Network


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

Hotspot


Dive into the research topics where Zongmin Zhou is active.

Publication


Featured researches published by Zongmin Zhou.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Hydrogen sulfide is an endogenous stimulator of angiogenesis

Andreas Papapetropoulos; Anastasia Pyriochou; Zaid Altaany; Guangdong Yang; Antonia Marazioti; Zongmin Zhou; Mark G. Jeschke; Ludwik K. Branski; David N. Herndon; Rui Wang; Csaba Szabó

The goal of the current study was to investigate the role of exogenous and endogenous hydrogen sulfide (H2S) on neovascularization and wound healing in vitro and in vivo. Incubation of endothelial cells (ECs) with H2S enhanced their angiogenic potential, evidenced by accelerated cell growth, migration, and capillary morphogenesis on Matrigel. Treatment of chicken chorioallantoic membranes (CAMS) with H2S increased vascular length. Exposure of ECs to H2S resulted in increased phosphorylation of Akt, ERK, and p38. The KATP channel blocker glibenclamide or the p38 inhibitor SB203580 abolished H2S-induced EC motility. Since glibenclamide inhibited H2S-triggered p38 phosphorylation, we propose that KATP channels lay upstream of p38 in this process. When CAMs were treated with H2S biosynthesis inhibitors dl-propylargylglycine or beta-cyano-L-alanine, a reduction in vessel length and branching was observed, indicating that H2S serves as an endogenous stimulator of the angiogenic response. Stimulation of ECs with vascular endothelial growth factor (VEGF) increased H2S release, while pharmacological inhibition of H2S production or KATP channels or silencing of cystathionine gamma-lyase (CSE) attenuated VEGF signaling and migration of ECs. These results implicate endothelial H2S synthesis in the pro-angiogenic action of VEGF. Aortic rings isolated from CSE knockout mice exhibited markedly reduced microvessel formation in response to VEGF when compared to wild-type littermates. Finally, in vivo, topical administration of H2S enhanced wound healing in a rat model, while wound healing was delayed in CSE−/− mice. We conclude that endogenous and exogenous H2S stimulates EC-related angiogenic properties through a KATP channel/MAPK pathway.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Hydrogen Sulfide Is an Endogenous Inhibitor of Phosphodiesterase Activity

Mariarosaria Bucci; Andreas Papapetropoulos; Valentina Vellecco; Zongmin Zhou; Anastasia Pyriochou; Charis Roussos; Fiorentina Roviezzo; Vincenzo Brancaleone; Giuseppe Cirino

Objective—Recent studies have demonstrated that hydrogen sulfide (H2S) is produced within the vessel wall from l-cysteine regulating several aspects of vascular homeostasis. H2S generated from cystathione &ggr;-lyase (CSE) contributes to vascular tone; however, the molecular mechanisms underlying the vasorelaxing effects of H2S are still under investigation. Methods and Results—Using isolated aortic rings, we observed that addition of l-cysteine led to a concentration-dependent relaxation that was prevented by the CSE inhibitors dl-propargylglyicine (PAG) and &bgr;-cyano-l-alanine (BCA). Moreover, incubation with PAG or BCA resulted in a rightward shift in sodium nitroprusside-and isoproterenol-induced relaxation. Aortic tissues exposed to PAG or BCA contained lower levels of cGMP, exposure of cells to exogenous H2S or overexpression of CSE raised cGMP concentration. RNA silencing of CSE expression reduced intracellular cGMP levels confirming a positive role for endogenous H2S on cGMP accumulation. The ability of H2S to enhance cGMP levels was greatly reduced by the nonselective phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine. Finally, addition of H2S to a cell-free system inhibited both cGMP and cAMP breakdown. Conclusion—These findings provide direct evidence that H2S acts as an endogenous inhibitor of phosphodiesterase activity and reinforce the notion that this gasotransmitter could be therapeutically exploited.


British Journal of Pharmacology | 2013

Selectivity of commonly used pharmacological inhibitors for cystathionine β synthase (CBS) and cystathionine γ lyase (CSE)

Antonia Asimakopoulou; Panagiotis Panopoulos; Christos T. Chasapis; Ciro Coletta; Zongmin Zhou; Giuseppe Cirino; Athanassios Giannis; Csaba Szabó; Georgios A. Spyroulias; Andreas Papapetropoulos

Hydrogen sulfide (H2S) is a signalling molecule that belongs to the gasotransmitter family. Two major sources for endogenous enzymatic production of H2S are cystathionine β synthase (CBS) and cystathionine γ lyase (CSE). In the present study, we examined the selectivity of commonly used pharmacological inhibitors of H2S biosynthesis towards CSE and CBS.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Hydrogen sulfide replacement therapy protects the vascular endothelium in hyperglycemia by preserving mitochondrial function

Kunihiro Suzuki; Gabor Olah; Katalin Módis; Ciro Coletta; Gabriella Kulp; Domokos Gero; Petra Szoleczky; Tuanjie Chang; Zongmin Zhou; Lingyun Wu; Rui Wang; Andreas Papapetropoulos; Csaba Szabó

The goal of the present studies was to investigate the role of changes in hydrogen sulfide (H2S) homeostasis in the pathogenesis of hyperglycemic endothelial dysfunction. Exposure of bEnd3 microvascular endothelial cells to elevated extracellular glucose (in vitro “hyperglycemia”) induced the mitochondrial formation of reactive oxygen species (ROS), which resulted in an increased consumption of endogenous and exogenous H2S. Replacement of H2S or overexpression of the H2S-producing enzyme cystathionine-γ-lyase (CSE) attenuated the hyperglycemia-induced enhancement of ROS formation, attenuated nuclear DNA injury, reduced the activation of the nuclear enzyme poly(ADP-ribose) polymerase, and improved cellular viability. In vitro hyperglycemia resulted in a switch from oxidative phosphorylation to glycolysis, an effect that was partially corrected by H2S supplementation. Exposure of isolated vascular rings to high glucose in vitro induced an impairment of endothelium-dependent relaxations, which was prevented by CSE overexpression or H2S supplementation. siRNA silencing of CSE exacerbated ROS production in hyperglycemic endothelial cells. Vascular rings from CSE−/− mice exhibited an accelerated impairment of endothelium-dependent relaxations in response to in vitro hyperglycemia, compared with wild-type controls. Streptozotocin-induced diabetes in rats resulted in a decrease in the circulating level of H2S; replacement of H2S protected from the development of endothelial dysfunction ex vivo. In conclusion, endogenously produced H2S protects against the development of hyperglycemia-induced endothelial dysfunction. We hypothesize that, in hyperglycemic endothelial cells, mitochondrial ROS production and increased H2S catabolism form a positive feed-forward cycle. H2S replacement protects against these alterations, resulting in reduced ROS formation, improved endothelial metabolic state, and maintenance of normal endothelial function.


British Journal of Pharmacology | 2003

Angiopoietin‐1 inhibits endothelial permeability, neutrophil adherence and IL‐8 production

Lara Pizurki; Zongmin Zhou; Konstantinos Glynos; Charis Roussos; Andreas Papapetropoulos

Angiopoietin‐1 (Ang1) is an angiogenic growth factor that binds to the Tie2 receptor on vascular endothelium, promoting blood vessel maturation and integrity. In the present study, we have investigated whether Ang1 also possesses anti‐inflammatory properties by determining its effects on endothelial barrier function, neutrophil (PMN) adherence to endothelial cells (EC) and production of the PMN chemotactic factor interleukin‐8 (IL‐8). Pretreatment of endothelial monolayers with Ang1 attenuated the permeability increase induced by thrombin in both lung microvascular cells and a human endothelial cell line. Similarly, Ang1 prevented the permeability‐inducing effects of platelet‐activating factor, bradykinin and histamine. Pretreatment of EC with Ang1 also reduced the adherence of PMN to EC stimulated by thrombin. In contrast to its ability to counteract the increase in monolayer permeability brought about by various inflammatory agents, Ang1 did not affect the ability of histamine, PAF, or tumor necrosis factor‐α to stimulate PMN adherence to EC. In addition to its ability to inhibit PMN adherence, Ang1 diminished IL‐8 production from EC challenged with thrombin in a concentration‐dependent manner. When EC were preincubated with the specific Rho kinase (ROCK) inhibitor Y‐27632, we observed a reduction in PMN adherence in response to thrombin, as well as a decrease in thrombin‐stimulated IL‐8 production. Coincubation of monolayers with Y‐27632 and Ang1 did not further attenuate the above‐mentioned responses. However, Ang‐1 failed to inhibit the activation of RhoA in response to thrombin, suggesting that inhibition of EC adhesiveness for PMN and IL‐8 production by Ang1 does not result from reduced ROCK activation. We conclude that Ang1 can counteract several aspects of the inflammatory response, including endothelial permeability, PMN adherence to EC as well as inhibition of IL‐8 production by EC.


PLOS ONE | 2012

cGMP-dependent protein kinase contributes to hydrogen sulfide-stimulated vasorelaxation.

Mariarosaria Bucci; Andreas Papapetropoulos; Valentina Vellecco; Zongmin Zhou; Altaany Zaid; Panagiotis Giannogonas; Anna Cantalupo; Sandeep Dhayade; Katia P. Karalis; Rui Wang; Robert Feil; Giuseppe Cirino

A growing body of evidence suggests that hydrogen sulfide (H2S) is a signaling molecule in mammalian cells. In the cardiovascular system, H2S enhances vasodilation and angiogenesis. H2S-induced vasodilation is hypothesized to occur through ATP-sensitive potassium channels (KATP); however, we recently demonstrated that it also increases cGMP levels in tissues. Herein, we studied the involvement of cGMP-dependent protein kinase-I in H2S-induced vasorelaxation. The effect of H2S on vessel tone was studied in phenylephrine-contracted aortic rings with or without endothelium. cGMP levels were determined in cultured cells or isolated vessel by enzyme immunoassay. Pretreatment of aortic rings with sildenafil attenuated NaHS-induced relaxation, confirming previous findings that H2S is a phosphodiesterase inhibitor. In addition, vascular tissue levels of cGMP in cystathionine gamma lyase knockouts were lower than those in wild-type control mice. Treatment of aortic rings with NaHS, a fast releasing H2S donor, enhanced phosphorylation of vasodilator-stimulated phosphoprotein in a time-dependent manner, suggesting that cGMP-dependent protein kinase (PKG) is activated after exposure to H2S. Incubation of aortic rings with a PKG-I inhibitor (DT-2) attenuated NaHS-stimulated relaxation. Interestingly, vasodilatory responses to a slowly releasing H2S donor (GYY 4137) were unaffected by DT-2, suggesting that this donor dilates mouse aorta through PKG-independent pathways. Dilatory responses to NaHS and L-cysteine (a substrate for H2S production) were reduced in vessels of PKG-I knockout mice (PKG-I−/−). Moreover, glibenclamide inhibited NaHS-induced vasorelaxation in vessels from wild-type animals, but not PKG-I−/−, suggesting that there is a cross-talk between KATP and PKG. Our results confirm the role of cGMP in the vascular responses to NaHS and demonstrate that genetic deletion of PKG-I attenuates NaHS and L-cysteine-stimulated vasodilation.


Journal of the National Cancer Institute | 2008

A Central Role for Tumor-derived Monocyte Chemoattractant Protein-1 in Malignant Pleural Effusion

Georgios T. Stathopoulos; Ioannis Psallidas; Ardiana Moustaki; Charalampos Moschos; Androniki Kollintza; Sophia P. Karabela; Ilias Porfyridis; Spyridoula Vassiliou; Marilena Karatza; Zongmin Zhou; Myungsoo Joo; Timothy S. Blackwell; Charis Roussos; Daniel Graf; Ioannis Kalomenidis

BACKGROUND Tumor cells in malignant pleural effusions (MPEs) are an important source of monocyte chemoattractant protein (MCP)-1. However, the role of tumor-derived MCP-1 in the pathogenesis and progression of MPE has not been determined. METHODS B16 mouse skin melanoma cells, which are deficient in MCP-1 expression, and mouse Lewis lung cancer (LLC) cells, which express high levels of MCP-1, were engineered to stably express MCP-1 and short hairpin RNAs (shRNAs) targeting the MCP-1 transcript, respectively. Cells were injected into the pleural cavities of syngeneic immunocompetent mice, and MPE volume and pleural tumors were quantified at necropsy (day 14). MCP-1 and other mediators were determined by cytometric bead array and enzyme-linked immunosorbent assay, and mononuclear and endothelial cells were identified by immunolabeling of F4/80 and factor VIII-related antigen respectively. Mouse survival was assessed using Kaplan-Meier analysis. Vascular permeability in mice with MPE was assessed using albumin-binding Evans blue. Statistical tests were two-sided. RESULTS LLC cells expressing shRNA against MCP-1 elaborated less than 5% of the MCP-1 level in cells expressing nonspecific shRNA (control cells), and intrapleural delivery of these cells resulted in less MPE (mean MPE volume = 86 and 585 muL, respectively; difference = 499 muL; 95% confidence interval [CI] = 331 to 669 muL; P < .001), reduced MCP-1 levels in the pleural fluid, and lower mortality than when control cells were delivered. Overexpression of MCP-1 in intrapleurally injected B16 melanoma cells led to increased MPE and reduced survival. In mice with MPE, MCP-1 was a potent inducer of vascular permeability, mononuclear recruitment, and, in pleural tumors, of angiogenesis. CONCLUSION MCP-1 produced by tumor cells is an important determinant of their capacity to induce the formation of MPE and may be a useful target for the treatment of malignant pleural disease.


Journal of Pharmacology and Experimental Therapeutics | 2006

Soluble Guanylyl Cyclase Activation Promotes Angiogenesis

Anastasia Pyriochou; Dimitris Beis; Vasiliki Koika; Christos Potytarchou; Evangelia Papadimitriou; Zongmin Zhou; Andreas Papapetropoulos

Soluble guanylyl cyclase (sGC) is a cGMP-generating enzyme carrying a heme prosthetic group that functions as a nitric oxide (NO) sensor. sGC is present in most cells types, including the vascular endothelium, where its biological functions remain largely unexplored. Herein, we have investigated the role of sGC in angiogenesis and angiogenesis-related properties of endothelial cells (EC). Initially, we determined that sGC was present and enzymatically active in the chicken chorioallantoic membrane (CAM) during the days of maximal angiogenesis. In the CAM, inhibition of endogenous sGC inhibited neovascularization, whereas activation promoted neovessel formation. Using zebrafish as a model for vascular development, we did not detect any effect on vasculogenesis upon sGC blockade, but we did observe an abnormal angiogenic response involving the cranial and intersegmental vessels, as well as the posterior cardinal vein. In vitro, pharmacological activation of sGC or adenovirus-mediated sGC gene transfer promoted EC proliferation and migration, whereas sGC inhibition blocked tube-like network formation. In addition, sGC inhibition blocked the migratory response to vascular EC growth factor. Cells infected with sGC-expressing adenoviruses exhibited increased extracellular signal-regulated kinase 1/2 and p38 MAPK activation that was sensitive to sGC inhibition by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, suggesting that these mitogen-activated protein kinases are downstream effectors of sGC in EC. A functional role for p38 in cGMP-stimulated migration was demonstrated using SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole]; pharmacological inhibition of p38 attenuated BAY 41-2272 [5-cyclopropyl-2-[1-(2-fluoro-benzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]-pyrimidin-4-ylamine] and sGC overexpression-induced EC mobilization. We conclude that sGC activation promotes the expression of angiogenesis-related properties by EC and that sGC might represent a novel target to modulate neovessel formation.


Journal of Cellular Physiology | 2007

The phosphodiesterase 5 inhibitor sildenafil stimulates angiogenesis through a protein kinase G/MAPK pathway

Anastasia Pyriochou; Zongmin Zhou; Vasiliki Koika; Christos Petrou; Paul Cordopatis; William C. Sessa; Andreas Papapetropoulos

cGMP‐degrading pathways have received little attention in the context of angiogenesis. In the present study we set out to determine whether cGMP‐specific phosphodiesterase 5 (PDE5) inhibition affects new blood vessel growth. Incubation of chicken chorioallantoic membranes (CAMs) in vivo with sildenafil increased vascular length in a dose‐dependent manner. Moreover, incubation of cultured endothelial cells (ECs) with the PDE5 inhibitor promoted proliferation, migration, and organization into tube‐like structures. The effects of sildenafil on the angiogenesis‐related properties of EC could be blocked by pre‐treatment with the soluble guanylyl cyclase (sGC) inhibitor ODQ or the protein kinase G (PKG) I inhibitor DT‐3. In addition, over‐expression of sGC in EC led to an enhanced growth and migratory response to sildenafil. To study the signaling pathways implicated in the sildenafil‐stimulated angiogenic responses we determined the phosphorylation status of mitogen‐activated protein kinase (MAPK) members. Incubation of cells with sildenafil increased both extracellular signal regulated kinase 1/2 (ERK1/2) and p38 phosphorylation in a time‐dependent manner. Inhibition of MEK by PD98059 and p38 with SB203580 blocked sildenafil‐induced proliferation and migration, respectively, suggesting that these MAPK members are downstream of PDE5 and mediate the angiogenic effects of sildenafil. PDE5 inhibitors could, thus, be used in disease states where neo‐vessel growth is desired. J. Cell. Physiol. 211: 197–204, 2007.


British Journal of Pharmacology | 2009

Regulation of the expression of soluble guanylyl cyclase by reactive oxygen species

Christina Gerassimou; Anastasia Kotanidou; Zongmin Zhou; D D C Simoes; Charis Roussos; Andreas Papapetropoulos

British Journal of Pharmacology (2007) 150, 1092. doi:10.1038/sj.bjp.0707256

Collaboration


Dive into the Zongmin Zhou's collaboration.

Top Co-Authors

Avatar

Andreas Papapetropoulos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Charis Roussos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Theodoros P. Vassilakopoulos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Anastasia Kotanidou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Christina Magkou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Csaba Szabó

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

David Fulton

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppe Cirino

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge