Network


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

Hotspot


Dive into the research topics where Zhiming Zhu is active.

Publication


Featured researches published by Zhiming Zhu.


Cell Metabolism | 2010

Activation of TRPV1 by Dietary Capsaicin Improves Endothelium-Dependent Vasorelaxation and Prevents Hypertension

Dachun Yang; Zhidan Luo; Shuangtao Ma; Wing Tak Wong; Liqun Ma; Jian Zhong; Hongbo He; Zhigang Zhao; Tingbing Cao; Zhencheng Yan; Daoyan Liu; William J. Arendshorst; Yu Huang; Martin Tepel; Zhiming Zhu

Some plant-based diets lower the cardiometabolic risks and prevalence of hypertension. New evidence implies a role for the transient receptor potential vanilloid 1 (TRPV1) cation channel in the pathogenesis of cardiometabolic diseases. Little is known about impact of chronic TRPV1 activation on the regulation of vascular function and blood pressure. Here we report that chronic TRPV1 activation by dietary capsaicin increases the phosphorylation of protein kinase A (PKA) and eNOS and thus production of nitric oxide (NO) in endothelial cells, which is calcium dependent. TRPV1 activation by capsaicin enhances endothelium-dependent relaxation in wild-type mice, an effect absent in TRPV1-deficient mice. Long-term stimulation of TRPV1 can activate PKA, which contributes to increased eNOS phosphorylation, improves vasorelaxation, and lowers blood pressure in genetically hypertensive rats. We conclude that TRPV1 activation by dietary capsaicin improves endothelial function. TRPV1-mediated increase in NO production may represent a promising target for therapeutic intervention of hypertension.


Journal of Molecular Cell Biology | 2012

Activation of the cold-sensing TRPM8 channel triggers UCP1-dependent thermogenesis and prevents obesity

Shuangtao Ma; Hao Yu; Zhigang Zhao; Zhidan Luo; Jing Chen; Yinxing Ni; Rongbing Jin; Liqun Ma; Peijian Wang; Zhenyu Zhu; Li Li; Jian Zhong; Daoyan Liu; Bernd Nilius; Zhiming Zhu

Brown adipose tissue (BAT) is an energy-expending organ that produces heat. Expansion or activation of BAT prevents obesity and diabetes. Chronic cold exposure enhances thermogenesis in BAT through uncoupling protein 1 (UCP1) activation triggered via a β-adrenergic pathway. Here, we report that the cold-sensing transient receptor potential melastatin 8 (TRPM8) is functionally present in mouse BAT. Challenging brown adipocytes with menthol, a TRPM8 agonist, up-regulates UCP1 expression and requires protein kinase A activation. Upon mimicking long-term cold exposure with chronic dietary menthol application, menthol significantly increased the core temperatures and locomotor activity in wild-type mice; these effects were absent in both TRPM8(-/-) and UCP1(-/-) mice. Dietary obesity and glucose abnormalities were also prevented by menthol treatment. Our results reveal a previously unrecognized role for TRPM8, suggesting that stimulation of this channel mediates BAT thermogenesis, which could constitute a promising way to treat obesity.


Hypertension | 2012

Dipeptidyl Peptidase 4 Inhibitor Sitagliptin Protects Endothelial Function in Hypertension Through a Glucagon–Like Peptide 1–Dependent Mechanism

Limei Liu; Jian Liu; Wing Tak Wong; Xiao Yu Tian; Chi Wai Lau; Yi-Xiang J. Wang; Gang Xu; Yunfei Pu; Zhiming Zhu; Aimin Xu; Karen S.L. Lam; Zhen-Yu Chen; Chi Fai Ng; Xiaoqiang Yao; Yu Huang

Sitagliptin, a selective dipeptidyl peptidase 4 inhibitor, inhibits the inactivation and degradation of glucagon like peptide 1 (GLP-1), which is used for the treatment of type 2 diabetes mellitus. However, little is known about the role of GLP-1 in hypertension. This study investigated whether the activation of GLP-1 signaling protects endothelial function in hypertension. Two-week sitagliptin treatment (10 mg/kg per day, oral gavage) improved endothelium-dependent relaxation in renal arteries, restored renal blood flow, and reduced systolic blood pressure in spontaneously hypertensive rats. In vivo sitagliptin treatment elevated GLP-1 and GLP-1 receptor expressions, increased cAMP level, and subsequently activated protein kinase A, liver kinase B1, AMP-activated protein kinase-&agr; and endothelial NO synthase in spontaneously hypertensive rat renal arteries. Inhibition of GLP-1 receptor, adenylyl cyclase, protein kinase A, AMP-activated protein kinase-&agr;, or NO synthase reversed the protective effects of sitagliptin. We also demonstrate that GLP-1 receptor agonist exendin 4 in vitro treatment had similar vasoprotective effects in spontaneously hypertensive rat renal arteries and increased NO production in spontaneously hypertensive rat aortic endothelial cells. Studies using transient expressions of wild-type and dominant-negative AMP-activated protein kinase-&agr;2 support the critical role of AMP-activated protein kinase-&agr; in mediating the effect of GLP-1 in endothelial cells. Ex vivo exendin 4 treatment also improved endothelial function of renal arteries from hypertensive patients. Our results elucidate that upregulation of GLP-1 and related agents improve endothelial function in hypertension by restoring NO bioavailability, suggesting that GLP-1 signaling could be a therapeutic target in hypertension-related vascular events.


Diabetes, Obesity and Metabolism | 2013

Blood pressure-lowering effects of GLP-1 receptor agonists exenatide and liraglutide: a meta-analysis of clinical trials

Bin Wang; Jian Zhong; H. Lin; Zhigang Zhao; Zhencheng Yan; Hongbo He; Yinxing Ni; Daoyan Liu; Zhiming Zhu

Aside from lowering blood glucose, glucagon‐like peptide‐1 receptor agonists (GLP‐1 RAs) attract much attention because of their cardioprotective effects. The aim of this study was to assess the blood pressure‐lowering effects of the GLP‐1 RAs exenatide and liraglutide compared with other common drugs used to treat type 2 diabetes (T2DM) based on randomized controlled trials (RCTs) including data describing complete blood pressure (BP) changes from baseline.


Hypertension | 2009

Increased Transient Receptor Potential Canonical Type 3 Channels in Vasculature From Hypertensive Rats

Daoyan Liu; Dachun Yang; Hongbo He; Xiaoping Chen; Tingbing Cao; Xiaoli Feng; Liqun Ma; Zhidan Luo; Lijuan Wang; Zhencheng Yan; Zhiming Zhu; Martin Tepel

We tested the hypothesis that transient receptor potential canonical type 3 (TRPC3) channels are increased in vascular smooth muscle cells and aortic tissue from spontaneously hypertensive rats (SHR) compared with normotensive Wistar Kyoto rats. Expression of TRPC3 was analyzed by immunohistochemistry and Western blotting. TRPC3 gene knockdown was performed by specific small interfering RNA and TRPC3 overexpression using the pAdEasy-1 system. Cytosolic calcium was measured using fluorescence spectrophotometry and vasoconstriction of aortic rings using a force transducer. In SHR, the expression of TRPC3 channel protein was significantly higher in aortic rings (1.48±0.05 versus 1.00±0.06; each n=6; P<0.01) and vascular smooth muscle cells (1.28±0.08 versus 1.00±0.03; each n=6; P<0.05) compared with Wistar Kyoto rats. Knockdown of TRPC3 gene expression by specific small interfering RNA significantly reduced the angiotensin II–induced calcium influx by 30±3% (n=6; P<0.01), whereas TRPC3 overexpression significantly increased it by 55±3% (n=6; P<0.01). The angiotensin II–induced calcium increase was significantly enhanced in vascular smooth muscle cells from SHR compared with Wistar Kyoto rats, even in the presence of the calcium channel blocker amlodipine. Angiotensin II significantly elevated the TRPC3 channel protein expression in vascular smooth muscle cells from SHR from 1.28±0.08 to 1.61±0.08 (each n=6; P<0.01). Angiotensin II–induced TRPC3 expression was prevented by telmisartan. Administration of telmisartan to SHR for 4 weeks significantly reduced blood pressure, angiotensin II–induced vasoconstriction, and TRPC3 channel protein expression in aortic tissue. TRPC3 expression was not significantly reduced after reduction of blood pressure in SHR using amlodipine. In conclusion, we give experimental evidence that increased TRPC3 channel protein expression in the vasculature is important for elevated blood pressure.


Hypertension | 2010

Telmisartan Prevents Weight Gain and Obesity Through Activation of Peroxisome Proliferator-Activated Receptor-δ–Dependent Pathways

Hongbo He; Dachun Yang; Liqun Ma; Zhidan Luo; Shuangtao Ma; Xiaoli Feng; Tingbing Cao; Zhencheng Yan; Daoyan Liu; Martin Tepel; Zhiming Zhu

Telmisartan shows antihypertensive and several pleiotropic effects that interact with metabolic pathways. In the present study we tested the hypothesis that telmisartan prevents adipogenesis in vitro and weight gain in vivo through activation of peroxisome proliferator-activated receptor (PPAR)-&dgr;-dependent pathways in several tissues. In vitro, telmisartan significantly upregulated PPAR-&dgr; expression in 3T3-L1 preadipocytes in a time- and dose-dependent manner. Other than enhancing PPAR-&dgr; expression by 68.2±17.3% and PPAR-&dgr; activity by 102.0±9.0%, telmisartan also upregulated PPAR-&ggr; expression, whereas neither candesartan nor losartan affected PPAR-&dgr; expression. In vivo, long-term administration of telmisartan significantly reduced visceral fat and prevented high-fat diet-induced obesity in wild-type mice and hypertensive rats but not in PPAR-&dgr; knockout mice. Administration of telmisartan did not influence food intake in mice. Telmisartan influenced several lipolytic and energy uncoupling related proteins (UCPs) and enhanced phosphorylated protein kinase A and hormone sensitive lipase but reduced perilipin expression and finally inhibited adipogenesis in 3T3-L1 preadipocytes. Telmisartan-associated reduction of adipogenesis in preadipocytes was significantly blocked after PPAR-&dgr; gene knockout. Chronic telmisartan treatment upregulated the expressions of protein kinase A, hormone-sensitive lipase, and uncoupling protein 1 but reduced perilipin expression in adipose tissue and increased uncoupling protein 2 and 3 expression in skeletal muscle in wild-type mice but not in PPAR-&dgr; knockout mice. We conclude that telmisartan prevents adipogenesis and weight gain through activation of PPAR-&dgr;-dependent lipolytic pathways and energy uncoupling in several tissues.


Cell Research | 2012

TRPV1 activation improves exercise endurance and energy metabolism through PGC-1α upregulation in mice

Zhidan Luo; Liqun Ma; Zhigang Zhao; Hongbo He; Dachun Yang; Xiaoli Feng; Shuangtao Ma; Xiaoping Chen; Tianqi Zhu; Tingbing Cao; Daoyan Liu; Bernd Nilius; Yu Huang; Zhencheng Yan; Zhiming Zhu

Impaired aerobic exercise capacity and skeletal muscle dysfunction are associated with cardiometabolic diseases. Acute administration of capsaicin enhances exercise endurance in rodents, but the long-term effect of dietary capsaicin is unknown. The capsaicin receptor, the transient receptor potential vanilloid 1 (TRPV1) cation channel has been detected in skeletal muscle, the role of which remains unclear. Here we report the function of TRPV1 in cultured C2C12 myocytes and the effect of TRPV1 activation by dietary capsaicin on energy metabolism and exercise endurance of skeletal muscles in mice. In vitro, capsaicin increased cytosolic free calcium and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) expression in C2C12 myotubes through activating TRPV1. In vivo, PGC-1α in skeletal muscle was upregulated by capsaicin-induced TRPV1 activation or genetic overexpression of TRPV1 in mice. TRPV1 activation increased the expression of genes involved in fatty acid oxidation and mitochondrial respiration, promoted mitochondrial biogenesis, increased oxidative fibers, enhanced exercise endurance and prevented high-fat diet-induced metabolic disorders. Importantly, these effects of capsaicin were absent in TRPV1-deficient mice. We conclude that TRPV1 activation by dietary capsaicin improves energy metabolism and exercise endurance by upregulating PGC-1α in skeletal muscles. The present results indicate a novel therapeutic strategy for managing metabolic diseases and improving exercise endurance.


Hypertension Research | 2010

Perivascular fat-mediated vascular dysfunction and remodeling through the AMPK/mTOR pathway in high-fat diet-induced obese rats

Liqun Ma; Shuangtao Ma; Hongbo He; Dachun Yang; Xiaoping Chen; Zhidan Luo; Daoyan Liu; Zhiming Zhu

Perivascular adipose tissue (PVAT) is implicated in the regulation of vascular function in the physiological state, but the modulatory effect of PVAT on vasculature during obesity is poorly understood. Endothelial nitric oxide synthase (eNOS), AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) participate in the regulation of vascular function. We therefore investigated whether PVAT has a potential role through the AMPK/mTOR pathway in obesity-related vascular dysfunction. Wistar rats on a high-fat diet (HFD) for 6 months had higher periaortic fat mass compared with rats on a chow diet (3.31±0.56 vs. 2.34±0.28 g, P<0.05). Obesity-related impairment of endothelium-dependent relaxation of the aorta was markedly attenuated by temporary periaortic fat removal whereas obesity-related enhancement of contractile performance was unaffected. Rats on an HFD had thicker aortic tunica medias (180.06±7.56 vs. 128.14±13.21 μm for rats on a chow diet, P<0.01) and larger periaortic adipocytes than rats on a chow diet (1209.00±62.65 vs. 447.20±21.31 μm2, respectively, P<0.01). Furthermore, mesenteric arterial rings incubated with periaortic fat from rats on an HFD demonstrated lower endothelium-dependent relaxation. This effect was absent in mesenteric arterial rings incubated with periaortic fat from rats on a chow diet. Moreover, an HFD led to a downregulation of AMPK and eNOS in the aorta with a concurrent upregulation of mTOR. In a parallel in vitro study, culturing vascular smooth muscle cells with periaortic adipocytes from rats on an HFD reduced the AMPK phosphorylation and increased mTOR phosphorylation, and the latter one was blocked by the incubation of compound C. We conclude that PVAT likely impacts obesity-related vascular dysfunction and remodeling through impairment of eNOS-mediated vasodilatation and the AMPK/mTOR pathway.


Journal of Hypertension | 2006

Transient receptor potential channels in essential hypertension.

Daoyan Liu; Alexandra Scholze; Zhiming Zhu; Katharina Krueger; Florian Thilo; Antje Burkert; Katrin Streffer; Stefan Holz; Christian Harteneck; Walter Zidek; Martin Tepel

Objective The role of nonselective cation channels of the transient receptor potential channel (TRPC) family in essential hypertension has not yet been investigated. Methods We studied TRPCs in 51 patients with essential hypertension and 51 age-matched and sex-matched normotensive control subjects. Calcium and gadolinium influx into human monocytes was determined using the fluorescent dye technique. TRPC expression was measured using reverse transcriptase-polymerase chain reaction and in-cell western assay. Gene silencing by small interfering RNA for specific TRPC knockdown was also performed. Results We observed an increased gadolinium/calcium-influx ratio through TRPC in essential hypertensive patients compared with normotensive control subjects [cation influx ratio (mean ± SEM), 125 ± 14 versus 80 ± 7%; each n = 51; P < 0.01], due to an increase of gadolinium influx in hypertensive patients compared with normotensive control subjects (48 ± 4 versus 36 ± 3%; each n = 51; P < 0.05). We observed a significant increase of TRPC3 and TRPC5 protein expression in essential hypertensive patients compared with normotensive control subjects (normalized TRPC3 expression, 3.21 ± 0.59 versus 1.36 ± 0.07; each n = 20; P < 0.01; normalized TRPC5 expression, 2.10 ± 0.28 versus 1.40 ± 0.52; each n = 12; P < 0.05). We used small interfering RNA for knockdown of TRPC5. The thereby reduced channel expression caused a significant attenuation of calcium and gadolinium influx. Conclusion This study points to an important role of TRPCs in essential hypertension.


Cardiovascular Research | 2011

Activation of TRPV1 reduces vascular lipid accumulation and attenuates atherosclerosis

Liqun Ma; Jian Zhong; Zhigang Zhao; Zhidan Luo; Shuangtao Ma; Jing Sun; Hongbo He; Tianqi Zhu; Daoyan Liu; Zhiming Zhu; Martin Tepel

AIMS Activation of transient receptor potential vanilloid type-1 (TRPV1) channels may affect lipid storage and the cellular inflammatory response. Now, we tested the hypothesis that activation of TRPV1 channels attenuates atherosclerosis in apolipoprotein E knockout mice (ApoE(-/-)) but not ApoE(-/-)TRPV1(-/-) double knockout mice on a high-fat diet. METHODS AND RESULTS Both TRPV1 mRNA and protein expression were identified in vascular smooth muscle cells (VSMC) and in aorta from C57BL/6J mice using RT-PCR, immunoblotting, and immunohistochemistry. In vitro, activation of TRPV1 by the specific agonists capsaicin and resiniferatoxin dose-dependently increased cytosolic calcium and significantly reduced the accumulation of lipids in VSMC from C57BL/6J mice but not from TRPV1(-/-) mice. TRPV1 activation increased ATP-binding cassette transporter A1 (ABCA1) expression and reduced low-density lipoprotein-related protein 1 (LRP1) expression in VSMC by calcium-dependent and calcineurin- and protein kinase A-dependent mechanisms. These results showed increased cellular cholesterol efflux and reduced cholesterol uptake. In vivo, long-term activation of TRPV1 by capsaicin for 24 weeks increased ABCA1 and reduced LRP1 expression in aorta from ApoE(-/-) mice on a high-fat diet. Long-term activation of TRPV1 significantly reduced lipid storage and atherosclerotic lesions in the aortic sinus and in the thoracoabdominal aorta from ApoE(-/-) mice but not from ApoE(-/-)TRPV1(-/-) mice on a high-fat diet. These findings indicated that TRPV1 activation ameliorates high-fat diet-induced atherosclerosis. CONCLUSION Activation of TRPV1 may be a novel therapeutic tool to attenuate atherosclerosis caused by a high-fat diet.

Collaboration


Dive into the Zhiming Zhu's collaboration.

Top Co-Authors

Avatar

Daoyan Liu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhigang Zhao

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhencheng Yan

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongbo He

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhidan Luo

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jian Zhong

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Liqun Ma

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiang Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yinxing Ni

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge