Your browser doesn't support javascript.
loading
Hepatic protein-tyrosine phosphatase 1B disruption and pharmacological inhibition attenuate ethanol-induced oxidative stress and ameliorate alcoholic liver disease in mice.
Hsu, Ming-Fo; Koike, Shinichiro; Mello, Aline; Nagy, Laura E; Haj, Fawaz G.
Affiliation
  • Hsu MF; Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA. Electronic address: mfhsu@ucdavis.edu.
  • Koike S; Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA.
  • Mello A; Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA.
  • Nagy LE; Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA.
  • Haj FG; Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, University of California Dav
Redox Biol ; 36: 101658, 2020 09.
Article in En | MEDLINE | ID: mdl-32769011
ABSTRACT
Alcoholic liver disease (ALD) is a major health problem and a significant cause of liver-related death. Currently, the mainstay for ALD therapy is alcohol abstinence highlighting the need to develop pharmacotherapeutic approaches. Protein-tyrosine phosphatase 1B (PTP1B) is an established regulator of hepatic functions, but its role in ALD is mostly unexplored. In this study, we used mice with liver-specific PTP1B disruption as well as pharmacological inhibition to investigate the in vivo function of this phosphatase in ALD. We report upregulation of hepatic PTP1B in the chronic plus binge mouse model and, importantly, in liver biopsies of alcoholic hepatitis patients. Also, mice with hepatic PTP1B disruption attenuated ethanol-induced injury, inflammation, and steatosis compared with ethanol-fed control animals. Moreover, PTP1B deficiency was associated with decreased ethanol-induced oxidative stress in vivo and ex vivo. Further, pharmacological modulation of oxidative balance in hepatocytes identified diminished oxidative stress as a contributor to the salutary effects of PTP1B deficiency. Notably, PTP1B pharmacological inhibition elicited beneficial effects and mitigated hepatic injury, inflammation, and steatosis caused by ethanol feeding. In summary, these findings causally link hepatic PTP1B and ALD and define a potential therapeutic target for the management of this disease.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Ethanol / Liver Diseases, Alcoholic Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Redox Biol Year: 2020 Type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Ethanol / Liver Diseases, Alcoholic Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Redox Biol Year: 2020 Type: Article