Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Vsevolod E. Kostrubsky is active.

Publication


Featured researches published by Vsevolod E. Kostrubsky.


Toxicological Sciences | 2009

Role of Hepatic Transporters in the Disposition and Hepatotoxicity of a HER2 Tyrosine Kinase Inhibitor CP-724,714

Bo Feng; Jinghai J. Xu; Yi-an Bi; Rouchelle Mireles; Ralph E. Davidson; David B. Duignan; Scott D. Campbell; Vsevolod E. Kostrubsky; Margaret C. Dunn; Arthur R. Smith; Huifen F. Wang

CP-724,714, a potent and selective orally active HER2 tyrosine kinase inhibitor, was discontinued from clinical development due to unexpected hepatotoxicity in cancer patients. Based on the clinical manifestation of the toxicity, CP-724,714 likely exerted its hepatotoxicity via both hepatocellular injury and hepatobiliary cholestatic mechanisms. The direct cytotoxic effect, hepatobiliary disposition of CP-724,714, and its inhibition of active canalicular transport of bile constituents were evaluated in established human hepatocyte models and in vitro transporter systems. CP-724,714 exhibited direct cytotoxicity using human hepatocyte imaging assay technology with mitochondria identified as a candidate organelle for its off-target toxicity. Additionally, CP-724,714 was rapidly taken up into human hepatocytes, partially via an active transport process, with an uptake clearance approximately fourfold higher than efflux clearance. The major human hepatic uptake transporter, OATP1B1, and efflux transporters, multidrug resistance protein 1 (MDR1) and breast cancer resistance protein, were involved in hepatobiliary clearance of CP-724,714. Furthermore, CP-724,714 displayed a concentration-dependent inhibition of cholyl-lysyl fluorescein and taurocholate (TC) efflux into canaliculi in cryopreserved and fresh cultured human hepatocytes, respectively. Likewise, CP-724,714 inhibited TC transport in membrane vesicles expressing human bile salt export pump with an IC(50) of 16 microM. Finally, CP-724,714 inhibited the major efflux transporter in bile canaliculi, MDR1, with an IC(50) of approximately 28 microM. These results suggest that inhibition of hepatic efflux transporters contributed to hepatic accumulation of drug and bile constituents leading to hepatocellular injury and hepatobiliary cholestasis. This study provides likely explanations for clinically observed adverse liver effects of CP-724,714.


Biochemical Pharmacology | 1998

Alcohol-mediated increases in acetaminophen hepatotoxicity: role of CYP2E and CYP3A.

J F Sinclair; Elizabeth H. Jeffery; Steven A. Wrighton; Vsevolod E. Kostrubsky; Juliana G. Szakacs; Sheryl G. Wood; Peter R. Sinclair

This commentary focuses on the roles of CYP3A and CYP2E in alcohol-mediated increases in acetaminophen hepatotoxicity. CYP2E has been considered to be the main form of P450 responsible for such toxicity in animals and humans. However, CYP3A, which is also induced by alcohol, has been shown to have a greater affinity for acetaminophen than CYP2E. Previous experiments implicating CYP2E in alcohol-mediated increases in acetaminophen hepatotoxicity have used inhibitors of this form of P450 that are now proving to be non-specific. Triacetyloleandomycin (TAO) is a potent inhibitor of CYP3A that maintains specificity in vitro over a large concentration range. In rats treated with ethanol or the combination of ethanol and isopentanol, the major higher chain alcohol in alcoholic beverages, TAO protects animals from increases in acetaminophen hepatotoxicity, suggesting a major role of CYP3A. CYP2E may not have a major role due to the rapid loss of induced levels in the absence of continued exposure to ethanol. Knockout mice, which are being used to define the role of particular proteins in biological responses, have been developed for CYP2E1 and CYP1A2 but not CYP3A. Cyp2e1(-/-) and Cyp1a2(-/-) mice are more resistant to acetaminophen hepatotoxicity than wild-type strains, even though the amounts of the other forms of P450s are unaltered in the liver. These findings suggest that the relative amounts of P450s and not just kinetic characteristics determine their role in acetaminophen hepatotoxicity. The clinical implications of the findings that CYP3A can have a major role in acetaminophen-mediated hepatotoxicity are discussed.


Journal of Hepatology | 1999

Epidermal growth factor- and hepatocyte growth factor-receptor activity in serum-free cultures of human hepatocytes.

Dorothee M. Runge; Dieter Runge; Kenneth Dorko; Liubomir A. Pisarov; Kerstin Leckel; Vsevolod E. Kostrubsky; David Thomas; Stephen C. Strom; George K. Michalopoulos

BACKGROUND/AIMS Serum-free primary cultures of hepatocytes are a useful tool to study factors triggering hepatocyte proliferation and regeneration. We have developed a chemically defined serum-free system that allows human hepatocyte proliferation in the presence of epidermal growth factor and hepatocyte growth factor. METHODS DNA synthesis and accumulation were determined by [3H]thymidine incorporation and fluorometry, respectively. Western blot analyses and co-immunoprecipitations were used to investigate the association of proteins involved in epidermal growth factor and hepatocyte growth factor activation and signaling: epidermal growth factor receptor, hepatocyte growth factor receptor (MET), urokinase-type plasminogen activator and its receptor, and a member of the signal transducer and activator of transcription family, STAT-3. RESULTS Primary human hepatocytes proliferated under serum-free conditions in a chemically defined medium for up to 12 days. Epidermal growth factor-receptor and MET were present and functional, decreasing over time. MET, urokinase-type plasminogen activator and urokinase-type plasminogen activator receptor co-precipitated to varying degrees during the culture period. STAT-3 co-precipitated with epidermal growth factor-receptor and MET to varying degrees. CONCLUSIONS Proliferation of human hepatocytes can improve by modification of a chemically defined medium originally used for rat hepatocyte cultures. In these long-term cultures of human hepatocytes, hepatocyte growth factor and epidermal growth factor can stimulate growth and differentiation by interacting with their receptors and initiating downstream signaling. This involves complex formation of the receptors with other plasma membrane components for MET (urokinase-type plasminogen activator in context of its receptor) and activation of STAT-3 for both receptors.


Drug Metabolism and Disposition | 2007

Role of CYP3A and CYP2E1 in alcohol-mediated increases in acetaminophen hepatotoxicity : Comparison of wild-type and Cyp2e1(-/-) mice

Kristina K. Wolf; Sheryl G. Wood; Jenna L. Allard; Jane A. Hunt; Nadia Gorman; Brooke W. Walton-Strong; Juliana G. Szakacs; Su X. Duan; Qin Hao; Michael H. Court; Lisa L. von Moltke; David J. Greenblatt; Vsevolod E. Kostrubsky; Elizabeth H. Jeffery; Steven A. Wrighton; Frank J. Gonzalez; Peter R. Sinclair; Jacqueline F. Sinclair

CYP2E1 is widely accepted as the sole form of cytochrome P450 responsible for alcohol-mediated increases in acetaminophen (APAP) hepatotoxicity. However, we previously found that alcohol [ethanol and isopentanol (EIP)] causes increases in APAP hepatotoxicity in Cyp2e1(–/–) mice, indicating that CYP2E1 is not essential. Here, using wild-type and Cyp2e1(–/–) mice, we investigated the relative roles of CYP2E1 and CYP3A in EIP-mediated increases in APAP hepatotoxicity. We found that EIP-mediated increases in APAP hepatotoxicity occurred at lower APAP doses in wild-type mice (300 mg/kg) than in Cyp2e1(–/–) mice (600 mg/kg). Although this result suggests that CYP2E1 has a role in the different susceptibilities of these mouse lines, our findings that EIP-mediated increases in CYP3A activities were greater in wild-type mice compared with Cyp2e1(–/–) mice raises the possibility that differential increases in CYP3A may also contribute to the greater APAP sensitivity in EIP-pretreated wild-type mice. At the time of APAP administration, which followed an 11 h withdrawal from the alcohols, alcohol-induced levels of CYP3A were sustained in both mouse lines, whereas CYP2E1 was decreased to constitutive levels in wild-type mice. The CYP3A inhibitor triacetyloleandomycin (TAO) decreased APAP hepatotoxicity in EIP-pretreated wild-type and Cyp2e1(–/–) mice. TAO treatment in vivo resulted in inhibition of microsomal CYP3A-catalyzed activity, measured in vitro, with no inhibition of CYP1A2 and CYP2E1 activities. In conclusion, these findings suggest that both CYP3A and CYP2E1 contribute to APAP hepatotoxicity in alcohol-treated mice.


Biochemical Pharmacology | 2000

Acetaminophen hepatotoxicity precipitated by short-term treatment of rats with ethanol and isopentanol: Protection by triacetyloleandomycin

Jacqueline F. Sinclair; Juliana G. Szakacs; Sheryl G. Wood; Vsevolod E. Kostrubsky; Elizabeth H. Jeffery; Steven A. Wrighton; William J. Bement; Dane Wright; Peter R. Sinclair

Ethanol and isopentanol are the predominant alcohols in alcoholic beverages. We have reported previously that pretreatment of rats with a liquid diet containing 6.3% ethanol plus 0.5% isopentanol for 7 days results in a synergistic increase in acetaminophen hepatotoxicity, compared with rats treated with either alcohol alone. Here, we investigated the role of CYP3A in acetaminophen hepatotoxicity associated with the combined alcohol treatment. Triacetyloleandomycin, a specific inhibitor of CYP3A, protected rats pretreated with ethanol along with isopentanol from acetaminophen hepatotoxicity. At both 0.25 and 0.5 g acetaminophen/kg, triacetyloleandomycin partially prevented elevations in serum levels of alanine aminotransferase. At 0.25 g acetaminophen/kg, triacetyloleandomycin completely protected 6 of 8 rats from histologically observed liver damage, and partially protected the remaining 2 rats. At 0.5 g acetaminophen/kg, triacetyloleandomycin decreased histologically observed liver damage in 7 of 15 rats. In rats pretreated with ethanol plus isopentanol, CYP3A, measured immunohistochemically, was decreased by acetaminophen treatment. This effect was prevented by triacetyloleandomycin. These results suggest that CYP3A has a major role in acetaminophen hepatotoxicity in animals administered the combined alcohol treatment. We also found that exposure to ethanol along with 0.1% isopentanol for only 3 days resulted in maximal increases in acetaminophen hepatotoxicity by the combined alcohol treatment, suggesting that short-term consumption of alcoholic beverages rich in isopentanol may be a risk for developing liver damage from acetaminophen.


Biochemical Pharmacology | 1995

Acute hepatotoxicity of acetaminophen in rats treated with ethanol plus isopentanol

Vsevolod E. Kostrubsky; Sheryl G. Wood; Matthew D. Bush; Juliana G. Szakacs; William J. Bement; Peter R. Sinclair; Elizabeth H. Jeffery; Jacqueline F. Sinclair

Acetaminophen (APAP) hepatotoxicity was investigated in rats fed ethanol and isopentanol alone or in combination in a liquid diet for 7 days. Serum levels of aspartate aminotransferase (AST) and histological examination of liver slices were used to assess hepatotoxicity. At 7 hr after intragastric administration of 0.5 or 1.0 g APAP/kg, there was no significant increase in serum levels of AST in rats treated with APAP alone, or in rats pretreated with ethanol or isopentanol alone followed by APAP. There was mild central lobular congestion in the livers of rats pretreated with ethanol alone followed by APAP. In contrast, in rats pretreated with the combination of ethanol and isopentanol, administration of APAP caused a dramatic increase in serum levels of AST, along with marked central lobular necrosis, including steatosis and ischemic changes. Hepatic glutathione levels were decreased to 40-50% of control values in APAP-treated rats that had been pretreated with ethanol either alone or in combination with isopentanol. The serum concentrations of APAP were significantly lower in rats pretreated with the combination of ethanol and isopentanol followed by 1 g APAP/kg than in rats treated with APAP alone, suggesting a greater rate of APAP metabolism. We had reported previously that combined treatment of rats with ethanol and isopentanol resulted in additive to synergistic increases in CYP3A, with no further increases in CYP2E than that caused by ethanol alone. CYP3A may, therefore, be responsible for the increased APAP hepatotoxicity caused by the combined alcohol treatment.


Drug Metabolism and Disposition | 1999

The use of human hepatocyte cultures to study the induction of cytochrome P-450.

Vsevolod E. Kostrubsky; Raman Venkataramanan; Kenneth Dorko; James E. Esplen; Shimin Zhang; Jacqueline F. Sinclair; Steven A. Wrighton; Stephen C. Strom


Hepatology | 1999

Morphogenetic events in mixed cultures of rat hepatocytes and nonparenchymal cells maintained in biological matrices in the presence of hepatocyte growth factor and epidermal growth factor

George K. Michalopoulos; William C. Bowen; Valerie F. Zajac; Donna Beer-Stolz; Simon C. Watkins; Vsevolod E. Kostrubsky; Steven C. Strom


Biochemical and Biophysical Research Communications | 2000

Induction of cytochrome P450 (CYP)1A1, CYP1A2, and CYP3A4 but not of CYP2C9, CYP2C19, multidrug resistance (MDR-1) and multidrug resistance associated protein (MRP-1) by prototypical inducers in human hepatocytes.

Dieter Runge; Christoph Köhler; Vsevolod E. Kostrubsky; Dana Jäger; Thomas Lehmann; Dorothee M. Runge; Ursula May; Donna B. Stolz; Stephen C. Strom; Wolfgang E. Fleig; George K. Michalopoulos


Archives of Biochemistry and Biophysics | 1995

Ethanol and Isopentanol Increase CYP3A and CYP2E in Primary Cultures of Human Hepatocytes

Vsevolod E. Kostrubsky; Stephen C. Strom; Sheryl G. Wood; Steven A. Wrighton; Peter R. Sinclair; Jacqueline F. Sinclair

Collaboration


Dive into the Vsevolod E. Kostrubsky's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sheryl G. Wood

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William J. Bement

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dieter Runge

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge