Your browser doesn't support javascript.
loading
Hepatocyte mARC1 promotes fatty liver disease.
Lewis, Lara C; Chen, Lingyan; Hameed, L Shahul; Kitchen, Robert R; Maroteau, Cyrielle; Nagarajan, Shilpa R; Norlin, Jenny; Daly, Charlotte E; Szczerbinska, Iwona; Hjuler, Sara Toftegaard; Patel, Rahul; Livingstone, Eilidh J; Durrant, Tom N; Wondimu, Elisabeth; BasuRay, Soumik; Chandran, Anandhakumar; Lee, Wan-Hung; Hu, Sile; Gilboa, Barak; Grandi, Megan E; Toledo, Enrique M; Erikat, Abdullah H A; Hodson, Leanne; Haynes, William G; Pursell, Natalie W; Coppieters, Ken; Fleckner, Jan; Howson, Joanna M M; Andersen, Birgitte; Ruby, Maxwell A.
Affiliation
  • Lewis LC; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Chen L; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Hameed LS; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Kitchen RR; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Maroteau C; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Nagarajan SR; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK.
  • Norlin J; Novo Nordisk A/S, Måløv, Denmark.
  • Daly CE; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Szczerbinska I; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Hjuler ST; Novo Nordisk A/S, Måløv, Denmark.
  • Patel R; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Livingstone EJ; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Durrant TN; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Wondimu E; Dicerna Pharmaceuticals Inc., Lexington, MA, USA.
  • BasuRay S; Dicerna Pharmaceuticals Inc., Lexington, MA, USA.
  • Chandran A; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Lee WH; Dicerna Pharmaceuticals Inc., Lexington, MA, USA.
  • Hu S; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Gilboa B; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Grandi ME; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Toledo EM; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Erikat AHA; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Hodson L; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK.
  • Haynes WG; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, UK.
  • Pursell NW; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Coppieters K; Dicerna Pharmaceuticals Inc., Lexington, MA, USA.
  • Fleckner J; Novo Nordisk A/S, Måløv, Denmark.
  • Howson JMM; Novo Nordisk A/S, Måløv, Denmark.
  • Andersen B; Novo Nordisk Research Centre Oxford, Oxford, UK.
  • Ruby MA; Novo Nordisk A/S, Måløv, Denmark.
JHEP Rep ; 5(5): 100693, 2023 May.
Article in En | MEDLINE | ID: mdl-37122688
ABSTRACT
Background &

Aims:

Non-alcoholic fatty liver disease (NAFLD) has a prevalence of ∼25% worldwide, with significant public health consequences yet few effective treatments. Human genetics can help elucidate novel biology and identify targets for new therapeutics. Genetic variants in mitochondrial amidoxime-reducing component 1 (MTARC1) have been associated with NAFLD and liver-related mortality; however, its pathophysiological role and the cell type(s) mediating these effects remain unclear. We aimed to investigate how MTARC1 exerts its effects on NAFLD by integrating human genetics with in vitro and in vivo studies of mARC1 knockdown.

Methods:

Analyses including multi-trait colocalisation and Mendelian randomisation were used to assess the genetic associations of MTARC1. In addition, we established an in vitro long-term primary human hepatocyte model with metabolic readouts and used the Gubra Amylin NASH (GAN)-diet non-alcoholic steatohepatitis mouse model treated with hepatocyte-specific N-acetylgalactosamine (GalNAc)-siRNA to understand the in vivo impacts of MTARC1.

Results:

We showed that genetic variants within the MTARC1 locus are associated with liver enzymes, liver fat, plasma lipids, and body composition, and these associations are attributable to the same causal variant (p.A165T, rs2642438 G>A), suggesting a shared mechanism. We demonstrated that increased MTARC1 mRNA had an adverse effect on these traits using Mendelian randomisation, implying therapeutic inhibition of mARC1 could be beneficial. In vitro mARC1 knockdown decreased lipid accumulation and increased triglyceride secretion, and in vivo GalNAc-siRNA-mediated knockdown of mARC1 lowered hepatic but increased plasma triglycerides. We found alterations in pathways regulating lipid metabolism and decreased secretion of 3-hydroxybutyrate upon mARC1 knockdown in vitro and in vivo.

Conclusions:

Collectively, our findings from human genetics, and in vitro and in vivo hepatocyte-specific mARC1 knockdown support the potential efficacy of hepatocyte-specific targeting of mARC1 for treatment of NAFLD. Impact and implications We report that genetically predicted increases in MTARC1 mRNA associate with poor liver health. Furthermore, knockdown of mARC1 reduces hepatic steatosis in primary human hepatocytes and a murine NASH model. Together, these findings further underscore the therapeutic potential of targeting hepatocyte MTARC1 for NAFLD.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Clinical_trials / Prognostic_studies / Risk_factors_studies Language: En Journal: JHEP Rep Year: 2023 Document type: Article Affiliation country: United kingdom

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Clinical_trials / Prognostic_studies / Risk_factors_studies Language: En Journal: JHEP Rep Year: 2023 Document type: Article Affiliation country: United kingdom