Preclinical/Animal Transporters

Mdr1a - rat

Mdr1a/b (multidrug resistance protein 1a/b), rat


Aliases: Mdr1a: Abcb4, Evi32, Mdr3, P-gp, Pgp, Pgy-3, Pgy3, mdr-3; Mdr1b: Abcb1, Mdr1, Mdr1b, Pgy-1, Pgy1, mdr
Gene names: ATP-binding family, sub-family B (MDR/TAP), member 1A/B (Abcb1a/b)

While humans have only one MDR1 gene to encode a single P-gp drug transporter, there are two genes in rodents, Mdr1a and Mdr1b, that encode P-gps with largely overlapping functions and extensive sequence identity to the single human gene [1]. The 775-kb-long mMdr gene cluster is located on mouse chromosome 5 [2], while the rMdr cluster is found in the rat chromosomal region 4q11-12 [3].
All rodent Mdr1 transporters consist of 1272-1277 amino acids, have a molecular mass of 170 kDa [4], [5], [6], [7] and are arranged in two homologous halves with six transmembrane domains each [8]. Their protein structure is highly similar to human MDR1: mouse Mdr1a and Mdr1b share 87% and 80% amino acid homology, respectively, with human MDR1, while rat Mdr1 transporter amino acid sequences are 80% homologous to human [7], [9]. The amino acid-level homology between mouse and rat Mdr1 transporters is as high as 93% [10]. 
Murine Mdr1 transporters show a similar tissue distribution to that seen in humans. mMdr1a is predominantly expressed in the intestine, lung, liver, as well as the blood-brain and the blood-testes barriers, while mMdr1b is highly expressed in the adrenal, placenta, ovary, and the pregnant uterus [11]. Both genes are expressed at similar levels in the mouse kidney. These distribution data suggest that the two murine Mdr1 genes jointly accomplish the same function as the single MDR1 in humans.
Rat Mdr1 proteins display a slightly different tissue distribution compared to human or mouse. Expression of rMdr1a was primarily detected in the gastrointestinal tract and ovary, with lower levels in the brain, testis, kidney, lung and liver, whereas rMdr1b was found to reside mainly in the distal gastrointestinal tract, ovary, lung, and heart, with lower levels in the ileum, spleen, thymus, liver, kidney, and the embryo [12]. In the brain, rMdr1a was found to be expressed on the luminal (apical) membrane of capillary endothelial cells, whereas rMdr1b was detected in the parenchyma [13].
There is a significant overlap in the substrate-interacting residues of the MDR1/Mdr1 ligand binding site across human, rat and mouse [14]. Consistently, murine Mdr1 transporters have wide substrate specificities overlapping with human MDR1: they mainly accept amphiphilic and hydrophobic substrates such as anthracyclines, vinca alcaloids, epipodophyllotoxins and other chemoterapy drugs, various antibiotics, as well as endogenous compounds (e.g. steroids, cytokines, bilirubin) and natural products [15-21]. There are some exceptions, however, such as the antiepileptic drugs phenytoin and levetiracetam that were directionally transported by mMdr1a but not by hMDR1 [22]. Bisphenol A is a substrate for rMdr1b but not for hMDR1 or rMdr1a [23]. An in vitro directional transport study revealed that human, mouse and rat Mdr1 transporter liabilities for some compounds may be species-specific [7]. Also, some studies have reported that mouse Mdr1a and Mdr1b have different propensities for the transport of progesterone [24], ramosetron [25], diltiazem, cyclosporin A, or dexamethasone [26]. Overexpression of human MDR1 versus mouse Mdr1a or Mdr1b in CHO cells conferred different levels of drug resistance due to the distinct capacities of these transporters [27]. Cutler et al. have shown that in order to obtain the same inhibition as in rats and mice, higher blood concentration of the P-gp inhibitor elacridar was needed in guinea pigs [28]. It was also demonstrated that the brain penetration of [18F]altanserin and [11C]GR205171 was 4.5 and 8.6 times lower in rodents than in humans [29]. These results indicate higher activity and/or expression of Mdr1 isoforms in the murine and rat BBB compared to humans. Differences between human, rodent and other species were also found with regard to the inhibitory effects of quinidine and verapamil on Mdr1-mediated daunorubicin, digoxin and cyclosporin A transport in Mdr1-transfected cell lines [30]. These differences may result from divergent amino acid residues at the drug binding sites or distinct tissue-specific expression patterns between species.
For a long time, little was known of Mdr1 function in non-human species. The generation of Mdr1a-knockout mice marked a breakthrough in investigating the in vivo role of Mdr1 and its importance to drug transport [31]. These mice were viable and fertile; however, Mdr1 substrates tended to accumulate in their brains and they were thus more susceptible than wild-type to central neurotoxicity.
Mdr1a(-/-) / Mdr1b(-/-) mice generated by Schinkel et al. [32] were also healthy as long as left unchallenged, but turned out to be 50-100 times more sensitive to the neurotoxic effects of the pesticide ivermectin, and the accumulation of this drug in their brains was 80-100-fold increased over wild-types. In the double knock-outs vinblastine was 3-fold more likely to cause sensitivity, and acute arsenic toxicity was more pronounced compared to wild-type mice [33]. Furthermore, Mdr1 double-deficient mice showed increased oral drug absorption and impaired rate of drug elimination [34]. Some drugs penetrating into the CNS, such as cyclosporine A, were lethal to the double knock-outs [31], [35]. Hence, Mdr1a/1b(-/-) mice were utilized in a large number of studies as a model to explore the role of Mdr1 transporters in the in vivo disposition and toxicity of various organic chemicals including trabectedin [36] (which caused severe liver toxicity in Mdr1-deficient mice), digitoxin, vincristine, taxol, tributylmethyl ammonium or asimadoline [33]. The intestinal and hepatobiliary clearance of cationic amphiphilic drugs was reduced in double knock-outs [37], [38]. These animals showed increased bioavailability of paclitaxel [37], and elevated tissue levels of digoxin [39], dexamethasone [40], rifampicin [41], and morphine [42] in comparison with wild-type mice. 
When Mdr1 transporters were down-regulated in normal mice, significantly increased triptolide exposure in the liver and plasma was strongly correlated with enhanced hepatotoxicity [43]. Increases in xenobiotic penetration have also been observed when mouse Mdr1 transporters were inhibited with cyclosporine A [44], [45].
A novel MDR1-humanized mouse model was generated on the Mdr1a/b double knockout background by using a mouse artificial chromosome (MAC) vector. hMDR1-MAC mice are the first to express functional human MDR1 at the blood-brain barrier, and can be used estimate the human brain distribution of P-gp-substrate drugs in a nonhuman in vivo system [46]. 
Multiple rat Mdr1 knock-out strains developed by different laboratories are becoming employed in pharmacokinetic studies more often than knock-out mice, due to the practical advantages of using rats instead of mice, as well as the superior relevance of rats to drug development studies. Quantitatively consistent with murine models, distribution of some Mdr1 substrates to the brain was increased in both SAGE-type and Wistar-type Mdr1-deficient rats [47], [48]. Despite some changes observed in the gene expression of SAGE knock-out rats, they offer a useful tool for the study of transporter-mediated pharmacokinetics [49]. Recently, Mdr1a/b double-knockout rats were generated by the CRISPR/Cas9 system without any off-target effect, and significantly different digoxin absorption was observed in the KO animals [50]. 

References

1.    Hsu, S.I., L. Lothstein, and S.B. Horwitz, Differential overexpression of three mdr gene family members in multidrug-resistant J774.2 mouse cells. Evidence that distinct P-glycoprotein precursors are encoded by unique mdr genes. J Biol Chem, 1989. 264(20): p. 12053-62.
2.    Raymond, M. and P. Gros, Mammalian multidrug-resistance gene: correlation of exon organization with structural domains and duplication of an ancestral gene. Proc Natl Acad Sci U S A, 1989. 86(17): p. 6488-92.
3.    Popescu, N.C., J.A. Silverman, and S.S. Thorgeirsson, Mapping of a rat multidrug resistance gene by fluorescence in situ hybridization. Genomics, 1993. 15(1): p. 182-4.
4.    Devault, A. and P. Gros, Two members of the mouse mdr gene family confer multidrug resistance with overlapping but distinct drug specificities. Mol Cell Biol, 1990. 10(4): p. 1652-63.
5.    Hsu, S.I., et al., Structural analysis of the mouse mdr1a (P-glycoprotein) promoter reveals the basis for differential transcript heterogeneity in multidrug-resistant J774.2 cells. Mol Cell Biol, 1990. 10(7): p. 3596-606.
6.    Silverman, J.A., et al., Cloning and characterization of a member of the rat multidrug resistance (mdr) gene family. Gene, 1991. 106(2): p. 229-36.
7.    Booth-Genthe, C.L., et al., Development and characterization of LLC-PK1 cells containing Sprague-Dawley rat Abcb1a (Mdr1a): comparison of rat P-glycoprotein transport to human and mouse. J Pharmacol Toxicol Methods, 2006. 54(1): p. 78-89.
8.    Gottesman, M.M. and I. Pastan, The multidrug transporter, a double-edged sword. J Biol Chem, 1988. 263(25): p. 12163-6.
9.    van der Bliek, A.M., et al., Sequence of mdr3 cDNA encoding a human P-glycoprotein. Gene, 1988. 71(2): p. 401-11.
10.    Hubbard, T.J., et al., Ensembl 2009. Nucleic Acids Res, 2009. 37(Database issue): p. D690-7.
11.    Schinkel, A.H., et al., Multidrug resistance and the role of P-glycoprotein knockout mice. Eur J Cancer, 1995. 31A(7-8): p. 1295-8.
12.    Brady, J.M., et al., Tissue distribution and chemical induction of multiple drug resistance genes in rats. Drug Metab Dispos, 2002. 30(7): p. 838-44.
13.    Regina, A., et al., Mrp1 multidrug resistance-associated protein and P-glycoprotein expression in rat brain microvessel endothelial cells. J Neurochem, 1998. 71(2): p. 705-15.
14.    Jain, S., M. Grandits, and G.F. Ecker, Interspecies comparison of putative ligand binding sites of human, rat and mouse P-glycoprotein. Eur J Pharm Sci, 2018. 122: p. 134-143.
15.    Gottesman, M.M. and I. Pastan, Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem, 1993. 62: p. 385-427.
16.    Ambudkar, S.V., et al., Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol, 1999. 39: p. 361-98.
17.    van Kalken, C.K., et al., Cortisol is transported by the multidrug resistance gene product P-glycoprotein. Br J Cancer, 1993. 67(2): p. 284-9.
18.    Watchko, J.F., M.J. Daood, and T.W. Hansen, Brain bilirubin content is increased in P-glycoprotein-deficient transgenic null mutant mice. Pediatr Res, 1998. 44(5): p. 763-6.
19.    Drach, J., et al., Involvement of P-glycoprotein in the transmembrane transport of interleukin-2 (IL-2), IL-4, and interferon-gamma in normal human T lymphocytes. Blood, 1996. 88(5): p. 1747-54.
20.    Romiti, N., et al., Effects of curcumin on P-glycoprotein in primary cultures of rat hepatocytes. Life Sci, 1998. 62(25): p. 2349-58.
21.    Jodoin, J., M. Demeule, and R. Beliveau, Inhibition of the multidrug resistance P-glycoprotein activity by green tea polyphenols. Biochim Biophys Acta, 2002. 1542(1-3): p. 149-59.
22.    Baltes, S., et al., Differences in the transport of the antiepileptic drugs phenytoin, levetiracetam and carbamazepine by human and mouse P-glycoprotein. Neuropharmacology, 2007. 52(2): p. 333-46.
23.    Mazur, C.S., et al., Human and rat ABC transporter efflux of bisphenol a and bisphenol a glucuronide: interspecies comparison and implications for pharmacokinetic assessment. Toxicol Sci, 2012. 128(2): p. 317-25.
24.    Yang, C.P., et al., Differential transport properties of two mdr gene products are distinguished by progesterone. J Biol Chem, 1990. 265(18): p. 10282-8.
25.    Yamamoto, C., et al., Contribution of P-glycoprotein to efflux of ramosetron, a 5-HT3 receptor antagonist, across the blood-brain barrier. J Pharm Pharmacol, 2002. 54(8): p. 1055-63.
26.    Katoh, M., et al., Kinetic analyses for species differences in P-glycoprotein-mediated drug transport. J Pharm Sci, 2006. 95(12): p. 2673-83.
27.    Tang-Wai, D.F., et al., Human (MDR1) and mouse (mdr1, mdr3) P-glycoproteins can be distinguished by their respective drug resistance profiles and sensitivity to modulators. Biochemistry, 1995. 34(1): p. 32-9.
28.    Cutler, L., et al., Development of a P-glycoprotein knockout model in rodents to define species differences in its functional effect at the blood-brain barrier. J Pharm Sci, 2006. 95(9): p. 1944-53.
29.    Syvanen, S., et al., Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos, 2009. 37(3): p. 635-43.
30.    Suzuyama, N., et al., Species differences of inhibitory effects on P-glycoprotein-mediated drug transport. J Pharm Sci, 2007. 96(6): p. 1609-18.
31.    Schinkel, A.H., et al., Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell, 1994. 77(4): p. 491-502.
32.    Schinkel, A.H., et al., Normal viability and altered pharmacokinetics in mice lacking mdr1-type (drug-transporting) P-glycoproteins. Proc Natl Acad Sci U S A, 1997. 94(8): p. 4028-33.
33.    Liu, J., et al., Multidrug-resistance mdr1a/1b double knockout mice are more sensitive than wild type mice to acute arsenic toxicity, with higher arsenic accumulation in tissues. Toxicology, 2002. 170(1-2): p. 55-62.
34.    Schinkel, A.H., et al., Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest, 1995. 96(4): p. 1698-705.
35.    Kwei, G.Y., et al., Disposition of ivermectin and cyclosporin A in CF-1 mice deficient in mdr1a P-glycoprotein. Drug Metab Dispos, 1999. 27(5): p. 581-7.
36.    Beumer, J.H., et al., Disposition and toxicity of trabectedin (ET-743) in wild-type and mdr1 gene (P-gp) knock-out mice. Invest New Drugs, 2010. 28(2): p. 145-55.
37.    Sparreboom, A., et al., Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci U S A, 1997. 94(5): p. 2031-5.
38.    Smit, J.W., et al., Absence or pharmacological blocking of placental P-glycoprotein profoundly increases fetal drug exposure. J Clin Invest, 1999. 104(10): p. 1441-7.
39.    Kawahara, M., et al., Physiologically based pharmacokinetics of digoxin in mdr1a knockout mice. J Pharm Sci, 1999. 88(12): p. 1281-7.
40.    Meijer, O.C., et al., Penetration of dexamethasone into brain glucocorticoid targets is enhanced in mdr1A P-glycoprotein knockout mice. Endocrinology, 1998. 139(4): p. 1789-93.
41.    Schuetz, E.G., et al., P-glycoprotein: a major determinant of rifampicin-inducible expression of cytochrome P4503A in mice and humans. Proc Natl Acad Sci U S A, 1996. 93(9): p. 4001-5.
42.    Zong, J. and G.M. Pollack, Morphine antinociception is enhanced in mdr1a gene-deficient mice. Pharm Res, 2000. 17(6): p. 749-53.
43.    Kong, L.L., et al., Inhibition of P-glycoprotein Gene Expression and Function Enhances Triptolide-induced Hepatotoxicity in Mice. Sci Rep, 2015. 5: p. 11747.
44.    Hendrikse, N.H. and W. Vaalburg, Dynamics of multidrug resistance: P-glycoprotein analyses with positron emission tomography. Methods, 2002. 27(3): p. 228-33.
45.    Kemper, E.M., et al., Increased penetration of paclitaxel into the brain by inhibition of P-Glycoprotein. Clin Cancer Res, 2003. 9(7): p. 2849-55.
46.    Yamasaki, Y., et al., Characterization of P-Glycoprotein Humanized Mice Generated by Chromosome Engineering Technology: Its Utility for Prediction of Drug Distribution to the Brain in Humans. Drug Metab Dispos, 2018. 46(11): p. 1756-1766.
47.    Bundgaard, C., C.J. Jensen, and M. Garmer, Species comparison of in vivo P-glycoprotein-mediated brain efflux using mdr1a-deficient rats and mice. Drug Metab Dispos, 2012. 40(3): p. 461-6.
48.    Chu, X., et al., Characterization of multidrug resistance 1a/P-glycoprotein knockout rats generated by zinc finger nucleases. Mol Pharmacol, 2012. 81(2): p. 220-7.
49.    Zamek-Gliszczynski, M.J., et al., Minor compensatory changes in SAGE Mdr1a (P-gp), Bcrp, and Mrp2 knockout rats do not detract from their utility in the study of transporter-mediated pharmacokinetics. Drug Metab Dispos, 2013. 41(6): p. 1174-8.
50.    Liang, C., et al., Development and Characterization of MDR1 (Mdr1a/b) CRISPR/Cas9 Knockout Rat Model. Drug Metab Dispos, 2019. 47(2): p. 71-79.

Solvo Transporter Book 4th Edition
Transporter Book 4th edition
  • 63 transporters
  • over 1500 references
  • comprehensive information on holistic models and proteomics for transporter research
  • changes in the regulatory landscape and scientific insights

Get the Book