Network


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

Hotspot


Dive into the research topics where Chunjiong Wang is active.

Publication


Featured researches published by Chunjiong Wang.


International Journal of Endocrinology | 2010

Cytokines in the Progression of Pancreatic β-Cell Dysfunction

Chunjiong Wang; Youfei Guan; Jichun Yang

The dysfunction of pancreatic β-cell and the reduction in β-cell mass are the decisive events in the progression of type 2 diabetes. There is increasing evidence that cytokines play important roles in the procedure of β-cell failure. Cytokines, such as IL-1β, IFN-γ, TNF-α, leptin, resistin, adiponectin, and visfatin, have been shown to diversely regulate pancreatic β-cell function. Recently, islet-derived cytokine PANcreatic DERived factor (PANDER or FAM3B) has also been demonstrated to be a regulator of islet β-cell function. The change in cytokine profile in islet and plasma is associated with pancreatic β-cell dysfunction and apoptosis. In this paper, we summarize and discuss the recent studies on the effects of certain important cytokines on pancreatic β-cell function. The imbalance in deleterious and protective cytokines plays pivotal roles in the development and progression of pancreatic β-cell dysfunction under insulin-resistant conditions.


Hepatology | 2011

Pancreatic-derived factor promotes lipogenesis in the mouse liver: Role of the Forkhead box 1 signaling pathway†‡

Jing Li; Yujing Chi; Chunjiong Wang; Jing Wu; Hang Yang; Dongjuan Zhang; Yi Zhu; Nanping Wang; Jichun Yang; Youfei Guan

Pancreatic‐derived factor (PANDER) is a pancreatic islet‐specific cytokine that cosecretes with insulin and is important for β cell function. Here, we show that PANDER is constitutively expressed in hepatocytes, and its expression is significantly increased in steatotic livers of diabetic insulin‐resistant db/db mice and mice fed a high‐fat diet. Overexpression of PANDER in the livers of C57Bl/6 mice promoted lipogenesis, with increased Forkhead box 1 (FOXO1) expression, whereas small interfering RNA–mediated knockdown of hepatic PANDER significantly attenuated steatosis, with reduced FOXO1 expression in db/db mice. Hepatic PANDER silencing also attenuated insulin resistance and hyperglycemia in db/db mice. In cultured hepatocytes, PANDER overexpression induced lipid deposition, increased FOXO1 expression, and suppressed insulin‐stimulated Akt activation and FOXO1 inactivation. Moreover, FOXO1 overexpression increased PANDER expression in cultured hepatocytes and mouse livers. Conclusion: PANDER promotes lipogenesis and compromises insulin signaling in the liver by increasing FOXO1 activity. PANDER may represent a potential therapeutic target for the treatment of fatty liver and insulin resistance. (HEPATOLOGY 2011;)


Hepatology | 2014

FAM3A activates PI3K p110α/Akt signaling to ameliorate hepatic gluconeogenesis and lipogenesis

Chunjiong Wang; Yujing Chi; Jing Li; Yifei Miao; Sha Li; Wen Su; Shi Jia; Zhenzhen Chen; Shengnan Du; Xiaoyan Zhang; Yunfeng Zhou; Wenhan Wu; Mingyan Zhu; Zhiwei Wang; Huaqian Yang; Guoheng Xu; Shiqiang Wang; Jichun Yang; Youfei Guan

FAM3A belongs to a novel cytokine‐like gene family, and its physiological role remains largely unknown. In our study, we found a marked reduction of FAM3A expression in the livers of db/db and high‐fat diet (HFD)‐induced diabetic mice. Hepatic overexpression of FAM3A markedly attenuated hyperglycemia, insulin resistance, and fatty liver with increased Akt (pAkt) signaling and repressed gluconeogenesis and lipogenesis in the livers of those mice. In contrast, small interfering RNA (siRNA)‐mediated knockdown of hepatic FAM3A resulted in hyperglycemia with reduced pAkt levels and increased gluconeogenesis and lipogenesis in the livers of C57BL/6 mice. In vitro study revealed that FAM3A was mainly localized in the mitochondria, where it increases adenosine triphosphate (ATP) production and secretion in cultured hepatocytes. FAM3A activated Akt through the p110α catalytic subunit of PI3K in an insulin‐independent manner. Blockade of P2 ATP receptors or downstream phospholipase C (PLC) and IP3R and removal of medium calcium all significantly reduced FAM3A‐induced increase in cytosolic free Ca2+ levels and attenuated FAM3A‐mediated PI3K/Akt activation. Moreover, FAM3A‐induced Akt activation was completely abolished by the inhibition of calmodulin (CaM). Conclusion: FAM3A plays crucial roles in the regulation of glucose and lipid metabolism in the liver, where it activates the PI3K‐Akt signaling pathway by way of a Ca2+/CaM‐dependent mechanism. Up‐regulating hepatic FAM3A expression may represent an attractive means for the treatment of insulin resistance, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD). (Hepatology 2014;59:1779–1790)


FEBS Letters | 2009

PANDER binds to the liver cell membrane and inhibits insulin signaling in HepG2 cells

Jichun Yang; Chunjiong Wang; Jing Li; Brant R. Burkhardt; Claudia E. Robert-Cooperman; Camella G. Wilson; Zhiyong Gao; Bryan A. Wolf

PANDER is a cytokine co‐secreted with insulin from islet β‐cells. To date, the physiological function of PANDER remains largely unknown. Here we show that PANDER binds to the liver membrane by 125I‐PANDER saturation and competitive binding assays. In HepG2 cells, pre‐treatment with PANDER ranging from 4 pM to 4 nM for 8 h resulted in a maximal inhibition of insulin‐stimulated activation of insulin receptor and insulin receptor substrate 1 by 52% and 63%, respectively. Moreover, PANDER treatment also reduced insulin‐stimulated PI3K and pAkt levels by 55% and 48%, respectively. In summary, we have identified the liver as a novel target for PANDER, and PANDER may be involved in the progression of diabetes by regulating hepatic insulin signaling pathways.


Hepatology | 2013

Thyroid hormone-responsive SPOT 14 homolog promotes hepatic lipogenesis, and its expression is regulated by liver X receptor α through a sterol regulatory element-binding protein 1c-dependent mechanism in mice.

Jing Wu; Chunjiong Wang; Shuo Li; Sha Li; Wanyi Wang; Jing Li; Yujing Chi; Hang Yang; Xiaomu Kong; Yunfeng Zhou; Chengyan Dong; Fan Wang; Guoheng Xu; Jichun Yang; Jan Åke Gustafsson; Youfei Guan

The protein, thyroid hormone‐responsive SPOT 14 homolog (Thrsp), has been reported to be a lipogenic gene in cultured hepatocytes, implicating an important role of Thrsp in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Thrsp expression is known to be regulated by a variety of transcription factors, including thyroid hormone receptor, pregnane X receptor, and constitutive androstane receptor. Emerging in vitro evidence also points to a critical role of liver X receptor (LXR) in regulating Thrsp transcription in hepatocytes. In the present study, we showed that Thrsp was up‐regulated in livers of db/db mice and high‐fat‐diet–fed mice, two models of murine NAFLD. Hepatic overexpression of Thrsp increased triglyceride accumulation with enhanced lipogenesis in livers of C57Bl/6 mice, whereas hepatic Thrsp gene silencing attenuated the fatty liver phenotype in db/db mice. LXR activator TO901317 induced Thrsp expression in livers of wild‐type (WT) and LXR‐β gene‐deficient mice, but not in LXR‐α or LXR‐α/β double‐knockout mice. TO901317 treatment significantly enhanced hepatic sterol regulatory element‐binding protein 1c (SREBP‐1c) expression and activity in WT mice, but failed to induce Thrsp expression in SREBP‐1c gene‐deficient mice. Sequence analysis revealed four LXR response‐element–like elements and one sterol regulatory element (SRE)‐binding site within a −2,468 ∼+1‐base‐pair region of the Thrsp promoter. TO901317 treatment and LXR‐α overexpression failed to induce, whereas overexpression of SREBP‐1c significantly increased Thrsp promoter activity. Moreover, deletion of the SRE site completely abolished SREBP‐1c–induced Thrsp transcription. Conclusion: Thrsp is a lipogenic gene in the liver that is induced by the LXR agonist through an LXR‐α–mediated, SREBP‐1c–dependent mechanism. Therefore, Thrsp may represent a potential therapeutic target for the treatment of NAFLD. (Hepatology 2013;58:617–628)


Nutrition Reviews | 2012

Role of pancreatic-derived factor in type 2 diabetes: evidence from pancreatic β cells and liver

Chunjiong Wang; Brant R. Burkhardt; Youfei Guan; Jichun Yang

Pancreatic-derived factor (PANDER) is a cytokine-like protein that is highly expressed in pancreatic islets. In vitro, PANDER pretreatment or viral-mediated overexpression promotes apoptosis of islet β cells. Under conditions of insulin resistance, chronic hyperglycemia potently activates PANDER expression and stimulates the cosecretion of insulin and PANDER in β cells. PANDER binds to the liver cell membrane and induces insulin resistance, resulting in increased gluconeogenesis. Recently, PANDER was found to be expressed in rodent and human liver, and its expression is increased in the liver of diabetic mice and rats. Hepatic overexpression of PANDER promotes lipogenesis in the liver and induces insulin resistance in C57BL/6 mice, whereas the inactivation of hepatic PANDER markedly reduces steatosis, insulin resistance, and hyperglycemia in db/db mice. PANDER deficiency protects mice from high-fat-diet-induced hyperglycemia by decreasing gluconeogenesis in the liver. In summary, PANDER plays an important role in the progression of type 2 diabetes by negatively regulating islet β-cell function and insulin sensitivity in the liver.


Diabetes | 2014

Hepatic Overexpression of ATP Synthase β Subunit Activates PI3K/Akt Pathway to Ameliorate Hyperglycemia of Diabetic Mice

Chunjiong Wang; Zhenzhen Chen; Sha Li; Yuan Zhang; Shi Jia; Jing Li; Yujing Chi; Yifei Miao; Youfei Guan; Jichun Yang

ATP synthase β subunit (ATPSβ) had been previously shown to play an important role in controlling ATP synthesis in pancreatic β-cells. This study aimed to investigate the role of ATPSβ in regulation of hepatic ATP content and glucose metabolism in diabetic mice. ATPSβ expression and ATP content were both reduced in the livers of type 1 and type 2 diabetic mice. Hepatic overexpression of ATPSβ elevated cellular ATP content and ameliorated hyperglycemia of streptozocin-induced diabetic mice and db/db mice. ATPSβ overexpression increased phosphorylated Akt (pAkt) levels and reduced PEPCK and G6pase expression levels in the livers. Consistently, ATPSβ overexpression repressed hepatic glucose production in db/db mice. In cultured hepatocytes, ATPSβ overexpression increased intracellular and extracellular ATP content, elevated the cytosolic free calcium level, and activated Akt independent of insulin. The ATPSβ-induced increase in cytosolic free calcium and pAkt levels was attenuated by inhibition of P2 receptors. Notably, inhibition of calmodulin (CaM) completely abolished ATPSβ-induced Akt activation in liver cells. Inhibition of P2 receptors or CaM blocked ATPSβ-induced nuclear exclusion of forkhead box O1 in liver cells. In conclusion, a decrease in hepatic ATPSβ expression in the liver, leading to the attenuation of ATP-P2 receptor-CaM-Akt pathway, may play an important role in the progression of diabetes.


Biochimica et Biophysica Acta | 2013

FAM3A is a target gene of peroxisome proliferator-activated receptor gamma

Yanlan Zhou; Shi Jia; Chunjiong Wang; Zhenzhen Chen; Yujing Chi; Jing Li; Guoheng Xu; Youfei Guan; Jichun Yang

BACKGROUND To date, the biological function of FAM3A, the first member of FAM3 gene family, remains unknown. We aimed to investigate whether the expression of FAM3A in liver cells is regulated by peroxisome proliferator-activated receptors (PPARs). METHODS AND RESULTS The transcriptional activity of human and mouse FAM3A gene promoters was determined by luciferase reporter assay system. PPARγ agonist rosiglitazone induced FAM3A expression in primary cultured mouse hepatocytes and human HepG2 cells. PPARγ antagonism blocked rosiglitazone-induced FAM3A expression, whereas PPARγ overexpression stimulated FAM3A expression in HepG2 cells. In contrast, PPARα agonist fenofibrate or PPARβ agonist GW0742 failed to affect FAM3A expression in HepG2 cells. The transcriptional activities of human and mouse FAM3A promoters were markedly stimulated by PPARγ activation, but not by PPARα and PPARβ activation. Chromatin immunoprecipitation (ChIP) assay revealed a direct binding of PPARγ to the putative peroxisome proliferator response element (PPRE) located at -1258/-1246 in the human FAM3A promoter. Site-directed mutagenesis of this PPRE-like motif abolished PPARγs stimulatory effect on the transcriptional activity of human FAM3A promoter. In vivo, oral rosiglitazone treatment upregulated FAM3A expression in the livers of C57BL/6 mice and db/db mice. Moreover, upregulation of FAM3A by PPARγ activation was correlated with increased level of phosphorylated Akt (pAkt) in liver cells. CONCLUSIONS FAM3A as a novel target gene of PPARγ. Upregulation of FAM3A by PPARγ activation is correlated with increased pAkt level in liver cells. GENERAL SIGNIFICANCE Upregulation of FAM3A might contribute to PPARγs metabolic effects in the liver.


Journal of Diabetes | 2014

Liver X receptor activation increases hepatic fatty acid desaturation by the induction of SCD1 expression through an LXRα-SREBP1c-dependent mechanism (肝X受体活化可通过LXRα-SREBP1c依赖的机制诱导SCD1表达来增加肝脏脂肪酸不饱和度): LXRα activation induces hepatic SCD1 expression

Xiaoyan Zhang; Jia Liu; Wen Su; Jing Wu; Chunjiong Wang; Xiaomu Kong; Jan Åke Gustafsson; Jie Ding; Xiaosong Ma; Youfei Guan

Liver X receptors (LXRs) including LXRα and LXRβ are members of the nuclear hormone receptor superfamily of ligand activated transcription factors, which serve as lipid sensors to regulate expression of genes controlling many aspects of cholesterol and fatty acid metabolism. The liver is the central organ in controlling lipid metabolism. In the present study, we aimed at elucidating the role of LXR activation in hepatic fatty acid homeostasis.


Journal of Diabetes | 2014

Intracellular and extracellular adenosine triphosphate in regulation of insulin secretion from pancreaticβcells (细胞内外三磷酸腺苷对胰岛β细胞胰岛素分泌的调控作用): ATP regulating insulin from pancreaticβcells

Chunjiong Wang; Bin Geng; Qinghua Cui; Youfei Guan; Jichun Yang

Adenosine triphosphate (ATP) synthesis and release in mitochondria play critical roles in regulating insulin secretion in pancreatic β cells. Mitochondrial dysfunction is mainly characterized by a decrease in ATP production, which is a central event in the progression of pancreatic β cell dysfunction and diabetes. ATP has been demonstrated to regulate insulin secretion via several pathways: (i) Intracellular ATP directly closes ATP‐sensitive potassium channel to open L‐type calcium channel, leading to an increase in free cytosolic calcium levels and exocytosis of insulin granules; (ii) A decrease in ATP production is always associated with an increase in production of reactive oxygen species, which exerts deleterious effects on pancreatic β cell survival and insulin secretion; and (iii) ATP can be co‐secreted with insulin from pancreatic β cells, and the released ATP functions as an autocrine signal to modulate insulin secretory process via P2 receptors on the cell membrane. In this review, the recent findings regarding the role and mechanism of ATP synthesis and release in regulation of insulin secretion from pancreatic β cells will be summarized and discussed.

Collaboration


Dive into the Chunjiong Wang'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