Review
Mechanisms of renal anionic drug transport

https://doi.org/10.1016/j.ejphar.2008.02.085Get rights and content

Abstract

By utilizing filtration, active secretion and reabsorption processes, the kidney can conserve essential nutrients, and eliminate drugs and potentially toxic compounds. Active uptake of organic anions and cations across the basolateral membrane, and their extrusion into the urine across the brush border membrane mainly takes place in the renal proximal tubule cells, and is facilitated via a range of substrate-specific tubular transporters. Many drugs and their phase II conjugates are anionic compounds, and therefore renal organic anion transporters are important determinants of their distribution and elimination. Competition for renal excretory transporters may cause drugs to accumulate in the body leading to toxicity, which is a potential hazard of concomitant drug administration. Here, we present a brief update on the most prominent human proximal tubule organic anion transporters, which either belong to the ATP-binding cassette (ABC) or the solute carrier transporter (SLC) families. We focus on the participation of the individual transporters in renal anionic drug elimination, in an attempt to understand their overall biological and pharmacological significance, hoping to inspire further studies in the renal transporters field.

Introduction

The kidney plays a critical role in conserving essential nutrients and eliminating natural end products, toxins, drugs and drug metabolites. It consists of approximately 1 million functional units, which are called nephrons. The sum of the processes of glomerular filtration, active tubular secretion and reabsorption that a compound can undergo in each of the nephrons, determines its net renal excretion rate. Active drug secretion and reabsorption mainly take place in the proximal tubule cells, which are equipped with separate transport systems for organic anions and cations, each consisting of multiple transporters localised in the plasma membrane at both sides of the cell and with overlapping substrate specificities. Both systems are characterised by a high clearance capacity and broad diversity of substances accepted.

This review is devoted to the transporters that drive renal organic anion excretion. Recent research has provided much insight into their molecular characteristics. Because the renal organic anion transport system also accepts many phase II metabolites in addition to unconjugated anionic compounds, they play a critical role in the elimination of a large number of xenobiotics. The renal organic cation transporters have been recently reviewed by others (Koepsell et al., 2007) (Terada and Inui, 2007). Several groups of organic anion transporters in the proximal tubule cell mediate the uptake of compounds from blood across the basolateral membrane (BLM), followed by their secretion into the urine through the brush border membrane (BBM, also known as apical or luminal membrane) (Fig. 1). It has been suggested that the rate-limiting step for excretion of organic anions is the uptake step at the BLM, although their concentration inside the proximal tubule cells is higher than outside, either at the BLM or BBM side (Wright and Dantzler, 2004). This suggests that net transport across both membranes facilitates the intracellular accumulation of organic anions, and that the BBM controls the final exit into the urine via ATP-powered transporters and bidirectional exchangers. Furthermore, reabsorption processes may also contribute to intracellular organic anion accumulation, which in turn can affect BLM gradient-sensitive processes.

Several methods have been employed to study carrier-mediated organic anion transport. The recent use of knockout animals provides a powerful tool, yet one that is not devoid of limitations. In addition to possible up- or down-regulation of compensatory transporters and the possible inter-species variations (Johnson et al., 2006) (Chu et al., 2006), other feedback mechanisms, as for example the saturation of metabolic pathways, can restrain the use of such models. Investigations using the isolated perfused kidney from transporter deficient animals (Masereeuw et al., 2003) aimed at circumventing some of these factors. Yet, the actual transport across the cell membrane may be the most solid proof to characterize a compound as a substrate of a certain transporter protein. With the recent advances in molecular cloning methodologies, several cellular models have evolved that are transfected to over-express one or more transporters, including Xenopus laevis oocytes, Spodoptera frugiperda insect (Sf9) cells, and mammalian cell lines of animal and human origin (Masereeuw and Russel, 2004). These different cell types have been used for transport studies in the form of cellular monolayers, transwell transport studies or transport in isolated inside-out membrane vesicles. Ultimately, combining the results of diverse molecular, cellular and in vivo studies are required to identify the role of individual transporters in the overall renal handling of drugs. In this review, we present an updated overview of the major human organic anion transporter proteins involved in renal proximal tubular drug handling, focusing on members of the ATP-binding cassette (ABC) C and solute carrier (SLC) 22A subfamilies.

Section snippets

ATP binding cassette proteins

ATP-binding cassette (ABC) proteins are members of the largest and most important superfamily of membrane transport proteins, which are named after their highly conserved ATP-binding motif. About 100 putative ABC transporters have been identified throughout nature, 49 of which in humans. The ABC superfamily is further divided into 7 subfamilies: ABCA to ABCG (see for more details the ABC protein links at http://emb1.bcc.univie.ac.at/cgi-bin/ABC-DB/Welcome.cgi). The ABCC subfamily members encode

Solute carrier proteins

The solute carrier (SLC) family consists of 46 gene subfamilies, with a total of 360 family members. These genes encode for a large number of uniporter, symporter and antiporter protein transporters (see http://www.bioparadigms.org/slc/menu.asp). The SLC22A gene subfamily encodes the organic anion transporters (OATs) that are critical in the transport of anionic drugs throughout the body, but particularly in the kidney. Another SLC subfamily that may have an additional role in renal drug

Conclusions and future perspectives

The renal excretion of organic anions is handled by a well-organised sophisticated system that can excrete xenobiotics and their metabolites, while preserving essential molecules. During excretion, transporters in the proximal tubule BLM control the blood-to-cell transport, and in the BBM the cell-to-urine transport, mediating the final exit of organic anions from the body. On the other hand, the route of organic anions is reversed in case of reabsorption, where BBM transporters selectively

Acknowledgements

This work was supported in part by grants from the Dutch Kidney Foundation, NWO/ZonMW and a scholarship granted by the Egyptian Ministry of Higher Education (to A.A.K.E-S).

References (187)

  • EkaratanawongS. et al.

    Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules

    J. Pharmacol. Sci.

    (2004)
  • EralyS.A. et al.

    Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock-out mice

    J. Biol. Chem.

    (2006)
  • FrommM.F. et al.

    The effect of rifampin treatment on intestinal expression of human MRP transporters

    Am. J. Pathol.

    (2000)
  • GedeonC. et al.

    Transport of glyburide by placental ABC transporters: implications in fetal drug exposure

    Placenta

    (2006)
  • GopalE. et al.

    Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney

    Biochim. Biophys. Acta

    (2007)
  • HinoshitaE. et al.

    Decreased expression of an ATP-binding cassette transporter, MRP2, in human livers with hepatitis C virus infection

    J. Hepatol.

    (2001)
  • HongM. et al.

    Critical amino acid residues in transmembrane domain 1 of the human organic anion transporter hOAT1

    J. Biol. Chem.

    (2004)
  • HongM. et al.

    Human organic anion transporter hOAT1 forms homooligomers

    J. Biol. Chem.

    (2005)
  • HorikawaM. et al.

    The potential for an interaction between MRP2 (ABCC2) and various therapeutic agents: probenecid as a candidate inhibitor of the biliary excretion of irinotecan metabolites

    Eur. J. Drug Metab. Pharmacokinet.

    (2002)
  • IchidaK. et al.

    Urate transport via human PAH transporter hOAT1 and its gene structure

    Kidney Int.

    (2003)
  • JedlitschkyG. et al.

    The nucleotide transporter MRP4 (ABCC4) is highly expressed in human platelets and present in dense granules, indicating a role in mediator storage

    Blood

    (2004)
  • JungK.Y. et al.

    Characterization of ochratoxin A transport by human organic anion transporters

    Life Sci.

    (2001)
  • JutabhaP. et al.

    Identification of a novel voltage-driven organic anion transporter present at apical membrane of renal proximal tubule

    J. Biol. Chem.

    (2003)
  • KarlaP.K. et al.

    Molecular evidence and functional expression of a novel drug efflux pump (ABCC2) in human corneal epithelium and rabbit cornea and its role in ocular drug efflux

    Int. J. Pharm.

    (2007)
  • KatoY.

    Xenobiotic transporter–adaptor network

    Drug Metab. Pharmacokinet.

    (2007)
  • KawabeT. et al.

    Enhanced transport of anticancer agents and leukotriene C4 by the human canalicular multispecific organic anion transporter (cMOAT/MRP2)

    FEBS Lett.

    (1999)
  • KhamdangS. et al.

    Interaction of human and rat organic anion transporter 2 with various cephalosporin antibiotics

    Eur. J. Pharmacol.

    (2003)
  • KhamdangS. et al.

    Interactions of human- and rat-organic anion transporters with pravastatin and cimetidine

    J. Pharmacol. Sci.

    (2004)
  • AltenbergG.A.

    The engine of ABC proteins

    News Physiol. Sci.

    (2003)
  • AndersonP.L. et al.

    Pharmacogenetic characteristics of indinavir, zidovudine, and lamivudine therapy in HIV-infected adults: a pilot study

    J. Acquir. Immune Defic. Syndr.

    (2006)
  • AnzaiN. et al.

    Functional characterization of rat organic anion transporter 5 (Slc22a19) at the apical membrane of renal proximal tubules

    J. Pharmacol. Exp. Ther.

    (2005)
  • AnzaiN. et al.

    New insights into renal transport of urate

    Curr. Opin. Rheumatol.

    (2007)
  • ApiwattanakulN. et al.

    Transport properties of nonsteroidal anti-inflammatory drugs by organic anion transporter 1 expressed in Xenopus laevis oocytes

    Mol. Pharmacol.

    (1999)
  • BakhiyaA. et al.

    Human organic anion transporter 3 (hOAT3) can operate as an exchanger and mediate secretory urate flux

    Cell Physiol. Biochem.

    (2003)
  • BakosE. et al.

    Interactions of the human multidrug resistance proteins MRP1 and MRP2 with organic anions

    Mol. Pharmacol.

    (2000)
  • BelinskyM.G. et al.

    Characterization of the drug resistance and transport properties of multidrug resistance protein 6 (MRP6, ABCC6)

    Cancer Res.

    (2002)
  • BergenA.A. et al.

    Mutations in ABCC6 cause pseudoxanthoma elasticum

    Nat. Genet.

    (2000)
  • BleasbyK. et al.

    Functional consequences of single nucleotide polymorphisms in the human organic anion transporter hOAT1 (SLC22A6)

    J. Pharmacol. Exp. Ther.

    (2005)
  • BorstP. et al.

    Multidrug resistance-associated proteins 3, 4, and 5

    Pflugers Arch.

    (2007)
  • BryanJ. et al.

    ABCC8 and ABCC9: ABC transporters that regulate K+ channels

    Pflugers Arch.

    (2007)
  • CaiC. et al.

    Androgen induces expression of the multidrug resistance protein gene MRP4 in prostate cancer cells

    Prostate Cancer Prostatic Dis.

    (2007)
  • ChaS.H. et al.

    Identification and characterization of human organic anion transporter 3 expressing predominantly in the kidney

    Mol. Pharmacol.

    (2001)
  • ChenZ.S. et al.

    Effect of multidrug resistance-reversing agents on transporting activity of human canalicular multispecific organic anion transporter

    Mol. Pharmacol.

    (1999)
  • ChenZ.S. et al.

    Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system

    Cancer Res.

    (2002)
  • ChoiJ.H. et al.

    MRP2 haplotypes confer differential susceptibility to toxic liver injury

    Pharmacogenet.Genomics

    (2007)
  • ChuX.Y. et al.

    Transport of ethinylestradiol glucuronide and ethinylestradiol sulfate by the multidrug resistance proteins MRP1, MRP2, and MRP3

    J. Pharmacol. Exp. Ther.

    (2004)
  • ChuX.Y. et al.

    Characterization of mice lacking the multidrug resistance protein MRP2 (ABCC2)

    J. Pharmacol. Exp. Ther.

    (2006)
  • ChuX.Y. et al.

    Transport of the dipeptidyl peptidase-4 inhibitor sitagliptin by human organic anion transporter 3, organic anion transporting polypeptide 4C1, and multidrug resistance P-glycoprotein

    J. Pharmacol. Exp. Ther.

    (2007)
  • CiL. et al.

    Involvement of MRP4 (ABCC4) in the luminal efflux of ceftizoxime and cefazolin in the kidney

    Mol. Pharmacol.

    (2007)
  • CorpechotC. et al.

    Identification of a novel 974C ® G nonsense mutation of the MRP2/ABCC2 gene in a patient with Dubin–Johnson syndrome and analysis of the effects of rifampicin and ursodeoxycholic acid on serum bilirubin and bile acids

    Am. J. Gastroenterol.

    (2006)
  • Cited by (102)

    • Advances in kidney-targeted drug delivery systems

      2020, International Journal of Pharmaceutics
      Citation Excerpt :

      A variety of transporters and receptors using to transport drugs are found on the apical membrane of proximal tubular cells as shown in Fig. 4. These transporters are primarily involved in the uptake of small molecules such as glucose, peptides, neurotransmitters, and some large proteins molecules (Terada and Inui, 2008; Lee and Kim, 2004; Hagenbuch, 2009; El-Sheikh et al., 2008; Dresser et al., 2001). Delivering drugs to proximal tubular cells via receptor-mediated endocytosis seems to be an effective method.

    • Expression of xenobiotic transporters in the human renal proximal tubule cell line RPTEC/TERT1

      2015, Toxicology in Vitro
      Citation Excerpt :

      The proximal tubule is the major site of the nephron for the reabsorption and secretion of endogenous and exogenous substances, which underpins its susceptibility to xenobiotics. The concerted activities of several classes of transporters, including the SLC and the ABC transporter families, play a key role (El-Sheikh et al., 2008; Koepsell et al., 2007; Lee and Kim, 2004; Perri et al., 2003; Wright and Dantzler, 2004). In the proximal tubule of the human kidney, uptake of organic anions is mainly mediated by basolaterally expressed organic anion transporters (OAT) OAT1 (SLC22A6) and OAT3 (SLC22A8).

    View all citing articles on Scopus
    View full text