Your browser doesn't support javascript.
loading
Hepatocellular cystathionine γ lyase/hydrogen sulfide attenuates nonalcoholic fatty liver disease by activating farnesoid X receptor.
Xu, Wenjing; Cui, Changting; Cui, Chunmei; Chen, Zhenzhen; Zhang, Haizeng; Cui, Qinghua; Xu, Guoheng; Fan, Jianglin; Han, Yu; Tang, Liangjie; Targher, Giovanni; Byrne, Christopher D; Zheng, Ming-Hua; Yang, Liming; Cai, Jun; Geng, Bin.
Afiliación
  • Xu W; Department of Pathology, School of Basic Medical ScienceXi'an Medical UniversityShanxiChina.
  • Cui C; Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular DiseasesFuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina.
  • Cui C; Department of Bioinformatics, Physiology and Pathophysiology, School of Basic Medical SciencesPeking UniversityBeijingChina.
  • Chen Z; Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular DiseasesFuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina.
  • Zhang H; Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular DiseasesFuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina.
  • Cui Q; Department of Bioinformatics, Physiology and Pathophysiology, School of Basic Medical SciencesPeking UniversityBeijingChina.
  • Xu G; Department of Bioinformatics, Physiology and Pathophysiology, School of Basic Medical SciencesPeking UniversityBeijingChina.
  • Fan J; Department of Pathology, School of Basic Medical ScienceXi'an Medical UniversityShanxiChina.
  • Han Y; Department of Gastrointestinal Surgerythe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina.
  • Tang L; NAFLD Research Center, Department of Hepatologythe First Affiliated Hospital of Wenzhou Medical University; the Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang ProvinceWenzhouChina.
  • Targher G; Section of Endocrinology, Diabetes and Metabolism, Department of MedicineUniversity and Azienda Ospedaliera Universitaria Integrata of VeronaVeronaItaly.
  • Byrne CD; Southampton National Institute for Health and Care Research Biomedical Research CentreUniversity Hospital Southampton, Southampton General HospitalSouthamptonUK.
  • Zheng MH; NAFLD Research Center, Department of Hepatologythe First Affiliated Hospital of Wenzhou Medical University; the Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang ProvinceWenzhouChina.
  • Yang L; Department of PathophysiologyHarbin Medical University-DaqingDaqingChina.
  • Cai J; Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular DiseasesFuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina.
  • Geng B; Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular DiseasesFuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina.
Hepatology ; 76(6): 1794-1810, 2022 12.
Article en En | MEDLINE | ID: mdl-35586979
ABSTRACT
BACKGROUND AND

AIMS:

Hydrogen sulfide (H2 S) plays a protective role in NAFLD. However, whether cystathionine γ lyase (CSE), a dominant H2 S generating enzyme in hepatocytes, has a role in the pathogenesis of NAFLD is currently unclear. APPROACH AND

RESULTS:

We showed that CSE protein expression is dramatically downregulated, especially in fibrotic areas, in livers from patients with NAFLD. In high-fat diet (HFD)-induced NAFLD mice or an oleic acid-induced hepatocyte model, the CSE/H2 S pathway is also downregulated. To illustrate a regulatory role for CSE in NAFLD, we generated a hepatocyte-specific CSE knockout mouse (CSELKO ). Feeding an HFD to CSELKO mice, they showed more hepatic lipid deposition with increased activity of the fatty acid de novo synthesis pathway, increased hepatic insulin resistance, and higher hepatic gluconeogenic ability compared to CSELoxp control mice. By contrast, H2 S donor treatment attenuated these phenotypes. Furthermore, the protection conferred by H2 S was blocked by farnesoid X receptor (FXR) knockdown. Consistently, serum deoxycholic acid and lithocholic acid (FXR antagonists) were increased, and tauro-ß-muricholic acid (FXR activation elevated) was reduced in CSELKO . CSE/H2 S promoted a post-translation modification (sulfhydration) of FXR at Cys138/141 sites, thereby enhancing its activity to modulate expression of target genes related to lipid and glucose metabolism, inflammation, and fibrosis. Sulfhydration proteomics in patients' livers supported the CSE/H2 S modulation noted in the CSELKO mice.

CONCLUSIONS:

FXR sulfhydration is a post-translational modification affected by hepatic endogenous CSE/H2 S that may promote FXR activity and attenuate NAFLD. Hepatic CSE deficiency promotes development of nonalcoholic steatohepatitis. The interaction between H2 S and FXR may be amenable to therapeutic drug treatment in NAFLD.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Asunto principal: Carcinoma Hepatocelular / Enfermedad del Hígado Graso no Alcohólico / Sulfuro de Hidrógeno / Neoplasias Hepáticas Límite: Animals Idioma: En Revista: Hepatology Año: 2022 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Asunto principal: Carcinoma Hepatocelular / Enfermedad del Hígado Graso no Alcohólico / Sulfuro de Hidrógeno / Neoplasias Hepáticas Límite: Animals Idioma: En Revista: Hepatology Año: 2022 Tipo del documento: Article