Network


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

Hotspot


Dive into the research topics where Bing Shen is active.

Publication


Featured researches published by Bing Shen.


Molecular Nutrition & Food Research | 2015

Increased TRPP2 expression in vascular smooth muscle cells from high-salt intake hypertensive rats: The crucial role in vascular dysfunction

Ren Zhao; Muyao Zhou; Jie Li; Xia Wang; Kunli Su; Juncheng Hu; Yong Ye; Jinhang Zhu; Gongliang Zhang; Kai Wang; Juan Du; Liecheng Wang; Bing Shen

SCOPE High-salt intake is a major risk factor in the development of hypertension. The underlying mechanism of high sodium on the cardiovascular system has received extensive attention. TRPP2 (Polycystin-2) is a Ca(2+) permeable nonselective cation channel that mediates Ca(2+) mobilization to control vascular smooth muscle cells (VSMCs) contraction. Here, we investigated TRPP2 expression change in VSMCs from high-salt intake hypertensive rats and role of TRPP2 in the development of high-salt diet-induced hypertension. METHODS AND RESULTS After 4 ws of dietary treatment, systolic blood pressure was significantly elevated in high-salt intake rats (132 ± 3 mmHg) compared with regular diet control rats (104 ± 2 mmHg). Results from vessel tension and diameter measurements show that high-salt intake potentiated phenylephrine-induced contraction in denuded mesenteric artery and thoracic aorta. Immunoblot and immunofluorescence data indicate that TRPP2 expression in VSMCs from mesenteric artery and thoracic aorta was significantly increased in high-salt intake-induced hypertensive rats. However, agonist-induced contractions in denuded mesenteric artery and thoracic aorta were markedly decreased if TRPP2 was knocked down by specific shRNA. CONCLUSION Our data demonstrate that high-salt intake increased TRPP2 expression in VSMCs, which in turn potentiated blood vessel response to contractors; this may participate in the development of hypertension.


Cellular Physiology and Biochemistry | 2016

TRPP2 Enhances Metastasis by Regulating Epithelial-Mesenchymal Transition in Laryngeal Squamous Cell Carcinoma

Kaile Wu; Bing Shen; Feifei Jiang; Lin Xia; Taotao Fan; Maolin Qin; Lianqiang Yang; Jizheng Guo; Yifan Li; Min Zhu; Juan Du; Yehai Liu

Background/Aim: Surgery and chemotherapy treatments of human laryngeal squamous cell carcinoma (HLSCC) may fail due to metastasis, in which epithelial-mesenchymal transition (EMT) plays an important role. TRPP2, a nonselective cation channel, is expressed in various cell types and participates in many biological processes. Here, we show that TRPP2 enhanced metastasis by regulating EMT. Methods: We used immunohistochemistry, western blotting, Ca2+ imaging, transwell and wound healing assays to investigate TRPP2 expression levels in HLSCC tissue, and the role of TRPP2 in invasion and metastasis of a human laryngocarcinoma cell line (Hep2 cell). Results: We found that TRPP2 protein expression levels were significantly increased in HLSCC tissue; higher TRPP2 levels were associated with decreased patient survival time and degree of differentiation and advanced clinical stage. Knockdown of TRPP2 by transfection with TRPP2 siRNA markedly suppressed ATP-induced Ca2+ release, wound healing, and cell invasion in Hep2 cells. Moreover, TRPP2 siRNA significantly decreased vimentin expression but increased E-cadherin expression in Hep2 cells. In the EMT signalling pathway, TRPP2 siRNA significantly decreased Smad4, STAT3, SNAIL, SLUG and TWIST expression in Hep2 cells. Conclusion: We revealed a previously unknown function of TRPP2 in cancer development and a TRPP2-dependent mechanism underlying laryngocarcinoma cell invasion and metastasis. Our results suggest that TRPP2 may be used as a biomarker for evaluating patient prognosis and as a novel therapeutic target in HLSCC.


Scientific Reports | 2016

Increasing TRPV4 expression restores flow-induced dilation impaired in mesenteric arteries with aging

Juan Du; Xia Wang; Jie Li; Jizheng Guo; Limei Liu; Dejun Yan; Yunyun Yang; Zhongwen Li; Jinhang Zhu; Bing Shen

The flow-stimulated intracellular Ca2+ concentration ([Ca2+]i) rise in endothelial cells is an important early event leading to flow-induced blood vessel dilation. Transient receptor potential vanilloid subtype 4 (TRPV4), a Ca2+-permeable cation channel, facilitates the flow-stimulated [Ca2+]i rise. To determine whether TRPV4 is involved in age-related flow-induced blood vessel dilation impairment, we measured blood vessel diameter and nitric oxide (NO) levels and performed Ca2+ imaging, immunoblotting, and immunostaining assays in rats. We found that the flow-induced and TRPV4 activator 4α-PDD-induced dilation of mesenteric arteries from aged rats were significantly decreased compared with those from young rats. The flow- or 4α-PDD-induced [Ca2+]i rise was also markedly reduced in primary cultured mesenteric artery endothelial cells (MAECs) from aged rats. Immunoblotting and immunostaining results showed an age-related decrease of TRPV4 expression levels in MAECs. Additionally, the 4α-PDD-induced NO production was significantly reduced in aged MAECs. Compared with lentiviral GFP-treated aged rats, lentiviral vector delivery of TRPV4 increased TRPV4 expression level in aged MAECs and restored the flow- and 4α-PDD-induced vessel dilation in aged mesenteric arteries. We concluded that impaired TRPV4-mediated Ca2+ signaling causes endothelial dysfunction and that TRPV4 is a potential target for clinical treatment of age-related vascular system diseases.


Journal of Biological Chemistry | 2015

Organelle-specific subunit interactions of the vertebrate two-pore channel family.

Oluseye A. Ogunbayo; Yingmin Zhu; Bing Shen; Ejaife Agbani; Jie Li; Jianjie Ma; Michael X. Zhu; A. Mark Evans

Background: The role of two-pore channels (TPCs) in endolysosomal signaling remains controversial. Results: TPCs are targeted to different subpopulations of endolysosomes, and this determines subunit interaction and Ca2+ signaling by NAADP. Conclusion: All vertebrate TPC subtypes support Ca2+ signaling; lysosome-targeted, but not endosome-targeted, TPCs permit endoplasmic reticulum coupling. Significance: Organellar targeting and interaction of TPCs are likely critical to endolysosomal signaling in health and disease. The organellar targeting of two-pore channels (TPCs) and their capacity to associate as homo- and heterodimers may be critical to endolysosomal signaling. A more detailed understanding of the functional association of vertebrate TPC1–3 is therefore necessary. We report here that when stably expressed in HEK293 cells, human (h) TPC1 and chicken (c) TPC3 were specifically targeted to different subpopulations of endosomes, hTPC2 was specifically targeted to lysosomes, and rabbit (r) TPC3 was specifically targeted to both endosomes and lysosomes. Intracellular dialysis of NAADP evoked a Ca2+ transient in HEK293 cells that stably overexpressed hTPC1, hTPC2, and rTPC3, but not in cells that stably expressed cTPC3. The Ca2+ transients induced in cells that overexpressed endosome-targeted hTPC1 were abolished upon depletion of acidic Ca2+ stores by bafilomycin A1, but remained unaffected following depletion of endoplasmic reticulum stores by thapsigargin. In contrast, Ca2+ transients induced via lysosome-targeted hTPC2 and endolysosome-targeted rTPC3 were abolished by bafilomycin A1 and markedly attenuated by thapsigargin. NAADP induced marked Ca2+ transients in HEK293 cells that stably coexpressed hTPC2 with hTPC1 or cTPC3, but failed to evoke any such response in cells that coexpressed interacting hTPC2 and rTPC3 subunits. We therefore conclude that 1) all three TPC subtypes may support Ca2+ signaling from their designate acidic stores, and 2) lysosome-targeted (but not endosome-targeted) TPCs support coupling to the endoplasmic reticulum.


Acta Pharmacologica Sinica | 2016

The functions of TRPP2 in the vascular system.

Juan Du; Jie Fu; Xian-Ming Xia; Bing Shen

TRPP2 (polycystin-2, PC2 or PKD2), encoded by the PKD2 gene, is a non-selective cation channel with a large single channel conductance and high Ca2+ permeability. In cell membrane, TRPP2, along with polycystin-1, TRPV4 and TRPC1, functions as a mechanotransduction channel. In the endoplasmic reticulum, TRPP2 modulates intracellular Ca2+ release associated with IP3 receptors and the ryanodine receptors. Noteworthily, TRPP2 is widely expressed in vascular endothelial and smooth muscle cells of all major vascular beds, and contributes to the regulation of vessel function. The mutation of the PKD2 gene is a major cause of autosomal dominant polycystic kidney disease (ADPKD), which is not only a common genetic disease of the kidney but also a systemic disorder associated with abnormalities in the vasculature; cardiovascular complications are the leading cause of mortality and morbidity in ADPKD patients. This review provides an overview of the current knowledge regarding the TRPP2 protein and its possible role in cardiovascular function and related diseases.


Science China-life Sciences | 2015

Canonical transient receptor potential 4 and its small molecule modulators.

Jie Fu; Zhaobing Gao; Bing Shen; Michael X. Zhu

Canonical transient receptor potential 4 (TRPC4) forms non-selective cation channels that contribute to phospholipase C-dependent Ca2+ entry into cells following stimulation of G protein coupled receptors and receptor tyrosine kinases. Moreover, the channels are regulated by pertussis toxin-sensitive Gi/o proteins, lipids, and various other signaling mechanisms. TRPC4-containing channels participate in the regulation of a variety of physiological functions, including excitability of both gastrointestinal smooth muscles and brain neurons. This review is to present recent advances in the understanding of physiology and development of small molecular modulators of TRPC4 channels.


Journal of Cardiovascular Pharmacology | 2015

Contrasting Patterns of Agonist-induced Store-operated Ca2+ Entry and Vasoconstriction in Mesenteric Arteries and Aorta With Aging.

Yong Yang; Jinhang Zhu; Xia Wang; Nairui Xue; Juan Du; Xiangling Meng; Bing Shen

Abstract: Ca2+ is a crucial factor in the regulation of smooth muscle contraction. Store-operated Ca2+ entry (SOCE) is one pathway that mediates Ca2+ influx and smooth muscle contraction. Vessel contraction function usually alters with aging to cause severe vascular-related diseases. However, the underlying mechanism is still not fully understood. Here, we assessed intracellular Ca2+ and vessel tension and found that SOCE and SOCE-mediated contraction of vascular smooth muscle cells (VSMCs) was reduced in aorta but increased in mesenteric arteries from aged rats. The results of Western blot and immunofluorescence staining show that the expression levels of Orai1, a store-operated Ca2+ channel, were increased in VSMCs of mesenteric arteries but were reduced in VSMCs of aorta with aging. In conclusion, we demonstrated that the changing pattern of SOCE and SOCE-mediated contraction of VSMCs is completely reversed in mesenteric arteries and aorta with aging, providing a potential therapeutic target for clinical treatment in age-related vascular diseases.


Journal of Medicinal Chemistry | 2017

Pyrazolopyrimidines as Potent Stimulators for Transient Receptor Potential Canonical 3/6/7 Channels

Chunrong Qu; Mingmin Ding; Yingmin Zhu; Yungang Lu; Juan Du; Melissa Miller; Jinbin Tian; Jinmei Zhu; Jian Xu; Meng Wen; A. G.A. Er-Bu; Jule Wang; Yuling Xiao; Meng Wu; Owen B. McManus; Min Li; Jilin Wu; Huai-Rong Luo; Zhengyu Cao; Bing Shen; Hongbo Wang; Michael X. Zhu; Xuechuan Hong

Transient receptor potential canonical 3/6/7 (TRPC3/6/7) are highly homologous receptor-operated nonselective cation channels. Despite their physiological significance, very few selective and potent agonists are available for functional examination of these channels. Using a cell-based high throughput screening approach, a lead compound with the pyrazolopyrimidine skeleton was identified as a TRPC6 agonist. Synthetic schemes for the lead and its analogues were established, and structural-activity relationship studies were carried out. A series of potent and direct agonists of TRPC3/6/7 channels were identified, and among them, 4m-4p have a potency order of TRPC3 > C7 > C6, with 4n being the most potent with an EC50 of <20 nM on TRPC3. Importantly, these compounds exhibited no stimulatory activity on related TRP channels. The potent and selective compounds described here should be suitable for evaluation of the roles of TRPC channels in the physiology and pathogenesis of diseases, including glomerulosclerosis and cancer.


Physiological Reports | 2016

Orai1 forms a signal complex with BKCa channel in mesenteric artery smooth muscle cells

Meihua Chen; Jie Li; Feifei Jiang; Jie Fu; Xian-Ming Xia; Juan Du; Min Hu; Junhao Huang; Bing Shen

Orai1, a specific nonvoltage‐gated Ca2+ channel, has been found to be one of key molecules involved in store‐operated Ca2+ entry (SOCE). Orai1 may associate with other proteins to form a signaling complex, which is essential for regulating a variety of physiological functions. In this study, we studied the possible interaction between Orai1 and large conductance Ca2+‐activated potassium channel (BKCa). Using RNA interference technique, we demonstrated that the SOCE and its associated membrane hyperpolarization were markedly suppressed after knockdown of Orai1 with a specific Orai1 siRNA in rat mesenteric artery smooth muscle. Moreover, isometric tension measurements showed that agonist‐induced vasocontraction was increased after Orai1 was knocked down or the tissue was incubated with BKCa blocker iberiotoxin. Coimmunoprecipitation data revealed that BKCa and Orai1 could reciprocally pull down each other. In situ proximity ligation assay further demonstrated that Orai1 and BKCa are in close proximity. Taken together, these results indicate that Orai1 physically associates with BKCa to form a signaling complex in the rat mesenteric artery smooth muscle. Ca2+ influx via Orai1 stimulates BKCa, leading to membrane hyperpolarization. This hyperpolarizing effect of Orai1‐BKCa coupling could contribute to reduce agonist‐induced membrane depolarization, therefore preventing excessive contraction of the rat mesenteric artery smooth muscle in response to contractile agonists.


Cellular Physiology and Biochemistry | 2015

Role of Na+-K+-2Cl- Cotransporter 1 in Phenylephrine-Induced Rhythmic Contraction in the Mouse Aorta: Regulation of Na+-K+-2Cl- Cotransporter 1 by Ca2+ Sparks and KCa Channels

Bing Shen; Jie Fu; Jizheng Guo; Jie Zhang; Xia Wang; Xiang Pan; Meihua Chen; Yifan Zhou; Min Zhu; Juan Du

Background/Aims: Vasoconstrictor-induced rhythmic contraction of arteries or veins has been observed both in vivo and in vitro. Many studies have reported that gap junctions, ryanodine receptors, Na+, K+-ATPase and other factors are involved in vasoconstrictor-induced rhythmic contraction in vascular smooth muscle. However, the mechanism is still not completely understood. Methods: We used vessel tension measurements, intracellular recordings and intracellular Cl- concentration ([Cl-]i) measurements to investigate the mechanism underlying phenylephrine (PE)-induced rhythmic contraction in the mouse aorta. Results: We found that Na+-K+-2Cl- cotransporter 1 (NKCC1) inhibitor bumetanide abolished PE-induced rhythmic contraction. The Cl- channel blockers DIDS and niflumic acid initially augmented the amplitude of PE-induced rhythmic contraction but later inhibited the rhythmic contraction. The large Ca2+-activated K+ channel blocker TEA and iberiotoxin increased the amplitude of PE-induced rhythmic contraction. The voltage-dependent Ca2+ channel blocker, nifedipine, and a Ca2+-free solution abolished PE-induced rhythmic contraction. The inhibitor of ryanodine receptors in the sarcoplasmic reticulum, ryanodine, inhibited PE-induced rhythmic contraction. Moreover, bumetanide hyperpolarized the membrane potential of vascular smooth muscle cells in a resting state or after PE pre-treatment. Bumetanide, niflumic acid, ryanodine, iberiotoxin, nifedipine and Ca2+-free buffer significantly suppressed the PE-induced [Cl-]i increase. Conclusion: These data indicate that NKCC1 is involved in the formation of PE-induced rhythmic contraction, and we also provide a method with which to indirectly observe the NKCC1 activity in isolated intact mouse thoracic aortas.

Collaboration


Dive into the Bing Shen's collaboration.

Top Co-Authors

Avatar

Juan Du

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Jie Fu

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Jie Li

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinhang Zhu

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Jizheng Guo

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Xia Wang

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Feifei Jiang

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Kaile Wu

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Meihua Chen

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Xian-Ming Xia

Anhui Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge