, and R. Weinander. (1998). Phosphate permeases of Saccharomyces cerevisiae. Biochim. Biophys. Acta 1365: 23-30. 9693717
Persson, B.L., J. Petersson, U. Fristedt, R. Weinander, A. Berhe, and J. Pattison. (1999). Phosphate permeases of Saccharomyces cerevisiae: structure, function and regulation. Biochim. Biophys. Acta 1422: 255-272. 10548719
Philip, M., S.A. Funkhouser, E.Y. Chiu, S.R. Phelps, J.J. Delrow, J. Cox, P.J. Fink, and J.L. Abkowitz. (2015). Heme Exporter FLVCR Is Required for T Cell Development and Peripheral Survival. J Immunol 194: 1677-1685. 25582857
Pietrancosta, N., C. Anne, H. Prescher, R. Ruivo, C. Sagné, C. Debacker, H.O. Bertrand, R. Brossmer, F. Acher, and B. Gasnier. (2012). Successful prediction of substrate-binding pocket in SLC17 transporter sialin. J. Biol. Chem. 287: 11489-11497. 22334707
Pietrancosta, N., M. Djibo, S. Daumas, S. El Mestikawy, and J.D. Erickson. (2020). Molecular, Structural, Functional, and Pharmacological Sites for Vesicular Glutamate Transporter Regulation. Mol Neurobiol. [Epub: Ahead of Print] 32474835
Pimentel-Schmitt, E.F., K. Jahreis, M.P. Eddy, J. Amon, A. Burkovski, and F. Titgemeyer. (2009). Identification of a glucose permease from Mycobacterium smegmatis mc2 155. J. Mol. Microbiol. Biotechnol. 16: 169-175. 18311074
Pina, C., P. Goncalves, C. Prista, and M.C. Loureiro-Dias. (2004). Ffz1, a new transporter specific for fructose from Zygosaccharomyces bailii. Microbiology 150: 2429-2433. 15256584
Plourde-Owobi, L., S. Durner, J.L. Parrou, R. Wieczorke, G. Goma, and J. François. (1999). AGT1, encoding an α-glucoside transporter involved in uptake and intracellular accumulation of trehalose in Saccharomyces cerevisiae. J. Bacteriol. 181: 3830-3832. 10368160
Pochini, L., M. Scalise, M. Galluccio, and C. Indiveri. (2013). OCTN cation transporters in health and disease: role as drug targets and assay development. J Biomol Screen 18: 851-867. 23771822
Pochini, L., M. Scalise, S. Di Silvestre, S. Belviso, A. Pandolfi, A. Arduini, M. Bonomini, and C. Indiveri. (2015). Acetylcholine and acetylcarnitine transport in peritoneum: role of the SLC22A4 (OCTN1) transporter. Biochim. Biophys. Acta. [Epub: Ahead of Print] 26724204
Posavi, M., D. Gulisija, J.B. Munro, J.C. Silva, and C.E. Lee. (2020). Rapid evolution of genome-wide gene expression and plasticity during saline to freshwater invasions by the copepod Eurytemora affinis species complex. Mol Ecol. [Epub: Ahead of Print] 33047351
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. 25209865
Price, D.R., H.S. Wilkinson, and J.A. Gatehouse. (2007). Functional expression and characterisation of a gut facilitative glucose transporter, NlHT1, from the phloem-feeding insect Nilaparvata lugens (rice brown planthopper). Insect Biochem Mol Biol 37: 1138-1148. 17916500
Psakis, G., M. Saidijam, K. Shibayama, J. Polaczek, K.E. Bettaney, J.M. Baldwin, S.A. Baldwin, R. Hope, L.O. Essen, R.C. Essenberg, and P.J. Henderson. (2009). The sodium-dependent D-glucose transport protein of Helicobacter pylori. Mol. Microbiol. 71: 391-403. 19161491
Puri, S. and K. Juvale. (2020). Monocarboxylate transporter 1 and 4 inhibitors as potential therapeutics for treating solid tumours: A review with structure-activity relationship insights. Eur J Med Chem 199: 112393. [Epub: Ahead of Print] 32388280
Qiao, Y., C. Li, X. Lu, H. Zong, and B. Zhuge. (2021). Identification of key residues for efficient glucose transport by the hexose transporter CgHxt4 in high sugar fermentation yeast Candida glycerinogenes. Appl. Microbiol. Biotechnol. [Epub: Ahead of Print] 34515842
Qin, L., X. Liu, Q. Sun, Z. Fan, D. Xia, G. Ding, H.L. Ong, D. Adams, W.A. Gahl, C. Zheng, S. Qi, L. Jin, C. Zhang, L. Gu, J. He, D. Deng, I.S. Ambudkar, and S. Wang. (2012). Sialin (SLC17A5) functions as a nitrate transporter in the plasma membrane. Proc. Natl. Acad. Sci. USA 109: 13434-13439. 22778404
Qin, Y., J. Wang, Q. Lv, and B. Han. (2023). Recent Progress in Research on Mitochondrion-Targeted Antifungal Drugs: a Review. Antimicrob. Agents Chemother. e0000323. [Epub: Ahead of Print] 37195189
Qiu, A., M. Jansen, A. Sakaris, S.H. Min, S. Chattopadhyay, E. Tsai, C. Sandoval, R. Zhao, M.H. Akabas, and I.D. Goldman. (2006). Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption. Cell 127: 917-928. 17129779
Quistgaard, E.M., C. Löw, P. Moberg, and P. Nordlund. (2013). Metal-mediated crystallization of the xylose transporter XylE from Escherichia coli in three different crystal forms. J Struct Biol 184: 375-378. 24060988
Radestock, S. and L.R. Forrest. (2011). The alternating-access mechanism of MFS transporters arises from inverted-topology repeats. J. Mol. Biol. 407: 698-715. 21315728
Rajamohan, G., V.B. Srinivasan, and W.A. Gebreyes. (2010). Molecular and functional characterization of a novel efflux pump, AmvA, mediating antimicrobial and disinfectant resistance in Acinetobacter baumannii. J Antimicrob Chemother 65: 1919-1925. 20573661
Ramìrez, S., R. Moreno, O. Zafra, P. Castàn, C. Vallès, and J. Berenguer. (2000). Two nitrate/nitrite transporters are encoded within the mobilizable plasmid for nitrate respiration of Thermus thermophilus HB8. J. Bacteriol. 182: 2179-2183. 10735860
Ramon-Garcia S., Martin C., Thompson CJ. and Ainsa JA. (2009). Role of the Mycobacterium tuberculosis P55 efflux pump in intrinsic drug resistance, oxidative stress responses, and growth. Antimicrob Agents Chemother. 53(9):3675-82. 19564371
Ramzan, R., M. Safiullah Virk, Z. Muhammad, A.M.M. Ahmed, X. Yuan, and F. Chen. (2019). Genetic Modification of Gene Stimulating the Putative Penicillin Production in M7 and Exhibiting the Sensitivity towards Precursor Amino Acids of Penicillin Pathway. Microorganisms 7:. 31554331
Rana, N., M.A. Aziz, R.A.T. Serya, D.S. Lasheen, N. Samir, F. Wuest, K.A.M. Abouzid, and F.G. West. (2023). A Fluorescence-Based Assay to Probe Inhibitory Effect of Fructose Mimics on GLUT5 Transport in Breast Cancer Cells. ACS Bio Med Chem Au 3: 51-61. 37101605
Rani, M., S. Raj, V. Dayaman, M. Kumar, M. Dua, and A.K. Johri. (2016). Functional Characterization of a Hexose Transporter from Root Endophyte Piriformospora indica. Front Microbiol 7: 1083. 27499747
Rappold, P.M., M. Cui, A.S. Chesser, J. Tibbett, J.C. Grima, L. Duan, N. Sen, J.A. Javitch, and K. Tieu. (2011). Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3. Proc. Natl. Acad. Sci. USA 108: 20766-20771. 22143804
Raschdorf, O., F.D. Müller, M. Pósfai, J.M. Plitzko, and D. Schüler. (2013). The magnetosome proteins MamX, MamZ and MamH are involved in redox control of magnetite biomineralization in Magnetospirillum gryphiswaldense. Mol. Microbiol. 89: 872-886. 23889511
Raven, L.M., C.A. Muir, C. Kessler Iglesias, N.K. Bart, K. Muthiah, E. Kotlyar, P. Macdonald, C.S. Hayward, A. Jabbour, and J.R. Greenfield. (2023). Sodium glucose co-transporter 2 inhibition with empagliflozin on metabolic, cardiac and renal outcomes in recent cardiac transplant recipients (EMPA-HTx): protocol for a randomised controlled trial. BMJ Open 13: e069641. 36990488
Ray, A., J. Wen, L. Yammine, J. Culver, I.S. Parida, J. Garren, L. Xue, K. Hales, Q. Xiang, M.J. Birnbaum, B.B. Zhang, M. Monetti, and T.E. McGraw. (2023). Regulated dynamic subcellular GLUT4 localization revealed by proximal proteome mapping in human muscle cells. J Cell Sci 136:. 38126809
Raymond-Bouchard I., Carroll CS., Nesbitt JR., Henry KA., Pinto LJ., Moinzadeh M., Scott JK. and Moore MM. (2012). Structural requirements for the activity of the MirB ferrisiderophore transporter of Aspergillus fumigatus. Eukaryot Cell. 11(11):1333-44. 22903978
Reddy, A., L.H.M. Bozi, O.K. Yaghi, E.L. Mills, H. Xiao, H.E. Nicholson, M. Paschini, J.A. Paulo, R. Garrity, D. Laznik-Bogoslavski, J.C.B. Ferreira, C.S. Carl, K.A. Sjøberg, J.F.P. Wojtaszewski, J.F. Jeppesen, B. Kiens, S.P. Gygi, E.A. Richter, D. Mathis, and E.T. Chouchani. (2020). pH-Gated Succinate Secretion Regulates Muscle Remodeling in Response to Exercise. Cell 183: 62-75.e17. 32946811
Redhu, A.K., A. Banerjee, A.H. Shah, A. Moreno, M.K. Rawal, R. Nair, P. Falson, and R. Prasad. (2018). Molecular Basis of Substrate Polyspecificity of the Candida albicans Mdr1p Multidrug/H Antiporter. J. Mol. Biol. 430: 682-694. 29341887
Reihl, P. and J. Stolz. (2005). The monocarboxylate transporter homolog Mch5p catalyzes riboflavin (vitamin B2) uptake in Saccharomyces cerevisiae. J. Biol. Chem. 280: 39809-39817. 16204239
Reinders, A., J.A. Panshyshyn, and J.M. Ward. (2005). Analysis of transport activity of Arabidopsis sugar alcohol permease homolog AtPLT5. J. Biol. Chem. 280: 1594-1602. 15525644
Remm, S., D. De Vecchis, J. Schöppe, C.A.J. Hutter, I. Gonda, M. Hohl, S. Newstead, L.V. Schäfer, and M.A. Seeger. (2023). Structural basis for triacylglyceride extraction from mycobacterial inner membrane by MFS transporter Rv1410. Nat Commun 14: 6449. 37833269
Remy, E., T.R. Cabrito, P. Baster, R.A. Batista, M.C. Teixeira, J. Friml, I. Sá-Correia, and P. Duque. (2013). A major facilitator superfamily transporter plays a dual role in polar auxin transport and drought stress tolerance in Arabidopsis. Plant Cell 25: 901-926. 23524662
Remy, E., T.R. Cabrito, R.A. Batista, M.A. Hussein, M.C. Teixeira, A. Athanasiadis, I. Sá-Correia, and P. Duque. (2014). Intron retention in the 5''UTR of the novel ZIF2 transporter enhances translation to promote zinc tolerance in arabidopsis. PLoS Genet 10: e1004375. 24832541
Ren F., Zhao CZ., Liu CS., Huang KL., Guo QQ., Chang LL., Xiong H. and Li XB. (2014). A Brassica napus PHT1 phosphate transporter, BnPht1;4, promotes phosphate uptake and affects roots architecture of transgenic Arabidopsis. Plant Mol Biol. 86(6):595-607. 25194430
Reynes, J.P., T. Calmels, D. Drocourt, and G. Tiraby. (1988). Cloning, expression in Escherichia coli and nucleotide sequence of a tetracycline-resistance gene from Streptomyces rimosus. J. Gen. Microbiol. 134: 585-598. 3053973
Ricard-Blum, S. and J.R. Couchman. (2023). Conformations, interactions and functions of intrinsically disordered syndecans. Biochem Soc Trans 51: 1083-1096. 37334846
Rizwan, A.N., W. Krick, G. Burckhardt. (2007). The chloride dependence of the human organic anion transporter 1 (hOAT1) is blunted by mutation of a single amino acid. J. Biol. Chem. 282: 13402-13409. 17353191
Roca, I., S. Marti, P. Espinal, P. Martínez, I. Gibert, and J. Vila. (2009). CraA, a major facilitator superfamily efflux pump associated with chloramphenicol resistance in Acinetobacter baumannii. Antimicrob. Agents Chemother. 53: 4013-4014. 19581458
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. 12376536
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. 20836887
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. 18931129
Rodionov, D.A., X. Li, I.A. Rodionova, C. Yang, L. Sorci, E. Dervyn, D. Martynowski, H. Zhang, M.S. Gelfand, and A.L. Osterman. (2008). Transcriptional regulation of NAD metabolism in bacteria: genomic reconstruction of NiaR (YrxA) regulon. Nucleic Acids Res 36: 2032-2046. 18276644
Rodionova, I.A., Y. Gao, A. Sastry, Y. Hefner, H.G. Lim, D.A. Rodionov, M.H. Saier, Jr, and B.O. Palsson. (2021). Identification of a transcription factor, PunR, that regulates the purine and purine nucleoside transporter punC in E. coli. Commun Biol 4: 991. 34413462
Rodrigues, L., J. Ramos, I. Couto, L. Amaral, and M. Viveiros. (2011). Ethidium bromide transport across Mycobacterium smegmatis cell-wall: correlation with antibiotic resistance. BMC Microbiol 11: 35. 21332993
Rodriguez-Contreras, D., H. Aslan, X. Feng, K. Tran, P.A. Yates, S. Kamhawi, and S.M. Landfear. (2015). Regulation and biological function of a flagellar glucose transporter in Leishmania mexicana: a potential glucose sensor. FASEB J. 29: 11-24. 25300620
Rolland, S., M. Hnatova, M. Lemaire, J. Leal-Sanchez, and M. Wésolowski-Louvel. (2006). Connection between the Rag4 glucose sensor and the KlRgt1 repressor in Kluyveromyces lactis. Genetics 174: 617-626. 16783006
Roohparvar, R., M.A. De Waard, G.H. Kema, and L.H. Zwiers. (2007). MgMfs1, a major facilitator superfamily transporter from the fungal wheat pathogen Mycosphaerella graminicola, is a strong protectant against natural toxic compounds and fungicides. Fungal Genet Biol 44: 378-388. 17107817
Ropka-Molik, K., M. Stefaniuk-Szmukier, T. Szmatoła, K. Piórkowska, and M. Bugno-Poniewierska. (2019). The use of the SLC16A1 gene as a potential marker to predict race performance in Arabian horses. BMC Genet 20: 73. 31510920
Roshanbin, S., F.A. Lindberg, E. Lekholm, M.M. Eriksson, E. Perland, J. Åhlund, A. Raine, and R. Fredriksson. (2016). Histological characterization of orphan transporter MCT14 (SLC16A14) shows abundant expression in mouse CNS and kidney. BMC Neurosci 17: 43. 27364523
Rottmann, T., W. Zierer, C. Subert, N. Sauer, and R. Stadler. (2016). STP10 encodes a high-affinity monosaccharide transporter and is induced under low-glucose conditions in pollen tubes of Arabidopsis. J Exp Bot. [Epub: Ahead of Print] 26893494
Rubio, L., J. Díaz-García, V. Amorim-Silva, A.P. Macho, M.A. Botella, and J.A. Fernández. (2019). Molecular Characterization of , the Putative Sodium Dependent High-Affinity Nitrate Transporter of L. Int J Mol Sci 20:. 31357380
Ruiz-Pavón, L., P.M. Karlsson, J. Carlsson, D. Samyn, B. Persson, B.L. Persson, and C. Spetea. (2010). Functionally important amino acids in the Arabidopsis thylakoid phosphate transporter: homology modeling and site-directed mutagenesis. Biochemistry 49: 6430-6439. 20565143
Ruscitto, A., K. Honma, V.M. Veeramachineni, K. Nishikawa, G.P. Stafford, and A. Sharma. (2017). Regulation and Molecular Basis of Environmental Muropeptide Uptake and Utilization in Fastidious Oral Anaerobe. Front Microbiol 8: 648. 28446907
Ryu, N., B. Sagong, H.J. Park, M.A. Kim, K.Y. Lee, J.Y. Choi, and U.K. Kim. (2016). Screening of the SLC17A8 gene as a causative factor for autosomal dominant non-syndromic hearing loss in Koreans. BMC Med Genet 17: 6. 26797701
Sa-Nogueira I. and Ramos S.S. (1997). Cloning, functional analysis, and transcriptional regulation of the Bacillus subtilis araE gene involved in L-arabinose utilization. J. of Bacteriol. 179:7705-7711. 9401028
Sachs, M., A.R. Quijada-Rodriguez, S. Hans, and D. Weihrauch. (2022). Characterization of two novel ammonia transporters, HIAT1α and HIAT1β, in the American Horseshoe Crab, Limulus polyphemus. Comp Biochem Physiol A Mol Integr Physiol 278: 111365. [Epub: Ahead of Print] 36577451
Sager, G., N. Smaglyukova, and O.M. Fuskevaag. (2018). The role of OAT2 (SLC22A7) in the cyclic nucleotide biokinetics of human erythrocytes. J Cell Physiol 233: 5972-5980. 29244191
Sagor, G.H., T. Berberich, S. Kojima, M. Niitsu, and T. Kusano. (2016). Spermine modulates the expression of two probable polyamine transporter genes and determines growth responses to cadaverine in Arabidopsis. Plant Cell Rep 35: 1247-1257. 26905725
Saha, M., A.K. Pragasam, S. Kumari, J. Verma, B. Das, and R.K. Bhadra. (2024). Genomic and functional insights into antibiotic resistance genes and linked with the SXT element of non-O1/non-O139. Microbiology (Reading) 170:. 38180462
Saha, P., S. Sikdar, G. Krishnamoorthy, H.I. Zgurskaya, and V.V. Rybenkov. (2020). Drug Permeation against Efflux by Two Transporters. ACS Infect Dis 6: 747-758. 32039579
Said, H.M., K. Van Voorhis, F.K. Ghishan, N. Abumurad, W. Nylander, and R. Redha. (1989). Transport characteristics of glutamine in human intestinal brush-border membrane vesicles. Am. J. Physiol. 256: G240-245. 2492158
Saier, M.H., Jr, D.L. Wentzel, B.U. Feucht, and J.J. Judice. (1975). A transport system for phosphoenolpyruvate, 2-phosphoglycerate, and 3-phosphoglycerate in Salmonella typhimurium. J. Biol. Chem. 250: 5089-5096. 238977
Saitoh H., Hirabuchi A., Fujisawa S., Mitsuoka C., Terauchi R. and Takano Y. (2014). MoST1 encoding a hexose transporter-like protein is involved in both conidiation and mycelial melanization of Magnaporthe oryzae. FEMS Microbiol Lett. 352(1):104-13. 24372780
Sakamoto, T., K. Inoue-Sakamoto, and D.A. Bryant. (1999). A novel nitrate/nitrite permease in the marine cyanobacterium Synechococcus sp. strain PCC 7002. J. Bacteriol. 181: 7363-7372. 10572142
Salcedo-Sora, J.E., S. Jindal, S. O''Hagan, and D.B. Kell. (2021). A palette of fluorophores that are differentially accumulated by wild-type and mutant strains of : surrogate ligands for profiling bacterial membrane transporters. Microbiology (Reading) 167:. 33406033
Salveridou, E., S. Mayerl, S.M. Sundaram, B. Markova, and H. Heuer. (2020). Tissue-Specific Function of Thyroid Hormone Transporters: New Insights from Mouse Models. Exp Clin Endocrinol Diabetes 128: 423-427. 31724131
Samodelov, S.L., G.A. Kullak-Ublick, Z. Gai, and M. Visentin. (2020). Organic Cation Transporters in Human Physiology, Pharmacology, and Toxicology. Int J Mol Sci 21:. 33114309
Sandoval, G.M., J.S. Duerr, J. Hodgkin, J.B. Rand, and G. Ruvkun. (2006). A genetic interaction between the vesicular acetylcholine transporter VAChT/UNC-17 and synaptobrevin/SNB-1 in C. elegans. Nat Neurosci 9: 599-601. 16604067
Sano, R., Y. Shinozaki, and T. Ohta. (2020). Sodium-glucose cotransporters: Functional properties and pharmaceutical potential. J Diabetes Investig. [Epub: Ahead of Print] 32196987
Santiviago, C.A., J.A. Fuentes, S.M. Bueno, A.N. Trombert, A.A. Hildago, L.T. Socias, P. Youderian, and G.C. Mora. (2002). The Salmonella entrica sv. Typhimurium smvA, yddG and ompD (porin) genes are required for the efficient efflux of methyl viologen. Mol. Microbiol. 46: 687-698. 12410826
Sanyal, S. and M. Ramaswami. (2002). Spinsters, synaptic defects, and amaurotic idiocy. Neuron 36: 335-338. 12408836
Sapunaric, F.M. and S.B. Levy. (2005). Substitutions in the interdomain loop of the Tn10 TetA efflux transporter alter tetracycline resistance and substrate specificity. Microbiology 151: 2315-2322. 16000721
Sardar, D., Y.T. Cheng, J. Woo, D.J. Choi, Z.F. Lee, W. Kwon, H.C. Chen, B. Lozzi, A. Cervantes, K. Rajendran, T.W. Huang, A. Jain, B.R. Arenkiel, I. Maze, and B. Deneen. (2023). Induction of astrocytic Slc22a3 regulates sensory processing through histone serotonylation. Science 380: eade0027. 37319217
Sargiacomo, M., M. Valtieri, M. Gabbianelli, E. Pelosi, U. Testa, A. Camagna, and C. Peschle. (1991). Pure human hematopoietic progenitors: direct inhibitory effect of transforming growth factors-beta 1 and -beta 2. Ann. N.Y. Acad. Sci. 628: 84-91. 2069325
Sasa, K., K. Yoshimura, A. Yamada, D. Suzuki, Y. Miyamoto, H. Imai, K. Nagayama, K. Maki, M. Yamamoto, and R. Kamijo. (2018). Monocarboxylate transporter-1 promotes osteoblast differentiation via suppression of p53, a negative regulator of osteoblast differentiation. Sci Rep 8: 10579. 30002387
Sasaki, M. and Y. Kurusu. (2004). Analysis of spontaneous base substitutions generated in mutator strains of Bacillus subtilis. FEMS Microbiol. Lett. 234: 37-42. 15109717
Sato, M. and M. Mueckler. (1999). A conserved amino acid motif (R-X-G-R-R) in the Glut1 glucose transporter is an important determinant of membrane topology. J. Biol. Chem. 274: 24721-24725. 10455140
Sauer, J.-D., Bachman, M.A., and Swanson, M.S. (2005). The phagosomal transporter A couples threonine acquisition to differentiation and replication of Legionella pneumophila in macrophages. Proc. Natl. Acad. Sci. USA 102: 9924-9929. 15998735
Savalas, L.R., B. Gasnier, M. Damme, T. Lübke, C. Wrocklage, C. Debacker, A. Jézégou, T. Reinheckel, A. Hasilik, P. Saftig, and B. Schröder. (2011). Disrupted in renal carcinoma 2 (DIRC2), a novel transporter of the lysosomal membrane, is proteolytically processed by cathepsin L. Biochem. J. 439: 113-128. 21692750
Sawada, K., N. Echigo, N. Juge, T. Miyaji, M. Otsuka, H. Omote, A. Yamamoto, and Y. Moriyama. (2008). Identification of a vesicular nucleotide transporter. Proc. Natl. Acad. Sci. USA. 105: 5683-5686. 18375752
Schaedler TA. and van Veen HW. (2010). A flexible cation binding site in the multidrug major facilitator superfamily transporter LmrP is associated with variable proton coupling. FASEB J. 24(10):3653-61. 20472749
Scharenberg, S.G., W. Dong, A. Ghoochani, K. Nyame, R. Levin-Konigsberg, A.R. Krishnan, E.S. Rawat, K. Spees, M.C. Bassik, and M. Abu-Remaileh. (2023). An SPNS1-dependent lysosomal lipid transport pathway that enables cell survival under choline limitation. Sci Adv 9: eadf8966. 37075117
Scharff-Poulsen, P., H. Moriya, and M. Johnston. (2018). Genetic Analysis of Signal Generation by the Rgt2 Glucose Sensor of. G3 (Bethesda). [Epub: Ahead of Print] 29954842
Schaub, R.E. and J.P. Dillard. (2019). The Pathogenic Neisseria Use a Streamlined Set of Peptidoglycan Degradation Proteins for Peptidoglycan Remodeling, Recycling, and Toxic Fragment Release. Front Microbiol 10: 73. 30766523
Scheepers, A., S. Schmidt, A. Manolescu, C.I. Cheeseman, A. Bell, C. Zahn, H.G. Joost, and A. Schürmann. (2005). Characterization of the human SLC2A11 (GLUT11) gene: alternative promoter usage, function, expression, and subcellular distribution of three isoforms, and lack of mouse orthologue. Mol. Membr. Biol. 22: 339-351. 16154905
Schilling, S. and C. Oesterhelt. (2007). Structurally reduced monosaccharide transporters in an evolutionarily conserved red alga. Biochem. J. 406: 325-331. 17497961
Schlessinger, A., N. Zatorski, K. Hutchinson, and C. Colas. (2023). Targeting SLC transporters: small molecules as modulators and therapeutic opportunities. Trends. Biochem. Sci. [Epub: Ahead of Print] 37355450
Schmidl, S., S.A. Tamayo Rojas, C.V. Iancu, J.Y. Choe, and M. Oreb. (2020). Functional Expression of the Human Glucose Transporters GLUT2 and GLUT3 in Yeast Offers Novel Screening Systems for GLUT-Targeting Drugs. Front Mol Biosci 7: 598419. 33681287
Schmitt, B.M. and H. Koepsell. (2005). Alkali cation binding and permeation in the rat organic cation transporter rOCT2. J. Biol. Chem. 280: 24481-24490. 15878879
Schneider, S., A. Schneidereit, K.R. Konrad, M.R. Hajirezaei, M. Gramann, R. Hedrich, and N. Sauer. (2006). Arabidopsis INOSITOL TRANSPORTER4 mediates high-affinity H+ symport of myoinositol across the plasma membrane. Plant Physiol. 141:565-577. 16603666
Schneider, S., A. Schneidereit, P. Udvardi, U. Hammes, M. Gramann, P. Dietrich, and N. Sauer. (2007). Arabidopsis INOSITOL TRANSPORTER2 Mediates H+-Symport of Different Inositol Epimers and Derivatives across the Plasma Membrane. Plant Physiol. 145: 1395-1407. 17951450
Schneider, S., D. Beyhl, R. Hedrich, and N. Sauer. (2008). Functional and Physiological Characterization of Arabidopsis INOSITOL TRANSPORTER1, a Novel Tonoplast-Localized Transporter for myo-Inositol. Plant Cell 20: 1073-1087. 18441213
Schneidereit, A., J. Scholz-Starke, N. Sauer, and M. Büttner. (2005). AtSTP11, a pollen tube-specific monosaccharide transporter in Arabidopsis. Planta.221: 48-55. 15565288
Scholz-Starke J., M. Büttner, and N. Sauer. (2003). AtSTP6, a new pollen-specific H+-monosaccharide symporter from Arabidopsis. Plant Physiol. 131:70-77. 12529516
Schouler, C., A. Taki, I. Chouikha, M. Moulin-Schouleur, and P. Gilot. (2009). A genomic island of an extraintestinal pathogenic Escherichia coli Strain enables the metabolism of fructooligosaccharides, which improves intestinal colonization. J. Bacteriol. 191: 388-393. 18978057
Schubbe, S., M. Kube, A. Scheffel, C. Wawer, U. Heyen, A. Meyerdierks, M.H. Madkour, F. Mayer, R. Reinhardt, and D. Schüler. (2003). Characterization of a spontaneous nonmagnetic mutant of Magnetospirillum gryphiswaldense reveals a large deletion comprising a putative magnetosome island. J. Bacteriol. 185: 5779–5790. 13129949
Schuldiner, S., A. Shirvan, and M. Linial. (1995). Vesicular neurotransmitter transporters: from bacteria to humans. Physiol. Rev. 75: 369-392. 7724667
Schulz, A., D. Beyhl, I. Marten, A. Wormit, E. Neuhaus, G. Poschet, M. Büttner, S. Schneider, N. Sauer, and R. Hedrich. (2011). Proton-driven sucrose symport and antiport are provided by the vacuolar transporters SUC4 and TMT1/2. Plant J. 68: 129-136. 21668536
Schussler, A., H. Martin, D. Cohen, M. Fitz, and D. Wipf. (2006). Characterization of a carbohydrate transporter from symbiotic glomeromycotan fungi. Nature 444: 933-936. 17167486
Schweizer U. and Kohrle J. (2013). Function of thyroid hormone transporters in the central nervous system. Biochim Biophys Acta. 1830(7):3965-73. 22890106
Schwöppe, C., H.H. Winkler, and H.E. Neuhaus. (2002). Properties of the glucose-6-phosphate transporter from Chlamydia pneumoniae (HPTcp) and the glucose-6-phosphate sensor from Escherichia coli (UhpC). J. Bacteriol. 184: 2108-2115. 11914341
Scisciola, L., F. Taktaz, R.A. Fontanella, A. Pesapane, Surina, V. Cataldo, P. Ghosh, M. Franzese, A. Puocci, P. Paolisso, C. Rafaniello, R. Marfella, M.R. Rizzo, E. Barbato, M. Vanderheyden, and M. Barbieri. (2023). Targeting high glucose-induced epigenetic modifications at cardiac level: the role of SGLT2 and SGLT2 inhibitors. Cardiovasc Diabetol 22: 24. 36732760
Sealover, N.R., B. Felts, C.P. Kuntz, R.E. Jarrard, G.H. Hockerman, E.L. Barker, and L.K. Henry. (2016). The external gate of the human and Drosophila serotonin transporters requires a basic/acidic amino acid pair for 3,4-methylenedioxymethamphetamine (MDMA) translocation and the induction of substrate efflux. Biochem Pharmacol 120: 46-55. 27638414
Seatter, M.J., S.A. De la Rue, L.M. Porter, and G.W. Gould. (1998). QLS motif in transmembrane helix VII of the glucose transporter family interacts with the C-1 position of D-glucose and is involved in substrate selection at the exofacial binding site. Biochemistry 37: 1322-1326. 9477959
Seeger, C., C. Poulsen, and G. Dandanell. (1995). Identification and characterization of genes (xapA, xapB, and xapR) involved in xanthosine catabolism in Escherichia coli. J. Bacteriol. 177: 5506-5516. 7559336
Seka, D.J., A.K. Schulz, T.M. Thaker, and T.M. Tomasiak. (2024). The N-terminal signature motif on the transporter MCT1 is critical for CD147-mediated trafficking. J. Biol. Chem. 300: 107333. 38820650
Sekine, T., S.H. Cha, M. Tsuda, N. Apiwattanakul, N. Nakajima, Y. Kanai, and H. Endou. (1998). Identification of multispecific organic anion transporter 2 expressed predominantly in the liver. FEBS Lett. 429: 179-182. 9650585
Seol, W. and A.J. Shatkin. (1992). Escherichia coli α-ketoglutarate permease is a constitutively expressed proton symporter. J. Biol. Chem. 267: 6409-6413. 1556144
Seol, W. and A.J. Shatkin. (1992). Site-directed mutants of Escherichia coli α-ketoglutarate permease (KgtP). Biochemistry 31: 3550-3554. 1554735
Serdiuk, T., S.A. Mari, and D.J. Müller. (2017). Pull-and-Paste of Single Transmembrane Proteins. Nano Lett. [Epub: Ahead of Print] 28627175
Serrano-Saiz, E., M.C. Vogt, S. Levy, Y. Wang, K.K. Kaczmarczyk, X. Mei, G. Bai, A. Singson, B.D. Grant, and O. Hobert. (2019). SLC17A6/7/8 Vesicular Glutamate Transporter Homologs in Nematodes. Genetics. [Epub: Ahead of Print] 31776169
Serrano-Saiz, E., R.J. Poole, T. Felton, F. Zhang, E.D. De La Cruz, and O. Hobert. (2013). Modular control of glutamatergic neuronal identity in C. elegans by distinct homeodomain proteins. Cell 155: 659-673. 24243022
Shaheen, A., F. Ismat, M. Iqbal, A. Haque, Z. Ul-Haq, O. Mirza, R. De Zorzi, T. Walz, and M. Rahman. (2021). Characterization of the multidrug efflux transporter styMdtM from Salmonella enterica serovar Typhi. Proteins 89: 1193-1204. 33983672
Shaji, D. (2021). Identification of Inhibitors Based on Molecular Docking: Thyroid Hormone Transmembrane Transporter MCT8 as a Target. Curr Drug Discov Technol 18: 105-112. 31774046
Sharifi, A., M. Kousi, C. Sagné, G.C. Bellenchi, L. Morel, M. Darmon, H. Hulková, R. Ruivo, C. Debacker, S. El Mestikawy, M. Elleder, A.E. Lehesjoki, A. Jalanko, B. Gasnier, and A. Kyttälä. (2010). Expression and lysosomal targeting of CLN7, a major facilitator superfamily transporter associated with variant late-infantile neuronal ceroid lipofuscinosis. Hum Mol Genet 19: 4497-4514. 20826447
Sharlin, D.S., L. Ng, F. Verrey, T.J. Visser, Y. Liu, R.T. Olszewski, M. Hoa, H. Heuer, and D. Forrest. (2018). Deafness and loss of cochlear hair cells in the absence of thyroid hormone transporters Slc16a2 (Mct8) and Slc16a10 (Mct10). Sci Rep 8: 4403. 29535325
Sharma, S., A. Banerjee, A. Moreno, A.K. Redhu, P. Falson, and R. Prasad. (2022). Spontaneous Suppressors against Debilitating Transmembrane Mutants of Mdr1 Disclose Novel Interdomain Communication via Signature Motifs of the Major Facilitator Superfamily. J Fungi (Basel) 8:. 35628792
Sharma, V., C.E. Noriega, and J.J. Rowe. (2006). Involvement of NarK1 and NarK2 proteins in transport of nitrate and nitrite in the denitrifying bacterium Pseudomonas aeruginosa PAO1. Appl. Environ. Microbiol. 72: 695-701. 16391109
Shayeghi, M., G.O. Latunde-Dada, J.S. Oakhill, A.H. Laftah, K. Takeuchi, N. Halliday, Y. Khan, A. Warley, F.E. McCann, R.C. Hider, D.M. Frazer, G.J. Anderson, C.D. Vulpe, R.J. Simpson, and A.T. McKie. (2005). Identification of an intestinal heme transporter. Cell 122: 789-801. 16143108
Sheng, Q., X. Wang, Z. Hou, B. Liu, M. Jiang, M. Ren, J. Fu, M. He, J. Zhang, Y. Xiang, Q. Zhang, L. Zhou, Y. Deng, and X. Shen. (2024). Novel functions of o-cymen-5-ol nanoemulsion in reversing colistin resistance in multidrug-resistant Klebsiella pneumoniae infections. Biochem Pharmacol 227: 116384. 38909787
Sheremet, A.S., S.V. Gronskiy, R.A. Akhmadyshin, A.E. Novikova, V.A. Livshits, R.S. Shakulov, and N.P. Zakataeva. (2011). Enhancement of extracellular purine nucleoside accumulation by Bacillus strains through genetic modifications of genes involved in nucleoside export. J Ind Microbiol Biotechnol 38: 65-70. 20814730
Shi, K., C. Li, C. Rensing, X. Dai, X. Fan, and G. Wang. (2018). A novel efflux transporter, ArsK, is responsible for bacterial resistance to arsenite, antimonite, trivalent roxarsone and methylarsenite. Appl. Environ. Microbiol. [Epub: Ahead of Print] 30315082
Shi, Z., H. Zeng, B. Zhao, C. Zeng, F. Zhang, Z. Liu, H.Y. Kwan, and T. Su. (2024). Sulforaphane reverses the enhanced NSCLC metastasis by regulating the miR-7-5p/c-Myc/LDHA axis in the acidic tumor microenvironment. Phytomedicine 133: 155874. 39079314
Shim, M.S., J.Y. Kim, K.H. Lee, H.K. Jung, B.A. Carlson, X.M. Xu, D.L. Hatfield, and B.J. Lee. (2011). l(2)01810 is a novel type of glutamate transporter that is responsible for megamitochondrial formation. Biochem. J. 439: 277-286. 21728998
Shimazu, M., T. Sekito, K. Akiyama, Y. Ohsumi, and Y. Kakinuma. (2005). A family of basic amino acid transporters of the vacuolar membrane from Saccharomyces cerevisiae. J. Biol. Chem. 280: 4851-4857. 15572352
Shin DS., Zhao R., Fiser A. and Goldman DI. (2012). Functional roles of the A335 and G338 residues of the proton-coupled folate transporter (PCFT-SLC46A1) mutated in hereditary folate malabsorption. Am J Physiol Cell Physiol. 303(8):C834-42. 22843796
Shin, D.S., R. Zhao, E.H. Yap, A. Fiser, and I.D. Goldman. (2012). A P425R mutation of the proton-coupled folate transporter causing hereditary folate malabsorption produces a highly selective alteration in folate binding. Am. J. Physiol. Cell Physiol. 302: C1405-1412. 22345511
Shin, D.S., S.H. Min, L. Russell, R. Zhao, A. Fiser, and I.D. Goldman. (2010). Functional roles of aspartate residues of the proton-coupled folate transporter (PCFT-SLC46A1); a D156Y mutation causing hereditary folate malabsorption. Blood 116: 5162-5169. 20805364
Shin, H.J., N. Anzai, A. Enomoto, X. He, do K. Kim, H. Endou, and Y. Kanai. (2007). Novel liver-specific organic anion transporter OAT7 that operates the exchange of sulfate conjugates for short chain fatty acid butyrate. Hepatology. 45: 1046-1055. 17393504
Shinnick, S.G., S.A. Perez, and M.F. Varela. (2003). Altered substrate selection of the melibiose transporter (MelY) of Enterobacter cloacae involving point mutations in Leu-88, Leu-91, and Ala-182 that confer enhanced maltose transport. J. Bacteriol. 185: 3672-3677. 12775706
Shiraya, K., T. Hirata, R. Hatano, S. Nagamori, P. Wiriyasermkul, P. Jutabha, M. Matsubara, S. Muto, H. Tanaka, S. Asano, N. Anzai, H. Endou, A. Yamada, H. Sakurai, and Y. Kanai. (2010). A novel transporter of SLC22 family specifically transports prostaglandins and co-localizes with 15-hydroxyprostaglandin dehydrogenase in renal proximal tubules. J. Biol. Chem. 285: 22141-22151. 20448048
Shishmarev, D., C.Q. Fontenelle, I. Kuprov, B. Linclau, and P.W. Kuchel. (2018). Transmembrane Exchange of Fluorosugars: Characterization of Red Cell GLUT1 Kinetics Using F NMR. Biophys. J. 115: 1906-1919. 30366625
Šikić, K., T. M A Peters, E. Marušić, I. Čulo Čagalj, D. Petković Ramadža, T. Žigman, K. Fumić, E. Fernandez, K. Gevaert, &.#.3.8.1.;. Debeljak, R. A Wevers, and I. Barić. (2022). Abnormal concentrations of acetylated amino acids in cerebrospinal fluid in acetyl-CoA transporter deficiency. J Inherit Metab Dis. [Epub: Ahead of Print] 35999711
Sim, S.Y., E.J. Hong, Y. Kim, and H.S. Lee. (2014). Analysis of cepA encoding an efflux pump-like protein in Corynebacterium glutamicum. J Microbiol 52: 278-283. 24535744
Singh, A.K. and R. Singh. (2020). Cardiovascular outcomes with SGLT-2 inhibitors and GLP-1 receptor agonist in Asians with type 2 diabetes: A systematic review and meta-analysis of cardiovascular outcome trials. Diabetes Metab Syndr 14: 715-722. [Epub: Ahead of Print] 32470852
Singh, G. and Y. Akhter. (2021). Molecular insights into the differential efflux mechanism of Rv1634 protein, a multidrug transporter of major facilitator superfamily in Mycobacterium tuberculosis. Proteins. [Epub: Ahead of Print] 34601761
Sionov, R.V., S. Banerjee, S. Bogomolov, R. Smoum, R. Mechoulam, and D. Steinberg. (2022). Targeting the Achilles'' Heel of Multidrug-Resistant by the Endocannabinoid Anandamide. Int J Mol Sci 23:. 35887146
Siskind, L.J., L. Feinstein, T. Yu, J.S. Davis, D. Jones, J. Choi, J.E. Zuckerman, W. Tan, R.B. Hill, J.M. Hardwick, and M. Colombini. (2008). Anti-apoptotic Bcl-2 Family proteins disassemble ceramide channels. J. Biol. Chem. 283: 6622-6630. 18171672
Skelly, P.J., J.W. Kim, J. Cunningham, and C.B. Shoemaker. (1994). Cloning, characterization, and functional expression of cDNAs encoding glucose transporter proteins from the human parasite Schistosoma mansoni. J. Biol. Chem. 269: 4247-4253. 8307988
Slavetinsky, C.J., A. Peschel, and C.M. Ernst. (2012). Alanyl-phosphatidylglycerol and lysyl-phosphatidylglycerol are translocated by the same MprF flippases and have similar capacities to protect against the antibiotic daptomycin in Staphylococcus aureus. Antimicrob. Agents Chemother. 56: 3492-3497. 22491694
Sloothaak, J., M. Schilders, P.J. Schaap, and L.H. de Graaff. (2014). Overexpression of the Aspergillus niger GatA transporter leads to preferential use of D-galacturonic acid over D-xylose. AMB Express 4: 66. 25177540
Smirnova, I., V. Kasho, and H.R. Kaback. (2011). Lactose permease and the alternating access mechanism. Biochemistry 50: 9684-9693. 21995338
Smirnova, I., V. Kasho, J. Sugihara, and H.R. Kaback. (2011). Opening the periplasmic cavity in lactose permease is the limiting step for sugar binding. Proc. Natl. Acad. Sci. USA 108: 15147-15151. 21896727
Smit, A., S.G. Moses, I.S. Pretorius, and R.R. Cordero Otero. (2008). The Thr505 and Ser557 residues of the AGT1-encoded α- glucoside transporter are critical for maltotriose transport in Saccharomyces cerevisiae. J. Appl. Microbiol. 104: 1103-1111. 18179544
Smith, D.J., J. Park, J.M. Tiedje, and W.W. Mohn. (2007). A large gene cluster in Burkholderia xenovorans encoding abietane diterpenoid catabolism. J. Bacteriol. 189: 6195-6204. 17586638
Smith, K.P., S. Kumar, and M.F. Varela. (2009). Identification, cloning, and functional characterization of EmrD-3, a putative multidrug efflux pump of the major facilitator superfamily from Vibrio cholerae O395. Arch. Microbiol. 191: 903-911. 19876617
Soares-Silva, I., J. Sá-Pessoa, V. Myrianthopoulos, E. Mikros, M. Casal, and G. Diallinas. (2011). A substrate translocation trajectory in a cytoplasm-facing topological model of the monocarboxylate/H⁺ symporter Jen1p. Mol. Microbiol. 81: 805-817. 21651629
Soares-Silva, I., S. Paiva, P. Kötter, K.D. Entian, and M. Casal. (2013). The disruption of JEN1 from Candida albicans impairs the transport of lactate. Mol. Membr. Biol. 21: 403-411. 15764370
Son, H.Y., S.W. Sohn, S.H. Im, H.J. Kim, M.K. Lee, B. Gombojav, H.S. Kwon, D.S. Park, H.L. Kim, K.U. Min, J. Sung, J.S. Seo, and J.I. Kim. (2015). Family-Based Association Study of Pulmonary Function in a Population in Northeast Asia. PLoS One 10: e0139716. 26430897
Soo, V.W., P. Hanson-Manful, and W.M. Patrick. (2011). Artificial gene amplification reveals an abundance of promiscuous resistance determinants in Escherichia coli. Proc. Natl. Acad. Sci. USA 108: 1484-1489. 21173244
Srinivas, S.R., P.D. Prasad, N.S. Umapathy, V. Ganapathy, and P.S. Shekhawat. (2007). Transport of butyryl-L-carnitine, a potential prodrug, via the carnitine transporter OCTN2 and the amino acid transporter ATB0,+. Am. J. Physiol. Gastrointest Liver Physiol 293: G1046-1053. 17855766
Stadler, R., M. Büttner, P. Ache, R. Hedrich, N. Ivashikina, M. Melzer, S.M. Shearson, S.M. Smith, and N. Sauer. (2003). Diurnal and light-regulated expression of AtSTP1 in guard cells of Arabidopsis. Plant Physiol. 133: 528-537. 12972665
Stasyk, O.G., M.M. Maidan, O.V. Stasyk, P. Van Dijck, J.M. Thevelein, and A.A. Sibirny. (2008). Identification of hexose transporter-like sensor HXS1 and functional hexose transporter HXT1 in the methylotrophic yeast Hansenula polymorpha. Eukaryot. Cell. 7(4): 735-746. 18310355
Stasyk, O.V., O.G. Stasyk, J. Komduur, M. Veenhuis, J.M. Cregg, and A.A. Sibirny. (2004). A hexose transporter homologue controls glucose repression in the methylotrophic yeast Hansenula polymorpha. J. Biol. Chem. 279: 8116-8125. 14660581
Staub, J.M., L. Brand, M. Tran, Y. Kong, and S.G. Rogers. (2012). Bacterial glyphosate resistance conferred by overexpression of an E. coli membrane efflux transporter. J Ind Microbiol Biotechnol 39: 641-647. 22089966
Staubitz, P., H. Neumann, T. Schneider, I. Wiedemann, and A. Peschel. (2004). MprF-mediated biosynthesis of lysylphosphatidylglycerol, an important determinant in staphylococcal defensin resistance. FEMS Microbiol. Lett. 231: 67-71. 14769468
Stolz, J. (2003). Isolation and characterization of the plasma membrane biotin transporter from Schizosaccharomyces pombe. Yeast. 20: 221-231. 12557275
Stolz, J., H.J. Wöhrmann, and C. Vogl. (2005). Amiloride uptake and toxicity in fission yeast are caused by the pyridoxine transporter encoded by bsu1+ (car1+). Eukaryot. Cell. 4: 319-326. 15701794
Stolz, J., T. Caspari, A.M. Carr, N. Sauer. (2004). Cell division defects of Schizosaccharomyces pombe liz1- mutants are caused by defects in pantothenate uptake. Euk. Cell 3: 406-412. 15075270
Stolz, J., U. Hoja, S, Meier, N. Sauer, and E. Schweizer. (2000). Identification of the plasma membrane H+-biotin symporter of Saccharomyces cerevisiae by rescue of a fatty acid-auxotrophic mutant. J. Biol. Chem. 274: 18741-18746. 10373489
Stout, K.A., A.R. Dunn, C. Hoffman, and G.W. Miller. (2019). The Synaptic Vesicle Glycoprotein 2: Structure, Function, and Disease Relevance. ACS Chem Neurosci 10: 3927-3938. 31394034
Strohm, A.K., L.M. Vaughn, and P.H. Masson. (2015). Natural variation in the expression of ORGANIC CATION TRANSPORTER 1 affects root length responses to cadaverine in Arabidopsis. J Exp Bot 66: 853-862. 25403917
Stroobants, K., J.R. Kumita, N.J. Harris, D.Y. Chirgadze, C.M. Dobson, P.J. Booth, and M. Vendruscolo. (2017). Amyloid-like Fibrils from an α-Helical Transmembrane Protein. Biochemistry 56: 3225-3233. 28493669
Sturm, A., V. Gorboulev, D. Gorbunov, T. Keller, C. Volk, B.M. Schmitt, P. Schlachtbauer, G. Ciarimboli, and H. Koepsell. (2007). Identification of cysteines in rat organic cation transporters rOCT1 (C322, C451) and rOCT2 (C451) critical for transport activity and substrate affinity. Am. J. Physiol. Renal Physiol 293: F767-779. 17567940
Su X. and Tsang JS. (2013). Existence of a robust haloacid transport system in a Burkholderia species bacterium. Biochim Biophys Acta. 1828(2):187-92. 23022134
Su, X., R. Li, K.F. Kong, and J.S. Tsang. (2016). Transport of haloacids across biological membranes. Biochim. Biophys. Acta. 1858: 3061-3070. 27668346
Subramanian, V.S., J.S. Marchant, and H.M. Said. (2008). Apical membrane targeting and trafficking of the human proton-coupled transporter in polarized epithelia. Am. J. Physiol. Cell Physiol. 294: C233-240. 18003745
Sugihara, J., I. Smirnova, V. Kasho, and H.R. Kaback. (2011). Sugar recognition by CscB and LacY. Biochemistry 50: 11009-11014. 22106930
Sugimoto, N., T. Iwaki, S. Chardwiriyapreecha, M. Shimazu, M. Kawano, T. Sekito, K. Takegawa, and Y. Kakinuma. (2011). Atg22p, a vacuolar membrane protein involved in the amino acid compartmentalization of Schizosaccharomyces pombe. Biosci. Biotechnol. Biochem. 75: 385-387. 21307582
Sugita, T. and K. Koketsu. (2022). Transporter Engineering Enables the Efficient Production of Lacto--triose II and Lacto--tetraose in. J Agric Food Chem 70: 5106-5114. 35426313
Suman, T.Y., S.Y. Kim, D.H. Yeom, Y. Jang, T.Y. Jeong, and J. Jeon. (2023). Transcriptome and computational approaches highlighting the molecular regulation of Zacco platypus induced by mesocosm exposure to common disinfectant chlorine. Chemosphere 319: 137989. 36736481
Sun, F., X. Cao, D. Yu, D. Hu, Z. Yan, Y. Fan, C. Wang, and A. Wu. (2022). AaTAS1 and AaMFS1 genes for biosynthesis or efflux transport of tenuazonic acid and pathogenicity of Alternaria alternata. Mol. Plant Microbe Interact. [Epub: Ahead of Print] 35175146
Sun, L., X. Zeng, C. Yan, X. Sun, X. Gong, Y. Rao, and N. Yan. (2012). Crystal structure of a bacterial homologue of glucose transporters GLUT1-4. Nature 490: 361-366. 23075985
Sun, M. and Q. Zheng. (2019). Key Factors in Conformation Transformation of an Important Neuron.ic Protein Glucose Transport 3 Revealed by Molecular Dynamics Simulation. ACS Chem Neurosci. [Epub: Ahead of Print] 31617996
Sun, Y. and C.K. Vanderpool. (2011). Regulation and function of Escherichia coli sugar efflux transporter A (SetA) during glucose-phosphate stress. J. Bacteriol. 193: 143-153. 20971900
Sutter, M.L., L. Console, A.F. Fahner, S.L. Samodelov, Z. Gai, G. Ciarimboli, C. Indiveri, G.A. Kullak-Ublick, and M. Visentin. (2021). The role of cholesterol recognition (CARC/CRAC) mirror codes in the allosterism of the human organic cation transporter 2 (OCT2, SLC22A2). Biochem Pharmacol 194: 114840. 34774844
Suzawa, K., A. Yukita, T. Hayata, T. Goto, H. Danno, T. Michiue, K.W. Cho, and M. Asashima. (2007). Xenopus glucose transporter 1 (xGLUT1) is required for gastrulation movement in Xenopus laevis. Int J Dev Biol 51: 183-190. 17486538
Sveinsdóttir, H.S., A. Decker, C. Christensen, P.B. Lucena, H. Þorsteinsson, E. Richert, V.H. Maier, R. Cornell, and K.&.#.1.9.8.;. Karlsson. (2022). Motility phenotype in a zebrafish vmat2 mutant. PLoS One 17: e0259753. 34986152
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. 23024344
Swapna, G.V.T., N. Dube, M.J. Roth, and G.T. Montelione. (2024). Modeling Alternative Conformational States of Pseudo-Symmetric Solute Carrier Transporters using Methods from Machine Learning. bioRxiv. 39071413
Sweet, D.H. and J.B. Pritchard. (1999). rOCT2 is a basolateral potential-driven carrier, not an organic cation/proton exchanger. Am. J. Physiol. 277: F890-898. 10600936
Tahlan, K., S.K. Ahn, A. Sing, T.D. Bodnaruk, A.R. Willems, A.R. Davidson, and J.R. Nodwell. (2007). Initiation of actinorhodin export in Streptomyces coelicolor. Mol. Microbiol. 63: 937-940. 17338074
Tailor, C.S., B.J. Willett, and D. Kabat. (1999). A putatve cell surface receptor for anemia-inducing feline leukemia virus subgroup C is a member of a transporter superfamily. Virology 73: 6500-6505. 10400745
Takahashi, M., H. Kishimoto, Y. Shirasaka, and K. Inoue. (2020). Functional characterization of monocarboxylate transporter 12 (SLC16A12/MCT12) as a facilitative creatine transporter. Drug Metab Pharmacokinet. [Epub: Ahead of Print] 32249133
Takanaga H. and Frommer WB. (2010). Facilitative plasma membrane transporters function during ER transit. FASEB J. 24(8):2849-58. 20354141
Tamai, I., R. Ohashi, J. Nezu, H. Yabuuchi, A. Oku, M. Shimane, Y. Sai, and A. Tsuji. (1998). Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J. Biol. Chem. 273: 20378-20382. 9685390
Tamayo, E., B. Nada, I. Hafermann, and J.P. Benz. (2024). Correlating sugar transporter expression and activities to identify transporters for an orphan sugar substrate. Appl. Microbiol. Biotechnol. 108: 83. 38189952
Tan, G. and F. Meier-Abt. (2021). Differential expression of hydroxyurea transporters in normal and polycythemia vera hematopoietic stem and progenitor cell subpopulations. Exp Hematol. [Epub: Ahead of Print] 33677043
Tan, M., Q. Pan, C. Yu, X. Zhai, J. Gu, L. Tao, and D. Xu. (2024). PIGT promotes cell growth, glycolysis, and metastasis in bladder cancer by modulating GLUT1 glycosylation and membrane trafficking. J Transl Med 22: 5. 38169393
Tan, P.K., T.M. Ostertag, and J.N. Miner. (2016). Mechanism of high affinity inhibition of the human urate transporter URAT1. Sci Rep 6: 34995. 27713539
Tanabe, T., H. Nakao, T. Kuroda, T. Tsuchiya, and S. Yamamoto. (2006). Involvement of the Vibrio parahaemolyticus pvsC gene in export of the siderophore vibrioferrin. Microbiol Immunol 50: 871-876. 17116982
Tanabe, T., T. Funahashi, H. Nakao, S.-I. 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. 14617658
Tanaka, M., Y. Miyamoto, K. Sasa, K. Yoshimura, A. Yamada, T. Shirota, and R. Kamijo. (2022). Low oxygen tension suppresses the death of chondrocyte-like ATDC5 cells induced by interleukin-1ß. In Vitro Cell Dev Biol Anim. [Epub: Ahead of Print] 35925448
Tang, G., B. Jiang, Y. Huang, M. Fu, L. Wu, and R. Wang. (2011). Identification of a novel bacterial k(+) channel. J. Membr. Biol. 242: 153-164. 21744086
Tanner, W. (2000). The Chlorella hexose/H+-symporters. Int Rev Cytol 200: 101-141. 10965467
Tavoulari, S. and S. Frillingos. (2008). Substrate selectivity of the melibiose permease (MelY) from Enterobacter cloacae. J. Mol. Biol. 376: 681-693. 18177889
Taylor, C.A., K.N. Stanley, and A.D. Shirras. (1997). The Orct gene of Drosophila melanogaster codes for a putative organic cation transporter with six or 12 transmembrane domains. Gene 201: 69-74. 9409773
Teijeira, F., R.V. Ullán, S.M. Guerra, C. García-Estrada, I. Vaca, and J.F. Martín. (2009). The transporter CefM involved in translocation of biosynthetic intermediates is essential for cephalosporin production. Biochem. J. 418: 113-124. 18840096
Tejada-Jiménez, M., A. Galván, and E. Fernández. (2011). Algae and humans share a molybdate transporter. Proc. Natl. Acad. Sci. USA 108: 6420-6425. 21464289
Tenreiro, S., P.A. Nunes, C.A. Viegas, M.S. Neves, M.C. Teixeira, M.G. Cabral, and I. Sá-Correia. (2002). AQR1 gene (ORF YNL065w) encodes a plasma membrane transporter of the major facilitator superfamily that confers resistance to short-chain monocarboxylic acids and quinidine in Saccharomyces cerevisiae. Biochem. Biophys. Res. Commun. 292: 741-748. 11922628
Teranishi, Y., M. Inoue, N.G. Yamamoto, T. Kihara, B. Wiehager, T. Ishikawa, B. Winblad, S. Schedin-Weiss, S. Frykman, and L.O. Tjernberg. (2015). Proton myo-inositol cotransporter is a novel γ-secretase associated protein that regulates Aβ production without affecting Notch cleavage. FEBS J. 282: 3438-3451. 26094765
Thévenod, F., R. Herbrechter, C. Schlabs, A. Pethe, W.K. Lee, N.A. Wolff, and E. Roussa. (2023). Role of the SLC22A17/lipocalin-2 receptor in renal endocytosis of proteins/metalloproteins: A focus on iron- and cadmium-binding proteins. Am. J. Physiol. Renal Physiol. [Epub: Ahead of Print] 37589051
Thompson, S.A., E.V. Maani, A.H. Lindell, C.J. King, and J.V. McArthur. (2007). Novel tetracycline resistance determinant isolated from an environmental strain of Serratia marcescens. Appl. Environ. Microbiol. 73: 2199-2206. 17308196
Tian, W., J. Qin, C. Lian, Q. Yao, and X. Wang. (2022). Identification of a major facilitator superfamily protein that is beneficial to L-lactic acid production by Bacillus coagulans at low pH. BMC Microbiol 22: 310. 36536285
Tirosh, O., N. Sigal, A. Gelman, N. Sahar, N. Fluman, S. Siemion, and E. Bibi. (2012). Manipulating the drug/proton antiport stoichiometry of the secondary multidrug transporter MdfA. Proc. Natl. Acad. Sci. USA 109: 12473-12478. 22802625
Tomitori, H., K. Kashiwagi, K. Sakata, Y. Kakinuma, and K. Igarashi. (1999). Identification of a gene for a polyamine transport protein in yeast. J. Biol. Chem. 274: 3265-3267. 9920864
Truernit, E., J. Schmid, P. Epple, J. Illig, and N. Sauer. (1996). The sink-specific and stress-regulated Arabidopsis STP4 gene: enhanced expression of a gene encoding a monosaccharide transporter by wounding, elicitors, and pathogen challenge. Plant Cell 8: 2169-2182. 8989877
Truernit, E., R. Stadler, K. Baier, and N. Sauer. (1999). A male gametophyte-specific monosaccharide transporter in Arabidopsis. Plant J. 17: 191-201. 10074716
Truong, D.M., G. Kaler, A. Khandelwal, P.W. Swaan, and S.K. Nigam. (2008). Multi-level analysis of organic anion transporters 1, 3, and 6 reveals major differences in structural determinants of antiviral discrimination. J. Biol. Chem. 283(13): 8654-8663. 18174163
Truong-Bolduc, Q.C., J. Strahilevitz, and D.C. Hooper. (2006). NorC, a new efflux pump regulated by MgrA of Staphylococcus aureus. Antimicrob. Agents Chemother. 50: 1104-1107. 16495280
Truong-Bolduc, Q.C., P.M. Dunman, J. Strahilevitz, S.J. Projan, and D.C. Hooper. (2005). MgrA is a multiple regulator of two new efflux pumps in Staphylococcus aureus. J. Bacteriol. 187: 2395-2405. 15774883
Truong-Bolduc, Q.C., Y. Wang, and D.C. Hooper. (2021). Staphylococcus aureus Tet38 Efflux Pump Structural Modeling and Roles of Essential Residues in Drug Efflux and Host Cell Internalization. Infect. Immun. 89:. 33619028
Tse, Y.M., M. Yu, and J.S. Tsang. (2009). Topological analysis of a haloacid permease of a Burkholderia sp. bacterium with a PhoA-LacZ reporter. BMC Microbiol 9: 233. 19878597
Tsuchida, H., N. Anzai, H.J. Shin, M.F. Wempe, P. Jutabha, A. Enomoto, S.H. Cha, T. Satoh, M. Ishida, H. Sakurai, and H. Endou. (2010). Identification of a novel organic anion transporter mediating carnitine transport in mouse liver and kidney. Cell Physiol Biochem 25: 511-522. 20332632
Tsutsumi, R., B. Ueberheide, F.X. Liang, B.G. Neel, R. Sakai, and Y. Saito. (2024). Endocytic vesicles act as vehicles for glucose uptake in response to growth factor stimulation. Nat Commun 15: 2843. 38565573
Tucker, D.F., J.T. Sullivan, K.A. Mattia, C.R. Fisher, T. Barnes, M.N. Mabila, R. Wilf, C. Sulli, M. Pitts, R.J. Payne, M. Hall, D. Huston-Paterson, X. Deng, E. Davidson, S.H. Willis, B.J. Doranz, R. Chambers, and J.B. Rucker. (2018). Isolation of state-dependent monoclonal antibodies against the 12-transmembrane domain glucose transporter 4 using virus-like particles. Proc. Natl. Acad. Sci. USA. [Epub: Ahead of Print] 29769329
Turner, M.S. and J.D. Helmann. (2000). Mutations in multidrug efflux homologs, sugar isomerases, and antimicrobial biosynthesis genes differentially elevate activity of the σX and σW factors in Bacillus subtilis. J. Bacteriol. 182: 5202-5210. 10960106
Uebanso, T., M. Fukui, C. Naito, T. Shimohata, K. Mawatari, and A. Takahashi. (2023). SLC16a6, mTORC1, and Autophagy Regulate Ketone Body Excretion in the Intestinal Cells. Biology (Basel) 12:. 38132294
Uebe, R. and D. Schüler. (2016). Magnetosome biogenesis in magnetotactic bacteria. Nat. Rev. Microbiol. 14: 621-637. 27620945
Uehara, M., A. Fukumoto, H. Omote, and M. Hiasa. (2024). Polyamine release and vesicular polyamine transporter expression in megakaryoblastic cells and platelets. Biochim. Biophys. Acta. Gen Subj 1868: 130610. 38527572
Uemura, T., K. Tachihara, H. Tomitori, K. Kashiwagi, K. Igarashi. (2005). Characteristics of the polyamine transporter TPO1 and regulation of its activity and cellular localization by phosphorylation. J. Biol. Chem. 280: 9646-9652. 15637075
Uldry, M., M. Ibberson, J.D. Horisberger, J.Y. Chatton, B.M. Riederer, and B. Thorens. (2001). Identification of a mammalian H+-myo-inositol symporter expressed predominantly in the brain. EMBO J. 20: 4467-4477. 11500374
Uldry, M., M. Ibberson, M. Hosokawa, and B. Thorens. (2002). GLUT2 is a high affinity glucosamine transporter. FEBS Lett. 524: 199-203. 12135767
Ullán, R.V., G. Liu, J. Casqueiro, S. Gutiérrez, O. Bañuelos, and J.F. Martín. (2002). The cefT gene of Acremonium chrysogenum C10 encodes a putative multidrug efflux pump protein that significantly increases cephalosporin C production. Mol. Genet. Genomics 267: 673-683. 12172807
Unkles SE., Karabika E., Symington VF., Cecile JL., Rouch DA., Akhtar N., Cromer BA. and Kinghorn JR. (2012). Alanine scanning mutagenesis of a high-affinity nitrate transporter highlights the requirement for glycine and asparagine residues in the two nitrate signature motifs. Biochem J. 447(1):35-42. 22738143
Unkles, S.E., K.L. Hawker, C. Grieve, E.I. Campbell, P. Montague, and J.R. Kinghorn. (1991). crnA encodes a nitrate transporter in Aspergillus nidulans. Proc. Natl. Acad. Sci. USA 88: 204-208. 1986367
Unkles, S.E., V.F. Symington, Z. Kotur, Y. Wang, M.Y. Siddiqi, J.R. Kinghorn, and A.D. Glass. (2011). Physiological and biochemical characterization of AnNitA, the Aspergillus nidulans high-affinity nitrite transporter. Eukaryot. Cell. 10: 1724-1732. 22021238
Unno, K., K. Taguchi, Y. Takagi, T. Hase, S. Meguro, and Y. Nakamura. (2023). Mouse Models with SGLT2 Mutations: Toward Understanding the Role of SGLT2 beyond Glucose Reabsorption. Int J Mol Sci 24:. 37047250
Uwai, Y., Y. Ozeki, T. Isaka, H. Honjo, and K. Iwamoto. (2011). Inhibitory effect of caffeic acid on human organic anion transporters hOAT1 and hOAT3: a novel candidate for food-drug interaction. Drug Metab Pharmacokinet 26: 486-493. 21697612
Vale, C., P.J. Godolphin, D. Fisher, P.W. Horby, M.N. Kosiborod, J.S. Hochman, K. Webster, J.P.T. Higgins, A.D. Althouse, O. Berwanger, R.H.M. Furtado, S.B. Gasparyan, R. Haynes, G.G. Koch, M. Landray, E. Leifer, J. Marshall, S. Murthy, M.D. Neal, N. Staplin, J. Diaz, J.A.C. Sterne, M. Shankar-Hari, and. (2024). Sodium-glucose co-transporter-2 inhibitors for hospitalised patients with COVID-19: a prospective meta-analysis of randomised trials. Lancet Diabetes Endocrinol 12: 735-747. 39250923
Valifard, M., A.R. Fernie, A. Kitashova, T. Nägele, R. Schröder, M. Meinert, B. Pommerrenig, D. Mehner-Breitfeld, C.P. Witte, T. Brüser, I. Keller, and H.E. Neuhaus. (2023). The novel chloroplast glucose transporter pGlcT2 affects adaptation to extended light periods. J. Biol. Chem. 299: 104741. 37088133
Vallon, V. (2020). Glucose transporters in the kidney in health and disease. Pflugers Arch. [Epub: Ahead of Print] 32144488
Valmeekam, V., Y.L. Loh, and M.J. San Francisco. (2001). Control of exuT activity for galacturonate transport by the negative regulator ExuR in Erwinia chrysanthemi EC16. Mol. Plant Microbe Interact. 14: 816-820. 11386378
Van Camp, B.M., R.R. Crow, Y. Peng, and M.F. Varela. (2007). Amino acids that confer transport of raffinose and maltose sugars in the raffinose permease (RafB) of Escherichia coli as implicated by spontaneous mutations at Val-35, Ser-138, Ser-139, Gly-389 and Ile-391. J. Membr. Biol. 220: 87-95. 18008022
van Geest, F.S., M.E. Meima, K.E. Stuurman, N.I. Wolf, M.S. van der Knaap, C.F. Lorea, F.O. Poswar, F. Vairo, N. Brunetti-Pierri, G. Cappuccio, P. Bakhtiani, S.A. de Munnik, R.P. Peeters, W.E. Visser, and S. Groeneweg. (2020). Clinical and functional consequences of C-terminal variants in MCT8: a case series. J Clin Endocrinol Metab. [Epub: Ahead of Print] 33141165
van Vliet, A.R., S. De Tito, E. Almacellas, and S.A. Tooze. (2023). Imaging ATG9A, a Multi-Spanning Membrane Protein. J Vis Exp. 37395569
van Wezel, G.P., K. Mahr, M. König, B.A. Traag, E.F. Pimentel-Schmitt, A. Willimek, and F. Titgemeyer. (2005). GlcP constitutes the major glucose uptake system of Streptomyces coelicolor A3(2). Mol. Microbiol. 55: 624-636. 15659175
Vancamp, P. and V.M. Darras. (2017). From zebrafish to human: A comparative approach to elucidate the role of the thyroid hormone transporter MCT8 during brain development. Gen Comp Endocrinol. [Epub: Ahead of Print] 29183795
Vardy, E., S. Steiner-Mordoch, and S. Schuldiner. (2005). Characterization of bacterial drug antiporters homologous to mammalian neurotransmitter transporters. J. Bacteriol. 187: 7518-7525. 16237035
Vasamsetti, B.M.K., K. Chon, C.Y. Yoon, J. Kim, J.Y. Choi, S. Hwang, and K.H. Park. (2023). Transcriptome Profiling of Etridiazole-Exposed Zebrafish () Embryos Reveals Pathways Associated with Cardiac and Ocular Toxicities. Int J Mol Sci 24:. 37894748
Västermark, &.#.1.9.7.;., J.A. Jacobsson, &.#.1.9.7.;. Johansson, R. Fredriksson, U. Gyllensten, and H.B. Schiöth. (2012). Polymorphisms in sh2b1 and spns1 loci are associated with triglyceride levels in a healthy population in northern Sweden. J Genet 91: 237-240. 22942098
Västermark, A. and M.H. Saier. (2014). Major Facilitator Superfamily (MFS) evolved without 3-transmembrane segment unit rearrangements. Proc. Natl. Acad. Sci. USA 111: E1162-1163. 24567407
Västermark, A., B. Lunt, and M. Saier. (2014). Major facilitator superfamily porters, LacY, FucP and XylE of Escherichia coli appear to have evolved positionally dissimilar catalytic residues without rearrangement of 3-TMS repeat units. J. Mol. Microbiol. Biotechnol. 24: 82-90. 24603210
Vela-Corcía, D., D. Aditya Srivastava, A. Dafa-Berger, N. Rotem, O. Barda, and M. Levy. (2019). MFS transporter from Botrytis cinerea provides tolerance to glucosinolate-breakdown products and is required for pathogenicity. Nat Commun 10: 2886. 31253809
Verheijen, F.W., E. Verbeek, N. Aula, C.E. Beerens, A.C. Havelaar, M. Joosse, L. Peltonen, R. Aula, H. Galjaard, P.J. van der Spek, and G.M. Mancini. (1999). A new gene, encoding an anion transporter, is mutated in sialic acid storage diseases. Nature Genet. 23: 462-465. 10581036
Versaw, W.K. (1995). A phosphate-repressible, high-affinity phosphate permease is encoded by the pho-5+ gene of Neurospora crassa. Gene 153: 135-139. 7883177
Vieira, N., M. Casal, B. Johansson, D.M. MacCallum, A.J. Brown, and S. Paiva. (2010). Functional specialization and differential regulation of short-chain carboxylic acid transporters in the pathogen Candida albicans. Mol. Microbiol. 75: 1337-1354. 19968788
Viigand, K., K. Tammus, and T. Alamäe. (2005). Clustering of MAL genes in Hansenula polymorpha: cloning of the maltose permease gene and expression from the divergent intergenic region between the maltose permease and maltase genes. FEMS Yeast Res 5: 1019-1028. 16103021
Vilhena, C., E. Kaganovitch, J.Y. Shin, A. Grünberger, S. Behr, I. Kristoficova, S. Brameyer, D. Kohlheyer, and K. Jung. (2017). A single-cell view of the BtsSR/YpdAB pyruvate sensing network in Escherichia coli and its biological relevance. J. Bacteriol. [Epub: Ahead of Print] 29038258
Villagra, N.A., A.A. Hidalgo, C.A. Santiviago, C.P. Saavedra, and G.C. Mora. (2008). SmvA, and not AcrB, is the major efflux pump for acriflavine and related compounds in Salmonella enterica serovar Typhimurium. J Antimicrob Chemother 62: 1273-1276. 18819967
Visser, W.E., N.J. Philp, T.B. van Dijk, W. Klootwijk, E.C. Friesema, J. Jansen, P.W. Beesley, A.G. Ianculescu, and T.J. Visser. (2009). Evidence for a homodimeric structure of human monocarboxylate transporter 8. Endocrinology 150: 5163-5170. 19797118
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. 17173541
Vitrac, H., V.K.P.S. Mallampalli, M. Bogdanov, and W. Dowhan. (2019). The lipid-dependent structure and function of LacY can be recapitulated and analyzed in phospholipid-containing detergent micelles. Sci Rep 9: 11338. 31383935
Vitrac, H., V.K.P.S. Mallampalli, S. Azinas, and W. Dowhan. (2020). Structural and Functional Adaptability of Sucrose and Lactose Permeases from to the Membrane Lipid Composition. Biochemistry. [Epub: Ahead of Print] 32363862
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. 12136096
Vogl, C., C.M. Klein, A.F. Batke, M.E. Schweingruber, and J. Stolz. (2008). Characterization of Thi9, a novel thiamine (Vitamin B1) transporter from Schizosaccharomyces pombe. J. Biol. Chem. 283: 7379-7389. 18201975
Volkova M., Mandikova J., Barta P., Navratilova L., Laznickova A. and Trejtnar F. (2015). The in vivo disposition and in vitro transmembrane transport of two model radiometabolites of DOTA-conjugated receptor-specific peptides labelled with (177) Lu. J Labelled Comp Radiopharm. 58(13-14):483-9. 26526343
von Rozycki, T., M.R. Yen, E.E. Lende, and M.H. Saier, Jr. (2004). The YedZ family: possible heme binding proteins that can be fused to transporters and electron carriers. J. Mol. Microbiol. Biotechnol. 8: 129-140. 16088215
Wagai, S., A. Kasamatsu, M. Iyoda, F. Hayashi, K. Hiroshima, S. Yoshimura, I. Miyamoto, D. Nakashima, Y. Endo-Sakamoto, M. Shiiba, H. Tanzawa, and K. Uzawa. (2019). UNC93B1 promotes tumoral growth by controlling the secretion level of granulocyte macrophage colony-stimulating factor in human oral cancer. Biochem. Biophys. Res. Commun. 513: 81-87. 30935694
Wagenaars, F., P. Cenijn, M. Scholze, C. Frädrich, K. Renko, J. Köhrle, and T. Hamers. (2024). Screening for endocrine disrupting chemicals inhibiting monocarboxylate 8 (MCT8) transporter facilitated thyroid hormone transport using a modified nonradioactive assay. Toxicol In Vitro 96: 105770. 38151217
Walters, D.K., B.K. Arendt, and D.F. Jelinek. (2013). CD147 regulates the expression of MCT1 and lactate export in multiple myeloma cells. Cell Cycle 12: 3175-3183. 24013424
Wambo, T.O., L.Y. Chen, C. Phelix, and G. Perry. (2017). Affinity and path of binding xylopyranose unto E. coli xylose permease. Biochem. Biophys. Res. Commun. [Epub: Ahead of Print] 29032199
Wang C., Shen Y., Hou J., Suo F. and Bao X. (2013). An assay for functional xylose transporters in Saccharomyces cerevisiae. Anal Biochem. 442(2):241-8. 23928049
Wang M., Qi H., Li J., Xu Y. and Zhang H. (2015). Transmembrane transport of steviol glucuronide and its potential interaction with selected drugs and natural compounds. Food Chem Toxicol. 86:217-24. 26525112
Wang, C., F.Q. Zhao, J. Liu, and H. Liu. (2020). Short communication: The essential role of N-glycosylation in the transport activity of bovine peptide transporter 2. J Dairy Sci. [Epub: Ahead of Print] 32331895
Wang, C., L. Yang, A.A. Shah, E.S. Choi, and S.W. Kim. (2015). Dynamic interplay of multidrug transporters with TolC for isoprenol tolerance in Escherichia coli. Sci Rep 5: 16505. 26563610
Wang, C., X. Bao, Y. Li, C. Jiao, J. Hou, Q. Zhang, W. Zhang, W. Liu, and Y. Shen. (2015). Data set for cloning and characterization of heterologous transporters in Saccharomyces cerevisiae and identification of important amino acids for xylose utilization. Data Brief 4: 119-126. 26217774
Wang, D., H. Wu, J. Yang, M. Li, C. Ling, Z. Gao, H. Lu, H. Shen, and Y. Tang. (2022). Loss of SLC46A1 decreases tumor iron content in hepatocellular carcinoma. Hepatol Commun. [Epub: Ahead of Print] 35811443
Wang, J., T. Li, X. Wu, and Z. Zhao. (2014). Molecular cloning and functional analysis of a H+-dependent phosphate transporter gene from the ectomycorrhizal fungus Boletus edulis in southwest China. Fungal Biol 118: 453-461. 24863474
Wang, N., X. Jiang, S. Zhang, A. Zhu, Y. Yuan, H. Xu, J. Lei, and C. Yan. (2020). Structural basis of human monocarboxylate transporter 1 inhibition by anti-cancer drug candidates. Cell. [Epub: Ahead of Print] 33333023
Wang, Q. and M.E. Morris. (2007). The role of monocarboxylate transporter 2 and 4 in the transport of γ-hydroxybutyric acid in mammalian cells. Drug Metab Dispos 35: 1393-1399. 17502341
Wang, Q., H. Li, and A.F. Post. (2000). Nitrate assimilation genes of the marine diazotrophic, filamentous cyanobacterium Trichodesmium sp. strain WH9601. J. Bacteriol. 182: 1764-1767. 10692386
Wang, S. and L. Qin. (2022). Homeostatic medicine: a strategy for exploring health and disease. Curr Med (Cham) 1: 16. 36189427
Wang, S., K. Wang, K. Song, P. Li, D. Li, Y. Sun, Y. Mei, C. Xu, and M. Liao. (2024). Structures of the essential efflux pump EfpA from reveal the mechanisms of substrate transport and small-molecule inhibition. Res Sq. 38260587
Wang, T., J. Wang, X. Hu, X.J. Huang, and G.X. Chen. (2020). Current understanding of glucose transporter 4 expression and functional mechanisms. World J. Biol. Chem. 11: 76-98. 33274014
Wang, T., Y. Wang, A. Montero-Pedrazuela, L. Prensa, A. Guadaño-Ferraz, and E. Rausell. (2023). Thyroid Hormone Transporters MCT8 and OATP1C1 Are Expressed in Projection Neuron.s and Interneurons of Basal Ganglia and Motor Thalamus in the Adult Human and Macaque Brains. Int J Mol Sci 24:. 37298594
Wang, W. and H.W. van Veen. (2012). Basic residues R260 and K357 affect the conformational dynamics of the major facilitator superfamily multidrug transporter LmrP. PLoS One 7: e38715. 22761697
Wang, W., A.A. Guffanti, Y. Wei, M. Ito, and T.A. Krulwich. (2000). Two types of Bacillus subtilis tetA(L) deletion strains reveal the physiological importance of TetA(L) in K+ acquisition as well as in Na+, alkali, and tetracycline resistance. J. Bacteriol. 182: 2088-2095. 10735849
Wang, W., Z. Li, S. Yuan, Z. Du, J. Li, H. Peng, and S. Ru. (2024). A Potential Neurotoxic Mechanism: Bisphenol S-Induced Inhibition of Glucose Transporter 1 Leads to ATP Excitotoxicity in the Zebrafish Brain. Environ Sci Technol 58: 15463-15474. 39167196
Wang, W.A. and N. Demaurex. (2022). The mammalian trafficking chaperone protein UNC93B1 maintains the ER calcium sensor STIM1 in a dimeric state primed for translocation to the ER cortex. J. Biol. Chem. 298: 101607. 35065962
Wang, X., R.I. Sarker, and P.C. Maloney. (2006). Analysis of substrate-binding elements in OxlT, the oxalate:formate antiporter of Oxalobacter formigenes. Biochemistry 45: 10344-10350. 16922510
Wang, Y., T. Wang, A. Montero-Pedrazuela, A. Guadaño-Ferraz, and E. Rausell. (2023). Thyroid Hormone Transporters MCT8 and OATP1C1 Are Expressed in Pyramidal Neuron.s and Interneurons in the Adult Motor Cortex of Human and Macaque Brain. Int J Mol Sci 24:. 36834621
Wang, Y., W. Li, Y. Siddiqi, V.F. Symington, J.R. Kinghorn, S.E. Unkles, and A.D. Glass. (2008). Nitrite transport is mediated by the nitrite-specific high-affinity NitA transporter and by nitrate transporters NrtA, NrtB in Aspergillus nidulans. Fungal Genet Biol 45: 94-102. 18024100
Watson, E.T., M.M. Pauers, M.J. Seibert, J.D. Vevea, and E.R. Chapman. (2023). Synaptic vesicle proteins are selectively delivered to axons in mammalian neurons. Elife 12:. 36729040
Wei, H., K. Vienken, R. Weber, S. Bunting, N. Requena, and R. Fischer. (2004). A putative high affinity hexose transporter, hxtA, of Aspergillus nidulans is induced in vegetative hyphae upon starvation and in ascogenous hyphae during cleistothecium formation. Fungal Genet Biol 41: 148-156. 14732261
Wei, X., Y. Fu, R. Yu, L. Wu, Z. Wu, P. Tian, S. Li, X. Yang, and M. Yang. (2022). Comprehensive sequence and expression profile analysis of the phosphate transporter gene family in soybean. Sci Rep 12: 20883. 36463363
Weijers, R.N. (2020). Fundamentals about onset and progressive disease character of type 2 diabetes mellitus. World J Diabetes 11: 165-181. 32477453
Wichers, J.S., P. Mesén-Ramírez, G. Fuchs, J. Yu-Strzelczyk, J. Stäcker, H. von Thien, A. Alder, I. Henshall, B. Liffner, G. Nagel, C. Löw, D. Wilson, T. Spielmann, S. Gao, T.W. Gilberger, A. Bachmann, and J. Strauss. (2022). PMRT1, a -Specific Parasite Plasma Membrane Transporter, Is Essential for Asexual and Sexual Blood Stage Development. mBio 13: e0062322. 35404116
Widiasih Widiyanto, T., X. Chen, S. Iwatani, H. Chibana, and S. Kajiwara. (2019). Role of major facilitator superfamily transporter Qdr2p in biofilm formation by Candida glabrata. Mycoses 62: 1154-1163. 31519064
Wieczorke, R., S. Krampe, T. Weierstall, K. Freidel, C.P. Hollenberg, and E. Boles. (1999). Concurrent knock-out of at least 20 transporter genes is required to block uptake of hexoses in Saccharomyces cerevisiae. FEBS Lett. 464: 123-128. 10618490
Williams, P.A. and L.E. Shaw. (1997). mucK, a gene in Acinetobacter calcoaceticus ADP1 (BD413), encodes the ability to grow on exogenous cis,cis-muconate as the sole carbon source. J. Bacteriol. 179: 5935-5942. 9294455
Wilpert, N.M., M. Krueger, R. Opitz, D. Sebinger, S. Paisdzior, B. Mages, A. Schulz, J. Spranger, E.K. Wirth, H. Stachelscheid, P. Mergenthaler, P. Vajkoczy, H. Krude, P. Kühnen, I. Bechmann, and H. Biebermann. (2020). Spatiotemporal Changes of Cerebral Monocarboxylate Transporter 8 Expression. Thyroid. [Epub: Ahead of Print] 32143555
Wilson MR., Kugel S., Huang J., Wilson LJ., Wloszczynski PA., Ye J., Matherly LH. and Hou Z. (2015). Structural determinants of human proton-coupled folate transporter oligomerization: role of GXXXG motifs and identification of oligomeric interfaces at transmembrane domains 3 and 6. Biochem J. 469(1):33-44. 25877470
Wilson, M.C., D. Meredith, J.E. Fox, C. Manoharan, A.J. Davies, and A.P. Halestrap. (2005). Basigin (CD147) is the target for organomercurial inhibition of monocarboxylate transporter isoforms 1 and 4: the ancillary protein for the insensitive MCT2 is EMBIGIN (gp70). J. Biol. Chem. 280: 27213-27221. 15917240
Wilson, M.C., V.N. Jackson, C. Heddle, N.T. Price, H. Pilegaard, C. Juel, A. Bonen, I. Montgomery, O.F. Hutter, and A.P. Halestrap. (1998). Lactic acid efflux from white skeletal muscle is catalyzed by the monocarboxylate transporter isoform MCT3. J. Biol. Chem. 273: 15920-15926. 9632638
Wilson, M.R., Z. Hou, and L.H. Matherly. (2014). Substituted Cysteine Accessibility Reveals a Novel Transmembrane 2-3 Reentrant Loop and Functional Role for Transmembrane Domain 2 in the Human Proton-coupled Folate Transporter. J. Biol. Chem. 289: 25287-25295. 25053408
Wilson-O''Brien, A.L., N. Patron, and S. Rogers. (2010). Evolutionary ancestry and novel functions of the mammalian glucose transporter (GLUT) family. BMC Evol Biol 10: 152. 20487568
Wirth, E.K., S.Y. Sheu, J. Chiu-Ugalde, R. Sapin, M.O. Klein, I. Mossbrugger, L. Quintanilla-Martinez, M.H. de Angelis, H. Krude, T. Riebel, K. Rothe, J. Köhrle, K.W. Schmid, U. Schweizer, and A. Grüters. (2011). Monocarboxylate transporter 8 deficiency: altered thyroid morphology and persistent high triiodothyronine/thyroxine ratio after thyroidectomy. Eur J Endocrinol 165: 555-561. 21813593
Wirth, E.K., U. Schweizer, and J. Köhrle. (2014). Transport of thyroid hormone in brain. Front Endocrinol (Lausanne) 5: 98. 25009532
Wirth, J., F. Chopin, V. Santoni, G. Viennois, P. Tillard, A. Krapp, L. Lejay, F. Daniel-Vedele, and A. Gojon. (2007). Regulation of root nitrate uptake at the NRT2.1 protein level in Arabidopsis thaliana. J. Biol. Chem. 282: 23541-23552. 17573350
Witkowska K., Smith KM., Yao SY., Ng AM., O'Neill D., Karpinski E., Young JD. and Cheeseman CI. (2012). Human SLC2A9a and SLC2A9b isoforms mediate electrogenic transport of urate with different characteristics in the presence of hexoses. Am J Physiol Renal Physiol. 303(4):F527-39. 22647630
Wolf, L., M. Föller, and M. Feger. (2023). The impact of SGLT2 inhibitors on αKlotho in renal MDCK and HK-2 cells. Front Endocrinol (Lausanne) 14: 1069715. 36967770
Wollack, J.B., B. Makori, S. Ahlawat, R. Koneru, S.C. Picinich, A. Smith, I.D. Goldman, A. Qiu, P.D. Cole, J. Glod, and B. Kamen. (2008). Characterization of folate uptake by choroid plexus epithelial cells in a rat primary culture model. J Neurochem 104(6): 1494-1503. 18086128
Wood, N.J., T. Alizadeh, D.J. Richardson, S.J. Ferguson, and J.W.B. Moir. (2002). Two domains of a dual-function NarK protein are required for nitrate uptake, the first step of denitrification in Paracoccus pantotrophus. Mol. Microbiol. 44: 157-170. 11967076
Woolley, R.C., G. Vediyappan, M. Anderson, M. Lackey, B. Ramasubramanian, B. Jiangping, T. Borisova, J.A. Colmer, A.N. Hamood, C.S. McVay, and J.A. Fralick. (2005). Characterization of the Vibrio cholerae vceCAB multiple-drug resistance efflux operon in Escherichia coli. J. Bacteriol. 187: 5500-5503. 16030246
Woolridge, D.P., N. Vazquez-Laslop, P.N. Markham, M.S. Chevalier, E.W. Gerner, and A.A. Neyfakh. (1997). Efflux of the natural polyamine spermidine facilitated by the Bacillus subtilis multidrug transporter Blt. J. Biol. Chem. 272: 8864-8866. 9083003
Wright DJ. and Tate CG. (2015). Isolation and characterisation of transport-defective substrate-binding mutants of the tetracycline antiporter TetA(B). Biochim Biophys Acta. 1848(10 Pt A):2261-2270. 26143388
Wright, D.C. (2007). Mechanisms of calcium-induced mitochondrial biogenesis and GLUT4 synthesis. Appl Physiol Nutr Metab 32: 840-845. 18059607
Wright, M.B., E.A. Howell, and R.F. Gaber. (1996). Amino acid substitutions in membrane-spanning domains of Hol1, a member of the major facilitator superfamily of transporters, confer nonselective cation uptake in Saccharomyces cerevisiae. J. Bacteriol. 178: 7197-7205. 8955402
Wu, W., K.T. Bush, H.C. Liu, C. Zhu, R. Abagyan, and S.K. Nigam. (2015). Shared Ligands Between Organic Anion Transporters (OAT1 and OAT6) and Odorant Receptors. Drug Metab Dispos 43: 1855-1863. 26358290
Wu, W., N. Jamshidi, S.A. Eraly, H.C. Liu, K.T. Bush, B.O. Palsson, and S.K. Nigam. (2013). Multispecific drug transporter slc22a8 (oat3) regulates multiple metabolic and signaling pathways. Drug Metab Dispos 41: 1825-1834. 23920220
Wu, X., R. Kekuda, W. Huang, Y.J. Fei, F.H. Leibach, J. Chen, S.J. Conway, and V. Ganapathy. (1998). Identity of the organic cation transporter OCT3 as the extraneuronal monoamine transporter (uptake2) and evidence for the expression of the transporter in the brain. J. Biol. Chem. 273: 32776-32786. 9830022
Wu, X., Y.J. Fei, W. Huang, C. Chancy, F.H. Leibach, and V. Ganapathy. (1999). Identity of the F52F12.1 gene product in Caenorhabditis elegans as an organic cation transporter. Biochim. Biophys. Acta. 1418: 239-244. 10209228
Wuchiyama, J., M. Kimura, and I. Yamaguchi. (2000). A trichothecene efflux pump encoded by Tri102 in the biosynthesis gene cluster of Fusarium graminearum. J Antibiot (Tokyo) 53: 196-200. 10805582
Wunderlich, J. (2022). Updated List of Transport Proteins in. Front Cell Infect Microbiol 12: 926541. 35811673
Xi, Y., T. Zhan, H. Xu, J. Chen, C. Bi, F. Fan, and X. Zhang. (2021). Characterization of JEN family carboxylate transporters from the acid-tolerant yeast Pichia kudriavzevii and their applications in succinic acid production. Microb Biotechnol. [Epub: Ahead of Print] 33629807
Xie, X., H. Lin, X. Peng, C. Xu, Z. Sun, K. Jiang, A. Huang, X. Wu, N. Tang, A. Salvioli, P. Bonfante, and B. Zhao. (2016). Arbuscular Mycorrhizal Symbiosis Requires a Phosphate Transceptor in the Gigaspora margarita Fungal Symbiont. Mol Plant 9: 1583-1608. 27688206
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. 29065880
Xiong, Y., D. Delic, S. Zeng, X. Chen, C. Chu, A.A. Hasan, B.K. Krämer, T. Klein, L. Yin, and B. Hocher. (2022). Regulation of SARS CoV-2 host factors in the kidney and heart in rats with 5/6 nephrectomy-effects of salt, ARB, DPP4 inhibitor and SGLT2 blocker. BMC Nephrol 23: 117. 35331159
Xu Y., Chen B., Chao H. and Zhou NY. (2013). mhpT encodes an active transporter involved in 3-(3-hydroxyphenyl)propionate catabolism by Escherichia coli K-12. Appl Environ Microbiol. 79(20):6362-8. 23934492
Xu, X., J. Chen, H. Xu, and D. Li. (2014). Role of a major facilitator superfamily transporter in adaptation capacity of Penicillium funiculosum under extreme acidic stress. Fungal Genet Biol 69: 75-83. 24959657
Yadav, V., M. Kumar, D.K. Deep, H. Kumar, R. Sharma, T. Tripathi, N. Tuteja, A.K. Saxena, and A.K. Johri. (2010). A phosphate transporter from the root endophytic fungus Piriformospora indica plays a role in phosphate transport to the host plant. J. Biol. Chem. 285: 26532-26544. 20479005
Yaffe, D., S. Radestock, Y. Shuster, L.R. Forrest, and S. Schuldiner. (2013). Identification of molecular hinge points mediating alternating access in the vesicular monoamine transporter VMAT2. Proc. Natl. Acad. Sci. USA 110: E1332-1341. 23530208
Yamada, K., Y. Saijo, H. Nakagami, and Y. Takano. (2016). Regulation of sugar transporter activity for antibacterial defense in Arabidopsis. Science 354: 1427-1430. 27884939
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. 16820466
Yamada, Y., J. Sakuma, I. Takeuchi, Y. Yasukochi, K. Kato, M. Oguri, T. Fujimaki, H. Horibe, M. Muramatsu, M. Sawabe, Y. Fujiwara, Y. Taniguchi, S. Obuchi, H. Kawai, S. Shinkai, S. Mori, T. Arai, and M. Tanaka. (2017). Identification of TNFSF13, SPATC1L, SLC22A25 and SALL4 as novel susceptibility loci for atrial fibrillation by an exome‑wide association study. Mol Med Rep 16: 5823-5832. 28849223
Yamada, Y., K. Hideka, S. Shiota, T. Kuroda, and T. Tsuchiya. (2006). Gene cloning and characterization of SdrM, a chromosomally-encoded multidrug efflux pump, from Staphylococcus aureus. Biol Pharm Bull 29: 554-556. 16508166
Yamada, Y., S. Shiota, T. Mizushima, T. Kuroda, and T. Tsuchiya. (2006). Functional gene cloning and characterization of MdeA, a multidrug efflux pump from Staphylococcus aureus. Biol Pharm Bull 29: 801-804. 16595922
Yamagata, A., K. Ito, T. Suzuki, N. Dohmae, T. Terada, and M. Shirouzu. (2024). Structural basis for antiepileptic drugs and botulinum neurotoxin recognition of SV2A. Nat Commun 15: 3027. 38637505
Yamaguchi, A., Y. Mukai, T. Sakuma, A. Furugen, K. Narumi, and M. Kobayashi. (2023). Atorvastatin Exerts More Selective Inhibitory Effects on hMCT2 than on hMCT1 and hMCT4. Anticancer Res 43: 3015-3022. 37351987
Yanagisawa, H., T. Miyashita, Y. Nakano, and D. Yamamoto. (2003). HSpin1, a transmembrane protein interacting with Bcl-2/Bcl-xL, induces a caspase-independent autophagic cell death. Cell Death Differ. 10: 798-807. 12815463
Yanatori, I., Y. Yasui, K. Miura, and F. Kishi. (2012). Mutations of FLVCR1 in posterior column ataxia and retinitis pigmentosa result in the loss of heme export activity. Blood Cells Mol Dis 49: 60-66. 22483575
Yang, C., D.A. Rodionov, X. Li, O.N. Laikova, M.S. Gelfand, O.P. Zagnitko, M.F. Romine, A.Y. Obraztsova, K.H. Nealson, and A.L. Osterman. (2006). Comparative genomics and experimental characterization of N-acetylglucosamine utilization pathway of Shewanella oneidensis. J. Biol. Chem. 281: 29872-29885. 16857666
Yang, H., G. Qin, Z. Luo, X. Kong, C. Gan, R. Zhang, and W. Jiang. (2022). MFSD4A inhibits the malignant progression of nasopharyngeal carcinoma by targeting EPHA2. Cell Death Dis 13: 332. 35410462
Yang, J., X. Xu, and G. Liu. (2012). Amplification of an MFS Transporter Encoding Gene penT Significantly Stimulates Penicillin Production and Enhances the Sensitivity of Penicillium chrysogenum to Phenylacetic Acid. J Genet Genomics 39: 593-602. 23177147
Yang, L., J. Li, Y. Li, Y. Zhou, Z. Wang, D. Zhang, J. Liu, and X. Zhang. (2021). Diclofenac impairs the proliferation and glucose metabolism of triple-negative breast cancer cells by targeting the c-Myc pathway. Exp Ther Med 21: 584. 33850556
Yang, L., Z. Chen, W. Xiong, H. Ren, E. Zhai, K. Xu, H. Yang, Z. Zhang, L. Ding, Y. He, X. Song, and J. Liu. (2019). High expression of SLC17A9 correlates with poor prognosis in colorectal cancer. Hum Pathol 84: 62-70. 30236596
Yang, R., T. Viswanatham, S. Huang, Y. Li, Y. Yu, J. Zhang, J. Chen, M. Herzberg, R. Feng, B.P. Rosen, and C. Rensing. (2024). A Sb(III)-specific efflux transporter from Ensifer adhaerens E-60. Microbiol Res 286: 127830. [Epub: Ahead of Print] 39004025
Yang, Y.J., R.P. Singh, X. Lan, C.S. Zhang, D.H. Sheng, and Y.Q. Li. (2019). Whole transcriptome analysis and gene deletion to understand the chloramphenicol resistance mechanism and develop a screening method for homologous recombination in Myxococcus xanthus. Microb Cell Fact 18: 123. 31291955
Yao, J. and S.M. Bajjalieh. (2009). SVOP is a nucleotide binding protein. PLoS One 4: e5315. 19390693
Yao, X., L. Liu, W. Shao, M. Bai, X. Ding, G. Wang, S. Wang, L. Zheng, Y. Sun, G. Wang, Y. Huang, C. Yu, Z. Song, Y. Bao, S. Yang, and L. Sun. (2023). Tectorigenin targets PKACα to promote GLUT4 expression in skeletal muscle and improve insulin resistance and. Int J Biol Sci 19: 1579-1596. 37056927
Yasuda, S., S. Hasui, M. Kobayashi, S. Itagaki, T. Hirano, and K. Iseki. (2008). The mechanism of carrier-mediated transport of folates in BeWo cells: the involvement of heme carrier protein 1 in placental folate transport. Biosci. Biotechnol. Biochem. 72: 329-334. 18256483
Ye, L. and P.C. Maloney. (2002). Structure/function relationships in OxlT, the oxalate/formate antiporter of Oxalobacter formigenes: assignment of transmembrane helix 2 to the translocation pathway. J. Biol. Chem. 277: 20372-20378. 11919184
Ye, L., Z. Jia, T. Jung, and P.C. Maloney. (2001). Toplogy of OxlT, the oxalate transporter of Oxalobacter formigenes, determined by site-directed fluorescence labeling. J. Bacteriol. 183: 2490-2496. 11274108
Yee, S.W., C. Macdonald, D. Mitrovic, X. Zhou, M.L. Koleske, J. Yang, D.B. Silva, P.R. Grimes, D. Trinidad, S.S. More, L. Kachuri, J.S. Witte, L. Delemotte, K.M. Giacomini, and W. Coyote-Maestas. (2023). The full spectrum of OCT1 (SLC22A1) mutations bridges transporter biophysics to drug pharmacogenomics. bioRxiv. 37333090
Yee, S.W., D. Buitrago, A. Stecula, H.X. Ngo, H.C. Chien, L. Zou, M.L. Koleske, and K.M. Giacomini. (2020). Deorphaning a solute carrier 22 family member, SLC22A15, through functional genomic studies. FASEB J. 34: 15734-15752. 33124720
Yi, Y., H. Zhang, M. Chen, B. Chen, Y. Chen, P. Li, H. Zhou, Z. Ma, and H. Jiang. (2023). Inhibition of multiple uptake transporters in cardiomyocytes/mitochondria alleviates doxorubicin-induced cardiotoxicity. Chem Biol Interact 382: 110627. [Epub: Ahead of Print] 37453608
Yin, Y., X. He, P. Szewczyk, T. Nguyen, and G. Chang. (2006). Structure of the multidrug transporter EmrD from Escherichia coli. Science 312: 741-744. 16675700
Yonezawa, A., S. Masuda, S. Yokoo, T. Katsura, and K. Inui. (2006). Cisplatin and oxaliplatin, but not carboplatin and nedaplatin, are substrates for human organic cation transporters (SLC22A1-3 and multidrug and toxin extrusion family). J Pharmacol Exp Ther 319: 879-886. 16914559
Yong, Z., Z. Kotur, and A.D. Glass. (2010). Characterization of an intact two-component high-affinity nitrate transporter from Arabidopsis roots. Plant J. 63: 739-748. 20561257
Yoshida, K., Y. Yamamoto, K. Omae, M. Yamamoto, and Y. Fujita. (2002). Identification of two myo-inositol transporter genes of Bacillus subtilis. J. Bacteriol. 184: 983-991. 11807058
Young, C.S. and J.T. Beatty. (1998). A topological model of the Rhodobacter capsulatus light-harvesting I complex assembly protein LlaA (previously known as ORF1696). J. Bacteriol. 180: 4742-4745. 9721320
Young, K.G., R. Hopkins, B.M. Shields, N.J. Thomas, A.P. McGovern, J.M. Dennis, and. (2023). Recent UK type 2 diabetes treatment guidance represents a near whole population indication for SGLT2-inhibitor therapy. Cardiovasc Diabetol 22: 302. 37919773
Yousefzadeh, N., S. Jeddi, M. Zarkesh, K. Kashfi, and A. Ghasemi. (2023). Altered sialin mRNA gene expression in type 2 diabetic male Wistar rats: implications for nitric oxide deficiency. Sci Rep 13: 4013. 36899088
Yu, J., D. Bhatnagar, and T.E. Cleveland. (2004). Completed sequence of aflatoxin pathway gene cluster in Aspergillus parasiticus. FEBS Lett. 564: 126-130. 15094053
Yu, J., P.K. Chang, K.C. Ehrlich, J.W. Cary, D. Bhatnagar, T.E. Cleveland, G.A. Payne, J.E. Linz, C.P. Woloshuk, and J.W. Bennett. (2004). Clustered pathway genes in aflatoxin biosynthesis. Appl. Environ. Microbiol. 70: 1253-1262. 15006741
Yu, M., Y.W. Faan, W.Y. Chung, and J.S. Tsang. (2007). Isolation and characterization of a novel haloacid permease from Burkholderia cepacia MBA4. Appl. Environ. Microbiol. 73: 4874-4880. 17545323
Yu, S., A. Vit, S. Devenish, H.K. Mahanty, A. Itzen, R.S. Goody, and W. Blankenfeldt. (2011). Atomic resolution structure of EhpR: phenazine resistance in Enterobacter agglomerans Eh1087 follows principles of bleomycin/mitomycin C resistance in other bacteria. BMC Struct Biol 11: 33. 21849072
Yu, W., L. Wang, W.Y. Ren, H.X. Xu, N.N. Wu, D.H. Yu, R.J. Reiter, W.L. Zha, Q.D. Guo, and J. Ren. (2023). SGLT2 inhibitor empagliflozin alleviates cardiac remodeling and contractile anomalies in a FUNDC1-dependent manner in experimental Parkinson''s disease. Acta Pharmacol Sin. [Epub: Ahead of Print] 37679644
Yu, Z., H. Wang, and G. You. (2023). The regulation of human organic anion transporter 4 by insulin-like growth factor 1 and protein kinase B signaling. Biochem Pharmacol 115702. [Epub: Ahead of Print] 37487877
Yuan, Y., F. Kong, H. Xu, A. Zhu, N. Yan, and C. Yan. (2022). Cryo-EM structure of human glucose transporter GLUT4. Nat Commun 13: 2671. 35562357
Yun, Y., P. Guo, J. Zhang, H. You, P. Guo, H. Deng, Y. Hao, L. Zhang, X. Wang, Y.S. Abubakar, J. Zhou, G. Lu, Z. Wang, and W. Zheng. (2020). Flippases play specific but distinct roles in the development, pathogenicity, and secondary metabolism of Fusarium graminearum. Mol Plant Pathol 21: 1307-1321. 32881238
Yusuf, I.H., M.E. Shanks, P. Clouston, and R.E. MacLaren. (2018). A splice-site variant in FLVCR1 produces retinitis pigmentosa without posterior column ataxia. Ophthalmic Genet 39: 263-267. 29192808
Zada, D., E. Blitz, and L. Appelbaum. (2017). Zebrafish - An emerging model to explore thyroid hormone transporters and psychomotor retardation. Mol. Cell Endocrinol. [Epub: Ahead of Print] 28274736
Zähner, D., X. Zhou, S.T. Chancey, J. Pohl, W.M. Shafer, and D.S. Stephens. (2010). Human antimicrobial peptide LL-37 induces MefE/Mel-mediated macrolide resistance in Streptococcus pneumoniae. Antimicrob. Agents Chemother. 54: 3516-3519. 20498319
Zakataeva, N.P., E.A. Kutukova, S.V. Gronskiĭ, P.V. Troshin, V.A. Livshits, and V.V. Aleshin. (2006). [Export of metabolites by the proteins of the DMT and RhtB families and its possible role in intercellular communication]. Mikrobiologiia 75: 509-520. 17025177
Zakataeva, N.P., S.V. Gronskiy, A.S. Sheremet, E.A. Kutukova, A.E. Novikova, and V.A. Livshits. A new function for the Bacillus PbuE purine base efflux pump: efflux of purine nucleosides. Res. Microbiol. 158(8-9): 659-665. 17935948
Zannad, F., J.P. Ferreira, J. Butler, G. Filippatos, J.L. Januzzi, M. Sumin, M. Zwick, M. Saadati, S.J. Pocock, N. Sattar, S.D. Anker, and M. Packer. (2022). Effect of Empagliflozin on Circulating Proteomics in Heart Failure: Mechanistic Insights from the EMPEROR Program. Eur Heart J. [Epub: Ahead of Print] 36017745
Zhan, C., S. Wang, Y. Sun, X. Dai, X. Liu, L. Harvey, B. McNeil, Y. Yang, and Z. Bai. (2016). The Pichia pastoris transmembrane protein GT1 is a glycerol transporter and relieves the repression of glycerol on AOX1 expression. FEMS Yeast Res 16:. 27189360
Zhang Z., Wang R. and Xie J. (2015). Mycobacterium smegmatis MSMEG_3705 encodes a selective major facilitator superfamily efflux pump with multiple roles. Curr Microbiol. 70(6):801-9. 25697715
Zhang, C., X. Wei, G.S. Omenn, and Y. Zhang. (2018). Structure and Protein Interaction-based Gene Ontology Annotations Reveal Likely Functions of Uncharacterized Proteins on Human Chromosome 17. J Proteome Res. [Epub: Ahead of Print] 30265558
Zhang, C.C., M.C. Durand, R. Jeanjean, and F. Joset. (1989). Molecular and genetical analysis of the fructose-glucose transport system in the cyanobacterium Synechocystis PCC6803. Mol. Microbiol. 3: 1221-1229. 2507869
Zhang, F.X., S.N. Ge, Y.L. Dong, J. Shi, Y.P. Feng, Y. Li, Y.Q. Li, and J.L. Li. (2018). Vesicular glutamate transporter isoforms: The essential players in the somatosensory systems. Prog Neurobiol 171: 72-89. 30273635
Zhang, J., X. Xu, Y. Zhao, C. Ren, M. Gu, H. Zhang, P. Wu, Y. Wang, L. Kong, and C. Han. (2023). Target Separation and Potential Anticancer Activity of Withanolide-Based Glucose Transporter Protein 1 Inhibitors from var. J Nat Prod. [Epub: Ahead of Print] 38117981
Zhang, L. and L. Xu. (2023). Fgf2 and Ptpn11 play a role in cerebral injury caused by sevoflurane anesthesia. Medicine (Baltimore) 102: e36108. 37960778
Zhang, L., Q. Yang, Y. Zhang, X. Li, R. Dong, C. Song, L. Cheng, and H. Zhao. (2020). [Effects of resveratrol and soy isoflavones on learning and memory ability and expression of glucose transporter in aging model rats]. Wei Sheng Yan Jiu 49: 249-253. 32290941
Zhang, L., T. Gui, L. Console, M. Scalise, C. Indiveri, S. Hausler, G.A. Kullak-Ublick, Z. Gai, and M. Visentin. (2020). Cholesterol stimulates the cellular uptake of L-carnitine by the carnitine/organic cation transporter novel 2 (OCTN2). J. Biol. Chem. [Epub: Ahead of Print] 33334877
Zhang, L., W. Guo, Y. Lu, T. Zhou, Y. Wang, X. Tang, and J. Zhang. (2023). Genome-wide characterization of the inositol transporters gene family in Populus and functional characterization of PtINT1b in response to salt stress. Int J Biol Macromol 228: 197-206. 36572075
Zhang, P., P. Azad, D.C. Engelhart, G.G. Haddad, and S.K. Nigam. (2021). SLC22 Transporters in the Fly Renal System Regulate Response to Oxidative Stress In Vivo. Int J Mol Sci 22:. 34948211
Zhang, W., S. Wang, F. Yu, J. Tang, L. Yu, H. Wang, and J. Li. (2019). Genome-Wide Identification and Expression Profiling of Sugar Transporter Protein (STP) Family Genes in Cabbage (Brassica oleracea var. capitata L.) Reveals their Involvement in Clubroot Disease Responses. Genes (Basel) 10:. 30669698
Zhang, W., Y. Liao, P. Shao, Y. Yang, L. Huang, Z. Du, C. Zhang, Y. Wang, Y. Lin, and J. Zhu. (2024). Integrated analysis of differently expressed microRNAs and mRNAs at different postnatal stages reveals intramuscular fat deposition regulation in goats (Capra hircus). Anim Genet. [Epub: Ahead of Print] 38175181
Zhang, X., J. Feng, R. Zhao, H. Cheng, J. Ashraf, Q. Wang, L. Lv, Y. Zhang, G. Song, and D. Zuo. (2023). Functional characterization of the gene reveals its significant role in improving nitrogen use efficiency in. PeerJ 11: e15152. 37009157
Zhang, X., K. Garbett, K. Veeraraghavalu, B. Wilburn, R. Gilmore, K. Mirnics, and S.S. Sisodia. (2012). A role for presenilins in autophagy revisited: normal acidification of lysosomes in cells lacking PSEN1 and PSEN2. J. Neurosci. 32: 8633-8648. 22723704
Zhang, X., X. Wen, Y. Si, D. Li, C. Yang, L. Wang, and L. Song. (2024). Regulation Mechanisms of the Glutamate Transporter in the Response of Pacific Oyster upon High-Temperature Stress. Int J Mol Sci 25:. 39518895
Zhang, X.C., Y. Zhao, J. Heng, and D. Jiang. (2015). Energy coupling mechanisms of MFS transporters. Protein. Sci. 24: 1560-1579. 26234418
Zhang, Y., Q. Bao, L.A. Gagnon, A. Huletsky, A. Oliver, S. Jin, and T. Langaee. (2010). ampG gene of Pseudomonas aeruginosa and its role in β-lactamase expression. Antimicrob. Agents Chemother. 54: 4772-4779. 20713660
Zhang, Z., X. Li, F. Yang, C. Chen, P. Liu, Y. Ren, P. Sun, Z. Wang, Y. You, Y.X. Zeng, and X. Li. (2021). DHHC9-mediated GLUT1 S-palmitoylation promotes glioblastoma glycolysis and tumorigenesis. Nat Commun 12: 5872. 34620861
Zhao R., Shin DS., Fiser A. and Goldman ID. (2012). Identification of a functionally critical GXXG motif and its relationship to the folate binding site of the proton-coupled folate transporter (PCFT-SLC46A1). Am J Physiol Cell Physiol. 303(6):C673-81. 22785121
Zhao R., Visentin M., Suadicani SO. and Goldman ID. (2013). Inhibition of the proton-coupled folate transporter (PCFT-SLC46A1) by bicarbonate and other anions. Mol Pharmacol. 84(1):95-103. 23609145
Zhao, J., Y. Liu, L. Zhu, J. Li, Y. Liu, J. Luo, T. Xie, and D. Chen. (2023). Tumor cell membrane-coated continuous electrochemical sensor for GLUT1 inhibitor screening. J Pharm Anal 13: 673-682. 37440905
Zhao, R. and I.D. Goldman. (2007). The molecular identity and characterization of a proton-coupled folate transporter--PCFT; biological ramifications and impact on the activity of pemetrexed. Cancer Metastasis. Rev. 26: 129-139. 17340171
Zhao, R., E.S. Unal, D.S. Shin, and I.D. Goldman. (2010). Membrane topological analysis of the proton-coupled folate transporter (PCFT-SLC46A1) by the substituted cysteine accessibility method. Biochemistry 49: 2925-2931. 20225891
Zhao, R., M. Najmi, A. Fiser, and I.D. Goldman. (2016). Identification of an Extracellular Gate for the Proton-Coupled Folate Transporter (SLC46A1) by Cysteine Cross-Linking. J. Biol. Chem. [Epub: Ahead of Print] 26884338
Zhao, R., M. Najmi, S. Aluri, D.C. Spray, and I.D. Goldman. (2018). Concentrative Transport of Antifolates Mediated by the Proton-Coupled Folate Transporter (SLC46A1); Augmentation by a HEPES Buffer. Mol Pharmacol 93: 208-215. 29326243
Zhao, R., N. Diop-Bove, M. Visentin, and I.D. Goldman. (2011). Mechanisms of membrane transport of folates into cells and across epithelia. Annu. Rev. Nutr. 31: 177-201. 21568705
Zhao, S., Z. Guo, L. Zhu, J. Fan, B. Yang, W. Chai, H. Sun, F. Feng, Y. Liang, C. Zou, X. Jiang, W. Zhao, J. Lü, and C. Zhang. (2023). [Identification, expression and DNA variation analysis of high affinity nitrate transporter / gene family in ]. Sheng Wu Gong Cheng Xue Bao 39: 2743-2761. 37584129
Zhao, Y., B. Veysman, K. Antolijao, Y. Zhao, Y. Papagni, H. Wang, R. Ross, T. Tibbot, D. Povrzenic, and R. Fox. (2024). Increase in the Expression of Glucose Transporter 2 (GLUT2) on the Peripheral Blood Insulin-Producing Cells (PB-IPC) in Type 1 Diabetic Patients after Receiving Stem Cell Educator Therapy. Int J Mol Sci 25:. 39125908
Zheng, H., G. Wisedchaisri, and T. Gonen. (2013). Crystal structure of a nitrate/nitrite exchanger. Nature 497: 647-651. 23665960
Zheng, H., J. Taraska, A.J. Merz, and T. Gonen. (2010). The prototypical H+/galactose symporter GalP assembles into functional trimers. J. Mol. Biol. 396: 593-601. 20006622
Zhou, H., G. Lei, Y. Chen, M. You, and S. You. (2022). Affects the Temperature Adaptability of a Cosmopolitan Pest by Altering Trehalose Tissue Distribution. Int J Mol Sci 23:. 36012281
Zhou, J., E. Fernández, A. Galván, and A.J. Miller. (2000). A high affinity nitrate/nitrite transport system from Chlamydomonas requires two gene products. FEBS Lett. 466: 225-227. 10682832
Zhou, W., Y.M. Jiang, H.J. Wang, L.H. Kuang, Z.Q. Hu, H. Shi, M. Shu, and C.M. Wa. (2014). Erythromycin-resistant genes in group A β-haemolytic Streptococci in Chengdu, Southwestern China. Indian J. Med. Microbiol. 32: 290-293. 25008823
Zhou, X., A. Hilk, N.V. Solis, N. Pereira De Sa, B.M. Hogan, T.A. Bierbaum, M. Del Poeta, S.G. Filler, L.S. Burrack, and A. Selmecki. (2024). Erg251 has complex and pleiotropic effects on sterol composition, azole susceptibility, filamentation, and stress response phenotypes. PLoS Pathog 20: e1012389. [Epub: Ahead of Print] 39078851
Zhou, Y., Z. Li, C. Chi, C. Li, M. Yang, and B. Liu. (2023). Identification of Hub Genes and Potential Molecular Pathogenesis in Substantia Nigra in Parkinson''s Disease via Bioinformatics Analysis. Parkinsons Dis 2023: 6755569. 37089789
Zhou, Z., L. Ma, J. Zhou, Z. Song, J. Zhang, K. Wang, B. Chen, D. Pan, Z. Li, C. Li, and Y. Shi. (2018). Renal hypouricemia caused by novel compound heterozygous mutations in the SLC22A12 gene: a case report with literature review. BMC Med Genet 19: 142. 30097038
Zhu, X., K. Ren, Y.Z. Zeng, Z. Zheng, and G.H. Yi. (2018). Biological function of SPNS2: From zebrafish to human. Mol Immunol 103: 55-62. 30196234
Zorzano, A., M. Palacín, and A. Gumà. (2005). Mechanisms regulating GLUT4 glucose transporter expression and glucose transport in skeletal muscle. Acta Physiol Scand 183: 43-58. 15654919