Different transport properties between famotidine and cimetidine by human renal organic ion transporters (SLC22A)

https://doi.org/10.1016/j.ejphar.2004.09.032Get rights and content

Abstract

Histamine H2 receptor antagonist famotidine and cimetidine are commonly used for treatment of gastrointestinal ulcer diseases. Inasmuch as these drugs are mainly secreted by renal tubules, dosages have been adjusted according to renal function. Although many studies have been performed on the molecular mechanisms of renal handling of cimetidine, little is known about that of famotidine. In this study, to examine the recognition and transport of famotidine by human organic anion transporters (OATs; hOAT1, hOAT3) and human organic cation transporter (OCT; hOCT2), the uptake studies using Xenopus laevis oocytes were performed in comparison with cimetidine. The half-maximal inhibitory concentrations of famotidine for [3H]estrone sulfate transport by hOAT3 and [14C]tetraethylammonium transport by hOCT2 (300 μM and 1.8 mM, respectively) were higher than those of cimetidine (53 and 67 μM, respectively). While cimetidine inhibited p-[14C]aminohippurate transport by hOAT1 in a concentration dependent manner, famotidine did not affect it at 5 mM. In addition, hOAT3 mediated famotidine uptake, but hOAT1 and hOCT2 did not show famotidine transport. These results indicate that there are marked differences between famotidine and cimetidine in the recognition and transport by organic ion transporters and that hOAT3 contributes to the renal tubular secretion of famotidine. Present findings should be useful information to understand the renal handling of famotidine and cimetidine.

Introduction

A histamine H2 receptor antagonist famotidine is commonly used in the treatment for gastrointestinal ulcer diseases. Famotidine shows beneficial characteristics, including stronger inhibition of gastric acid secretion, longer acting and less interaction with P-450, compared with another histamine H2 receptor antagonist cimetidine (Howard et al., 1985, Humphries, 1987). Pharmacokinetic studies revealed that famotidine was mainly excreted into the urine as an unmetabolized form (Takabatake et al., 1985, Lin et al., 1988, Dowling et al., 2001). Renal clearance of famotidine exceeded the creatinine clearance about three times in subjects with normal renal function, indicating the efficient tubular secretion of the drug. In patients with renal insufficiency, plasma elimination and renal clearance of famotidine were significantly decreased (Takabatake et al., 1985, Lin et al., 1988). To avoid the accumulation and undesirable effects, famotidine dosage should be reduced according to the renal functions.

In this decade, organic anion transporters (OATs) and organic cation transporters (OCTs) in the kidney were identified, and their characteristics have been clarified. These transporters mediate endogenous and exogenous substances, such as uremic toxins, diuretics, methotrexate, antibiotics, antivirals and nonsteroidal antiinflammatory drugs, suggesting that OATs and OCTs are responsible for the tubular secretion of these compounds (Inui et al., 2000, Sekine et al., 2000, Dresser et al., 2001). Recently, we quantified mRNA levels of organic ion transporters in the human kidney cortex and revealed that hOAT1 (SLC22A6), hOAT3 (SLC22A8) and hOCT2 (SLC22A2) mRNA were higher than other organic ion transporters (Motohashi et al., 2002). Moreover, hOAT1, hOAT3 and hOCT2 protein localized at the basolateral membrane of the renal proximal tubules. Accordingly, these transporters play important roles for the renal uptake of organic compounds from the circulation.

It has been assumed that the renal tubular secretion of famotidine and cimetidine would be mediated by OCTs due to their cationic property at the physiological pH (Fig. 1). Indeed, cimetidine inhibits the renal secretion of various organic cations (Somogyi et al., 1983, Christian et al., 1984, van Crugten et al., 1986) and Urakami et al., 2001, Urakami et al., 2002 indicated that rat and human organic cation transporters rOCT1, rOCT2 and hOCT2 transported cimetidine. However, in addition to hOCT2, hOAT1, hOAT3 and rOAT3 were shown to transport the cimetidine. (Kusuhara et al., 1999, Cha et al., 2001, Burckhardt et al., 2003). Therefore, it was supposed that these three transporters were concerned with renal secretion of cimetidine. While famotidine is more frequently used than cimetidine because of it beneficial characteristics, adverse effects associated with famotidine, such as thrombocytopenia (Wade et al., 2002) or central nerve system reaction (Yoshimoto et al., 1994), have been reported. Although dosage should be adjusted according to renal function, molecular mechanisms of renal secretion of famotidine have not been investigated in contrast to cimetidine. To understand the renal handling of famotidine, it should be examined whether hOATs or hOCTs recognize and transport famotidine.

The purpose of the present study is to clarify the recognition and transport properties of famotidine by hOAT1, hOAT3 and hOCT2 in comparison with cimetidine. We performed transport experiments of famotidine and cimetidine using Xenopus laevis oocytes expressing these organic ion transporters.

Section snippets

Materials

p-[Glycyl-14C]aminohippurate (PAH; 1.9 GBq/mmol), [1-14C]tetraethylammonium bromide (TEA; 88.8 MBq/mmol) and [6,7-3H(N)]estrone sulfate were obtained from Perkin-Elmer Life Science Products (Boston, MA, USA). Famotidine and cimetidine were purchased from Wako Pure Chemical Industries (Osaka, Japan) and Nacalai Tesque (Kyoto, Japan), respectively. Unlabeled PAH and estrone 3-sulfate were from Sigma (St. Louis, MO, USA). 1-Methyl-4-phenylpyridinium (MPP) was from Research Biochemicals

Inhibitory effects of famotidine and cimetidine on hOAT1, hOAT3 and hOCT2

To assess whether famotidine interacts with hOAT1, hOAT3 and hOCT2, we examined its inhibitory effects on organic ion transporters (Fig. 2). Famotidine at 5 mM completely inhibited [3H]estrone sulfate transport by hOAT3 and [14C]TEA transport by hOCT2 but did not affect [14C]PAH uptake by hOAT1. To compare the inhibitory potencies of famotidine for the transport activity of hOAT3 and hOCT2 with those of cimetidine, half-maximal inhibitory concentrations (IC50) were estimated. As shown in Fig. 2

Discussion

In this study, we examined whether basolateral organic ion transporters such as hOAT1, hOAT3 and hOCT2 recognized famotidine as substrate in comparison with cimetidine. It was shown that there were marked differences between famotidine and cimetidine in the recognition and transport properties by renal organic ion transporters.

It was indicated that efficient tubular secretion contributed to renal excretion of famotidine and cimetidine in subjects with normal renal function. Inasmuch as these

Acknowledgements

This study was supported by a grant-in-aid for Comprehensive Research on Aging and Health from the Ministry of Health, Labor, and Welfare of Japan, by a grant-in-aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan, and by 21st Century COE Program “Knowledge Information Infrastructure for Genome Science.”

References (30)

  • S.H. Cha et al.

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

    Mol. Pharmacol.

    (2001)
  • C.D. Christian et al.

    Cimetidine inhibits renal procainamide clearance

    Clin. Pharmacol. Ther.

    (1984)
  • L.G. Gisclon et al.

    The effect of probenecid on the renal elimination of cimetidine

    Clin. Pharmacol. Ther.

    (1989)
  • T.J. Humphries

    Famotidine: a notable lack of drug interactions

    Scand. J. Gastroenterol.

    (1987)
  • N. Inotsume et al.

    The inhibitory effect of probenecid on renal excretion of famotidine in young, healthy volunteers

    J. Clin. Pharmacol.

    (1990)
  • Cited by (41)

    • The promiscuous binding of pharmaceutical drugs and their transporter-mediated uptake into cells: What we (need to) know and how we can do so

      2013, Drug Discovery Today
      Citation Excerpt :

      While a study of 12,000 gene tags, 443 carriers and no proper hold-out set is not capable of explaining robustly [272,273] the permeability properties of just 26 drugs, our focus lies on the drugs studied. Specifically, the list of compounds that were not removed, and thus presumably taken to lack significant interactions with carriers, is as follows, but now with non-exhaustive references added by us to show that all of them possess, or interact with, known (and, often, multiple) transporters: furosemide (six transporters, e.g. http://www.drugbank.ca/drugs/DB00695#transporters, [274,275]), hydrochlorothiazide [275–280], atenolol [281–284]; cimetidine (12 transporters http://www.drugbank.ca/drugs/DB00501#transporters, e.g. [60,164,285–294]), mannitol (believed to be transported, if at all, via a paracellular route [295–297]), terbutaline [294], metoprolol [298–300], propranolol [299–304], desipramine [305–307], piroxicam [308–315], ketoprofen [312,314,316–318] and naproxen [308,312,316,319]. As before (Fig. 2b of [5]), we would point out that claims about the absence (or that ignore the presence) of a transporter interacting with a named drug may often be dismissed following a simple literature search or an inspection of public databases [320–322] such as DrugBank (http://www.drugbank.ca/) or others in Table 1 and elsewhere.

    • Drug transport by Organic Anion Transporters (OATs)

      2012, Pharmacology and Therapeutics
      Citation Excerpt :

      Sitagliptin did not inhibit human OAT1 and was not transported, as was the organic cation metformin (Kimura et al., 2005; Chu et al., 2007). Cimetidine and ranitidine, but not famotidine, were transported by human OAT1 (Burckhardt et al., 2003; Motohashi et al., 2004; Tahara et al., 2005a). For rat Oat1, transport (Nagata et al., 2002) or no transport (Prueksaritanont et al., 2004) of cimetidine was reported and, for mouse Oat1, no cimetidine transport was found (Ahn et al., 2009).

    • Interaction and transport characteristics of mycophenolic acid and its glucuronide via human organic anion transporters hOAT1 and hOAT3

      2007, Biochemical Pharmacology
      Citation Excerpt :

      However, in patients with hypoalbuminemia or renal impairment, care should be taken to avoid MMF-related drug interaction via hOAT1 and hOAT3, because there is a possibility that plasma concentrations of unbound MPA and MPAG will reach levels inhibitory to the transporters. Among drugs frequently administered to recipients of organ transplantations, ganciclovir and famotidine as well as acyclovir are substrates of hOAT1 or hOAT3 [16,17,32] and are myelosuppressive in addition to MPA. Furthermore, the present study shows that MPAG is a substrate of hOAT3, suggesting that hOAT3 contributes to the renal tubular secretion of MPAG.

    • Solute Carrier Transportome in Chemotherapy-Induced Adverse Drug Reactions

      2022, Reviews of Physiology, Biochemistry and Pharmacology
    View all citing articles on Scopus
    View full text