Search Results
You are looking at 1 - 10 of 19 items for
- Author: Wei Li x
- Refine by access: All content x
Search for other papers by Liqiong Song in
Google Scholar
PubMed
Search for other papers by Wei Xia in
Google Scholar
PubMed
Search for other papers by Zhao Zhou in
Google Scholar
PubMed
Search for other papers by Yuanyuan Li in
Google Scholar
PubMed
Search for other papers by Yi Lin in
Google Scholar
PubMed
Search for other papers by Jie Wei in
Google Scholar
PubMed
Search for other papers by Zhengzheng Wei in
Google Scholar
PubMed
Search for other papers by Bing Xu in
Google Scholar
PubMed
Search for other papers by Jie Shen in
Google Scholar
PubMed
Search for other papers by Weiyong Li in
Google Scholar
PubMed
Search for other papers by Shunqing Xu in
Google Scholar
PubMed
Phenolic estrogen pollutants, a class of typical endocrine-disrupting chemicals, have attracted public attention due to their estrogenic activities of imitating steroid hormone 17β-estradiol (E2) effects. Exposure to these pollutants may disrupt insulin secretion and be a risk factor for type 2 diabetes. In this study, we investigated the direct effects of phenolic estrogen diethylstilbestrol (DES), octylphenol (OP), nonylphenol (NP), and bisphenol A (BPA) on rat pancreatic islets in vitro, whose estrogenic activities were DES>NP>OP>BPA. Isolated β-cells were exposed to E2, DES, OP, NP, or BPA (0, 0.1, 0.5, 2.5, 25, and 250 μg/l) for 24 h. Parameters of insulin secretion, content, and morphology of β-cells were measured. In the glucose-stimulated insulin secretion test, E2 and DES increased insulin secretion in a dose-dependent manner in a 16.7 mM glucose condition. However, for BPA, NP, or OP with lower estrogenic activity, the relationship between the doses and insulin secretion was an inverted U-shape. Moreover, OP, NP, or BPA (25 μg/l) impaired mitochondrial function in β-cells and induced remarkable swelling of mitochondria with loss of distinct cristae structure within the membrane, which was accompanied by disruption of mRNA expression of genes playing a key role in β-cell function (Glut2 (Slc2a2), Gck, Pdx1, Hnf1 α, Rab27a, and Snap25), and mitochondrial function (Ucp2 and Ogdh). Therefore, these phenolic estrogens can disrupt islet morphology and β-cell function, and mitochondrial dysfunction is suggested to play an important role in the impairment of β-cell function.
Search for other papers by Jian-Ting Ke in
Google Scholar
PubMed
Search for other papers by Mi Li in
Google Scholar
PubMed
Search for other papers by Shi-Qing Xu in
Google Scholar
PubMed
Search for other papers by Wen-Jian Zhang in
Google Scholar
PubMed
Search for other papers by Yong-Wei Jiang in
Google Scholar
PubMed
Search for other papers by Lan-yun Cheng in
Google Scholar
PubMed
Search for other papers by Li Chen in
Google Scholar
PubMed
Search for other papers by Jin-Ning Lou in
Google Scholar
PubMed
Search for other papers by Wei Wu in
Google Scholar
PubMed
The efficacy of gliquidone for the treatment of diabetic nephropathy was investigated by implanting micro-osmotic pumps containing gliquidone into the abdominal cavities of Goto-Kakizaki (GK) rats with diabetic nephropathy. Blood glucose, 24 h urinary protein, and 24 h urinary albumin levels were measured weekly. After 4 weeks of gliquidone therapy, pathological changes in the glomerular basement membrane (GBM) were examined using an electron microscope. Real-time PCR, western blotting, and immunohistochemistry were employed to detect glomerular expression of receptors for advanced glycation end products (RAGE) (AGER), protein kinase C β (PKCβ), and protein kinase A (PKA) as well as tubular expression of the albumin reabsorption-associated proteins: megalin and cubilin. Human proximal tubular epithelial cells (HK-2 cells) were used to analyze the effects of gliquidone and advanced glycation end products (AGEs) on the expression of megalin and cubilin and on the absorption of albumin. Gliquidone lowered blood glucose, 24 h urinary protein, and 24 h urinary albumin levels in GK rats with diabetic nephropathy. The level of plasma C-peptide increased markedly and GBM and podocyte lesions improved dramatically after gliquidone treatment. Glomerular expression of RAGE and PKCβ decreased after gliquidone treatment, while PKA expression increased. AGEs markedly suppressed the expression of megalin and cubulin and the absorption of albumin in HK-2 cells in vitro, whereas the expression of megalin and cubilin and the absorption of albumin were all increased in these cells after gliquidone treatment. In conclusion, gliquidone treatment effectively reduced urinary protein in GK rats with diabetic nephropathy by improving glomerular lesions and promoting tubular reabsorption.
Search for other papers by Min Liu in
Google Scholar
PubMed
Search for other papers by Shuo Xie in
Google Scholar
PubMed
Search for other papers by Weiwei Liu in
Google Scholar
PubMed
Search for other papers by Jingjin Li in
Google Scholar
PubMed
Search for other papers by Chao Li in
Google Scholar
PubMed
Search for other papers by Wei Huang in
Google Scholar
PubMed
Search for other papers by Hexin Li in
Google Scholar
PubMed
Search for other papers by Jinghai Song in
Google Scholar
PubMed
Search for other papers by Hong Zhang in
Google Scholar
PubMed
Obesity is a worldwide health problem. Semaphorins are involved in axonal guidance; however, the role of secretory semaphorin 3G (SEMA3G) in regulating adipocyte differentiation remains unclear. Microarray analysis showed that the SEMA3G gene was upregulated in an in vitro model of adipogenesis. In this study, SEMA3G was highly expressed in the white adipose tissue and liver. Analysis of 3T3-L1 cell and primary mouse preadipocyte differentiation showed that SEMA3G mRNA and protein levels were increased during the middle stage of cell development. In vitro experiments also showed that adipocyte differentiation was promoted by SEMA3G; however, SEMA3G inhibition using a recombinant lentiviral vector expressing a specific shRNA showed the opposite results. Mice were fed a chow or high-fat diet (HFD); knockdown of SEMA3G was found to inhibit weight gain, reduce fat mass in the tissues, prevent lipogenesis in the liver tissue, reduce insulin resistance and ameliorate glucose tolerance in HFD mice. Additionally, the effect of SEMA3G on HFD-induced obesity was activated through PI3K/Akt/GSK3β signaling in the adipose tissue and the AMPK/SREBP-1c pathway in the liver. Moreover, the plasma concentrations of SEMA3G and leptin were measured in 20 obese and 20 non-obese human subjects. Both proteins were increased in obese subjects, who also exhibited a lower level of adiponectin and presented with insulin resistance. In summary, we demonstrated that SEMA3G is an adipokine essential for adipogenesis, lipogenesis, and insulin resistance and is associated with obesity. SEMA3G inhibition may, therefore, be useful for treating diet-induced obesity and its complications.
Department of Integrated Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China
Search for other papers by Xuemei Tang in
Google Scholar
PubMed
Department of Integrated Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China
Search for other papers by Jingwen Li in
Google Scholar
PubMed
Search for other papers by Wei Xiang in
Google Scholar
PubMed
Search for other papers by Ye Cui in
Google Scholar
PubMed
Search for other papers by Bin Xie in
Google Scholar
PubMed
Search for other papers by Xiaodong Wang in
Google Scholar
PubMed
Search for other papers by Zihui Xu in
Google Scholar
PubMed
Search for other papers by Lixia Gan in
Google Scholar
PubMed
In addition to the ascertained efficacy as antidiabetic drug, metformin is increasingly being used as weight-loss agent in obesity, and as insulin sensitizer in nonalcoholic fatty liver disease (NAFLD). However, the mechanisms underlying these effects are still incompletely understood. Emerging evidence suggest metformin as leptin sensitizer to mediate the weight-loss effect in the brain. In this study, we investigated effects of metformin on expression of leptin receptors in liver and kidney in mice. C57BL/6 mice were fed with chow diet (CD) or high-fat diet (HF) for 5months. Afterward, mice were treated with metformin (50mg/kg or 200mg/kg) for 15days. Metabolic parameters and hepatic gene expression were analyzed at the end of the treatment. We also tested the effects of metformin on plasma-soluble leptin receptor (sOB-R) levels in newly diagnosed type 2 diabetes mellitus (T2DM) patients, and assessed its effect on hepatosteatosis in mice. Results showed that metformin upregulates the expression of leptin receptors (OB-Ra, -Rb, -Rc, and -Rd) in liver but not kidney. The stimulation effect is dose-dependent in both chow and HF mice. Upregulation of OB-Rb, long signaling isoform, needs a relatively higher dose of metformin. This effect was paralleled by increased sOBR levels in mice and T2DM patients, and decreased hepatic triglyceride (TG) content and lipogenic gene expression, including sterol regulatory element-binding protein 1c (SREBP-1c), fatty acid synthase (FAS) and acetyl-CoA carboxylase-1 (ACC-1). Taken together, these data identify hepatic leptin receptor as target gene being upregulated by metformin which may enhance leptin sensitivity in liver to alleviate steatosis.
Search for other papers by Akhilesh K Pandey in
Google Scholar
PubMed
Search for other papers by Wei Li in
Google Scholar
PubMed
Search for other papers by Xiangling Yin in
Google Scholar
PubMed
Search for other papers by Douglas M Stocco in
Google Scholar
PubMed
Search for other papers by Paula Grammas in
Google Scholar
PubMed
Search for other papers by XingJia Wang in
Google Scholar
PubMed
Previous studies have reported the roles of Ca2+ in steroidogenesis. The present study has investigated an inhibitory effect of Ca2+ influx through L-type Ca2+ channels on gene expression of steroidogenic acute regulatory (STAR) protein that regulates the transfer of substrate cholesterol to the inner mitochondrial membrane for steroidogenesis. Blocking Ca2+ influx through L-type Ca2+ channels using the selective Ca2+ channel blocker, nifedipine, markedly enhanced cAMP-induced STAR protein expression and progesterone production in MA-10 mouse Leydig cells. This was confirmed by utilization of different L-type Ca2+ channel blockers. Reverse transcription-PCR analyses of Star mRNA and luciferase assays of Star promoter activity indicated that blocking Ca2+ influx through L-type Ca2+ channels acted at the level of Star gene transcription. Further studies showed that blocking the Ca2+ channel enhanced Star gene transcription by depressing the expression of DAX-1 (NR0B1 as listed in the MGI Database) protein, a transcriptional repressor of Star gene expression. It was also observed that there is a synergistic interaction between nifedipine and cAMP. Normally, sub-threshold levels of cAMP are unable to induce steroidogenesis, but in the presence of the L-type Ca2+ channel blocker, they increased STAR protein and steroid hormone to the maximal levels. However, in the absence of minimal levels of cAMP, none of the L-type Ca2+ channel blockers are able to induce Star gene expression. These observations indicate that Ca2+ influx through L-type Ca2+ channels is involved in an inhibitory effect on Star gene expression. Blocking L-type Ca2+ channel attenuated the inhibition and reduced the threshold of cAMP-induced Star gene expression in Leydig cells.
Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
Search for other papers by Min Hu in
Google Scholar
PubMed
Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
Search for other papers by Yuehui Zhang in
Google Scholar
PubMed
Search for other papers by Jiaxing Feng in
Google Scholar
PubMed
Search for other papers by Xue Xu in
Google Scholar
PubMed
Search for other papers by Jiao Zhang in
Google Scholar
PubMed
Search for other papers by Wei Zhao in
Google Scholar
PubMed
Search for other papers by Xiaozhu Guo in
Google Scholar
PubMed
Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
Search for other papers by Juan Li in
Google Scholar
PubMed
Search for other papers by Edvin Vestin in
Google Scholar
PubMed
Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, Shanghai, China
Institute of Integrative Medicine of Fudan University, Shanghai, China
Search for other papers by Peng Cui in
Google Scholar
PubMed
Department of Gynecology Obstetrics and Gynecology, Hospital of Fudan University, Shanghai, China
Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
Search for other papers by Xin Li in
Google Scholar
PubMed
Search for other papers by Xiao-ke Wu in
Google Scholar
PubMed
Search for other papers by Mats Brännström in
Google Scholar
PubMed
Search for other papers by Linus R Shao in
Google Scholar
PubMed
Search for other papers by Håkan Billig in
Google Scholar
PubMed
Impaired progesterone (P4) signaling is linked to endometrial dysfunction and infertility in women with polycystic ovary syndrome (PCOS). Here, we report for the first time that elevated expression of progesterone receptor (PGR) isoforms A and B parallels increased estrogen receptor (ER) expression in PCOS-like rat uteri. The aberrant PGR-targeted gene expression in PCOS-like rats before and after implantation overlaps with dysregulated expression of Fkbp52 and Ncoa2, two genes that contribute to the development of uterine P4 resistance. In vivo and in vitro studies of the effects of metformin on the regulation of the uterine P4 signaling pathway under PCOS conditions showed that metformin directly inhibits the expression of PGR and ER along with the regulation of several genes that are targeted dependently or independently of PGR-mediated uterine implantation. Functionally, metformin treatment corrected the abnormal expression of cell-specific PGR and ER and some PGR-target genes in PCOS-like rats with implantation. Additionally, we documented how metformin contributes to the regulation of the PGR-associated MAPK/ERK/p38 signaling pathway in the PCOS-like rat uterus. Our data provide novel insights into how metformin therapy regulates uterine P4 signaling molecules under PCOS conditions.
Search for other papers by Bin Li in
Google Scholar
PubMed
Beijing Institute of Hepatology, Beijing, China
Search for other papers by Jiming Yin in
Google Scholar
PubMed
Search for other papers by Jing Chang in
Google Scholar
PubMed
Search for other papers by Jia Zhang in
Google Scholar
PubMed
Search for other papers by Yangjia Wang in
Google Scholar
PubMed
Search for other papers by Haixia Huang in
Google Scholar
PubMed
Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
Search for other papers by Wei Wang in
Google Scholar
PubMed
Search for other papers by Xiangjun Zeng in
Google Scholar
PubMed
Microcirculatory injuries had been reported to be involved in diabetic cardiomyopathy, which was mainly related to endothelial cell dysfunction. Apelin, an adipokine that is upregulated in diabetes mellitus, was reported to improve endothelial cell dysfunction and attenuate cardiac insufficiency induced by ischemia and reperfusion. Therefore, it is hypothesized that apelin might be involved in alleviating endothelial cell dysfunction and followed cardiomyopathy in diabetes mellitus. The results showed that apelin improved endothelial cell dysfunction via decreasing apoptosis and expression of adhesion molecules and increasing proliferation, angiogenesis, and expression of E-cadherin, VEGFR 2 and Tie-2 in endothelial cells, which resulted in the attenuation of the capillary permeability in cardiac tissues and following diabetic cardiomyopathy. Meanwhile, the results from endothelial cell-specific APJ knockout mice and cultured endothelial cells confirmed that the effects of apelin on endothelial cells were dependent on APJ and the downstream NFκB pathways. In conclusion, apelin might reduce microvascular dysfunction induced by diabetes mellitus via improving endothelial dysfunction dependent on APJ activated NFκB pathways.
Search for other papers by Sung Wook Park in
Google Scholar
PubMed
Search for other papers by Shawna D Persaud in
Google Scholar
PubMed
Search for other papers by Stanislas Ogokeh in
Google Scholar
PubMed
Search for other papers by Tatyana A Meyers in
Google Scholar
PubMed
Search for other papers by DeWayne Townsend in
Google Scholar
PubMed
Search for other papers by Li-Na Wei in
Google Scholar
PubMed
Excessive and/or persistent activation of calcium-calmodulin protein kinase II (CaMKII) is detrimental in acute and chronic cardiac injury. However, intrinsic regulators of CaMKII activity are poorly understood. We find that cellular retinoic acid-binding protein 1 (CRABP1) directly interacts with CaMKII and uncover a functional role for CRABP1 in regulating CaMKII activation. We generated Crabp1-null mice (CKO) in C57BL/6J background for pathophysiological studies. CKO mice develop hypertrophy as adults, exhibiting significant left ventricular dilation with reduced ejection fraction at the baseline cardiac function. Interestingly, CKO mice have elevated basal CaMKII phosphorylation at T287, and phosphorylation on its substrate phospholamban (PLN) at T17. Acute isoproterenol (ISO) challenge (80 mg/kg two doses in 1 day) causes more severe apoptosis and necrosis in CKO hearts, and treatment with a CaMKII inhibitor KN-93 protects CKO mice from this injury. Chronic (30 mg/kg/day) ISO challenge also significantly increases hypertrophy and fibrosis in CKO mice as compared to WT. In wild-type mice, CRABP1 expression is increased in early stages of ISO challenge and eventually reduces to the basal level. Mechanistically, CRABP1 directly inhibits CaMKII by competing with calmodulin (CaM) for CaMKII interaction. This study demonstrates increased susceptibility of CKO mice to ISO-induced acute and chronic cardiac injury due to, at least in part, elevated CaMKII activity. Deleting Crabp1 results in reduced baseline cardiac function and aggravated damage challenged with acute and persistent β-adrenergic stimulation. This is the first report of a physiological role of CRABP1 as an endogenous regulator of CaMKII, which protects the heart from ISO-induced damage.
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by You-Hua Xu in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
Search for other papers by Chen-Lin Gao in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Heng-Li Guo in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Wen-Qian Zhang in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
Search for other papers by Wei Huang in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Shan-Shan Tang in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Wen-Jun Gan in
Google Scholar
PubMed
Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
Search for other papers by Yong Xu in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Hua Zhou in
Google Scholar
PubMed
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
Search for other papers by Quan Zhu in
Google Scholar
PubMed
Endotoxemia has been recognized to be closely accompanied with type 2 diabetes mellitus (T2DM) and is responsible for many diabetic complications. Recent study suggests the potential role of butyrate, a short-chain fatty acid (SCFA) from microbiota metabolite, on T2DM. Gut-leak is a key event in diabetic-endotoxemia. To investigate if butyrate could ameliorate diabetic-endotoxemia, both in vivo and in vitro experiments were carried out in the present study. The effect of butyrate supplementation on blood HbA1c and inflammatory cytokines were determined in db/db mice; gut barrier integrity and expression of tight junction proteins were investigated both in vivo and in vitro. Oral butyrate administration significantly decreased blood HbA1c, inflammatory cytokines and LPS in db/db mice; inflammatory cell infiltration was reduced, and gut integrity and intercellular adhesion molecules were increased as detected by HE staining, immunohistochemistry and Western blot. By gut microbiota assay, ratio of Firmicutes:Bacteroidetes for gut microbiota was reduced by butyrate. In Caco-2 cells, butyrate significantly promoted cell proliferation, decreased inflammatory cytokines’ secretion, enhanced cell anti-oxidative stress ability and preserved the epithelial monocellular integrity, which was damaged by LPS. The present findings demonstrated that butyrate supplementation could ameliorate diabetic-endotoxemia in db/db mice via restoring composition of gut microbiota and preserving gut epithelial barrier integrity.
State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
Search for other papers by Shu-Fang Xia in
Google Scholar
PubMed
Shandong Sport Training Center, Jinan, China
Search for other papers by Xiao-Mei Duan in
Google Scholar
PubMed
Search for other papers by Xiang-Rong Cheng in
Google Scholar
PubMed
Search for other papers by Li-Mei Chen in
Google Scholar
PubMed
Search for other papers by Yan-Jun Kang in
Google Scholar
PubMed
Search for other papers by Peng Wang in
Google Scholar
PubMed
Search for other papers by Xue Tang in
Google Scholar
PubMed
Search for other papers by Yong-Hui Shi in
Google Scholar
PubMed
Search for other papers by Guo-Wei Le in
Google Scholar
PubMed
The study was designed to investigate the possible mechanisms of hepatic microRNAs (miRs) in regulating local thyroid hormone (TH) action and ultimately different propensities to high-fat diet (HFD)-induced obesity. When obesity-prone (OP) and obesity-resistant (OR) mice were fed HFD for 7 weeks, OP mice showed apparent hepatic steatosis, with significantly higher body weight and lower hepatic TH receptor b (TRb) expression and type 1 deiodinase (DIO1) activity than OR mice. Next-generation sequencing technology revealed that 13 miRs in liver were dysregulated between the two phenotypes, of which 8 miRs were predicted to target on Dio1 or TRb. When mice were fed for 17 weeks, OR mice had mild hepatic steatosis and increased Dio1 and TRb expression than OP mice, with downregulation of T3 target genes (including Srebp1c, Acc1, Scd1 and Fasn) and upregulation of Cpt1α, Atp5c1, Cox7c and Cyp7a1. A stem-loop qRT-PCR analysis confirmed that the levels of miR-383, miR-34a and miR-146b were inversely correlated with those of DIO1 or TRb. Down-regulated expression of miR-383 or miR-146b by miR-383 inhibitor (anti-miR-383) or miR-146b inhibitor (anti-miR-146b) in free fatty acid-treated primary mouse hepatocytes led to increased DIO1 and TRb expressions, respectively, and subsequently decreased cellular lipid accumulation, while miR-34a inhibitor (anti-miR-34a) transfection had on effects on TRb expression. Luciferase reporter assay illustrated that miR-146b could directly target TRb 3′untranslated region (3′UTR). These findings suggested that miR-383 and miR-146b might play critical roles in different propensities to diet-induced obesity via targeting on Dio1 and TRb, respectively.