메뉴 건너뛰기




Volumn , Issue , 2019, Pages 135-204

Drug-Drug Interactions Involving the Membrane Transport Process

Author keywords

[No Author keywords available]

Indexed keywords


EID: 77955260077     PISSN: None     EISSN: None     Source Type: Book    
DOI: 10.1201/9780429131967-5     Document Type: Chapter
Times cited : (4)

References (398)
  • 1
    • 29144436223 scopus 로고    scopus 로고
    • Membrane transporters and drug response
    • Brunton LL, Lazo JS, Parker KL, eds., 11th ed. New York: McGraw-Hill
    • Giacomini KM, Sugiyama, Y. Membrane transporters and drug response. In: Brunton LL, Lazo JS, Parker KL, eds. Goodman & Gilman's The Pharmacological Basis of Therapeutics. 11th ed. New York: McGraw-Hill, 2006:41-70.
    • (2006) Goodman & Gilman's the Pharmacological Basis of Therapeutics , pp. 41-70
    • Giacomini, K.M.1    Sugiyama, Y.2
  • 2
    • 0035012079 scopus 로고    scopus 로고
    • Inhibition of biliary excretion of methotrexate by probenecid in rats: Quantitative prediction of interaction from in vitro data
    • Ueda K, Kato Y, Komatsu K, et al. Inhibition of biliary excretion of methotrexate by probenecid in rats: quantitative prediction of interaction from in vitro data. J Pharmacol Exp Ther 2001; 297:1036-1043.
    • (2001) J Pharmacol Exp Ther , vol.297 , pp. 1036-1043
    • Ueda, K.1    Kato, Y.2    Komatsu, K.3
  • 3
    • 0031777718 scopus 로고    scopus 로고
    • Quantative prediction of in vivo drug clearance and drug interactions from in vitro data on metabolism together with binding and transport
    • Ito K, Iwatsubo T, Ueda K, et al. Quantative prediction of in vivo drug clearance and drug interactions from in vitro data on metabolism together with binding and transport. Ann Rev Pharmacol Toxicol 1998; 38:461-499.
    • (1998) Ann Rev Pharmacol Toxicol , vol.38 , pp. 461-499
    • Ito, K.1    Iwatsubo, T.2    Ueda, K.3
  • 4
    • 0031723235 scopus 로고    scopus 로고
    • Prediction of pharmacokinetic alterations caused by drug-drug interactions: Metabolic interaction in the liver
    • Ito K, Iwatsubo T, Kanamitsu S, et al. Prediction of pharmacokinetic alterations caused by drug-drug interactions: metabolic interaction in the liver. Pharmacol Rev 1998; 50:387-412.
    • (1998) Pharmacol Rev , vol.50 , pp. 387-412
    • Ito, K.1    Iwatsubo, T.2    Kanamitsu, S.3
  • 5
    • 0033026478 scopus 로고    scopus 로고
    • Hepatobiliary transport governs overall elimination of peptidic endothelin antagonists in rats
    • Kato Y, Akhteruzzaman S, Hisaka A, et al. Hepatobiliary transport governs overall elimination of peptidic endothelin antagonists in rats. J Pharmacol Exp Ther 1999; 288:568-574.
    • (1999) J Pharmacol Exp Ther , vol.288 , pp. 568-574
    • Kato, Y.1    Akhteruzzaman, S.2    Hisaka, A.3
  • 6
    • 85024471761 scopus 로고    scopus 로고
    • Function of uptake transporters for taurocholate and estradiol 17b - D-glucuronide in cryopreserved human hepatocytes
    • Shitara Y, Li AP, Kato Y, et al. Function of uptake transporters for taurocholate and estradiol 17b - D-glucuronide in cryopreserved human hepatocytes. Drug Metab Pharmacokinet 2003; 18:33-41.
    • (2003) Drug Metab Pharmacokinet , vol.18 , pp. 33-41
    • Shitara, Y.1    Li, A.P.2    Kato, Y.3
  • 7
    • 4644364576 scopus 로고    scopus 로고
    • Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans
    • Hirano M, Maeda K, Shitara Y, et al. Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther 2004; 311:139-146.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 139-146
    • Hirano, M.1    Maeda, K.2    Shitara, Y.3
  • 8
    • 0029608831 scopus 로고
    • Evaluation of the uptake of pravastatin by perfused rat liver and primary cultured rat hepatocytes
    • Ishigami M, Tokui T, Komai T, et al. Evaluation of the uptake of pravastatin by perfused rat liver and primary cultured rat hepatocytes. Pharm Res 1995; 12: 1741-1745.
    • (1995) Pharm Res , vol.12 , pp. 1741-1745
    • Ishigami, M.1    Tokui, T.2    Komai, T.3
  • 9
    • 0028001860 scopus 로고
    • Formation of extensive canalicular networks by rat hepatocytes cultured in collagen-sandwich configuration
    • LeCluyse EL, Audus KL, Hochman JH. Formation of extensive canalicular networks by rat hepatocytes cultured in collagen-sandwich configuration. Am J Physiol 1994; 266:C1764-C1774.
    • (1994) Am J Physiol , vol.266 , pp. C1764-C1774
    • Lecluyse, E.L.1    Audus, K.L.2    Hochman, J.H.3
  • 10
    • 0033017134 scopus 로고    scopus 로고
    • Correlation of biliary excretion in sandwichcultured rat hepatocytes and in vivo in rats
    • Liu X, Chism JP, LeCluyse EL, et al. Correlation of biliary excretion in sandwichcultured rat hepatocytes and in vivo in rats. Drug Metab Dispos 1999; 27:637-644.
    • (1999) Drug Metab Dispos , vol.27 , pp. 637-644
    • Liu, X.1    Chism, J.P.2    Lecluyse, E.L.3
  • 11
    • 0032589267 scopus 로고    scopus 로고
    • Use of Ca2+ modulation to evaluate biliary excretion in sandwich- cultured rat hepatocytes
    • Liu X, LeCluyse EL, Brouwer KR, et al. Use of Ca2+ modulation to evaluate biliary excretion in sandwich- cultured rat hepatocytes. J Pharmacol Exp Ther 1999; 289:1592-1599.
    • (1999) J Pharmacol Exp Ther , vol.289 , pp. 1592-1599
    • Liu, X.1    Lecluyse, E.L.2    Brouwer, K.R.3
  • 12
    • 3242699783 scopus 로고    scopus 로고
    • P-glycoprotein expression, localization, and function in sandwich-cultured primary rat and human hepatocytes: Relevance to the hepatobiliary disposition of a model opioid peptide
    • Hoffmaster KA, Turncliff RZ, LeCluyse EL, et al. P-glycoprotein expression, localization, and function in sandwich-cultured primary rat and human hepatocytes: relevance to the hepatobiliary disposition of a model opioid peptide. Pharm Res 2004; 21:1294-1302.
    • (2004) Pharm Res , vol.21 , pp. 1294-1302
    • Hoffmaster, K.A.1    Turncliff, R.Z.2    Lecluyse, E.L.3
  • 13
    • 0033036416 scopus 로고    scopus 로고
    • GSH transport in immortalized mouse brain endothelial cells: Evidence for apical localization of a sodium-dependent GSH transporter
    • Kannan R, Mittur A, Bao Y, et al. GSH transport in immortalized mouse brain endothelial cells: evidence for apical localization of a sodium-dependent GSH transporter. J Neurochem 1999; 73:390-399.
    • (1999) J Neurochem , vol.73 , pp. 390-399
    • Kannan, R.1    Mittur, A.2    Bao, Y.3
  • 14
    • 0031809205 scopus 로고    scopus 로고
    • Characterization of efflux transport of organic anions in a mouse brain capillary endothelial cell line
    • Kusuhara H, Suzuki H, Naito M, et al. Characterization of efflux transport of organic anions in a mouse brain capillary endothelial cell line. J Pharmacol Exp Ther 1998; 285:1260-1265.
    • (1998) J Pharmacol Exp Ther , vol.285 , pp. 1260-1265
    • Kusuhara, H.1    Suzuki, H.2    Naito, M.3
  • 15
    • 0027133590 scopus 로고
    • Mechanisms mediating renal secretion of organic anions and cations
    • Pritchard JB, Miller DS. Mechanisms mediating renal secretion of organic anions and cations. Physiol Rev 1993; 73:765-796.
    • (1993) Physiol Rev , vol.73 , pp. 765-796
    • Pritchard, J.B.1    Miller, D.S.2
  • 16
    • 0027049962 scopus 로고
    • Neutral amino acid transport by the blood-brain barrier. Membrane vesicle studies
    • Sanchez del Pino MM, Hawkins RA, Peterson DR. Neutral amino acid transport by the blood-brain barrier. Membrane vesicle studies. J Biol Chem 1992; 267: 25951-25957.
    • (1992) J Biol Chem , vol.267 , pp. 25951-25957
    • Sanchez Del Pino, M.M.1    Hawkins, R.A.2    Peterson, D.R.3
  • 17
    • 0024348932 scopus 로고
    • Transport studies with renal proximal tubular and small intestinal brush border and basolateral membrane vesicles: Vesicle heterogeneity, coexistence of transport system
    • Murer H, Gmaj P, Steiger B, et al. Transport studies with renal proximal tubular and small intestinal brush border and basolateral membrane vesicles: vesicle heterogeneity, coexistence of transport system. Methods Enzymol 1989; 172:346-364.
    • (1989) Methods Enzymol , vol.172 , pp. 346-364
    • Murer, H.1    Gmaj, P.2    Steiger, B.3
  • 18
    • 0025684803 scopus 로고
    • Characterizing mechanisms of hepatic bile acid transport utilizing isolated membrane vesicles
    • Boyer JL, Meier PJ. Characterizing mechanisms of hepatic bile acid transport utilizing isolated membrane vesicles. Methods Enzymol 1990; 192:517-533.
    • (1990) Methods Enzymol , vol.192 , pp. 517-533
    • Boyer, J.L.1    Meier, P.J.2
  • 19
    • 0025613205 scopus 로고
    • Isolation of lumenal and contralumenal plasma membrane vesicles from kidney
    • Kinne-Saffran E, Kinne RK. Isolation of lumenal and contralumenal plasma membrane vesicles from kidney. Methods Enzymol 1990; 191:450-469.
    • (1990) Methods Enzymol , vol.191 , pp. 450-469
    • Kinne-Saffran, E.1    Kinne, R.K.2
  • 20
    • 0025606834 scopus 로고
    • Preparation of basolateral (sinusoidal) and canalicular plasma membrane vesicles for the study of hepatic transport processes
    • Meier PJ, Boyer JL. Preparation of basolateral (sinusoidal) and canalicular plasma membrane vesicles for the study of hepatic transport processes. Methods Enzymol 1990; 192:534-545.
    • (1990) Methods Enzymol , vol.192 , pp. 534-545
    • Meier, P.J.1    Boyer, J.L.2
  • 21
    • 0021250574 scopus 로고
    • St. Meier-Abt A, Barrett C, et al. Mechanisms of taurocholate transport in canalicular and basolateral rat liver plasma membrane vesicles. Evidence for an electrogenic canalicular organic anion carrier
    • Meier PJ, St. Meier-Abt A, Barrett C, et al. Mechanisms of taurocholate transport in canalicular and basolateral rat liver plasma membrane vesicles. Evidence for an electrogenic canalicular organic anion carrier. J Biol Chem 1984; 259:10614-10622.
    • (1984) J Biol Chem , vol.259 , pp. 10614-10622
    • Meier, P.J.1
  • 22
    • 0024371025 scopus 로고
    • The function of Gpl70, the multidrug resistance gene product, in rat liver canalicular membrane vesicles
    • Kamimoto Y, Gatmaitan Z, Hsu J, et al. The function of Gpl70, the multidrug resistance gene product, in rat liver canalicular membrane vesicles. J Biol Chem 1989; 264:11693-11698.
    • (1989) J Biol Chem , vol.264 , pp. 11693-11698
    • Kamimoto, Y.1    Gatmaitan, Z.2    Hsu, J.3
  • 23
    • 0015615109 scopus 로고
    • The distribution of p-aminohippuric acid in rat kidney slices. I. Tubular localization
    • Wedeen RP, Weiner B. The distribution of p-aminohippuric acid in rat kidney slices. I. Tubular localization. Kidney Int 1973; 3:205-213.
    • (1973) Kidney Int , vol.3 , pp. 205-213
    • Wedeen, R.P.1    Weiner, B.2
  • 24
    • 0036175127 scopus 로고    scopus 로고
    • Functional involvement of rat organic anion transporter 3 (rOat3; Slc22a8) in the renal uptake of organic anions
    • Hasegawa M, Kusuhara H, Sugiyama D, et al. Functional involvement of rat organic anion transporter 3 (rOat3; Slc22a8) in the renal uptake of organic anions. J Pharmacol Exp Ther 2002; 300:746-753.
    • (2002) J Pharmacol Exp Ther , vol.300 , pp. 746-753
    • Hasegawa, M.1    Kusuhara, H.2    Sugiyama, D.3
  • 25
    • 0034512016 scopus 로고    scopus 로고
    • Ex vivo stimulation of renal tubular PAH transport by dexamethasone and triiodothyronine in human renal cell carcinoma
    • Fleck C, Bachner B, Gockeritz S, et al. Ex vivo stimulation of renal tubular PAH transport by dexamethasone and triiodothyronine in human renal cell carcinoma. Urol Res 2000; 28:383-390.
    • (2000) Urol Res , vol.28 , pp. 383-390
    • Fleck, C.1    Bachner, B.2    Gockeritz, S.3
  • 26
    • 0030981071 scopus 로고    scopus 로고
    • Tubular PAH transport capacity in human kidney tissue and in renal cell carcinoma: Correlation with various clinical and morphological parameters of the tumor
    • Reck C, Gockeritz S, Schubert J. Tubular PAH transport capacity in human kidney tissue and in renal cell carcinoma: correlation with various clinical and morphological parameters of the tumor. Urol Res 1997; 25:167-171.
    • (1997) Urol Res , vol.25 , pp. 167-171
    • Reck, C.1    Gockeritz, S.2    Schubert, J.3
  • 27
    • 0036924482 scopus 로고    scopus 로고
    • Ex vivo stimulation of renal transport of the cytostatic drugs methotrexate, cisplatin, topotecan (Hycamtin) and raltitrexed (Tomudex) by dexamethasone, T3 and EGF in intact human and rat kidney tissue and in human renal cell carcinoma
    • Reck C, Hilger R, Jurkutat S, et al. Ex vivo stimulation of renal transport of the cytostatic drugs methotrexate, cisplatin, topotecan (Hycamtin) and raltitrexed (Tomudex) by dexamethasone, T3 and EGF in intact human and rat kidney tissue and in human renal cell carcinoma. Urol Res 2002; 30:256-262.
    • (2002) Urol Res , vol.30 , pp. 256-262
    • Reck, C.1    Hilger, R.2    Jurkutat, S.3
  • 28
    • 33947426344 scopus 로고    scopus 로고
    • Characterization of the uptake of OAT1 and OAT3 substrates by human kidney slices
    • Nozaki Y, Kusuhara H, Kondo T, et al. Characterization of the uptake of OAT1 and OAT3 substrates by human kidney slices. J Pharmacol Exp Ther 2007; 321:362-369.
    • (2007) J Pharmacol Exp Ther , vol.321 , pp. 362-369
    • Nozaki, Y.1    Kusuhara, H.2    Kondo, T.3
  • 29
    • 6544259277 scopus 로고
    • The use of sacs of everted small intestine for the study of the transference of substances from the mucosal to serosal surface
    • Wilson T, Wiselman G. The use of sacs of everted small intestine for the study of the transference of substances from the mucosal to serosal surface. J Physiol 1954; 123:116-125.
    • (1954) J Physiol , vol.123 , pp. 116-125
    • Wilson, T.1    Wiselman, G.2
  • 30
    • 0014734572 scopus 로고
    • Intestinal drug absorption and metabolism. II. Kinetic aspects of intestinal glucuronide conjugation
    • Barr WH, Riegelman S. Intestinal drug absorption and metabolism. II. Kinetic aspects of intestinal glucuronide conjugation. J Pharm Sci 1970; 59:164-168.
    • (1970) J Pharm Sci , vol.59 , pp. 164-168
    • Barr, W.H.1    Riegelman, S.2
  • 31
    • 0017066737 scopus 로고
    • Ion transport by rabbit colon. I. Active and passive components
    • Frizzell RA, Koch MJ, Schultz SG. Ion transport by rabbit colon. I. Active and passive components. J Membr Biol 1976; 27:297-316.
    • (1976) J Membr Biol , vol.27 , pp. 297-316
    • Frizzell, R.A.1    Koch, M.J.2    Schultz, S.G.3
  • 32
    • 0015311097 scopus 로고
    • Schultz SG. Ionic conductances of extracellular shunt pathway in rabbit ileum. Influence of shunt on transmural sodium transport and electrical potential differences
    • Frizzell RA, Schultz SG. Ionic conductances of extracellular shunt pathway in rabbit ileum. Influence of shunt on transmural sodium transport and electrical potential differences. J Gen Physiol 1972; 59:318-346.
    • (1972) J Gen Physiol , vol.59 , pp. 318-346
    • Frizzell, R.A.1
  • 33
    • 0025046860 scopus 로고
    • Uptake of the cephalosporin, cephalexin, by a dipeptide transport carrier in the human intestinal cell line, Caco-2
    • Dantzig AH, Bergin L. Uptake of the cephalosporin, cephalexin, by a dipeptide transport carrier in the human intestinal cell line, Caco-2. Biochim Biophys Acta 1990; 1027:211-217.
    • (1990) Biochim Biophys Acta , vol.1027 , pp. 211-217
    • Dantzig, A.H.1    Bergin, L.2
  • 34
    • 0025332938 scopus 로고
    • Cell culture techniques for the study of drug transport
    • Wilson G. Cell culture techniques for the study of drug transport. Eur J Drug Metab Pharmacokinet 1990; 15:159-163.
    • (1990) Eur J Drug Metab Pharmacokinet , vol.15 , pp. 159-163
    • Wilson, G.1
  • 35
    • 0033278620 scopus 로고    scopus 로고
    • Gastrointestinal absorption of drugs: Methods and studies
    • Barthe L, Woodley J, Houin G. Gastrointestinal absorption of drugs: methods and studies. Fundam Clin Pharmacol 1999; 13:154-168.
    • (1999) Fundam Clin Pharmacol , vol.13 , pp. 154-168
    • Barthe, L.1    Woodley, J.2    Houin, G.3
  • 36
    • 0028786481 scopus 로고
    • The human intestinal epithelial cell line Caco-2; pharmacological and pharmacokinetic applications
    • Meunier V, Bourne M, Berger Y, et al. The human intestinal epithelial cell line Caco-2; pharmacological and pharmacokinetic applications. Cell Biol Toxicol 1995; 11:187-194.
    • (1995) Cell Biol Toxicol , vol.11 , pp. 187-194
    • Meunier, V.1    Bourne, M.2    Berger, Y.3
  • 37
    • 0031780682 scopus 로고    scopus 로고
    • Correlation between oral drug absorption in humans, and apparent drug permeability in TC-7 cells, a human epithelial intestinal cell line: Comparison with the parental Caco-2 cell line
    • Gres MC, Julian B, Bourrie M, et al. Correlation between oral drug absorption in humans, and apparent drug permeability in TC-7 cells, a human epithelial intestinal cell line: comparison with the parental Caco-2 cell line. Pharm Res 1998; 15:726-733.
    • (1998) Pharm Res , vol.15 , pp. 726-733
    • Gres, M.C.1    Julian, B.2    Bourrie, M.3
  • 38
    • 0031941496 scopus 로고    scopus 로고
    • Development and utility of anti-PepTl antipeptide polyclonal antibodies
    • Basu SK, Shen J, Elbert KJ, et al. Development and utility of anti-PepTl antipeptide polyclonal antibodies. Pharm Res 1998; 15:338-342.
    • (1998) Pharm Res , vol.15 , pp. 338-342
    • Basu, S.K.1    Shen, J.2    Elbert, K.J.3
  • 39
    • 0030761681 scopus 로고    scopus 로고
    • Molecular and functional characterization of intestinal Na(-b)-dependent neutral amino acid transporter BO
    • Kekuda R, Torres-Zamorano V, Fei YJ, et al. Molecular and functional characterization of intestinal Na(-b)-dependent neutral amino acid transporter BO. Am J Physiol 1997; 272:G1463-G1472.
    • (1997) Am J Physiol , vol.272 , pp. G1463-G1472
    • Kekuda, R.1    Torres-Zamorano, V.2    Fei, Y.J.3
  • 40
    • 0029562420 scopus 로고
    • Proton-cotransport of pravastatin across intestinal brush-border membrane
    • Tamai I, Takanaga H, Maeda H, et al. Proton-cotransport of pravastatin across intestinal brush-border membrane. Pharm Res 1995; 12:1727-1732.
    • (1995) Pharm Res , vol.12 , pp. 1727-1732
    • Tamai, I.1    Takanaga, H.2    Maeda, H.3
  • 41
    • 0027181852 scopus 로고
    • Functional expression of P-glycoprotein in apical membranes of human intestinal Caco-2 cells. Kinetics of vinblastine secretion and interaction with modulators
    • Hunter J, Jepson MA, Tsuruo T, et al. Functional expression of P-glycoprotein in apical membranes of human intestinal Caco-2 cells. Kinetics of vinblastine secretion and interaction with modulators. J Biol Chem 1993; 268:14991-14997.
    • (1993) J Biol Chem , vol.268 , pp. 14991-14997
    • Hunter, J.1    Jepson, M.A.2    Tsuruo, T.3
  • 42
    • 0033985306 scopus 로고    scopus 로고
    • Function and expression of multidrug resistance-associated protein family in human colon adenocarcinoma cells (Caco-2)
    • Hirohashi T, Suzuki H, Chu XY, et al. Function and expression of multidrug resistance-associated protein family in human colon adenocarcinoma cells (Caco-2). J Pharmacol Exp Ther 2000; 292:265-270.
    • (2000) J Pharmacol Exp Ther , vol.292 , pp. 265-270
    • Hirohashi, T.1    Suzuki, H.2    Chu, X.Y.3
  • 43
    • 17844383698 scopus 로고    scopus 로고
    • Expression, localization, and functional characteristics of breast cancer resistance protein in Caco-2 cells
    • Xia CQ, Liu N, Yang D, et al. Expression, localization, and functional characteristics of breast cancer resistance protein in Caco-2 cells. Drug Metab Dispos 2005; 33:637-643.
    • (2005) Drug Metab Dispos , vol.33 , pp. 637-643
    • Xia, C.Q.1    Liu, N.2    Yang, D.3
  • 44
    • 0027180795 scopus 로고
    • Drug absorption limited by P-glycoproteinmediated secretory drug transport in human intestinal epithelial Caco-2 cell layer
    • Hunter J, Hirst BH, Simmons NL. Drug absorption limited by P-glycoproteinmediated secretory drug transport in human intestinal epithelial Caco-2 cell layer. Pharm Res 1993; 10:743-749.
    • (1993) Pharm Res , vol.10 , pp. 743-749
    • Hunter, J.1    Hirst, B.H.2    Simmons, N.L.3
  • 45
    • 0026669192 scopus 로고
    • Functional involvement of P-glycoprotein in blood-brain barrier
    • Tatsuta T, Naito M, Oh-hara T, et al. Functional involvement of P-glycoprotein in blood-brain barrier. J Biol Chem 1992; 267:20383-20391.
    • (1992) J Biol Chem , vol.267 , pp. 20383-20391
    • Tatsuta, T.1    Naito, M.2    Oh-Hara, T.3
  • 46
    • 0034452792 scopus 로고    scopus 로고
    • mRNA expression and transport characterization of conditionally immortalized rat brain capillary endothelial cell lines; a new in vitro BBB model for drug targeting
    • Hosoya KI, Takashima T, Tetsuka K, et al. mRNA expression and transport characterization of conditionally immortalized rat brain capillary endothelial cell lines; a new in vitro BBB model for drug targeting. J Drug Target 2000; 8:357-370.
    • (2000) J Drug Target , vol.8 , pp. 357-370
    • Hosoya, K.I.1    Takashima, T.2    Tetsuka, K.3
  • 47
    • 0028892249 scopus 로고
    • Comparisons of P-glycoprotein expression in isolated rat brain microvessels and in primary cultures of endothelial cells derived from microvasculature of rat brain, epididymal fat pad and from aorta
    • Barrand MA, Robertson KJ, von Weikersthal SF. Comparisons of P-glycoprotein expression in isolated rat brain microvessels and in primary cultures of endothelial cells derived from microvasculature of rat brain, epididymal fat pad and from aorta. FEBS Lett 1995; 374:179-183.
    • (1995) FEBS Lett , vol.374 , pp. 179-183
    • Barrand, M.A.1    Robertson, K.J.2    von Weikersthal, S.F.3
  • 48
    • 0031891475 scopus 로고    scopus 로고
    • Multidrug resistance-related transport proteins in isolated human brain microvessels and in cells cultured from these isolates
    • Seetharaman S, Barrand MA, Maskell L, et al. Multidrug resistance-related transport proteins in isolated human brain microvessels and in cells cultured from these isolates. J Neurochem 1998; 70:1151-1159.
    • (1998) J Neurochem , vol.70 , pp. 1151-1159
    • Seetharaman, S.1    Barrand, M.A.2    Maskell, L.3
  • 49
    • 0031724003 scopus 로고    scopus 로고
    • Mrpl multidrug resistance-associated protein and P-glycoprotein expression in rat brain micro vessel endothelial cells
    • Regina A, Koman A, Piciotti M, et al. Mrpl multidrug resistance-associated protein and P-glycoprotein expression in rat brain micro vessel endothelial cells. J Neurochem 1998; 71:705-715.
    • (1998) J Neurochem , vol.71 , pp. 705-715
    • Regina, A.1    Koman, A.2    Piciotti, M.3
  • 50
    • 0032422456 scopus 로고    scopus 로고
    • Contribution of sodium taurocholate cotransporting polypeptide to the uptake of its possible substrates into rat hepatocytes
    • Kouzuki H, Suzuki H, Ito K, et al. Contribution of sodium taurocholate cotransporting polypeptide to the uptake of its possible substrates into rat hepatocytes. J Pharmacol Exp Ther 1998; 286:1043-1050.
    • (1998) J Pharmacol Exp Ther , vol.286 , pp. 1043-1050
    • Kouzuki, H.1    Suzuki, H.2    Ito, K.3
  • 51
    • 0033063815 scopus 로고    scopus 로고
    • Contribution of organic anion transporting polypeptide to uptake of its possible substrates into rat hepatocytes
    • Kouzuki H, Suzuki H, Ito K, et al. Contribution of organic anion transporting polypeptide to uptake of its possible substrates into rat hepatocytes. J Pharmacol Exp Ther 1999; 288:627-634.
    • (1999) J Pharmacol Exp Ther , vol.288 , pp. 627-634
    • Kouzuki, H.1    Suzuki, H.2    Ito, K.3
  • 52
    • 33745243715 scopus 로고    scopus 로고
    • Drug-drug interaction between pitavastatin and various drugs via OATP1B1
    • Hirano M, Maeda K, Shitara Y, et al. Drug-drug interaction between pitavastatin and various drugs via OATP1B1. Drug Metab Dispos 2006; 34:1229-1236.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1229-1236
    • Hirano, M.1    Maeda, K.2    Shitara, Y.3
  • 53
    • 0038137530 scopus 로고    scopus 로고
    • Contribution of organic anion transporters to the renal uptake of anionic compounds and nucleoside derivatives in rat
    • Hasegawa M, Kusuhara H, Endou H, et al. Contribution of organic anion transporters to the renal uptake of anionic compounds and nucleoside derivatives in rat. J Pharmacol Exp Ther 2003; 305:1087-1097.
    • (2003) J Pharmacol Exp Ther , vol.305 , pp. 1087-1097
    • Hasegawa, M.1    Kusuhara, H.2    Endou, H.3
  • 54
    • 0037155204 scopus 로고    scopus 로고
    • Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and multidrug resistance-associated protein 2 (MRP2/ABCC2)
    • Sasaki M, Suzuki H, Ito K, et al. Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and multidrug resistance-associated protein 2 (MRP2/ABCC2). J Biol Chem 2002; 277:6497-6503.
    • (2002) J Biol Chem , vol.277 , pp. 6497-6503
    • Sasaki, M.1    Suzuki, H.2    Ito, K.3
  • 55
    • 25144494314 scopus 로고    scopus 로고
    • Human hepatobiliary transport of organic anions analyzed by quadruple-transfected cells
    • Kopplow K, Letschert K, Konig J, et al. Human hepatobiliary transport of organic anions analyzed by quadruple-transfected cells. Mol Pharmacol 2005; 68:1031-1038.
    • (2005) Mol Pharmacol , vol.68 , pp. 1031-1038
    • Kopplow, K.1    Letschert, K.2    Konig, J.3
  • 56
    • 22944446686 scopus 로고    scopus 로고
    • Identification of the hepatic efflux transporters of organic anions using double-transfected Madin-Darby canine kidney II cells expressing human organic anion-transporting polypeptide 1B1 (OATPlBl)/multidrug resistance-associated protein 2, OATPlBl/multidrug resistance 1, and OATP1B1/ breast cancer resistance protein
    • Matsushima S, Maeda K, Kondo C, et al. Identification of the hepatic efflux transporters of organic anions using double-transfected Madin-Darby canine kidney II cells expressing human organic anion-transporting polypeptide 1B1 (OATPlBl)/multidrug resistance-associated protein 2, OATPlBl/multidrug resistance 1, and OATP1B1/ breast cancer resistance protein. J Pharmacol Exp Ther 2005; 314:1059-1067.
    • (2005) J Pharmacol Exp Ther , vol.314 , pp. 1059-1067
    • Matsushima, S.1    Maeda, K.2    Kondo, C.3
  • 57
    • 0034753042 scopus 로고    scopus 로고
    • Vectorial transport by double-transfected cells expressing the human uptake transporter SLC21A8 and the apical export pump ABCC2
    • Cui Y, Konig J, Keppler D. Vectorial transport by double-transfected cells expressing the human uptake transporter SLC21A8 and the apical export pump ABCC2. Mol Pharmacol 2001; 60:934-943.
    • (2001) Mol Pharmacol , vol.60 , pp. 934-943
    • Cui, Y.1    Konig, J.2    Keppler, D.3
  • 58
    • 33845930051 scopus 로고    scopus 로고
    • Organic anion transporting polypeptide 2B1 and breast cancer resistance protein interact in the transepithelial transport of steroid sulfates in human placenta
    • Grube M, Reuther S, Meyer Zu, et al. Organic anion transporting polypeptide 2B1 and breast cancer resistance protein interact in the transepithelial transport of steroid sulfates in human placenta. Drug Metab Dispos 2007; 35:30-35.
    • (2007) Drug Metab Dispos , vol.35 , pp. 30-35
    • Grube, M.1    Reuther, S.2    Meyer, Z.3
  • 59
    • 4944225107 scopus 로고    scopus 로고
    • Prediction of in vivo biliary clearance from the in vitro transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II monolayer expressing both rat organic anion transporting polypeptide 4 and multidrug resistance associated protein 2
    • Sasaki M, Suzuki H, Aoki J, et al. Prediction of in vivo biliary clearance from the in vitro transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II monolayer expressing both rat organic anion transporting polypeptide 4 and multidrug resistance associated protein 2. Mol Pharmacol 2004; 66:450-459.
    • (2004) Mol Pharmacol , vol.66 , pp. 450-459
    • Sasaki, M.1    Suzuki, H.2    Aoki, J.3
  • 60
    • 10844270655 scopus 로고    scopus 로고
    • Vectorial transport of bile salts across MDCK cells expressing both rat Na+-taurocholate cotransporting polypeptide and rat bile salt export pump
    • Mita S, Suzuki H, Akita H, et al. Vectorial transport of bile salts across MDCK cells expressing both rat Na+-taurocholate cotransporting polypeptide and rat bile salt export pump. Am J Physiol Gastrointest Liver Physiol 2005; 288: G159-G167.
    • (2005) Am J Physiol Gastrointest Liver Physiol , vol.288 , pp. G159-G167
    • Mita, S.1    Suzuki, H.2    Akita, H.3
  • 61
    • 33644980997 scopus 로고    scopus 로고
    • Vectorial transport of unconjugated and conjugated bile salts by monolayers of LLC-PK1 cells doubly transfected with human NTCP and BSEP or with rat Ntcp and Bsep
    • Mita S, Suzuki H, Akita H, et al. Vectorial transport of unconjugated and conjugated bile salts by monolayers of LLC-PK1 cells doubly transfected with human NTCP and BSEP or with rat Ntcp and Bsep. Am J Physiol Gastrointest Liver Physiol 2006; 290:G550-G556.
    • (2006) Am J Physiol Gastrointest Liver Physiol , vol.290 , pp. G550-G556
    • Mita, S.1    Suzuki, H.2    Akita, H.3
  • 62
    • 3543124167 scopus 로고    scopus 로고
    • The renal-specific transporter mediates facilitative transport of organic anions at the brush border membrane of mouse renal tubules
    • Imaoka T, Kusuhara H, Adachi-Akahane S, et al. The renal-specific transporter mediates facilitative transport of organic anions at the brush border membrane of mouse renal tubules. J Am Soc Nephrol 2004; 15:2012-2022.
    • (2004) J am Soc Nephrol , vol.15 , pp. 2012-2022
    • Imaoka, T.1    Kusuhara, H.2    Adachi-Akahane, S.3
  • 63
    • 14844291348 scopus 로고    scopus 로고
    • The heteromeric organic solute transporter alpha-beta, Ostalpha-Ostbeta, is an ileal basolateral bile acid transporter
    • Dawson PA, Hubbert M, Haywood J, et al. The heteromeric organic solute transporter alpha-beta, Ostalpha-Ostbeta, is an ileal basolateral bile acid transporter. J Biol Chem 2005; 280:6960-6968.
    • (2005) J Biol Chem , vol.280 , pp. 6960-6968
    • Dawson, P.A.1    Hubbert, M.2    Haywood, J.3
  • 64
    • 0037427972 scopus 로고    scopus 로고
    • The superfamily of organic anion transporting polypeptides
    • Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003; 1609:1-18.
    • (2003) Biochim Biophys Acta , vol.1609 , pp. 1-18
    • Hagenbuch, B.1    Meier, P.J.2
  • 65
    • 0028140393 scopus 로고
    • Expression cloning of a rat liver Na(+)-independent organic anion transporter
    • Jacquemin E, Hagenbuch B, Stieger B, et al. Expression cloning of a rat liver Na(+)-independent organic anion transporter. Proc Natl Acad Sci U S A 1994; 91:133-137.
    • (1994) Proc Natl Acad Sci U S A , vol.91 , pp. 133-137
    • Jacquemin, E.1    Hagenbuch, B.2    Stieger, B.3
  • 66
    • 16044374493 scopus 로고    scopus 로고
    • Immunologic distribution of an organic anion transport protein in rat liver and kidney
    • Bergwerk AJ, Shi X, Ford AC, et al. Immunologic distribution of an organic anion transport protein in rat liver and kidney. Am J Physiol 1996; 271:G231-G238.
    • (1996) Am J Physiol , vol.271 , pp. G231-G238
    • Bergwerk, A.J.1    Shi, X.2    Ford, A.C.3
  • 67
    • 0032825736 scopus 로고    scopus 로고
    • Poly specific organic anion transporting polypeptides mediate hepatic uptake of amphipathic type II organic cations
    • van Montfoort JE, Hagenbuch B, Fattinger KE, et al. Poly specific organic anion transporting polypeptides mediate hepatic uptake of amphipathic type II organic cations. J Pharmacol Exp Ther 1999; 291:147-152.
    • (1999) J Pharmacol Exp Ther , vol.291 , pp. 147-152
    • van Montfoort, J.E.1    Hagenbuch, B.2    Fattinger, K.E.3
  • 68
    • 0032568838 scopus 로고    scopus 로고
    • Identification of glutathione as a driving force and leukotriene C4 as a substrate for oatpl, the hepatic sinusoidal organic solute transporter
    • Li L, Lee TK, Meier PJ, et al. Identification of glutathione as a driving force and leukotriene C4 as a substrate for oatpl, the hepatic sinusoidal organic solute transporter. J Biol Chem 1998; 273:16184-16191.
    • (1998) J Biol Chem , vol.273 , pp. 16184-16191
    • Li, L.1    Lee, T.K.2    Meier, P.J.3
  • 69
    • 0029786681 scopus 로고    scopus 로고
    • Cloning and functional characterization of a novel rat organic anion transporter mediating basolateral uptake of methotrexate in the kidney
    • Saito H, Masuda S, Inui K. Cloning and functional characterization of a novel rat organic anion transporter mediating basolateral uptake of methotrexate in the kidney. J Biol Chem 1996; 271:20719-20725.
    • (1996) J Biol Chem , vol.271 , pp. 20719-20725
    • Saito, H.1    Masuda, S.2    Inui, K.3
  • 70
    • 0032943652 scopus 로고    scopus 로고
    • Cloning and functional characterization of a new multispecific organic anion transporter, OAT-K2, in rat kidney
    • Masuda S, Ibaramoto K, Takeuchi A, et al. Cloning and functional characterization of a new multispecific organic anion transporter, OAT-K2, in rat kidney. Mol Pharmacol 1999; 55:743-752.
    • (1999) Mol Pharmacol , vol.55 , pp. 743-752
    • Masuda, S.1    Ibaramoto, K.2    Takeuchi, A.3
  • 71
    • 0031589135 scopus 로고    scopus 로고
    • mRNA distribution and membrane localization of the OAT-K1 organic anion transporter in rat renal tubules
    • Masuda S, Saito H, Nonoguchi H, et al. mRNA distribution and membrane localization of the OAT-K1 organic anion transporter in rat renal tubules. FEBS Lett 1997; 407:127-131.
    • (1997) FEBS Lett , vol.407 , pp. 127-131
    • Masuda, S.1    Saito, H.2    Nonoguchi, H.3
  • 72
    • 0030886124 scopus 로고    scopus 로고
    • Isolation of a multispecific organic anion and cardiac glycoside transporter from rat brain
    • Noe B, Hagenbuch B, Stieger B, et al. Isolation of a multispecific organic anion and cardiac glycoside transporter from rat brain. Proc Natl Acad Sci U S A 1997; 94:10346-10350.
    • (1997) Proc Natl Acad Sci U S A , vol.94 , pp. 10346-10350
    • Noe, B.1    Hagenbuch, B.2    Stieger, B.3
  • 73
    • 15444342300 scopus 로고    scopus 로고
    • Molecular characterization and tissue distribution of a new organic anion transporter subtype (oatp3) that transports thyroid hormones and taurocholate and comparison with oatp2
    • Abe T, Kakyo M, Sakagami H, et al. Molecular characterization and tissue distribution of a new organic anion transporter subtype (oatp3) that transports thyroid hormones and taurocholate and comparison with oatp2. J Biol Chem 1998; 273:22395-22401.
    • (1998) J Biol Chem , vol.273 , pp. 22395-22401
    • Abe, T.1    Kakyo, M.2    Sakagami, H.3
  • 74
    • 22344452137 scopus 로고    scopus 로고
    • Tissue distribution and ontogeny of mouse organic anion transporting polypeptides (Oatps)
    • Cheng X, Maher J, Chen C, et al. Tissue distribution and ontogeny of mouse organic anion transporting polypeptides (Oatps). Drug Metab Dispos 2005; 33:1062-1073.
    • (2005) Drug Metab Dispos , vol.33 , pp. 1062-1073
    • Cheng, X.1    Maher, J.2    Chen, C.3
  • 75
    • 0032819955 scopus 로고    scopus 로고
    • Localization and function of the organic anion-transporting polypeptide Oatp2 in rat liver
    • Reichel C, Gao B, Van Montfoort J, et al. Localization and function of the organic anion-transporting polypeptide Oatp2 in rat liver. Gastroenterology 1999; 117: 688-695.
    • (1999) Gastroenterology , vol.117 , pp. 688-695
    • Reichel, C.1    Gao, B.2    van Montfoort, J.3
  • 76
    • 0032859340 scopus 로고    scopus 로고
    • Localization of the organic anion transporting polypeptide 2 (Oatp2) in capillary endothelium and choroid plexus epithelium of rat brain
    • Gao B, Stieger B, Noe B, et al. Localization of the organic anion transporting polypeptide 2 (Oatp2) in capillary endothelium and choroid plexus epithelium of rat brain. J Histochem Cytochem 1999; 47:1255-1264.
    • (1999) J Histochem Cytochem , vol.47 , pp. 1255-1264
    • Gao, B.1    Stieger, B.2    Noe, B.3
  • 77
    • 0033932502 scopus 로고    scopus 로고
    • Organic anion-transporting polypeptides mediate transport of opioid peptides across blood-brain barrier
    • Gao B, Hagenbuch B, Kullak-Ublick GA, et al. Organic anion-transporting polypeptides mediate transport of opioid peptides across blood-brain barrier. J Pharmacol Exp Ther 2000; 294:73-79.
    • (2000) J Pharmacol Exp Ther , vol.294 , pp. 73-79
    • Gao, B.1    Hagenbuch, B.2    Kullak-Ublick, G.A.3
  • 78
    • 0032437785 scopus 로고    scopus 로고
    • Efflux of taurocholic acid across the bloodbrain barrier: Interaction with cyclic peptides
    • Kitazawa T, Terasaki T, Suzuki H, et al. Efflux of taurocholic acid across the bloodbrain barrier: interaction with cyclic peptides. J Pharmacol Exp Ther 1998; 286:890-895.
    • (1998) J Pharmacol Exp Ther , vol.286 , pp. 890-895
    • Kitazawa, T.1    Terasaki, T.2    Suzuki, H.3
  • 79
    • 0034978374 scopus 로고    scopus 로고
    • Characterization of the efflux transport of 17beta-estradiol-D-17beta- glucuronide from the brain across the blood-brain barrier
    • Sugiyama D, Kusuhara H, Shitara Y, et al. Characterization of the efflux transport of 17beta-estradiol-D-17beta- glucuronide from the brain across the blood-brain barrier. J Pharmacol Exp Ther 2001; 298:316-322.
    • (2001) J Pharmacol Exp Ther , vol.298 , pp. 316-322
    • Sugiyama, D.1    Kusuhara, H.2    Shitara, Y.3
  • 80
    • 0033794507 scopus 로고    scopus 로고
    • Blood-brain barrier is involved in the efflux transport of a neuroactive steroid, dehydroepiandrosterone sulfate, via organic anion transporting polypeptide 2
    • Asaba H, Hosoya K, Takanaga H, et al. Blood-brain barrier is involved in the efflux transport of a neuroactive steroid, dehydroepiandrosterone sulfate, via organic anion transporting polypeptide 2. J Neurochem 2000; 75:1907-1916.
    • (2000) J Neurochem , vol.75 , pp. 1907-1916
    • Asaba, H.1    Hosoya, K.2    Takanaga, H.3
  • 81
    • 10044219529 scopus 로고    scopus 로고
    • Involvement of multiple transporters in the efflux of 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors across the bloodbrain barrier
    • Kikuchi R, Kusuhara H, Abe T, et al. Involvement of multiple transporters in the efflux of 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors across the bloodbrain barrier. J Pharmacol Exp Ther 2004; 311:1147-1153.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 1147-1153
    • Kikuchi, R.1    Kusuhara, H.2    Abe, T.3
  • 82
    • 0034874453 scopus 로고    scopus 로고
    • Effect of mdrla P-glycoprotein gene disruption, gender, and substrate concentration on brain uptake of selected compounds
    • Dagenais C, Zong J, Ducharme J, et al. Effect of mdrla P-glycoprotein gene disruption, gender, and substrate concentration on brain uptake of selected compounds. Pharm Res 2001; 18:957-963.
    • (2001) Pharm Res , vol.18 , pp. 957-963
    • Dagenais, C.1    Zong, J.2    Ducharme, J.3
  • 83
    • 0036229943 scopus 로고    scopus 로고
    • Tissue expression, ontogeny, and inducibility of rat organic anion transporting polypeptide 4
    • Li N, Hartley DP, Cherrington NJ, et al. Tissue expression, ontogeny, and inducibility of rat organic anion transporting polypeptide 4. J Pharmacol Exp Ther 2002; 301:551-560.
    • (2002) J Pharmacol Exp Ther , vol.301 , pp. 551-560
    • Li, N.1    Hartley, D.P.2    Cherrington, N.J.3
  • 84
    • 0037406304 scopus 로고    scopus 로고
    • Expression and functional involvement of organic anion transporting polypeptide subtype 3 (Slc21a7) in rat choroid plexus
    • Kusuhara H, He Z, Nagata Y, et al. Expression and functional involvement of organic anion transporting polypeptide subtype 3 (Slc21a7) in rat choroid plexus. Pharm Res 2003; 20:720-727.
    • (2003) Pharm Res , vol.20 , pp. 720-727
    • Kusuhara, H.1    He, Z.2    Nagata, Y.3
  • 85
    • 0033638544 scopus 로고    scopus 로고
    • Expression, transport properties, and chromosomal location of organic anion transporter subtype 3
    • Walters HC, Craddock AL, Fusegawa H, et al. Expression, transport properties, and chromosomal location of organic anion transporter subtype 3. Am J Physiol Gastrointest Liver Physiol 2000; 279:G1188-G1200.
    • (2000) Am J Physiol Gastrointest Liver Physiol , vol.279 , pp. G1188-G1200
    • Walters, H.C.1    Craddock, A.L.2    Fusegawa, H.3
  • 86
    • 3342999996 scopus 로고    scopus 로고
    • Localization of organic anion transporting polypeptide 3 (oatp3) in mouse brain parenchymal and capillary endothelial cells
    • Ohtsuki S, Takizawa T, Takanaga H, et al. Localization of organic anion transporting polypeptide 3 (oatp3) in mouse brain parenchymal and capillary endothelial cells. J Neurochem 2004; 90:743-749.
    • (2004) J Neurochem , vol.90 , pp. 743-749
    • Ohtsuki, S.1    Takizawa, T.2    Takanaga, H.3
  • 87
    • 0035188883 scopus 로고    scopus 로고
    • Localization of organic anion transporting polypeptide 4 (Oatp4) in rat liver and comparison of its substrate specificity with Oatpl, Oatp2 and Oatp3
    • Cattori V, van Montfoort JE, Stieger B, et al. Localization of organic anion transporting polypeptide 4 (Oatp4) in rat liver and comparison of its substrate specificity with Oatpl, Oatp2 and Oatp3. Pflugers Arch 2001; 443:188-195.
    • (2001) Pflugers Arch , vol.443 , pp. 188-195
    • Cattori, V.1    van Montfoort, J.E.2    Stieger, B.3
  • 88
    • 34548628779 scopus 로고    scopus 로고
    • Transporter-mediated intestinal absorption of fexofenadine in rats
    • Kikuchi A, Nozawa T, Wakasawa T, et al. Transporter-mediated intestinal absorption of fexofenadine in rats. Drug Metab Pharmacokinet 2006; 21:308-314.
    • (2006) Drug Metab Pharmacokinet , vol.21 , pp. 308-314
    • Kikuchi, A.1    Nozawa, T.2    Wakasawa, T.3
  • 89
    • 0029091022 scopus 로고
    • Molecular and functional characterization of an organic anion transporting polypeptide cloned from human liver
    • Kullak-Ublick GA, Hagenbuch B, Stieger B, et al. Molecular and functional characterization of an organic anion transporting polypeptide cloned from human liver. Gastroenterology 1995; 109:1274-1282.
    • (1995) Gastroenterology , vol.109 , pp. 1274-1282
    • Kullak-Ublick, G.A.1    Hagenbuch, B.2    Stieger, B.3
  • 90
    • 0040368659 scopus 로고    scopus 로고
    • Identification of a novel gene family encoding human liver-specific organic anion transporter LST-1
    • Abe T, Kakyo M, Tokui T, et al. Identification of a novel gene family encoding human liver-specific organic anion transporter LST-1. J Biol Chem 1999; 274:17159-17163.
    • (1999) J Biol Chem , vol.274 , pp. 17159-17163
    • Abe, T.1    Kakyo, M.2    Tokui, T.3
  • 91
    • 0036827601 scopus 로고    scopus 로고
    • Transporter gene expression in lactating and nonlactating human mammary epithelial cells using real-time reverse transcriptionpolymerase chain reaction
    • Alcorn J, Lu X, Moscow JA, et al. Transporter gene expression in lactating and nonlactating human mammary epithelial cells using real-time reverse transcriptionpolymerase chain reaction. J Pharmacol Exp Ther 2002; 303:487-496.
    • (2002) J Pharmacol Exp Ther , vol.303 , pp. 487-496
    • Alcorn, J.1    Lu, X.2    Moscow, J.A.3
  • 92
    • 29244441701 scopus 로고    scopus 로고
    • ABCC drug efflux pumps and organic anion uptake transporters in human gliomas and the blood-tumor barrier
    • Bronger H, Konig J, Kopplow K, et al. ABCC drug efflux pumps and organic anion uptake transporters in human gliomas and the blood-tumor barrier. Cancer Res 2005; 65:11419-11428.
    • (2005) Cancer Res , vol.65 , pp. 11419-11428
    • Bronger, H.1    Konig, J.2    Kopplow, K.3
  • 93
    • 0030296680 scopus 로고    scopus 로고
    • Multispecific amphipathic substrate transport by an organic anion transporter of human liver
    • Bossuyt X, Muller M, Meier PJ. Multispecific amphipathic substrate transport by an organic anion transporter of human liver. J Hepatol 1996; 25:733-738.
    • (1996) J Hepatol , vol.25 , pp. 733-738
    • Bossuyt, X.1    Muller, M.2    Meier, P.J.3
  • 94
    • 0037733365 scopus 로고    scopus 로고
    • A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane
    • Konig J, Cui Y, Nies AT, et al. A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane. Am J Physiol Gastrointest Liver Physiol 2000; 278:G156-G164.
    • (2000) Am J Physiol Gastrointest Liver Physiol , vol.278 , pp. G156-G164
    • Konig, J.1    Cui, Y.2    Nies, A.T.3
  • 95
    • 0033601257 scopus 로고    scopus 로고
    • A novel human hepatic organic anion transporting polypeptide (OATP2). Identification of a liver-specific human organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-CoA reductase inhibitor transporters
    • Hsiang B, Zhu Y, Wang Z, et al. A novel human hepatic organic anion transporting polypeptide (OATP2). Identification of a liver-specific human organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-CoA reductase inhibitor transporters. J Biol Chem 1999; 274:37161-37168.
    • (1999) J Biol Chem , vol.274 , pp. 37161-37168
    • Hsiang, B.1    Zhu, Y.2    Wang, Z.3
  • 96
    • 0034709561 scopus 로고    scopus 로고
    • Molecular identification and characterization of novel members of the human organic anion transporter (OATP) family
    • Tamai I, Nezu J, Uchino H, et al. Molecular identification and characterization of novel members of the human organic anion transporter (OATP) family. Biochem Biophys Res Commun 2000; 273:251-260.
    • (2000) Biochem Biophys Res Commun , vol.273 , pp. 251-260
    • Tamai, I.1    Nezu, J.2    Uchino, H.3
  • 97
    • 0034725617 scopus 로고    scopus 로고
    • Localization and genomic organization of a new hepatocellular organic anion transporting polypeptide
    • Konig J, Cui Y, Nies AT, et al. Localization and genomic organization of a new hepatocellular organic anion transporting polypeptide. J Biol Chem 2000; 275:23161-23168.
    • (2000) J Biol Chem , vol.275 , pp. 23161-23168
    • Konig, J.1    Cui, Y.2    Nies, A.T.3
  • 98
    • 0035006408 scopus 로고    scopus 로고
    • LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers
    • Abe T, Unno M, Onogawa T, et al. LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers. Gastroenterology 2001; 120:1689-1699.
    • (2001) Gastroenterology , vol.120 , pp. 1689-1699
    • Abe, T.1    Unno, M.2    Onogawa, T.3
  • 99
    • 0034595593 scopus 로고    scopus 로고
    • Identification of organic anion transporting polypeptide 4 (Oatp4) as a major full-length isoform of the liverspecific transporter-1 (rlst-1) in rat liver
    • Cattori V, Hagenbuch B, Hagenbuch N, et al. Identification of organic anion transporting polypeptide 4 (Oatp4) as a major full-length isoform of the liverspecific transporter-1 (rlst-1) in rat liver. FEBS Lett 2000; 474:242-245.
    • (2000) FEBS Lett , vol.474 , pp. 242-245
    • Cattori, V.1    Hagenbuch, B.2    Hagenbuch, N.3
  • 100
    • 0032869932 scopus 로고    scopus 로고
    • Molecular characterization and functional regulation of a novel rat liver-specific organic anion transporter rlst-1
    • Kakyo M, Unno M, Tokui T, et al. Molecular characterization and functional regulation of a novel rat liver-specific organic anion transporter rlst-1. Gastroenterology 1999; 117:770-775.
    • (1999) Gastroenterology , vol.117 , pp. 770-775
    • Kakyo, M.1    Unno, M.2    Tokui, T.3
  • 101
    • 33745253947 scopus 로고    scopus 로고
    • Predominant contribution of OATP1B3 to the hepatic uptake of telmisartan, an angiotensin II receptor antagonist, in humans
    • Ishiguro N, Maeda K, Kishimoto W, et al. Predominant contribution of OATP1B3 to the hepatic uptake of telmisartan, an angiotensin II receptor antagonist, in humans. Drug Metab Dispos 2006; 34:1109-1115.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1109-1115
    • Ishiguro, N.1    Maeda, K.2    Kishimoto, W.3
  • 102
    • 33745231761 scopus 로고    scopus 로고
    • Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II ATI-receptor, in humans
    • Yamashiro W, Maeda K, Hirouchi M, et al. Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II ATI-receptor, in humans. Drug Metab Dispos 2006; 34:1247-1254.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1247-1254
    • Yamashiro, W.1    Maeda, K.2    Hirouchi, M.3
  • 103
    • 0035971239 scopus 로고    scopus 로고
    • Hepatic uptake of bilirubin and its conjugates by the human organic anion transporter SLC21A6
    • Cui Y, Konig J, Leier I, et al. Hepatic uptake of bilirubin and its conjugates by the human organic anion transporter SLC21A6. J Biol Chem 2001; 276:9626-9630.
    • (2001) J Biol Chem , vol.276 , pp. 9626-9630
    • Cui, Y.1    Konig, J.2    Leier, I.3
  • 104
    • 0034754481 scopus 로고    scopus 로고
    • Hepatic uptake of cholecystokinin octapeptide by organic anion-transporting polypeptides OATP4 and OATP8 of rat and human liver
    • Ismair MG, Stieger B, Cattori V, et al. Hepatic uptake of cholecystokinin octapeptide by organic anion-transporting polypeptides OATP4 and OATP8 of rat and human liver. Gastroenterology 2001; 121:1185-1190.
    • (2001) Gastroenterology , vol.121 , pp. 1185-1190
    • Ismair, M.G.1    Stieger, B.2    Cattori, V.3
  • 105
    • 0242384851 scopus 로고    scopus 로고
    • Functional characterization of rat brain-specific organic anion transporter (Oatpl4) at the blood-brain barrier: High affinity transporter for thyroxine
    • Sugiyama D, Kusuhara H, Taniguchi H, et al. Functional characterization of rat brain-specific organic anion transporter (Oatpl4) at the blood-brain barrier: high affinity transporter for thyroxine. J Biol Chem 2003; 278:43489-43495.
    • (2003) J Biol Chem , vol.278 , pp. 43489-43495
    • Sugiyama, D.1    Kusuhara, H.2    Taniguchi, H.3
  • 106
    • 0036793106 scopus 로고    scopus 로고
    • Identification of a novel human organic anion transporting polypeptide as a high affinity thyroxine transporter
    • Pizzagalli F, Hagenbuch B, Stieger B, et al. Identification of a novel human organic anion transporting polypeptide as a high affinity thyroxine transporter. Mol Endocrinol 2002; 16:2283-2296.
    • (2002) Mol Endocrinol , vol.16 , pp. 2283-2296
    • Pizzagalli, F.1    Hagenbuch, B.2    Stieger, B.3
  • 107
    • 4344581457 scopus 로고    scopus 로고
    • Involvement of multispecific organic anion transporter, Oatpl4 (Slc21al4), in the transport of thyroxine across the blood-brain barrier
    • Tohyama K, Kusuhara H, Sugiyama Y. Involvement of multispecific organic anion transporter, Oatpl4 (Slc21al4), in the transport of thyroxine across the blood-brain barrier. Endocrinology 2004; 145:4384-4391.
    • (2004) Endocrinology , vol.145 , pp. 4384-4391
    • Tohyama, K.1    Kusuhara, H.2    Sugiyama, Y.3
  • 108
    • 0035125407 scopus 로고    scopus 로고
    • Organic anion-transporting polypeptide B (OATP-B) and its functional comparison with three other OATPs of human liver
    • Kullak-Ublick GA, Ismair MG, Stieger B, et al. Organic anion-transporting polypeptide B (OATP-B) and its functional comparison with three other OATPs of human liver. Gastroenterology 2001; 120:525-533.
    • (2001) Gastroenterology , vol.120 , pp. 525-533
    • Kullak-Ublick, G.A.1    Ismair, M.G.2    Stieger, B.3
  • 109
    • 0038637144 scopus 로고    scopus 로고
    • Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane
    • Kobayashi D, Nozawa T, Imai K, et al. Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane. J Pharmacol Exp Ther 2003; 306:703-708.
    • (2003) J Pharmacol Exp Ther , vol.306 , pp. 703-708
    • Kobayashi, D.1    Nozawa, T.2    Imai, K.3
  • 110
    • 33746048844 scopus 로고    scopus 로고
    • Predominant contribution of organic anion transporting polypeptide OATP-B (OATP2B1) to apical uptake of estrone-3-sulfate by human intestinal Caco-2 cells
    • Sai Y, Kaneko Y, Ito S, et al. Predominant contribution of organic anion transporting polypeptide OATP-B (OATP2B1) to apical uptake of estrone-3-sulfate by human intestinal Caco-2 cells. Drug Metab Dispos 2006; 34:1423-1431.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1423-1431
    • Sai, Y.1    Kaneko, Y.2    Ito, S.3
  • 111
    • 12144288563 scopus 로고    scopus 로고
    • Isolation and characterization of a digoxin transporter and its rat homologue expressed in the kidney
    • Mikkaichi T, Suzuki T, Onogawa T, et al. Isolation and characterization of a digoxin transporter and its rat homologue expressed in the kidney. Proc Natl Acad Sci U S A 2004; 101:3569-3574.
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 3569-3574
    • Mikkaichi, T.1    Suzuki, T.2    Onogawa, T.3
  • 112
    • 0028063302 scopus 로고
    • Drug excretion mediated by a new prototype of polyspecific transporter
    • Grundemann D, Gorboulev V, Gambaryan S, et al. Drug excretion mediated by a new prototype of polyspecific transporter. Nature 1994; 372:549-552.
    • (1994) Nature , vol.372 , pp. 549-552
    • Grundemann, D.1    Gorboulev, V.2    Gambaryan, S.3
  • 113
    • 0036146331 scopus 로고    scopus 로고
    • Tissue distribution and renal developmental changes in rat organic cation transporter mRNA levels
    • Slitt AL, Cherrington NJ, Hartley DP, et al. Tissue distribution and renal developmental changes in rat organic cation transporter mRNA levels. Drug Metab Dispos 2002; 30:212-219.
    • (2002) Drug Metab Dispos , vol.30 , pp. 212-219
    • Slitt, A.L.1    Cherrington, N.J.2    Hartley, D.P.3
  • 114
    • 0030763035 scopus 로고    scopus 로고
    • Cloning and characterization of two human polyspecific organic cation transporters
    • Gorboulev V, Ulzheimer JC, Akhoundova A, et al. Cloning and characterization of two human polyspecific organic cation transporters. DNA Cell Biol 1997; 16:871-881.
    • (1997) DNA Cell Biol , vol.16 , pp. 871-881
    • Gorboulev, V.1    Ulzheimer, J.C.2    Akhoundova, A.3
  • 115
    • 0032456205 scopus 로고    scopus 로고
    • Functional characteristics and membrane localization of rat multispecific organic cation transporters, OCT1 and OCT2, mediating tubular secretion of cationic drugs
    • Urakami Y, Okuda M, Masuda S, et al. Functional characteristics and membrane localization of rat multispecific organic cation transporters, OCT1 and OCT2, mediating tubular secretion of cationic drugs. J Pharmacol Exp Ther 1998; 287:800-805.
    • (1998) J Pharmacol Exp Ther , vol.287 , pp. 800-805
    • Urakami, Y.1    Okuda, M.2    Masuda, S.3
  • 116
    • 0032581363 scopus 로고    scopus 로고
    • Membrane localization of the electrogenic cation transporter rOCTl in rat liver
    • Meyer-Wentrup F, Karbach U, Gorboulev V, et al. Membrane localization of the electrogenic cation transporter rOCTl in rat liver. Biochem Biophys Res Commun 1998; 248:673-678.
    • (1998) Biochem Biophys Res Commun , vol.248 , pp. 673-678
    • Meyer-Wentrup, F.1    Karbach, U.2    Gorboulev, V.3
  • 117
    • 0036070666 scopus 로고    scopus 로고
    • Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin
    • Wang DS, Jonker JW, Kato Y, et al. Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin. J Pharmacol Exp Ther 2002; 302:510-515.
    • (2002) J Pharmacol Exp Ther , vol.302 , pp. 510-515
    • Wang, D.S.1    Jonker, J.W.2    Kato, Y.3
  • 118
    • 0034935038 scopus 로고    scopus 로고
    • Reduced hepatic uptake and intestinal excretion of organic cations in mice with a targeted disruption of the organic cation transporter 1 (Octl [Slc22al]) gene
    • Jonker JW, Wagenaar E, Mol CA, et al. Reduced hepatic uptake and intestinal excretion of organic cations in mice with a targeted disruption of the organic cation transporter 1 (Octl [Slc22al]) gene. Mol Cell Biol 2001; 21:5471-5477.
    • (2001) Mol Cell Biol , vol.21 , pp. 5471-5477
    • Jonker, J.W.1    Wagenaar, E.2    Mol, C.A.3
  • 119
    • 33344454764 scopus 로고    scopus 로고
    • Tissue distribution and ontogeny of organic cation transporters in mice
    • Alnouti Y, Petrick JS, Klaassen CD. Tissue distribution and ontogeny of organic cation transporters in mice. Drug Metab Dispos 2006; 34:477-482.
    • (2006) Drug Metab Dispos , vol.34 , pp. 477-482
    • Alnouti, Y.1    Petrick, J.S.2    Klaassen, C.D.3
  • 120
    • 25644452793 scopus 로고    scopus 로고
    • A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters
    • Tahara H, Kusuhara H, Endou H, et al. A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters. J Pharmacol Exp Ther 2005; 315:337-345.
    • (2005) J Pharmacol Exp Ther , vol.315 , pp. 337-345
    • Tahara, H.1    Kusuhara, H.2    Endou, H.3
  • 121
    • 0042886894 scopus 로고    scopus 로고
    • Selective substrates for non-neuronal monoamine transporters
    • Grundemann D, Liebich G, Kiefer N, et al. Selective substrates for non-neuronal monoamine transporters. Mol Pharmacol 1999; 56:1-10.
    • (1999) Mol Pharmacol , vol.56 , pp. 1-10
    • Grundemann, D.1    Liebich, G.2    Kiefer, N.3
  • 122
    • 0036175142 scopus 로고    scopus 로고
    • Human organic anion transporters and human organic cation transporters mediate renal antiviral transport
    • Takeda M, Khamdang S, Narikawa S, et al. Human organic anion transporters and human organic cation transporters mediate renal antiviral transport. J Pharmacol Exp Ther 2002; 300:918-924.
    • (2002) J Pharmacol Exp Ther , vol.300 , pp. 918-924
    • Takeda, M.1    Khamdang, S.2    Narikawa, S.3
  • 123
    • 0036205460 scopus 로고    scopus 로고
    • Gene expression levels and immunolocalization of organic ion transporters in the human kidney
    • Motohashi H, Sakurai Y, Saito H, et al. Gene expression levels and immunolocalization of organic ion transporters in the human kidney. J Am Soc Nephrol 2002; 13:866-874.
    • (2002) J am Soc Nephrol , vol.13 , pp. 866-874
    • Motohashi, H.1    Sakurai, Y.2    Saito, H.3
  • 124
    • 0033711928 scopus 로고    scopus 로고
    • Basolateral localization of organic cation transporter 2 in intact renal proximal tubules
    • Sweet DH, Miller DS, Pritchard JB. Basolateral localization of organic cation transporter 2 in intact renal proximal tubules. Am J Physiol Renal Physiol 2000; 279:F826-F834.
    • (2000) Am J Physiol Renal Physiol , vol.279 , pp. F826-F834
    • Sweet, D.H.1    Miller, D.S.2    Pritchard, J.B.3
  • 125
    • 33644664085 scopus 로고    scopus 로고
    • Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1
    • Kimura N, Masuda S, Tanihara Y, et al. Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1. Drug Metab Pharmacokinet 2005; 20:379-386.
    • (2005) Drug Metab Pharmacokinet , vol.20 , pp. 379-386
    • Kimura, N.1    Masuda, S.2    Tanihara, Y.3
  • 126
    • 0142123151 scopus 로고    scopus 로고
    • Deficiency in the organic cation transporters 1 and 2 (Octl/Oct2 [Slc22al/Slc22a2]) in mice abolishes renal secretion of organic cations
    • Jonker JW, Wagenaar E, Van Eijl S, et al. Deficiency in the organic cation transporters 1 and 2 (Octl/Oct2 [Slc22al/Slc22a2]) in mice abolishes renal secretion of organic cations. Mol Cell Biol 2003; 23:7902-7908.
    • (2003) Mol Cell Biol , vol.23 , pp. 7902-7908
    • Jonker, J.W.1    Wagenaar, E.2    van Eijl, S.3
  • 127
    • 0032569029 scopus 로고    scopus 로고
    • Cloning and functional characterization of a potential-sensitive, polyspecific organic cation transporter (OCT3) most abundantly expressed in placenta
    • Kekuda R, Prasad PD, Wu X, et al. Cloning and functional characterization of a potential-sensitive, polyspecific organic cation transporter (OCT3) most abundantly expressed in placenta. J Biol Chem 1998; 273:15971-15979.
    • (1998) J Biol Chem , vol.273 , pp. 15971-15979
    • Kekuda, R.1    Prasad, P.D.2    Wu, X.3
  • 128
    • 0034975789 scopus 로고    scopus 로고
    • Impaired activity of the extraneuronal monoamine transporter system known as uptake-2 in Orct3/Slc22a3-deficient mice
    • Zwart R, Verhaagh S, Buitelaar M, et al. Impaired activity of the extraneuronal monoamine transporter system known as uptake-2 in Orct3/Slc22a3-deficient mice. Mol Cell Biol 2001; 21:4188-4196.
    • (2001) Mol Cell Biol , vol.21 , pp. 4188-4196
    • Zwart, R.1    Verhaagh, S.2    Buitelaar, M.3
  • 129
    • 0030855487 scopus 로고    scopus 로고
    • Expression cloning and characterization of a novel multispecific organic anion transporter
    • Sekine T, Watanabe N, Hosoyamada M, et al. Expression cloning and characterization of a novel multispecific organic anion transporter. J Biol Chem 1997; 272:18526-18529.
    • (1997) J Biol Chem , vol.272 , pp. 18526-18529
    • Sekine, T.1    Watanabe, N.2    Hosoyamada, M.3
  • 130
    • 0032979273 scopus 로고    scopus 로고
    • Immunohistochemical localization of multispecific renal organic anion transporter 1 in rat kidney
    • Tojo A, Sekine T, Nakajima N, et al. Immunohistochemical localization of multispecific renal organic anion transporter 1 in rat kidney. J Am Soc Nephrol 1999; 10:464-471.
    • (1999) J am Soc Nephrol , vol.10 , pp. 464-471
    • Tojo, A.1    Sekine, T.2    Nakajima, N.3
  • 131
    • 0032931612 scopus 로고    scopus 로고
    • Transport properties of nonsteroidal anti-inflammatory drugs by organic anion transporter 1 expressed in Xenopus laevis oocytes
    • Apiwattanakul N, Sekine T, Chairoungdua A, et al. Transport properties of nonsteroidal anti-inflammatory drugs by organic anion transporter 1 expressed in Xenopus laevis oocytes. Mol Pharmacol 1999; 55:847-854.
    • (1999) Mol Pharmacol , vol.55 , pp. 847-854
    • Apiwattanakul, N.1    Sekine, T.2    Chairoungdua, A.3
  • 132
    • 0032780906 scopus 로고    scopus 로고
    • The interaction and transport of beta-lactam antibiotics with the cloned rat renal organic anion transporter 1
    • Jariyawat S, Sekine T, Takeda M, et al. The interaction and transport of beta-lactam antibiotics with the cloned rat renal organic anion transporter 1. J Pharmacol Exp Ther 1999; 290:672-677.
    • (1999) J Pharmacol Exp Ther , vol.290 , pp. 672-677
    • Jariyawat, S.1    Sekine, T.2    Takeda, M.3
  • 133
    • 24344461838 scopus 로고    scopus 로고
    • Human organic anion transporter hOAT3 is a potent transporter of cephalosporin antibiotics, in comparison with hOATl
    • Ueo H, Motohashi H, Katsura T, et al. Human organic anion transporter hOAT3 is a potent transporter of cephalosporin antibiotics, in comparison with hOATl. Biochem Pharmacol 2005; 70:1104-1113.
    • (2005) Biochem Pharmacol , vol.70 , pp. 1104-1113
    • Ueo, H.1    Motohashi, H.2    Katsura, T.3
  • 134
    • 0032856603 scopus 로고    scopus 로고
    • The antiviral nucleotide analogs cidofovir and adefovir are novel substrates for human and rat renal organic anion transporter 1
    • Cihlar T, Lin DC, Pritchard JB, et al. The antiviral nucleotide analogs cidofovir and adefovir are novel substrates for human and rat renal organic anion transporter 1. Mol Pharmacol 1999; 56:570-580.
    • (1999) Mol Pharmacol , vol.56 , pp. 570-580
    • Cihlar, T.1    Lin, D.C.2    Pritchard, J.B.3
  • 135
    • 33646165531 scopus 로고    scopus 로고
    • Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock-out mice
    • Eraly SA, Vallon V, Vaughn DA, et al. Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock-out mice. J Biol Chem 2006; 281:5072-5083.
    • (2006) J Biol Chem , vol.281 , pp. 5072-5083
    • Eraly, S.A.1    Vallon, V.2    Vaughn, D.A.3
  • 136
    • 0036198703 scopus 로고    scopus 로고
    • Gender-specific and developmental influences on the expression of rat organic anion transporters
    • Buist SC, Cherrington NJ, Choudhuri S, et al. Gender-specific and developmental influences on the expression of rat organic anion transporters. J Pharmacol Exp Ther 2002; 301:145-151.
    • (2002) J Pharmacol Exp Ther , vol.301 , pp. 145-151
    • Buist, S.C.1    Cherrington, N.J.2    Choudhuri, S.3
  • 137
    • 0036073213 scopus 로고    scopus 로고
    • Gender difference in the urinary excretion of organic anions in rats
    • Kato Y, Kuge K, Kusuhara H, et al. Gender difference in the urinary excretion of organic anions in rats. J Pharmacol Exp Ther 2002; 302:483-89.
    • (2002) J Pharmacol Exp Ther , vol.302 , pp. 483-489
    • Kato, Y.1    Kuge, K.2    Kusuhara, H.3
  • 138
    • 0036084956 scopus 로고    scopus 로고
    • Isolation, characterization and differential gene expression of multispecific organic anion transporter 2 in mice
    • Kobayashi Y, Ohshiro N, Shibusawa A, et al. Isolation, characterization and differential gene expression of multispecific organic anion transporter 2 in mice. Mol Pharmacol 2002; 62:7-14.
    • (2002) Mol Pharmacol , vol.62 , pp. 7-14
    • Kobayashi, Y.1    Ohshiro, N.2    Shibusawa, A.3
  • 139
    • 2442700344 scopus 로고    scopus 로고
    • Rat and mouse differences in gender-predominant expression of organic anion transporter (Oatl-3; Slc22a6-8) mRNA levels
    • Buist SC, Klaassen CD. Rat and mouse differences in gender-predominant expression of organic anion transporter (Oatl-3; Slc22a6-8) mRNA levels. Drug Metab Dispos 2004; 32:620-625.
    • (2004) Drug Metab Dispos , vol.32 , pp. 620-625
    • Buist, S.C.1    Klaassen, C.D.2
  • 140
    • 0031984212 scopus 로고    scopus 로고
    • Role of lipoprotein in the plasma binding of SDZ PSC 833, a novel multidrug resistance-reversing cyclosporin
    • Simon N, Dailly E, Combes S, et al. Role of lipoprotein in the plasma binding of SDZ PSC 833, a novel multidrug resistance-reversing cyclosporin. Br J Clin Pharmacol 1998; 45:173-175.
    • (1998) Br J Clin Pharmacol , vol.45 , pp. 173-175
    • Simon, N.1    Dailly, E.2    Combes, S.3
  • 141
    • 17144449057 scopus 로고    scopus 로고
    • Identification of multispecific organic anion transporter 2 expressed predominantly in the liver
    • Sekine T, Cha SH, Tsuda M, et al. Identification of multispecific organic anion transporter 2 expressed predominantly in the liver. FEBS Lett 1998; 429:179-182.
    • (1998) FEBS Lett , vol.429 , pp. 179-182
    • Sekine, T.1    Cha, S.H.2    Tsuda, M.3
  • 142
    • 22944438439 scopus 로고    scopus 로고
    • Functional involvement of rat organic anion transporter 2 (Slc22a7) in the hepatic uptake of the nonsteroidal anti-inflammatory drug ketoprofen
    • Morita N, Kusuhara H, Nozaki Y, et al. Functional involvement of rat organic anion transporter 2 (Slc22a7) in the hepatic uptake of the nonsteroidal anti-inflammatory drug ketoprofen. Drug Metab Dispos 2005; 33:1151-1157.
    • (2005) Drug Metab Dispos , vol.33 , pp. 1151-1157
    • Morita, N.1    Kusuhara, H.2    Nozaki, Y.3
  • 143
    • 0034892457 scopus 로고    scopus 로고
    • Functional characterization of rat organic anion transporter 2 in LLC-PK1 cells
    • Morita N, Kusuhara H, Sekine T, et al. Functional characterization of rat organic anion transporter 2 in LLC-PK1 cells. J Pharmacol Exp Ther 2001; 298:1179-1184.
    • (2001) J Pharmacol Exp Ther , vol.298 , pp. 1179-1184
    • Morita, N.1    Kusuhara, H.2    Sekine, T.3
  • 144
    • 0033532065 scopus 로고    scopus 로고
    • Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain
    • Kusuhara H, Sekine T, Utsunomiya-Tate N, et al. Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain. J Biol Chem 1999; 274:13675-13680.
    • (1999) J Biol Chem , vol.274 , pp. 13675-13680
    • Kusuhara, H.1    Sekine, T.2    Utsunomiya-Tate, N.3
  • 145
    • 0033574034 scopus 로고    scopus 로고
    • Molecular cloning and characterization of two novel human renal organic anion transporters (hOATl and hOAT3)
    • Race JE, Grassl SM, Williams WJ, et al. Molecular cloning and characterization of two novel human renal organic anion transporters (hOATl and hOAT3). Biochem Biophys Res Commun 1999; 255:508-514.
    • (1999) Biochem Biophys Res Commun , vol.255 , pp. 508-514
    • Race, J.E.1    Grassl, S.M.2    Williams, W.J.3
  • 146
    • 0035039326 scopus 로고    scopus 로고
    • Identification and characterization of human organic anion transporter 3 expressing predominantly in the kidney
    • Cha SH, Sekine T, Fukushima JI, et al. Identification and characterization of human organic anion transporter 3 expressing predominantly in the kidney. Mol Pharmacol 2001; 59:1277-1286.
    • (2001) Mol Pharmacol , vol.59 , pp. 1277-1286
    • Cha, S.H.1    Sekine, T.2    Fukushima, J.I.3
  • 147
    • 0037383621 scopus 로고    scopus 로고
    • Rat organic anion transporter 3 (rOAT3) is responsible for brain-to-blood efflux of homovanillic acid at the abluminal membrane of brain capillary endothelial cells
    • Mori S, Takanaga H, Ohtsuki S, et al. Rat organic anion transporter 3 (rOAT3) is responsible for brain-to-blood efflux of homovanillic acid at the abluminal membrane of brain capillary endothelial cells. J Cereb Blood Flow Metab 2003; 23:432-440.
    • (2003) J Cereb Blood Flow Metab , vol.23 , pp. 432-440
    • Mori, S.1    Takanaga, H.2    Ohtsuki, S.3
  • 148
    • 0038274495 scopus 로고    scopus 로고
    • Contribution of organic anion transporter 3 (Slc22a8) to the elimination of p-aminohippuric acid and benzylpenicillin across the blood-brain barrier
    • Kikuchi R, Kusuhara H, Sugiyama D, et al. Contribution of organic anion transporter 3 (Slc22a8) to the elimination of p-aminohippuric acid and benzylpenicillin across the blood-brain barrier. J Pharmacol Exp Ther 2003; 306:51-58.
    • (2003) J Pharmacol Exp Ther , vol.306 , pp. 51-58
    • Kikuchi, R.1    Kusuhara, H.2    Sugiyama, D.3
  • 149
    • 0036235889 scopus 로고    scopus 로고
    • Expression and functional characterization of rat organic anion transporter 3 (rOat3) in the choroid plexus
    • Nagata Y, Kusuhara H, Endou H, et al. Expression and functional characterization of rat organic anion transporter 3 (rOat3) in the choroid plexus. Mol Pharmacol 2002; 61:982-988.
    • (2002) Mol Pharmacol , vol.61 , pp. 982-988
    • Nagata, Y.1    Kusuhara, H.2    Endou, H.3
  • 150
    • 4143108317 scopus 로고    scopus 로고
    • Carrier-mediated uptake of H2-receptor antagonists by the rat choroid plexus: Involvement of rat organic anion transporter 3
    • Nagata Y, Kusuhara H, Hirono S, et al. Carrier-mediated uptake of H2-receptor antagonists by the rat choroid plexus: involvement of rat organic anion transporter 3. Drug Metab Dispos 2004; 32:1040-1047.
    • (2004) Drug Metab Dispos , vol.32 , pp. 1040-1047
    • Nagata, Y.1    Kusuhara, H.2    Hirono, S.3
  • 151
    • 0347362498 scopus 로고    scopus 로고
    • Characterization of uremic toxin transport by organic anion transporters in the kidney
    • Deguchi T, Kusuhara H, Takadate A, et al. Characterization of uremic toxin transport by organic anion transporters in the kidney. Kidney Int 2004; 65:162-174.
    • (2004) Kidney Int , vol.65 , pp. 162-174
    • Deguchi, T.1    Kusuhara, H.2    Takadate, A.3
  • 152
    • 33644749355 scopus 로고    scopus 로고
    • Is the monkey an appropriate animal model to examine drug-drug interactions involving renal clearance? Effect of probenecid on the renal elimination of H2 receptor antagonists
    • Tahara H, Kusuhara H, Chida M, et al. Is the monkey an appropriate animal model to examine drug-drug interactions involving renal clearance? Effect of probenecid on the renal elimination of H2 receptor antagonists. J Pharmacol Exp Ther 2006; 316:1187-1194.
    • (2006) J Pharmacol Exp Ther , vol.316 , pp. 1187-1194
    • Tahara, H.1    Kusuhara, H.2    Chida, M.3
  • 153
    • 0027378438 scopus 로고
    • Bisubstrates: Substances that interact with renal contraluminal organic anion and organic cation transport systems. I. Amines, piperidines, piperazines, azepines, pyridines, quinolines, imidazoles, thiazoles, guanidines and hydrazines
    • Ullrich KJ, Rumrich G, David C, et al. Bisubstrates: substances that interact with renal contraluminal organic anion and organic cation transport systems. I. Amines, piperidines, piperazines, azepines, pyridines, quinolines, imidazoles, thiazoles, guanidines and hydrazines. Pflugers Arch 1993; 425:280-299.
    • (1993) Pflugers Arch , vol.425 , pp. 280-299
    • Ullrich, K.J.1    Rumrich, G.2    David, C.3
  • 154
    • 0037178852 scopus 로고    scopus 로고
    • Impaired organic anion transport in kidney and choroid plexus of organic anion transporter 3 (Oat3 (Slc22a8)) knockout mice
    • Sweet DH, Miller DS, Pritchard JB, et al. Impaired organic anion transport in kidney and choroid plexus of organic anion transporter 3 (Oat3 (Slc22a8)) knockout mice. J Biol Chem 2002; 277:26934-26943.
    • (2002) J Biol Chem , vol.277 , pp. 26934-26943
    • Sweet, D.H.1    Miller, D.S.2    Pritchard, J.B.3
  • 155
    • 0000518648 scopus 로고    scopus 로고
    • Molecular cloning and characterization of multispecific organic anion transporter 4 expressed in the placenta
    • Cha SH, Sekine T, Kusuhara H, et al. Molecular cloning and characterization of multispecific organic anion transporter 4 expressed in the placenta. J Biol Chem 2000; 275:4507-4512.
    • (2000) J Biol Chem , vol.275 , pp. 4507-4512
    • Cha, S.H.1    Sekine, T.2    Kusuhara, H.3
  • 156
    • 0037067159 scopus 로고    scopus 로고
    • Role of human organic anion transporter 4 in the transport of ochratoxin A
    • Babu E, Takeda M, Narikawa S, et al. Role of human organic anion transporter 4 in the transport of ochratoxin A. Biochim Biophys Acta 2002; 1590:64-75.
    • (2002) Biochim Biophys Acta , vol.1590 , pp. 64-75
    • Babu, E.1    Takeda, M.2    Narikawa, S.3
  • 157
    • 27144483941 scopus 로고    scopus 로고
    • Functional characterization of rat organic anion transporter 5 (Slc22al9) at the apical membrane of renal proximal tubules
    • Anzai N, Jutabha P, Enomoto A, et al. Functional characterization of rat organic anion transporter 5 (Slc22al9) at the apical membrane of renal proximal tubules. J Pharmacol Exp Ther 2005; 315:534-544.
    • (2005) J Pharmacol Exp Ther , vol.315 , pp. 534-544
    • Anzai, N.1    Jutabha, P.2    Enomoto, A.3
  • 158
    • 0031434178 scopus 로고    scopus 로고
    • Cloning and characterization of a novel human pH-dependent organic cation transporter, OCTN1
    • Tamai I, Yabuuchi H, Nezu J, et al. Cloning and characterization of a novel human pH-dependent organic cation transporter, OCTN1. FEBS Lett 1997; 419:107-111.
    • (1997) FEBS Lett , vol.419 , pp. 107-111
    • Tamai, I.1    Yabuuchi, H.2    Nezu, J.3
  • 159
    • 0032935034 scopus 로고    scopus 로고
    • Novel membrane transporter OCTN1 mediates multispecific, bidirectional, and pH-dependent transport of organic cations
    • Yabuuchi H, Tamai I, Nezu J, et al. Novel membrane transporter OCTN1 mediates multispecific, bidirectional, and pH-dependent transport of organic cations. J Pharmacol Exp Ther 1999; 289:768-773.
    • (1999) J Pharmacol Exp Ther , vol.289 , pp. 768-773
    • Yabuuchi, H.1    Tamai, I.2    Nezu, J.3
  • 160
    • 0032577202 scopus 로고    scopus 로고
    • cDNA sequence, transport function, and genomic organization of human OCTN2, a new member of the organic cation transporter family
    • Wu X, Prasad PD, Leibach FH, et al. cDNA sequence, transport function, and genomic organization of human OCTN2, a new member of the organic cation transporter family. Biochem Biophys Res Commun 1998; 246:589-595.
    • (1998) Biochem Biophys Res Commun , vol.246 , pp. 589-595
    • Wu, X.1    Prasad, P.D.2    Leibach, F.H.3
  • 161
    • 0032493741 scopus 로고    scopus 로고
    • Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2
    • Tamai I, Ohashi R, Nezu J, et al. Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J Biol Chem 1998; 273:20378-20382.
    • (1998) J Biol Chem , vol.273 , pp. 20378-20382
    • Tamai, I.1    Ohashi, R.2    Nezu, J.3
  • 162
    • 0032767691 scopus 로고    scopus 로고
    • Functional characteristics and tissue distribution pattern of organic cation transporter 2 (OCTN2), an organic cation/carnitine transporter
    • Wu X, Huang W, Prasad PD, et al. Functional characteristics and tissue distribution pattern of organic cation transporter 2 (OCTN2), an organic cation/carnitine transporter. J Pharmacol Exp Ther 1999; 290:1482-1492.
    • (1999) J Pharmacol Exp Ther , vol.290 , pp. 1482-1492
    • Wu, X.1    Huang, W.2    Prasad, P.D.3
  • 163
    • 0032696186 scopus 로고    scopus 로고
    • Na(+)-dependent carnitine transport by organic cation transporter (OCTN2): Its pharmacological and toxicological relevance
    • Ohashi R, Tamai I, Yabuuchi H, et al. Na(+)-dependent carnitine transport by organic cation transporter (OCTN2): its pharmacological and toxicological relevance. J Pharmacol Exp Ther 1999; 291:778-784.
    • (1999) J Pharmacol Exp Ther , vol.291 , pp. 778-784
    • Ohashi, R.1    Tamai, I.2    Yabuuchi, H.3
  • 164
    • 0034695673 scopus 로고    scopus 로고
    • Beta-lactam antibiotics as substrates for OCTN2, an organic cation/carnitine transporter
    • Ganapathy ME, Huang W, Rajan DP, et al. Beta-lactam antibiotics as substrates for OCTN2, an organic cation/carnitine transporter. J Biol Chem 2000; 275:1699-1707.
    • (2000) J Biol Chem , vol.275 , pp. 1699-1707
    • Ganapathy, M.E.1    Huang, W.2    Rajan, D.P.3
  • 165
    • 0032953645 scopus 로고    scopus 로고
    • Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter
    • Nezu J, Tamai I, Oku A, et al. Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter. Nat Genet 1999; 21:91-94.
    • (1999) Nat Genet , vol.21 , pp. 91-94
    • Nezu, J.1    Tamai, I.2    Oku, A.3
  • 166
    • 0035132434 scopus 로고    scopus 로고
    • Molecular and physiological evidence for multifunctionality of carnitine/organic cation transporter OCTN2
    • Ohashi R, Tamai I, Nezu Ji J, et al. Molecular and physiological evidence for multifunctionality of carnitine/organic cation transporter OCTN2. Mol Pharmacol 2001; 59:358-366.
    • (2001) Mol Pharmacol , vol.59 , pp. 358-366
    • Ohashi, R.1    Tamai, I.2    Nezu Ji, J.3
  • 167
    • 29144523534 scopus 로고    scopus 로고
    • A human transporter protein that mediates the final excretion step for toxic organic cations
    • Otsuka M, Matsumoto T, Morimoto R, et al. A human transporter protein that mediates the final excretion step for toxic organic cations. Proc Natl Acad Sci U S A 2005; 102:17923-17928.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 17923-17928
    • Otsuka, M.1    Matsumoto, T.2    Morimoto, R.3
  • 168
    • 33746548445 scopus 로고    scopus 로고
    • Identification and functional characterization of a new human kidney-specific H+/organic cation antiporter, kidneyspecific multidrug and toxin extrusion 2
    • Masuda S, Terada T, Yonezawa A, et al. Identification and functional characterization of a new human kidney-specific H+/organic cation antiporter, kidneyspecific multidrug and toxin extrusion 2. J Am Soc Nephrol 2006; 17:2127-2135.
    • (2006) J am Soc Nephrol , vol.17 , pp. 2127-2135
    • Masuda, S.1    Terada, T.2    Yonezawa, A.3
  • 169
    • 0028333611 scopus 로고
    • Expression cloning of a mammalian protoncoupled oligopeptide transporter
    • Fei YJ, Kanai Y, Nussberger S, et al. Expression cloning of a mammalian protoncoupled oligopeptide transporter. Nature 1994; 368:563-566.
    • (1994) Nature , vol.368 , pp. 563-566
    • Fei, Y.J.1    Kanai, Y.2    Nussberger, S.3
  • 170
    • 0029979149 scopus 로고    scopus 로고
    • Immuno-localization of H-f /peptide cotransporter in rat digestive tract
    • Ogihara H, Saito H, Shin BC, et al. Immuno-localization of H-f /peptide cotransporter in rat digestive tract. Biochem Biophys Res Commun 1996; 220:848-852.
    • (1996) Biochem Biophys Res Commun , vol.220 , pp. 848-852
    • Ogihara, H.1    Saito, H.2    Shin, B.C.3
  • 171
    • 0032967296 scopus 로고    scopus 로고
    • Localization of PEPT1 and PEPT2 protoncoupled oligopeptide transporter mRNA and protein in rat kidney
    • Shen H, Smith DE, Yang T, et al. Localization of PEPT1 and PEPT2 protoncoupled oligopeptide transporter mRNA and protein in rat kidney. Am J Physiol 1999; 276:F658-F665.
    • (1999) Am J Physiol , vol.276 , pp. F658-F665
    • Shen, H.1    Smith, D.E.2    Yang, T.3
  • 172
    • 0034825881 scopus 로고    scopus 로고
    • Intestinal peptide transport: Ex vivo uptake studies and localization of peptide carrier PEPT1
    • Groneberg DA, Doring F, Eynott PR, et al. Intestinal peptide transport: ex vivo uptake studies and localization of peptide carrier PEPT1. Am J Physiol Gastrointest Liver Physiol 2001; 281:G697-G704.
    • (2001) Am J Physiol Gastrointest Liver Physiol , vol.281 , pp. G697-G704
    • Groneberg, D.A.1    Doring, F.2    Eynott, P.R.3
  • 173
    • 0027967880 scopus 로고
    • Expression cloning of a cDNA from rabbit small intestine related to proton-coupled transport of peptides, beta-lactam antibiotics and ACE- inhibitors
    • Boll M, Markovich D, Weber WM, et al. Expression cloning of a cDNA from rabbit small intestine related to proton-coupled transport of peptides, beta-lactam antibiotics and ACE- inhibitors. Pflugers Arch 1994; 429:146-149.
    • (1994) Pflugers Arch , vol.429 , pp. 146-149
    • Boll, M.1    Markovich, D.2    Weber, W.M.3
  • 174
    • 0031981201 scopus 로고    scopus 로고
    • Carrier-mediated intestinal absorption of valacyclovir, the L-valyl ester prodrug of acyclovir: 1. Interactions with peptides, organic anions and organic cations in rats
    • Sinko PJ, Balimane PV. Carrier-mediated intestinal absorption of valacyclovir, the L-valyl ester prodrug of acyclovir: 1. Interactions with peptides, organic anions and organic cations in rats. Biopharm Drug Dispos 1998; 19:209-217.
    • (1998) Biopharm Drug Dispos , vol.19 , pp. 209-217
    • Sinko, P.J.1    Balimane, P.V.2
  • 175
    • 0028883462 scopus 로고
    • On N, et al. Absolute bioavailability and metabolic disposition of valaciclovir, the L-valyl ester of acyclovir, following oral administration to humans
    • Soul-Lawton J, Seaber E, On N, et al. Absolute bioavailability and metabolic disposition of valaciclovir, the L-valyl ester of acyclovir, following oral administration to humans. Antimicrob Agents Chemother 1995; 39:2759-2764.
    • (1995) Antimicrob Agents Chemother , vol.39 , pp. 2759-2764
    • Soul-Lawton, J.1    Seaber, E.2
  • 176
    • 0027768955 scopus 로고
    • Pharmacokinetics of the acyclovir pro-drug valaciclovir after escalating single- and multiple-dose administration to normal volunteers
    • Weller S, Blum MR, Doucette M, et al. Pharmacokinetics of the acyclovir pro-drug valaciclovir after escalating single- and multiple-dose administration to normal volunteers. Clin Pharmacol Ther 1993; 54:595-605.
    • (1993) Clin Pharmacol Ther , vol.54 , pp. 595-605
    • Weller, S.1    Blum, M.R.2    Doucette, M.3
  • 177
    • 0031662109 scopus 로고    scopus 로고
    • Rhie JK, et al. 5'-Amino acid esters of antiviral nucleosides, acyclovir, and AZT are absorbed by the intestinal PEPT1 peptide transporter
    • Han H, de Vrueh RL, Rhie JK, et al. 5'-Amino acid esters of antiviral nucleosides, acyclovir, and AZT are absorbed by the intestinal PEPT1 peptide transporter. Pharm Res 1998; 15:1154-1159.
    • (1998) Pharm Res , vol.15 , pp. 1154-1159
    • Han, H.1    de Vrueh, R.L.2
  • 178
    • 0032546467 scopus 로고    scopus 로고
    • Valacyclovir: A substrate for the intestinal and renal peptide transporters PEPT1 and PEPT2
    • Ganapathy ME, Huang W, Wang H, et al. Valacyclovir: a substrate for the intestinal and renal peptide transporters PEPT1 and PEPT2. Biochem Biophys Res Commun 1998; 246:470-475.
    • (1998) Biochem Biophys Res Commun , vol.246 , pp. 470-475
    • Ganapathy, M.E.1    Huang, W.2    Wang, H.3
  • 179
    • 0031773397 scopus 로고    scopus 로고
    • Improvement of L-dopa absorption by dipeptidyl derivation, utilizing peptide transporter PepTl
    • Tamai I, Nakanishi T, Nakahara H, et al. Improvement of L-dopa absorption by dipeptidyl derivation, utilizing peptide transporter PepTl. J Pharm Sci 1998; 87:1542-1546.
    • (1998) J Pharm Sci , vol.87 , pp. 1542-1546
    • Tamai, I.1    Nakanishi, T.2    Nakahara, H.3
  • 180
    • 0029919966 scopus 로고    scopus 로고
    • Molecular cloning and tissue distribution of rat peptide transporter PEPT2
    • Saito H, Terada T, Okuda M, et al. Molecular cloning and tissue distribution of rat peptide transporter PEPT2. Biochim Biophys Acta 1996; 1280:173-177.
    • (1996) Biochim Biophys Acta , vol.1280 , pp. 173-177
    • Saito, H.1    Terada, T.2    Okuda, M.3
  • 181
    • 0030026812 scopus 로고    scopus 로고
    • Expression cloning and functional characterization of the kidney cortex high-affinity proton-coupled peptide transporter
    • Boll M, Herget M, Wagener M, et al. Expression cloning and functional characterization of the kidney cortex high-affinity proton-coupled peptide transporter. Proc Natl Acad Sci U S A 1996; 93:284-289.
    • (1996) Proc Natl Acad Sci U S A , vol.93 , pp. 284-289
    • Boll, M.1    Herget, M.2    Wagener, M.3
  • 182
    • 0031713192 scopus 로고    scopus 로고
    • Tubular localization and tissue distribution of peptide transporters in rat kidney
    • Smith DE, Pavlova A, Berger UV, et al. Tubular localization and tissue distribution of peptide transporters in rat kidney. Pharm Res 1998; 15:1244-1249.
    • (1998) Pharm Res , vol.15 , pp. 1244-1249
    • Smith, D.E.1    Pavlova, A.2    Berger, U.V.3
  • 183
    • 0003119124 scopus 로고    scopus 로고
    • The peptide transporter PepT2 is expressed in rat brain and mediates the accumulation of the florescent dipeptide derivative
    • Dieck ST, Heuer H, Ehrchen J, et al. The peptide transporter PepT2 is expressed in rat brain and mediates the accumulation of the florescent dipeptide derivative. Glia 1998; 25:10-20.
    • (1998) Glia , vol.25 , pp. 10-20
    • Dieck, S.T.1    Heuer, H.2    Ehrchen, J.3
  • 184
    • 0034131371 scopus 로고    scopus 로고
    • Mechanisms of 5-aminolevulinic acid uptake at the choroid plexus
    • Novotny A, Xiang J, Stummer W, et al. Mechanisms of 5-aminolevulinic acid uptake at the choroid plexus. J Neurochem 2000; 75:321-328.
    • (2000) J Neurochem , vol.75 , pp. 321-328
    • Novotny, A.1    Xiang, J.2    Stummer, W.3
  • 185
    • 0033613850 scopus 로고    scopus 로고
    • Stoichiometry and kinetics of the high-affinity H+-coupled peptide transporter PepT2
    • Chen XZ, Zhu T, Smith DE, et al. Stoichiometry and kinetics of the high-affinity H+-coupled peptide transporter PepT2. J Biol Chem 1999; 274:2773-2779.
    • (1999) J Biol Chem , vol.274 , pp. 2773-2779
    • Chen, X.Z.1    Zhu, T.2    Smith, D.E.3
  • 186
    • 0030661969 scopus 로고    scopus 로고
    • Recognition of beta-lactam antibiotics by rat peptide transporters, PEPT1 and PEPT2, in LLC-PK1 cells
    • Terada T, Saito H, Mukai M, et al. Recognition of beta-lactam antibiotics by rat peptide transporters, PEPT1 and PEPT2, in LLC-PK1 cells. Am J Physiol 1997; 273:F706-F711.
    • (1997) Am J Physiol , vol.273 , pp. F706-F711
    • Terada, T.1    Saito, H.2    Mukai, M.3
  • 187
    • 0028822394 scopus 로고
    • Proton/peptide cotransporter (PEPT 2) from human kidney: Functional characterization and chromosomal localization
    • Ramamoorthy S, Liu W, Ma YY, et al. Proton/peptide cotransporter (PEPT 2) from human kidney: functional characterization and chromosomal localization. Biochim Biophys Acta 1995; 1240:1-4.
    • (1995) Biochim Biophys Acta , vol.1240 , pp. 1-4
    • Ramamoorthy, S.1    Liu, W.2    Ma, Y.Y.3
  • 188
    • 0032430799 scopus 로고    scopus 로고
    • Interaction of beta-lactam antibiotics with H+/peptide cotransporters in rat renal brush-border membranes
    • Takahashi K, Nakamura N, Terada T, et al. Interaction of beta-lactam antibiotics with H+/peptide cotransporters in rat renal brush-border membranes. J Pharmacol Exp Ther 1998; 286:1037-1042.
    • (1998) J Pharmacol Exp Ther , vol.286 , pp. 1037-1042
    • Takahashi, K.1    Nakamura, N.2    Terada, T.3
  • 189
    • 0029896232 scopus 로고    scopus 로고
    • Expression of a renal type I sodium/ phosphate transporter (NaPi-1) induces a conductance in Xenopus oocytes permeable for organic and inorganic anions
    • Busch AE, Schuster A, Waldegger S, et al. Expression of a renal type I sodium/ phosphate transporter (NaPi-1) induces a conductance in Xenopus oocytes permeable for organic and inorganic anions. Proc Natl Acad Sci U S A 1996; 93:5347-5351.
    • (1996) Proc Natl Acad Sci U S A , vol.93 , pp. 5347-5351
    • Busch, A.E.1    Schuster, A.2    Waldegger, S.3
  • 190
    • 0032160713 scopus 로고    scopus 로고
    • Hepatic sinusoidal membrane transport of anionic drugs mediated by anion transporter Nptl
    • Yabuuchi H, Tamai I, Morita K, et al. Hepatic sinusoidal membrane transport of anionic drugs mediated by anion transporter Nptl. J Pharmacol Exp Ther 1998; 286:1391-1396.
    • (1998) J Pharmacol Exp Ther , vol.286 , pp. 1391-1396
    • Yabuuchi, H.1    Tamai, I.2    Morita, K.3
  • 191
    • 0015893137 scopus 로고
    • Active outward transport of daunomycin in resistant Ehrlich ascites tumor cells
    • Dano K. Active outward transport of daunomycin in resistant Ehrlich ascites tumor cells. Biochim Biophys Acta 1973; 323:466-483.
    • (1973) Biochim Biophys Acta , vol.323 , pp. 466-483
    • Dano, K.1
  • 192
    • 0018200435 scopus 로고
    • Mechanisms of resistance to daunorubicin in Ehrlich ascites tumor cells
    • Skovsgaard T. Mechanisms of resistance to daunorubicin in Ehrlich ascites tumor cells. Cancer Res 1978; 38:1785-1791.
    • (1978) Cancer Res , vol.38 , pp. 1785-1791
    • Skovsgaard, T.1
  • 193
    • 0031691234 scopus 로고    scopus 로고
    • The role of P-glycoprotein and canalicular multispecific organic anion transporter in the hepatobiliary excretion of drugs
    • 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; 87:1025-1040.
    • (1998) J Pharm Sci , vol.87 , pp. 1025-1040
    • Kusuhara, H.1    Suzuki, H.2    Sugiyama, Y.3
  • 194
    • 0029082150 scopus 로고
    • Hepatobiliary secretion of organic compounds; molecular mechanisms of membrane transport
    • Oude Elferink RP, Meijer DK, Kuipers F, et al. Hepatobiliary secretion of organic compounds; molecular mechanisms of membrane transport. Biochim Biophys Acta 1995; 1241:215-268.
    • (1995) Biochim Biophys Acta , vol.1241 , pp. 215-268
    • Oude Elferink, R.P.1    Meijer, D.K.2    Kuipers, F.3
  • 195
    • 0343247810 scopus 로고    scopus 로고
    • Blood-brain barrier function of P-glycoprotein
    • Tsuji A, Tamai I. Blood-brain barrier function of P-glycoprotein. Adv Drug Deliv Rev 1997; 25:285-298.
    • (1997) Adv Drug Deliv Rev , vol.25 , pp. 285-298
    • Tsuji, A.1    Tamai, I.2
  • 196
    • 0342618583 scopus 로고    scopus 로고
    • Renal secretion of xenobiotics mediated by P-glycoprotein: Importance to renal function in health and exploitation for targeted drug delivery to epithelial cysts in polycystic kidney disease
    • Simons NL, Hunter J, Jepson MA. Renal secretion of xenobiotics mediated by P-glycoprotein: importance to renal function in health and exploitation for targeted drug delivery to epithelial cysts in polycystic kidney disease. Adv Drug Deliv Rev 1997; 25:243-256.
    • (1997) Adv Drug Deliv Rev , vol.25 , pp. 243-256
    • Simons, N.L.1    Hunter, J.2    Jepson, M.A.3
  • 197
    • 0343488673 scopus 로고    scopus 로고
    • Hirst BH. Intestinal secretion of drugs. The role of P-glycoprotein and related drug efflux systems in limiting oral drug absorption
    • Hunter J, Hirst BH. Intestinal secretion of drugs. The role of P-glycoprotein and related drug efflux systems in limiting oral drug absorption. Adv Drug Deliv Rev 1997; 25:129-157.
    • (1997) Adv Drug Deliv Rev , vol.25 , pp. 129-157
    • Hunter, J.1
  • 198
    • 0042468096 scopus 로고    scopus 로고
    • Impact of drug transporter studies on drug discovery and development
    • Mizuno N, Niwa T, Yotsumoto Y, et al. Impact of drug transporter studies on drug discovery and development. Pharmacol Rev 2003; 55:425-461.
    • (2003) Pharmacol Rev , vol.55 , pp. 425-461
    • Mizuno, N.1    Niwa, T.2    Yotsumoto, Y.3
  • 199
    • 0035703732 scopus 로고    scopus 로고
    • The roles of P-glycoprotein and MRP1 in the blood-brain and bloodcerebrospinal fluid barriers
    • Schinkel AH. The roles of P-glycoprotein and MRP1 in the blood-brain and bloodcerebrospinal fluid barriers. Adv Exp Med Biol 2001; 500:365-372.
    • (2001) Adv Exp Med Biol , vol.500 , pp. 365-372
    • Schinkel, A.H.1
  • 200
    • 0028229150 scopus 로고
    • Disruption of the mouse mdrla P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs
    • Schinkel AH, Smit JJ, van Tellingen O, et al. Disruption of the mouse mdrla P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 1994; 77:491-502.
    • (1994) Cell , vol.77 , pp. 491-502
    • Schinkel, A.H.1    Smit, J.J.2    van Tellingen, O.3
  • 201
    • 23944475593 scopus 로고    scopus 로고
    • Permeability dominates in vivo intestinal absorption of P-gp substrate with high solubility and high permeability
    • Cao X, Yu LX, Barbaciru C, et al. Permeability dominates in vivo intestinal absorption of P-gp substrate with high solubility and high permeability. Mol Pharmacol 2005; 2:329-340.
    • (2005) Mol Pharmacol , vol.2 , pp. 329-340
    • Cao, X.1    Yu, L.X.2    Barbaciru, C.3
  • 202
    • 33747821713 scopus 로고    scopus 로고
    • P-glycoprotein and an unstirred water layer barring digoxin absorption in the vascularly perfused rat small intestine preparation: Induction studies with pregnenolone-16alpha-carbonitrile
    • Liu S, Tam D, Chen X, et al. P-glycoprotein and an unstirred water layer barring digoxin absorption in the vascularly perfused rat small intestine preparation: induction studies with pregnenolone-16alpha-carbonitrile. Drug Metab Dispos 2006; 34:1468-1479.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1468-1479
    • Liu, S.1    Tam, D.2    Chen, X.3
  • 203
    • 33645827555 scopus 로고    scopus 로고
    • Effect of P-glycoprotein on intestinal absorption and brain penetration of antiallergic agent bepotastine besilate
    • Ohashi R, Kamikozawa Y, Sugiura M, et al. Effect of P-glycoprotein on intestinal absorption and brain penetration of antiallergic agent bepotastine besilate. Drug Metab Dispos 2006; 34:793-799.
    • (2006) Drug Metab Dispos , vol.34 , pp. 793-799
    • Ohashi, R.1    Kamikozawa, Y.2    Sugiura, M.3
  • 204
    • 13444252326 scopus 로고    scopus 로고
    • Mapping of multidrug resistance gene 1 and multidrug resistance-associated protein isoform 1 to 5 mRNA expression along the human intestinal tract
    • Zimmermann C, Gutmann H, Hruz P, et al. Mapping of multidrug resistance gene 1 and multidrug resistance-associated protein isoform 1 to 5 mRNA expression along the human intestinal tract. Drug Metab Dispos 2005; 33:219-224.
    • (2005) Drug Metab Dispos , vol.33 , pp. 219-224
    • Zimmermann, C.1    Gutmann, H.2    Hruz, P.3
  • 205
    • 21744436852 scopus 로고    scopus 로고
    • Cytochromes P450 and MDR1 mRNA expression along the human gastrointestinal tract
    • Thorn M, Finnstrom N, Lundgren S, et al. Cytochromes P450 and MDR1 mRNA expression along the human gastrointestinal tract. Br J Clin Pharmacol 2005; 60:54-60.
    • (2005) Br J Clin Pharmacol , vol.60 , pp. 54-60
    • Thorn, M.1    Finnstrom, N.2    Lundgren, S.3
  • 206
    • 33750181267 scopus 로고    scopus 로고
    • Regional levels of drug transporters along the human intestinal tract: Co-expression of ABC and SLC transporters and comparison with Caco-2 cells
    • Englund G, Rorsman F, Ronnblom A, et al. Regional levels of drug transporters along the human intestinal tract: co-expression of ABC and SLC transporters and comparison with Caco-2 cells. Eur J Pharm Sei 2006; 29:269-277.
    • (2006) Eur J Pharm Sei , vol.29 , pp. 269-277
    • Englund, G.1    Rorsman, F.2    Ronnblom, A.3
  • 207
    • 0036229852 scopus 로고    scopus 로고
    • Drugs as P-glycoprotein substrates, inhibitors, and inducers
    • Kim RB. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev 2002; 34:47-54.
    • (2002) Drug Metab Rev , vol.34 , pp. 47-54
    • Kim, R.B.1
  • 208
    • 0031921312 scopus 로고    scopus 로고
    • Contribution of the murine mdrla p-glycoprotein to hepatobiliary and intestinal elimination of cationic drugs as measured in mice with an mdrla gene disruption
    • Smit JW, Schinkel AH, Muller M, et al. Contribution of the murine mdrla p-glycoprotein to hepatobiliary and intestinal elimination of cationic drugs as measured in mice with an mdrla gene disruption. Hepatology 1998; 27:1056-1063.
    • (1998) Hepatology , vol.27 , pp. 1056-1063
    • Smit, J.W.1    Schinkel, A.H.2    Muller, M.3
  • 209
    • 0029860174 scopus 로고    scopus 로고
    • Substantial excretion of digoxin via the intestinal mucosa and prevention of long-term digoxin accumulation in the brain by the mdr la P-glycoprotein
    • Mayer U, Wagenaar E, Beijnen JH, et al. Substantial excretion of digoxin via the intestinal mucosa and prevention of long-term digoxin accumulation in the brain by the mdr la P-glycoprotein. Br J Pharmacol 1996; 119:1038-1044.
    • (1996) Br J Pharmacol , vol.119 , pp. 1038-1044
    • Mayer, U.1    Wagenaar, E.2    Beijnen, J.H.3
  • 210
    • 4243801613 scopus 로고    scopus 로고
    • Normal viability and altered pharmacokinetics in mice lacking mdr 1-type (drug-transporting) P-glycoproteins
    • Schinkel AH, Mayer U, Wagenaar E, et al. Normal viability and altered pharmacokinetics in mice lacking mdr 1-type (drug-transporting) P-glycoproteins. Proc Natl Acad Sei U S A 1997; 94:4028-4033.
    • (1997) Proc Natl Acad Sei U S A , vol.94 , pp. 4028-4033
    • Schinkel, A.H.1    Mayer, U.2    Wagenaar, E.3
  • 211
    • 12644278301 scopus 로고    scopus 로고
    • Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine
    • Sparreboom A, van Asperen J, Mayer U, et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sei U S A 1997; 94:2031-2035.
    • (1997) Proc Natl Acad Sei U S A , vol.94 , pp. 2031-2035
    • Sparreboom, A.1    van Asperen, J.2    Mayer, U.3
  • 212
    • 0032793959 scopus 로고    scopus 로고
    • OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine
    • Cvetkovic M, Leake B, Fromm MF, et al. OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine. Drug Metab Dispos 1999; 27:866-871.
    • (1999) Drug Metab Dispos , vol.27 , pp. 866-871
    • Cvetkovic, M.1    Leake, B.2    Fromm, M.F.3
  • 213
    • 0042125510 scopus 로고    scopus 로고
    • Quantitative evaluation of the function of small intestinal P-glycoprotein: Comparative studies between in situ and in vitro
    • Adachi Y, Suzuki H, Sugiyama Y. Quantitative evaluation of the function of small intestinal P-glycoprotein: comparative studies between in situ and in vitro. Pharm Res 2003; 20:1163-1169.
    • (2003) Pharm Res , vol.20 , pp. 1163-1169
    • Adachi, Y.1    Suzuki, H.2    Sugiyama, Y.3
  • 214
    • 0031757312 scopus 로고    scopus 로고
    • BBB transport and P-glycoprotein functionality using MDR1A (-/-) and wild-type mice. Total brain versus microdialysis concentration profiles of rhodamine-123
    • de Lange EC, de Bock G, Schinkel AH, et al. BBB transport and P-glycoprotein functionality using MDR1A (-/-) and wild-type mice. Total brain versus microdialysis concentration profiles of rhodamine-123. Pharm Res 1998; 15:1657-1665.
    • (1998) Pharm Res , vol.15 , pp. 1657-1665
    • de Lange, E.C.1    de Bock, G.2    Schinkel, A.H.3
  • 215
    • 0034724324 scopus 로고    scopus 로고
    • Functional polymorphisms of the human multidrug-resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo
    • Hoffmeyer S, Burk O, von Richter O, et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 2000; 97:3473-3478.
    • (2000) Proc Natl Acad Sci U S A , vol.97 , pp. 3473-3478
    • Hoffmeyer, S.1    Burk, O.2    von Richter, O.3
  • 216
    • 0036693051 scopus 로고    scopus 로고
    • Role of human MDR1 gene polymorphism in bioavailability and interaction of digoxin, a substrate of P-glycoprotein
    • Kurata Y, Ieiri I, Kimura M, et al. Role of human MDR1 gene polymorphism in bioavailability and interaction of digoxin, a substrate of P-glycoprotein. Clin Pharmacol Ther 2002; 72:209-219.
    • (2002) Clin Pharmacol Ther , vol.72 , pp. 209-219
    • Kurata, Y.1    Ieiri, I.2    Kimura, M.3
  • 217
    • 0033833430 scopus 로고    scopus 로고
    • Increased drug delivery to the brain by P-glycoprotein inhibition
    • Sadeque AJ, Wandel C, He H, et al. Increased drug delivery to the brain by P-glycoprotein inhibition. Clin Pharmacol Ther 2000; 68:231-237.
    • (2000) Clin Pharmacol Ther , vol.68 , pp. 231-237
    • Sadeque, A.J.1    Wandel, C.2    He, H.3
  • 218
    • 20444480695 scopus 로고    scopus 로고
    • Imaging P-glycoprotein transport activity at the human blood-brain barrier with positron emission tomography
    • Sasongko L, Link JM, Muzi M, et al. Imaging P-glycoprotein transport activity at the human blood-brain barrier with positron emission tomography. Clin Pharmacol Ther 2005; 77:503-514.
    • (2005) Clin Pharmacol Ther , vol.77 , pp. 503-514
    • Sasongko, L.1    Link, J.M.2    Muzi, M.3
  • 219
    • 0027095653 scopus 로고
    • Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line [see comments]
    • Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line [see comments]. Science 1992; 258:1650-1654.
    • (1992) Science , vol.258 , pp. 1650-1654
    • Cole, S.P.1    Bhardwaj, G.2    Gerlach, J.H.3
  • 220
    • 0027172915 scopus 로고
    • Analysis of the expression of MRP, the gene for a new putative transmembrane drug transporter, in human multidrug resistant lung cancer cell lines
    • Zaman GJ, Versantvoort CH, Smit JJ, et al. Analysis of the expression of MRP, the gene for a new putative transmembrane drug transporter, in human multidrug resistant lung cancer cell lines. Cancer Res 1993; 53:1747-1750.
    • (1993) Cancer Res , vol.53 , pp. 1747-1750
    • Zaman, G.J.1    Versantvoort, C.H.2    Smit, J.J.3
  • 221
    • 0030775395 scopus 로고    scopus 로고
    • Pharmacological characterization of the murine and human orthologs of multidrug-resistance protein in transfected human embryonic kidney cells
    • Stride BD, Grant CE, Loe DW, et al. Pharmacological characterization of the murine and human orthologs of multidrug-resistance protein in transfected human embryonic kidney cells. Mol Pharmacol 1997; 52:344-353.
    • (1997) Mol Pharmacol , vol.52 , pp. 344-353
    • Stride, B.D.1    Grant, C.E.2    Loe, D.W.3
  • 222
    • 0027960078 scopus 로고
    • Pharmacological characterization of multidrug resistant MRP-transfected human tumor cells
    • Cole SP, Sparks KE, Fraser K, et al. Pharmacological characterization of multidrug resistant MRP-transfected human tumor cells. Cancer Res 1994; 54:5902-5910.
    • (1994) Cancer Res , vol.54 , pp. 5902-5910
    • Cole, S.P.1    Sparks, K.E.2    Fraser, K.3
  • 223
    • 31844455959 scopus 로고    scopus 로고
    • Substrate recognition and transport by multidrug resistance protein 1 (ABCC1)
    • Deeley RG, Cole SP. Substrate recognition and transport by multidrug resistance protein 1 (ABCC1). FEBS Lett 2006; 580:1103-1111.
    • (2006) FEBS Lett , vol.580 , pp. 1103-1111
    • Deeley, R.G.1    Cole, S.P.2
  • 224
    • 0033975098 scopus 로고    scopus 로고
    • Multidrug resistance protein 1 protects the choroid plexus epithelium and contributes to the blood-cerebrospinal fluid barrier
    • Wijnholds J, de Lange EC, Scheffer GL, et al. Multidrug resistance protein 1 protects the choroid plexus epithelium and contributes to the blood-cerebrospinal fluid barrier. J Clin Invest 2000; 105:279-285.
    • (2000) J Clin Invest , vol.105 , pp. 279-285
    • Wijnholds, J.1    de Lange, E.C.2    Scheffer, G.L.3
  • 225
    • 0141790752 scopus 로고    scopus 로고
    • Involvement of multidrug resistance associated protein 1 (Mrpl) in the efflux transport of 17beta estradiol-D-17betaglucuronide (E217betaG) across the blood-brain barrier
    • Sugiyama D, Kusuhara H, Lee YJ, et al. Involvement of multidrug resistance associated protein 1 (Mrpl) in the efflux transport of 17beta estradiol-D-17betaglucuronide (E217betaG) across the blood-brain barrier. Pharm Res 2003; 20: 1394-1400.
    • (2003) Pharm Res , vol.20 , pp. 1394-1400
    • Sugiyama, D.1    Kusuhara, H.2    Lee, Y.J.3
  • 226
    • 0347124816 scopus 로고    scopus 로고
    • Do multidrug resistance-associated protein-1 and -2 play any role in the elimination of estradiol-17 beta-glucuronide and 2,4-dinitrophenyl-S-glutathione across the blood-cerebrospinal fluid barrier?
    • Lee YJ, Kusuhara H, Sugiyama Y. Do multidrug resistance-associated protein-1 and -2 play any role in the elimination of estradiol-17 beta-glucuronide and 2,4-dinitrophenyl-S-glutathione across the blood-cerebrospinal fluid barrier? J Pharm Sci 2004; 93:99-107.
    • (2004) J Pharm Sci , vol.93 , pp. 99-107
    • Lee, Y.J.1    Kusuhara, H.2    Sugiyama, Y.3
  • 227
    • 0000053197 scopus 로고
    • The characters of a new mutant in rats with hyperbilirubinemic syndrome
    • Mikami T, Nozaki T, Tagaya O, et al. The characters of a new mutant in rats with hyperbilirubinemic syndrome. Cong Anom 1986; 26:250-251.
    • (1986) Cong Anom , vol.26 , pp. 250-251
    • Mikami, T.1    Nozaki, T.2    Tagaya, O.3
  • 228
    • 0021804301 scopus 로고
    • Hereditary chronic conjugated hyperbilirubinemia in mutant rats caused by defective hepatic anion transport
    • Jansen PL, Peters WH, Lamers WH. Hereditary chronic conjugated hyperbilirubinemia in mutant rats caused by defective hepatic anion transport. Hepatology 1985; 5:573-579.
    • (1985) Hepatology , vol.5 , pp. 573-579
    • Jansen, P.L.1    Peters, W.H.2    Lamers, W.H.3
  • 229
    • 0026519238 scopus 로고
    • A new rat mutant with chronic conjugated hyperbilirubinemia and renal glomerular lesions
    • Hosokawa S, Tagaya O, Mikami T, et al. A new rat mutant with chronic conjugated hyperbilirubinemia and renal glomerular lesions. Lab Anim Sci 1992; 42:27-34.
    • (1992) Lab Anim Sci , vol.42 , pp. 27-34
    • Hosokawa, S.1    Tagaya, O.2    Mikami, T.3
  • 230
    • 0024312957 scopus 로고
    • Hepatobiliary transport of glutathione and glutathione conjugate in rats with hereditary hyperbilirubinemia
    • Elferink RP, Ottenhoff R, Liefting W, et al. Hepatobiliary transport of glutathione and glutathione conjugate in rats with hereditary hyperbilirubinemia. J Clin Invest 1989; 84:476-483.
    • (1989) J Clin Invest , vol.84 , pp. 476-483
    • Elferink, R.P.1    Ottenhoff, R.2    Liefting, W.3
  • 231
    • 0027382061 scopus 로고
    • Kinetic analysis of hepatobiliary transport of organic anions in Eisai hyperbilirubinemic mutant rats
    • Sathirakul K, Suzuki H, Yasuda K, et al. Kinetic analysis of hepatobiliary transport of organic anions in Eisai hyperbilirubinemic mutant rats. J Pharmacol Exp Ther 1993; 265:1301-1312.
    • (1993) J Pharmacol Exp Ther , vol.265 , pp. 1301-1312
    • Sathirakul, K.1    Suzuki, H.2    Yasuda, K.3
  • 232
    • 0032434272 scopus 로고    scopus 로고
    • Excretion of GSSG and glutathione conjugates mediated by MRP1 and cMOAT/MRP2
    • Suzuki H, Sugiyama Y. Excretion of GSSG and glutathione conjugates mediated by MRP1 and cMOAT/MRP2. Semin Liver Dis 1998; 18:359-376.
    • (1998) Semin Liver Dis , vol.18 , pp. 359-376
    • Suzuki, H.1    Sugiyama, Y.2
  • 233
    • 0029981430 scopus 로고    scopus 로고
    • Congenital jaundice in rats with mutation in a multidrug resistance-associated protein gene
    • Paulusma CC, Bosma PJ, Zaman GJ, et al. Congenital jaundice in rats with mutation in a multidrug resistance-associated protein gene. Science 1996; 271: 1126-1128.
    • (1996) Science , vol.271 , pp. 1126-1128
    • Paulusma, C.C.1    Bosma, P.J.2    Zaman, G.J.3
  • 234
    • 0031031247 scopus 로고    scopus 로고
    • Molecular cloning of canalicular multispecific organic anion transporter defective in EHBR
    • Ito K, Suzuki H, Hirohashi T, et al. Molecular cloning of canalicular multispecific organic anion transporter defective in EHBR. Am J Physiol 1997; 272:G16-G22.
    • (1997) Am J Physiol , vol.272 , pp. G16-G22
    • Ito, K.1    Suzuki, H.2    Hirohashi, T.3
  • 235
    • 0040761368 scopus 로고    scopus 로고
    • cDNA cloning of the hepatocyte canalicular isoform of the multidrug resistance protein, cMrp, reveals a novel conjugate export pump deficient in hyperbilirubinemic mutant rats
    • Buchler M, Konig J, Brom M, et al. cDNA cloning of the hepatocyte canalicular isoform of the multidrug resistance protein, cMrp, reveals a novel conjugate export pump deficient in hyperbilirubinemic mutant rats. J Biol Chem 1996; 271:15091-15098.
    • (1996) J Biol Chem , vol.271 , pp. 15091-15098
    • Buchler, M.1    Konig, J.2    Brom, M.3
  • 236
    • 0030994468 scopus 로고    scopus 로고
    • A mutation in the human canalicular multispecific organic anion transporter gene causes the Dubin-Johnson syndrome
    • Paulusma CC, Kool M, Bosma PJ, et al. A mutation in the human canalicular multispecific organic anion transporter gene causes the Dubin-Johnson syndrome. Hepatology 1997; 25:1539-1542.
    • (1997) Hepatology , vol.25 , pp. 1539-1542
    • Paulusma, C.C.1    Kool, M.2    Bosma, P.J.3
  • 237
    • 0032974409 scopus 로고    scopus 로고
    • Sugiyama. Primary active transport of organic anions on bile canalicular membrane in humans
    • Niinuma K, Kato Y, Suzuki H, et al. Sugiyama. Primary active transport of organic anions on bile canalicular membrane in humans. Am J Physiol 1999; 276:G1153-G1164.
    • (1999) Am J Physiol , vol.276 , pp. G1153-G1164
    • Niinuma, K.1    Kato, Y.2    Suzuki, H.3
  • 238
    • 22344432207 scopus 로고    scopus 로고
    • Tissue distribution and hepatic and renal ontogeny of the multidrug resistance-associated protein (Mrp) family in mice
    • Maher JM, Slitt AL, Cherrington NJ, et al. Tissue distribution and hepatic and renal ontogeny of the multidrug resistance-associated protein (Mrp) family in mice. Drug Metab Dispos 2005; 33:947-955.
    • (2005) Drug Metab Dispos , vol.33 , pp. 947-955
    • Maher, J.M.1    Slitt, A.L.2    Cherrington, N.J.3
  • 239
    • 0036080426 scopus 로고    scopus 로고
    • Expression and localization of the multidrug resistance-associated protein 3 in rat small and large intestine
    • Rost D, Mahner S, Sugiyama Y, et al. Expression and localization of the multidrug resistance-associated protein 3 in rat small and large intestine. Am J Physiol Gastrointest Liver Physiol 2002; 282:G720-G726.
    • (2002) Am J Physiol Gastrointest Liver Physiol , vol.282 , pp. G720-G726
    • Rost, D.1    Mahner, S.2    Sugiyama, Y.3
  • 240
    • 0033958156 scopus 로고    scopus 로고
    • Involvement of an organic anion transporter (canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2) in gastrointestinal secretion of glutathione conjugates in rats
    • Gotoh Y, Suzuki H, Kinoshita S, et al. Involvement of an organic anion transporter (canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2) in gastrointestinal secretion of glutathione conjugates in rats. J Pharmacol Exp Ther 2000; 292:433-439.
    • (2000) J Pharmacol Exp Ther , vol.292 , pp. 433-439
    • Gotoh, Y.1    Suzuki, H.2    Kinoshita, S.3
  • 241
    • 0033032730 scopus 로고    scopus 로고
    • Expression of the MRP2 gene-encoded conjugate export pump in human kidney proximal tubules and in renal cell carcinoma
    • Schaub TP, Kartenbeck J, Konig J, et al. Expression of the MRP2 gene-encoded conjugate export pump in human kidney proximal tubules and in renal cell carcinoma. J Am Soc Nephrol 1999; 10:1159-1169.
    • (1999) J am Soc Nephrol , vol.10 , pp. 1159-1169
    • Schaub, T.P.1    Kartenbeck, J.2    Konig, J.3
  • 242
    • 0030832802 scopus 로고    scopus 로고
    • Expression of the conjugate export pump encoded by the mrp2 gene in the apical membrane of kidney proximal tubules
    • Schaub TP, Kartenbeck J, Konig J, et al. Expression of the conjugate export pump encoded by the mrp2 gene in the apical membrane of kidney proximal tubules. J Am Soc Nephrol 1997; 8:1213-1221.
    • (1997) J am Soc Nephrol , vol.8 , pp. 1213-1221
    • Schaub, T.P.1    Kartenbeck, J.2    Konig, J.3
  • 243
    • 0142213937 scopus 로고    scopus 로고
    • Impaired renal secretion of substrates for the multidrug resistance protein 2 in mutant transport-deficient (TR-) rats
    • Masereeuw R, Notenboom S, Smeets PH, et al. Impaired renal secretion of substrates for the multidrug resistance protein 2 in mutant transport-deficient (TR-) rats. J Am Soc Nephrol 2003; 14:2741-2749.
    • (2003) J am Soc Nephrol , vol.14 , pp. 2741-2749
    • Masereeuw, R.1    Notenboom, S.2    Smeets, P.H.3
  • 244
    • 21044455712 scopus 로고    scopus 로고
    • A mutation in the drug transporter gene ABCC2 associated with impaired methotrexate elimination
    • Hulot JS, Villard E, Maguy A, et al. A mutation in the drug transporter gene ABCC2 associated with impaired methotrexate elimination. Pharmacogenet Genom 2005; 15:277-285.
    • (2005) Pharmacogenet Genom , vol.15 , pp. 277-285
    • Hulot, J.S.1    Villard, E.2    Maguy, A.3
  • 245
    • 33644910207 scopus 로고    scopus 로고
    • Mice lacking Mrp3 (Abcc3) have normal bile salt transport, but altered hepatic transport of endogenous glucuronides
    • Zelcer N, van de Wetering K, de Waart R, et al. Mice lacking Mrp3 (Abcc3) have normal bile salt transport, but altered hepatic transport of endogenous glucuronides. J Hepatol 2006; 44:768-775.
    • (2006) J Hepatol , vol.44 , pp. 768-775
    • Zelcer, N.1    van de Wetering, K.2    de Waart, R.3
  • 246
    • 0031866357 scopus 로고    scopus 로고
    • Hepatic expression of multidrug resistanceassociated protein-like proteins maintained in eisai hyperbilirubinemic rats
    • Hirohashi T, Suzuki H, Ito K, et al. Hepatic expression of multidrug resistanceassociated protein-like proteins maintained in eisai hyperbilirubinemic rats. Mol Pharmacol 1998; 53:1068-1075.
    • (1998) Mol Pharmacol , vol.53 , pp. 1068-1075
    • Hirohashi, T.1    Suzuki, H.2    Ito, K.3
  • 247
    • 0031838805 scopus 로고    scopus 로고
    • cDNA cloning and inducible expression of human multidrug resistance associated protein 3 (MRP3)
    • Kiuchi Y, Suzuki H, Hirohashi T, et al. cDNA cloning and inducible expression of human multidrug resistance associated protein 3 (MRP3). FEBS Lett 1998; 433:149-152.
    • (1998) FEBS Lett , vol.433 , pp. 149-152
    • Kiuchi, Y.1    Suzuki, H.2    Hirohashi, T.3
  • 248
    • 0032501054 scopus 로고    scopus 로고
    • Isolation of a novel human canalicular multispecific organic anion transporter, cMOAT2/MRP3, and its expression in cisplatin-resistant cancer cells with decreased ATP-dependent drug transport
    • Uchiumi T, Hinoshita E, Haga S, et al. Isolation of a novel human canalicular multispecific organic anion transporter, cMOAT2/MRP3, and its expression in cisplatin-resistant cancer cells with decreased ATP-dependent drug transport. Biochem Biophys Res Commun 1998; 252:103-110.
    • (1998) Biochem Biophys Res Commun , vol.252 , pp. 103-110
    • Uchiumi, T.1    Hinoshita, E.2    Haga, S.3
  • 250
    • 0032926354 scopus 로고    scopus 로고
    • Characterization of the human multidrug resistance protein isoform MRP3 localized to the basolateral hepatocyte membrane
    • Konig J, Rost D, Cui Y, et al. Characterization of the human multidrug resistance protein isoform MRP3 localized to the basolateral hepatocyte membrane. Hepatology 1999; 29:1156-1163.
    • (1999) Hepatology , vol.29 , pp. 1156-1163
    • Konig, J.1    Rost, D.2    Cui, Y.3
  • 251
    • 0033591235 scopus 로고    scopus 로고
    • Characterization of the transport properties of cloned rat multidrug resistance-associated protein 3 (MRP3)
    • Hirohashi T, Suzuki H, Sugiyama Y. Characterization of the transport properties of cloned rat multidrug resistance-associated protein 3 (MRP3). J Biol Chem 1999; 274:15181-15185.
    • (1999) J Biol Chem , vol.274 , pp. 15181-15185
    • Hirohashi, T.1    Suzuki, H.2    Sugiyama, Y.3
  • 252
    • 0034723163 scopus 로고    scopus 로고
    • ATP-dependent transport of bile salts by rat multidrug resistance-associated protein 3 (Mrp3)
    • Hirohashi T, Suzuki H, Takikawa H, et al. ATP-dependent transport of bile salts by rat multidrug resistance-associated protein 3 (Mrp3). J Biol Chem 2000; 275: 2905-2910.
    • (2000) J Biol Chem , vol.275 , pp. 2905-2910
    • Hirohashi, T.1    Suzuki, H.2    Takikawa, H.3
  • 253
    • 0035476729 scopus 로고    scopus 로고
    • Transport of methotrexate (MTX) and folates by multidrug resistance protein (MRP) 3 and MRP1: Effect of polyglutamylation on MTX transport
    • Zeng H, Chen ZS, Belinsky MG, et al. Transport of methotrexate (MTX) and folates by multidrug resistance protein (MRP) 3 and MRP1: effect of polyglutamylation on MTX transport. Cancer Res 2001; 61:7225-7232.
    • (2001) Cancer Res , vol.61 , pp. 7225-7232
    • Zeng, H.1    Chen, Z.S.2    Belinsky, M.G.3
  • 254
    • 0036149075 scopus 로고    scopus 로고
    • Transport activity of human MRP3 expressed in Sf9 cells: Comparative studies with rat MRP3
    • Akita H, Suzuki H, Hirohashi T, et al. Transport activity of human MRP3 expressed in Sf9 cells: comparative studies with rat MRP3. Pharm Res 2002; 19:34-41.
    • (2002) Pharm Res , vol.19 , pp. 34-41
    • Akita, H.1    Suzuki, H.2    Hirohashi, T.3
  • 255
    • 0034802573 scopus 로고    scopus 로고
    • Sinusoidal efflux of taurocholate is enhanced in Mrp2-deficient rat liver
    • Akita H, Suzuki H, Sugiyama Y. Sinusoidal efflux of taurocholate is enhanced in Mrp2-deficient rat liver. Pharm Res 2001; 18:1119-1125.
    • (2001) Pharm Res , vol.18 , pp. 1119-1125
    • Akita, H.1    Suzuki, H.2    Sugiyama, Y.3
  • 256
    • 21044435938 scopus 로고    scopus 로고
    • Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception
    • Zelcer N, van de Wetering K, Hillebrand M, et al. Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception. Proc Natl Acad Sci U S A 2005; 102:7274-7279.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 7274-7279
    • Zelcer, N.1    van de Wetering, K.2    Hillebrand, M.3
  • 257
    • 33751199707 scopus 로고    scopus 로고
    • Evaluation of the role of multidrug resistance-associated protein (Mrp) 3 and Mrp4 in hepatic basolateral excretion of sulfate and glucuronide metabolites of acetaminophen, 4-methylumbelliferone, and harmol in Abcc3-/- and Abcc4-/- mice
    • Zamek-Gliszczynski MJ, Nezasa K, Tian X, et al. Evaluation of the role of multidrug resistance-associated protein (Mrp) 3 and Mrp4 in hepatic basolateral excretion of sulfate and glucuronide metabolites of acetaminophen, 4-methylumbelliferone, and harmol in Abcc3-/- and Abcc4-/- mice. J Pharmacol Exp Ther 2006; 319:1485-1491.
    • (2006) J Pharmacol Exp Ther , vol.319 , pp. 1485-1491
    • Zamek-Gliszczynski, M.J.1    Nezasa, K.2    Tian, X.3
  • 258
    • 27744574913 scopus 로고    scopus 로고
    • Altered disposition of acetaminophen in mice with a disruption of the Mrp3 gene
    • Manautou JE, de Waart DR, Kunne C, et al. Altered disposition of acetaminophen in mice with a disruption of the Mrp3 gene. Hepatology 2005; 42:1091-1098.
    • (2005) Hepatology , vol.42 , pp. 1091-1098
    • Manautou, J.E.1    de Waart, D.R.2    Kunne, C.3
  • 259
    • 4644349805 scopus 로고    scopus 로고
    • ATP-dependent transport of organic anions into isolated basolateral membrane vesicles from rat intestine
    • Shoji T, Suzuki H, Kusuhara H, et al. ATP-dependent transport of organic anions into isolated basolateral membrane vesicles from rat intestine. Am J Physiol Gastrointest Liver Physiol 2004; 287:G749-G756.
    • (2004) Am J Physiol Gastrointest Liver Physiol , vol.287 , pp. G749-G756
    • Shoji, T.1    Suzuki, H.2    Kusuhara, H.3
  • 260
    • 0030841332 scopus 로고    scopus 로고
    • Analysis of expression of cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues of the multidrug resistance-associated protein gene (MRP1), in human cancer cell lines
    • Kool M, de Haas M, Scheffer GL, et al. Analysis of expression of cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues of the multidrug resistance-associated protein gene (MRP1), in human cancer cell lines. Cancer Res 1997; 57:3537-3547.
    • (1997) Cancer Res , vol.57 , pp. 3537-3547
    • Kool, M.1    de Haas, M.2    Scheffer, G.L.3
  • 261
    • 0042766881 scopus 로고    scopus 로고
    • Cotransport of reduced glutathione with bile salts by MRP4 (ABCC4) localized to the basolateral hepatocyte membrane
    • Rius M, Nies AT, Hummel-Eisenbeiss J, et al. Cotransport of reduced glutathione with bile salts by MRP4 (ABCC4) localized to the basolateral hepatocyte membrane. Hepatology 2003; 38:374-384.
    • (2003) Hepatology , vol.38 , pp. 374-384
    • Rius, M.1    Nies, A.T.2    Hummel-Eisenbeiss, J.3
  • 262
    • 4344632950 scopus 로고    scopus 로고
    • Mrp4 confers resistance to topotecan and protects the brain from chemotherapy
    • Leggas M, Adachi M, Scheffer GL, et al. Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol Cell Biol 2004; 24:7612-7621.
    • (2004) Mol Cell Biol , vol.24 , pp. 7612-7621
    • Leggas, M.1    Adachi, M.2    Scheffer, G.L.3
  • 263
    • 0036186107 scopus 로고    scopus 로고
    • The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: Putative efflux pump for urinary cAMP and cGMP
    • van Aubel RA, Smeets PH, Peters JH, et al. The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP. J Am Soc Nephrol 2002; 13:595-603.
    • (2002) J am Soc Nephrol , vol.13 , pp. 595-603
    • van Aubel, R.A.1    Smeets, P.H.2    Peters, J.H.3
  • 264
    • 0038752694 scopus 로고    scopus 로고
    • Characterization of the transport of nucleoside analog drugs by the human multidrug resistance proteins MRP4 and MRP5
    • Reid G, Wielinga P, Zelcer N, et al. Characterization of the transport of nucleoside analog drugs by the human multidrug resistance proteins MRP4 and MRP5. Mol Pharmacol 2003; 63:1094-1103.
    • (2003) Mol Pharmacol , vol.63 , pp. 1094-1103
    • Reid, G.1    Wielinga, P.2    Zelcer, N.3
  • 265
    • 0036606023 scopus 로고    scopus 로고
    • Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system
    • Chen ZS, Lee K, Walther S, et al. Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system. Cancer Res 2002; 62:3144-3150.
    • (2002) Cancer Res , vol.62 , pp. 3144-3150
    • Chen, Z.S.1    Lee, K.2    Walther, S.3
  • 266
    • 12144269640 scopus 로고    scopus 로고
    • Contribution of multidrug resistance protein 2 (MRP2/ABCC2) to the renal excretion of p-aminohippurate (PAH) and identification of MRP4 (ABCC4) as a novel PAH transporter
    • Smeets PH, van Aubel RA, Wouterse AC, et al. Contribution of multidrug resistance protein 2 (MRP2/ABCC2) to the renal excretion of p-aminohippurate (PAH) and identification of MRP4 (ABCC4) as a novel PAH transporter. J Am Soc Nephrol 2004; 15:2828-2835.
    • (2004) J am Soc Nephrol , vol.15 , pp. 2828-2835
    • Smeets, P.H.1    van Aubel, R.A.2    Wouterse, A.C.3
  • 267
    • 33846019659 scopus 로고    scopus 로고
    • Multidrug resistance-associated protein 4 is involved in the urinary excretion of hydrochlorothiazide and furosemide
    • Hasegawa M, Kusuhara H, Adachi M, et al. Multidrug resistance-associated protein 4 is involved in the urinary excretion of hydrochlorothiazide and furosemide. J Am Soc Nephrol 2007; 18:37-45.
    • (2007) J am Soc Nephrol , vol.18 , pp. 37-45
    • Hasegawa, M.1    Kusuhara, H.2    Adachi, M.3
  • 268
    • 33846420624 scopus 로고    scopus 로고
    • Functional involvement of multidrug resistance associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs, adefovir and tenofovir
    • Imaoka T, Kusuhara H, Adachi M, et al. Functional involvement of multidrug resistance associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs, adefovir and tenofovir. Mol Pharmacol 2007; 71:619-627.
    • (2007) Mol Pharmacol , vol.71 , pp. 619-627
    • Imaoka, T.1    Kusuhara, H.2    Adachi, M.3
  • 269
    • 33846664466 scopus 로고    scopus 로고
    • ATP-binding cassette, subfamily G (ABCG family)
    • Kusuhara H. Sugiyama Y. ATP-binding cassette, subfamily G (ABCG family). Pflugers Arch 2007; 453:735-744.
    • (2007) Pflugers Arch , vol.453 , pp. 735-744
    • Kusuhara, H.1    Sugiyama, Y.2
  • 270
    • 0042068263 scopus 로고    scopus 로고
    • A functional study on polymorphism of the ATPbinding cassette transporter ABCG2: Critical role of arginine-482 in methotrexate transport
    • Mitomo H, Kato R, Ito A, et al. A functional study on polymorphism of the ATPbinding cassette transporter ABCG2: critical role of arginine-482 in methotrexate transport. Biochem J 2003; 373:767-774.
    • (2003) Biochem J , vol.373 , pp. 767-774
    • Mitomo, H.1    Kato, R.2    Ito, A.3
  • 271
    • 0037050736 scopus 로고    scopus 로고
    • Dominant-negative inhibition of breast cancer resistance protein as drug efflux pump through the inhibition of S-S dependent homodimerization
    • Kage K, Tsukahara S, Sugiyama T, et al. Dominant-negative inhibition of breast cancer resistance protein as drug efflux pump through the inhibition of S-S dependent homodimerization. Int J Cancer 2002; 97:626-630.
    • (2002) Int J Cancer , vol.97 , pp. 626-630
    • Kage, K.1    Tsukahara, S.2    Sugiyama, T.3
  • 272
    • 0032404092 scopus 로고    scopus 로고
    • A human placenta-specific ATPbinding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance
    • Allikmets R, Schriml LM, Hutchinson A, et al. A human placenta-specific ATPbinding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res 1998; 58:5337-5339.
    • (1998) Cancer Res , vol.58 , pp. 5337-5339
    • Allikmets, R.1    Schriml, L.M.2    Hutchinson, A.3
  • 273
    • 13044249156 scopus 로고    scopus 로고
    • A multidrug resistance transporter from human MCF-7 breast cancer cells
    • Doyle LA, Yang W, Abruzzo LV, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 1998; 95:15665-15670.
    • (1998) Proc Natl Acad Sci U S A , vol.95 , pp. 15665-15670
    • Doyle, L.A.1    Yang, W.2    Abruzzo, L.V.3
  • 274
    • 0035870289 scopus 로고    scopus 로고
    • Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues
    • Maliepaard M, Scheffer GL, Faneyte IF, et al. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res 2001; 61:3458-3464.
    • (2001) Cancer Res , vol.61 , pp. 3458-3464
    • Maliepaard, M.1    Scheffer, G.L.2    Faneyte, I.F.3
  • 275
    • 0037180446 scopus 로고    scopus 로고
    • The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria
    • Jonker JW, Buitelaar M, Wagenaar E, et al. The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria. Proc Natl Acad Sci U S A 2002; 99:15649-15654.
    • (2002) Proc Natl Acad Sci U S A , vol.99 , pp. 15649-15654
    • Jonker, J.W.1    Buitelaar, M.2    Wagenaar, E.3
  • 276
    • 3342996554 scopus 로고    scopus 로고
    • Functional expression of rat ABCG2 on the luminal side of brain capillaries and its enhancement by astrocyte-derived soluble factor(s)
    • Hon S, Ohtsuki S, Tachikawa M, et al. Functional expression of rat ABCG2 on the luminal side of brain capillaries and its enhancement by astrocyte-derived soluble factor(s). J Neurochem 2004; 90:526-536.
    • (2004) J Neurochem , vol.90 , pp. 526-536
    • Hon, S.1    Ohtsuki, S.2    Tachikawa, M.3
  • 277
    • 11844267237 scopus 로고    scopus 로고
    • Investigation of efflux transport of dehydroepiandrosterone sulfate and mitoxantrone at the mouse blood-brain barrier: A minor role of breast cancer resistance protein
    • Lee YJ, Kusuhara H, Jonker JW, et al. Investigation of efflux transport of dehydroepiandrosterone sulfate and mitoxantrone at the mouse blood-brain barrier: a minor role of breast cancer resistance protein. J Pharmacol Exp Ther 2005; 312: 44-52.
    • (2005) J Pharmacol Exp Ther , vol.312 , pp. 44-52
    • Lee, Y.J.1    Kusuhara, H.2    Jonker, J.W.3
  • 278
    • 0345688604 scopus 로고    scopus 로고
    • Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2)
    • Doyle LA, Ross DD. Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene 2003; 22:7340-7358.
    • (2003) Oncogene , vol.22 , pp. 7340-7358
    • Doyle, L.A.1    Ross, D.D.2
  • 279
    • 14644409841 scopus 로고    scopus 로고
    • The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk
    • Jonker JW, Merino G, Musters S, et al. The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 2005; 11:127-129.
    • (2005) Nat Med , vol.11 , pp. 127-129
    • Jonker, J.W.1    Merino, G.2    Musters, S.3
  • 280
    • 33644682781 scopus 로고    scopus 로고
    • Breast cancer resistance protein (Bcrp/abcg2) is a major determinant of sulfasalazine absorption and elimination in the mouse
    • Zaher H, Khan AA, Palandra J, et al. Breast cancer resistance protein (Bcrp/abcg2) is a major determinant of sulfasalazine absorption and elimination in the mouse. Mol Pharmacol 2006; 3:55-61.
    • (2006) Mol Pharmacol , vol.3 , pp. 55-61
    • Zaher, H.1    Khan, A.A.2    Palandra, J.3
  • 281
    • 33645108568 scopus 로고    scopus 로고
    • Alvarez Al, Pulido MM, et al. Breast cancer resistance protein (BCRP/ ABCG2) transports fluoroquinolone antibiotics and affects their oral availability, pharmacokinetics, and milk secretion
    • Merino G, Alvarez Al, Pulido MM, et al. Breast cancer resistance protein (BCRP/ ABCG2) transports fluoroquinolone antibiotics and affects their oral availability, pharmacokinetics, and milk secretion. Drug Metab Dispos 2006; 34:690-695.
    • (2006) Drug Metab Dispos , vol.34 , pp. 690-695
    • Merino, G.1
  • 282
    • 23944475903 scopus 로고    scopus 로고
    • Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin
    • Hirano M, Maeda K, Matsushima S, et al. Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol Pharmacol 2005; 68:800-807.
    • (2005) Mol Pharmacol , vol.68 , pp. 800-807
    • Hirano, M.1    Maeda, K.2    Matsushima, S.3
  • 283
    • 29744441900 scopus 로고    scopus 로고
    • Breast cancer resistance protein (Bcrpl/Abcg2) reduces systemic exposure of the dietary carcinogens aflatoxin Bl, IQ and Trp-P-1 but also mediates their secretion into breast milk
    • van Herwaarden AE, Wagenaar E, Karnekamp B, et al. Breast cancer resistance protein (Bcrpl/Abcg2) reduces systemic exposure of the dietary carcinogens aflatoxin Bl, IQ and Trp-P-1 but also mediates their secretion into breast milk. Carcinogenesis 2006; 27:123-130.
    • (2006) Carcinogenesis , vol.27 , pp. 123-130
    • van Herwaarden, A.E.1    Wagenaar, E.2    Karnekamp, B.3
  • 284
    • 0141919551 scopus 로고    scopus 로고
    • The breast cancer resistance protein (Bcrpl/Abcg2) restricts exposure to the dietary carcinogen 2-amino-lmethyl-6-phenylimidazo[4,5-b]pyridine
    • van Herwaarden AE, Jonker JW, Wagenaar E, et al. The breast cancer resistance protein (Bcrpl/Abcg2) restricts exposure to the dietary carcinogen 2-amino-lmethyl-6-phenylimidazo[4,5-b]pyridine. Cancer Res 2003; 63:6447-6452.
    • (2003) Cancer Res , vol.63 , pp. 6447-6452
    • van Herwaarden, A.E.1    Jonker, J.W.2    Wagenaar, E.3
  • 285
    • 14944364661 scopus 로고    scopus 로고
    • Role of breast cancer resistance protein (Bcrpl/Abcg2) in the extrusion of glucuronide and sulfate conjugates from enterocytes to intestinal lumen
    • Adachi Y, Suzuki H, Schinkel AH, et al. Role of breast cancer resistance protein (Bcrpl/Abcg2) in the extrusion of glucuronide and sulfate conjugates from enterocytes to intestinal lumen. Mol Pharmacol 2005; 67:923-928.
    • (2005) Mol Pharmacol , vol.67 , pp. 923-928
    • Adachi, Y.1    Suzuki, H.2    Schinkel, A.H.3
  • 286
    • 20944433769 scopus 로고    scopus 로고
    • Lack of improvement of oral absorption of ME3277 by prodrug formation is ascribed to the intestinal efflux mediated by breast cancer resistant protein (BCRP/ABCG2)
    • Kondo C, Onuki R, Kusuhara H, et al. Lack of improvement of oral absorption of ME3277 by prodrug formation is ascribed to the intestinal efflux mediated by breast cancer resistant protein (BCRP/ABCG2). Pharm Res 2005; 22:613-618.
    • (2005) Pharm Res , vol.22 , pp. 613-618
    • Kondo, C.1    Onuki, R.2    Kusuhara, H.3
  • 287
    • 17844365262 scopus 로고    scopus 로고
    • The breast cancer resistance protein (BCRP/ABCG2) affects pharmacokinetics, hepatobiliary excretion, and milk secretion of the antibiotic nitrofurantoin
    • Merino G, Jonker JW, Wagenaar E, et al. The breast cancer resistance protein (BCRP/ABCG2) affects pharmacokinetics, hepatobiliary excretion, and milk secretion of the antibiotic nitrofurantoin. Mol Pharmacol 2005; 67:1758-1764.
    • (2005) Mol Pharmacol , vol.67 , pp. 1758-1764
    • Merino, G.1    Jonker, J.W.2    Wagenaar, E.3
  • 288
    • 4143142207 scopus 로고    scopus 로고
    • Mechanism of the pharmacokinetic interaction between methotrexate and benzimidazoles: Potential role for breast cancer resistance protein in clinical drug-drug interactions
    • Breedveld P, Zelcer N, Pluim D, et al. Mechanism of the pharmacokinetic interaction between methotrexate and benzimidazoles: potential role for breast cancer resistance protein in clinical drug-drug interactions. Cancer Res 2004; 64:5804-5811.
    • (2004) Cancer Res , vol.64 , pp. 5804-5811
    • Breedveld, P.1    Zelcer, N.2    Pluim, D.3
  • 289
    • 16844384057 scopus 로고    scopus 로고
    • The effect of Bcrpl (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): Implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients
    • Breedveld P, Pluim D, Cipriani G, et al. The effect of Bcrpl (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients. Cancer Res 2005; 65:2577-2582.
    • (2005) Cancer Res , vol.65 , pp. 2577-2582
    • Breedveld, P.1    Pluim, D.2    Cipriani, G.3
  • 290
    • 4143107239 scopus 로고    scopus 로고
    • Impaired renal excretion of 6-hydroxy-5,7-dimethyl-2-methylamino-4-(3-pyridylmethyl) benzothiazole (e3040) sulfate in breast cancer resistance protein (bcrpl/abcg2) knockout mice
    • Mizuno N, Suzuki M, Kusuhara H, et al. Impaired renal excretion of 6-hydroxy-5,7-dimethyl-2-methylamino-4-(3-pyridylmethyl) benzothiazole (e3040) sulfate in breast cancer resistance protein (bcrpl/abcg2) knockout mice. Drug Metab Dispos 2004; 32:898-901.
    • (2004) Drug Metab Dispos , vol.32 , pp. 898-901
    • Mizuno, N.1    Suzuki, M.2    Kusuhara, H.3
  • 291
    • 0025092259 scopus 로고
    • Interactions in the renal and biliary elimination of digoxin: Stereoselective difference between quinine and quinidine
    • Hedman A, Angelin B, Arvidsson A, et al. Interactions in the renal and biliary elimination of digoxin: stereoselective difference between quinine and quinidine. Clin Pharmacol Ther 1990; 47:20-26.
    • (1990) Clin Pharmacol Ther , vol.47 , pp. 20-26
    • Hedman, A.1    Angelin, B.2    Arvidsson, A.3
  • 292
    • 0026092531 scopus 로고
    • Digoxin-verapamil interaction: Reduction of biliary but not renal digoxin clearance in humans
    • Hedman A, Angelin B, Arvidsson A, et al. Digoxin-verapamil interaction: reduction of biliary but not renal digoxin clearance in humans. Clin Pharmacol Ther 1991; 49:256-262.
    • (1991) Clin Pharmacol Ther , vol.49 , pp. 256-262
    • Hedman, A.1    Angelin, B.2    Arvidsson, A.3
  • 293
    • 0022338030 scopus 로고
    • Effects of verapamil, diltiazem, and nifedipine on plasma levels and renal excretion of digitoxin
    • Kuhlmann J. Effects of verapamil, diltiazem, and nifedipine on plasma levels and renal excretion of digitoxin. Clin Pharmacol Ther 1985; 38:667-673.
    • (1985) Clin Pharmacol Ther , vol.38 , pp. 667-673
    • Kuhlmann, J.1
  • 294
    • 0031836110 scopus 로고    scopus 로고
    • Characterization of the uptake of rocuronium and digoxin in human hepatocytes: Carrier specificity and comparison with in vivo data
    • Olinga P, Merema M, Hof IH, et al. Characterization of the uptake of rocuronium and digoxin in human hepatocytes: carrier specificity and comparison with in vivo data. J Pharmacol Exp Ther 1998; 285:506-510.
    • (1998) J Pharmacol Exp Ther , vol.285 , pp. 506-510
    • Olinga, P.1    Merema, M.2    Hof, I.H.3
  • 295
    • 0031926050 scopus 로고    scopus 로고
    • Stereoselective inhibition by the diastereomers quinidine and quinine of uptake of cardiac glycosides into isolated rat hepatocytes
    • Hedman A, Meijer DK. Stereoselective inhibition by the diastereomers quinidine and quinine of uptake of cardiac glycosides into isolated rat hepatocytes. J Pharm Sei 1998; 87:457-461.
    • (1998) J Pharm Sei , vol.87 , pp. 457-461
    • Hedman, A.1    Meijer, D.K.2
  • 296
    • 0027818964 scopus 로고
    • Role of P-glycoprotein in renal tubular secretion of digoxin in the isolated perfused rat kidney
    • Hon R, Okamura N, Aiba T, et al. Role of P-glycoprotein in renal tubular secretion of digoxin in the isolated perfused rat kidney. J Pharmacol Exp Ther 1993; 266:1620-1625.
    • (1993) J Pharmacol Exp Ther , vol.266 , pp. 1620-1625
    • Hon, R.1    Okamura, N.2    Aiba, T.3
  • 297
    • 0033514320 scopus 로고    scopus 로고
    • Inhibition of P-glycoprotein-mediated drug transport: A unifying mechanism to explain the interaction between digoxin and quinidine [see comments]
    • Fromm MF, Kim RB, Stein CM, et al. Inhibition of P-glycoprotein-mediated drug transport: a unifying mechanism to explain the interaction between digoxin and quinidine [see comments]. Circulation 1999; 99:552-557.
    • (1999) Circulation , vol.99 , pp. 552-557
    • Fromm, M.F.1    Kim, R.B.2    Stein, C.M.3
  • 298
    • 0031900997 scopus 로고    scopus 로고
    • Role of P-glycoprotein as a secretory mechanism in quinidine absorption from rat small intestine
    • Emi Y, Tsunashima D, Ogawara K, et al. Role of P-glycoprotein as a secretory mechanism in quinidine absorption from rat small intestine. J Pharm Sei 1998; 87:295-299.
    • (1998) J Pharm Sei , vol.87 , pp. 295-299
    • Emi, Y.1    Tsunashima, D.2    Ogawara, K.3
  • 299
    • 1642457252 scopus 로고    scopus 로고
    • Functional characterization of pH-sensitive organic anion transporting polypeptide OATP-B in human
    • Nozawa T, Imai K, Nezu J, et al. Functional characterization of pH-sensitive organic anion transporting polypeptide OATP-B in human. J Pharmacol Exp Ther 2004; 308:438-445.
    • (2004) J Pharmacol Exp Ther , vol.308 , pp. 438-445
    • Nozawa, T.1    Imai, K.2    Nezu, J.3
  • 300
    • 24944482460 scopus 로고    scopus 로고
    • Contribution of oatp (organic aniontransporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans
    • Shimizu M, Fuse K, Okudaira K, et al. Contribution of oatp (organic aniontransporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans. Drug Metab Dispos 2005; 33:1477-1481.
    • (2005) Drug Metab Dispos , vol.33 , pp. 1477-1481
    • Shimizu, M.1    Fuse, K.2    Okudaira, K.3
  • 301
    • 33645798634 scopus 로고    scopus 로고
    • Inhibition of oat3-mediated renal uptake as mechanism for drug-drug interaction between fexofenadine and probenecid
    • Tahara H, Kusuhara H, Maeda K, et al. Inhibition of oat3-mediated renal uptake as mechanism for drug-drug interaction between fexofenadine and probenecid. Drug Metab Dispos 2006; 34:743-747.
    • (2006) Drug Metab Dispos , vol.34 , pp. 743-747
    • Tahara, H.1    Kusuhara, H.2    Maeda, K.3
  • 302
    • 22344436690 scopus 로고    scopus 로고
    • P-glycoprotein plays a major role in the efflux of fexofenadine in the small intestine and blood-brain barrier, but only a limited role in its biliary excretion
    • Tahara H, Kusuhara H, Fuse E, et al. P-glycoprotein plays a major role in the efflux of fexofenadine in the small intestine and blood-brain barrier, but only a limited role in its biliary excretion. Drug Metab Dispos 2005; 33:963-968.
    • (2005) Drug Metab Dispos , vol.33 , pp. 963-968
    • Tahara, H.1    Kusuhara, H.2    Fuse, E.3
  • 303
    • 33751095399 scopus 로고    scopus 로고
    • Lack of dose-dependent effects of itraconazole on the pharmacokinetic interaction with fexofenadine
    • Uno T, Shimizu M, Sugawara K, et al. Lack of dose-dependent effects of itraconazole on the pharmacokinetic interaction with fexofenadine. Drug Metab Dispos 2006; 34:1875-1879.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1875-1879
    • Uno, T.1    Shimizu, M.2    Sugawara, K.3
  • 304
    • 33644513697 scopus 로고    scopus 로고
    • The effect of short- and long-term administration of verapamil on the disposition of cytochrome P450 3A and P-glycoprotein substrates
    • Lemma GL, Wang Z, Hamman MA, et al. The effect of short- and long-term administration of verapamil on the disposition of cytochrome P450 3A and P-glycoprotein substrates. Clin Pharmacol Ther 2006; 79:218-230.
    • (2006) Clin Pharmacol Ther , vol.79 , pp. 218-230
    • Lemma, G.L.1    Wang, Z.2    Hamman, M.A.3
  • 305
    • 33745070264 scopus 로고    scopus 로고
    • Time-dependent interaction between lopinavir/ritonavir and fexofenadine
    • van Heeswijk RP, Bourbeau M, Campbell P, et al. Time-dependent interaction between lopinavir/ritonavir and fexofenadine. J Clin Pharmacol 2006; 46:758-767.
    • (2006) J Clin Pharmacol , vol.46 , pp. 758-767
    • van Heeswijk, R.P.1    Bourbeau, M.2    Campbell, P.3
  • 306
    • 0037464458 scopus 로고    scopus 로고
    • Comparison of in vitro P-glycoprotein screening assays: Recommendations for their use in drug discovery
    • Schwab D, Fischer H, Tabatabaei A, et al. Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery. J Med Chem 2003; 46:1716-1725.
    • (2003) J Med Chem , vol.46 , pp. 1716-1725
    • Schwab, D.1    Fischer, H.2    Tabatabaei, A.3
  • 307
    • 0036135397 scopus 로고    scopus 로고
    • Interaction of common azole antifungals with P glycoprotein
    • Wang EJ, Lew K, Casciano CN, et al. Interaction of common azole antifungals with P glycoprotein. Antimicrob Agents Chemother 2002; 46:160-165.
    • (2002) Antimicrob Agents Chemother , vol.46 , pp. 160-165
    • Wang, E.J.1    Lew, K.2    Casciano, C.N.3
  • 308
    • 4344594700 scopus 로고    scopus 로고
    • Transport characteristics of fexofenadine in the Caco-2 cell model
    • Petri N, Tannergren C, Rungstad D, et al. Transport characteristics of fexofenadine in the Caco-2 cell model. Pharm Res 2004; 21:1398-1404.
    • (2004) Pharm Res , vol.21 , pp. 1398-1404
    • Petri, N.1    Tannergren, C.2    Rungstad, D.3
  • 309
    • 0034121122 scopus 로고    scopus 로고
    • Pharmacological inhibition of P-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes
    • Choo EF, Leake B, Wandel C, et al. Pharmacological inhibition of P-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes. Drug Metab Dispos 2000; 28:655-660.
    • (2000) Drug Metab Dispos , vol.28 , pp. 655-660
    • Choo, E.F.1    Leake, B.2    Wandel, C.3
  • 310
    • 13944275994 scopus 로고    scopus 로고
    • Effect of grapefruit juice volume on the reduction of fexofenadine bioavailability: Possible role of organic anion transporting polypeptides
    • Dresser GK, Kim RB, Bailey DG. Effect of grapefruit juice volume on the reduction of fexofenadine bioavailability: possible role of organic anion transporting polypeptides. Clin Pharmacol Ther 2005; 77:170-177.
    • (2005) Clin Pharmacol Ther , vol.77 , pp. 170-177
    • Dresser, G.K.1    Kim, R.B.2    Bailey, D.G.3
  • 311
    • 0036736505 scopus 로고    scopus 로고
    • Evaluation of peppermint oil and ascorbyl palmitate as inhibitors of cytochrome P4503A4 activity in vitro and in vivo
    • Dresser GK, Wacher V, Wong S, et al. Evaluation of peppermint oil and ascorbyl palmitate as inhibitors of cytochrome P4503A4 activity in vitro and in vivo. Clin Pharmacol Ther 2002; 72:247-255.
    • (2002) Clin Pharmacol Ther , vol.72 , pp. 247-255
    • Dresser, G.K.1    Wacher, V.2    Wong, S.3
  • 312
    • 0036091282 scopus 로고    scopus 로고
    • Grapefruit juice reduces the oral bioavailability of fexofenadine but not desloratadine
    • Banfield C, Gupta S, Marino M, et al. Grapefruit juice reduces the oral bioavailability of fexofenadine but not desloratadine. Clin Pharmacokinet 2002; 41:311-318.
    • (2002) Clin Pharmacokinet , vol.41 , pp. 311-318
    • Banfield, C.1    Gupta, S.2    Marino, M.3
  • 313
    • 0033064071 scopus 로고    scopus 로고
    • Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients
    • Muck W, Mai I, Fritsche L, et al. Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporine-treated kidney transplant recipients. Clin Pharmacol Ther 1999; 65:251-261.
    • (1999) Clin Pharmacol Ther , vol.65 , pp. 251-261
    • Muck, W.1    Mai, I.2    Fritsche, L.3
  • 314
    • 0037310871 scopus 로고    scopus 로고
    • Inhibition of transporter-mediated hepatic uptake as a mechanism for drug-drug interaction between cerivastatin and cyclosporin A
    • Shitara Y, Itoh T, Sato H, et al. Inhibition of transporter-mediated hepatic uptake as a mechanism for drug-drug interaction between cerivastatin and cyclosporin A. J Pharmacol Exp Ther 2003; 304:610-616.
    • (2003) J Pharmacol Exp Ther , vol.304 , pp. 610-616
    • Shitara, Y.1    Itoh, T.2    Sato, H.3
  • 315
    • 0038209381 scopus 로고    scopus 로고
    • Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: Consequences for pravastatin pharmacokinetics
    • Nishizato Y, Ieiri I, Suzuki H, et al. Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin Pharmacol Ther 2003; 73:554-565.
    • (2003) Clin Pharmacol Ther , vol.73 , pp. 554-565
    • Nishizato, Y.1    Ieiri, I.2    Suzuki, H.3
  • 316
    • 25844530136 scopus 로고    scopus 로고
    • Effect of OATP1B1 (SLCOIBI) variant alleles on the pharmacokinetics of pitavastatin in healthy volunteers
    • Chung JY, Cho JY, Yu KS, et al. Effect of OATP1B1 (SLCOIBI) variant alleles on the pharmacokinetics of pitavastatin in healthy volunteers. Clin Pharmacol Ther 2005; 78:342-350.
    • (2005) Clin Pharmacol Ther , vol.78 , pp. 342-350
    • Chung, J.Y.1    Cho, J.Y.2    Yu, K.S.3
  • 317
    • 33751096467 scopus 로고    scopus 로고
    • SLCOIBI polymorphism markedly affects the pharmacokinetics of simvastatin acid
    • Pasanen MK, Neuvonen M, Neuvonen PJ, et al. SLCOIBI polymorphism markedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet Genom 2006; 16:873-879.
    • (2006) Pharmacogenet Genom , vol.16 , pp. 873-879
    • Pasanen, M.K.1    Neuvonen, M.2    Neuvonen, P.J.3
  • 318
    • 33749049136 scopus 로고    scopus 로고
    • SLCOIBI polymorphism and sex affect the pharmacokinetics of pravastatin but not fluvastatin
    • Niemi M, Pasanen MK, Neuvonen PJ. SLCOIBI polymorphism and sex affect the pharmacokinetics of pravastatin but not fluvastatin. Clin Pharmacol Ther 2006; 80:356-366.
    • (2006) Clin Pharmacol Ther , vol.80 , pp. 356-366
    • Niemi, M.1    Pasanen, M.K.2    Neuvonen, P.J.3
  • 319
    • 33748042671 scopus 로고    scopus 로고
    • Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: Drugdrug interactions and interindividual differences in transporter and metabolic enzyme functions
    • Shitara Y, Sugiyama Y. Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drugdrug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol Ther 2006; 112:71-105.
    • (2006) Pharmacol Ther , vol.112 , pp. 71-105
    • Shitara, Y.1    Sugiyama, Y.2
  • 320
    • 33646205286 scopus 로고    scopus 로고
    • Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril
    • Maeda K, Ieiri I, Yasuda K, et al. Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril. Clin Pharmacol Ther 2006; 79:427-439.
    • (2006) Clin Pharmacol Ther , vol.79 , pp. 427-439
    • Maeda, K.1    Ieiri, I.2    Yasuda, K.3
  • 321
    • 20444466590 scopus 로고    scopus 로고
    • Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics
    • Niemi M, Backman JT, Kajosaari LI, et al. Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin Pharmacol Ther 2005; 77:468-478.
    • (2005) Clin Pharmacol Ther , vol.77 , pp. 468-478
    • Niemi, M.1    Backman, J.T.2    Kajosaari, L.I.3
  • 322
    • 25844438751 scopus 로고    scopus 로고
    • Cyclosporine markedly raises the plasma concentrations of repaglinide
    • Kajosaari LI, Niemi M, Neuvonen M, et al. Cyclosporine markedly raises the plasma concentrations of repaglinide. Clin Pharmacol Ther 2005; 78:388-399.
    • (2005) Clin Pharmacol Ther , vol.78 , pp. 388-399
    • Kajosaari, L.I.1    Niemi, M.2    Neuvonen, M.3
  • 323
    • 4644301418 scopus 로고    scopus 로고
    • Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATPlBl:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: Analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil
    • Shitara Y, Hirano M, Sato H, et al. Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATPlBl:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug-drug interaction between cerivastatin and gemfibrozil. J Pharmacol Exp Ther 2004; 311:228-236.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 228-236
    • Shitara, Y.1    Hirano, M.2    Sato, H.3
  • 324
    • 29944446317 scopus 로고    scopus 로고
    • Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: Implications for drug-drug interactions
    • Ogilvie BW, Zhang D, Li W, et al. Glucuronidation converts gemfibrozil to a potent, metabolism-dependent inhibitor of CYP2C8: implications for drug-drug interactions. Drug Metab Dispos 2006; 34:191-197.
    • (2006) Drug Metab Dispos , vol.34 , pp. 191-197
    • Ogilvie, B.W.1    Zhang, D.2    Li, W.3
  • 325
    • 0036146750 scopus 로고    scopus 로고
    • Multiple interactions of Cimetidine and probenecid with valaciclovir and its metabolite acyclovir
    • De Bony F, Tod M, Bidault R, et al. Multiple interactions of Cimetidine and probenecid with valaciclovir and its metabolite acyclovir. Antimicrob Agents Chemother 2002; 46:458-463.
    • (2002) Antimicrob Agents Chemother , vol.46 , pp. 458-463
    • de Bony, F.1    Tod, M.2    Bidault, R.3
  • 326
    • 0020325272 scopus 로고
    • Effects of probenecid on the pharmacokinetics and elimination of acyclovir in humans
    • Laskin OL, de Miranda P, King DH, et al. Effects of probenecid on the pharmacokinetics and elimination of acyclovir in humans. Antimicrob Agents Chemother 1982; 21:804-807.
    • (1982) Antimicrob Agents Chemother , vol.21 , pp. 804-807
    • Laskin, O.L.1    de Miranda, P.2    King, D.H.3
  • 327
    • 0027233424 scopus 로고
    • Effects of probenecid on the pharmacokinetics of allopurinol riboside
    • Were JB, Shapiro TA. Effects of probenecid on the pharmacokinetics of allopurinol riboside. Antimicrob Agents Chemother 1993; 37:1193-1196.
    • (1993) Antimicrob Agents Chemother , vol.37 , pp. 1193-1196
    • Were, J.B.1    Shapiro, T.A.2
  • 328
    • 0021082401 scopus 로고
    • Coupling between renal tubular secretion and effect of bumetanide
    • Odlind B, Beermann B, Lindstrom B. Coupling between renal tubular secretion and effect of bumetanide. Clin Pharmacol Ther 1983; 34:805-809.
    • (1983) Clin Pharmacol Ther , vol.34 , pp. 805-809
    • Odlind, B.1    Beermann, B.2    Lindstrom, B.3
  • 330
    • 0024519265 scopus 로고
    • Pharmacokinetics of intravenously administered cefmetazole and cefoxitin and effects of probenecid on cefmetazole elimination
    • Ko H, Cathcart KS, Griffith DL, et al. Pharmacokinetics of intravenously administered cefmetazole and cefoxitin and effects of probenecid on cefmetazole elimination. Antimicrob Agents Chemother 1989; 33:356-361.
    • (1989) Antimicrob Agents Chemother , vol.33 , pp. 356-361
    • Ko, H.1    Cathcart, K.S.2    Griffith, D.L.3
  • 331
    • 0018893606 scopus 로고
    • Effect of orally administered probenecid on the pharmacokinetics of cefoxitin
    • Vlasses PH, Holbrook AM, Schrogie JJ, et al. Effect of orally administered probenecid on the pharmacokinetics of cefoxitin. Antimicrob Agents Chemother 1980; 17:847-855.
    • (1980) Antimicrob Agents Chemother , vol.17 , pp. 847-855
    • Vlasses, P.H.1    Holbrook, A.M.2    Schrogie, J.J.3
  • 332
    • 0026640488 scopus 로고
    • Pharmacokinetic interactions of cefprozil with food, propantheline, metoclopramide, and probenecid in healthy volunteers
    • Shukla UA, Pittman KA, Barbhaiya RH. Pharmacokinetic interactions of cefprozil with food, propantheline, metoclopramide, and probenecid in healthy volunteers. J Clin Pharmacol 1992; 32:725-731.
    • (1992) J Clin Pharmacol , vol.32 , pp. 725-731
    • Shukla, U.A.1    Pittman, K.A.2    Barbhaiya, R.H.3
  • 333
    • 0020520248 scopus 로고
    • Effect of probenecid on the pharmacokinetics of ceftizoxime
    • LeBel M, Paone RP, Lewis GP. Effect of probenecid on the pharmacokinetics of ceftizoxime. J Antimicrob Chemother 1983; 12:147-155.
    • (1983) J Antimicrob Chemother , vol.12 , pp. 147-155
    • Lebel, M.1    Paone, R.P.2    Lewis, G.P.3
  • 334
    • 0031438533 scopus 로고    scopus 로고
    • Comparison of dose doubling with probenecid for sustaining serum cefuroxime levels
    • Garton AM, Rennie RP, Gilpin J, et al. Comparison of dose doubling with probenecid for sustaining serum cefuroxime levels. J Antimicrob Chemother 1997; 40: 903-906.
    • (1997) J Antimicrob Chemother , vol.40 , pp. 903-906
    • Garton, A.M.1    Rennie, R.P.2    Gilpin, J.3
  • 335
    • 0029000871 scopus 로고
    • Clinical pharmacokinetics of cidofovir in human immunodeficiency virus-infected patients
    • Cundy KC, Petty BG, Flaherty J, et al. Clinical pharmacokinetics of cidofovir in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother 1995; 39:1247-1252.
    • (1995) Antimicrob Agents Chemother , vol.39 , pp. 1247-1252
    • Cundy, K.C.1    Petty, B.G.2    Flaherty, J.3
  • 336
    • 0028822282 scopus 로고
    • Effect of probenecid on the distribution and elimination of ciprofloxacin in humans
    • Jaehde U, Sorgel F, Reiter A, et al. Effect of probenecid on the distribution and elimination of ciprofloxacin in humans. Clin Phamacol Ther 1995; 58:532-541.
    • (1995) Clin Phamacol Ther , vol.58 , pp. 532-541
    • Jaehde, U.1    Sorgel, F.2    Reiter, A.3
  • 337
    • 0025055624 scopus 로고
    • The inhibitory effect of probenecid on renal excretion of famotidine in young, healthy volunteers
    • Inotsume N, Nishimura M, Nakano M, et al. The inhibitory effect of probenecid on renal excretion of famotidine in young, healthy volunteers. J Clin Pharmacol 1990; 30:50-56.
    • (1990) J Clin Pharmacol , vol.30 , pp. 50-56
    • Inotsume, N.1    Nishimura, M.2    Nakano, M.3
  • 338
    • 11344279702 scopus 로고    scopus 로고
    • Different effects of three transporting inhibitors, verapamil, Cimetidine, and probenecid, on fexofenadine pharmacokinetics
    • Yasui-Furukori N, Uno T, Sugawara K, et al. Different effects of three transporting inhibitors, verapamil, Cimetidine, and probenecid, on fexofenadine pharmacokinetics. Clin Pharmacol Ther 2005; 77:17-23.
    • (2005) Clin Pharmacol Ther , vol.77 , pp. 17-23
    • Yasui-Furukori, N.1    Uno, T.2    Sugawara, K.3
  • 340
    • 0036135044 scopus 로고    scopus 로고
    • The anti-influenza drug oseltamivir exhibits low potential to induce pharmacokinetic drug interactions via renal secretion-correlation of in vivo and in vitro studies
    • Hill G, Cihlar T, Oo C, et al. The anti-influenza drug oseltamivir exhibits low potential to induce pharmacokinetic drug interactions via renal secretion-correlation of in vivo and in vitro studies. Drug Metab Dispos 2002; 30:13-19.
    • (2002) Drug Metab Dispos , vol.30 , pp. 13-19
    • Hill, G.1    Cihlar, T.2    Oo, C.3
  • 342
    • 0017652767 scopus 로고
    • Effects of probenecid on furosemide kinetics and natriuresis in man
    • Honari J, Blair AD, Cutler RE. Effects of probenecid on furosemide kinetics and natriuresis in man. Clin Pharmacol Ther 1977; 22:395-401.
    • (1977) Clin Pharmacol Ther , vol.22 , pp. 395-401
    • Honari, J.1    Blair, A.D.2    Cutler, R.E.3
  • 343
    • 0034662273 scopus 로고    scopus 로고
    • Fluorescence-based assay for the interaction of small molecules with the human renal organic anion transporter 1
    • Cihlar T, Ho ES. Fluorescence-based assay for the interaction of small molecules with the human renal organic anion transporter 1. Anal Biochem 2000; 283:49-55.
    • (2000) Anal Biochem , vol.283 , pp. 49-55
    • Cihlar, T.1    Ho, E.S.2
  • 344
    • 0037213731 scopus 로고    scopus 로고
    • Urate transport via human PAH transporter hOATl and its gene structure
    • Ichida K, Hosoyamada M, Kimura H, et al. Urate transport via human PAH transporter hOATl and its gene structure. Kidney Int 2003; 63:143-155.
    • (2003) Kidney Int , vol.63 , pp. 143-155
    • Ichida, K.1    Hosoyamada, M.2    Kimura, H.3
  • 345
    • 0034008388 scopus 로고    scopus 로고
    • Cytotoxicity of antiviral nucleotides adefovir and cidofovir is induced by the expression of human renal organic anion transporter 1
    • Ho ES, Lin DC, Mendel DB, et al. Cytotoxicity of antiviral nucleotides adefovir and cidofovir is induced by the expression of human renal organic anion transporter 1. J Am Soc Nephrol 2000; 11:383-393.
    • (2000) J am Soc Nephrol , vol.11 , pp. 383-393
    • Ho, E.S.1    Lin, D.C.2    Mendel, D.B.3
  • 346
    • 0024511555 scopus 로고
    • The effect of probenecid on the renal elimination of Cimetidine
    • Gisclon LG, Boyd RA, Williams RL, et al. The effect of probenecid on the renal elimination of Cimetidine. Clin Pharmacol Ther 1989; 45:444-452.
    • (1989) Clin Pharmacol Ther , vol.45 , pp. 444-452
    • Gisclon, L.G.1    Boyd, R.A.2    Williams, R.L.3
  • 348
    • 0023838424 scopus 로고
    • Kinetic studies on the competition between famotidine and Cimetidine in rats. Evidence of multiple renal secretory systems for organic cations
    • Lin JH, Los LE, Ulm EH, et al. Kinetic studies on the competition between famotidine and Cimetidine in rats. Evidence of multiple renal secretory systems for organic cations. Drug Metab Dispos 1988; 16:52-56.
    • (1988) Drug Metab Dispos , vol.16 , pp. 52-56
    • Lin, J.H.1    Los, L.E.2    Ulm, E.H.3
  • 349
    • 0031923577 scopus 로고    scopus 로고
    • A physiologically based kidney model for the renal clearance of ranitidine and the interaction with Cimetidine and probenecid in the dog
    • Boom SP, Meyer I, Wouterse AC, et al. A physiologically based kidney model for the renal clearance of ranitidine and the interaction with Cimetidine and probenecid in the dog. Biopharm Drug Dispos 1998; 19:199-208.
    • (1998) Biopharm Drug Dispos , vol.19 , pp. 199-208
    • Boom, S.P.1    Meyer, I.2    Wouterse, A.C.3
  • 351
    • 0023129704 scopus 로고
    • Drug inhibition of penicillin tubular secretion: Concordance between in vitro and clinical findings
    • Nierenerg DW. Drug inhibition of penicillin tubular secretion: concordance between in vitro and clinical findings. J Pharmacol Exp Ther 1987; 240:712-716.
    • (1987) J Pharmacol Exp Ther , vol.240 , pp. 712-716
    • Nierenerg, D.W.1
  • 352
    • 0016413786 scopus 로고
    • Inhibition of renal tubular transport of methotrexate by probenecid
    • Bourke RS, Chheda G, Bremer A, et al. Inhibition of renal tubular transport of methotrexate by probenecid. Cancer Res 1975; 35:110-116.
    • (1975) Cancer Res , vol.35 , pp. 110-116
    • Bourke, R.S.1    Chheda, G.2    Bremer, A.3
  • 353
    • 0021364392 scopus 로고
    • Effect of penicillin on the renal tubular secretion of methotrexate in the monkey
    • Williams WM, Chen TS, Huang KC. Effect of penicillin on the renal tubular secretion of methotrexate in the monkey. Cancer Res 1984; 44:1913-1917.
    • (1984) Cancer Res , vol.44 , pp. 1913-1917
    • Williams, W.M.1    Chen, T.S.2    Huang, K.C.3
  • 354
    • 0036076780 scopus 로고    scopus 로고
    • Characterization of methotrexate transport and its drug interactions with human organic anion transporters
    • Takeda M, Khamdang S, Narikawa S, et al. Characterization of methotrexate transport and its drug interactions with human organic anion transporters. J Pharmacol Exp Ther 2002; 302:666-671.
    • (2002) J Pharmacol Exp Ther , vol.302 , pp. 666-671
    • Takeda, M.1    Khamdang, S.2    Narikawa, S.3
  • 355
    • 1642388976 scopus 로고    scopus 로고
    • Quantitative evaluation of the drug-drug interactions between methotrexate and nonsteroidal anti-inflammatory drugs in the renal uptake process based on the contribution of organic anion transporters and reduced folate carrier
    • Nozaki Y, Kusuhara H, Endou H, et al. Quantitative evaluation of the drug-drug interactions between methotrexate and nonsteroidal anti-inflammatory drugs in the renal uptake process based on the contribution of organic anion transporters and reduced folate carrier. J Pharmacol Exp Ther 2004; 309:226-234.
    • (2004) J Pharmacol Exp Ther , vol.309 , pp. 226-234
    • Nozaki, Y.1    Kusuhara, H.2    Endou, H.3
  • 356
    • 33845876683 scopus 로고    scopus 로고
    • Interaction of nonsteroidal anti-inflammatory drugs with multidrug resistance protein (MRP) 2/ABCC2- and MRP4/ABCC4-mediated methotrexate transport
    • El-Sheikh AA, van den Heuvel JJ, Koenderink JB, et al. Interaction of nonsteroidal anti-inflammatory drugs with multidrug resistance protein (MRP) 2/ABCC2- and MRP4/ABCC4-mediated methotrexate transport. J Pharmacol Exp Ther 2007; 320:229-235.
    • (2007) J Pharmacol Exp Ther , vol.320 , pp. 229-235
    • El-Sheikh, A.A.1    van den Heuvel, J.J.2    Koenderink, J.B.3
  • 357
    • 0025868537 scopus 로고
    • Dose-dependent absorption and elimination of Cefadroxil in man
    • Garrigues TM, Martin U, Peris-Ribera JE, et al. Dose-dependent absorption and elimination of Cefadroxil in man. Eur J Clin Pharmacol 1991; 41:179-183.
    • (1991) Eur J Clin Pharmacol , vol.41 , pp. 179-183
    • Garrigues, T.M.1    Martin, U.2    Peris-Ribera, J.E.3
  • 358
    • 0026758261 scopus 로고
    • Structural specificity of mucosal-cell transport and metabolism of peptide drugs: Implication for oral peptide drug delivery
    • Bai JPF, Amidon GL. Structural specificity of mucosal-cell transport and metabolism of peptide drugs: implication for oral peptide drug delivery. Pharm Res 1992; 9:969-978.
    • (1992) Pharm Res , vol.9 , pp. 969-978
    • Bai, J.P.F.1    Amidon, G.L.2
  • 359
    • 0023807777 scopus 로고
    • Amidon GL. Characterization of the oral absorption of beta-lactam antibiotics. I. Cephalosporins: Determination of intrinsic membrane absorption parameters in the rat intestine in situ
    • Sinko PJ, Amidon GL. Characterization of the oral absorption of beta-lactam antibiotics. I. Cephalosporins: determination of intrinsic membrane absorption parameters in the rat intestine in situ. Pharm Res 1988; 5:645-650.
    • (1988) Pharm Res , vol.5 , pp. 645-650
    • Sinko, P.J.1
  • 360
    • 0030782511 scopus 로고    scopus 로고
    • Positively cooperative sites for drug transport by P-glycoprotein with distinct drug specificities
    • Shapiro AB, Ling V. Positively cooperative sites for drug transport by P-glycoprotein with distinct drug specificities. Eur J Biochem 1997; 250:130-137.
    • (1997) Eur J Biochem , vol.250 , pp. 130-137
    • Shapiro, A.B.1    Ling, V.2
  • 361
    • 31144434780 scopus 로고    scopus 로고
    • In vivo evaluation of P-glycoprotein function at the blood-brain barrier in nonhuman primates using [11C]verapamil
    • Lee YJ, Maeda J, Kusuhara H, et al. In vivo evaluation of P-glycoprotein function at the blood-brain barrier in nonhuman primates using [11C]verapamil. J Pharmacol Exp Ther 2006; 316:647-653.
    • (2006) J Pharmacol Exp Ther , vol.316 , pp. 647-653
    • Lee, Y.J.1    Maeda, J.2    Kusuhara, H.3
  • 362
    • 0023845228 scopus 로고
    • Protein distribution diet restores motor function in patients with dopa-resistant" off ' period
    • Pincus JH, Barry K. Protein distribution diet restores motor function in patients with dopa-resistant" off ' period. Neurology 1988; 38:481-483.
    • (1988) Neurology , vol.38 , pp. 481-483
    • Pincus, J.H.1    Barry, K.2
  • 363
    • 0022454431 scopus 로고
    • Characterization of the large neutral amino acid transport system of bovine brain microvessel endothelial cell monolayers
    • Audus KL, Borchardt RT. Characterization of the large neutral amino acid transport system of bovine brain microvessel endothelial cell monolayers. J Neurochem 1986; 47:484-488.
    • (1986) J Neurochem , vol.47 , pp. 484-488
    • Audus, K.L.1    Borchardt, R.T.2
  • 364
    • 0023130976 scopus 로고
    • Facilitated transport of melphalan at the rat blood-brain barrier by the large neutral amino acid carrier system
    • Greig NH, Momma S, Sweeney DJ, et al. Facilitated transport of melphalan at the rat blood-brain barrier by the large neutral amino acid carrier system. Cancer Res 1987; 47:1571-1576.
    • (1987) Cancer Res , vol.47 , pp. 1571-1576
    • Greig, N.H.1    Momma, S.2    Sweeney, D.J.3
  • 365
    • 11144358423 scopus 로고    scopus 로고
    • Genipin enhances Mrp2 (Abcc2)-mediated bile formation and organic anion transport in rat liver
    • Shoda J, Miura T, Utsunomiya H, et al. Genipin enhances Mrp2 (Abcc2)-mediated bile formation and organic anion transport in rat liver. Hepatology 2004; 39: 167-178.
    • (2004) Hepatology , vol.39 , pp. 167-178
    • Shoda, J.1    Miura, T.2    Utsunomiya, H.3
  • 366
    • 33745774784 scopus 로고    scopus 로고
    • Role of nuclear receptors in the adaptive response to bile acids and cholestasis: Pathogenetic and therapeutic considerations
    • Zollner G, Marschall HU, Wagner M, et al. Role of nuclear receptors in the adaptive response to bile acids and cholestasis: pathogenetic and therapeutic considerations. Mol Pharmacol 2006; 3:231-251.
    • (2006) Mol Pharmacol , vol.3 , pp. 231-251
    • Zollner, G.1    Marschall, H.U.2    Wagner, M.3
  • 367
    • 30144433535 scopus 로고    scopus 로고
    • Coordinate transcriptional regulation of transport and metabolism
    • Eloranta JJ, Meier PJ, Kullak-Ublick GA. Coordinate transcriptional regulation of transport and metabolism. Methods Enzymol 2005; 400:511-530.
    • (2005) Methods Enzymol , vol.400 , pp. 511-530
    • Eloranta, J.J.1    Meier, P.J.2    Kullak-Ublick, G.A.3
  • 369
    • 33746706762 scopus 로고    scopus 로고
    • PXR and CAR: Nuclear receptors which play a pivotal role in drug disposition and chemical toxicity
    • Stanley LA, Horsburgh BC, Ross J, et al. PXR and CAR: nuclear receptors which play a pivotal role in drug disposition and chemical toxicity. Drug Metab Rev 2006; 38:515-597.
    • (2006) Drug Metab Rev , vol.38 , pp. 515-597
    • Stanley, L.A.1    Horsburgh, B.C.2    Ross, J.3
  • 370
    • 33646713469 scopus 로고    scopus 로고
    • Phénobarbital confers its diverse effects by activating the orphan nuclear receptor car
    • Kodama S, Negishi M. Phénobarbital confers its diverse effects by activating the orphan nuclear receptor car. Drug Metab Rev 2006; 38:75-87.
    • (2006) Drug Metab Rev , vol.38 , pp. 75-87
    • Kodama, S.1    Negishi, M.2
  • 371
    • 33646498651 scopus 로고    scopus 로고
    • The quantitative prediction of in vivo enzymeinduction caused by drug exposure from in vitro information on human hepatocytes
    • Kato M, Chiba K, Horikawa M, et al. The quantitative prediction of in vivo enzymeinduction caused by drug exposure from in vitro information on human hepatocytes. Drug Metab Pharmacokinet 2005; 20:236-243.
    • (2005) Drug Metab Pharmacokinet , vol.20 , pp. 236-243
    • Kato, M.1    Chiba, K.2    Horikawa, M.3
  • 372
    • 0032754284 scopus 로고    scopus 로고
    • The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin
    • Greiner B, Eichelbaum M, Fritz P, et al. The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Invest 1999; 104:147-153.
    • (1999) J Clin Invest , vol.104 , pp. 147-153
    • Greiner, B.1    Eichelbaum, M.2    Fritz, P.3
  • 373
    • 0033753435 scopus 로고    scopus 로고
    • Induction of P-glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: A new type of drug/drug interaction
    • Westphal K, Weinbrenner A, Zschiesche M, et al. Induction of P-glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: a new type of drug/drug interaction. Clin Pharmacol Ther 2000; 68:345-355.
    • (2000) Clin Pharmacol Ther , vol.68 , pp. 345-355
    • Westphal, K.1    Weinbrenner, A.2    Zschiesche, M.3
  • 374
    • 0035107449 scopus 로고    scopus 로고
    • The effect of rifampin administration on the disposition of fexofenadine
    • Hamman MA, Bruce MA, Haehner-Daniels BD, et al. The effect of rifampin administration on the disposition of fexofenadine. Clin Pharmacol Ther 2001; 69:114-121.
    • (2001) Clin Pharmacol Ther , vol.69 , pp. 114-121
    • Hamman, M.A.1    Bruce, M.A.2    Haehner-Daniels, B.D.3
  • 375
    • 0035805536 scopus 로고    scopus 로고
    • Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin
    • Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem 2001; 276:14581-14587.
    • (2001) J Biol Chem , vol.276 , pp. 14581-14587
    • Geick, A.1    Eichelbaum, M.2    Burk, O.3
  • 376
    • 0033724658 scopus 로고    scopus 로고
    • The effect of rifampin treatment on intestinal expression of human MRP transporters
    • Fromm MF, Kauffmann HM, Fritz P, et al. The effect of rifampin treatment on intestinal expression of human MRP transporters. Am J Pathol 2000; 157: 1575-1580.
    • (2000) Am J Pathol , vol.157 , pp. 1575-1580
    • Fromm, M.F.1    Kauffmann, H.M.2    Fritz, P.3
  • 377
    • 0037169551 scopus 로고    scopus 로고
    • Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor
    • Kast HR, Goodwin B, Tarr PT, et al. Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor. J Biol Chem 2002; 277: 2908-2915.
    • (2002) J Biol Chem , vol.277 , pp. 2908-2915
    • Kast, H.R.1    Goodwin, B.2    Tarr, P.T.3
  • 378
    • 0035823528 scopus 로고    scopus 로고
    • Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR
    • Dussault I, Lin M, Hollister K, et al. Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR. J Biol Chem 2001; 276:33309-33312.
    • (2001) J Biol Chem , vol.276 , pp. 33309-33312
    • Dussault, I.1    Lin, M.2    Hollister, K.3
  • 379
    • 33748894676 scopus 로고    scopus 로고
    • Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drugsensing receptors in primary human hepatocytes
    • Jigorel E, Le Vee M, Boursier-Neyret C, et al. Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drugsensing receptors in primary human hepatocytes. Drug Metab Dispos 2006; 34:1756-1763.
    • (2006) Drug Metab Dispos , vol.34 , pp. 1756-1763
    • Jigorel, E.1    Le Vee, M.2    Boursier-Neyret, C.3
  • 380
    • 23044436687 scopus 로고    scopus 로고
    • CAR and PXR agonists stimulate hepatic bile acid and bilirubin detoxification and elimination pathways in mice
    • Wagner M, Halilbasic E, Marschall HU, et al. CAR and PXR agonists stimulate hepatic bile acid and bilirubin detoxification and elimination pathways in mice. Hepatology 2005; 42:420-430.
    • (2005) Hepatology , vol.42 , pp. 420-430
    • Wagner, M.1    Halilbasic, E.2    Marschall, H.U.3
  • 381
    • 0037403746 scopus 로고    scopus 로고
    • Regulation of drug transporter gene expression by nuclear receptors
    • Staudinger JL, Madan A, Carol KM, et al. Regulation of drug transporter gene expression by nuclear receptors. Drug Metab Dispos 2003; 31:523-527.
    • (2003) Drug Metab Dispos , vol.31 , pp. 523-527
    • Staudinger, J.L.1    Madan, A.2    Carol, K.M.3
  • 382
    • 0034982907 scopus 로고    scopus 로고
    • Differential effects of microsomal enzyme-inducing chemicals on the hepatic expression of rat organic anion transporters, OATP1 and OATP2
    • Rausch-Derra LC, Hartley DP, Meier PJ, et al. Differential effects of microsomal enzyme-inducing chemicals on the hepatic expression of rat organic anion transporters, OATP1 and OATP2. Hepatology 2001; 33:1469-1478.
    • (2001) Hepatology , vol.33 , pp. 1469-1478
    • Rausch-Derra, L.C.1    Hartley, D.P.2    Meier, P.J.3
  • 383
    • 23944438892 scopus 로고    scopus 로고
    • Regulation of mouse organic anion-transporting polypeptides (Oatps) in liver by prototypical microsomal enzyme inducers that activate distinct transcription factor pathways
    • Cheng X, Maher J, Dieter MZ, et al. Regulation of mouse organic anion-transporting polypeptides (Oatps) in liver by prototypical microsomal enzyme inducers that activate distinct transcription factor pathways. Drug Metab Dispos 2005; 33:1276-1282.
    • (2005) Drug Metab Dispos , vol.33 , pp. 1276-1282
    • Cheng, X.1    Maher, J.2    Dieter, M.Z.3
  • 384
    • 0036137072 scopus 로고    scopus 로고
    • Organ distribution of multidrug resistance proteins 1, 2, and 3 (Mrpl, 2, and 3) mRNA and hepatic induction of Mrp3 by constitutive androstane receptor activators in rats
    • Cherrington NJ, Hartley DP, Li N, et al. Organ distribution of multidrug resistance proteins 1, 2, and 3 (Mrpl, 2, and 3) mRNA and hepatic induction of Mrp3 by constitutive androstane receptor activators in rats. J Pharmacol Exp Ther 2002; 300:97-104.
    • (2002) J Pharmacol Exp Ther , vol.300 , pp. 97-104
    • Cherrington, N.J.1    Hartley, D.P.2    Li, N.3
  • 385
    • 22344433760 scopus 로고    scopus 로고
    • Induction of the multidrug resistance-associated protein family of transporters by chemical activators of receptor-mediated pathways in mouse liver
    • Maher JM, Cheng X, Slitt AL, et al. Induction of the multidrug resistance-associated protein family of transporters by chemical activators of receptor-mediated pathways in mouse liver. Drug Metab Dispos 2005; 33:956-962.
    • (2005) Drug Metab Dispos , vol.33 , pp. 956-962
    • Maher, J.M.1    Cheng, X.2    Slitt, A.L.3
  • 386
    • 2542440464 scopus 로고    scopus 로고
    • Interactions between hepatic Mrp4 and Sult2a as revealed by the constitutive androstane receptor and Mrp4 knockout mice
    • Assem M, Schuetz EG, Leggas M, et al. Interactions between hepatic Mrp4 and Sult2a as revealed by the constitutive androstane receptor and Mrp4 knockout mice. J Biol Chem 2004; 279:22250-22257.
    • (2004) J Biol Chem , vol.279 , pp. 22250-22257
    • Assem, M.1    Schuetz, E.G.2    Leggas, M.3
  • 387
    • 0034958172 scopus 로고    scopus 로고
    • Effect of phénobarbital on the expression of bile salt and organic anion transporters of rat liver
    • Hagenbuch N, Reichel C, Stieger B, et al. Effect of phénobarbital on the expression of bile salt and organic anion transporters of rat liver. J Hepatol 2001; 34:881-887.
    • (2001) J Hepatol , vol.34 , pp. 881-887
    • Hagenbuch, N.1    Reichel, C.2    Stieger, B.3
  • 388
    • 0142181031 scopus 로고    scopus 로고
    • Induction of multidrug resistance protein 3 (mrp3) in vivo is independent of constitutive androstane receptor
    • Cherrington NJ, Slitt AL, Maher JM, et al. Induction of multidrug resistance protein 3 (mrp3) in vivo is independent of constitutive androstane receptor. Drug Metab Dispos 2003; 31:1315-1319.
    • (2003) Drug Metab Dispos , vol.31 , pp. 1315-1319
    • Cherrington, N.J.1    Slitt, A.L.2    Maher, J.M.3
  • 389
    • 0036328387 scopus 로고    scopus 로고
    • Role of constitutive androstane receptor in the in vivo induction of Mrp3 and CYP2B1/2 by phénobarbital
    • Xiong H, Yoshinari K, Brouwer KL, et al. Role of constitutive androstane receptor in the in vivo induction of Mrp3 and CYP2B1/2 by phénobarbital. Drug Metab Dispos 2002; 30:918-923.
    • (2002) Drug Metab Dispos , vol.30 , pp. 918-923
    • Xiong, H.1    Yoshinari, K.2    Brouwer, K.L.3
  • 390
    • 0036080163 scopus 로고    scopus 로고
    • Human organic anion transporting polypeptide 8 promoter is transactivated by the farnesoid X receptor/bile acid receptor
    • Jung D, Podvinec M, Meyer UA, et al. Human organic anion transporting polypeptide 8 promoter is transactivated by the farnesoid X receptor/bile acid receptor. Gastroenterology 2002; 122:1954-1966.
    • (2002) Gastroenterology , vol.122 , pp. 1954-1966
    • Jung, D.1    Podvinec, M.2    Meyer, U.A.3
  • 391
    • 33744809847 scopus 로고    scopus 로고
    • Farnesoid X receptor, hepatocyte nuclear factors 1 alpha and 3beta are essential for transcriptional activation of the liverspecific organic anion transporter-2 gene
    • Ohtsuka H, Abe T, Onogawa T, et al. Farnesoid X receptor, hepatocyte nuclear factors 1 alpha and 3beta are essential for transcriptional activation of the liverspecific organic anion transporter-2 gene. J Gastroenterol 2006; 41:369-377.
    • (2006) J Gastroenterol , vol.41 , pp. 369-377
    • Ohtsuka, H.1    Abe, T.2    Onogawa, T.3
  • 392
    • 0037371120 scopus 로고    scopus 로고
    • Hepatocyte nuclear factor 1 alpha: A key mediator of the effect of bile acids on gene expression
    • Jung D, Kullak-Ublick GA. Hepatocyte nuclear factor 1 alpha: a key mediator of the effect of bile acids on gene expression. Hepatology 2003; 37:622-631.
    • (2003) Hepatology , vol.37 , pp. 622-631
    • Jung, D.1    Kullak-Ublick, G.A.2
  • 393
    • 0035813209 scopus 로고    scopus 로고
    • Characterization of the human OATP-C (SLC21A6) gene promoter and regulation of liver-specific OATP genes by hepatocyte nuclear factor 1 alpha
    • Jung D, Hagenbuch B, Gresh L, et al. Characterization of the human OATP-C (SLC21A6) gene promoter and regulation of liver-specific OATP genes by hepatocyte nuclear factor 1 alpha. J Biol Chem 2001; 276:37206-37214.
    • (2001) J Biol Chem , vol.276 , pp. 37206-37214
    • Jung, D.1    Hagenbuch, B.2    Gresh, L.3
  • 394
    • 33645865553 scopus 로고    scopus 로고
    • Induction of hepatic transporters multidrug resistance-associated proteins (Mrp) 3 and 4 by clofibrate is regulated by peroxisome proliferator-activated receptor alpha
    • Moffit JS, Aleksunes LM, Maher JM, et al. Induction of hepatic transporters multidrug resistance-associated proteins (Mrp) 3 and 4 by clofibrate is regulated by peroxisome proliferator-activated receptor alpha. J Pharmacol Exp Ther 2006; 317:537-545.
    • (2006) J Pharmacol Exp Ther , vol.317 , pp. 537-545
    • Moffit, J.S.1    Aleksunes, L.M.2    Maher, J.M.3
  • 395
    • 16544381367 scopus 로고    scopus 로고
    • Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies
    • Nishimura M, Naito S, Yokoi T. Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet 2004; 19:135-149.
    • (2004) Drug Metab Pharmacokinet , vol.19 , pp. 135-149
    • Nishimura, M.1    Naito, S.2    Yokoi, T.3
  • 396
    • 34548103547 scopus 로고    scopus 로고
    • Species difference in the inhibitory effect of non-steroidal anti-inflammatory drugs on the uptake of methotrexate by human kidney slices
    • Nozaki Y, Kusuhara H, Kondo T, et al. Species difference in the inhibitory effect of non-steroidal anti-inflammatory drugs on the uptake of methotrexate by human kidney slices. J. Pharmacol Exp Ther 2007; 322:1162-70.
    • (2007) J. Pharmacol Exp Ther , vol.322 , pp. 1162-1170
    • Nozaki, Y.1    Kusuhara, H.2    Kondo, T.3
  • 397
    • 0038265441 scopus 로고    scopus 로고
    • ABCG2 transports sulfated conjugates of steroids and xenobiotics
    • Suzuki M, Suzuki H, Sugimoto Y, et al. ABCG2 transports sulfated conjugates of steroids and xenobiotics. J Biol Chem 2003; 278:22644-9.
    • (2003) J Biol Chem , vol.278 , pp. 22644-22649
    • Suzuki, M.1    Suzuki, H.2    Sugimoto, Y.3
  • 398
    • 0022631053 scopus 로고
    • Selectivity of the cimetidine-induced alterations in the renal handling of organic substrates in humans. Studies with anionic, cationic and zwitterionic drugs
    • Crugten J, Bochner F, Keal J, et al. Selectivity of the cimetidine-induced alterations in the renal handling of organic substrates in humans. Studies with anionic, cationic and zwitterionic drugs. J Pharmacol Exp Ther. 1986; 236:481-7.
    • (1986) J Pharmacol Exp Ther , vol.236 , pp. 481-487
    • Crugten, J.1    Bochner, F.2    Keal, J.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.