Elsevier

Pharmacological Research

Volume 111, September 2016, Pages 237-246
Pharmacological Research

Review
Polyspecific organic cation transporters and their impact on drug intracellular levels and pharmacodynamics

https://doi.org/10.1016/j.phrs.2016.06.002Get rights and content

Abstract

Most drugs are intended to act on molecular targets residing within a specific tissue or cell type. Therefore, the drug concentration within the target tissue or cells is most relevant to its pharmacological effect. Increasing evidences suggest that drug transporters not only play a significant role in governing systemic drug levels, but are also an important gate keeper for intra-tissue and intracellular drug concentrations. This review focuses on polyspecific organic cation transporters, which include the organic cation transporters 1⿿3 (OCT1-3), the multidrug and toxin extrusion proteins 1⿿2 (MATE1-2) and the plasma membrane monoamine transporter (PMAT). Following an overview of the tissue distribution, transport mechanisms, and functional characteristics of these transporters, we highlight the studies demonstrating the ability of locally expressed OCTs to impact intracellular drug concentrations and directly influence their pharmacological and toxicological activities. Specifically, OCT1-mediated metformin access to its site of action in the liver is impacted by genetic polymorphisms and chemical inhibition of OCT1. The impact of renal OCT2 and MATE1/2-K in cisplatin intrarenal accumulation and nephrotoxicity is reviewed. New data demonstrating the role of OCT3 in salivary drug accumulation and secretion is discussed. Whenever possible, the pharmacodynamic response and toxicological effects is presented and discussed in light of intra-tissue and intracellular drug exposure. Current challenges, knowledge gaps, and future research directions are discussed. Understanding the impact of transporters on intra-tissue and intracellular drug concentrations has important implications for rational-based optimization of drug efficacy and safety.

Introduction

The ability of a drug molecule to move through cell membranes is a vital property affecting its pharmacokinetic and pharmacodynamic properties. Lipophilic drugs generally have high membrane permeability and their movement across cell membranes occurs primarily through passive diffusion, a non-mediated process discussed in great details elsewhere in this issue. Hydrophilic drugs, on the other hand, have low membrane permeability, and their efficient uptake into cells and tissues often involve facilitated mechanisms mediated by membrane transporters (also known as carriers). Different from passive diffusion where a drug molecule moves across membranes down its concentration gradient without energy input, carrier-mediated transport can be coupled to a cellular energy source to power uphill transport against the drug concentration gradient. Further, carrier-mediated drug transport is saturable, inhibitable, and highly dependent on the functional characteristics of the membrane transporters expressed in the specific tissues or cell types. In mammalian cells, there are two major types of membrane proteins involved in drug and solute transport: the solute carrier (SLC) and the ATP-binding cassette (ABC) transporters. The past two decades have witnessed an explosion of knowledge in our understandings of the basic biology and pharmacology of various SLC and ABC drug transporters. The in vivo roles of these transporters in drug disposition, efficacy, and toxicity are increasingly being appreciated. The clinical significance of transporters as a site of drug⿿drug interaction and a source for interindividual variability in drug response is also begining to be acknowledged [1], [2], [3].

Most drugs are intended to act on targets residing within a specific tissue or cells. While some drugs bind to external cell surface targets (e.g. G protein-coupled receptors), others act on intracellular enzymes and receptors residing inside the cell. Thus, it is the unbound drug concentration within the target tissue or cells that is directly responsible for eliciting its pharmacological effect. However, in the clinical setting, direct measurement of drug concentrations in target tissues and cells is difficult to achieve. Measurement of blood or plasma drug concentrations is thus commonly used to establish pharmacokinetic⿿pharmacodynamic relationships. For drugs that rapidly cross membranes by passive diffusion, plasma concentration is often a good surrogate for tissue concentration because the unbound drug concentration in tissue/cells is at equilibrium with its unbound concentration in plasma at steady state [4], [5]. However, if a drug is transported by active uptake and/or efflux drug transporters, such a relationship may no longer exist. For drugs that are substrates of uptake transporters, tissue and/or intracellular drug concentrations can be much higher than drug concentrations in plasma. Conversely, for drugs that are substrates of efflux transporters, concentrations in tissues and cells may be substantially lower than predicted from plasma levels. Increasing evidences suggest that transporters expressed in specific tissues and cells can exert a great impact on local and intracellular drug concentrations, directly influencing their pharmacological and toxicological activities [4], [5].

This review focuses on a special group of SLC drug transporters⿿the polyspecific organic cation transporters, which mediate cellular uptake and efflux of a broad spectrum of drugs, toxins, and endogenous compounds. We first briefly review the molecular and functional characteristics of major organic cation transporters with a special emphasis on their tissue distribution, cellular localization and transport mechanisms. We then highlight the impact of these transporters in controlling tissue and intracellular drug concentrations using literature examples where the roles of locally expressed organic cation transporters have been clearly demonstrated in several tissues (liver, kidney, salivary glands) in in vivo or clinical studies. The resulting consequence on pharmacodynamic response and toxicological effects of clinically used organic cation drugs is presented and discussed alongside. Lastly, the current challenges, knowledge gaps and future research directions in this field are briefly summarized and discussed.

Section snippets

Molecular and functional characteristics of polyspecific organic cation transporters

Organic cations are structurally diverse endogenous compounds (e.g. biogenic amines) and xenobiotics (e.g. drugs, environmental toxins) that carry a net positive charge at physiological pH. About 40% of the commonly prescribed drugs exist as organic cations at physiological pH [6]. Many organic cations are hydrophilic and rely on transporters to move across cell membranes. In humans and other mammals, there are a number of SLC transporters that appear to be evolved specifically to handle these

Impact of OCT1/Oct1 on hepatic drug levels and action

Located in the sinusoidal membrane of hepatocytes, OCT1 has been identified as a main organic cation transporter in the liver and is responsible for the uptake of basic compounds in hepatocytes [3], [29], [56], [57]. Although OCT3 and MATE1 are also expressed in hepatocytes, their roles in hepatic drug disposition and elimination have not been well established as compared to OCT1 [29], [56], [58], [59]. MATE1 appears to mediate some biliary excretion but its activity with in vivo probes is

Conclusions

In the past two decades, great progress has been made in molecular and functional characterization of drug transporters and understanding their roles in drug disposition and response. It is now becoming increasingly recognized that locally expressed transporters can exert a large impact on tissue and intracellular drug levels, directly influencing their pharmacological and toxicological activities. Drug concentrations in target tissues do not always correlate with plasma drug concentrations

Acknowledgements

This study was supported by the National Institutes of Health National Institute on Drug Abuse Grant P01DA032507 (JW) and National Institutes of Health General Medical Sciences Grants R01GM066233 (JW) and T32GM07750 (DW). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

References (141)

  • a. Dahlin et al.

    Expression and immunolocalization of the plasma membrane monoamine transporter in the brain

    Neuroscience

    (2007)
  • H. Duan et al.

    Impaired monoamine and organic cation uptake in choroid plexus in mice with targeted disruption of the plasma membrane monoamine transporter (Slc29a4) gene

    J. Biol. Chem.

    (2013)
  • T. Budiman et al.

    Mechanism of electrogenic cation transport by the cloned organic cation transporter 2 from rat

    J. Biol. Chem.

    (2000)
  • S.K. Moule et al.

    Regulation of the plasma membrane potential in hepatocytes⿿mechanism and physiological significance

    Biochim. Biophys. Acta

    (1990)
  • H. Koepsell

    The SLC22 family with transporters of organic cations, anions and zwitterions

    Mol. Aspects Med.

    (2013)
  • F. Staud et al.

    Multidrug and toxin extrusion proteins (MATE/SLC47); role in pharmacokinetics

    Int. J. Biochem. Cell Biol.

    (2013)
  • W. Abbud et al.

    Stimulation of AMP-activated protein kinase (AMPK) is associated with enhancement of Glut1-mediated glucose transport

    Arch. Biochem. Biophys.

    (2000)
  • A. Kunze et al.

    Interaction of the antiviral drug telaprevir with renal and hepatic drug transporters

    Biochem. Pharmacol.

    (2012)
  • G. Minuesa et al.

    Drug uptake transporters in antiretroviral therapy

    Pharmacol. Ther.

    (2011)
  • F. Müller et al.

    Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine

    Biochem. Pharmacol.

    (2013)
  • A. Yonezawa et al.

    Organic cation transporter OCT/SLC22A and H+/organic cation antiporter MATE/SLC47A are key molecules for nephrotoxicity of platinum agents

    Biochem. Pharmacol.

    (2011)
  • Z.H. Siddik et al.

    The comparative pharmacokinetics of carboplatin and cisplatin in mice and rats

    Biochem. Pharmacol.

    (1987)
  • M.K. DeGorter et al.

    Drug transporters in drug efficacy and toxicity

    Annu. Rev. Pharmacol. Toxicol.

    (2012)
  • K.M. Giacomini et al.

    Membrane transporters in drug development

    Nat. Rev. Drug Discov.

    (2010)
  • X. Chu et al.

    Intracellular drug concentrations and transporters: measurement, modeling, and implications for the liver

    Clin Pharmacol. Ther.

    (2013)
  • D. a Smith et al.

    The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery

    Nat. Rev. Drug Discov.

    (2010)
  • S. Neuhoff et al.

    pH-dependent bidirectional transport of weakly basic drugs across Caco-2 monolayers: implications for drug⿿drug interactions

    Pharm. Res.

    (2003)
  • H. Koepsell et al.

    The SLC22 drug transporter family

    Pflugers Arch.

    (2004)
  • S.H. Wright et al.

    Molecular and cellular physiology of renal organic cation and anion transport

    Physiol. Rev.

    (2004)
  • M. Otsuka et al.

    A human transporter protein that mediates the final excretion step for toxic organic cations

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • K. Engel et al.

    Interaction of organic cations with a newly identified plasma membrane monoamine transporter

    Mol. Pharmacol.

    (2005)
  • M. Sala-Rabanal et al.

    Polyamine transport by the polyspecific organic cation transporters OCT1, OCT2, and OCT3

    Mol. Pharm.

    (2013)
  • H. Koepsell et al.

    Structure and function of renal organic cation transporters

    News Physiol. Sci.

    (1998)
  • A.T. Nies et al.

    Multidrug and toxin extrusion proteins as transporters of antimicrobial drugs

    Expert Opin. Drug Metab. Toxicol.

    (2012)
  • S. Masuda 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)
  • H. Motohashi et al.

    Organic cation transporter OCTs (SLC22) and MATEs (SLC47) in the human kidney

    AAPS J.

    (2013)
  • H.T. Ho et al.

    Molecular analysis and structure-activity relationship modeling of the substrate/inhibitor interaction site of plasma membrane monoamine transporter

    J. Pharmacol. Exp. Ther.

    (2011)
  • M. Zhou et al.

    Metformin transport by a newly cloned proton-stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine

    Drug Metab. Dispos.

    (2007)
  • H. Koepsell

    Substrate recognition and translocation by polyspecific organic cation transporters

    Biol. Chem.

    (2011)
  • H. Koepsell et al.

    Organic cation transporters

    Rev. Physiol. Biochem. Pharmacol.

    (2003)
  • G. Ciarimboli

    Role of organic cation transporters in drug-induced toxicity

    Expert Opin. Drug Metab. Toxicol.

    (2011)
  • H.-C. Chien et al.

    Rapid method to determine intracellular drug concentrations in cellular uptake assays: application to metformin in OCT1-transfected HEK cells

    Drug Metab. Dispos.

    (2015)
  • S. Itagaki et al.

    Electrophysiological characterization of the polyspecific organic cation transporter plasma membrane monoamine transporter

    Drug Metab. Dispos.

    (2012)
  • M. Zhou et al.

    Adenosine transport by plasma membrane monoamine transporter: reinvestigation and comparison with organic cations

    Drug Metab. Dispos.

    (2010)
  • S.H. Wright et al.

    Transport of tetraethylammonium by rabbit renal brush-border and basolateral membrane vesicles

    Am. J. Physiol.

    (1987)
  • D. Grundemann et al.

    Drug excretion mediated by a new prototype of polyspecific transporter

    Nature

    (1994)
  • H. Koepsell et al.

    Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications

    Pharm. Res.

    (2007)
  • H. Motohashi et al.

    Gene expression levels and immunolocalization of organic ion transporters in the human kidney

    J. Am. Soc. Nephrol.

    (2002)
  • A.T. Nies et al.

    Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver

    Hepatology

    (2009)
  • V. Gorboulev et al.

    Cloning and characterization of two human polyspecific organic cation transporters

    DNA Cell Biol.

    (1997)
  • Cited by (73)

    View all citing articles on Scopus
    View full text