Literature
Home医源资料库在线期刊分子药理学杂志2006年第68卷第1期

Analysis of the In Vivo Functions of Mrp3

来源:分子药理学杂志
摘要:ToelucidatetheroleofMrp3intheseprocesses,theMrp3gene(Abcc3)wasdisruptedbyhomologousrecombination。Mrp3eC/eCmicedidnotexhibitenhancedlethalitytoetoposidephosphate,althoughananalysisoftransfectedhumanembryonickidney293cellsindicatedthatthepotencyofmurineM......

点击显示 收起

    Medical Science (M.G.B., I.S., Z.-S.C., A.K.-S., G.D.K.) and Basic Science Divisions (A.L.), Fox Chase Cancer Center, Philadelphia, Pennsylvania
    Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (P.A.D.)
    Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas (L.J.B.)
    British Columbia Cancer Agency, Vancouver, British Columbia, Canada (R.W., V.L.)
    National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland (A.G., H.W.)

    Abstract

    Multidrug resistance protein 3 (MRP3) is an ATP-binding cassette transporter that is able to confer resistance to anticancer agents such as etoposide and to transport lipophilic anions such as bile acids and glucuronides. These capabilities, along with the induction of the MRP3 protein on hepatocyte sinusoidal membranes in cholestasis and the expression of MRP3 in enterocytes, have led to the hypotheses that MRP3 may function in the body to protect normal tissues from etoposide, to protect cholestatic hepatocytes from endobiotics, and to facilitate bile-acid reclamation from the gut. To elucidate the role of Mrp3 in these processes, the Mrp3 gene (Abcc3) was disrupted by homologous recombination. Homozygous null animals were healthy and physically indistinguishable from wild-type mice. Mrp3eC/eC mice did not exhibit enhanced lethality to etoposide phosphate, although an analysis of transfected human embryonic kidney 293 cells indicated that the potency of murine Mrp3 toward etoposide (2.0- to 2.5-fold) is comparable with that of human MRP3. After induction of cholestasis by bile duct ligation, Mrp3eC/eC mice had 1.5-fold higher levels of liver bile acids and 3.1-fold lower levels of serum bilirubin glucuronide compared with ligated wild-type mice, whereas significant differences were not observed between the respective sham-operated mice. Bile acid excretion, pool size, and fractional turnover rates were similar in Mrp3eC/eC and wild-type mice. We conclude that Mrp3 functions as an alternative route for the export of bile acids and glucuronides from cholestatic hepatocytes, that the pump does not play a major role in the enterohepatic circulation of bile acids and that the lack of chemosensitivity is probably attributable to functional redundancy with other pumps.

    MRP3 belongs to a group of nine related ATP-binding cassette transporters that constitute the MRP family. Members of this family function as efflux pumps for lipophilic anions and hydrophobic compounds (Kruh and Belinsky, 2003). The in vivo functions of the first two members of this family have been investigated in gene-disrupted mice in the case of Mrp1 and hereditarily deficient rats (EHBR and TReC rats) and humans (Dubin-Johnson syndrome) in the case of Mrp2. These studies indicate that Mrp1 functions as an in vivo resistance factor for anticancer agents and in inflammatory responses mediated by leukotriene C4 (Lorico et al., 1997; Wijnholds et al., 1997; Johnson et al., 2001; Schultz et al., 2001) and that Mrp2 is involved in the hepatobiliary elimination of endogenous compounds, such as bilirubin glucuronide, and in the hepatobiliary and renal elimination of xenobiotics (Gerk and Vore, 2002). In contrast to MRP1 and MRP2, the in vivo functions of MRP3 have not been established. However, studies in our and other laboratories on the pump's substrate selectivity, resistance capabilities, and tissue-expression pattern have allowed speculation as to its functions in the body. In cellular models, ectopic expression of human MRP3 is able to confer resistance to etoposide and methotrexate, results which are consistent with the hypothesis that the pump is an in vivo resistance factor that protects normal tissues from chemotherapeutic agents (Kool et al., 1999; Zeng et al., 1999, 2001). The substrate selectivity of human and rat Mrp3 is similar to that of MRP1 and MRP2 with respect to the ability to transport glucuronate and glutathione conjugates, but in contrast to the latter two pumps, monoanionic bile acids are also good substrates of MRP3 (Hirohashi et al., 1999, 2000; Zeng et al., 2000). MRP3 assumes basolateral subcellular localization in polarized cells and is expressed in a variety of tissues including kidney, gut, and pancreas (Belinsky et al., 1998; Rost et al., 2002; Scheffer et al., 2002). It is noteworthy that its expression is dramatically increased at the hepatocyte sinusoidal membrane in cholestatic conditions (Donner and Keppler, 2001; Soroka et al., 2001). The ability of MRP3 to transport bile acids, glucuronides, and other compounds that are normally transported across the canalicular membrane by MRP2 and the bile salt export pump in combination with the induction of MRP3 in cholestasis have led to the hypothesis that MRP3 protects hepatocytes by functioning as an alternate route of elimination when the canalicular route of detoxification is impaired. This notion is also supported by studies showing that Mrp3 is induced by bile acids and that Mrp3 expression is linked to signal transduction and transcriptional pathways that govern bile acid homeostatic mechanisms (Inokuchi et al., 2001; Bohan et al., 2003; Zollner et al., 2003). Although mice deficient in some of these transcriptional pathways are more susceptible to liver damage when made cholestatic, the extent to which attenuation of Mrp3 induction may contribute to this process, as opposed to impairments in the pleiotropic mechanisms of bile acid homeostasis that are regulated by these transcriptional pathways, is unknown (Bohan et al., 2003; Zhang et al., 2004). MRP3 has also been implicated in the enterohepatic circulation of bile acids. Bile acids are transported from the intestinal lumen across the enterocyte brush-border membrane by the well characterized ileal apical bile acid transporter (ASBT). However, the mechanisms responsible for bile acid transport across the enterocyte basolateral membrane have not been fully identified. By virtue of its basolateral membrane expression in enterocytes and its bile acid transport properties, it has been proposed that MRP3 functions as the intestinal basolateral bile acid transporter that moves bile acids from the enterocyte into the portal circulation.

    Here, Mrp3eC/eC mice were generated and analyzed to investigate the in vivo functions of Mrp3. Mrp3eC/eC mice did not exhibit increased sensitivity to etoposide phosphate, although transfection experiments showed that expression of the murine protein in HEK293 cells is capable of conferring resistance to this agent. Although Mrp3 null mice are healthy and have normal bile flow, these animals accumulate increased levels of hepatic bile acids and have reduced serum levels of bilirubin glucuronide after bile duct ligation. Measurements of overall bile acid metabolism, including fecal bile acid excretion, bile acid pool size, and bile acid fractional turnover rate were similar in Mrp3eC/eC and wild-type mice. From these findings, we concluded that Mrp3 functions as an alternative route for the export of bile acids and glucuronides in cholestatic hepatocytes, that the pump does not play a major role in the enterohepatic circulation of bile acids, and that the lack of chemosensitivity in Mrp3eC/eC mice may be attributable to functional redundancy with other efflux pumps.

    Materials and Methods

    Targeted Disruption of the Mrp3 Gene and Generation of Mrp3eC/eC Mice. A 1-kb fragment containing the 5' end of the Mrp3 coding sequence was used to screen a mouse strain 129-derived  phage genomic library, and a 9-kb Mrp3 clone was isolated. The Mrp3 genomic clone was sequenced using an ABI 377 DNA sequencer (Applied Biosystems, Foster City, CA) and encompassed exons 2 to 11 of the Mrp3 gene, corresponding to nucleotides 100 to 1482 of the coding sequence. The left and right arms of a targeting vector were generated by polymerase chain reaction and were inserted, respectively, to the 5' and 3' of the pgk-neo cassette in the PNT plasmid. The vector was designed to delete exons 6 to 8, encoding nucleotides 613 to 996 of the coding sequence, and to introduce a frame-shift into the transcribed RNA sequence. The nucleotide sequence of the cloned arms was confirmed, and the resulting 12-kb vector was digested with NotI. The linearized DNA was electroporated into strain 129-derived R1 embryonic stem cells. Individual colonies isolated after positive/negative selection with G418 and gancyclovir were screened by Southern blot analysis using 5' and 3' probes and genomic DNA digested with XhoI/XmnI. In addition, the absence of randomly integrated vector sequences was confirmed by Southern blot analysis using a neo probe. Two correctly targeted ES cell lines were injected into C57BL/6J blastocysts, and the blastocysts were implanted in pseudopregnant females. Male chimeric progeny were crossed with female C57BL/6J (in-houseeCbred) mice. Germ-line transmission of the targeted allele was confirmed by Southern blot analysis, and subsequent genotyping was accomplished by PCR analysis of tail DNA. The latter reaction was carried out in a single tube using three primers: 5'-gttctgtgccctcatcctgtcc-3', 5'-gggaggg-ggcaagtcaggcc-3', and 5'-aattgacctgcaggggccctcg-3'. The former two primers generate a 790-bp wild-type product, and the latter two generate a 330-bp product from the targeted allele. The Mrp3 null allele was subsequently backcrossed for eight generations into the C57BL/6J background. As indicated below, Mrp3 null mice in the mixed C57BL/6J x 129 background and the C57BL/6J background were used for these studies.

    Isolation of Mrp3 cDNA, Expression Vector Construction, and Transfection. A PCR product corresponding to nucleotides 600 to 1440 of the human MRP3 coding sequence (AF104943 ) was used to screen a bacteriophage library prepared from mouse testis, and a clone encompassing the entire coding region was isolated. The cDNA was sequenced, and the resulting sequences were assembled using the Sequencher program (Gene Codes Corporation, Ann Arbor, MI). The Mrp3 cDNA sequence has been deposited in GenBank (accession no. AY841885 ). A 4.8-kb fragment encompassing the 4.6-kb coding sequence was cloned into the PEAK10 vector (Edge Biosystems, Gaithersburg, MD) to create PEAK10-Mrp3. Human embryonic kidney cells (HEK293/EBNA) were electroporated with 10 e of either PEAK10-Mrp3 or the parental plasmid, and individual puromycin-resistant colonies were isolated and expanded for analysis of Mrp3 protein expression. Cells were grown in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum.

    Generation of Mrp3 Polyclonal Antibody and Immunoblot Analysis. A cDNA fragment encoding amino acids 858 to 945 of the linker region of Mrp3 was inserted downstream of the glutathione S-transferase coding sequence in the vector pGEX2T, and the induced fusion protein was purified using glutathione Sepharose beads (Amersham Biosciences Inc., Piscataway, NJ). Rabbits were immunized with the recombinant protein, and the immune sera were used for immunoblot analysis. Total cellular lysates prepared from cultured cells, and membrane fractions prepared from liver were subjected to SDS-polyacrylamide gel electrophoresis and were electrotransferred to nitrocellulose filters. Mrp3 antibody was used at a 1:500 dilution, and horseradish peroxidase-conjugated secondary antibody (PerkinElmer Life and Analytical Sciences, Boston, MA) was used at 1:2500. Mrp2 monoclonal antibody M2III-5 (kindly provided by George Scheffer, Free University, Amsterdam, The Netherlands) and a previously described polyclonal antibody against the bile salt export pump (Wang et al., 2001) were used at 1:1000 dilutions. Affinity-purified Mrp4 antisera were raised against amino acids NVDPRTDELIQQKIREK conjugated to keyhole limpet hemocyanin (Invitrogen, Carlsbad, CA) and were used at a dilution of 1:500. Antibody to -actin (Sigma-Aldrich, St. Louis, MO) was used at a dilution of 1:1000. Immunoblots were developed using the enhanced chemiluminescence method (Amersham).

    Blood Chemistries, Hematology, and Histopathology. Animals (mixed 129 x C57BL/6J background from the F3 and F4 generation) were maintained in the Fox Chase laboratory animal facility and were housed in a temperature- and humidity-controlled environment under 12-h light/dark cycles. Mice were fed a standard rodent diet (Lab Diet 5013; PMI Nutrition, Brentwood, MO) and had free access to water. The Fox Chase Institutional Animal Care and Use Committee approved the protocol. Peripheral blood was obtained by orbital bleeding of anesthetized mice. Blood chemistry and hematology parameters were determined at Antech Diagnostics (Farmingdale, NY). For histological analysis, tissues were fixed in 10% phosphate-buffered formalin, embedded in paraffin, sectioned, and stained with hematoxylin/eosin.

    In Vivo Etoposide Toxicity. Groups of male mice (mixed 129 x C57BL/6J background) aged 8 to 12 weeks were treated with single intraperitoneal injections of etoposide phosphate (Bristol-Myers Squibb Co., Princeton, NJ). Mice were observed daily for a period of 4 weeks. For analysis of white blood cell counts, groups consisting of five male mice were treated with a single intraperitoneal injection (150 mg/kg body weight), and blood samples (20 e) were taken daily by orbital bleeding for a period of 6 days. White blood cell counts were analyzed using a Coulter Z1 Series Particle Counter (Beckman Coulter, Miami, FL).

    In Vitro Cytotoxicity Assays. Drug sensitivity was analyzed using the CellTiter 96 Cell Proliferation Assay (Promega, Madison WI). Cells seeded overnight in triplicate at 3000 cells/well were treated with drugs at various concentrations, and the proliferation assays were performed after 72 h of drug exposure. The nonparametric two-tailed Wilcoxon test was used to make inferences about the significance of the data.

    Bile Duct Ligation and Analysis of Liver and Serum Bile Acids. Mice (mixed 129 x C57BL/6J background) were anesthetized using vaporized isofluorane applied through a nose cone, and surgery was performed aseptically, using steam-sterilized instruments within a disinfected surgical area. The abdomen was shaved and disinfected with an iodine-based surgical scrub and was covered with sterile drapes. A 2.0-cm transverse incision was made through the skin of the ventral abdomen at a point starting just below the sternum. A similar incision was made through the body wall. The liver was displaced upward to expose the bile duct, which was tied off using 6-0 silk, and the body wall was closed with interrupted sutures of 6-0 Vicryl. The skin was closed with wound clips, and mice were placed on a warming tray until they recovered from the anesthesia. After 3 days, peripheral blood was sampled from fasting (4 h) animals, and livers were removed from animals that were sacrificed. Livers were washed in saline, flash-frozen in liquid nitrogen, and stored at eC80°C. Bile acids from preweighed, pulverized liver samples were extracted overnight in tert-butanol/water (1:1), and liver and serum bile acids were determined using a colorimetric assay (Sigma Diagnostics, St. Louis MO). Serum bilirubin was determined at Antech Diagnostics.

    Kruskall-Wallis tests, a nonparametric analog of the analysis of variance, were used to compare the distributions of measurements across the four mouse type-treatment combinations. These tests were conducted at a 5% significance level. Next, two-sample Wilcoxon tests were used to make comparisons between all possible pairs of mouse type-treatment combinations. To account for multiple comparisons, Bonferroni adjustment was used to control the experiment-wise type I error. In particular, each individual null hypothesis was rejected if the significance level was less than  = (0.05/6) = 0.0083. These analyses were performed separately for serum-conjugated bilirubin, serum bile acid, and liver bile acid measurements.

    Analysis of Bile Flow. Bile duct cannulation of mice (mixed 129 x C57BL/6J background) and collection of bile were performed as described previously (Wang et al., 2001). The gallbladder was cannulated after ligation of the common bile duct, and bile was collected at 5-min intervals. At 10 min, taurocholate (100 e/kg body weight) was injected as a bolus into the jugular vein. Bile was then collected at 2-min intervals for 10 min and then at 10-min intervals for 20 min.

    Analysis of Fecal Bile Acid Excretion, Bile Acid Pool Size, and Composition. Wild-type and Mrp3eC/eC male mice (3eC4 months; C57BL/6J background) were individually housed in wire-bottom cages, and stools were collected for 3 days. The stools were extracted as described previously (Turley et al., 1996) and were used to determine the total bile acid content by an enzymatic method (Mashige et al., 1981). Pool size was determined as the bile acid content of the small intestine, liver, and gallbladder. These tissues were removed and extracted in ethanol as described previously (Schwarz et al., 1998). The extract was filtered, and bile acid composition was determined using high-performance liquid chromatography as described previously (Torchia et al., 2001). Individual bile acid species were measured using an evaporative light-scatter detector (Alltech ELSD 800; Alltech-Applied Science Labs, State College, PA). Bile acids were identified and quantified by comparison to known amounts of authentic standards purchased from Steraloids (Newport, RI).

    Analysis of Asbt, Ost, Ost, and Ibabp Expression in Intestine. RNA was prepared from intestines taken from wild-type and Mrp3eC/eC male mice aged 3 to 4 months (C57BL/6J background). cDNA synthesis was initiated from 1 e of RNA using random hexamer primers and Omniscript transcription reagents (QIAGEN, Valencia, CA). For each real-time PCR reaction, cDNA synthesized from 25 ng of RNA was mixed with 2x SYBR Green PCR Master Mix (Applied Biosystems) containing 500 nM concentrations of specific primers. The PCR reactions were carried out in triplicate, and samples were analyzed on an ABI 7900 sequence detection system. The oligonucleotide primer sequences were the following: Asbt, 5'-tgggtttcttcctggctagact-3' and 5'-tgttctgcattccagtttccaa-3'; Ost, 5'-tacaagaacaccctttgccc-3' and 5'-cgaggaatccagagaccaaa-3'; Ost, 5'-gtattttcgtgcagaagatgcg-3' and 5'-tttctgtttgccaggatgctc-3'; Ibabp, 5'-caaggctaccgtgaagatgga-3' and 5'-cccacgacctccgaagtct-3'; and GAPDH, 5'-tgtgtccgtcgtggatctga-3' and 5'-cctgcttcaccaccttcttgat-3'.

    Results

    Inactivation of the Mrp3 Gene in Mice. A targeting construct designed to delete a 2.1-kb fragment encoding exons 6 to 8 of the Mrp3 gene was generated (Fig. 1A). The intended deletion removes three exons encoding amino acids 205 to 332 of the Mrp3 protein and introduces a frame shift in the reading frame should splicing take place between the exons that immediately border the neo marker. The construct was electroporated into R1 embryonic stem cells, and G418/gancyclovir-resistant colonies were isolated. Southern blot analysis of DNA preparations from two ES clones in which proper targeting took place (Fig. 1B) revealed the predicted 5.0- (left) and 5.7-kb (right) bands for the disrupted allele, in addition to the 10.7-kb band for the wild-type allele (left and right). Chimeric mice were generated, and germ-line transmission of the disrupted allele was achieved in animals derived from each cell line. Homozygous null mice were identified in litters of backcrossed heterozygote animals (Fig. 1C). Inactivation of the gene was confirmed by immunoblot analysis using polyclonal antisera directed toward Mrp3 protein. Immunoreactive protein of the expected molecular weight was readily detected in crude membranes prepared from liver tissue of wild-type mice, whereas the Mrp3-specific band was absent in Mrp3eC/eC mice (Fig. 1D). Haploinsufficiency was apparent in heterozygote mice in which the level of Mrp3 protein in liver was 50% of that in wild-type mice (Fig. 1D, lane 4).

    Heterozygote crosses yielded a distribution of genotypes approximating the expected Mendelian ratios (24.8% wild type, 49.9% heterozygote, 25.4% null; n = 351). Mrp3eC/eC mice were grossly indistinguishable from their wild-type counterparts, had normal viability, and produced litters that were similar in size to those of wild-type mice. The average weight at weaning of male and female Mrp3eC/eC mice was not significantly different from wild-type mice. Histopathological analysis of tissues taken from Mrp3eC/eC mice at 6 weeks and 6 months of age did not reveal obvious abnormalities (data not shown). Serum chemical and hematological parameters measured in 10-week-old (Table 1) and 9-month-old mice (data not shown) did not differ significantly between wild-type and Mrp3eC/eC mice.

    Blood and serum were analyzed for the indicated parameters. Groups consisted of three to five 10-week-old mice.

    Sensitivity of Mrp3-Deficient Mice to Etoposide. The sensitivity of Mrp3eC/eC mice toward anticancer agents was analyzed to ascertain the contribution of the protein to protecting normal tissues. Etoposide was selected as the chemotherapeutic agent in these experiments because human MRP3 is capable of conferring resistance to this compound (Kool et al., 1999; Zeng et al., 1999). Etoposide phosphate, a water-soluble ester of etoposide that is converted to etoposide in plasma, was administered via a single intraperitoneal injection, and the sensitivities of wild-type and Mrp3eC/eC mice were compared. Preliminary experiments indicated that the LD50 values of wild-type and Mrp3eC/eC mice were similar and fell between 125 and 250 mg/kg (data not shown). When this dosage range was examined in detail, significant differences in the chemosensitivity of wild-type and Mrp3eC/eC mice were not observed (Table 2). The estimated LD50 for both groups of mice was 193 mg/kg body weight.

    Groups of male mice (aged 8eC12 weeks) were treated with varying amounts of etoposide phosphate administered as a single intraperitoneal injection. Dose values shown are milligrams of etoposide phosphate per kilogram of body weight. The values represent the number of surviving animals per group 4 weeks after treatment.

    Damage to hematopoietic tissue is a result of the toxicity of etoposide. Therefore, white blood cell counts were analyzed in the two groups of treated mice. In accordance with the results described above, the effects of this agent on white blood cell counts of wild-type and Mrp3eC/eC mice were indistinguishable with respect to the time and depth of white blood cell count depression and the time until recovery (Fig. 2).

    Analysis of the in Vitro Drug Resistance Activity of Mrp3. Although the drug-resistance activity of human MRP3 has been determined, the activity of the rodent pump has not been reported. To confirm that the results of the chemosensitivity experiments were not attributable to species-specific differences in resistance properties, the Mrp3 cDNA was isolated [Mrp3 protein was 81% identical with human MRP3 (Belinsky et al., 1998) and 92% identical with rat Mrp3 (NM080581)] and expressed in HEK293 cells. HEK-Mrp3-1 and HEK-Mrp3-2, two Mrp3-transfectants (Fig. 1D, lane 2; data not shown), exhibited 2.5- and 2.0-fold resistance toward etoposide, respectively, compared with parental vector-transfected cells (p < 0.05; data not shown). This activity was comparable with that of human MRP3 expressed in the same cellular background, for which we previously reported 3- to 4-fold resistance to this agent (Zeng et al., 1999).

    Analysis of Liver Function in Bile Duct-Ligated Mrp3eC/eC Mice. To determine whether Mrp3 is able to protect cholestatic liver by functioning as a basolateral export system for bile acids and other conjugates, the effects of bile duct ligation in wild-type and Mrp3eC/eC mice were analyzed. Bile acids and glucuronides, including bilirubin glucuronide, are established substrates of MRP3 (Hirohashi et al., 1999, 2000; Zeng et al., 2000; Lee et al., 2004). Therefore, levels of serum bilirubin glucuronide, a compound that is formed in the liver by glucuronidation, and of liver and serum bile acids were analyzed. The Mrp3 null mice exhibited abnormalities in each of these parameters that were consistent with the proposed hepatic basolateral export function.

    Cholestasis was achieved after 3 days of bile duct ligation, as indicated by marked elevations in liver bile acids in both bile duct-ligated wild-type and Mrp3 null mice by comparison with their respective sham-ligated controls (Fig. 3A). However, under these conditions, liver bile acid levels in the bile duct-ligated Mrp3eC/eC mice were elevated 1.5-fold in comparison with the bile duct-ligated wild-type mice (p = 0.0074). Under non-bile duct ligation (i.e., sham surgery) conditions, liver bile acids were also elevated in Mrp3eC/eC mice compared with wild-type mice (Fig. 3A), and serum bile acids were depressed in bile duct-ligated Mrp3eC/eC mice compared with bile duct-ligated control mice (Fig. 3B), but these differences did not reach statistical significance. Serum levels of conjugated bilirubin, which were too low to be measured under non-bile duct ligation conditions, were significantly depressed (67%; p = 0.0005) in bile duct-ligated Mrp3eC/eC mice compared with bile duct-ligated wild-type animals (Fig. 3C). Comparable levels of necrosis, inflammation, and portal expansion were observed in the livers of the bile duct-ligated Mrp3eC/eC and corresponding wild-type mice (data not shown).

    The effects of bile duct ligation on protein expression levels of Mrp3 (wild-type mice), as well as on the expression of other ATP-binding cassette transporters whose activities could impact liver parameters, were analyzed (Fig. 4). These pumps included Mrp2 and the bile salt export pump (Bsep), two canalicular transporters that efflux glucuronides and bile acids, respectively, and Mrp4, a sinusoidal pump that transports both of these types of compounds (Gerloff et al., 1998; Cui et al., 1999; Chen et al., 2001; Rius et al., 2003). Mrp3 levels were comparable in the sham and bile duct-ligated wild-type mice. Mrp2 levels were slightly depressed in both wild-type and Mrp3eC/eC bile duct-ligated mice compared with the respective sham-operated control mice, as expected (Paulusma et al., 2000), whereas expression levels of the bile salt export pump and Mrp4 were similar in the four groups of animals. No statistically significant differences were observed when protein levels were quantified and normalized to the actin control.

    Analysis of Bile Flow in Mrp3eC/eC Mice. In view of the abundance of Mrp3 in liver (Fig. 4), measurements were made to determine whether deficiency of Mrp3 affects bile flow. Differences in bile flow were not detected. Basal bile flow rates were 6.0 ± 1.4 and 6.7 ± 1.5 e/min/100 g body weight (p > 0.4), and taurocholate-stimulated bile flow rates increased to 13.0 ± 2.5 and 13.1 ± 2.3 e/min/100 g of body weight (p > 0.9) in wild-type and Mrp3eC/eC mice, respectively.

    Analysis of Bile Acid Pool Size and Fecal Excretion in Mrp3eC/eC Mice. It has been proposed that Mrp3 functions as an intestinal basolateral transporter in the enterohepatic circulation of bile acids. To test that hypothesis, bile acid metabolism was examined in the wild-type and Mrp3eC/eC mice. As shown in Fig. 5A, fecal bile acid secretion was not significantly different in the wild-type and Mrp3eC/eC mice (11.40 ± 2.85 versus 12.96 ± 3.96; p > 0.33). Further analysis revealed that the bile acid pool size and composition were also similar in wild-type and Mrp3eC/eC mice (Fig. 5B). The mass and percentage composition values for all of the major bile acid species, including taurocholate, tauro--muricholate, taurodeoxycholate, tauroursodeoxycholate, and taurochenodeoxycholate, were similar for the two genotypes, with taurocholate and tauro--muricholate accounting for 80% of the pool. A crude fractional turnover rate can be calculated, because the daily rate of fecal bile acid excretion and bile acid pool size were measured in the same animals. The bile acid fractional turnover rate (daily fecal excretion per pool size) was also similar in the two genotypes (Fig. 5C; wild-type, 0.27 ± 0.09 versus Mrp3eC/eC, 0.33 ± 0.08; p > 0.15).

    Analysis of Intestinal Bile Acid Transporter Gene Expression in Mrp3eC/eC Mice. To confirm that the lack of impairment in intestinal bile acid absorption by Mrp3eC/eC mice is not attributable to compensatory alterations in the expression of other intestinal basolateral bile acid transporters, mRNA expression of Asbt, which is responsible for the uptake of bile acids across the brush border, the recently described Ost/, which is a candidate basolateral bile acid transporter (Dawson et al., 2005), and Ibabp, an established farnesoid X receptor target gene and an indirect indicator of bile acid flux through the ileum (Grober et al., 1999; Chen et al., 2003), were analyzed in wild-type and Mrp3eC/eC mice. Asbt, Ost, Ost, and Ibabp mRNA expression did not significantly differ between the wild-type and Mrp3 null animals (data not shown).

    Discussion

    In the present study, Mrp3eC/eC mice generated by targeted disruption were analyzed to determine the in vivo functions of the transporter. Although human MRP3 is an established cellular resistance factor for etoposide, our experiments did not reveal enhanced chemosensitivity of Mrp3eC/eC mice. In addition, Mrp3eC/eC mice did not exhibit increased hematopoietic damage, a major toxicity of etoposide. These findings contrast with Mrp1eC/eC mice, which exhibit etoposide sensitivity associated with increased hematopoietic damage (Lorico et al., 1997; Wijnholds et al., 1997). The experiments showing that ectopic expression of Mrp3 in HEK293 cells is able to confer 2.0- to 2.5-fold resistance to etoposide, a potency that is only 2-fold lower than that reported for Mrp1 expressed in HEK293 cells (Stride et al., 1997), indicate that the lack of chemosensitivity is not attributable to the inability of the murine protein to transport etoposide and instead suggest that the presence of redundant functions mask the contribution of Mrp3 to protecting normal tissues. A report showing that MRP1 is expressed at higher levels than MRP3 in human hematopoietic cells suggests that relatively low levels of Mrp3 expression in chemosensitive cells may be an additional factor that bears on the lack of sensitization in Mrp3eC/eC mice (Laupeze et al., 2001). It is worth mentioning that our experiments on the impact of the pump on the in vivo chemosensitivity of normal tissues do not preclude the possibility that MRP3 may function as a resistance factor in tumors, a notion that is supported by studies showing that MRP3 is expressed in several cancers, and a report in which a correlation between MRP3 expression and clinical outcome was found (Nies et al., 2001; Steinbach et al., 2003).

    A major finding of our study is that Mrp3 functions to protect cholestatic hepatocytes from endobiotics. Serum levels of hepatic constituents such as bile acids and bilirubin glucuronide increase in cholestatic conditions, indicating that hepatocytes are able to deploy basolateral systems to efflux these compounds into sinusoidal blood. Several observations suggested that MRP3 functions as one of these systems. Conjugated bile acids, such as glycocholate in the case of human MRP3 and glycocholate and taurocholate in the case of the rat protein, are established substrates of MRP3 (Hirohashi et al., 2000; Zeng et al., 2000), and MRP3 is induced in sinusoidal membranes of hepatocytes in humans with Dubin-Johnson syndrome, a disorder caused by hereditary deficiency of MRP2 and whose principal manifestation is jaundice (Konig et al., 1999). Induction of Mrp3 has also been reported in rats with experimentally induced obstructive jaundice, in BsepeC/eC mice fed a diet supplemented with cholic acid, and in rat strains that are deficient in Mrp2, the latter of which facilitated the isolation of the rat Mrp3 cDNA from liver (Hirohashi et al., 1998; Donner and Keppler, 2001; Soroka et al., 2001; Wang et al., 2003). In addition, the induction of MRP3 is mediated by transcriptional pathways associated with bile acids. Bile acids and CAR activators induce the expression of Mrp3 in vivo, and induction of MRP3 in Caco2 cells was attributed to FTF-like elements in the MRP3 promoter (Inokuchi et al., 2001; Xiong et al., 2002; Zollner et al., 2003). Furthermore, TnfreC/eC mice fail to induce Mrp3, and induction is impaired in CareC/eC mice (Bohan et al., 2003; Zhang et al., 2004). Although these observations suggested that MRP3 may be an alternate pathway of bile-acid export from hepatocytes, a direct analysis of MRP3 function in vivo has not been reported. Here, we provide direct evidence that Mrp3 protects cholestatic hepatocytes from bile acids by showing that Mrp3eC/eC mice subjected to bile duct ligation have elevated levels of liver bile acids. Moreover, the finding that serum bilirubin levels are elevated in Mrp3eC/eC mice indicates that the protective function of Mrp3 extends to glucuronides as well. These differences in liver parameters were detectable in our experiments after only 3 days of bile duct ligation, a relatively short time frame that was selected so as to minimize the impact of progressive adaptive changes in liver homeostatic mechanisms on the phenotype of Mrp3eC/eC mice. It is noteworthy that this short time frame may explain why differences in liver pathology were not observed between wild-type and Mrp3eC/eC mice. In connection with our assessment of Mrp3 function in liver, it should be mentioned that in the absence of induction, Mrp3 expression in liver is very low or undetectable in rats and humans (Donner and Keppler, 2001; Soroka et al., 2001), whereas in mouse, Mrp3 expression is readily detectable in liver (Fig. 3). Hence, our data suggest that Mrp3-mediated protection of cholestatic mouse liver is afforded by the basal levels of murine Mrp3 expression. In addition, it is important to bear in mind that Mrp3 is likely to represent only one of the basolateral membrane systems that are deployed to protect cholestatic hepatocytes. Mrp4 may be another system, as suggested by reports showing that human MRP4 is able to transport glucuronides and bile acids and that mouse Mrp4 is localized to sinusoidal hepatocyte membranes and induced 7 to 14 days after bile duct ligation (Chen et al., 2001; Rius et al., 2003; Denk et al., 2004).

    In contrast to apical transport across the intestinal apical brush-border membrane, there is limited information regarding the mechanisms and transporters responsible for bile acid efflux across the basolateral membrane (Shneider, 2001). Previous in vitro studies using rat ileal basolateral membrane vesicles have demonstrated bile acid anion exchange (Weinberg et al., 1986) as well as ATP-dependent bile acid transport activity (Shoji et al., 2004). However, the relative in vivo contribution of these transport activities to basolateral bile acid export is unknown. Although Mrp3 was considered to be a candidate for the intestinal basolateral bile acid transporter on the basis of its intestinal expression, localization to the basolateral surface, and ability to transport various bile acid species (Rost et al., 1999; Hirohashi et al., 2000; Zeng et al., 2000; Scheffer et al., 2002), we found that disruption of Mrp3 did not result in significant changes in intestinal bile acid absorption, as indicated by the lack of significant changes in fecal bile acid excretion, bile acid pool size, or the calculated fractional turnover rate for bile acids. These results indicate that Mrp3 is not essential for efficient intestinal absorption of bile acids and that additional transporters must be present to mediate basolateral bile acid efflux in ileum and other Asbt-expressing tissues. One such carrier is the recently described organic solute carrier (OST) / (OST/) (Seward et al., 2003). In the mouse, Ost/ fulfills many of the criteria for a dedicated basolateral bile acid transporter, including tissue expression that closely parallels that of Asbt, basolateral membrane localization, positive regulation by bile acids, and the ability to efflux all the major species of bile acids (Dawson et al., 2005). Although it is possible that Mrp3 functions in conjunction with Ost/ to mediate intestinal basolateral bile acid transporter, the lack of any compensatory increase in Ost/ expression suggests that Mrp3 contributes little to the intestinal reclamation of bile acids.

    Acknowledgements

    We thank Andre Rogatko and Eric Ross for assistance with statistical treatment of our data and Ann L. Craddock and Jamie Haywood for outstanding technical assistance.

    doi:10.1124/mol.104.010587.

    1 Current address: College of Pharmacy and Allied Health Professions, St. John's University, Jamaica, New York.

    References

    Belinsky MG, Bain LJ, Balsara BB, Testa JR, and Kruh GD (1998) Characterization of MOAT-C and MOAT-D, new members of the MRP/cMOAT subfamily of transporter proteins. J Natl Cancer Inst 90: 1735eC1741.

    Bohan A, Chen WS, Denson LA, Held MA, and Boyer JL (2003) Tumor necrosis factor -dependent up-regulation of Lrh-1 and Mrp3(Abcc3) reduces liver injury in obstructive cholestasis. J Biol Chem 278: 36688eC36698.

    Chen F, Ma L, Dawson PA, Sinal CJ, Sehayek E, Gonzalez FJ, Breslow J, Ananthanarayanan M, and Shneider BL (2003) Liver receptor homologue-1 mediates species- and cell line-specific bile acid-dependent negative feedback regulation of the apical sodium-dependent bile acid transporter. J Biol Chem 278: 19909eC19916.

    Chen Z-S, Lee K, and Kruh GD (2001) Transport of cyclic nucleotides and estradiol 17--D-glucuronide by multidrug resistance protein 4. Resistance to 6-mercaptopurine and 6-thioguanine. J Biol Chem 276: 33747eC33754.

    Cui Y, Konig J, Buchholz JK, Spring H, Leier I, and Keppler D (1999) Drug resistance and ATP-dependent conjugate transport mediated by the apical multidrug resistance protein, MRP2, permanently expressed in human and canine cells. Mol Pharmacol 55: 929eC937.

    Dawson PA, Hubbert M, Haywood J, Craddock AL, Zerangue N, Christian WV, and Ballatori N (2005) The heteromeric organic solute transporter -, Ost-Ost, is an ileal basolateral bile acid transporter. J Biol Chem 280: 6960eC6968.

    Denk GU, Soroka CJ, Takeyama Y, Chen WS, Schuetz JD, and Boyer JL (2004) Multidrug resistance-associated protein 4 is up-regulated in liver but down-regulated in kidney in obstructive cholestasis in the rat. J Hepatol 40: 585eC591.

    Donner MG and Keppler D (2001) Up-regulation of basolateral multidrug resistance protein 3 (Mrp3) in cholestatic rat liver. Hepatology 34: 351eC359.

    Gerk PM and Vore M (2002) Regulation of expression of the multidrug resistance-associated protein 2 (MRP2) and its role in drug disposition. J Pharmacol Exp Ther 302: 407eC415.

    Gerloff T, Stieger B, Hagenbuch B, Madon J, Landmann L, Roth J, Hofmann AF, and Meier PJ (1998) The sister of P-glycoprotein represents the canalicular bile salt export pump of mammalian liver. J Biol Chem 273: 10046eC10050.

    Grober J, Zaghini I, Fujii H, Jones SA, Kliewer SA, Willson TM, Ono T, and Besnard P (1999) Identification of a bile acid-responsive element in the human ileal bile acid-binding protein gene. Involvement of the farnesoid X receptor/9-cis-retinoic acid receptor heterodimer. J Biol Chem 274: 29749eC29754.

    Hirohashi T, Suzuki H, Ito K, Ogawa K, Kume K, Shimizu T, and Sugiyama Y (1998) Hepatic expression of multidrug resistance-associated protein-like proteins maintained in Eisai hyperbilirubinemic rats. Mol Pharmacol 53: 1068eC1075.

    Hirohashi T, Suzuki H, and Sugiyama Y (1999) Characterization of the transport properties of cloned rat multidrug resistance-associated protein 3 (MRP3). J Biol Chem 274: 15181eC15185.

    Hirohashi T, Suzuki H, Takikawa H, and Sugiyama Y (2000) ATP-dependent transport of bile salts by rat multidrug resistance-associated protein 3 (Mrp3). J Biol Chem 275: 2905eC2910.

    Inokuchi A, Hinoshita E, Iwamoto Y, Kohno K, Kuwano M, and Uchiumi T (2001) Enhanced expression of the human multidrug resistance protein 3 by bile salt in human enterocytes: a transcriptional control of a plausible bile acid transporter. J Biol Chem 276: 46822eC46829.

    Johnson DR, Finch RA, Lin ZP, Zeiss CJ, and Sartorelli AC (2001) The pharmacological phenotype of combined multidrug-resistance mdr1a/1b- and mrp1-deficient mice. Cancer Res 61: 1469eC1476.

    Konig J, Rost D, Cui Y, and Keppler D (1999) Characterization of the human multidrug resistance protein isoform MRP3 localized to the basolateral hepatocyte membrane. Hepatology 29: 1156eC1163.

    Kool M, van der Linden M, de Haas M, Scheffer GL, de Vree JM, Smith AJ, Jansen G, Peters GJ, Ponne N, Scheper RJ, et al. (1999) MRP3, an organic anion transporter able to transport anti-cancer drugs. Proc Natl Acad Sci USA 96: 6914eC6919.

    Kruh GD and Belinsky MG (2003) The MRP family of drug efflux pumps. Oncogene 22: 7537eC7552.

    Laupeze B, Amiot L, Payen L, Drenou B, Grosset JM, Lehne G, Fauchet R, and Fardel O (2001) Multidrug resistance protein (MRP) activity in normal mature leukocytes and CD34-positive hematopoietic cells from peripheral blood. Life Sci 68: 1323eC1331.

    Lee YM, Cui Y, Konig J, Risch A, Jager B, Drings P, Bartsch H, Keppler D, and Nies AT (2004) Identification and functional characterization of the natural variant MRP3-Arg1297His of human multidrug resistance protein 3 (MRP3/ABCC3). Pharmacogenetics 14: 213eC223.

    Lorico A, Rappa G, Finch RA, Yang D, Flavell RA, and Sartorelli AC (1997) Disruption of the murine MRP (multidrug resistance protein) gene leads to increased sensitivity to etoposide (VP-16) and increased levels of glutathione. Cancer Res 57: 5238eC5242.

    Mashige F, Tanaka N, Maki A, Kamei S, and Yamanaka M (1981) Direct spectrophotometry of total bile acids in serum. Clin Chem 27: 1352eC1356.

    Nies AT, Konig J, Pfannschmidt M, Klar E, Hofmann WJ, and Keppler D (2001) Expression of the multidrug resistance proteins MRP2 and MRP3 in human hepatocellular carcinoma. Int J Cancer 94: 492eC499.

    Paulusma CC, Kothe MJ, Bakker CT, Bosma PJ, van Bokhoven I, van Marle J, Bolder U, Tytgat GN, and Oude Elferink RP (2000) Zonal down-regulation and redistribution of the multidrug resistance protein 2 during bile duct ligation in rat liver. Hepatology 31: 684eC693.

    Rius M, Nies AT, Hummel-Eisenbeiss J, Jedlitschky G, and Keppler D (2003) Cotransport of reduced glutathione with bile salts by MRP4 (ABCC4) localized to the basolateral hepatocyte membrane. Hepatology 38: 374eC384.

    Rost D, Kartenbeck J, and Keppler D (1999) Changes in the localization of the rat canalicular conjugate export pump Mrp2 in phalloidin-induced cholestasis. Hepatology 29: 814eC821.

    Rost D, Mahner S, Sugiyama Y, and Stremmel W (2002) Expression and localization of the multidrug resistance-associated protein 3 in rat small and large intestine. Am J Physiol 282: G720eCG726.

    Scheffer GL, Kool M, de Haas M, de Vree JM, Pijnenborg AC, Bosman DK, Elferink RP, van der Valk P, Borst P, and Scheper RJ (2002) Tissue distribution and induction of human multidrug resistant protein 3. Lab Investig 82: 193eC201.

    Schultz MJ, Wijnholds J, Peppelenbosch MP, Vervoordeldonk MJ, Speelman P, van Deventer SJ, Borst P, and van der Poll T (2001) Mice lacking the multidrug resistance protein 1 are resistant to Streptococcus pneumoniae-induced pneumonia. J Immunol 166: 4059eC5064.

    Schwarz M, Russell DW, Dietschy JM, and Turley SD (1998) Marked reduction in bile acid synthesis in cholesterol 7alpha-hydroxylase-deficient mice does not lead to diminished tissue cholesterol turnover or to hypercholesterolemia. J Lipid Res 39: 1833eC1843.

    Seward DJ, Koh AS, Boyer JL, and Ballatori N (2003) Functional complementation between a novel mammalian polygenic transport complex and an evolutionarily ancient organic solute transporter, OST-OST. J Biol Chem 278: 27473eC27482.

    Shneider BL (2001) Intestinal bile acid transport: biology, physiology and pathophysiology. J Pediatr Gastroenterol Nutr 32: 407eC417.

    Shoji T, Suzuki H, Kusuhara H, Watanabe Y, Sakamoto S, and Sugiyama Y (2004) ATP-dependent transport of organic anions into isolated basolateral membrane vesicles from rat intestine. Am J Physiol 287: G749eCG756.

    Soroka CJ, Lee JM, Azzaroli F, and Boyer JL (2001) Cellular localization and up-regulation of multidrug resistance-associated protein 3 in hepatocytes and cholangiocytes during obstructive cholestasis in rat liver. Hepatology 33: 783eC791.

    Steinbach D, Wittig S, Cario G, Viehmann S, Mueller A, Gruhn B, Haefer R, Zintl F, and Sauerbrey A (2003) The multidrug resistance-associated protein 3 (MRP3) is associated with a poor outcome in childhood ALL and may account for the worse prognosis in male patients and T-cell immunophenotype. Blood 102: 4493eC4498.

    Stride BD, Grant CE, Loe DW, Hipfner DR, Cole SPC, and Deeley RG (1997) Pharmacological characterization of the murine and human orthologs of multidrug-resistance protein in transfected human embryonic kidney cells. Mol Pharmacol 52: 344eC353.

    Torchia EC, Labonte ED, and Agellon LB (2001) Separation and quantitation of bile acids using an isocratic solvent system for high performance liquid chromatography coupled to an evaporative light scattering detector. Anal Biochem 298: 293eC298.

    Turley SD, Daggy BP, and Dietschy JM (1996) Effect of feeding psyllium and cholestyramine in combination on low density lipoprotein metabolism and fecal bile acid excretion in hamsters with dietary-induced hypercholesterolemia. J Cardiovasc Pharmacol 27: 71eC79.

    Wang R, Lam P, Liu L, Forrest D, Yousef IM, Mignault D, Phillips MJ, and Ling V (2003) Severe cholestasis induced by cholic acid feeding in knockout mice of sister of P-glycoprotein. Hepatology 38: 1489eC1499.

    Wang R, Salem M, Yousef IM, Tuchweber B, Lam P, Childs SJ, Helgason CD, Ackerley C, Phillips MJ, and Ling V (2001) Targeted inactivation of sister of P-glycoprotein gene (spgp) in mice results in nonprogressive but persistent intrahepatic cholestasis. Proc Natl Acad Sci USA 98: 2011eC2016.

    Weinberg SL, Burckhardt G, and Wilson FA (1986) Taurocholate transport by rat intestinal basolateral membrane vesicles. Evidence for the presence of an anion exchange transport system. J Clin Investig 78: 44eC50.

    Wijnholds J, Evers R, van Leusden MR, Mol CA, Zaman GJ, Mayer U, Beijnen JH, van der Valk M, Krimpenfort P, and Borst P (1997) Increased sensitivity to anticancer drugs and decreased inflammatory response in mice lacking the multidrug resistance-associated protein. Nat Med 3: 1275eC1279.

    Xiong H, Yoshinari K, Brouwer KL, and Negishi M (2002) Role of constitutive androstane receptor in the in vivo induction of Mrp3 and CYP2B1/2 by phenobarbital. Drug Metab Dispos 30: 918eC923.

    Zeng H, Bain LJ, Belinsky MG, and Kruh GD (1999) Expression of multidrug resistance protein-3 (multispecific organic anion transporter-D) in human embryonic kidney 293 cells confers resistance to anticancer agents. Cancer Res 59: 5964eC5967.

    Zeng H, Chen Z-S, Belinsky MG, Rea PA, and Kruh GD (2001) Transport of methotrexate (MTX) and folates by multidrug resistance protein (MRP) 3 and MRP1: effect of polyglutamylation on MTX transport. Cancer Res 61: 7225eC7232.

    Zeng H, Liu G, Rea PA, and Kruh GD (2000) Transport of amphipathic anions by human multidrug resistance protein 3. Cancer Res 60: 4779eC4784.

    Zhang J, Huang W, Qatanani M, Evans RM, and Moore DD (2004) The constitutive androstane receptor and pregnane X receptor function coordinately to prevent bile acid-induced hepatotoxicity. J Biol Chem 279: 49517eC49522.

    Zollner G, Fickert P, Fuchsbichler A, Silbert D, Wagner M, Arbeiter S, Gonzalez FJ, Marschall HU, Zatloukal K, Denk H, et al. (2003) Role of nuclear bile acid receptor, FXR, in adaptive ABC transporter regulation by cholic and ursodeoxycholic acid in mouse liver, kidney and intestine. J Hepatol 39: 480eC488.

作者: Martin G. Belinsky, Paul A. Dawson, Irina Shchavel 2007-5-15
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具