Your browser doesn't support javascript.
loading
Metabolic regulation of hepatic PNPLA3 expression and severity of liver fibrosis in patients with NASH.
Bruschi, Francesca V; Tardelli, Matteo; Herac, Merima; Claudel, Thierry; Trauner, Michael.
Afiliación
  • Bruschi FV; Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Internal Medicine III, Medical University of Vienna, Vienna, Austria.
  • Tardelli M; Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Internal Medicine III, Medical University of Vienna, Vienna, Austria.
  • Herac M; Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Cornell Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
  • Claudel T; Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
  • Trauner M; Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Internal Medicine III, Medical University of Vienna, Vienna, Austria.
Liver Int ; 40(5): 1098-1110, 2020 05.
Article en En | MEDLINE | ID: mdl-32043752
ABSTRACT
BACKGROUND AND

AIMS:

The genetic PNPLA3 polymorphism I148M has been extensively associated with higher risk for development and progression of NAFLD towards NASH.

METHODS:

PNPLA3 and α-SMA expression were quantified in liver biopsies collected from NASH patients (n = 26) with different fibrosis stages and PNPLA3 genotypes. To study the potential mechanisms driving PNPLA3 expression during NASH progression towards fibrosis, hepatocytes and hepatic stellate cells (HSCs) were cultivated in low and high glucose medium. Moreover, hepatocytes were treated with increasing concentrations of palmitic acid alone or in combination with glucose. Conditioned media were collected from challenged hepatocytes to stimulate HSCs.

RESULTS:

Tissue expression of PNPLA3 was significantly enhanced in biopsies of patients carrying the I148M polymorphism compared to wild type (WT). In NASH biopsies, PNPLA3 significantly correlated with fibrosis stage and α-SMA levels independently of PNPLA3 genotype. In line, PNPLA3 expression was higher in α-SMA positive cells. Low glucose increased PNPLA3 in HSCs, whereas high glucose induced PNPLA3 and de-novo lipogenesis-related genes expression in hepatocytes. Palmitic acid induced fat accumulation and cell stress markers in hepatocytes, which could be counteracted by oleic acid. Conditioned media collected from lipotoxic challenged hepatocytes markedly induced PNPLA3 mRNA and protein levels, fibrogenic and autophagic markers and promoted migration in HSCs. Notably, conditioned media collected from hepatocytes cultivated with both glucose and palmitic acid exacerbated HSCs migration, PNPLA3 and fibrogenic gene expression, promoting release of cytokines from HSCs.

CONCLUSIONS:

Collectively, our observations uncover the diverse metabolic regulation of PNPLA3 among different hepatic cell populations and support its relation to fibrosis progression.
Asunto(s)
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Enfermedad del Hígado Graso no Alcohólico / Lipasa / Proteínas de la Membrana Límite: Humans Idioma: En Revista: Liver Int Asunto de la revista: GASTROENTEROLOGIA Año: 2020 Tipo del documento: Article País de afiliación: Austria

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Enfermedad del Hígado Graso no Alcohólico / Lipasa / Proteínas de la Membrana Límite: Humans Idioma: En Revista: Liver Int Asunto de la revista: GASTROENTEROLOGIA Año: 2020 Tipo del documento: Article País de afiliación: Austria