Your browser doesn't support javascript.
loading
Ketohexokinase inhibition improves NASH by reducing fructose-induced steatosis and fibrogenesis.
Shepherd, Emma L; Saborano, Raquel; Northall, Ellie; Matsuda, Kae; Ogino, Hitomi; Yashiro, Hiroaki; Pickens, Jason; Feaver, Ryan E; Cole, Banumathi K; Hoang, Stephen A; Lawson, Mark J; Olson, Matthew; Figler, Robert A; Reardon, John E; Nishigaki, Nobuhiro; Wamhoff, Brian R; Günther, Ulrich L; Hirschfield, Gideon; Erion, Derek M; Lalor, Patricia F.
Affiliation
  • Shepherd EL; Centre for Liver and Gastroenterology Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
  • Saborano R; Centre for Liver and Gastroenterology Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
  • Northall E; Centre for Liver and Gastroenterology Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
  • Matsuda K; Takeda Pharmaceuticals Cardiovascular and Metabolic Drug Discovery Unit, Kanagawa, Japan.
  • Ogino H; Takeda Pharmaceuticals Cardiovascular and Metabolic Drug Discovery Unit, Kanagawa, Japan.
  • Yashiro H; Takeda Pharmaceuticals Gastroenterology Drug Discovery Unit, Cambridge, MA, USA.
  • Pickens J; Takeda Pharmaceuticals Gastroenterology Drug Discovery Unit, Cambridge, MA, USA.
  • Feaver RE; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Cole BK; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Hoang SA; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Lawson MJ; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Olson M; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Figler RA; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Reardon JE; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Nishigaki N; Takeda Pharmaceuticals Cardiovascular and Metabolic Drug Discovery Unit, Kanagawa, Japan.
  • Wamhoff BR; HemoShear Therapeutics, Charlottesville, VA, USA.
  • Günther UL; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
  • Hirschfield G; Centre for Liver and Gastroenterology Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
  • Erion DM; Toronto Centre for Liver Disease, University of Toronto, Toronto General Hospital, Toronto, Canada.
  • Lalor PF; Takeda Pharmaceuticals Gastroenterology Drug Discovery Unit, Cambridge, MA, USA.
JHEP Rep ; 3(2): 100217, 2021 Apr.
Article in En | MEDLINE | ID: mdl-33490936
ABSTRACT
BACKGROUND &

AIMS:

Increasing evidence highlights dietary fructose as a major driver of non-alcoholic fatty liver disease (NAFLD) pathogenesis, the majority of which is cleared on first pass through the hepatic circulation by enzymatic phosphorylation to fructose-1-phosphate via the ketohexokinase (KHK) enzyme. Without a current approved therapy, disease management emphasises lifestyle interventions, but few patients adhere to such strategies. New targeted therapies are urgently required.

METHODS:

We have used a unique combination of human liver specimens, a murine dietary model of NAFLD and human multicellular co-culture systems to understand the hepatocellular consequences of fructose administration. We have also performed a detailed nuclear magnetic resonance-based metabolic tracing of the fate of isotopically labelled fructose upon administration to the human liver.

RESULTS:

Expression of KHK isoforms is found in multiple human hepatic cell types, although hepatocyte expression predominates. KHK knockout mice show a reduction in serum transaminase, reduced steatosis and altered fibrogenic response on an Amylin diet. Human co-cultures exposed to fructose exhibit steatosis and activation of lipogenic and fibrogenic gene expression, which were reduced by pharmacological inhibition of KHK activity. Analysis of human livers exposed to 13C-labelled fructose confirmed that steatosis, and associated effects, resulted from the accumulation of lipogenic precursors (such as glycerol) and enhanced glycolytic activity. All of these were dose-dependently reduced by administration of a KHK inhibitor.

CONCLUSIONS:

We have provided preclinical evidence using human livers to support the use of KHK inhibition to improve steatosis, fibrosis, and inflammation in the context of NAFLD. LAY

SUMMARY:

We have used a mouse model, human cells, and liver tissue to test how exposure to fructose can cause the liver to store excess fat and become damaged and scarred. We have then inhibited a key enzyme within the liver that is responsible for fructose metabolism. Our findings show that inhibition of fructose metabolism reduces liver injury and fibrosis in mouse and human livers and thus this may represent a potential route for treating patients with fatty liver disease in the future.
Key words

Full text: 1 Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: JHEP Rep Year: 2021 Type: Article Affiliation country: United kingdom

Full text: 1 Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: JHEP Rep Year: 2021 Type: Article Affiliation country: United kingdom