Network


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

Hotspot


Dive into the research topics where Yuxiang Sun is active.

Publication


Featured researches published by Yuxiang Sun.


International Journal of Molecular Sciences | 2017

Suppression of GHS-R in AgRP Neurons Mitigates Diet-Induced Obesity by Activating Thermogenesis

Chia Shan Wu; Odelia Y. N. Bongmba; Jing Yue; Jong Han Lee; Li-Gen Lin; Kenji Saito; Geetali Pradhan; De Pei Li; Hui Lin Pan; Allison W. Xu; Shaodong Guo; Yong Xu; Yuxiang Sun

Ghrelin, an orexigenic hormone released primarily from the gut, signals the hypothalamus to stimulate growth hormone release, enhance appetite and promote weight gain. The ghrelin receptor, aka Growth Hormone Secretagogue Receptor (GHS-R), is highly expressed in the brain, with highest expression in Agouti-Related Peptide (AgRP) neurons of the hypothalamus. We recently reported that neuron-specific deletion of GHS-R completely prevents diet-induced obesity (DIO) in mice by activating non-shivering thermogenesis. To further decipher the specific neuronal circuits mediating the metabolic effects of GHS-R, we generated AgRP neuron-specific GHS-R knockout mice (AgRP-Cre;Ghsrf/f). Our data showed that GHS-R in AgRP neurons is required for ghrelin’s stimulatory effects on growth hormone secretion, acute food intake and adiposity, but not for long-term total food intake. Importantly, deletion of GHS-R in AgRP neurons attenuated diet-induced obesity (DIO) and enhanced cold-resistance in mice fed high fat diet (HFD). The HFD-fed knockout mice showed increased energy expenditure, and exhibited enhanced thermogenic activation in both brown and subcutaneous fat; this implies that GHS-R suppression in AgRP neurons enhances sympathetic outflow. In summary, our results suggest that AgRP neurons are key site for GHS-R mediated thermogenesis, and demonstrate that GHS-R in AgRP neurons plays crucial roles in governing energy utilization and pathogenesis of DIO.


Journal of Neurochemistry | 2017

Ghrelin receptors mediate ghrelin-induced excitation of agouti-related protein/neuropeptide Y but not pro-opiomelanocortin neurons

Shao Rui Chen; Hong Chen; Jing Jing Zhou; Geetali Pradhan; Yuxiang Sun; Hui Lin Pan; De Pei Li

Ghrelin increases food intake and body weight by stimulating orexigenic agouti‐related protein (AgRP)/neuropeptide Y (NPY) neurons and inhibiting anorexic pro‐opiomelanocortin (POMC) neurons in the hypothalamus. Growth hormone secretagogue receptor (Ghsr) mediates the effect of ghrelin on feeding behavior and energy homeostasis. However, the role of Ghsr in the ghrelin effect on these two populations of neurons is unclear. We hypothesized that Ghsr mediates the effect of ghrelin on AgRP and POMC neurons. In this study, we determined whether Ghsr similarly mediates the effects of ghrelin on AgRP/NPY and POMC neurons using cell type‐specific Ghsr‐knockout mice. Perforated whole‐cell recordings were performed on green fluorescent protein‐tagged AgRP/NPY and POMC neurons in the arcuate nucleus in hypothalamic slices. In Ghsr+/+ mice, ghrelin (100 nM) significantly increased the firing activity of AgRP/NPY neurons but inhibited the firing activity of POMC neurons. In Ghsr−/− mice, the excitatory effect of ghrelin on AgRP/NPY neurons was abolished. Ablation of Ghsr also eliminated ghrelin‐induced increases in the frequency of GABAergic inhibitory postsynaptic currents of POMC neurons. Strikingly, ablation of Ghsr converted the ghrelin effect on POMC neurons from inhibition to excitation. Des‐acylated ghrelin had no such effect on POMC neurons in Ghsr−/− mice. In both Ghsr+/+ and Ghsr−/− mice, blocking GABAA receptors with gabazine increased the basal firing activity of POMC neurons, and ghrelin further increased the firing activity of POMC neurons in the presence of gabazine. Our findings provide unequivocal evidence that Ghsr is essential for ghrelin‐induced excitation of AgRP/NPY neurons. However, ghrelin excites POMC neurons through an unidentified mechanism that is distinct from conventional Ghsr.


International Journal of Molecular Sciences | 2017

Suppression of Ghrelin Exacerbates HFCS-Induced Adiposity and Insulin Resistance

Xiaojun Ma; Li-Gen Lin; Jing Yue; Chia Shan Wu; Cathy A. Guo; Ruitao Wang; Kai Jiang Yu; Sridevi Devaraj; Peter S. Murano; Zheng Chen; Yuxiang Sun

High fructose corn syrup (HFCS) is widely used as sweetener in processed foods and soft drinks in the United States, largely substituting sucrose (SUC). The orexigenic hormone ghrelin promotes obesity and insulin resistance; ghrelin responds differently to HFCS and SUC ingestion. Here we investigated the roles of ghrelin in HFCS- and SUC-induced adiposity and insulin resistance. To mimic soft drinks, 10-week-old male wild-type (WT) and ghrelin knockout (Ghrelin−/−) mice were subjected to ad lib. regular chow diet supplemented with either water (RD), 8% HFCS (HFCS), or 10% sucrose (SUC). We found that SUC-feeding induced more robust increases in body weight and body fat than HFCS-feeding. Comparing to SUC-fed mice, HFCS-fed mice showed lower body weight but higher circulating glucose and insulin levels. Interestingly, we also found that ghrelin deletion exacerbates HFCS-induced adiposity and inflammation in adipose tissues, as well as whole-body insulin resistance. Our findings suggest that HFCS and SUC have differential effects on lipid metabolism: while sucrose promotes obesogenesis, HFCS primarily enhances inflammation and insulin resistance, and ghrelin confers protective effects for these metabolic dysfunctions.


Scientific Reports | 2017

Obestatin stimulates glucose-induced insulin secretion through ghrelin receptor GHS-R

Geetali Pradhan; Chia Shan Wu; Jong Han Lee; Preeti Kanikarla; Shaodong Guo; Vijay K. Yechoor; Susan L. Samson; Yuxiang Sun

Orexigenic hormone ghrelin and anorexic hormone obestatin are encoded by the same preproghrelin gene. While it is known that ghrelin inhibits glucose-stimulated insulin secretion (GSIS), the effect of obestatin on GSIS is unclear. Ghrelin’s effect is mediated by its receptor Growth Hormone Secretagogue Receptor (GHS-R), but the physiologically relevant receptor of obestatin remains debatable. Here we have investigated the effect of obestatin on GSIS in vitro, in vivo and ex vivo, and tested whether obestatin regulates insulin secretion through GHS-R. We found that under hyperglycemic condition, obestatin augments GSIS in rat insulinoma cells (INS-1) and in pancreatic islets from ghrelin−/− mice. Surprisingly, obestatin-induced GSIS was absent in β-cells in which GHS-R was suppressed. Obestatin-induced insulin secretion was abolished in the circulation of Ghsr−/− mice, and in pancreatic islets isolated from Ghsr−/− mice. We also found that obestatin-induced GSIS was attenuated in islets isolated from β-cell-specific Ghsr knockout MIP-Cre/ERT;Ghsrf/f mice. Our data collectively demonstrate that obestatin is a potent insulin secretagogue under hyperglycemic condition, and obestatin’s effect on insulin secretion is mediated by GHS-R in pancreatic β-cells. Our findings reveal an intriguing insight that obestatin and ghrelin have opposing effects on insulin secretion, and both are mediated through ghrelin receptor GHS-R.


BMC Physiology | 2017

Adiponectin is required for maintaining normal body temperature in a cold environment

Qiong Wei; Jong Han Lee; Hongying Wang; Odelia Y. N. Bongmba; Chia-Shan Wu; Geetali Pradhan; Zilin Sun; Lindsey Chew; Mandeep Bajaj; Lawrence Chan; Robert S. Chapkin; Miao-Hsueh Chen; Yuxiang Sun

BackgroundThermogenic impairment promotes obesity and insulin resistance. Adiponectin is an important regulator of energy homeostasis. While many beneficial metabolic effects of adiponectin resemble that of activated thermogenesis, the role of adiponectin in thermogenesis is not clear. In this study, we investigated the role of adiponectin in thermogenesis using adiponectin-null mice (Adipoq−/−).MethodsBody composition was measured using EchoMRI. Metabolic parameters were determined by indirect calorimetry. Insulin sensitivity was evaluated by glucose- and insulin- tolerance tests. Core body temperature was measured by a TH-8 temperature monitoring system. Gene expression was assessed by real-time PCR and protein levels were analyzed by Western blotting and immunohistochemistry. The mitochondrial density of brown adipose tissue was quantified by calculating the ratio of mtDNA:total nuclear DNA.ResultsUnder normal housing temperature of 24xa0°C and ad libitum feeding condition, the body weight, body composition, and metabolic profile of Adipoq−/− mice were unchanged. Under fasting condition, Adipoq−/− mice exhibited reduced energy expenditure. Conversely, under cold exposure, Adipoq−/− mice exhibited reduced body temperature, and the expression of thermogenic regulatory genes was significantly reduced in brown adipose tissue (BAT) and subcutaneous white adipose tissue (WAT). Moreover, we observed that mitochondrial content was reduced in BAT and subcutaneous WAT, and the expression of mitochondrial fusion genes was decreased in BAT of Adipoq−/− mice, suggesting that adiponectin ablation diminishes mitochondrial biogenesis and altered mitochondrial dynamics. Our study further revealed that adiponectin deletion suppresses adrenergic activation, and down-regulates β3-adrenergic receptor, insulin signaling, and the AMPK-SIRT1 pathway in BAT.ConclusionsOur findings demonstrate that adiponectin is an essential regulator of thermogenesis, and adiponectin is required for maintaining body temperature under cold exposure.


The FASEB Journal | 2018

Shift work cycle-induced alterations of circadian rhythms potentiate the effects of high fat diet on inflammation and metabolism

Sam-Moon Kim; Nichole Neuendorff; Robert C. Alaniz; Yuxiang Sun; Robert S. Chapkin; David J. Earnest

Based on genetic models with mutation or deletion of core clock genes, circadian disruption has been implicated in the pathophysiology of metabolic disorders. Thus, we examined whether circadian desynchronization in response to shift work–type schedules is sufficient to compromise metabolic homeostasis and whether inflammatory mediators provide a key link in the mechanism by which alterations of circadian timekeeping contribute to diet‐induced metabolic dysregulation. In high‐fat diet (HFD)‐fed mice, exposure to chronic shifts of the light‐dark cycle (12 h advance every 5 d): 1) disrupts photoentrainment of circadian behavior and modulates the period of spleen and macrophage clock gene rhythms; 2) potentiates HFD‐induced adipose tissue infiltration and activation of proinflammatory M1 macrophages; 3) amplifies macrophage proinflammatory cytokine expression in adipose tissue and bone marrow–derived macrophages; and 4) exacerbates diet‐induced increases in body weight, insulin resistance, and glucose intolerance in the absence of changes in total daily food intake. Thus, complete disruption of circadian rhythmicity or clock gene function as transcription factors is not requisite to the link between circadian and metabolic phenotypes. These findings suggest that macrophage proinflammatory activation and inflammatory signaling are key processes in the physiologic cascade by which dysregulation of circadian rhythmicity exacerbates diet‐induced systemic insulin resistance and glucose intolerance.—Kim, S.‐M., Neuendorff, N., Alaniz, R. C., Sun, Y., Chapkin, R. S., Earnest, D. J. Shift work cycle‐induced alterations of circadian rhythms potentiate the effects of high‐fat diet on inflammation and metabolism. FASEB J. 32, 3085–3095 (2018). www.fasebj.org


Diabetes | 2018

Novel Mechanism of Foxo1 Phosphorylation in Glucagon Signaling in Control of Glucose Homeostasis

Yuxin Wu; Quan Pan; Hui Yan; Kebin Zhang; Xiaoqin Guo; Zihui Xu; Wanbao Yang; Yajuan Qi; Cathy A. Guo; Caitlyn Hornsby; Lin Zhang; Aimin Zhou; Ling Li; Yunmei Chen; Weiping Zhang; Yuxiang Sun; Hongting Zheng; Fred Wondisford; Ling He; Shaodong Guo

Dysregulation of hepatic glucose production (HGP) serves as a major underlying mechanism for the pathogenesis of type 2 diabetes. The pancreatic hormone glucagon increases and insulin suppresses HGP, controlling blood glucose homeostasis. The forkhead transcription factor Foxo1 promotes HGP through increasing expression of genes encoding the rate-limiting enzymes responsible for gluconeogenesis. We previously established that insulin suppresses Foxo1 by Akt-mediated phosphorylation of Foxo1 at Ser256 in human hepatocytes. In this study, we found a novel Foxo1 regulatory mechanism by glucagon, which promotes Foxo1 nuclear translocation and stability via cAMP- and protein kinase A–dependent phosphorylation of Foxo1 at Ser276. Replacing Foxo1-S276 with alanine (A) or aspartate (D) to block or mimic phosphorylation, respectively, markedly regulates Foxo1 stability and nuclear localization in human hepatocytes. To establish in vivo function of Foxo1-Ser276 phosphorylation in glucose metabolism, we generated Foxo1-S273A and Foxo1-S273D knock-in (KI) mice. The KI mice displayed impaired blood glucose homeostasis, as well as the basal and glucagon-mediated HGP in hepatocytes. Thus, Foxo1-Ser276 is a new target site identified in the control of Foxo1 bioactivity and associated metabolic diseases.


Journal of Neurology & Neuromedicine | 2018

New Insights on Neuronal Functions of Ghrelin Receptor GHS-R in Obesity

Jong Han Lee; Sahar Eshghjoo; Jon Davis; Robert C. Alaniz; Yuxiang Sun; Incheon; Gyeonggi; Korea; Houston; Tx; sup>Integrative Physiology; Pullman Wa; blockquote


Diabetes | 2018

Foxo1-Ser253 Phosphorylation Regulates Glucose Homeostasis in Mice

Hui Yan; Yuxin Wu; Quan Pan; Zheng Shen; Hongting Zheng; Morris F. White; Yuxiang Sun; Shaodong Guo


Diabetes | 2018

Glucagon Regulates Hepatic Mitochondrial Biogenesis and Function through Foxo1

Wanbao Yang; Hui Yan; Quan Pan; Zheng Shen; Sudath A. Dahanayaka; Chaodong Wu; Yuxiang Sun; Shaodong Guo

Collaboration


Dive into the Yuxiang Sun's collaboration.

Top Co-Authors

Avatar

Geetali Pradhan

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jong Han Lee

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongting Zheng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Aimin Zhou

Cleveland State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge