Adipocyte-specific deletion of Tcf7l2 induces dysregulated lipid metabolism and impairs glucose tolerance in mice

Marie-Sophie Nguyen-Tu, Aida Martinez-Sanchez, Isabelle Leclerc, Guy A Rutter, Gabriela da Silva Xavier

Research output: Contribution to journalArticlepeer-review

203 Downloads (Pure)

Abstract

AIMS/HYPOTHESIS: Transcription factor 7-like 2 (TCF7L2) is a downstream effector of the Wnt/β-catenin signalling pathway implicated in type 2 diabetes risk through genome-wide association studies. Although its expression is critical for adipocyte development, the potential roles of changes in adipose tissue TCF7L2 levels in diabetes risk are poorly defined. Here, we investigated whether forced changes in Tcf7l2 expression in adipocytes affect whole body glucose or lipid metabolism and crosstalk between disease-relevant tissues.

METHODS: Tcf7l2 was selectively ablated in mature adipocytes in C57BL/6J mice using Cre recombinase under Adipoq promoter control to recombine Tcf7l2 alleles floxed at exon 1 (referred to as aTCF7L2 mice). aTCF7L2 mice were fed normal chow or a high-fat diet for 12 weeks. Glucose and insulin sensitivity, as well as beta cell function, were assessed in vivo and in vitro. Levels of circulating NEFA, selected hormones and adipokines were measured using standard assays.

RESULTS: Reduced TCF7L2 expression in adipocytes altered glucose tolerance and insulin secretion in male but not in female mice. Thus, on a normal chow diet, male heterozygote knockout mice (aTCF7L2het) exhibited impaired glucose tolerance at 16 weeks (p = 0.03) and increased fat mass (1.4 ± 0.1-fold, p = 0.007) but no changes in insulin secretion. In contrast, male homozygote knockout (aTCF7L2hom) mice displayed normal body weight but impaired oral glucose tolerance at 16 weeks (p = 0.0001). These changes were mechanistically associated with impaired in vitro glucose-stimulated insulin secretion (decreased 0.5 ± 0.1-fold vs control mice, p = 0.02) and decreased levels of the incretins glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide (0.6 ± 0.1-fold and 0.4 ± 0.1-fold vs control mice, p = 0.04 and p < 0.0001, respectively). Circulating levels of plasma NEFA and fatty acid binding protein 4 were increased by 1.3 ± 0.1-fold and 1.8 ± 0.3-fold vs control mice (p = 0.03 and p = 0.05, respectively). Following exposure to a high-fat diet for 12 weeks, male aTCF7L2hom mice exhibited reduced in vivo glucose-stimulated insulin secretion (0.5 ± 0.1-fold vs control mice, p = 0.02).

CONCLUSIONS/INTERPRETATION: Loss of Tcf7l2 gene expression selectively in adipocytes leads to a sexually dimorphic phenotype, with impairments not only in adipocytes, but also in pancreatic islet and enteroendocrine cells in male mice only. Our findings suggest novel roles for adipokines and incretins in the effects of diabetes-associated variants in TCF7L2, and further illuminate the roles of TCF7L2 in glucose homeostasis and diabetes risk. Graphical abstract.

Original languageEnglish
Pages (from-to)129-141
Number of pages13
JournalDiabetologia
Volume64
Issue number1
Early online date17 Oct 2020
DOIs
Publication statusPublished - Jan 2021

Bibliographical note

Funding Information:
This project was supported by an Early Career Research grant from the Society for Endocrinology to M-SN-T. GdSX was supported by Diabetes UK (BDA13/0004672), European Foundation for the Study of Diabetes (EFSD/Boehringer-Ingelheim and EFSD/Lilly), University of Birmingham (research start-up grant and Research Development Fund-Publication Award) and the Rosetrees Trust. GAR was supported by Wellcome Trust Senior Investigator (WT098424AIA) and Investigator (212625/Z/18/Z) Awards, MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1) an MRC Experimental Challenge Grant (DIVA, MR/L02036X/1), MRC (MR/N00275X/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) and Imperial Confidence in Concept (ICiC) grants, and a Royal Society Wolfson Research Merit Award. This project has received funding from the European Association for the Study of Diabetes, and University of Birmingham starter grant and Publication Data Award to GdSX, and European Union’s Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement No 115881 (RHAPSODY), which receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA, to GAR. The study sponsor/funders were not involved in the design of the study; the collection, analysis, and interpretation of data; writing the report; and did not impose any restrictions regarding the publication of the report. Acknowledgements Authors’ relationships and activities

Funding Information:
GAR has received grant funding from Sun Pharma and Les Laboratoires Servier. The remaining authors declare that there are no relationships or activities that might bias, or be perceived to bias, their work.

Publisher Copyright:
© 2020, The Author(s).

Keywords

  • Adipocytes/metabolism
  • Animals
  • Body Composition/genetics
  • Fatty Acid-Binding Proteins/blood
  • Fatty Acids, Nonesterified/blood
  • Female
  • Gene Expression
  • Glucose/pharmacology
  • Glucose Intolerance/genetics
  • Incretins/blood
  • Insulin Secretion/drug effects
  • Insulin-Secreting Cells/drug effects
  • Integrases/genetics
  • Lipid Metabolism/genetics
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Transcription Factor 7-Like 2 Protein/genetics
  • Type 2 diabetes
  • Adipocyte
  • Incretin
  • Fatty acid
  • TCF7L2
  • Insulin
  • Mouse
  • Beta cell

ASJC Scopus subject areas

  • Internal Medicine
  • Endocrinology, Diabetes and Metabolism

Fingerprint

Dive into the research topics of 'Adipocyte-specific deletion of Tcf7l2 induces dysregulated lipid metabolism and impairs glucose tolerance in mice'. Together they form a unique fingerprint.

Cite this