Abstract

ObjectiveAdipogenesis is critical for adipose tissue remodeling during the development of obesity. While the role of transcription factors in the orchestration of adipogenic pathways is already established, the involvement of coregulators that transduce regulatory signals into epigenome alterations and transcriptional responses remains poorly understood. The aim of our study was to investigate which pathways are controlled by G protein pathway suppressor 2 (GPS2) during the differentiation of human adipocytes. MethodsWe generated a unique loss-of-function model by RNAi depletion of GPS2 in human multipotent adipose-derived stem (hMADS) cells. We thoroughly characterized the coregulator depletion-dependent pathway alterations during adipocyte differentiation at the level of transcriptome (RNA-seq), epigenome (ChIP-seq H3K27ac), cistrome (ChIP-seq GPS2), and lipidome. We validated the in vivo relevance of the identified pathways in non-diabetic and diabetic obese patients. ResultsThe loss of GPS2 triggers the reprogramming of cellular processes related to adipocyte differentiation by increasing the responses to the adipogenic cocktail. In particular, GPS2 depletion increases the expression of BMP4, an important trigger for the commitment of fibroblast-like progenitors toward the adipogenic lineage and increases the expression of inflammatory and metabolic genes. GPS2-depleted human adipocytes are characterized by hypertrophy, triglyceride and phospholipid accumulation, and sphingomyelin depletion. These changes are likely a consequence of the increased expression of ATP-binding cassette subfamily G member 1 (ABCG1) that mediates sphingomyelin efflux from adipocytes and modulates lipoprotein lipase (LPL) activity. We identify ABCG1 as a direct transcriptional target, as GPS2 depletion leads to coordinated changes of transcription and H3K27 acetylation at promoters and enhancers that are occupied by GPS2 in wild-type adipocytes. We find that in omental adipose tissue of obese humans, GPS2 levels correlate with ABCG1 levels, type 2 diabetic status, and lipid metabolic status, supporting the in vivo relevance of the hMADS cell-derived in vitro data. ConclusionOur study reveals a dual regulatory role of GPS2 in epigenetically modulating the chromatin landscape and gene expression during human adipocyte differentiation and identifies a hitherto unknown GPS2-ABCG1 pathway potentially linked to adipocyte hypertrophy in humans.

Highlights

  • The rising prevalence of obesity and its strong association with comorbidities such as insulin resistance and type 2 diabetes has increased interest in adipose tissue biology and its therapeutic potential

  • We demonstrate that depletion of G protein pathway suppressor 2 (GPS2) induces upregulation of multiple genes involved in adipogenesis and lipid metabolism that is associated with a deep remodeling of the adipocyte lipidome, such as the accumulation of triglycerides, phosphatidylcholine, and phosphatidylethanolamine and depletion of sphingolipids

  • We show that the loss of GPS2 in preadipocytes causes increased ATP-binding cassette subfamily G member 1 (ABCG1) and lipoprotein lipase (LPL) gene expression, likely via de-repression of their promoter and enhancers and increase in LPL activity, two crucial elements for triglyceride accumulation linked to hypertrophy

Read more

Summary

Introduction

The rising prevalence of obesity and its strong association with comorbidities such as insulin resistance and type 2 diabetes has increased interest in adipose tissue biology and its therapeutic potential. Obesity is recognized as a pathological condition characterized by body fat accumulation with expansion of adipose depots in response to high caloric intake. Adipose tissue plasticity plays a critical role in systemic metabolic homeostasis and the ability of adipocytes to effectively store lipids, thereby protecting other tissues such as muscle and liver from lipotoxicity [1]. Adipose tissue enlargement is achieved either via hyperplasia, that is, the increased number of adipocytes. MOLECULAR METABOLISM 42 (2020) 101066 Ó 2020 The Authors.

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.