151
|
König J, Nies AT, Cui Y, Leier I, Keppler D. Conjugate export pumps of the multidrug resistance protein (MRP) family: localization, substrate specificity, and MRP2-mediated drug resistance. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1461:377-94. [PMID: 10581368 DOI: 10.1016/s0005-2736(99)00169-8] [Citation(s) in RCA: 544] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The membrane proteins mediating the ATP-dependent transport of lipophilic substances conjugated to glutathione, glucuronate, or sulfate have been identified as members of the multidrug resistance protein (MRP) family. Several isoforms of these conjugate export pumps with different kinetic properties and domain-specific localization in polarized human cells have been cloned and characterized. Orthologs of the human MRP isoforms have been detected in many different organisms. Studies in mutant rats lacking the apical isoform MRP2 (symbol ABCC2) indicate that anionic conjugates of endogenous and exogenous substances cannot exit from cells at a sufficient rate unless an export pump of the MRP family is present in the plasma membrane. Several mutations in the human MRP2 gene have been identified which lead to the absence of the MRP2 protein from the hepatocyte canalicular membrane and to the conjugated hyperbilirubinemia of Dubin-Johnson syndrome. Overexpression of recombinant MRP2 confers resistance to multiple chemotherapeutic agents. Because of its function in the terminal excretion of cytotoxic and carcinogenic substances, MRP2 as well as other members of the MRP family, play an important role in detoxification and chemoprevention.
Collapse
Affiliation(s)
- J König
- Division of Tumor Biochemistry, Deutsches Krebsforschungszentrum, D-69120, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
152
|
Hipfner DR, Deeley RG, Cole SP. Structural, mechanistic and clinical aspects of MRP1. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1461:359-76. [PMID: 10581367 DOI: 10.1016/s0005-2736(99)00168-6] [Citation(s) in RCA: 278] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The cDNA encoding ATP-binding cassette (ABC) multidrug resistance protein MRP1 was originally cloned from a drug-selected lung cancer cell line resistant to multiple natural product chemotherapeutic agents. MRP1 is the founder of a branch of the ABC superfamily whose members (from species as diverse as plants and yeast to mammals) share several distinguishing structural features that may contribute to functional and mechanistic similarities among this subgroup of transport proteins. In addition to its role in resistance to natural product drugs, MRP1 (and related proteins) functions as a primary active transporter of structurally diverse organic anions, many of which are formed by the biotransformation of various endo- and xenobiotics by Phase II conjugating enzymes, such as the glutathione S-transferases. MRP1 is involved in a number of glutathione-related cellular processes. Glutathione also appears to play a key role in MRP1-mediated drug resistance. This article reviews the discovery of MRP1 and its relationships with other ABC superfamily members, and summarizes current knowledge of the structure, transport functions and relevance of this protein to in vitro and clinical multidrug resistance.
Collapse
Affiliation(s)
- D R Hipfner
- Cancer Research Laboratories, Queen's University, Kingston, Ont., Canada
| | | | | |
Collapse
|
153
|
Tanaka T, Uchiumi T, Hinoshita E, Inokuchi A, Toh S, Wada M, Takano H, Kohno K, Kuwano M. The human multidrug resistance protein 2 gene: functional characterization of the 5'-flanking region and expression in hepatic cells. Hepatology 1999; 30:1507-12. [PMID: 10573531 DOI: 10.1002/hep.510300617] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
The human multidrug resistance protein 2 (MRP2), also termed as the canalicular multispecific organic anion transporter (cMOAT), is a member of the adenosine triphosphate-binding cassette transporter superfamily. In the liver, MRP2 mediates the multispecific efflux of various types of organic anions, including glucuronate, sulfate, and glutathione conjugates, across the canalicular hepatocyte membrane to the bile. To investigate how the MRP2 gene is expressed in liver cells, the 5'-flanking region of the human MRP2 gene was isolated from a human placental genomic library. Sequence analysis of the MRP2 promoter showed a number of consensus binding sites for both ubiquitous and liver-enriched transcription factors. Transfection of human hepatic HepG2 cells with a series of 5'-deleted promoter luciferase constructs identified a putative silencer element localized in the -1,659/-491 region and a liver-specific positive regulatory element localized in the -491/-258 region. This latter region contained the liver-abundant transcription factor CCAAT-enhancer binding protein beta (C/EBPbeta). The transcriptional activity of the promoter construct containing a mutation in the C/EBPbeta binding site was significantly decreased in HepG2 cells. This study suggests that C/EBPbeta (-356 to -343) may regulate the liver expression of the MRP2 gene.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/chemistry
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP-Binding Cassette Transporters/chemistry
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Amino Acid Sequence
- Base Sequence
- CCAAT-Enhancer-Binding Proteins
- Cloning, Molecular
- DNA-Binding Proteins/physiology
- Drug Resistance, Multiple/genetics
- Gene Expression Regulation/genetics
- Gene Silencing
- Genes, Reporter
- Genomic Library
- Humans
- Liver/cytology
- Liver/metabolism
- Molecular Sequence Data
- Multidrug Resistance-Associated Protein 2
- Nuclear Proteins/physiology
- Promoter Regions, Genetic/genetics
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Response Elements/genetics
- Sequence Deletion/genetics
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- T Tanaka
- Department of Biochemistry, Kyushu University School of Medicine, Fukuoka, University of Occupational Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Safa AR. Photoaffinity analogs for multidrug resistance-related transporters and their use in identifying chemosensitizers. Drug Resist Updat 1999; 2:371-381. [PMID: 11498353 DOI: 10.1054/drup.1999.0105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A major obstacle in cancer treatment is the development of resistance to multiple chemotherapeutic agents in tumor cells. The hallmark of this multidrug resistance (MDR) is overexpression of the MDR 1 P-glycoprotein or the multidrug resistance protein MRP1. It is well documented that these proteins confer MDR in cancer cells. Much evidence indicates that control of intracellular drug levels in MDR cells is determined by P-glycoprotein or MRP, and therefore these proteins are suitable targets for identifying MDR-reversing agents (MDR modulators). We originally explored the drug-binding ability of P-glycoprotein by synthesizing and using radioactive photoaffinity analogs of vinblastine. Since our initial discovery that P-glycoprotein binds to vinblastine photoaffinity analogs, many P-glycoprotein- and MRP-specific photoaffinity analogs have been developed. In this review, photoaffinity analogs which specifically bind to P-glycoprotein or MRP are discussed. Moreover, utilizing these photoprobes to identify, characterize and localize the drug binding sites of P-glycoprotein and MRP is described. Using P-glycoprotein-specific photoaffinity analogs in combination with site-directed antibodies to several domains of this protein has allowed the localization of the general binding domains of some of the cytotoxic agents an MDR modulators on P-glycoprotein. However, the molecular architecture of the drug binding sites, their exact location on the P-glycoprotein molecule, and the total number of the drug binding sites remain to be determined. This review discusses recent advances in delineating the structure of the drug-binding sites of P-glycoprotein. Moreover, novel MRP1 photoaffinity analogs are reviewed. Copyright 1999 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Ahmad R. Safa
- Indiana University Cancer Research Institute, 1044 West Walnut, Indianapolis, IN, 46202, USA
| |
Collapse
|
155
|
Courtois A, Payen L, Vernhet L, Morel F, Guillouzo A, Fardel O. Differential regulation of canalicular multispecific organic anion transporter (cMOAT) expression by the chemopreventive agent oltipraz in primary rat hepatocytes and in rat liver. Carcinogenesis 1999; 20:2327-30. [PMID: 10590227 DOI: 10.1093/carcin/20.12.2327] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Expression of the canalicular multispecific organic anion transporter (cMOAT), an efflux pump involved in biliary secretion of xenobiotics, was investigated in rat hepatocytes exposed to the chemopreventive agent oltipraz. Northern blotting indicated that this compound increased cMOAT mRNA levels in primary cultured hepatocytes. Such an induction of cMOAT transcripts was demonstrated to be dose-dependent and started as early as 4 h treatment; in addition, western blotting showed increased levels of 190 kDa cMOAT in oltipraz-treated primary rat hepatocytes when compared with their untreated counterparts. In contrast, administration of oltipraz to rats failed to enhance hepatic cMOAT mRNA and protein amounts whereas it was found to induce liver expression of glutathione S-transferase P1, a well-known oltipraz-regulated drug metabolizing enzyme. These data therefore suggest that cMOAT up-regulation occurring in rat hepatocytes in response to oltipraz may be restricted to in vitro situations and is therefore unlikely to be directly involved in the in vivo chemopreventive properties of oltipraz.
Collapse
Affiliation(s)
- A Courtois
- INSERM U456 'D¿etoxication et R¿eparation Tissulaire', Facult¿e de Pharmacie, 2 avenue du Pr L. Bernard, 35043 Rennes Cedex, France
| | | | | | | | | | | |
Collapse
|
156
|
Abstract
The release of glutathione S-conjugates from cells is an ATP-dependent process mediated by integral membrane glycoproteins belonging to the recently discovered multidrug-resistance protein (MRP) family. Many lipophilic compounds conjugated with glutathione, glucuronate, or sulfate are substrates for export pumps of the MRP family. In humans six MRP isoforms encoded by different genes have been cloned. Orthologs of MRP have been identified in many species including yeast, plants, and nematodes. Human MRP1 and MRP2 are currently best characterized with respect to substrate specificity by measurements of ATP-dependent transport into inside-out membrane vesicles. High-affinity substrates include the glutathione S-conjugate leukotriene C4, S-(2,4dinitrophenyl)glutathione, bilirubin glucuronosides, and 17beta-glucuronosyl estradiol. In addition, glutathione disulfide is transported by MRP1 and MRP2. Reduced glutathione may be released from cells in a process directly or indirectly mediated by members of the MRP family. Proteins of the MRP family are indispensable for transport of glutathione S-conjugates and glutathione disulfide into the extracellular space and play, therefore, a decisive role in detoxification and defense against oxidative stress.
Collapse
Affiliation(s)
- D Keppler
- Division of Tumor Biochemistry, Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| |
Collapse
|
157
|
Kakyo M, Unno M, Tokui T, Nakagomi R, Nishio T, Iwasashi H, Nakai D, Seki M, Suzuki M, Naitoh T, Matsuno S, Yawo H, Abe T. Molecular characterization and functional regulation of a novel rat liver-specific organic anion transporter rlst-1. Gastroenterology 1999; 117:770-5. [PMID: 10500057 DOI: 10.1016/s0016-5085(99)70333-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Recently, we isolated a new complementary DNA (cDNA) encoding human liver-specific organic anion transporter (LST-1), representing the multispecificity of human liver. The aim of this study was to isolate a rat counterpart of human LST-1 and examine the expression regulation of its messenger RNA (mRNA) to clarify the molecular basis of cholestasis. METHODS A rat liver cDNA library was screened with human LST-1 cDNA as a probe. Xenopus oocyte expression system was used for functional analysis. Northern blot analyses were performed using the isolated cDNA (termed rlst-1). The bile duct ligation model and the cecum ligation and puncture model were used for expression analyses. RESULTS rlst-1 encodes 652 amino acids, predicting at least 11 transmembrane regions. The overall homology with human LST-1 was 60.2%, which is the highest among all known organic anion transporters. rlst-1 also belongs to the same new gene family as human LST-1, located between the organic anion transporter family and the prostaglandin transporter. rlst-1 preferably transports taurocholate (K(m), 9.45 micromol/L) in an Na(+)-independent manner. The rlst-1 mRNA is exclusively expressed in the liver. In both the bile duct ligation model and the cecum ligation and puncture model, mRNA expression levels of rlst-1 were down-regulated. CONCLUSIONS rlst-1 is a counterpart of human LST-1 and is one of the important transporters in rat liver for the clearance of bile acid. The expression of rlst-1 may be under feedback regulation of cholestasis by biliary obstruction and/or sepsis.
Collapse
Affiliation(s)
- M Kakyo
- Department of Neurophysiology, Tohoku University School of Medicine, Sendai, Ltd., Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Mills CO, Milkiewicz P, Müller M, Roma MG, Havinga R, Coleman R, Kuipers F, Jansen PL, Elias E. Different pathways of canalicular secretion of sulfated and non-sulfated fluorescent bile acids: a study in isolated hepatocyte couplets and TR- rats. J Hepatol 1999; 31:678-84. [PMID: 10551392 DOI: 10.1016/s0168-8278(99)80348-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS Fluorescent bile acids have proved useful for characterizing bile salt transport mechanisms. The aim of this study was to further validate the use of lysyl-fluorescein conjugated bile acid analogues as surrogate bile acids. METHODS We analyzed biliary excretion kinetics of cholyl lysyl fluorescein (CLF), lithocholyl lysyl fluorescein (LLF) and sulfo-lithocholyl lysyl fluorescein (sLLF), both in the isolated rat hepatocyte couplet model and in TR- rats with a selective canalicular transport defect of non-bile acid organic anions. RESULTS CLF and LLF, which like their natural nonsulfated bile acid congeners are expected to be handled by the canalicular bile salt export pump, were transferred into the bile canaliculus much faster than sLLF, a putative substrate for the canalicular multispecific organic anion transporter in both the in vivo and the in vitro models employed. The contention that different transport systems are involved in sulfated and non-sulfated lysyl fluorescein conjugated bile acids biliary excretion was supported further by studies using TR- rats, in which the cumulative biliary excretion of sLLF was reduced to 6% as compared with that of normal Wistar rats, in good agreement with values for its naturally-occurring radiolabeled parent compound sulfoglycolithocholate. In contrast, CLF and LLF were reduced to 66% and 52%, similar values to these for their congeners, [14C] glycocholate and [14C] lithocholate. CONCLUSION The close similarity in behavior of lysyl fluorescein conjugated bile acids to that of their naturally-occurring parent compounds in these different models gives support for both sulfated and nonsulfated lysyl fluorescein conjugated bile acids as substitute molecules for studies of bile acid transport.
Collapse
Affiliation(s)
- C O Mills
- Liver Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Soroka CJ, Pate MK, Boyer JL. Canalicular export pumps traffic with polymeric immunoglobulin A receptor on the same microtubule-associated vesicle in rat liver. J Biol Chem 1999; 274:26416-24. [PMID: 10473600 DOI: 10.1074/jbc.274.37.26416] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Basolateral to apical vesicular transcytosis in the hepatocyte is an essential pathway for the delivery of compounds from the sinusoidal blood to the bile and to traffic newly synthesized resident apical membrane proteins to their site of function at the canalicular membrane front. To characterize this pathway better, microtubules in a hepatocyte homogenate were polymerized by addition of taxol, and associated membrane-bound vesicles were isolated. This fraction was enriched in polymeric immunoglobulin A receptor and contained apical membrane proteins. Immunoelectron microscopy demonstrated that polymeric immunoglobulin A receptor was localized predominantly on vesicles ranging from 100 to 160 nm and that the multidrug resistance protein 2 and the bile salt export pump co-localized on these vesicles. The minus-ended microtubule motor, dynein, was highly enriched in the fraction, and its intermediate chain could be released effectively by incubation with 1 mM ATP or GTP. However, the association of the transcytotic vesicles with the microtubules was not sensitive to hydrolyzable or non-hydrolyzable nucleotides. This study characterizes a fraction of microtubule-associated vesicles from rat hepatocytes and demonstrates that several resident apical membrane transport proteins and the polymeric immunoglobulin A receptor traffic on the same vesicle.
Collapse
Affiliation(s)
- C J Soroka
- Department of Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut 06520-8019, USA.
| | | | | |
Collapse
|
160
|
Martel F, Ribeiro L, Calhau C, Azevedo I. Inhibition by levamisole of the organic cation transporter rOCT1 in cultured rat hepatocytes. Pharmacol Res 1999; 40:275-9. [PMID: 10479473 DOI: 10.1006/phrs.1999.0506] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Levamisole is known to be subject to hepatic removal and metabolism and to biliary excretion. The aim of our work was to study the mechanism involved in the removal of this compound by the liver. For this purpose, we studied the influence of levamisole on the uptake and efflux of the model organic cation 1-methyl-4-phenylpyridinium (MPP(+)) by primary cultured rat hepatocytes. Levamisole (500 microm) was found to produce a strong inhibition (to 31+/-2% of control) of [(3)H]MPP(+)uptake. Moreover, efflux of [(3)H]MPP(+)was also potently reduced by levamisole (500 microm). Our results show that levamisole interferes with an hepatic organic cation transporter which accepts MPP(+)as a substrate. This mechanism most probably corresponds to rOCT1, and it might be responsible for the hepatic removal of levamisole from the blood circulation.
Collapse
Affiliation(s)
- F Martel
- Department of Biochemistry, Faculty of Medicine, Porto, 4200, Portugal
| | | | | | | |
Collapse
|
161
|
Martel F, Ribeiro L, Calhau C, Azevedo I. Characterization of the efflux of the organic cation MPP+ in cultured rat hepatocytes. Eur J Pharmacol 1999; 379:211-8. [PMID: 10497908 DOI: 10.1016/s0014-2999(99)00501-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The aim of this study was to characterize the efflux of organic cations from primary cultured rat hepatocytes, using 1-methyl-4-phenylpyridinium (MPP+) as a model compound. The efflux of [3H]MPP+ was temperature dependent, and pH and metabolic inhibition independent. It was either strongly reduced (verapamil, vinblastine and rhodamine123) or only moderately reduced (daunomycin) by other organic cations. The anti-P-glycoprotein antibody UIC2 (20 microg/ml) and the P-glycoprotein inhibitors vanadate and cyclosporine A had no effect on [3H]MPP+ efflux. Decynium22 and corticosterone, known inhibitors of rat Organic Cation Transporter 1 (rOCT1), markedly reduced [3H]MPP+ efflux. The uptake of [3H]MPP+ into hepatocytes, known to be mediated by rOCT1, was inhibited by verapamil and vinblastine (IC50s of 2.6 and 34.4 microM, respectively). In conclusion, [3H]MPP+ efflux from primary cultured rat hepatocytes appears to be mediated by rOCT1, a polyspecific organic cation transporter. Moreover, our results do not support the involvement of P-glycoprotein or of an organic cation/proton antiporter in the efflux of [3H]MPP+.
Collapse
Affiliation(s)
- F Martel
- Department of Biochemistry, Faculty of Medicine, Porto, Portugal
| | | | | | | |
Collapse
|
162
|
Demonstration of a coupled metabolism-efflux process at the choroid plexus as a mechanism of brain protection toward xenobiotics. J Neurosci 1999. [PMID: 10414957 DOI: 10.1523/jneurosci.19-15-06275.1999] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Brain homeostasis depends on the composition of both brain interstitial fluid and CSF. Whereas the former is largely controlled by the blood-brain barrier, the latter is regulated by a highly specialized blood-CSF interface, the choroid plexus epithelium, which acts either by controlling the influx of blood-borne compounds, or by clearing deleterious molecules and metabolites from CSF. To investigate mechanisms of brain protection at the choroid plexus, the blood-CSF barrier was reconstituted in vitro by culturing epithelial cells isolated from newborn rat choroid plexuses of either the fourth or the lateral ventricle. The cells grown in primary culture on semipermeable membranes established a pure polarized monolayer displaying structural and functional barrier features, (tight junctions, high electric resistance, low permeability to paracellular markers) and maintaining tissue-specific markers (transthyretin) and specific transporters for micronutriments (amino acids, nucleosides). In particular, the high enzymatic drug metabolism capacity of choroid plexus was preserved in the in vitro blood-CSF interface. Using this model, we demonstrated that choroid plexuses can act as an absolute blood-CSF barrier toward 1-naphthol, a cytotoxic, lipophilic model compound, by a coupled metabolism-efflux mechanism. This compound was metabolized in situ via uridine diphosphate glururonosyltransferase-catalyzed conjugation, and the cellular efflux of the glucurono-conjugate was mediated by a transporter predominantly located at the basolateral, i.e., blood-facing membrane. The transport process was temperature-dependent, probenecid-sensitive, and recognized other glucuronides. Efflux of 1-naphthol metabolite was inhibited by intracellular glutathione S-conjugates. This metabolism-polarized efflux process adds a new facet to the understanding of the protective functions of choroid plexuses.
Collapse
|
163
|
Kamisako T, Leier I, Cui Y, König J, Buchholz U, Hummel-Eisenbeiss J, Keppler D. Transport of monoglucuronosyl and bisglucuronosyl bilirubin by recombinant human and rat multidrug resistance protein 2. Hepatology 1999; 30:485-90. [PMID: 10421658 DOI: 10.1002/hep.510300220] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The secretion of bilirubin conjugates from hepatocytes into bile represents a decisive step in the prevention of hyperbilirubinemia. The bilirubin conjugates, monoglucuronosyl bilirubin (MGB) and bisglucuronosyl bilirubin (BGB), were previously suggested to be endogenous substrates for the apical multidrug resistance protein (MRP2), a member of the adenosine triphosphate (ATP)-binding cassette family of transporters (symbol ABCC2), also termed canalicular multispecific organic anion transporter. We have characterized this ATP-dependent transport using membrane vesicles from human embryonic kidney (HEK) cells expressing recombinant rat as well as human MRP2. MGB and BGB, (3)H-labeled in the glucuronosyl moiety, were synthesized enzymatically with recombinant UDP-glucuronosyltransferase 1A1, and stabilized with ascorbate. Rates for ATP-dependent transport of MGB and BGB (0.5 micromol/L each) by human MRP2 were 183 and 104 pmol x mg protein(-1) x min(-1), respectively. K(m) values were 0.7 and 0.9 micromol/L for human MRP2, and 0.8 and 0.5 micromol/L for rat MRP2, with MGB and BGB as substrates, respectively. Leukotriene C(4) and 17beta-glucuronosyl estradiol, which are both known high-affinity substrates for human MRP2, inhibited [(3)H]MGB transport with IC(50) values of 2.3 and 30 micromol/L, respectively. Cyclosporin A competitively inhibited human and rat MRP2-mediated transport of [(3)H]MGB, with K(i) values of 21 and 10 micromol/L, respectively. Our results provide direct evidence that recombinant MRP2, cloned from rat as well as human liver, mediates the primary-active ATP-dependent transport of the bilirubin conjugates MGB and BGB.
Collapse
Affiliation(s)
- T Kamisako
- Division of Tumor Biochemistry, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
164
|
Affiliation(s)
- M Trauner
- Department of Medicine, Karl Franzens University, Graz, Austria
| | | | | |
Collapse
|
165
|
Liu X, LeCluyse EL, Brouwer KR, Gan LS, Lemasters JJ, Stieger B, Meier PJ, Brouwer KL. Biliary excretion in primary rat hepatocytes cultured in a collagen-sandwich configuration. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:G12-21. [PMID: 10409146 DOI: 10.1152/ajpgi.1999.277.1.g12] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The objective of the present investigation was to examine the functional reestablishment of polarity in freshly isolated hepatocytes cultured between 2 layers of gelled collagen (sandwich configuration). Immunoblot analysis demonstrated that the canalicular multispecific organic anion transport protein (multidrug resistance-associated protein, Mrp2) was partially maintained in day 5 hepatocytes cultured in a sandwich configuration. Fluorescein-labeled taurocholate and carboxydichlorofluorescein were excreted into and concentrated in the bile canalicular lumen of day 5 sandwich-cultured hepatocytes, resulting in formation of fluorescent networks in standard buffer (intact bile canaliculi). Confocal microscopy studies demonstrated that 1) carboxydichlorofluorescein that had concentrated in the canalicular lumen was released into the incubation buffer in the presence of Ca(2+)-free buffer (disrupted bile canaliculi), and 2) rhodamine-dextran, an extracellular space marker, was only able to diffuse into the canalicular lumen in the presence of Ca(2+)-free buffer. The cumulative uptake of [(3)H]taurocholate in day 5 sandwich-cultured hepatocytes was significantly higher in standard buffer compared with Ca(2+)-free buffer, due to accumulation of taurocholate in canalicular spaces. When [(3)H]taurocholate was preloaded in the day 5 sandwich-cultured hepatocytes, taurocholate efflux was greater in Ca(2+)-free compared with standard buffer. The biliary excretion index of taurocholate, equivalent to the percentage of retained taurocholate in the canalicular networks, increased from approximately 8% at day 0 to approximately 60% at day 5 in sandwich-cultured hepatocytes. In summary, hepatocytes cultured in a collagen-sandwich configuration for up to 5 days establish intact canalicular networks, maintain Mrp2, reestablish polarized excretion of organic anions and bile acids, and represent a useful in vitro model system to investigate the hepatobiliary disposition of substrates.
Collapse
Affiliation(s)
- X Liu
- Division of Drug Delivery and Disposition, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
166
|
Kool M, van der Linden M, de Haas M, Scheffer GL, de Vree JM, Smith AJ, Jansen G, Peters GJ, Ponne N, Scheper RJ, Elferink RP, Baas F, Borst P. MRP3, an organic anion transporter able to transport anti-cancer drugs. Proc Natl Acad Sci U S A 1999; 96:6914-9. [PMID: 10359813 PMCID: PMC22016 DOI: 10.1073/pnas.96.12.6914] [Citation(s) in RCA: 443] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The human multidrug-resistance protein (MRP) gene family contains at least six members: MRP1, encoding the multidrug-resistance protein; MRP2 or cMOAT, encoding the canalicular multispecific organic anion transporter; and four homologs, called MRP3, MRP4, MRP5, and MRP6. In this report, we characterize MRP3, the closest homolog of MRP1. Cell lines were retrovirally transduced with MRP3 cDNA, and new monoclonal antibodies specific for MRP3 were generated. We show that MRP3 is an organic anion and multidrug transporter, like the GS-X pumps MRP1 and MRP2. In Madin-Darby canine kidney II cells, MRP3 routes to the basolateral membrane and mediates transport of the organic anion S-(2,4-dinitrophenyl-)glutathione toward the basolateral side of the monolayer. In ovarian carcinoma cells (2008), expression of MRP3 results in low-level resistance to the epipodophyllotoxins etoposide and teniposide. In short-term drug exposure experiments, MRP3 also confers high-level resistance to methotrexate. Neither 2008 cells nor Madin-Darby canine kidney II cells overexpressing MRP3 showed an increase in glutathione export or a decrease in the level of intracellular glutathione, in contrast to cells overexpressing MRP1 or MRP2. We discuss the possible function of MRP3 in (hepatic) physiology and its potential contribution to drug resistance of cancer cells.
Collapse
Affiliation(s)
- M Kool
- Division of Molecular Biology and Center of Biomedical Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Wehner F, Tinel H. Uptake of bromosulfophthalein via SO2-4/OH- exchange increases the K+ conductance of rat hepatocytes. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G1380-90. [PMID: 10362641 DOI: 10.1152/ajpgi.1999.276.6.g1380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
In confluent primary cultures of rat hepatocytes, micromolar concentrations of bromosulfophthalein (BSP) lead to a sizeable hyperpolarization of membrane voltage. The effect is a saturable function of BSP concentration yielding an apparent value of 226 micromol/l and a Vmax of -10.3 mV. The BSP-induced membrane hyperpolarization is inhibited by the K+ channel blocker Ba2+, and in cable-analysis and ion-substitution experiments it becomes evident that the effect is due to a significant increase in cell membrane K+ conductance. Voltage changes were attenuated by the simultaneous administration of SO2-4, succinate, and cholate (cis-inhibition) and increased after preincubation with SO2-4 and succinate (trans-stimulation), suggesting that the effect occurs via BSP uptake through the known SO2-4/OH- exchanger. Microfluorometric measurements reveal that BSP-induced activation of K+ conductance is not mediated by changes in cell pH, cell Ca2+, or cell volume. It is concluded that K+ channel activation by BSP (as well as by DIDS and indocyanine green) may reflect a physiological mechanism linking the sinusoidal uptake of certain anions to their electrogenic canalicular secretion.
Collapse
Affiliation(s)
- F Wehner
- Max-Planck-Institut für molekulare Physiologie, Abteilung Epithelphysiologie, 44139 Dortmund, Germany.
| | | |
Collapse
|
168
|
Abstract
Rat liver slices were employed as experimental model to characterise the system involved in the transport process which participates in liver tyramine uptake. The uptake of 0.4 micromol l-1of [3H]tyramine by rat liver slices was linear from 5 min up to the end of incubation. At 15 min the uptake was 4.58+/-0.18 pmol mg-1protein. The accumulation of [3H]tyramine was sensitive to temperature (69. 3+/-4.0% inhibition at 0 degrees C, P<0.001), to sodium omission replaced by 150 mmol l-1Tris or 110 mmol l-1Tris+40 mmol l-1choline (27.6+/-6.0%, P<0.01, and 24.6+/-3.8% inhibition, P<0.01, respectively), and the inhibition of Na+-K+-adenosine triphosphatase by 150 micromol l-1ouabain (20.4+/-2.6% decrease, P<0.01). Uptake of [3H]tyramine was cocaine- (10 micromol l-1) and desipramine- (1 micromol l-1) dependent (32.2+/-6.4%, P<0.05, and 31.6+/-4.0% inhibition, P<0.05, respectively). Uptake of [3H]tyramine in rat liver slices was not modified by 30 micromol l-1isoprenaline, 30 micromol l-1corticosterone, 30 micromol l-1normetanephrine and noradrenaline up to 4 micrometers at higher noradrenaline concentrations tyramine transport was diminished (P<0.05). Results achieved by incubation with increasing tyramine concentrations indicate that at the micromolar level hepatic uptake occurs by a combined passive diffusion and transport-mediated mechanism, whereas at greater tyramine concentrations passive transport predominates. These results suggest that both simple diffusion and a transport-mediated mechanism are involved in this uptake from hepatocytes, which presents features similar to those described for type 1 non-neuronal uptake systems.
Collapse
Affiliation(s)
- D M Tchercansky
- Cátedra de Farmacologiá, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, Argentina
| | | | | |
Collapse
|
169
|
Tuma PL, Finnegan CM, Yi JH, Hubbard AL. Evidence for apical endocytosis in polarized hepatic cells: phosphoinositide 3-kinase inhibitors lead to the lysosomal accumulation of resident apical plasma membrane proteins. J Cell Biol 1999; 145:1089-102. [PMID: 10352024 PMCID: PMC2133136 DOI: 10.1083/jcb.145.5.1089] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/1999] [Revised: 04/15/1999] [Indexed: 12/18/2022] Open
Abstract
The architectural complexity of the hepatocyte canalicular surface has prevented examination of apical membrane dynamics with methods used for other epithelial cells. By adopting a pharmacological approach, we have documented for the first time the internalization of membrane proteins from the hepatic apical surface. Treatment of hepatocytes or WIF-B cells with phosphoinositide 3-kinase inhibitors, wortmannin or LY294002, led to accumulation of the apical plasma membrane proteins, 5'-nucleotidase and aminopeptidase N in lysosomal vacuoles. By monitoring the trafficking of antibody-labeled molecules, we determined that the apical proteins in vacuoles came from the apical plasma membrane. Neither newly synthesized nor transcytosing apical proteins accumulated in vacuoles. In wortmannin-treated cells, transcytosing apical proteins traversed the subapical compartment (SAC), suggesting that this intermediate in the basolateral-to-apical transcytotic pathway remained functional. Ultrastructural analysis confirmed these results. However, apically internalized proteins did not travel through SAC en route to lysosomal vacuoles, indicating that SAC is not an intermediate in the apical endocytic pathway. Basolateral membrane protein distributions did not change in treated cells, uncovering another difference in endocytosis from the two domains. Similar effects were observed in polarized MDCK cells, suggesting conserved patterns of phosphoinositide 3-kinase regulation among epithelial cells. These results confirm a long-held but unproven assumption that lysosomes are the final destination of apical membrane proteins in hepatocytes. Significantly, they also confirm our hypothesis that SAC is not an apical endosome.
Collapse
Affiliation(s)
- P L Tuma
- Department of Cell Biology and Anatomy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
170
|
Hirohashi T, Suzuki H, Sugiyama Y. Characterization of the transport properties of cloned rat multidrug resistance-associated protein 3 (MRP3). J Biol Chem 1999; 274:15181-5. [PMID: 10329726 DOI: 10.1074/jbc.274.21.15181] [Citation(s) in RCA: 263] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We have previously cloned rat MRP3 as an inducible transporter in the liver (Hirohashi, T., Suzuki, H., Ito, K., Ogawa, K., Kume, K., Shimizu, T., and Sugiyama, Y. (1998) Mol. Pharmacol. 53, 1068-1075). In the present study, the function of rat MRP3 was investigated using membrane vesicles isolated from LLC-PK1 and HeLa cell population transfected with corresponding cDNA. The ATP-dependent uptake of both 17beta estradiol 17-beta-D-glucuronide ([3H]E217betaG) and glucuronide of [14C] 6-hydroxy-5, 7-dimethyl-2-methylamino-4-(3-pyridylmethyl) benzothiazole (E3040), but not that of [3H]leukotriene C4 and [3H]2, 4-dinitrophenyl-S-glutathione, was markedly stimulated by MRP3 transfection in both cell lines. The Km and Vmax values for the uptake of [3H]E217betaG were 67 +/- 14 microM and 415 +/- 73 pmol/min/mg of protein, respectively, for MRP3-expressing membrane vesicles and 3.0 +/- 0.7 microM and 3.4 +/- 0.4 pmol/min/mg of protein, respectively, for the endogenous transporter expressed on HeLa cells. [3H]E217betaG had also a similar Km value for MRP3 when LLC-PK1 cells were used as the host. All glucuronide conjugates examined (E3040 glucuronide, 4-methylumbelliferone glucuronide, and naphthyl glucuronide) and methotrexate inhibited MRP3-mediated [3H]E217betaG transport in LLC-PK1 cells. Moreover, [3H]methotrexate was transported via MRP3. The inhibitory effect of estrone sulfate, [3H]2,4-dinitrophenyl-S-glutathione, and [3H]leukotriene C4 was moderate or minimal, whereas N-acetyl-2,4-dinitrophenylcysteine had no effect on the uptake of [3H]E217betaG. The uptake of [3H]E217betaG was enhanced by E3040 sulfate and 4-methylumbelliferone sulfate. Thus we were able to demonstrate that several kinds of organic anions are transported via MRP3, although the substrate specificity of MRP3 differs from that of MRP1 and cMOAT/MRP2 in that glutathione conjugates are poor substrates for MRP3.
Collapse
Affiliation(s)
- T Hirohashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | |
Collapse
|
171
|
Canalicular multispecific organic anion transporter/multidrug resistance protein 2 mediates low-affinity transport of reduced glutathione. Biochem J 1999. [PMID: 10024515 DOI: 10.1042/bj3380393] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The canalicular multispecific organic anion transporter (cMOAT), a member of the ATP-binding cassette transporter family, mediates the transport of a broad range of non-bile salt organic anions from liver into bile. cMOAT-deficient Wistar rats (TR-) are mutated in the gene encoding cMOAT, leading to defective hepatobiliary transport of a whole range of substrates, including bilirubin glucuronide. These mutants also have impaired hepatobiliary excretion of GSH and, as a result, the bile flow in these animals is reduced. In the present work we demonstrate a role for cMOAT in the excretion of GSH both in vivo and in vitro. Biliary GSH excretion in rats heterozygous for the cMOAT mutation (TR/tr) was decreased to 63% of controls (TR/TR) (114+/-24 versus 181+/-20 nmol/min per kg body weight). Madin-Darby canine kidney (MDCK) II cells stably expressing the human cMOAT protein displayed >10-fold increase in apical GSH excretion compared with wild-type MDCKII cells (141+/-6.1 pmol/min per mg of protein versus 13.2+/-1.3 pmol/min per mg of protein in wild-type MDCKII cells). Similarly, MDCKII cells expressing the human multidrug resistance protein 1 showed a 4-fold increase in GSH excretion across the basolateral membrane. In several independent cMOAT-transfectants, the level of GSH excretion correlated with the expression level of the protein. Furthermore, we have shown, in cMOAT-transfected cells, that GSH is a low-affinity substrate for the transporter and that its excretion is reduced upon ATP depletion. In membrane vesicles isolated from cMOAT-expressing MDCKII cells, ATP-dependent S-(2,4-dinitrophenyl)glutathione uptake is competitively inhibited by high concentrations of GSH (Ki approximately 20 mM). We concluded that cMOAT mediates low-affinity transport of GSH. However, since hepatocellular GSH concentrations are high (5-10 mM), cMOAT might serve an important physiological function in maintenance of bile flow in addition to hepatic GSH turnover.
Collapse
|
172
|
Green RM, Lo K, Sterritt C, Beier DR. Cloning and functional expression of a mouse liver organic cation transporter. Hepatology 1999; 29:1556-62. [PMID: 10216142 DOI: 10.1002/hep.510290530] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Hepatic uptake of organic cations is essential for the metabolism and secretion of numerous endobiotics and drugs. Several hepatic organic cation transporters have been kinetically defined, yet have not been isolated or cloned. We have isolated a complementary DNA (cDNA) from both murine liver and kidney cDNA libraries (mOct1/Slc22a1), and have functionally expressed it in Xenopus laevis oocytes. Although mOct1/Slc22a1 is homologous to previously cloned rat and human organic cation transporters, organic cation transport kinetics differed markedly. mOct1/Slc22a1-RNA injection of oocytes resulted in the saturable, time- and temperature-dependent uptake of the quaternary organic cation [14C]-tetraethylammonium ([14C]-TEA), with a Km of 38 micromol/L. TEA uptake was inhibited by several other organic cation drugs, but was not inhibited by the organic cation n-methyl-nicotinamide (NMN), being instead stimulated by it (fourfold). [14C]-TEA uptake was also stimulated by an inside-outside proton gradient. mOct1/Slc22a1-injected oocytes transported the organic cations [3H]-1-methyl-4-phenylpyridium and [3H]-choline chloride, but did not transport other classes of organic compounds. mOct1/Slc22a1 encodes for a hepatic and renal organic cation transporter which may be important for the uptake and secretion of cationic drugs and endobiotics.
Collapse
Affiliation(s)
- R M Green
- Section of Digestive and Liver Diseases, University of Illinois at Chicago College of Medicine, Chicago, IL 60612-7323, USA.
| | | | | | | |
Collapse
|
173
|
Niinuma K, Kato Y, Suzuki H, Tyson CA, Weizer V, Dabbs JE, Froehlich R, Green CE, Sugiyama Y. Primary active transport of organic anions on bile canalicular membrane in humans. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G1153-64. [PMID: 10330006 DOI: 10.1152/ajpgi.1999.276.5.g1153] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Biliary excretion of several anionic compounds was examined by assessing their ATP-dependent uptake in bile canalicular membrane vesicles (CMV) prepared from six human liver samples. 2, 4-Dinitrophenyl-S-glutathione (DNP-SG), leukotriene C4 (LTC4), sulfobromophthalein glutathione (BSP-SG), E3040 glucuronide (E-glu), beta-estradiol 17-(beta-D-glucuronide) (E2-17G), grepafloxacin glucuronide (GPFXG), pravastatin, BQ-123, and methotrexate, which are known to be substrates for the rat canalicular multispecific organic anion transporter, and taurocholic acid (TCA), a substrate for the bile acid transporter, were used as substrates. ATP-dependent and saturable uptake of TCA, DNP-SG, LTC4, E-glu, E2-17G, and GPFXG was observed in all human CMV preparations examined, suggesting that these compounds are excreted in the bile via a primary active transport system in humans. Primary active transport of the other substrates was also seen in some of CMV preparations but was negligible in the others. The ATP-dependent uptake of all the compounds exhibited a large inter-CMV variation, and there was a significant correlation between the uptake of glutathione conjugates (DNP-SG, LTC4, and BSP-SG) and glucuronides (E-glu, E2-17G, and GPFXG). However, there was no significant correlation between TCA and the other organic anions, implying that the transporters for TCA and for organic anions are different also in humans. When the average value for the ATP-dependent uptake by each preparation of human CMVs was compared with that of rat CMVs, the uptake of glutathione conjugates and nonconjugated anions (pravastatin, BQ-123, and methotrexate) in humans was approximately 3- to 76-fold lower than that in rats, whereas the uptake of glucuronides was similar in the two species. Thus there is a species difference in the primary active transport of organic anions across the bile canalicular membrane that is less marked for glucuronides.
Collapse
Affiliation(s)
- K Niinuma
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Zucker SD, Goessling W, Hoppin AG. Unconjugated bilirubin exhibits spontaneous diffusion through model lipid bilayers and native hepatocyte membranes. J Biol Chem 1999; 274:10852-62. [PMID: 10196162 DOI: 10.1074/jbc.274.16.10852] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The liver is responsible for the clearance and metabolism of unconjugated bilirubin, the hydrophobic end-product of heme catabolism. Although several putative bilirubin transporters have been described, it has been alternatively proposed that bilirubin enters the hepatocyte by passive diffusion through the plasma membrane. In order to elucidate the mechanism of bilirubin uptake, we measured the rate of bilirubin transmembrane diffusion (flip-flop) using stopped-flow fluorescence techniques. Unconjugated bilirubin rapidly diffuses through model phosphatidylcholine vesicles, with a first-order rate constant of 5.3 s-1 (t(1)/(2) = 130 ms). The flip-flop rate is independent of membrane cholesterol content, phospholipid acyl saturation, and lipid packing, consistent with thermodynamic analyses demonstrating minimal steric constraint to bilirubin transmembrane diffusion. The coincident decrease in pH of the entrapped vesicle volume supports a mechanism whereby the bilirubin molecule crosses the lipid bilayer as the uncharged diacid. Transport of bilirubin by native rat hepatocyte membranes exhibits kinetics comparable with that in model vesicles, suggesting that unconjugated bilirubin crosses cellular membranes by passive diffusion through the hydrophobic lipid core. In contrast, there is no demonstrable flip-flop of bilirubin diglucuronide or bilirubin ditaurate in phospholipid vesicles, yet these compounds rapidly traverse isolated rat hepatocyte membranes, confirming the presence of a facilitated uptake system(s) for hydrophilic bilirubin conjugates.
Collapse
Affiliation(s)
- S D Zucker
- Division of Digestive Diseases, University of Cincinnati Medical Center, Cincinnati, Ohio 45267-0595, USA
| | | | | |
Collapse
|
175
|
Courtois A, Payen L, Lagadic D, Guillouzo A, Fardel O. Evidence for a multidrug resistance-associated protein 1 (MRP1)-related transport system in cultured rat liver biliary epithelial cells. Life Sci 1999; 64:763-74. [PMID: 10075109 DOI: 10.1016/s0024-3205(98)00618-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cellular accumulation and efflux of the anionic fluorescent dye carboxy-2',7'-dichlorofluorescein (CF) were studied in rat liver SDVI cells thought to derive from primitive bile ductules, in order to characterize carrier-related membrane transport of organic anions in epithelial cells. Probenecid, a common blocker of anion transport, was found to strongly enhance CF levels in SDVI cells in a dose-dependent manner through inhibition of dye efflux. Such an outwardly-directed transport was demonstrated to be temperature-dependent and down-regulated by various metabolic inhibitors, therefore outlining its requirement for energy; it was shown to be Na+- and membrane potential-independent and inhibited by anionic drugs such as indomethacin, indoprofen and rifamycin B. These functional features are closed to those described for multidrug resistance-associated protein 1 (MRP1) that was furthermore demonstrated, in contrast to P-glycoprotein, to be expressed in SDVI cells and to lower CF accumulation in MRP1-overexpressing drug-resistant tumor cells. These data therefore suggest that active membrane transport of organic anions such as CF occurs in epithelial cells like cultured liver biliary SDVI cells through a MRP1-related efflux system.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Animals, Newborn
- Bile Ducts, Intrahepatic/cytology
- Bile Ducts, Intrahepatic/drug effects
- Bile Ducts, Intrahepatic/metabolism
- Biological Transport
- Blotting, Northern
- Dose-Response Relationship, Drug
- Drug Resistance, Multiple
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Fluoresceins/metabolism
- Indomethacin/pharmacology
- Indoprofen/pharmacology
- Multidrug Resistance-Associated Proteins
- Probenecid/pharmacology
- RNA/analysis
- Rats
- Rats, Sprague-Dawley
- Rifamycins/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A Courtois
- INSERM U456 and Laboratoire de Physiologie Faculté de Pharmacie, Université de Rennes, France
| | | | | | | | | |
Collapse
|
176
|
Abstract
Bile acids undergo a unique enterohepatic circulation, which allows them to be efficiently reused with minimal loss. With the cloning of key bile acid transporter genes in the liver and intestine, clinicians now have a detailed understanding of how the different components in the enterohepatic circulation operate. These advances in basic knowledge of this process have directly led to a rapid and highly detailed understanding of rare genetic disorders of bile acid transport, which usually present as pediatric cholestatic disorders. Mutations in specific bile acid or lipid transporters have been identified within specific cholestatic disorders, which allows for genetic tests to be established for specific diseases and provides a unique opportunity to understand how these genes operate together. These same transporters may also prove useful for development of novel drug delivery systems, which can either enhance intestinal absorption of drugs or be used to target delivery to the liver or biliary system. Knowledge gained from these transporters will provide new therapeutic modalities to treat cholestatic disorders caused by common diseases.
Collapse
Affiliation(s)
- R J Bahar
- Department of Pediatrics, University of California-Los Angeles School of Medicine, USA
| | | |
Collapse
|
177
|
Linsdell P, Hanrahan JW. Substrates of multidrug resistance-associated proteins block the cystic fibrosis transmembrane conductance regulator chloride channel. Br J Pharmacol 1999; 126:1471-7. [PMID: 10217542 PMCID: PMC1565925 DOI: 10.1038/sj.bjp.0702458] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/1998] [Revised: 12/29/1998] [Accepted: 01/11/1999] [Indexed: 02/08/2023] Open
Abstract
1. The effects of physiological substrates of multidrug resistance-associated proteins (MRPs) on cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel currents were examined using patch clamp recording from CFTR-transfected mammalian cell lines. 2. Two MRP substrates, taurolithocholate-3-sulphate (TLCS) and beta-estradiol 17-(beta-D-glucuronide) (E217betaG) caused a voltage-dependent block of macroscopic CFTR Cl- currents when applied to the intracellular face of excised membrane patches, with mean apparent dissociation constants (KDs) of 96+/-10 and 563+/-103 microM (at 0 mV) respectively. The unconjugated bile salts taurocholate and cholate were also effective CFTR channel blockers under these conditions, with KDs of 453+/-44 and 3760+/-710 microM (at 0 mV) respectively. 3. Reducing the extracellular Cl- concentration from 154 to 20 mM decreased the KD for block intracellular TLCS to 54+/-1 microM, and also significantly reduced the voltage dependence of block, by suggesting that TLCS blocks Cl- permeation through CFTR by binding within the channel pore. 4. Intracellular TLCS reduced the apparent amplitude of CFTR single channel currents, suggesting that the duration of block is very fast compared to the gating of the channel. 5. The apparent affinity of block by TLCs is comparable to that of other well-known CFTR channel blockers, suggesting that MRP substrates may comprise a novel class of probes of the CFTR channel pore. 6. These results also suggest that the related proteins CFTR and MRP may share a structurally similar anion binding site at the cytoplasmic face of the membrane.
Collapse
Affiliation(s)
- P Linsdell
- Department of Physiology, McGill University, Montréal, Québec, Canada.
| | | |
Collapse
|
178
|
Toh S, Wada M, Uchiumi T, Inokuchi A, Makino Y, Horie Y, Adachi Y, Sakisaka S, Kuwano M. Genomic structure of the canalicular multispecific organic anion-transporter gene (MRP2/cMOAT) and mutations in the ATP-binding-cassette region in Dubin-Johnson syndrome. Am J Hum Genet 1999; 64:739-46. [PMID: 10053008 PMCID: PMC1377791 DOI: 10.1086/302292] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dubin-Johnson syndrome (DJS) is an autosomal recessive disease characterized by conjugated hyperbilirubinemia. Previous studies of the defects in the human canalicular multispecific organic anion transporter gene (MRP2/cMOAT) in patients with DJS have suggested that the gene defects are responsible for DJS. In this study, we determined the exon/intron structure of the human MRP2/cMOAT gene and further characterized mutations in patients with DJS. The human MRP2/cMOAT gene contains 32 exons, and it has a structure that is highly conserved with that of another ATP-binding-cassette gene, that for a multidrug resistance-associated protein. We then identified three mutations, including two novel ones. All mutations identified to date are in the cytoplasmic domain, which includes the two ATP-binding cassettes and the linker region, or adjacent putative transmembrane domain. Our results confirm that MRP2/cMOAT is the gene responsible for DJS. The finding that mutations are concentrated in the first ATP-binding-cassette domain strongly suggests that a disruption of this region is a critical route to loss of function.
Collapse
Affiliation(s)
- S Toh
- Department of Biochemistry, Kyushu University School of Medicine, Maidashi 3-1-1, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Rost D, Kartenbeck J, Keppler D. Changes in the localization of the rat canalicular conjugate export pump Mrp2 in phalloidin-induced cholestasis. Hepatology 1999; 29:814-21. [PMID: 10051484 DOI: 10.1002/hep.510290319] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Administration of phalloidin, one of the toxic peptides of the mushroom Amanita phalloides, leads to rapid and sustained cholestasis in rats. Although attributed to the interaction of phalloidin with microfilaments, the events leading to cholestasis are incompletely understood. The adenosine triphosphate (ATP)-dependent, apical conjugate export pump, termed multidrug resistance protein 2 (Mrp2) or canalicular multispecific organic anion transporter, is the major driving force for bile salt-independent bile flow. We investigated the role of Mrp2 in phalloidin-induced cholestasis. Bile flow decreased to 53% and 31% of control at 15 and 30 minutes after phalloidin (0.5 mg/kg), respectively. Mrp2-mediated [3H]leukotriene excretion into bile during the initial 45 minutes was reduced to 44% of control when [3H]LTC4 was injected 15 minutes after phalloidin treatment. Mrp2 was progressively lost from the hepatocyte canalicular membrane and detected predominantly on intracellular membrane structures together with other canalicular proteins including P-glycoproteins, ecto-ATPase, and dipeptidyl-peptidase IV. By contrast, structures involved in intercellular adhesion (zonula occludens, zonula adhaerens, and desmosomes) as well as intermediate filaments of the cytokeratin type appeared largely unaffected within 30 minutes after phalloidin. In line with the immunofluorescence analysis, immunoblots indicated a loss of Mrp2 and P-glycoproteins from the canalicular membrane and a 3- and 4.6-fold increase of these transport proteins in the microsomal fraction, respectively. Our results indicate that phalloidin induces marked alterations of the hepatocyte canalicular architecture and a loss of Mrp2 together with other proteins from the canalicular membrane. The resulting cholestasis can therefore be explained in part by the loss of export pumps, including Mrp2, from the canalicular membrane.
Collapse
Affiliation(s)
- D Rost
- Division of Tumor Biochemistry, Deutsches Krebsforschungszentrum, Heidelberg,
| | | | | |
Collapse
|
180
|
Abstract
Biliary lipid secretion is an important physiological event; not only for the disposal of cholesterol from the body, but also for the protection of cells lining the biliary tree against bile salts. Insight into the (patho)physiological role of biliary lipid secretion has been recently expanded through the study of a generation of mice with a disruption of the Mdr2 gene, who do not secrete lipids into bile. Mdr2 P-glycoprotein translocates phospholipids across the hepatocanalicular membrane. These animals suffer from progressive liver disease caused by the toxic detergent action of bile salts. Very recently, it has become clear that an analogous inherited human liver disease exists, which is caused by the absence of biliary lipid secretion. Patients with this disease, Progressive Familial Intrahepatic Cholestasis (PFIC) type 3, have a mutation in the MDR3 gene, which is the human homologue of the murine Mdr2 gene.
Collapse
Affiliation(s)
- R P Elferink
- Department of Gastrointestinal and Liver Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
181
|
Kubitz R, Wettstein M, Warskulat U, Häussinger D. Regulation of the multidrug resistance protein 2 in the rat liver by lipopolysaccharide and dexamethasone. Gastroenterology 1999; 116:401-10. [PMID: 9922322 DOI: 10.1016/s0016-5085(99)70138-1] [Citation(s) in RCA: 177] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND & AIMS Endotoxin lipopolysaccharide (LPS) induces cholestasis and down-regulates the multidrug resistance protein 2 (MRP2). This study intends to characterize the short-term effects of LPS on MRP2. METHODS The effects of LPS and dexamethasone on excretion of bromosulphalein (BSP), MRP2 messenger RNA (mRNA) levels, and subcellular MRP2 localization were studied by means of liver perfusion, Northern blots, and confocal microscopy. RESULTS LPS treatment for 3-12 hours decreased biliary BSP excretion (10 micromol/L) by 40%. Hyposmolarity stimulated BSP excretion to control levels 3 hours after LPS injection, but was ineffective after 12 hours or in saline-treated controls. LPS led to a strong decrease of MRP2 mRNA after 12 hours, but not during the first 6 hours. LPS induced the appearance of MRP2 in intracellular vesicles in the immediate vicinity of the canaliculi within 3 hours, and these vesicles were remote from the canaliculi after 6 and 12 hours. The MRP2-containing vesicles did not stain for dipeptidylpeptidase IV (DPPIV). Dexamethasone counteracted the LPS effects on MRP2 mRNA levels, subcellular distribution, and BSP excretion. CONCLUSIONS LPS induces cholestasis due to an early retrieval of MRP2 from the canalicular membrane, whereas down-regulation of MRP2 mRNA is a later event. LPS-induced MRP2 retrieval from the canalicular membrane is not associated with the retrieval of DPPIV, suggestive for selectivity of the process.
Collapse
Affiliation(s)
- R Kubitz
- Medizinische Universitätsklinik, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| | | | | | | |
Collapse
|
182
|
Hayes JH, Soroka CJ, Rios-Velez L, Boyer JL. Hepatic sequestration and modulation of the canalicular transport of the organic cation, daunorubicin, in the Rat. Hepatology 1999; 29:483-93. [PMID: 9918926 DOI: 10.1002/hep.510290216] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
In contrast to organic anions, substrates for the canalicular mdr1a and b are usually organic cations and are often sequestered in high concentrations in intracellular acidic compartments. Because many of these compounds are therapeutic agents, we investigated if their sequestration could be regulated. We used isolated perfused rat liver (IPRL), isolated rat hepatocyte couplets (IRHC), and WIF-B cells to study the cellular localization and biliary excretion of the fluorescent cation, daunorubicin (DNR). Despite rapid (within 15 minutes) and efficient (>90%) cellular uptake in the IPRL, only approximately 10% of the dose administered (0.2-20 micromol) was excreted in bile after 85 minutes. Confocal microscopy revealed fluorescence predominantly in vesicles in the pericanalicular region in IPRL, IRHC, and WIF-B cells. Treatment of these cells with chloroquine and bafilomycin A, agents that disrupt the pH gradient across the vesicular membrane, resulted in a loss of vesicular fluorescence, reversible in the case of bafilomycin A. Taurocholate (TC) and dibutyryl cAMP (DBcAMP), stimulators of transcytotic vesicular transport, increased the biliary recovery of DNR significantly above controls, by 70% and 35%, respectively. The microtubule destabilizer, nocodazole, decreased biliary excretion of DNR. No effect on secretion was noted in TR- mutant rats deficient in mrp2. Coadministration of verapamil, an inhibitor of mdr1, also decreased DNR excretion. While TC and DBcAMP did not affect the fluorescent intensity or pattern of distribution in IRHC, nocodazole resulted in redistribution of DNR to peripheral punctuate structures. These findings suggest that the organic cation, DNR, is largely sequestered in cells such as hepatocytes, yet its excretion can still be modulated.
Collapse
Affiliation(s)
- J H Hayes
- Department of Medicine and Liver Center, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | |
Collapse
|
183
|
Demeule M, Jodoin J, Beaulieu E, Brossard M, Béliveau R. Dexamethasone modulation of multidrug transporters in normal tissues. FEBS Lett 1999; 442:208-14. [PMID: 9929003 DOI: 10.1016/s0014-5793(98)01663-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The expression of P-glycoprotein (P-gp) and canalicular multispecific organic anion transporter (cMOAT or Mrp2) was evaluated by Western blotting analysis of rat tissues isolated following daily administration (1 mg kg(-1) day(-1)) of dexamethasone over 4 days. Dexamethasone rapidly increased P-gp expression more than 4.5- and 2-fold in liver and lung, respectively, while it was decreased 40% in kidney. cMOAT expression was increased 2-fold in liver and kidney following dexamethasone treatment. The levels of both proteins returned to control values by 6 days after the conclusion of dexamethasone administration. These results indicate that dexamethasone can modulate P-gp and cMOAT expression in specific rat tissues and may have significant relevance for patients treated with dexamethasone as a single agent or in combination therapy with other drugs.
Collapse
Affiliation(s)
- M Demeule
- Laboratoire d'Oncologie Moléculaire et Centre de Cancérologie Charles Bruneau, Université du Québec à Montréal, Canada
| | | | | | | | | |
Collapse
|
184
|
Rebbeor JF, Connolly GC, Dumont ME, Ballatori N. ATP-dependent transport of reduced glutathione on YCF1, the yeast orthologue of mammalian multidrug resistance associated proteins. J Biol Chem 1998; 273:33449-54. [PMID: 9837923 DOI: 10.1074/jbc.273.50.33449] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transport systems involved in the export of cellular reduced glutathione (GSH) have not been identified, although recent studies implicate a role for some of the multidrug resistance associated proteins (MRP), including MRP1 and MRP2. The present study examined the hypothesis that the yeast orthologue of MRP, Ycf1p, mediates ATP-dependent GSH transport. [3H]GSH transport was measured in vacuolar membrane vesicles isolated from a control strain of Saccharomyces cerevisiae (DTY165), the isogenic DTY167 strain that lacks a functional Ycf1p, and in DTY167 transformed with a 2-micrometer plasmid vector containing YCF1. GSH transport in control vacuolar membrane vesicles was mediated largely by an ATP-dependent, low affinity pathway (Km = 15 +/- 4 mM). ATP-dependent [3H]GSH transport was cis-inhibited by substrates of the yeast Ycf1p transporter and inhibited by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid, probenecid, and sulfinpyrazone, inhibitors of MRP1 and MRP2, but was minimally affected by membrane potential or pH gradient uncouplers. In contrast, ATP-dependent GSH transport was not seen in vacuolar membrane vesicles isolated from the DTY167 yeast strain without a functional Ycf1p but was restored to near wild-type levels in the DTY167 strain transformed with YCF1 and expressing the vacuolar Ycf1p transporter. On the other hand, expression and functional activity of a bile acid transporter, Bat1p, and of the V-type ATPase were similar in all three yeast strains. These results provide direct evidence for ATP-dependent low affinity transport of GSH by the yeast Ycf1p transporter. Because of the structural and functional homology between Ycf1p and MRP1 and MRP2, these data support the hypothesis that GSH efflux from mammalian cells is mediated by these membrane proteins.
Collapse
Affiliation(s)
- J F Rebbeor
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York 14642, USA.
| | | | | | | |
Collapse
|
185
|
Chung YB, Bae WT, Han K. Metabolism of an anionic fluorescent dye, 1-anilino-8-naphthalene sulfonate (ANS) by rat liver microsomes. Arch Pharm Res 1998; 21:677-82. [PMID: 9868536 DOI: 10.1007/bf02976756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The present study was designed to examine the metabolism of 1-anilino-8-naphthalene sulfonate (ANS), an anionic compound which is transported into liver via "multispecific organic anion transporter", with rat hepatic microsomes. TLC analysis indicated that the fluorescent metabolites were not produced to a measurable extent, which made it possible to assess the ANS metabolism by measuring the fluorescence disappearance. The metabolism of ANS was remarkably inhibited by the presence of SKF-525A as well as by the substitution of O2 by CO gas. ANS metabolism by microsomes also required NADPH as a cofactor. These results indicated that the microsomal monooxygenase system might be mainly responsible for the ANS metabolism. The maximum velocity (Vmax) and Michaelis constant (K(m)) were calculated to be 4.3 +/- 0.2 nmol/min/mg protein and 42.1 +/- 2.0 microM, respectively. Assuming that 1 g of liver contains 32 mg of microsomal protein, the Vmax value was extrapolated to that per g of liver (Vmax'). The intrinsic metabolic clearance (CLint) under linear conditions calculated from this in vitro metabolic study was 3.3 ml/min/g liver, being comparable with that (3.0 ml/min/g liver) calculated by analyzing the in vivo plasma disappearance curve in a previous study. Furthermore, the effects of other organic anions on the metabolism of ANS were examined. Bromophenolblue (BPB) and rose bengal (RB) competitively inhibited the metabolism of ANS, while BSP inhibited it only slightly. The inhibition constant (Ki) of BPB (6 microM) was much smaller than that of RB (200 microM). In conclusion, the microsomal monooxygenase system plays a major role in the metabolism of ANS, and other unmetabolizable organic anions (BPB and RB) compete for this metabolism.
Collapse
Affiliation(s)
- Y B Chung
- College of Pharmacy, Chungbuk National University, Korea
| | | | | |
Collapse
|
186
|
Vos TA, Hooiveld GJ, Koning H, Childs S, Meijer DK, Moshage H, Jansen PL, Müller M. Up-regulation of the multidrug resistance genes, Mrp1 and Mdr1b, and down-regulation of the organic anion transporter, Mrp2, and the bile salt transporter, Spgp, in endotoxemic rat liver. Hepatology 1998; 28:1637-44. [PMID: 9828229 DOI: 10.1002/hep.510280625] [Citation(s) in RCA: 276] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endotoxin-induced cholestasis is mainly caused by an impaired canalicular secretion. Mrp2, the canalicular multispecific organic anion transporter, is strongly down-regulated in this situation, and canalicular bile salt secretion is also reduced. We hypothesized that other adenosine triphosphate-binding cassette (ABC) transporters may compensate for the decreased transport activity to protect the cell from cytokine-induced oxidative damage. Therefore, we examined the expression of ABC-transport proteins in membrane fractions of whole liver and of isolated hepatocytes of endotoxin-treated rats and performed reverse-transcriptase polymerase chain reaction (RT-PCR) on mRNA isolated from these livers. In addition, the localization of these transporters was examined using confocal scanning laser microscopy. By 6 hours after endotoxin administration, we found a clear increase of mrp1 mRNA and protein, whereas mrp2 mRNA and protein were decreased. This was confirmed in isolated hepatocytes. In addition, mdr1b mRNA was strongly increased, whereas mdr1a and mdr2 mRNA did not change significantly. Both the mRNA and protein levels of the sister of P-glycoprotein (spgp), the recently cloned bile salt transporter, decreased. After endotoxin treatment, the normally sharply delineated canalicular staining of mrp2 and spgp had changed to a fuzzy pattern, suggesting localization in a subapical compartment. We conclude that endotoxin-induced cholestasis is caused by decreased mrp2 and spgp levels, as well as an abnormal localization of these proteins. The simultaneous up-regulation of mrp1 and mdr1b may confer resistance to hepatocytes against cytokine-induced metabolic stress.
Collapse
Affiliation(s)
- T A Vos
- Groningen Institute for Drug Studies, University Center for Pharmacy, University Hospital Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Kinoshita S, Suzuki H, Ito K, Kume K, Shimizu T, Sugiyama Y. Transfected rat cMOAT is functionally expressed on the apical membrane in Madin-Darby canine kidney (MDCK) cells. Pharm Res 1998; 15:1851-6. [PMID: 9892468 DOI: 10.1023/a:1011953906065] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE The purpose of the present study is to investigate the expression of canalicular multispecific organic anion transporter (cMOAT) by its cDNA transfection in polarized Madin-Darby canine kidney cells (MDCK). METHODS MDCK cells were transfected with an expression vector (pCXN2) containing the rat cMOAT cDNA with lipofectamine to obtain the stable transfectant under G418. Cells from a single colony whose cMOAT expression was the highest were seeded to form a tight epithelial monolayer on microporous membrane filters. Export of glutathione S-bimane (GS-B) from monolayers was determined after preloading its precursor, monochloro bimane (MCB). RESULTS A comparable amount of GS-B was excreted to the apical and basal compartments in the vector-transfected cells. In contrast, in cMOAT-transfected cells, the amount apically excreted was approximately twice that excreted into the basal compartment. Cyclosporin A (CsA) (30 microM), an inhibitor of cMOAT at higher concentrations, inhibited the preferential apical export of GS-B from cMOAT-transfected cells. CONCLUSIONS Rat cMOAT is functionally expressed on the apical membrane of MDCK cells after transfection.
Collapse
Affiliation(s)
- S Kinoshita
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
188
|
Nies AT, Cantz T, Brom M, Leier I, Keppler D. Expression of the apical conjugate export pump, Mrp2, in the polarized hepatoma cell line, WIF-B. Hepatology 1998; 28:1332-40. [PMID: 9794919 DOI: 10.1002/hep.510280523] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
The polarized rat hepatoma/human fibroblast hybrid cell line, WIF-B, forms apical vacuoles into which cholephilic substances are secreted. We studied expression, localization, and function of the apical conjugate export pump, Mrp2, in WIF-B cells. Mrp2, the apical isoform of the multidrug resistance protein, alternatively termed canalicular Mrp (cMrp) or canalicular multispecific organic anion transporter (cMoat), is a 190-kd membrane glycoprotein mediating adenosine triphosphate (ATP)-dependent transport of glucuronides, glutathione S-conjugates, and other amphiphilic anions across the hepatocyte canalicular membrane into bile. Expression of the rat mrp2 gene in WIF-B cells was shown by reverse-transcription polymerase chain reaction (PCR), followed by sequencing of the amplified 789-bp fragment. Immunoblotting, using antibodies reacting with the amino-terminal or with the carboxyl-terminal sequence of rat Mrp2, detected the 190-kd glycoprotein in WIF-B cell homogenates. Immunofluorescence microscopy localized Mrp2 to the apical membrane domain. Preloading of WIF-B cells with a membrane-permeable ester of the calcium-dependent fluorescent indicator, Fluo-3, was followed by Mrp2-mediated secretion of the amphiphilic anion, Fluo-3, into the apical vacuoles. This transport was potently inhibited by cyclosporin A added to the culture medium. Direct measurements of ATP-dependent transport into Mrp2-containing plasma membrane vesicles in comparison with Mrp2-deficient vesicles established that Fluo-3 is transported by Mrp2 with a Km value of 3.7 micromol/L. Our results indicate that the polarized WIF-B cells express the rat ortholog of the apical conjugate-transporting ATPase, Mrp2. The function of Mrp2 as well as the action of inhibitors can thus be analyzed by use of the fluorescent amphiphilic anion, Fluo-3.
Collapse
Affiliation(s)
- A T Nies
- Division of Tumor Biochemistry, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
189
|
Affiliation(s)
- M Trauner
- Department of Medicine, Karl Franzens University, Graz, Austria
| | | | | |
Collapse
|
190
|
Havelaar AC, de Gast IL, Snijders S, Beerens CE, Mancini GM, Verheijen FW. Characterization of a heavy metal ion transporter in the lysosomal membrane. FEBS Lett 1998; 436:223-7. [PMID: 9781683 DOI: 10.1016/s0014-5793(98)01133-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Lysosomes are thought to play a role in various aspects of heavy metal metabolism. In the present study we demonstrate for the first time the presence of a heavy metal ion transport protein in the lysosomal membrane. Uptake of radioactive silver both in highly purified lysosomal membrane vesicles and in purified intact lysosomes showed the typical kinetics of a carrier-mediated process. This transport was stimulated by ATP hydrolysis, and showed specificity for Ag+, Cu2+, and Cd2+. All biochemical properties of this lysosomal metal ion transporter could classify it as a heavy metal transporting P-type ATPase. Long Evans Cinnamon (LEC) rats, an animal model for the copper transport disorder Wilson disease, showed normal lysosomal silver transport.
Collapse
Affiliation(s)
- A C Havelaar
- Department of Clinical Genetics, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
191
|
Kusuhara H, Han YH, Shimoda M, Kokue E, Suzuki H, Sugiyama Y. Reduced folate derivatives are endogenous substrates for cMOAT in rats. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:G789-96. [PMID: 9756510 DOI: 10.1152/ajpgi.1998.275.4.g789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We examined the role of the canalicular multispecific organic anion transporter (cMOAT) in the biliary excretion of reduced folate derivatives in vivo and in vitro using normal [Sprague-Dawley rats (SDR)] and mutant [Eisai hyperbilirubinemic rats (EHBR)] rats whose cMOAT is hereditarily deficient. In vivo, the biliary excretion of endogenous tetrahydrofolate (H4PteGlu), 5-methyltetrahydrofolate (5-CH3-H4PteGlu), and 5,10-methylenetetrahydrofolate (5, 10-CH2-H4PteGlu) in EHBR was reduced to 8.2%, 1.9%, and 5.5% of those in SDR, respectively, whereas that of 10-formyltetrahydrofolate (10-HCO-H4PteGlu) was detected only in SDR and not in EHBR. Bile drainage caused reduction of endogenous plasma folate concentrations in SDR but not in EHBR. In vitro, significant ATP-dependent uptake of 3H-labeled 5-CH3-H4PteGlu into canalicular membrane vesicles was observed only in SDR. This ATP-dependent uptake was saturable with a Michaelis constant (Km) value of 126 microM, which was comparable with its inhibitor constant (Ki) value of 121 microM for the ATP-dependent uptake of a typical cMOAT substrate, 2,4-dinitrophenyl-S-glutathione (DNP-SG). Vice versa, DNP-SG inhibited the uptake of 5-CH3-H4PteGlu with a Ki of 35 microM, which was similar to its Km value. In addition, H4PteGlu and 5, 10-CH2-H4PteGlu also inhibited the ATP-dependent uptake of DNP-SG. These results indicate that 5-CH3-H4PteGlu and other derivatives are transported via cMOAT. Therefore, reduced folate derivatives are the first endogenous substrates for cMOAT that do not contain glutathione, glucuronide, or sulfate moieties.
Collapse
Affiliation(s)
- H Kusuhara
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113, Japan
| | | | | | | | | | | |
Collapse
|
192
|
Hisaka A, Sugiyama Y. Analysis of nonlinear and nonsteady state hepatic extraction with the dispersion model using the finite difference method. JOURNAL OF PHARMACOKINETICS AND BIOPHARMACEUTICS 1998; 26:495-519. [PMID: 10205768 DOI: 10.1023/a:1023294632129] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A numerical calculation method for dispersion models was developed to analyze nonlinear and nonsteady hepatic elimination of substances. The finite difference method (FDM), a standard numerical calculation technique, was utilized to solve nonlinear partial differential equations of the dispersion model. Using this method, flexible application of the dispersion model becomes possible, because (i) nonlinear kinetics can be incorporated anywhere, (ii) the input function can be altered arbitrarily, and (iii) the number of compartments can be increased as needed. This method was implemented in a multipurpose nonlinear least-squares fitting computer program, Napp (Numeric Analysis Program for Pharmacokinetics). We simulated dilution curves for several nonlinear two-compartment hepatic models in which the saturable process is assumed in transport or metabolism, and investigated whether they could definitely be discriminated from each other. Preliminary analysis of the rat liver perfusion data of a cyclic pentapeptide, BQ-123, was performed by this method to demonstrate its applicability.
Collapse
Affiliation(s)
- A Hisaka
- Development Research Laboratory, Banyu Pharmaceutical Company Limited, Saitama, Japan
| | | |
Collapse
|
193
|
Liu X, Brouwer KL, Gan LS, Brouwer KR, Stieger B, Meier PJ, Audus KL, LeCluyse EL. Partial maintenance of taurocholate uptake by adult rat hepatocytes cultured in a collagen sandwich configuration. Pharm Res 1998; 15:1533-9. [PMID: 9794494 DOI: 10.1023/a:1011994831139] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE This study was designed to characterize taurocholate uptake properties in primary cultures of rat hepatocytes maintained under different matrix conditions. METHODS Hepatocytes isolated from male Wistar rats (230-280 g) were cultured on a simple collagen film, on a substratum of gelled collagen or between two layers of gelled collagen (sandwich configuration). Hepatocyte morphology, taurocholate uptake properties, and expression of the sinusoidal transport protein. Na+/taurocholate-cotransporting polypeptide (Ntcp) were examined in these cultures at day 0 and day 5. RESULTS By day 5, monolayer integrity had deteriorated in simple collagen cultures. In contrast, cell morphology was preserved in hepatocytes maintained in a sandwich configuration. At day 5, taurocholate accumulation at 5 min in hepatocytes cultured on a simple collagen film, on a substratum of gelled collagen, and in a sandwich configuration was approximately 13%, 20% and 35% of day-0 levels, respectively, and occurred predominately by a Na+-dependent mechanism. The initial taurocholate uptake rate vs. concentration (1-200 microM) profile was best described by a combined Michaelis-Menten and first-order function. In all cases, the estimated apparent Km values were comparable for day-0 and day-5 hepatocytes (3241 microM). In contrast, the Vmax values of hepatocytes cultured on a simple collagen film, on gelled collagen and in a sandwich configuration were approximately 5, 6 and 14% of the values at day 0, respectively; values for the first-order rate constant were 5-, 3- and 2-fold lower, respectively. Immunoblot analysis indicated that at day 5 Ntcp expression in hepatocytes cultured in a sandwich configuration was greater than in hepatocytes cultured on a simple collagen film. CONCLUSIONS A collagen sandwich configuration reestablishes normal morphology and partially restores bile acid uptake properties in primary cultures of rat hepatocytes.
Collapse
Affiliation(s)
- X Liu
- Division of Drug Delivery and Disposition, School of Pharmacy, University of North Carolina at Chapel Hill, 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Gerloff T, Meier PJ, Stieger B. Taurocholate induces preferential release of phosphatidylcholine from rat liver canalicular vesicles. LIVER 1998; 18:306-12. [PMID: 9831358 DOI: 10.1111/j.1600-0676.1998.tb00810.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS/BACKGROUND Biliary phospholipid secretion involves predominant segregation of canalicular phosphatidylcholine into bile. We tested the hypothesis that micellar concentrations of the major physiologic bile salt taurocholate can preferentially solubilize phosphatidylcholine from the canalicular rat liver plasma membrane. METHODS Subcellular fractions from rat liver and kidney were isolated with standardized procedures, incubated in vitro with taurocholate or 3-[(3-cholamidopropyl)dimethylammonio]-propane-1-sulphonate (CHAPS) and released phospholipids determined after centrifugation. RESULTS After incubation of canalicular (cLPM) and basolateral (blLPM) rat liver plasma membrane vesicles with 6 and 8 mM taurocholate, the proportion of phosphatidylcholine released was about two-fold higher as compared with its relative contribution to the overall lipid composition of the membranes. Quantitatively, this taurocholate-induced preferential phosphatidylcholine release was about four-fold higher in cLPM (117 nmol) as compared with blLPM (28 nmol). Comparison of membranes from different organs showed that increased sphingomyelin content reduced taurocholate-induced phosphatidylcholine release. Furthermore, phosphatidylcholine release from cLPM did not fit an inverse exponential relationship between membrane sphingomyelin content and phosphatidylcholine release from different starting material, indicating that cLPM is especially prone to taurocholate-induced phosphatidylcholine release. In contrast, in rat liver microsomes and kidney brush border membranes, taurocholate released phospholipids in proportion of their membrane contents, indicating an unspecific membrane solubilizing effect only. Similarly, CHAPS had an unselective lipid solubilizing effects in cLPM and blLPM. CONCLUSION These results support the concept that the very last step of canalicular phospholipid secretion is mediated in vivo by bile salt-induced vesiculation of phosphatidylcholine-enriched microdomains from the outer leaflet of cLPM.
Collapse
Affiliation(s)
- T Gerloff
- Department of Internal Medicine, University Hospital, Zürich, Switzerland
| | | | | |
Collapse
|
195
|
Iyanagi T, Emi Y, Ikushiro S. Biochemical and molecular aspects of genetic disorders of bilirubin metabolism. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1407:173-84. [PMID: 9748558 DOI: 10.1016/s0925-4439(98)00044-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Bilirubin, the oxidative product of heme in mammals, is excreted into the bile after its esterification with glucuronic acid to polar mono- and diconjugated derivatives. The accumulation of unconjugated and conjugated bilirubin in the serum is caused by several types of hereditary disorder. The Crigler-Najjar syndrome is caused by a defect in the gene which encodes bilirubin UDP-glucuronosyltransferase (UGT), whereas the Dubin-Johnson syndrome is characterized by a defect in the gene which encodes the canalicular bilirubin conjugate export pump of hepatocytes. Animal models such as the unconjugated hyperbilirubinemic Gunn rat, the conjugated hyperbilirubinemic GY/TR-, and the Eisai hyperbilirubinemic rat, have contributed to the understanding of the molecular basis of hyperbilirubinemia in humans. Elucidation of both the structure of the UGT1 gene complex, and the Mrp2 (cMoat) gene which encodes the canalicular conjugate export pump, has led to a greater understanding of the genetic basis of hyperbilirubinemia.
Collapse
Affiliation(s)
- T Iyanagi
- Department of Life Science, Himeji Institute of Technology, Hyogo, Japan.
| | | | | |
Collapse
|
196
|
Breidert T, Spitzenberger F, Gründemann D, Schömig E. Catecholamine transport by the organic cation transporter type 1 (OCT1). Br J Pharmacol 1998; 125:218-24. [PMID: 9776363 PMCID: PMC1565608 DOI: 10.1038/sj.bjp.0702065] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Liver and kidney extract adrenaline and noradrenaline from the circulation by a mechanism which does not seem to be one of the classical catecholamine transporters. The hypothesis that OCT1 is involved the organic cation transporter type 1 which exists in rat kidney and liver-was tested. 2. Based on human embryonic kidney cells (293), we constructed a cell line which stably expresses OCT1r (293OCT1r cells). Transfection with OCT1 resulted in a transport activity not only for prototypical known substrates of OCT1 such as 3H-1-methyl-4-phenylpyridinium and 14C-tetraethylammonium but also for the catecholamines 3H-adrenaline, 3H-noradrenaline (3H-NA) and 3H-dopamine (3H-DA), the indoleamine 3H-5-hydroxytryptamine (3H-5HT) as well as the indirect sympathomimetic 14C-tyramine. 3. For 3H-DA, 3H-5HT and 3H-NA, at non-saturating concentrations, the rate constants for inwardly directed substrate flux (kin) were 6.9+/-0.8, 3.1+/-0.2, and 1.2+/-0.1 microl min(-1) mg protein(-1). In wild type cells (293WT) the corresponding kin's were considerably lower, being 0.94+/-0.40, 0.47+/-0.08 and 0.23+/-0.05 microl min(-1) mg protein ' (n=12). The indirectly determined half-saturating concentrations of DA, 5HT, and NA were 1.1 (95% c.i.: 0.8, 1.4), 0.65 (0.49, 0.86), and 2.8 (2.1, 3.7) mmol l(-1) (n=3). 4. Specific 3H-DA uptake in 293OCT1r cells was resistant to cocaine (1 micromoll(-1)), 3H-5HT uptake was resistant to citalopram (300 nmol l(-1)) and 3H-NA uptake was resistant to desipramine (100 nmoll(-1)), corticosterone (1 micromol l(-1)), and reserpine (10 nmoll(-1)) which rules out the involvement of classical transporters for biogenic amines. 5. The findings demonstrate that OCTI efficiently transports catecholamines and other biogenic amines and support the hypothesis that OCT1 is responsible for hepatic and renal inactivation of circulating catecholamines.
Collapse
Affiliation(s)
- T Breidert
- Department of Pharmacology, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
197
|
Kusuhara H, Suzuki H, Sugiyama Y. The role of P-glycoprotein and canalicular multispecific organic anion transporter in the hepatobiliary excretion of drugs. J Pharm Sci 1998; 87:1025-40. [PMID: 9724551 DOI: 10.1021/js970100b] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- H Kusuhara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113, Japan
| | | | | |
Collapse
|
198
|
Kiuchi Y, Suzuki H, Hirohashi T, Tyson CA, Sugiyama Y. cDNA cloning and inducible expression of human multidrug resistance associated protein 3 (MRP3). FEBS Lett 1998; 433:149-52. [PMID: 9738950 DOI: 10.1016/s0014-5793(98)00899-0] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Previously, we cloned rat MRP3 as a candidate for an inducible transporter for the biliary excretion of organic anions [Hirohashi et al. (1998) Mol. Pharmacol. 53, 1068-10751. In the present study, we cloned human MRP3 (1527 amino acids) from Caco-2 cells. Human MRP3 is predominantly expressed in liver, small intestine and colon; hepatic expression of MRP3 was observed in humans but not in normal rats. In HepG2 cells, the expression of MRP3 was induced by phenobarbital. These results suggest that MRP3 may act as an inducible transporter in the biliary and intestinal excretion of organic anions.
Collapse
Affiliation(s)
- Y Kiuchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Japan
| | | | | | | | | |
Collapse
|
199
|
Meyer-Wentrup F, Karbach U, Gorboulev V, Arndt P, Koepsell H. Membrane localization of the electrogenic cation transporter rOCT1 in rat liver. Biochem Biophys Res Commun 1998; 248:673-8. [PMID: 9703985 DOI: 10.1006/bbrc.1998.9034] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The polyspecific cation transporter rOCT1 in the rat was the first identified member of a new protein family with 12 presumed membrane-spanning alpha-helices and two large hydrophilic loops. Previous studies showed that rOCT1 is mainly expressed in liver and mediates electrogenic uptake of small organic cations into cells. Antibodies against partial sequences of rOCT1 were raised and their specificity was verified. Immunohistochemistry with rat liver and Western blots with isolated membranes showed that rOCT1 is localized within sinusoidal membranes of hepatocytes. Antibody reactions were also performed with intact and permeabilized human embryonic kidney cells that were stably transfected with rOCT1. They showed that the large hydrophilic loop after the first alpha-helix of rOCT1 is located extracellularly, while the C-terminus is located intracellularly. Translational regulation is suggested since the message of rOCT1 was distributed throughout the liver lobuli, whereas rOCT1 protein was observed only in hepatocytes surrounding the central veins.
Collapse
Affiliation(s)
- F Meyer-Wentrup
- Anatomisches Institut, Bayerischen Julius-Maximilians-Universität, Würzburg, Germany
| | | | | | | | | |
Collapse
|
200
|
Morán D, De Buitrago JM, Fernández E, Galán AI, Muñoz ME, Jiménez R. Inhibition of biliary glutathione secretion by cyclosporine A in the rat: possible mechanisms and role in the cholestasis induced by the drug. J Hepatol 1998; 29:68-77. [PMID: 9696494 DOI: 10.1016/s0168-8278(98)80180-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS Biliary glutathione appears to be a major osmotic factor in the generation of bile acid-independent bile flow. This study was designed to investigate its importance in cyclosporine A-induced cholestasis in both acute and short-term-treated rats. METHODS Adult male Wistar rats were treated as follows: (i) with a single i.v. dose of cyclosporine or its vehicle (acute assays); (ii) with cyclosporine, its vehicle or physiological saline, i.p., for 7 days once per day (short-term treatment assays). Bile flow and biliary glutathione levels were determined under anesthesia both before and after intrabiliary hydrolysis of the tripeptide had been inhibited. RESULTS Acute cyclosporine administration, at a dose of 20 mg/kg, brought about an abrupt and marked fall in bile flow and bile acid secretion simultaneously with a rapid decrease in the biliary concentration and secretion rates of total, reduced and oxidized glutathione. When the rats were treated with cyclosporine A for 1 week, at a dose of 10 mg/kg per day, similar cholestatic and inhibitory effects on the biliary secretion of glutathione were noted both before and after the intrabiliary catabolism of the tripeptide had been inhibited with acivicin; in addition, the hepatic content of glutathione was also reduced. The cholestatic effect of the drug was associated with reductions in the four bile flow fractions evaluated: bile acid- and glutathione-dependent bile flow and bile acid- and glutathione-independent bile flow. CONCLUSIONS These findings indicate that cyclosporine-induced cholestasis in the rat is due not only to alterations in the hepatobiliary transport of bile acids but also to an impairment of bile formation dependent on the biliary secretion of glutathione, possibly through inhibition of the canalicular transport of the tripeptide.
Collapse
Affiliation(s)
- D Morán
- Department of Physiology and Pharmacology, Universidad de Salamanca, Spain
| | | | | | | | | | | |
Collapse
|