RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.

In this study, we performed high throughput RNA sequencing at the preadipocyte (D0) and differentiated adipocyte (D7) stages of primary brown preadipocyte differentiation in order to characterize the transcriptional events regulating differentiation and function. Compared to the preadipocyte stage (...

Full description

Bibliographic Details
Main Authors: Min-Kyung Jang, Sunwoo Lee, Myeong Ho Jung
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2016-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC5017575?pdf=render
id doaj-6595d68dbcde44ea92fc193b28d0cb16
record_format Article
spelling doaj-6595d68dbcde44ea92fc193b28d0cb162020-11-25T01:41:58ZengPublic Library of Science (PLoS)PLoS ONE1932-62032016-01-01119e016223810.1371/journal.pone.0162238RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.Min-Kyung JangSunwoo LeeMyeong Ho JungIn this study, we performed high throughput RNA sequencing at the preadipocyte (D0) and differentiated adipocyte (D7) stages of primary brown preadipocyte differentiation in order to characterize the transcriptional events regulating differentiation and function. Compared to the preadipocyte stage (D0), 6,668 genes were identified as differentially expressed genes (DEGs) with a fold change of ≥ 2.0 at the differentiated adipocyte stage (D7). Several adipogenic genes including peroxisome proliferator-activated receptor-γ (PPARγ) and CCAAT/enhancer-binding protein-α (C/EBPα), and Krüppel-like factor (KLF) family genes were differentially expressed at D0 and D7. Since KLF16 gene expression was downregulated at day 7 and its adipogenic function has not been characterized, we investigated its role in adipogenesis. Knockdown of KLF16 stimulated the differentiation of both brown and 3T3-L1 preadipocytes, and led to increased PPARγ expression. However, overexpression of KLF16 had opposite effects. Furthermore, KLF16 downregulated PPARγ expression in brown adipocytes and inhibited its promoter activity. These results indicate that KLF16 inhibits adipogenesis through downregulation of PPARγ expression.http://europepmc.org/articles/PMC5017575?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Min-Kyung Jang
Sunwoo Lee
Myeong Ho Jung
spellingShingle Min-Kyung Jang
Sunwoo Lee
Myeong Ho Jung
RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
PLoS ONE
author_facet Min-Kyung Jang
Sunwoo Lee
Myeong Ho Jung
author_sort Min-Kyung Jang
title RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
title_short RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
title_full RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
title_fullStr RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
title_full_unstemmed RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis.
title_sort rna-seq analysis reveals a negative role of klf16 in adipogenesis.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2016-01-01
description In this study, we performed high throughput RNA sequencing at the preadipocyte (D0) and differentiated adipocyte (D7) stages of primary brown preadipocyte differentiation in order to characterize the transcriptional events regulating differentiation and function. Compared to the preadipocyte stage (D0), 6,668 genes were identified as differentially expressed genes (DEGs) with a fold change of ≥ 2.0 at the differentiated adipocyte stage (D7). Several adipogenic genes including peroxisome proliferator-activated receptor-γ (PPARγ) and CCAAT/enhancer-binding protein-α (C/EBPα), and Krüppel-like factor (KLF) family genes were differentially expressed at D0 and D7. Since KLF16 gene expression was downregulated at day 7 and its adipogenic function has not been characterized, we investigated its role in adipogenesis. Knockdown of KLF16 stimulated the differentiation of both brown and 3T3-L1 preadipocytes, and led to increased PPARγ expression. However, overexpression of KLF16 had opposite effects. Furthermore, KLF16 downregulated PPARγ expression in brown adipocytes and inhibited its promoter activity. These results indicate that KLF16 inhibits adipogenesis through downregulation of PPARγ expression.
url http://europepmc.org/articles/PMC5017575?pdf=render
work_keys_str_mv AT minkyungjang rnaseqanalysisrevealsanegativeroleofklf16inadipogenesis
AT sunwoolee rnaseqanalysisrevealsanegativeroleofklf16inadipogenesis
AT myeonghojung rnaseqanalysisrevealsanegativeroleofklf16inadipogenesis
_version_ 1725038580196179968