Associate Editor: L.H. Lash
Short-term regulation of organic anion transporters

https://doi.org/10.1016/j.pharmthera.2009.08.002Get rights and content

Abstract

Organic anion transporters (OATs), which belong to the superfamily SLC22A, are key determinants in the absorption, distribution, and excretion of a diverse array of environmental toxins, and clinically important drugs, and, therefore, are critical for the survival of mammalian species. Alteration in the function of these drug transporters plays important roles in intra- and inter-individual variability of the therapeutic efficacy and the toxicity of many drugs. As a result, the activity of OATs must be under tight regulation so as to carry out their normal functions. This review article highlights the recent advances from our laboratory as well as from others in delineating the short-term regulation of OATs. These advances provide important insights into strategies to maximize therapeutic efficacy in drug development.

Introduction

The organic anion transporter (OAT) family belongs to the amphiphilic solute carrier transporters family 22a (SLC22A), which transports a broad diversity of substrates including metabolites, toxins and clinical drugs such as β-lactam antibiotics, antivirals, ACE inhibitors, diuretics, and NSAIDs (You, 2004, Anzai et al., 2006, El-Sheikh et al., 2008, Srimaroeng et al., 2008). To date, ten members of the OAT family (OAT1-10) have been identified (Sweet et al., 1997, Sekine et al., 1997, Sekine et al., 1998, Kusuhara et al., 1999, Cha et al., 2000, Youngblood and Sweet, 2004, Schnabolk et al., 2006, Shin et al., 2007, Bahn et al., 2008, Yokoyama et al., 2008), which differ from each other by their localization, expression level, and substrate specificity. As the first cloned OAT, OAT1 is expressed predominantly at the basolateral membrane of renal proximal tubular cells (Sekine et al., 1997, Sweet et al., 1997, Lu et al., 1999, Race et al., 1999) and at low levels in the cerebral cortex and hippocampus as well as in the choroid plexus (Alebouyeh et al., 2003, Bahn et al., 2005). OAT2 is primarily expressed in the liver (Sekine et al., 1998) and kidney (Kojima et al., 2002). In the liver, OAT2 is expressed at the basolateral membrane of hepatocytes (Hilgendorf et al., 2007). In the kidney, however, the cellular location of OAT2 seems to be species-dependent. Human OAT2 is expressed similarly as hOAT1 at the basolateral membrane of the proximal tubule (Enomoto et al., 2002b), while in rat and mouse, OAT2 is located at the apical membrane of proximal tubular cells (Ljubojevic et al., 2007). OAT3 is highly expressed at the basolateral membrane of renal proximal tubular cells (Cha et al., 2001, Hasegawa et al., 2002, Kobayashi et al., 2004) and at the apical membrane of the choroid plexus (Sweet et al., 2002). OAT4 is expressed at the apical membrane of renal proximal tubular cells (Ekaratanawong et al., 2004), while in the placenta, OAT4 is expressed at the basal membrane of syncytiotrophoblast (Ugele et al., 2003). Expressed exclusively in kidneys, OAT5, similar to OAT4, is located at the apical membrane of renal proximal tubular cells (Youngblood & Sweet, 2004). The unique feature of OAT6 is its complete absence in the kidney and its expression in olfactory mucosa (Monte et al., 2004). Discovered as a liver-specific OAT, OAT7 localizes at the sinusoidal membrane of the hepatocyte (Shin et al., 2007). Recently, three other OATs were identified in the kidney. Rat OAT8 is expressed in rat renal collecting ducts and located at the apical membrane of renal intercalated cells (Yokoyama et al., 2008). OAT9 is expressed in the kidney and brain (Anzai et al., 2006). OAT10 is highly expressed at the apical side of renal proximal tubular cells (Bahn et al., 2008). The urate transporter URAT1 (SLC22A12), acting as an organic anion exchanger, is expressed at the apical membrane of the kidney proximal tubule and is believed to be responsible for renal urate reabsorption (Enomoto et al., 2002a).

In the kidney, OAT1 and OAT3 utilize a tertiary transport mechanism to move organic anions across the basolateral membrane into the proximal tubular cells for subsequent exit across the apical membrane into the urine for elimination (Fig. 1). Through this tertiary transport mechanism, Na+/K+-ATPase maintains an inwardly directed (blood-to-cell) Na+ gradient. The Na+ gradient then drives a sodium dicarboxylate cotransporter, sustaining an outwardly directed dicarboxylate gradient that is utilized by a dicarboxylate/organic anion exchanger, namely OAT, to move the organic anion substrate into the cell. This cascade of events indirectly links organic anion transport to metabolic energy and the Na+ gradient, allowing the entry of a negatively charged substrate against both its chemical concentration gradient and the electrical potential of the cell.

Structurally, all members of the OATs share some common features (Fig. 2) including 12 putative membrane-spanning segments, a cluster of potential glycosylation sites located in the first extracellular loop between transmembrane domains 1 and 2, and multiple presumptive phosphorylation sites in the intracellular loop between the sixth and the seventh transmembrane domains and in the carboxyl terminus.

One remarkable characteristic of OATs is their wide range of substrate recognition, from endogenous metabolites to xenobiotics and drugs including anti-HIV therapeutics, anti-tumor drugs, antibiotics, anti-hypertensives, and anti-inflammatories (You, 2004, Enomoto and Endou, 2005, Anzai et al., 2006, Srimaroeng et al., 2008). Given the critical role of OATs in the control of distribution, elimination and retention of such a diverse array of chemicals within the body, the activity of these transporters must be under tight regulation in order to accomplish their normal task. This review article highlights the recent advances from our laboratory as well as from others in unveiling the regulation of OATs, especially the short-term regulation of these transporters, as well as their clinical implications.

Section snippets

Regulation of organic anion transporters

OAT activity is subject to long- and short-term regulation. Long-term regulation, also called chronic regulation, usually occurs at transcriptional as well as translational levels within a time frame of hours to days. Long-term regulation usually happens when the body undergoes massive change, such as during development or the occurrence of disease (Buist et al., 2002, Sakurai et al., 2004, Naud et al., 2008). Short-term regulation, also called acute regulation, usually occurs at

Clinical implications

Members of the OAT family are known for their broad substrate specificity, including many drugs in clinical use. Therefore, the activity of OATs plays critical roles in determining the therapeutic efficacy and the toxicity of these drugs. Abnormal OAT expression and function have been associated with poor drug/toxin elimination in several pathophysiological conditions. Some of the abnormalities in organic anion transport are due to an altered expression of OAT genes and proteins, which was

Conclusions

OATs are key players in the body's disposition of a diverse array of environmental toxins and clinically important drugs. Alteration in the function of OATs plays important roles in intra- and inter-individual variability of the therapeutic efficacy and toxicity of many drugs. Although significant progress has been made in understanding the regulation of OATs (Table 1), this is just the beginning. We now know that OATs are subject to regulation by phosphorylation, yet the protein kinase(s)

Acknowledgment

This work was supported by grants (to Dr. Guofeng You) from the National Institute of Health (R01-DK 60034 and R01-GM 079123).

References (88)

  • M. Hao et al.

    Characterization of rapid membrane internalization and recycling

    J Biol Chem

    (2000)
  • Y. Kato et al.

    Regulation of drug transporters by PDZ adaptor proteins and nuclear receptors

    Eur J Pharm Sci

    (2006)
  • R. Khanna et al.

    Glycosylation increases potassium channel stability and surface expression in mammalian cells

    J Biol Chem

    (2001)
  • H. Kusuhara et al.

    Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain

    J Biol Chem

    (1999)
  • M.K. Loder et al.

    The dopamine transporter constitutively internalizes and recycles in a protein kinase C-regulated manner in stably transfected PC12 cell lines

    J Biol Chem

    (2003)
  • J.C. Monte et al.

    Identification of a novel murine organic anion transporter family member, OAT6, expressed in olfactory mucosa

    Biochem Biophys Res Commun

    (2004)
  • T. Nii-Kono et al.

    Indoxyl sulfate induces skeletal resistance to parathyroid hormone in cultured osteoblastic cells

    Kidney Int

    (2007)
  • F.Q. Nuttall et al.

    Regulation of glycogen synthesis in the liver

    Am J Med

    (1988)
  • R.J. Ott et al.

    Inhibition of N-linked glycosylation affects organic cation transport across the brush border membrane of opossum kidney (OK) cells

    J Biol Chem

    (1992)
  • J.E. Race et al.

    Molecular cloning and characterization of two novel human renal organic anion transporters (hOAT1 and hOAT3)

    Biochem Biophys Res Commun

    (1999)
  • T. Sekine et al.

    Expression cloning and characterization of a novel multispecific organic anion transporter

    J Biol Chem

    (1997)
  • T. Sekine et al.

    Identification of multispecific organic anion transporter 2 expressed predominantly in the liver

    FEBS Lett

    (1998)
  • P.M. Snyder et al.

    Mechanism by which Liddle's syndrome mutations increase activity of a human epithelial Na+ channel

    Cell

    (1995)
  • D.H. Sweet et al.

    Expression cloning and characterization of ROAT1. The basolateral organic anion transporter in rat kidney

    J Biol Chem

    (1997)
  • D.H. Sweet et al.

    Impaired organic anion transport in kidney and choroid plexus of organic anion transporter 3 (Oat3 (Slc22a8)) knockout mice

    J Biol Chem

    (2002)
  • K. Tanaka et al.

    Role of glycosylation in the organic anion transporter OAT1

    J Biol Chem

    (2004)
  • S.R. Villar et al.

    Altered expression of rat renal cortical OAT1 and OAT3 in response to bilateral ureteral obstruction

    Kidney Int

    (2005)
  • G. You et al.

    Regulation of mOAT-mediated organic anion transport by okadaic acid and protein kinase C in LLC-PK(1) cells

    J Biol Chem

    (2000)
  • N.R. Zahniser et al.

    Trafficking of dopamine transporters in psychostimulant actions

    Semin Cell Dev Biol

    (2009)
  • Q. Zhang et al.

    Organic anion transporter OAT1 undergoes constitutive and protein kinase C-regulated trafficking through a dynamin- and clathrin-dependent pathway

    J Biol Chem

    (2008)
  • A. Bahn et al.

    Murine renal organic anion transporters mOAT1 and mOAT3 facilitate the transport of neuroactive tryptophan metabolites

    Am J Physiol Cell Physiol

    (2005)
  • A.A. Bernardo et al.

    The renal cortical Na+/HCO3− cotransporter VI, the effect of chemical modification in cotransporter activity

    J Membr Biol

    (1997)
  • J. Biber et al.

    Protein/protein interactions (PDZ) in proximal tubules

    J Membr Biol

    (2005)
  • A. Bose et al.

    Glucose transporter recycling in response to insulin is facilitated by myosin Myo1c

    Nature

    (2002)
  • T.M. Buck et al.

    Evidence for stabilization of aquaporin-2 folding mutants by N-linked glycosylation in endoplasmic reticulum

    Am J Physiol Cell Physiol

    (2004)
  • S.C. Buist et al.

    Gender-specific and developmental influences on the expression of rat organic anion transporters

    J Pharmacol Exp Ther

    (2002)
  • S.H. Cha et al.

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

    Mol Pharmacol

    (2001)
  • J. Chen et al.

    Adaptive responses of renal organic anion transporter 3 (OAT3) during cholestasis

    Am J Physiol Renal Physiol

    (2008)
  • P. Coppo et al.

    BCR-ABL activates STAT3 via JAK and MEK pathways in human cells

    Br J Haematol

    (2006)
  • A. Dal Canton et al.

    Glomerular hemodynamics before and after release of 24-hour bilateral ureteral obstruction

    Kidney Int

    (1980)
  • C.B. Dugani et al.

    Glucose transporter 4, cycling, compartments and controversies

    EMBO Rep

    (2005)
  • A. Enomoto et al.

    Roles of organic anion transporters (OATs) and a urate transporter (URAT1) in the pathophysiology of human disease

    Clin Exp Nephrol

    (2005)
  • A. Enomoto et al.

    Molecular identification of a renal urate anion exchanger that regulates blood urate levels

    Nature

    (2002)
  • A. Enomoto et al.

    Interaction of human organic anion transporters 2 and 4 with organic anion transport inhibitors

    J Pharmacol Exp Ther

    (2002)
  • Cited by (15)

    • Regulation of organic anion transporters: Role in physiology, pathophysiology, and drug elimination

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      As a result, the functional diversities of the target proteins much exceed their molecular diversities. Various PTMs of OATs have been described in details in our previous review articles (P. Duan & You, 2010; D. Xu, Wang, & You, 2016b; D. Xu & You, 2017). In this review article, we will update the recent progress in uncovering the new PTMs of OATs, the relationship among these PTMs, and the regulatory network on OATs through remote sensing and signaling.

    • Renal Organic Anion Transporters 1 and 3 In Vitro: Gone but Not Forgotten

      2023, International Journal of Molecular Sciences
    • ORGANIC ANION TRANSPORTERS (OATs)

      2022, Drug Transporters: Molecular Characterization and Role in Drug Disposition: Third Edition
    • DRUG TRANSPORT IN THE BRAIN

      2022, Drug Transporters: Molecular Characterization and Role in Drug Disposition: Third Edition
    View all citing articles on Scopus
    View full text