Network


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

Hotspot


Dive into the research topics where Chunxiao Yu is active.

Publication


Featured researches published by Chunxiao Yu.


Hepatology | 2010

A novel role for thyroid-stimulating hormone: Up-regulation of hepatic 3-hydroxy-3-methyl-glutaryl-coenzyme a reductase expression through the cyclic adenosine monophosphate/protein kinase A/cyclic adenosine monophosphate–responsive element binding protein pathway†

Limin Tian; Yongfeng Song; Mingzhao Xing; Wei Zhang; Guang Ning; Xiaoying Li; Chunxiao Yu; Chengkong Qin; Jun Liu; Xingsong Tian; Xianglan Sun; Rui Fu; Lin Zhang; Xiujuan Zhang; Yan Lu; Jianwen Zou; Laicheng Wang; Qingbo Guan; Ling Gao; Jiajun Zhao

Elevated thyroid‐stimulating hormone (TSH) and hypercholesterolemia commonly coexist, as typically seen in hypothyroidism, but there is no known mechanism directly linking the two. Here, we demonstrated that in liver cells, TSH promoted the expression of 3‐hydroxy‐3‐methyl‐glutaryl coenzyme A reductase (HMGCR), a rate‐limiting enzyme in cholesterol synthesis, by acting on the TSH receptor in hepatocyte membranes and stimulating the cyclic adenosine monophosphate / protein kinase A / cyclic adenosine monophosphate–responsive element binding protein (cAMP/PKA/CREB) signaling system. In thyroidectomized rats, the production of endogenous thyroid hormone was eliminated and endogenous TSH was suppressed through pituitary suppression with constant administration of exogenous thyroid hormone, and hepatic HMGCR expression was increased by administration of exogenous TSH. These results suggested that TSH could up‐regulate hepatic HMGCR expression, which indicated a potential mechanism for hypercholesterolemia involving direct action of TSH on the liver. (HEPATOLOGY 2010)


The Journal of Clinical Endocrinology and Metabolism | 2012

Thyroid-Stimulating Hormone Levels within the Reference Range Are Associated with Serum Lipid Profiles Independent of Thyroid Hormones

Furong Wang; Yinyin Tan; Chenggang Wang; Xu Zhang; Yuanfei Zhao; Xinhong Song; Bingchang Zhang; Qingbo Guan; Jin Xu; Ji Zhang; Dongzhi Zhang; Haiyan Lin; Chunxiao Yu; Jiajun Zhao

CONTEXT AND OBJECTIVE Dyslipidemia in thyroid dysfunction has always been attributed to changes in thyroid hormone (TH) levels. We hypothesized that TSH plays an important role in lipid metabolism independent of TH. DESIGN AND SETTING We conducted a cross-sectional study to investigate the relationship between serum TSH levels and lipid profiles after controlling for free T(3), free T(4), total T(3), total T(4) and nonthyroid factors relevant to lipid metabolism in euthyroid Chinese subjects. MAIN OUTCOME MEASURES General linear analysis was performed to determine whether the impact of TSH on serum lipid levels is independent of the TH levels. Moreover, path analysis, an evolutionary multivariable regression technique, was conducted to test whether there is a direct and/or indirect effect between serum TSH and total cholesterol (TC) levels. Additionally, the odds ratios (95% confidence interval) for hypercholesterolemia in relation to TSH categories were calculated. RESULTS A total of 3664 euthyroid subjects were finally analyzed. There was a significant linear trend toward higher log TC (P = 0.021) and log triglyceride (P = 0.001) levels with increasing serum TSH levels within the reference range, which remained significant after adjusting for factors such as TH levels, age, and smoking. Most importantly, the total effect of TSH on TC levels (total effect(TC, TSH) = 0.05253) includes a direct effect (direct effect(TC, TSH) = 0.05979) and an indirect effect via TH. Compared with subjects in the lower part of the reference range (TSH level, 0.27-0.61 mIU/liter), the adjusted odds ratio for hypercholesterolemia was 3.239 (95% confidence interval, 1.392-7.538; P = 0.007) for those in the upper category (TSH level, 4.61-5.5 mIU/liter). CONCLUSIONS The variation in normal TSH levels is partially related to the lipid components and hypercholesterolemia in euthyroid subjects and includes both TH-dependent and TH-independent effects. Our study suggests the importance of controlling TSH in hypothyroid subjects.


Journal of Cardiovascular Pharmacology | 2012

Hydrogen sulfide protects against high-glucose-induced apoptosis in endothelial cells.

Qingbo Guan; Yuan Zhang; Chunxiao Yu; Yuantao Liu; Ling Gao; Jiajun Zhao

Hydrogen sulfide (H2S) is the third endogenous gaseous mediator identified after nitric oxide and carbon monoxide. It has been demonstrated that H2S has protective effects on myocardial ischemia/reperfusion-induced cell apoptosis. To date, little is known about the role of H2S in the pathophysiology of diabetic vascular complications. In this study, we investigated the effects of sodium hydrosulfide on high-glucose–induced apoptosis of primary human umbilical vein endothelium cells. Exposure to high glucose (25 mmole/L) for 48 hours resulted in the induction of apoptosis by 41.6% ± 1.01%, which was attenuated by pretreatment with sodium hydrosulfide (50 &mgr;mole/L) for 30 minutes. Further investigation of the apoptotic mechanisms in the cells demonstrated that high glucose upregulated the ratio of Bax/Bcl-2 and activated caspase-3 and also increased the levels of reactive oxygen species and malondialdehyde while reducing superoxide dismutase activity. All the above responses could be prevented by pretreatment with 50 &mgr;mole/L of sodium hydrosulfide. These results indicated that the protective effects of H2S on endothelial cells in the condition of high glucose might involve an antioxidative stress mechanism.


Journal of Hepatology | 2015

Thyroid-stimulating hormone regulates hepatic bile acid homeostasis via SREBP-2/HNF-4α/CYP7A1 axis

Yongfeng Song; Chao Xu; Shanshan Shao; Jun Liu; Wanjia Xing; Jin Xu; Chengkun Qin; Chunyou Li; Baoxiang Hu; Shounan Yi; Xuefeng Xia; Haiqing Zhang; Xiujuan Zhang; Tingting Wang; Wenfei Pan; Chunxiao Yu; Qiangxiu Wang; Xiaoyan Lin; Laicheng Wang; Ling Gao; Jiajun Zhao

BACKGROUND & AIMS Bile acids (BAs) play a crucial role in dietary fat digestion and in the regulation of lipid, glucose, and energy metabolism. Thyroid-stimulating hormone (TSH) is a hormone produced by the anterior pituitary gland that directly regulates several metabolic pathways. However, the impact of TSH on BA homeostasis remains largely unknown. METHODS We analyzed serum BA and TSH levels in healthy volunteers under strict control of caloric intake. Thyroidectomized rats were administered thyroxine and injected with different doses of TSH. Tshr(-/-) mice were supplemented with thyroxine, and C57BL/6 mice were injected with Tshr-siRNA via the tail vein. The serum BA levels, BA pool size, and fecal BA excretion rate were measured. The regulation of SREBP-2, HNF-4α, and CYP7A1 by TSH were analyzed using luciferase reporter, RNAi, EMSA, and CHIP assays. RESULTS A negative correlation was observed between the serum levels of TSH and the serum BA levels in healthy volunteers. TSH administration led to a decrease in BA content and CYP7A1 activity in thyroidectomized rats supplemented with thyroxine. When Tshr was silenced in mice, the BA pool size, fecal BA excretion rate, and serum BA levels all increased. Additionally, we found that HNF-4α acts as a critical molecule through which TSH represses CYP7A1 activity. We further confirmed that the accumulation of mature SREBP-2 protein could impair the capacity of nuclear HNF-4α to bind to the CYP7A1 promoter, a mechanism that appears to mediate the effects of TSH. CONCLUSIONS TSH represses hepatic BA synthesis via a SREBP-2/HNF-4α/CYP7A1 signaling pathway. This finding strongly supports the notion that TSH is an important pathophysiological regulator of liver BA homeostasis independently of thyroid hormones.


Nutrition & Metabolism | 2012

Thyroid stimulating hormone, independent of thyroid hormone, can elevate the serum total cholesterol level in patients with coronary heart disease: a cross-sectional design

Chao Xu; Xiaomei Yang; Wenhui Liu; Haitao Yuan; Chunxiao Yu; Ling Gao; Jiajun Zhao

BackgroundThe relationship between TSH and the lipid profile is contradictory because few studies have excluded the potential influence of the thyroid hormones (TH). The aim of the present study was to evaluate the relationship between serum TSH levels and the lipid profile independent of TH.Methods1302 CHD patients diagnosed by coronary angiography were retrospectively studied. The prevalence and distribution of thyroid dysfunction were analyzed first. To assess the impact of TSH on serum lipids, Pearson’s correlation analysis was performed after adjustments for classic factors and TH. To calculate the extent of the effect of TSH on the serum cholesterol level, the partial least squares method and additional statistical methods were used.ResultsAfter the exclusions, a total of 568 patients (270 males and 298 females with a mean age of 63.56 ± 11.376 years) were selected. The prevalence of thyroid dysfunction among the patients was 18.66%, and the prevalence of hypothyroidism (15.32%) was higher than that of hyperthyroidism (3.34%). Even after adjusting for confounding factors, such as sex, age, smoking status, fasting plasma glucose levels and TH, a significant positive impact of TSH on the serum total cholesterol (TC) level was revealed (r = 0.095, p = 0.036). Each 1 mIU/L increase in the TSH level might be linked to a 0.015580712 mmol/L elevation of the serum TC value.ConclusionsTSH can increase the TC level in CHD patients independent of TH. The present study suggests a potential physiological role of TSH and the importance of maintaining an appropriate TSH level in CHD patients.


PLOS ONE | 2014

Peroxisome Proliferator-Activated Receptor α Activation Induces Hepatic Steatosis, Suggesting an Adverse Effect

Fang Yan; Qi Wang; Chao Xu; Ming-feng Cao; Xiaoming Zhou; Tingting Wang; Chunxiao Yu; Fei Jing; Wenbin Chen; Ling Gao; Jiajun Zhao

Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic triglyceride accumulation, ranging from steatosis to steatohepatitis and cirrhosis. NAFLD is a risk factor for cardiovascular diseases and is associated with metabolic syndrome. Antihyperlipidemic drugs are recommended as part of the treatment for NAFLD patients. Although fibrates activate peroxisome proliferator-activated receptor α (PPARα), leading to the reduction of serum triglyceride levels, the effects of these drugs on NAFLD remain controversial. Clinical studies have reported that PPARα activation does not improve hepatic steatosis. In the present study, we focused on exploring the effect and mechanism of PPARα activation on hepatic triglyceride accumulation and hepatic steatosis. Male C57BL/6J mice, Pparα-null mice and HepG2 cells were treated with fenofibrate, one of the most commonly used fibrate drugs. Both low and high doses of fenofibrate were administered. Hepatic steatosis was detected through oil red O staining and electron microscopy. Notably, in fenofibrate-treated mice, the serum triglyceride levels were reduced and the hepatic triglyceride content was increased in a dose-dependent manner. Oil red O staining of liver sections demonstrated that fenofibrate-fed mice accumulated abundant neutral lipids. Fenofibrate also increased the intracellular triglyceride content in HepG2 cells. The expression of sterol regulatory element-binding protein 1c (SREBP-1c) and the key genes associated with lipogenesis were increased in fenofibrate-treated mouse livers and HepG2 cells in a dose-dependent manner. However, the effect was strongly impaired in Pparα-null mice treated with fenofibrate. Fenofibrate treatment induced mature SREBP-1c expression via the direct binding of PPARα to the DR1 motif of the SREBP-1c gene. Taken together, these findings indicate the molecular mechanism by which PPARα activation increases liver triglyceride accumulation and suggest an adverse effect of fibrates on the pathogenesis of hepatic steatosis.


Scientific Reports | 2013

First-principles prediction and experimental verification of glass-forming ability in Zr-Cu binary metallic glasses.

Chunxiao Yu; Xuanwen Liu; Juming Lu; Guang-Ping Zheng; Chuang Liu

In the field of metallic materials with amorphous structures, it is vitally important to understand the glass formation and to predict glass-forming ability (GFA) in terms of constituent elements and alloy compositions. In this study, an expression has been formulated from first-principles calculations to predict the trend of GFA by hybridizing both internal energies and atomic-scale defect structures. The prediction of GFA from compositions has been verified successfully by available experimental data in the model Zr-Cu alloy system. The physical scenario revealed here has extensive implications for the design of bulk metallic glasses with superior GFA.


The Journal of Clinical Endocrinology and Metabolism | 2014

Expression Profiles of Six Circulating MicroRNAs Critical to Atherosclerosis in Patients With Subclinical Hypothyroidism: A Clinical Study

Xinhuan Zhang; Shanshan Shao; Houfa Geng; Yong Yu; Chenggang Wang; Zhanfeng Liu; Chunxiao Yu; Xiuyun Jiang; Yangxin Deng; Ling Gao; Jiajun Zhao

CONTEXT Increasing evidence shows that subclinical hypothyroidism (SCH) is associated with atherosclerosis (ATH), but the association remains controversial. MicroRNAs (miRNAs) have been proved to be involved in atherosclerosis and dyslipidemia as gene regulators. OBJECTIVE The objective of the study was to determine the expression profiles of six serum miRNAs critical to atherosclerosis in SCH patients and reanalyze the association between atherosclerosis and SCH from a new perspective. OUTCOMES, DESIGN, AND PARTICIPANTS: MicroRNA profiling analysis was performed by real-time PCR in normal control subjects (NC; n = 22); patients with subclinical hypothyroidism alone (SCH; n = 20); SCH patients plus atherosclerosis (SCH+ATH; n = 21); and patients with atherosclerosis but without subclinical hypothyroidism (ATH; n = 22). RESULTS MiR-21-5p was up-regulated in SCH, SCH+ATH, and ATH groups than in the NC group. In addition, expression levels of miR-21-5p in SCH+ATH group were higher than in SCH alone and ATH alone groups, respectively. Both miR-125a-5p and miR-126-3p showed a decreased trend from NC to SCH and then to SCH+ATH or ATH subjects. MiR-221-3p and miR-222-3p were decreased in the SCH+ATH and ATH groups compared with either the NC or SCH groups. No differences were found in the levels of miR125a-5p, miR126-3p, miR221-3p, and miR222-3p between the ATH and SCH+ATH group. CONCLUSIONS MiR-21-5p showed the most specific expression patterns in all patients with subclinical hypothyroidism (SCH and SCH+ATH groups). Down-regulation of miR-125a-5p, miR-126-3p, miR-221-3p, and miR-222-3p may be a manifestation of atherosclerosis either in SCH+ATH or in ATH-alone patients. MiR-126-3p has the most specific expression patterns in all atherosclerosis patients (SCH+ATH and ATH groups).


Journal of Hepatology | 2014

Thyrotropin increases hepatic triglyceride content through upregulation of SREBP-1c activity

Fang Yan; Qi Wang; Ming Lu; Wenbin Chen; Yongfeng Song; Fei Jing; Youfei Guan; Laicheng Wang; Yanliang Lin; Tao Bo; Jie Zhang; Tingting Wang; Wei Xin; Chunxiao Yu; Qingbo Guan; Xinli Zhou; Ling Gao; Chao Xu; Jiajun Zhao

BACKGROUND & AIMS Hallmarks of non-alcoholic fatty liver disease (NAFLD) are increased triglyceride accumulation within hepatocytes. The prevalence of NAFLD increases steadily with increasing thyrotropin (TSH) levels. However, the underlying mechanisms are largely unknown. Here, we focused on exploring the effect and mechanism of TSH on the hepatic triglyceride content. METHODS As the function of TSH is mediated through the TSH receptor (TSHR), Tshr(-/-) mice (supplemented with thyroxine) were used. Liver steatosis and triglyceride content were analysed in Tshr(-/-) and Tshr(+/+) mice fed a high-fat or normal chow diet, as well as in Srebp-1c(-/-) and Tshr(-/-)Srebp-1c(-/-) mice. The expression levels of proteins and genes involved in liver triglyceride metabolism was measured. RESULTS Compared with control littermates, the high-fat diet induced a relatively low degree of liver steatosis in Tshr(-/-) mice. Even under chow diet, hepatic triglyceride content was decreased in Tshr(-/-) mice. TSH caused concentration- and time-dependent effects on intracellular triglyceride contents in hepatocytes in vitro. The activity of SREBP-1c, a key regulator involved in triglyceride metabolism and in the pathogenesis of NAFLD, was significantly lower in Tshr(-/-) mice. In Tshr(-/-)Srebp-1c(-/-) mice, the liver triglyceride content showed no significant difference compared with Tshr(+/+)Srebp-1c(-/-) mice. When mice were injected with forskolin (cAMP activator), H89 (inhibitor of PKA) or AICAR (AMPK activator), or HeG2 cells received MK886 (PPARα inhibitor), triglyceride contents presented in a manner dependent on SREBP-1c activity. The mechanism, underlying TSH-induced liver triglyceride accumulation, involved that TSH, through its receptor TSHR, triggered hepatic SREBP-1c activity via the cAMP/PKA/PPARα pathway associated with decreased AMPK, which further increased the expression of genes associated with lipogenesis. CONCLUSIONS TSH increased the hepatic triglyceride content, indicating an essential role for TSH in the pathogenesis of NAFLD.


Scientific Reports | 2015

Thyrotropin and Obesity: Increased Adipose Triglyceride Content Through Glycerol-3-Phosphate Acyltransferase 3

Shizhan Ma; Fei Jing; Chao Xu; Lingyan Zhou; Yongfeng Song; Chunxiao Yu; Dongqing Jiang; Ling Gao; Yujie Li; Qingbo Guan; Jiajun Zhao

Epidemiological evidence indicates that thyrotropin (TSH) is positively correlated with the severity of obesity. However, the mechanism remains unclear. Here, we show that TSH promoted triglyceride (TG) synthesis in differentiated adipocytes in a thyroid hormone-independent manner. Mice with subclinical hypothyroidism, which is characterized by elevated serum TSH but not thyroid hormone levels, demonstrated a 35% increase in the total white adipose mass compared with their wild-type littermates. Interestingly, Tshr KO mice, which had normal thyroid hormone levels after thyroid hormone supplementation, resisted high-fat diet-induced obesity. TSH could directly induce the activity of glycerol-3-phosphate-acyltransferase 3 (GPAT3), the rate-limiting enzyme in TG synthesis, in differentiated 3T3-L1 adipocytes. However, following either the knockdown of Tshr and PPARγ or the constitutive activation of AMPK, the changes to TSH-triggered GPAT3 activity and adipogenesis disappeared. The over-expression of PPARγ or the expression of an AMPK dominant negative mutant reversed the TSH-induced changes. Thus, TSH acted as a previously unrecognized master regulator of adipogenesis, indicating that modification of the AMPK/PPARγ/GPAT3 axis via the TSH receptor might serve as a potential therapeutic target for obesity.

Collaboration


Dive into the Chunxiao Yu'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

Jin Xu

Shandong University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge