Network


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

Hotspot


Dive into the research topics where Shuo Zheng is active.

Publication


Featured researches published by Shuo Zheng.


Hypertension | 2013

Gastrin and D1 Dopamine Receptor Interact to Induce Natriuresis and Diuresis

Yue Chen; Laureano D. Asico; Shuo Zheng; Van Anthony M. Villar; Duofen He; Lin Zhou; Chunyu Zeng; Pedro A. Jose

Oral NaCl produces a greater natriuresis and diuresis than the intravenous infusion of the same amount of NaCl. Gastrin is the major gastrointestinal hormone taken up by renal proximal tubule (RPT) cells. We hypothesized that renal gastrin and dopamine receptors interact to synergistically increase sodium excretion, an impaired interaction of which may be involved in the pathogenesis of hypertension. In Wistar-Kyoto rats, infusion of gastrin induced natriuresis and diuresis, which was abrogated in the presence of a gastrin (cholecystokinin B receptor [CCKBR]; CI-988) or a D1-like receptor antagonist (SCH23390). Similarly, the natriuretic and diuretic effects of fenoldopam, a D1-like receptor agonist, were blocked by SCH23390, as well as by CI-988. However, the natriuretic effects of gastrin and fenoldopam were not observed in spontaneously hypertensive rats. The gastrin/D1-like receptor interaction was also confirmed in RPT cells. In RPT cells from Wistar-Kyoto but not spontaneously hypertensive rats, stimulation of either D1-like receptor or gastrin receptor inhibited Na+-K+-ATPase activity, an effect that was blocked in the presence of SCH23390 or CI-988. In RPT cells from Wistar-Kyoto and spontaneously hypertensive rats, CCKBR and D1 receptor coimmunoprecipitated, which was increased after stimulation of either D1 receptor or CCKBR in RPT cells from Wistar-Kyoto rats; stimulation of one receptor increased the RPT cell membrane expression of the other receptor, effects that were not observed in spontaneously hypertensive rats. These data suggest that there is a synergism between CCKBR and D1-like receptors to increase sodium excretion. An aberrant interaction between the renal CCK BR and D1-like receptors (eg, D1 receptor) may play a role in the pathogenesis of hypertension.


Journal of the American Heart Association | 2016

Irisin Lowers Blood Pressure by Improvement of Endothelial Dysfunction via AMPK‐Akt‐eNOS‐NO Pathway in the Spontaneously Hypertensive Rat

Jinjuan Fu; Yu Han; Jialiang Wang; Yukai Liu; Shuo Zheng; Lin Zhou; Pedro A. Jose; Chunyu Zeng

Background Exercise is a major nonpharmacological treatment for hypertension, but its underlying mechanisms are still not completely elucidated. Irisin, a polypeptide containing 112 amino acids, which is secreted mainly by skeletal muscle cells during exercise, exerts a protective role in metabolic diseases, such as diabetes mellitus and obesity. Because of the close relationship between irisin and metabolic diseases, we hypothesized that irisin may play a role in the regulation of blood pressure. Methods and Results Blood pressures of male Wistar‐Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs) were monitored through the carotid artery. Our study found that acute intravenous injection of irisin reduced blood pressure in SHRs, but not WKY rats. Irisin, by itself, had no direct vasorelaxing effect in phenylephrine‐preconstricted mesenteric arteries from SHRs. However, irisin augmented the acetylcholine‐induced vasorelaxation in mesenteric arteries from SHRs that could be reversed by Nω‐nitro‐l‐arginine‐methyl ester (L‐NAME; 100 μmol/L), indicating a role of nitric oxide (NO) in this action. Indeed, irisin increased NO production and phosphorylation of endothelial nirtic oxide synthase (eNOS) in endothelial cells. 5′‐AMP‐activated protein kinase (AMPK) was involved in the vasorelaxing effect of irisin because compound C (20 μmol/L), an AMPK inhibitor, blocked the irisin‐mediated increase in phosphorylation of eNOS and protein kinase B (Akt) in endothelial cells and vasodilation in mesenteric arteries. Conclusions We conclude that acute administration of irisin lowers blood pressure of SHRs by amelioration of endothelial dysfunction of the mesenteric artery through the AMPK‐Akt‐eNOS‐NO signaling pathway.


Free Radical Biology and Medicine | 2014

Prenatal lipopolysaccharide exposure results in dysfunction of the renal dopamine D1 receptor in offspring.

Xinquan Wang; Hao Luo; Caiyu Chen; Ken Chen; Jialiang Wang; Yue Cai; Shuo Zheng; Xiaoli Yang; Lin Zhou; Pedro A. Jose; Chunyu Zeng

Adverse environment in early life can modulate the adult phenotype, including blood pressure. Lipopolysaccharide (LPS) exposure in utero results in increased blood pressure in the offspring, but the exact mechanisms are not clear. Studies have shown that the renal dopamine D1 receptor (D1R) plays an important role in maintaining sodium homeostasis and normal blood pressure; dysfunction of D1R is associated with oxidative stress and hypertension. In this study, we determined if dysfunction of the renal D1R is involved in fetal-programmed hypertension, and if oxidative stress contributes to this process. Pregnant Sprague-Dawley (SD) rats were intraperitoneally injected with LPS (0.79 mg/kg) or saline at gestation days 8, 10, and 12. As compared with saline-injected (control) dams, offspring of LPS-treated dams had increased blood pressure, decreased renal sodium excretion, and increased markers of oxidative stress. In addition, offspring of LPS-treated dams had decreased renal D1R expression, increased D1R phosphorylation, and G protein-coupled receptor kinase type 2 (GRK2) and type 4 (GRK4) protein expression, and impaired D1R-mediated natriuresis and diuresis. All of the findings in the offspring of LPS-treated dams were normalized after treatment with TEMPOL, an oxygen free radical scavenger. In conclusion, prenatal LPS exposure, via an increase in oxidative stress, impairs renal D1R function and leads to hypertension in the offspring. Normalization of renal D1R function by amelioration of oxidative stress may be a therapeutic target of fetal programming of hypertension.


American Journal of Physiology-renal Physiology | 2014

Regulation of renalase expression by D5 dopamine receptors in rat renal proximal tubule cells.

Shaoxiong Wang; Xi Lu; Jian Yang; Hongyong Wang; Caiyu Chen; Yu Han; Hongmei Ren; Shuo Zheng; Duofen He; Lin Zhou; Laureano D. Asico; Wei Eric Wang; Pedro A. Jose; Chunyu Zeng

The dopaminergic and sympathetic systems interact to regulate blood pressure. Our previous studies showed regulation of α1-adrenergic receptor function by D1-like dopamine receptors in vascular smooth muscle cells. Because renalase could regulate circulating epinephrine levels and dopamine production in renal proximal tubules (RPTs), we tested the hypothesis that D1-like receptors regulate renalase expression in kidney. The effect of D1-like receptor stimulation on renalase expression and function was measured in immortalized RPT cells from Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHRs). We found that the D1-like receptor agonist fenoldopam (10(-7)-10(-5) mol/l) increased renalase protein expression and function in WKY RPT cells but decreased them in SHR cells. Fenoldopam also increased renalase mRNA levels in WKY but not in SHR cells. In contrast, fenoldopam increased the degradation of renalase protein in SHR cells but not in WKY cells. The regulation of renalase by the D1-like receptor was mainly via the D5 receptor because silencing of the D5 but not D1 receptor by antisense oligonucleotides blocked the stimulatory effect of the D1-like receptor on renalase expression in WKY cells. Moreover, inhibition of PKC, by the PKC inhibitor 19-31, blocked the stimulatory effect of fenoldopam on renalase expression while stimulation of PKC, by a PKC agonist (PMA), increased renalase expression, indicating that PKC is involved in the process. Our studies suggest that the D5 receptor positively regulates renalase expression in WKY but not SHR RPT cells; aberrant regulation of renalase by the D5 receptor may be involved in the pathogenesis of hypertension.


Hypertension | 2015

Activation of D4 Dopamine Receptor Decreases Angiotensin II Type 1 Receptor Expression in Rat Renal Proximal Tubule CellsNovelty and Significance

Ken Chen; Kun Deng; Xiaoyan Wang; Zhen Wang; Shuo Zheng; Hongmei Ren; Duofen He; Yu Han; Laureano D. Asico; Pedro A. Jose; Chunyu Zeng

The dopaminergic and renin–angiotensin systems interact to regulate blood pressure. Disruption of the D4 dopamine receptor gene in mice produces hypertension that is associated with increased renal angiotensin type 1 (AT1) receptor expression. We hypothesize that the D4 receptor can inhibit AT1 receptor expression and function in renal proximal tubule cells from Wistar–Kyoto (WKY) rats, but the D4 receptor regulation of AT1 receptor is aberrant in renal proximal tubule cells from spontaneously hypertensive rats (SHRs). The D4 receptor agonist, PD168077, decreased AT1 receptor protein expression in a time- and concentration-dependent manner in WKY cells. By contrast, in SHR cells, PD168077 increased AT1 receptor protein expression. The inhibitory effect of D4 receptor on AT1 receptor expression in WKY cells was blocked by a calcium channel blocker, nicardipine, or calcium-free medium, indicating that calcium is involved in the D4 receptor–mediated signaling pathway. Angiotensin II increased Na+-K+ ATPase activity in WKY cells. Pretreatment with PD168077 decreased the stimulatory effect of angiotensin II on Na+-K+ ATPase activity in WKY cells. In SHR cells, the inhibitory effect of D4 receptor on angiotensin II–mediated stimulation of Na+-K+ ATPase activity was aberrant; pretreatment with PD168077 augmented the stimulatory effect of AT1 receptor on Na+-K+ ATPase activity in SHR cells. This was confirmed in vivo; pretreatment with PD128077 for 1 week augmented the antihypertensive and natriuretic effect of losartan in SHRs but not in WKY rats. We suggest that an aberrant interaction between D4 and AT1 receptors may play a role in the abnormal regulation of sodium excretion in hypertension.


Hypertension | 2015

Activation of D4 Dopamine Receptor Decreases Angiotensin II Type 1 Receptor Expression in Rat Renal Proximal Tubule Cells

Ken Chen; Kun Deng; Xiaoyan Wang; Zhen Wang; Shuo Zheng; Hongmei Ren; Duofen He; Yu Han; Laureano D. Asico; Pedro A. Jose; Chunyu Zeng

The dopaminergic and renin–angiotensin systems interact to regulate blood pressure. Disruption of the D4 dopamine receptor gene in mice produces hypertension that is associated with increased renal angiotensin type 1 (AT1) receptor expression. We hypothesize that the D4 receptor can inhibit AT1 receptor expression and function in renal proximal tubule cells from Wistar–Kyoto (WKY) rats, but the D4 receptor regulation of AT1 receptor is aberrant in renal proximal tubule cells from spontaneously hypertensive rats (SHRs). The D4 receptor agonist, PD168077, decreased AT1 receptor protein expression in a time- and concentration-dependent manner in WKY cells. By contrast, in SHR cells, PD168077 increased AT1 receptor protein expression. The inhibitory effect of D4 receptor on AT1 receptor expression in WKY cells was blocked by a calcium channel blocker, nicardipine, or calcium-free medium, indicating that calcium is involved in the D4 receptor–mediated signaling pathway. Angiotensin II increased Na+-K+ ATPase activity in WKY cells. Pretreatment with PD168077 decreased the stimulatory effect of angiotensin II on Na+-K+ ATPase activity in WKY cells. In SHR cells, the inhibitory effect of D4 receptor on angiotensin II–mediated stimulation of Na+-K+ ATPase activity was aberrant; pretreatment with PD168077 augmented the stimulatory effect of AT1 receptor on Na+-K+ ATPase activity in SHR cells. This was confirmed in vivo; pretreatment with PD128077 for 1 week augmented the antihypertensive and natriuretic effect of losartan in SHRs but not in WKY rats. We suggest that an aberrant interaction between D4 and AT1 receptors may play a role in the abnormal regulation of sodium excretion in hypertension.


American Journal of Hypertension | 2015

Enhanced Natriuresis and Diuresis in Wistar Rats Caused by the Costimulation of Renal Dopamine D3 and Angiotensin II Type 2 Receptors

Sufei Yang; Yu Han; Shuo Zheng; Xun Kou; Laureano D. Asico; Hefei Huang; Zhao Gao; Pedro A. Jose; Chunyu Zeng

BACKGROUND The kidney, via its regulation of sodium excretion, which is modulated by humoral factors, including the dopamine and renin-angiotensin systems, keeps the blood pressure in the normal range. We have reported a negative interaction between dopamine D3 and AT1 receptors (D3R and AT1R) in renal proximal tubule (RPT) cells. Here, we studied the interaction between D3R and AT2R in vitro and in vivo. METHODS AND RESULTS Stimulation of either the D3R or AT2R, by the intrarenal arterial infusion of PD128907, a D3R agonist, or CGP42112A, an AT2R agonist, induced natriuresis and diuresis that were enhanced by the simultaneous infusion of PD128907 and CGP42112A in Wistar rats. The D3/AT2 receptor interaction was confirmed in in vitro, i.e., stimulation of either the D3R or AT2R inhibited Na(+)-K(+)-ATPase activity that was enhanced by the costimulation of these receptors. D3R and AT2R colocalized and coimmunoprecipitated in kidney and RPT cells (RPTCs). Stimulation of one receptor increased the localization of the other receptor at the plasma cell membrane. ERK1/2-MAPK is involved in the signaling pathway of D3R and AT2R interaction because costimulation of D3R and AT2R significantly increased ERK1/2-MAPK expression in RPTCs; inhibition of ERK1/2-MAPK abolished the inhibition of Na(+)-K(+)-ATPase activity that was enhanced by D3R and AT2R costimulation. CONCLUSIONS Our current study indicates that D3R, in combination with AT2R, enhances natriuresis and diuresis, via ERK1/2-MAPK pathway, that may be involved in the regulation of blood pressure.


Clinical and Experimental Hypertension | 2014

Dopamine d1-like receptors suppress proliferation of vascular smooth muscle cell induced by insulin-like growth factor-1

Xun Kou; Yu Han; Di Yang; Yukai Liu; Jinjuan Fu; Shuo Zheng; Duofen He; Lin Zhou; Chunyu Zeng

Abstract Objective: Proliferation of vascular smooth muscle cells (VSMCs) participates in the pathogenesis and development of cardiovascular diseases, including essential hypertension and atherosclerosis. Our previous study found that stimulation of D1-like dopamine receptors inhibited insulin-induced proliferation of VSMCs. Insulin-like growth factor-1 (IGF-1) and insulin share similar structure and biological effect. However, whether or not there is any effect of D1-like receptors on IGF-1-induced proliferation of VSMCs is not known. Therefore, we investigated the inhibitory effect of D1-like dopamine receptors on the IGF-1-induced VSMCs proliferation in this study. Method: VSMC proliferation was determined by [3H]-thymidine incorporation, the uptake of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and cell number. Phosphorylated/non-phosphorylated IGF-1 receptor, Akt, mTOR and p70S6K expressions were determined by immunoblotting. The oligodeoxynucleotides were transfected to A10 cells to identify the effect of D1 and D5 receptors, respectively. Results: IGF-1 increased the proliferation of VSMCs, while in the presence of fenoldopam, IGF-1-mediated stimulatory effect was reduced. Use of either antisense for D1 or D5 receptor partially inhibited the fenoldopam-induced antiproliferation effect of VSMCs. Use of both D1 and D5 receptor antisenses completely blocked the inhibitory effect of fenoldopam. In the presence of PI3k and mTOR inhibitors, the IGF-1-mediated proliferation of VSMCs was blocked. Moreover, IGF-1 increased the phosphorylation of PI3k and mTOR. The inhibitory effect of fenoldopam on VSMC proliferation might be due to the inhibition of IGF-1 receptor expression and IGF-1 phosphorylation, because in the presence of fenoldopam, the stimulatory effect of IGF-1 on phosphorylation of IGF-1 receptor, PI3k and mTOR is reduced, the IGF-1 receptor expression was reduced in A10 cells. Conclusion: Activation of the D1-like receptors suppressed the proliferative effect of IGF-1 in A10 cells via the inhibition of the IGF-1R/Akt/mTOR/p70S6K pathway and downregulated the expression of IGF-1 receptor.


Hypertension Research | 2017

Activation of angiotensin II type 1 receptors increases D4 dopamine receptor expression in rat renal proximal tubule cells

Luxun Tang; Shuo Zheng; Hongmei Ren; Duofen He; Chunyu Zeng; Wei Eric Wang

Both the dopaminergic and renin–angiotensin systems play important roles in the regulation of blood pressure. Our previous study showed that the stimulation of dopaminergic D4 receptors reduced angiotensin II type 1 (AT1) receptor expression in renal proximal tubule (RPT) cells. In this study, we tested whether AT1 receptors, in return, would regulate D4 receptor expression and function in RPT cells. Expression of the D4 receptor from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) RPT cells and renal cortex tissues were determined by western blot, and Na+–K+ ATPase activity was determined using an enzyme assay. Urine volume and urine sodium of WKY rats and SHRs treated with or without D4 receptor stimulation were measured. Thus, activation of AT1 receptors with angiotensin II (Ang II) increased D4 receptor protein expression in RPT cells, and this increase was blocked by nicardipine, a calcium influx blocker. The D4 receptor agonist PD168077 inhibited Na+–K+ ATPase activity in WKY RPT cells but not in SHR RPT cells. Ang II pre-treatment promoted D4 receptor-mediated inhibition of Na+–K+ ATPase in RPT cells in WKY rats but not in SHRs. Meanwhile, Ang II pre-treatment augmented the natriuretic effect of PD168077 in WKY rats but not in SHRs. In conclusion, AT1 stimulation can regulate the expression and natriuretic function of dopaminergic D4 receptors in RPT cells and might be involved in the pathogenesis of essential hypertension.


Journal of the American Heart Association | 2018

Long‐Term Exposure of Fine Particulate Matter Causes Hypertension by Impaired Renal D1 Receptor–Mediated Sodium Excretion via Upregulation of G‐Protein–Coupled Receptor Kinase Type 4 Expression in Sprague‐Dawley Rats

Xi Lu; Zhengmeng Ye; Shuo Zheng; Hongmei Ren; Jing Zeng; Xinquan Wang; Pedro A. Jose; Ken Chen; Chunyu Zeng

Background Epidemiological evidence supports an important association between air pollution exposure and hypertension. However, the mechanisms are not clear. Methods and Results Our present study found that long‐term exposure to fine particulate matter (PM2.5) causes hypertension and impairs renal sodium excretion, which might be ascribed to lower D1 receptor expression and higher D1 receptor phosphorylation, accompanied with a higher G‐protein–coupled receptor kinase type 4 (GRK4) expression. The in vivo results were confirmed in in vitro studies (ie, PM 2.5 increased basal and decreased D1 receptor mediated inhibitory effect on Na+‐K+ ATPase activity, decreased D1 receptor expression, and increased D1 receptor phosphorylation in renal proximal tubule cells). The downregulation of D1 receptor expression and function might be attributable to a higher GRK4 expression after the exposure of renal proximal tubule cells to PM 2.5, because downregulation of GRK4 by small‐interfering RNA reversed the D1 receptor expression and function. Because of the role of reactive oxygen species on D1 receptor dysfunction and its relationship with air pollution exposure, we determined plasma reactive oxygen species and found the levels higher in PM 2.5‐treated Sprague‐Dawley rats. Inhibition of reactive oxygen species by tempol (4‐hydroxy‐2,2,6,6‐tetramethylpiperidin‐1‐oxyl) reduced blood pressure and increased sodium excretion in PM 2.5‐treated Sprague‐Dawley rats, accompanied by an increase in the low D1 receptor expression, and decreased the hyperphosphorylated D1 receptor and GRK4 expression. Conclusions Our present study indicated that long‐term exposure of PM 2.5 increases blood pressure by decreasing D1 receptor expression and function; reactive oxygen species, via regulation of GRK4 expression, plays an important role in the pathogenesis of PM 2.5‐induced hypertension.

Collaboration


Dive into the Shuo Zheng's collaboration.

Top Co-Authors

Avatar

Chunyu Zeng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Pedro A. Jose

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Duofen He

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Lin Zhou

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongmei Ren

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu Han

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caiyu Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ken Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yue Chen

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge