Abstract

BackgroundIncreased lower body fat is associated with reduced cardiometabolic risk. The molecular basis for depot-specific differences in gluteofemoral (GF) compared with abdominal (A) subcutaneous adipocyte function is poorly understood. In the current report, we used a combination of Assay for Transposase-Accessible Chromatin followed by sequencing (ATAC-seq), RNA-seq, and chromatin immunoprecipitation (ChIP)-qPCR analyses that provide evidence that depot-specific gene expression patterns are associated with differential epigenetic chromatin signatures.MethodsPreadipocytes cultured from A and GF adipose tissue obtained from premenopausal apple-shaped women were used to perform transcriptome analysis by RNA-seq and assess accessible chromatin regions by ATAC-seq. We measured mRNA expression and performed ChIP-qPCR experiments for histone modifications of active (H3K4me3) and repressed chromatin (H3K27me3) regions respectively on the promoter regions of differentially expressed genes.ResultsRNA-seq experiments revealed an A-fat and GF-fat selective gene expression signature, with 126 genes upregulated in abdominal preadipocytes and 90 genes upregulated in GF cells. ATAC-seq identified almost 10-times more A-specific chromatin-accessible regions. Using a combined analysis of ATAC-seq and global gene expression data, we identified 74 of the 126 abdominal-specific genes (59%) with A-specific accessible chromatin sites within 200 kb of the transcription start site (TSS), including HOXA3, HOXA5, IL8, IL1b, and IL6. Interestingly, only 14 of the 90 GF-specific genes (15%) had GF-specific accessible chromatin sites within 200 kb of the corresponding TSS, including HOXC13 and HOTAIR, whereas 25 of them (28%) had abdominal-specific accessible chromatin sites. ChIP-qPCR experiments confirmed that the active H3K4me3 chromatin mark was significantly enriched at the promoter regions of HOXA5 and HOXA3 genes in abdominal preadipocytes, while H3K27me3 was less abundant relative to chromatin from GF. This is consistent with their A-fat specific gene expression pattern. Conversely, analysis of the promoter regions of the GF specific HOTAIR and HOXC13 genes exhibited high H3K4me3 and low H3K27me3 levels in GF chromatin compared to A chromatin.ConclusionsGlobal transcriptome and open chromatin analyses of depot-specific preadipocytes identified their gene expression signature and differential open chromatin profile. Interestingly, A-fat-specific open chromatin regions can be observed in the proximity of GF-fat genes, but not vice versa.Trial registrationClinicaltrials.gov, NCT01745471. Registered 5 December 2012.

Highlights

  • As body weight increases, the distribution of body fat becomes an important factor that determines metabolic health [1]

  • Abdominal fat (A-fat)-specific open chromatin regions can be observed in the proximity of Gluteofemoral fat (GF-fat) genes, but not vice versa

  • We investigated depot-specific chromatin structure and gene expression using preadipocytes isolated from human subcutaneous adipose tissue

Read more

Summary

Introduction

The distribution of body fat becomes an important factor that determines metabolic health [1]. Excess intra-abdominal fat storage is wellknown to be associated with increased cardiovascular and metabolic risks [2, 3]. As compared to other fat depots, visceral adipose tissue has increased metabolic activity, both lipid uptake and lipolysis. A very recent study where the authors collected the data from the National Health and Nutrition Examination Survey for the years 1999–2006 established in the same manner a strong association between upper body fat and metabolic risk factors independent of visceral fat [8]. A common hypothesis is that lower body fat serves as a “sink” for excess energy sequestering lipid away from visceral adipose tissue and preventing fat accumulation in other unhealthy areas such as the liver, pancreas, and muscle [11]. We used a combination of Assay for Transposase-Accessible Chromatin followed by sequencing (ATAC-seq), RNA-seq, and chromatin immunoprecipitation (ChIP)-qPCR analyses that provide evidence that depot-specific gene expression patterns are associated with differential epigenetic chromatin signatures

Methods
Results
Conclusion
Full Text
Published version (Free)

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