Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zhi-Gang She is active.

Publication


Featured researches published by Zhi-Gang She.


Hepatology | 2014

Interferon regulatory factor 3 constrains IKKβ/NF-κB signaling to alleviate hepatic steatosis and insulin resistance.

Xin-An Wang; Ran Zhang; Zhi-Gang She; Xiao-Fei Zhang; Ding-Sheng Jiang; Tao Wang; Lu Gao; Wei Deng; Shu-Min Zhang; Li-Hua Zhu; Sen Guo; Ke Chen; Xiao-Dong Zhang; De-Pei Liu; Hongliang Li

Obesity and related metabolic diseases associated with chronic low‐grade inflammation greatly compromise human health. Previous observations on the roles of interferon regulatory factors (IRFs) in the regulation of metabolism prompted investigation of the involvement of a key family member, IRF3, in metabolic disorders. IRF3 expression in the liver is decreased in animals with diet‐induced and genetic obesity. The global knockout (KO) of IRF3 significantly promotes chronic high‐fat diet (HFD)‐induced hepatic insulin resistance and steatosis; in contrast, adenoviral‐mediated hepatic IRF3 overexpression preserves glucose and lipid homeostasis. Furthermore, systemic and hepatic inflammation, which is increased in IRF3 KO mice, is attenuated by the overexpression of hepatic IRF3. Importantly, inhibitor of nuclear factor kappa B kinase beta subunit / nuclear factor kappa B (IKKβ/NF‐κB) signaling is repressed by IRF3, and hepatic overexpression of the inhibitor of κB‐α (IκBα) reverses HFD‐induced insulin resistance and steatosis in IRF3 KO mice. Mechanistically, IRF3 interacts with the kinase domain of IKKβ in the cytoplasm and inhibits its downstream signaling. Moreover, deletion of the region of IRF3 responsible for the IRF3/IKKβ interaction inhibits the capacity of IRF3 to preserve glucose and lipid homeostasis. Conclusion: IRF3 interacts with IKKβ in the cytoplasm to inhibit IKKβ/NF‐κB signaling, thus alleviating hepatic inflammation, insulin resistance, and hepatic steatosis. (Hepatology 2014;59:870–885)


Hepatology | 2013

Interferon regulatory factor 9 protects against hepatic insulin resistance and steatosis in male mice

Xin An Wang; Ran Zhang; Ding-Sheng Jiang; Wei Deng; Shu-Min Zhang; Shan Deng; Jinfeng Zhong; Tao Wang; Li Hua Zhu; Li Yang; Shufen Hong; Sen Guo; Ke Chen; Xiao Fei Zhang; Zhi-Gang She; Yingjie Chen; Qinglin Yang; Xiao-Dong Zhang; Hongliang Li

Obesity is a calorie‐excessive state associated with high risk of diabetes, atherosclerosis, and certain types of tumors. Obesity may induce inflammation and insulin resistance (IR). We found that the expression of interferon (IFN) regulatory factor 9 (IRF9), a major transcription factor mediating IFN responses, was lower in livers of obese mice than in those of their lean counterparts. Furthermore, whole‐body IRF9 knockout (KO) mice were more obese and had aggravated IR, hepatic steatosis, and inflammation after chronic high‐fat diet feeding. In contrast, adenoviral‐mediated hepatic IRF9 overexpression in both diet‐induced and genetically (ob/ob) obese mice showed markedly improved hepatic insulin sensitivity and attenuated hepatic steatosis and inflammation. We further employed a yeast two‐hybrid screening system to investigate the interactions between IRF9 and its cofactors. Importantly, we identified that IRF9 interacts with peroxisome proliferator‐activated receptor alpha (PPAR‐α), an important metabolism‐associated nuclear receptor, to activate PPAR‐α target genes. In addition, liver‐specific PPAR‐α overexpression rescued insulin sensitivity and ameliorated hepatic steatosis and inflammation in IRF9 KO mice. Conclusion: IRF9 attenuates hepatic IR, steatosis, and inflammation through interaction with PPAR‐α. (Hepatology 2013;58:603–616)


American Journal of Physiology-endocrinology and Metabolism | 2013

Interferon regulatory factor 7 deficiency prevents diet-induced obesity and insulin resistance

Xin-An Wang; Ran Zhang; Shu-Min Zhang; Shan Deng; Ding-Sheng Jiang; Jinfeng Zhong; Li Yang; Tao Wang; Shufen Hong; Sen Guo; Zhi-Gang She; Xiao-Dong Zhang; Hongliang Li

Obesity-related inflammation has been implicated in the pathogenesis of insulin resistance and type 2 diabetes. In this study, we addressed the potential role of interferon regulatory factor 7 (IRF7), a master regulator of type I interferon-dependent immune responses, in the regulation of energy metabolism. The expression levels of IRF7 were increased in white adipose tissue, liver tissue, and gastrocnemius muscle of both diet-induced obese mice and ob/ob mice compared with their lean counterparts. After feeding a high-fat diet (HFD) for 24 wk, IRF7 knockout (KO) mice showed less weight gain and adiposity than wild-type controls. KO of IRF7 improved glucose and lipid homeostasis and insulin sensitivity. Additionally, KO of IRF7 ameliorated diet-induced hepatic steatosis. Next, we assessed the inflammatory state of the IRF7 KO mice on the HFD. These mice showed less macrophage infiltration into multiple organs and were protected from local and systemic inflammation. This study demonstrates a role for IRF7 in diet-induced alterations in energy metabolism and insulin sensitivity. Our results also suggest that IRF7 is involved in the etiology of metabolic abnormalities, which suggests a new strategy for treating obesity and type 2 diabetes.


Nature Medicine | 2017

Targeting CASP8 and FADD-like apoptosis regulator ameliorates nonalcoholic steatohepatitis in mice and nonhuman primates

Pi-Xiao Wang; Yan-Xiao Ji; Xiao-Jing Zhang; Ling-Ping Zhao; Zhen-Zhen Yan; Peng Zhang; Li-Jun Shen; Xia Yang; Jing Fang; Song Tian; Xueyong Zhu; Jun Gong; Xin Zhang; Qiao-Fang Wei; Yong Wang; Jing Li; Lu Wan; Qingguo Xie; Zhi-Gang She; Zhihua Wang; Zan Huang; Hongliang Li

Nonalcoholic steatohepatitis (NASH) is a progressive disease that is often accompanied by metabolic syndrome and poses a high risk of severe liver damage. However, no effective pharmacological treatment is currently available for NASH. Here we report that CASP8 and FADD-like apoptosis regulator (CFLAR) is a key suppressor of steatohepatitis and its metabolic disorders. We provide mechanistic evidence that CFLAR directly targets the kinase MAP3K5 (also known as ASK1) and interrupts its N-terminus-mediated dimerization, thereby blocking signaling involving ASK1 and the kinase MAPK8 (also known as JNK1). Furthermore, we identified a small peptide segment in CFLAR that effectively attenuates the progression of steatohepatitis and metabolic disorders in both mice and monkeys by disrupting the N-terminus-mediated dimerization of ASK1 when the peptide is expressed from an injected adenovirus-associated virus 8–based vector. Taken together, these findings establish CFLAR as a key suppressor of steatohepatitis and indicate that the development of CFLAR-peptide-mimicking drugs and the screening of small-molecular inhibitors that specifically block ASK1 dimerization are new and feasible approaches for NASH treatment.


Basic Research in Cardiology | 2014

Interferon regulatory factor 9 is an essential mediator of heart dysfunction and cell death following myocardial ischemia/reperfusion injury

Yan Zhang; Xiaoxiong Liu; Zhi-Gang She; Ding-Sheng Jiang; Nian Wan; Hao Xia; Xue-Hai Zhu; Xiang Wei; Xiao-Dong Zhang; Hongliang Li

This study aimed to investigate whether interferon regulatory factor 9 (IRF9) is involved in the pathogenesis of myocardial ischemia–reperfusion (I/R) injury and to explore the underlying molecular mechanisms of this process. Cell death plays a major role in myocardial I/R injury. We recently determined the importance of IRF9 in coordinating molecular events in response to hypertrophic stress in cardiomyocytes. However, the roles of IRF9 in lethal myocardial injury remain to be elucidated. The involvement of IRF9 was assessed via functional assays in a mouse myocardial I/R injury model by genetic knockout and cardiomyocyte-specific transgenic overexpression of IRF9, and its effects on cardiomyocyte apoptosis and inflammation were further studied in vivo and in vitro. IRF9 was upregulated in human ischemic heart tissue and mouse hearts after I/R injury. Ablation of IRF9 protected the heart against I/R-induced cardiomyocyte death, development of inflammation, and loss of heart function. In contrast, cardiomyocyte-specific transgenic overexpression of IRF9 aggravated myocardial reperfusion injury and inflammation. IRF9 negatively regulated the Sirt1-p53 axis under I/R conditions in vivo and in vitro. Downregulation of Sirt1 expression and its downstream apoptosis-related signaling cascade, which results from I/R, was ameliorated by loss of IRF9 and exacerbated by overexpression of IRF9. Cardiomyocyte-specific deletion of Sirt1 abolished the protective effect of IRF9 knockout against I/R injury, which further indicated that IRF9 mediated myocardial reperfusion injury by modulating the Sirt1-p53 axis. Thus, IRF9 may be a novel therapeutic target for the prevention of I/R injury resulting from revascularization therapy after acute myocardial infarction (MI).


Nature Medicine | 2017

Tmbim1 is a multivesicular body regulator that protects against non-alcoholic fatty liver disease in mice and monkeys by targeting the lysosomal degradation of Tlr4

Guang-Nian Zhao; Peng Zhang; Jun Gong; Xiao-Jing Zhang; Pi-Xiao Wang; Miao Yin; Zhou Jiang; Li-Jun Shen; Yan-Xiao Ji; Jingjing Tong; Yutao Wang; Qiao-Fang Wei; Yong Wang; Xueyong Zhu; Xin Zhang; Jing Fang; Qingguo Xie; Zhi-Gang She; Zhihua Wang; Zan Huang; Hongliang Li

Non-alcoholic steatohepatitis (NASH) is an increasingly prevalent liver pathology that can progress from non-alcoholic fatty liver disease (NAFLD), and it is a leading cause of cirrhosis and hepatocellular carcinoma. There is currently no pharmacological therapy for NASH. Defective lysosome-mediated protein degradation is a key process that underlies steatohepatitis and a well-recognized drug target in a variety of diseases; however, whether it can serve as a therapeutic target for NAFLD and NASH remains unknown. Here we report that transmembrane BAX inhibitor motif-containing 1 (TMBIM1) is an effective suppressor of steatohepatitis and a previously unknown regulator of the multivesicular body (MVB)-lysosomal pathway. Tmbim1 expression in hepatocytes substantially inhibited high-fat diet–induced insulin resistance, hepatic steatosis and inflammation in mice. Mechanistically, Tmbim1 promoted the lysosomal degradation of toll-like receptor 4 by cooperating with the ESCRT endosomal sorting complex to facilitate MVB formation, and the ubiquitination of Tmbim1 by the E3 ubiquitin ligase Nedd4l was required for this process. We also found that overexpression of Tmbim1 in the liver effectively inhibited a severe form of NAFLD in mice and NASH progression in monkeys. Taken together, these findings could lead to the development of promising strategies to treat NASH by targeting MVB regulators to properly orchestrate the lysosome-mediated protein degradation of key mediators of the disease.


Nature Medicine | 2017

The deubiquitinating enzyme TNFAIP3 mediates inactivation of hepatic ASK1 and ameliorates nonalcoholic steatohepatitis

Peng Zhang; Pi-Xiao Wang; Ling-Ping Zhao; Xin Zhang; Yan-Xiao Ji; Xiao-Jing Zhang; Chun Fang; Yue-Xin Lu; Xia Yang; Mao-Mao Gao; Yan Zhang; Song Tian; Xueyong Zhu; Jun Gong; Xin-Liang Ma; Feng Li; Zhihua Wang; Zan Huang; Zhi-Gang She; Hongliang Li

Activation of apoptosis signal-regulating kinase 1 (ASK1) in hepatocytes is a key process in the progression of nonalcoholic steatohepatitis (NASH) and a promising target for treatment of the condition. However, the mechanism underlying ASK1 activation is still unclear, and thus the endogenous regulators of this kinase remain open to be exploited as potential therapeutic targets. In screening for proteins that interact with ASK1 in the context of NASH, we identified the deubiquitinase tumor necrosis factor alpha–induced protein 3 (TNFAIP3) as a key endogenous suppressor of ASK1 activation, and we found that TNFAIP3 directly interacts with and deubiquitinates ASK1 in hepatocytes. Hepatocyte-specific ablation of Tnfaip3 exacerbated nonalcoholic fatty liver disease– and NASH-related phenotypes in mice, including glucose metabolism disorders, lipid accumulation and enhanced inflammation, in an ASK1-dependent manner. In contrast, transgenic or adeno-associated virus–mediated TNFAIP3 gene delivery in the liver in both mouse and nonhuman primate models of NASH substantially blocked the onset and progression of the disease. These results implicate TNFAIP3 as a functionally important endogenous suppressor of ASK1 hyperactivation in the pathogenesis of NASH and identify it as a potential new molecular target for NASH therapy.


Hepatology | 2017

USP18 protects against hepatic steatosis and insulin resistance through its deubiquitinating activity

Shimin An; Ling-Ping Zhao; Li-Jun Shen; Siyuan Wang; Kuo Zhang; Yu Qi; Jilin Zheng; Xiao-Jing Zhang; Xueyong Zhu; Rong Bao; Ling Yang; Yue-Xin Lu; Zhi-Gang She; Yi-Da Tang

Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, impaired insulin sensitivity, and chronic low‐grade inflammation. However, the pathogenic mechanism of NAFLD is poorly understood, which hinders the exploration of possible treatments. Here, we report that ubiquitin‐specific protease 18 (USP18), a member of the deubiquitinating enzyme family, plays regulatory roles in NAFLD progression. Expression of USP18 was down‐regulated in the livers of nonalcoholic steatohepatitis patients and high‐fat diet (HFD)–induced or genetically obese mice. When challenged with HFD, hepatocyte‐specific USP18 transgenic mice exhibited improved lipid metabolism and insulin sensitivity, whereas mice knocked out of USP18 expression showed adverse trends regarding hepatic steatosis and glucose metabolic disorders. Furthermore, the concomitant inflammatory response was suppressed in USP18–hepatocyte‐specific transgenic mice and promoted in USP18–hepatocyte‐specific knockout mice treated with HFD. Mechanistically, hepatocyte USP18 ameliorates hepatic steatosis by interacting with and deubiquitinating transforming growth factorβ‐activated kinase 1 (TAK1), which inhibits TAK1 activation and subsequently suppresses the downstream c‐Jun N‐terminal kinase and nuclear factor kappa B signaling pathways. This is further validated by alleviated steatotic phenotypes and highly activated insulin signaling in HFD‐fed USP18–hepatocyte‐specific knockout mice administered a TAK1 inhibitor. The therapeutic effect of USP18 on NAFLD relies on its deubiquitinating activity because HFD‐fed mice injected with active‐site mutant USP18 failed to inhibit hepatic steatosis. Conclusion: USP18 associates with and deubiquitinates TAK1 to protect against hepatic steatosis, insulin resistance, and the inflammatory response. (Hepatology 2017;66:1866–1884)


Endocrinology | 2013

CARD3 Deficiency Exacerbates Diet-Induced Obesity, Hepatosteatosis, and Insulin Resistance in Male Mice

Xin-An Wang; Shan Deng; Ding-Sheng Jiang; Ran Zhang; Shu-Min Zhang; Jinfeng Zhong; Li Yang; Tao Wang; Shufen Hong; Sen Guo; Zhi-Gang She; Xiao-Dong Zhang; Hongliang Li

Caspase activation and recruitment domain 3 (CARD3) is a 61-kDa protein kinase with an N-terminal serine/threonine kinase domain and a C-terminal CARD. Previous research on the function of CARD3 has focused on its role in the immune response and inflammatory diseases. Obesity is now a worldwide health problem and is generally recognized as an inflammatory disease. Unexpectedly, we found that CARD3 expression was lower during obesity. In this study, we explored the biological and genetic bases of obesity using CARD3-knockout (KO) and wild-type (WT) mice fed a high-fat diet (HFD) for 24 weeks. We demonstrate that KO mice were more obese than their WT littermates, and KO mice exhibited obvious visceral fat accumulation and liver weight gains after 24 weeks of HFD feeding. We also observed more severe hepatosteatosis in KO mice compared with the WT controls. Hepatic steatosis in the HFD-fed KO mice was linked to a significant increase in the expression of key lipogenic and cholesterol synthesis enzymes, whereas the expression of the enzymes involves in β-oxidation was dramatically reduced. Furthermore, we confirmed the repression of AMP-activated protein kinase signaling and activation of the endoplasmic reticulum stress response. Fatty liver impaired the global glucose and lipid metabolism, which further exacerbated the insulin resistance associated with the repression of Akt signaling and up-regulated systemic inflammation through the M1/M2 (pro- and anti-inflammation) type switch and the activation of the nuclear factor-κB pathway. Our studies demonstrate the crucial role of CARD3 in metabolism and indicate that CARD3 deficiency promotes the diet-induced phenotype of type 2 diabetes.


Nature Medicine | 2018

The deubiquitinating enzyme cylindromatosis mitigates nonalcoholic steatohepatitis

Yan-Xiao Ji; Zan Huang; Xia Yang; Xiaozhan Wang; Ling-Ping Zhao; Pi-Xiao Wang; Xiao-Jing Zhang; Michele Alves-Bezerra; Lin Cai; Peng Zhang; Yue-Xin Lu; Lan Bai; Mao-Mao Gao; Huan Zhao; Song Tian; Yong Wang; Zhi-Xiang Huang; Xueyong Zhu; Yan Zhang; Jun Gong; Zhi-Gang She; Feng Li; David E. Cohen; Hongliang Li

Nonalcoholic steatohepatitis (NASH) is a common clinical condition that can lead to advanced liver diseases. Lack of effective pharmacotherapies for NASH is largely attributable to an incomplete understanding of its pathogenesis. The deubiquitinase cylindromatosis (CYLD) plays key roles in inflammation and cancer. Here we identified CYLD as a suppressor of NASH in mice and in monkeys. CYLD is progressively degraded upon interaction with the E3 ligase TRIM47 in proportion to NASH severity. We observed that overexpression of Cyld in hepatocytes concomitantly inhibits lipid accumulation, insulin resistance, inflammation and fibrosis in mice with NASH induced in an experimental setting. Mechanistically, CYLD interacts directly with the kinase TAK1 and removes its K63-linked polyubiquitin chain, which blocks downstream activation of the JNK–p38 cascades. Notably, reconstitution of hepatic CYLD expression effectively reverses disease progression in mice with dietary or genetically induced NASH and in high-fat diet–fed monkeys predisposed to metabolic syndrome. Collectively, our findings demonstrate that CYLD mitigates NASH severity and identify the CYLD–TAK1 axis as a promising therapeutic target for management of the disease.

Collaboration


Dive into the Zhi-Gang She's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge