MATE transporters: involvement in drug pharmacokinetics and drug-drug interactions

DOI: https://doi.org/10.29296/25419218-2019-07-08
Issue: 
7
Year: 
2019

V.A. Evteev(1), A.B. Prokofyev(1, 2), N.D. Bunyatyan(1, 2), V.G. Kukes(1) 1-Research Center for Examination of Medical Products, Ministry of Health of the Russian Federation, 8, Petrovsky Boulevard, Build. 2, Moscow 127051, Russian Federation 2-I.M. Sechenov First Moscow State Medical University (Sechenov University), 8, Trubetskaya St., Build. 2, Moscow 119048, Russian Federation

Along with organic cation transporters (OCT) and organic anion transporters (OAT), MATE transporters belong to the solute carrier (SLC) family. They are transmembrane proteins and are located on the apical side of the cell membrane of the liver and kidney. Three human MATE isoforms, namely MATE1, MATE2, and MATE2-K, have been now studied. MATE1 is predominantly expressed in the liver, kidneys, and skeletal muscles. MATE2-K and MATE2 are expressed in the renal proximal tubular epithelial cells and hepatocytes, respectively. All the transporters of this subfamily are distinguishing. The transport mechanism has not yet been fully studied; the transport according to the principle of antiport in exchange for sodium cations (Na+) is currently considered most preferable. The direction of transport is shown to be pH-dependent. The MATE transporters have a broad range of transportable substrates, both endogenous and xenobiotic, mainly organic cations. The key substrates of MATE transporters among drugs are metformin, platinum preparations, acyclovir, cephalosporins, etc. It is worth noting that there is a great similarity to the spectrum of OCT substrates. The MATE transporters located on the apical membrane are most likely to form a functional unit with basolateral OCT transporters. This unit is capable of transporting organic cations through the hepatocyte and renal proximal tubular epithelium. The clinically significant genetic polymorphisms of MATE transporters have not yet been identified now. When drugs and MATE transporter substrates are used simultaneously, there may be a drug-drug interaction, which is manifested by the higher incidence of adverse drug reactions.

Keywords: 
transporters
MATE
metformin
cisplatin

References: 
  1. Terada T. et al. Molecular cloning, functional characterization and tissue distribution of rat H+/organic cation antiporter MATE1. Pharm. Res., 2006; 23(8): 1696–701.
  2. Masuda S.et al. Identification and functional characterization of a new human kidney-specific H+/organic cation antiporter, kidney-specific multidrug and toxin extrusion 2. J. Am. Soc. Nephrol., 2006; 17 (8): 2127–35.
  3. Lu M1., Symersky J., Radchenko M. et al. Structures of a Na+-coupled, substrate-bound MATE multidrug transporter. Proc. Natl. Acad. Sci.USA., 2013; 110 (6): 2099–104.
  4. Tsuda M.et al. Oppositely directed H+ gradient functions as a driving force of rat H+/organic cation antiporter MATE1. Am. J. Physiol. Renal. Physiol., 2007; 292 (2): 593–8.
  5. Tanihara Y. et al. Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions/H(+)-organic cation antiporters. Biochem. Pharmacol., 2007; 74 (2): 359–71.
  6. Sato T., Masuda S., Yonezawa A.et al. Transcellular transport of organic cations in double-transfected MDCK cells expressing human organic cation transporters hOCT1/hMATE1 and hOCT2/hMATE1. Biochem. Pharmacol., 2008;76: 894–903.
  7. Tsuda M. et al. Targeted disruption of the multidrug and toxin extrusion 1 (mate1) gene in mice reduces renal secretion of metformin. Mol. Pharmacol., 2009; 75 (6): 1280–6.
  8. Nakamura T. et al. Disruption of multidrug and toxin extrusion MATE1 potentiates cisplatin-induced nephrotoxicity. Biochem. Pharmacol., 2010; 80 (11): 1762–67.
  9. Tsuda M. et al. Involvement of human multidrug and toxin extrusion 1 in the drug interaction between cimetidine and metformin in renal epithelial cells. J. Pharmacol. Exp. Ther., 2009; 329 (1): 185–91.
  10. Yonezawa A., InuiK. Importance of the multidrug and toxin extrusion MATE/SLC47A family to pharmacokinetics, pharmacodynamics/toxicodynamics and pharmacogenomics. Br. J. Pharmacol., 2011; 164 (7): 1817–25.
  11. Kusuhara H. et al. Effects of a MATE protein inhibitor, pyrimethamine, on the renal elimination of metformin at oral microdose and at therapeutic dose in healthy subjects. Clin. Pharmacol. Ther., 2011; 89 (6): 837–44.
  12. Cho S.K., Chung J.Y. The MATE1 rs2289669 polymorphism affects the renal clearance of metformin following ranitidine treatment. Int. J. Clin. Pharmacol. Ther., 2016; 54 (4): 253–62.
  13. He R. et al., SLC47A1 gene rs2289669 G>A variants enhance the glucose-lowering effect of metformin via delaying its excretion in Chinese type 2 diabetes patients. Diabetes Res. Clin. Pract., 2015; 109 (1): 57–63.
  14. Tkac I. et al., Pharmacogenomic association between a variant in SLC47A1 gene and therapeutic response to metformin in type 2 diabetes. Diabetes Obes. Metab., 2013; 15(2): 189–91.
  15. Tsuda M. et al. Involvement of human multidrug and toxin extrusion 1 in the drug interaction between cimetidine and metformin in renal epithelial cells. J. Pharmacol. Exp. Ther., 2009; 329(1): 185–191.
  16. Kusuhara H. et al. Effects of a MATE protein inhibitor, pyrimethamine, on the renal elimination of metformin at oral microdose and at therapeutic dose in healthy subjects. Clin. Pharmacol. Ther., 2011; 89(6): 837-844.
  17. Oh J. et al. Inhibition of the multidrug and toxin extrusion (MATE) transporter by pyrimethamine increases the plasma concentration of metformin but does not increase antihyperglycaemic activity in humans. Diabetes Obes. Metab., 2016; 18(1): 104-108.
  18. Li Q. et al. Ondansetron can enhance cisplatin-induced nephrotoxicity via inhibition of multiple toxin and extrusion proteins (MATEs). Toxicol. Appl. Pharmacol., 2013; 273(1): 100-109.
  19. Hillgren K.M. et al. Emerging transporters of clinical importance: an update from the international transporter consortium. Clin. Pharmacol. Ther., 2013; 94(1): 52-63.
  20. Bukatina T.M., Pasternak E.Ju., Romanov B.K. i dr. Rekomendatsii zarubezhnyh reguljatornyh organov. Bezopasnost' i risk farmakoterapii. 2016; 4: 31–40. Bukatina T.M., Pasternak E.Y., Romanov B.K., Alyautdin R.N., Lepakhin V.K., Kazakov A.S., Zatolochina K.E., Snegireva I.I., Darmostukova M.A., Kolesnikova E.Y., Zhuravleva E.O., Romanova T.V., Velts N.Y., Maksimov M.L., Kutekhova G.V. Information on the decisions of foreign regulatory authorities. Bezopasnost' i risk farmakoterapii. 2016; (4): 31–40 (in Russian)]