High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1

碩士 === 國立陽明大學 === 生理學研究所 === 97 === Obesity is characterized by excess fat accumulation in white adipose tissue, which increases the risk of various diseases. Adipocytes release various adipokines such as adiponectin to regulate lipid homeostasis. High mobility group box 1 (HMGB1) is not only a key ...

Full description

Bibliographic Details
Main Authors: Kuan-Ting Chiu, 邱冠霆
Other Authors: Tzong-Shyuan Lee
Format: Others
Language:en_US
Published: 2009
Online Access:http://ndltd.ncl.edu.tw/handle/w72cax
id ndltd-TW-097YM005116001
record_format oai_dc
spelling ndltd-TW-097YM0051160012019-05-15T20:07:01Z http://ndltd.ncl.edu.tw/handle/w72cax High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1 高遷移率族蛋白負向調節脂肪細胞之脂質水解作用:第一型小窩蛋白的角色 Kuan-Ting Chiu 邱冠霆 碩士 國立陽明大學 生理學研究所 97 Obesity is characterized by excess fat accumulation in white adipose tissue, which increases the risk of various diseases. Adipocytes release various adipokines such as adiponectin to regulate lipid homeostasis. High mobility group box 1 (HMGB1) is not only a key protein for stabilization of chromosome and regulation of transcription in nucleus, but also released into the extracellular space via exocytosis to modulate inflammatory response. However, the exact role and underlying mechanisms of HMGB1 in adipogenesis are largely unknown. Caveolin-1 (Cav-1) is major structure protein of caveolae, which plays a crucial role in lipid metabolism and mediates secretion, internalization and vesicle trafficking of proteins by interacting with intracellular proteins. In the present study, we performed in vitro and in vivo experiments to explore whether Cav-1 interacts with HMGB1 to regulate adipogenesis. Western blotting showed that the levels of HMGB1, Cav-1 and phosphorylated Cav-1 were significantly increased during differentiation of 3T3-L1 adipocytes in a time-dependent manner. HMGB1 treatment did not affect preadipocyte proliferation and differentiation. By contrast, starvation- or 刍3-adrenoreceptor agonist CL-induced lipolysis and phosphorylation of hormone sensitive lipase were significantly attenuated upon HMGB1 treatment via inhibition on activation of protein kinase A and extracellular signal-regulated kinase phosphorylation in 3T3-L1 adipocytes. We also found that the increases in release of HMGB1, Cav-1 phosphorylation and interaction between HMGB1 and Cav-1 were Src kinase-dependent, as revealed by its abolition after inhibition using pharmacologic inhibitors. After starvation or fasting, secretion of HMGB1 was increased in Cav-1 deficient mouse embryonic fibroblast and mice compared to wildtypes. Collectively, the findings suggest that HMGB1 plays a critical role in adipocyte lipolysis via interaction with Cav-1. Tzong-Shyuan Lee 李宗玄 2009 學位論文 ; thesis 109 en_US
collection NDLTD
language en_US
format Others
sources NDLTD
description 碩士 === 國立陽明大學 === 生理學研究所 === 97 === Obesity is characterized by excess fat accumulation in white adipose tissue, which increases the risk of various diseases. Adipocytes release various adipokines such as adiponectin to regulate lipid homeostasis. High mobility group box 1 (HMGB1) is not only a key protein for stabilization of chromosome and regulation of transcription in nucleus, but also released into the extracellular space via exocytosis to modulate inflammatory response. However, the exact role and underlying mechanisms of HMGB1 in adipogenesis are largely unknown. Caveolin-1 (Cav-1) is major structure protein of caveolae, which plays a crucial role in lipid metabolism and mediates secretion, internalization and vesicle trafficking of proteins by interacting with intracellular proteins. In the present study, we performed in vitro and in vivo experiments to explore whether Cav-1 interacts with HMGB1 to regulate adipogenesis. Western blotting showed that the levels of HMGB1, Cav-1 and phosphorylated Cav-1 were significantly increased during differentiation of 3T3-L1 adipocytes in a time-dependent manner. HMGB1 treatment did not affect preadipocyte proliferation and differentiation. By contrast, starvation- or 刍3-adrenoreceptor agonist CL-induced lipolysis and phosphorylation of hormone sensitive lipase were significantly attenuated upon HMGB1 treatment via inhibition on activation of protein kinase A and extracellular signal-regulated kinase phosphorylation in 3T3-L1 adipocytes. We also found that the increases in release of HMGB1, Cav-1 phosphorylation and interaction between HMGB1 and Cav-1 were Src kinase-dependent, as revealed by its abolition after inhibition using pharmacologic inhibitors. After starvation or fasting, secretion of HMGB1 was increased in Cav-1 deficient mouse embryonic fibroblast and mice compared to wildtypes. Collectively, the findings suggest that HMGB1 plays a critical role in adipocyte lipolysis via interaction with Cav-1.
author2 Tzong-Shyuan Lee
author_facet Tzong-Shyuan Lee
Kuan-Ting Chiu
邱冠霆
author Kuan-Ting Chiu
邱冠霆
spellingShingle Kuan-Ting Chiu
邱冠霆
High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
author_sort Kuan-Ting Chiu
title High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
title_short High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
title_full High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
title_fullStr High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
title_full_unstemmed High Mobility Group Box 1 Negatively Regulates Lipolysis in Adipocytes : Role of Caveolin-1
title_sort high mobility group box 1 negatively regulates lipolysis in adipocytes : role of caveolin-1
publishDate 2009
url http://ndltd.ncl.edu.tw/handle/w72cax
work_keys_str_mv AT kuantingchiu highmobilitygroupbox1negativelyregulateslipolysisinadipocytesroleofcaveolin1
AT qiūguāntíng highmobilitygroupbox1negativelyregulateslipolysisinadipocytesroleofcaveolin1
AT kuantingchiu gāoqiānyílǜzúdànbáifùxiàngdiàojiézhīfángxìbāozhīzhīzhìshuǐjiězuòyòngdìyīxíngxiǎowōdànbáidejiǎosè
AT qiūguāntíng gāoqiānyílǜzúdànbáifùxiàngdiàojiézhīfángxìbāozhīzhīzhìshuǐjiězuòyòngdìyīxíngxiǎowōdànbáidejiǎosè
_version_ 1719097239252500480