Network


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

Hotspot


Dive into the research topics where Claudie Hecquet is active.

Publication


Featured researches published by Claudie Hecquet.


Circulation Research | 2008

Role of TRPM2 Channel in Mediating H2O2-Induced Ca2+ Entry and Endothelial Hyperpermeability

Claudie Hecquet; Gias U. Ahmmed; Stephen M. Vogel; Asrar B. Malik

Oxidative stress through the production of oxygen metabolites such as hydrogen peroxide (H2O2) increases vascular endothelial permeability. H2O2 stimulates ADP-ribose formation, which in turn opens transient receptor potential melastatin (TRPM)2 channels. Here, in endothelial cells, we demonstrate transcript and protein expression of TRPM2, a Ca2+-permeable, nonselective cation channel. We further show the importance of TRPM2 expression in signaling of increased endothelial permeability by oxidative stress. Exposure of endothelial cell monolayers to sublytic concentrations of H2O2 induced a cationic current measured by patch-clamp recording and Ca2+ entry detected by intracellular fura-2 fluorescence. H2O2 in a concentration-dependent manner also decreased trans-monolayer transendothelial electrical resistance for 3 hours (with maximal effect seen at 300 &mgr;mol/L H2O2), indicating opening of interendothelial junctions. The cationic current, Ca2+ entry, and transendothelial electrical resistance decrease elicited by H2O2 were inhibited by siRNA depleting TRPM2 or antibody blocking of TRPM2. H2O2 responses were attenuated by overexpression of the dominant-negative splice variant of TRPM2 or inhibition of ADP-ribose formation. Overexpression of the full-length TRPM2 enhanced H2O2-mediated Ca2+ entry, cationic current, and the transendothelial electrical resistance decrease. Thus, TRPM2 mediates H2O2-induced increase in endothelial permeability through the activation of Ca2+ entry via TRPM2. TRPM2 represents a novel therapeutic target directed against oxidant-induced endothelial barrier disruption.


Nature Immunology | 2012

The redox-sensitive cation channel TRPM2 modulates phagocyte ROS production and inflammation

Anke Di; Xiao Pei Gao; Feng Qian; Takeshi Kawamura; Jin Han; Claudie Hecquet; Richard D. Ye; Stephen M. Vogel; Asrar B. Malik

The NADPH oxidase activity of phagocytes and its generation of reactive oxygen species (ROS) is critical for host defense, but ROS overproduction can also lead to inflammation and tissue injury. Here we report that TRPM2, a nonselective and redox-sensitive cation channel, inhibited ROS production in phagocytic cells and prevented endotoxin-induced lung inflammation in mice. TRPM2-deficient mice challenged with endotoxin (lipopolysaccharide) had an enhanced inflammatory response and diminished survival relative to that of wild-type mice challenged with endotoxin. TRPM2 functioned by dampening NADPH oxidase–mediated ROS production through depolarization of the plasma membrane in phagocytes. As ROS also activate TRPM2, our findings establish a negative feedback mechanism for the inactivation of ROS production through inhibition of the membrane potential–sensitive NADPH oxidase.


Thrombosis and Haemostasis | 2009

Role of H2O2-activated TRPM2 calcium channel in oxidant-induced endothelial injury

Claudie Hecquet; Asrar B. Malik

The transient receptor potential (melastatin) 2 (TRPM2), is an oxidant-activated non-selective cation channel that is widely expressed in mammalian tissues including the vascular endothelium. Oxidative stress, through the generation of oxygen metabolites including H(2)O(2), stimulates intracellular ADP-ribose formation which, in turn, opens TRPM2 channels. These channels act as an endogenous redox sensor for mediating oxidative stress/ROS-induced Ca(2+) entry and the subsequent specific Ca(2+)-dependent cellular reactions such as endothelial hyperpermeability and apoptosis. This review summarizes recent findings on the mechanism by which oxidants induce TRPM2 activation, the role of these channels in the signalling vascular endothelial dysfunctions, and the modulation of oxidant-induced TRPM2 activation by PKCalpha and phospho-tyrosine phosphates L1.


Advances in Experimental Medicine and Biology | 2010

TRPM2 Channel Regulates Endothelial Barrier Function

Claudie Hecquet; Gias U. Ahmmed; Asrar B. Malik

Oxidative [Au1]stress, through the production of oxygen metabolites such as hydrogen peroxide[Au2] (H(2)O(2)), increases vascular endothelial permeability and plays a crucial role in several lung diseases. The transient receptor potential (melastatin) 2 (TRPM2) is an oxidant-sensitive, nonselective cation channel that is widely expressed in mammalian tissues, including the vascular endothelium. We have demonstrated the involvement of TRPM2 in mediating oxidant-induced calcium entry and endothelial hyperpermeability in cultured pulmonary artery endothelial cells. Here, we provide evidence that neutrophil activation-dependent increase in endothelial permeability and neutrophil extravasation requires TRPM2 in cultured endothelial cells. In addition, protein kinase Calpha (PKCalpha) that rapidly colocalizes with the short (nonconducting) TRPM2 isoform after exposure to hydrogen peroxide positively regulates calcium entry through the functional TRPM2 channel. Thus, increase in lung microvessel permeability and neutrophil sequestration depends on the activation of endothelial TRPM2 by neutrophilic oxidants and on PKCalpha regulation of TRPM2 channel activity. Manipulating TRPM2 function in the endothelium may represent a novel strategy aimed to prevent oxidative stress-related vascular dysfunction.


Circulation Research | 2014

Cooperative Interaction of trp Melastatin Channel Transient Receptor Potential (TRPM2) With Its Splice Variant TRPM2 Short Variant Is Essential for Endothelial Cell Apoptosis

Claudie Hecquet; Min Zhang; Manish Mittal; Stephen M. Vogel; Anke Di; Xiaopei Gao; Marcelo G. Bonini; Asrar B. Malik

Rationale: Oxidants generated by activated endothelial cells are known to induce apoptosis, a pathogenic feature of vascular injury and inflammation from multiple pathogeneses. The melastatin-family transient receptor potential 2 (TRPM2) channel is an oxidant-sensitive Ca2+ permeable channel implicated in mediating apoptosis; however, the mechanisms of gating of the supranormal Ca2+ influx required for initiating of apoptosis are not understood. Objective: Here, we addressed the role of TRPM2 and its interaction with the short splice variant TRPM2 short variant (TRPM2-S) in mediating the Ca2+ entry burst required for induction of endothelial cell apoptosis. Methods and Results: We observed that TRPM2-S was basally associated with TRPM2 in the endothelial plasmalemma, and this interaction functioned to suppress TRPM2-dependent Ca2+ gating constitutively. Reactive oxygen species production in endothelial cells or directly applying reactive oxygen species induced protein kinase C-&agr; activation and phosphorylation of TRPM2 at Ser 39. This in turn stimulated a large entry of Ca2+ and activated the apoptosis pathway. A similar TRPM2-dependent endothelial apoptosis mechanism was seen in intact vessels. The protein kinase C-&agr;–activated phosphoswitch opened the TRPM2 channel to allow large Ca2+ influx by releasing TRPM2-S inhibition of TRPM2, which in turn activated caspase-3 and cleaved the caspase substrate poly(ADP-ribose) polymerase. Conclusions: Here, we describe a fundamental mechanism by which activation of the trp superfamily TRPM2 channel induces apoptosis of endothelial cells. The signaling mechanism involves reactive oxygen species–induced protein kinase C-&agr; activation resulting in phosphorylation of TRPM2-S that allows enhanced TRPM2-mediated gating of Ca2+ and activation of the apoptosis program. Strategies aimed at preventing the uncoupling of TRPM2-S from TRPM2 and subsequent Ca2+ gating during oxidative stress may mitigate endothelial apoptosis and its consequences in mediating vascular injury and inflammation.


International Immunopharmacology | 2002

Kallikreins when activating bradykinin B2 receptor induce its redistribution on plasma membrane

Claudie Hecquet; Robert P. Becker; Fulong Tan; Ervin G. Erdös

The bradykinin (BK) B2 receptor (R) is directly activated by kallikreins and other serine proteases independent of BK release. Both the Galpha(i) and Galpha(q) proteins are involved, shown by the release of arachidonic acid and [Ca2+]i elevation. Site-directed mutagenesis of the receptor and the lack of heterogeneous desensitization of the human B2R by the BK and kallikrein emphasize among others the differences between activation by the proteases and the peptide. To characterize further the mechanism thereby kallikreins activate and desensitize the B2R we investigated the distribution of the human B2R tagged with the green fluorescent protein (B2-GFP(Ct)) on the plasma membrane of stably transfected Chinese hamster ovary (CHO) cells. We visualized the movement of B2-GFP(Ct) R with confocal fluorescence microscopy after activation by BK or a by serine protease. Continued exposure of the cells to BK led to B2R internalization within 15-20 min. Porcine pancreatic and human recombinant tissue kallikreins induced a rapid definite redistribution of receptors on the plasma membrane within 5 min, prior to internalization. These effects of kallikrein were blocked by the B2R antagonist HOE 140 and by the kallikrein inhibitor, aprotinin. The B2R was also activated by endoproteinases LysC and ArgC and trypsin, but these enzymes did not induce redistribution, only internalization. In control experiments, kallikrein had no effect on cells transfected to stably express the angiotensin-converting enzyme-green fluorescent protein (GFP). Thus, kallikreins when activating the BK B2R also trigger its redistribution on plasma membrane.


Circulation Research | 2017

Neutrophil Activation of Endothelial Cell-Expressed TRPM2 Mediates Transendothelial Neutrophil Migration and Vascular Injury.

Manish Mittal; Saroj Nepal; Yoshikazu Tsukasaki; Claudie Hecquet; Dheeraj Soni; Jalees Rehman; Chinnaswamy Tiruppathi; Asrar B. Malik

Rationale: TRPM2 (transient receptor potential melastatin-2) expressed in endothelial cells (ECs) is a cation channel mediating Ca2+ entry in response to intracellular generation of adenosine diphosphoribose—the TRPM2 ligand. Objective: Because polymorphonuclear neutrophils (PMN) interaction with ECs generates reactive oxygen species, we addressed the possible role of TRPM2 expressed in ECs in the mechanism of transendothelial migration of PMNs. Methods and Results: We observed defective PMN transmigration in response to lipopolysaccharide challenge in adult mice in which the EC expressed TRPM2 is conditionally deleted (Trpm2i&Dgr;EC). PMN interaction with ECs induced the entry of Ca2+ in ECs via the EC-expressed TRPM2. Prevention of generation of adenosine diphosphoribose in ECs significantly reduced Ca2+ entry in response to PMN activation of TRPM2 in ECs. PMNs isolated from gp91phox−/− mice significantly reduced Ca2+ entry in ECs via TRPM2 as compared with wild-type PMNs and failed to induce PMN transmigration. Overexpression of the adenosine diphosphoribose insensitive TRPM2 mutant channel (C1008→A) in ECs suppressed the Ca2+ entry response. Further, the forced expression of TRPM2 mutant channel (C1008→A) or silencing of poly ADP-ribose polymerase in ECs of mice prevented PMN transmigration. Conclusions: Thus, endotoxin-induced transmigration of PMNs was secondary to TRPM2-activated Ca2+ signaling and VE-cadherin phosphorylation resulting in the disassembly of adherens junctions and opening of the paracellular pathways. These results suggest blocking TRPM2 activation in ECs is a potentially important means of therapeutically modifying PMN-mediated vascular inflammation.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Novel Role of Reactive Oxygen Species–Activated trp Melastatin Channel-2 in Mediating Angiogenesis and Postischemic Neovascularization

Manish Mittal; Norifumi Urao; Claudie Hecquet; Min Zhang; Varadarajan Sudhahar; Xiao Pei Gao; Yulia Komarova; Masuko Ushio-Fukai; Asrar B. Malik

Objective— Transient receptor potential melastatin-2 (TRPM2) channel is a nonselective cation channel that mediates influx of Ca2+ and Na+ with relative permeability of PCa:PNa ≈0.6 in response to cellular oxidative stress. As angiogenesis and ischemic neovascularization are both significantly dependent on oxidant signaling, here we investigated the possible role of vascular endothelial growth factor (VEGF)–induced reactive oxygen species production in activating TRPM2-dependent Ca2+ signaling and in the mechanism of angiogenesis and ischemic neovascularization. Approach and Results— We observed that VEGF stimulation rapidly induced the association of TRPM2 and cellular Src kinase with vascular endothelial-cadherin forming a signalplex at vascular endothelial-cadherin junctions in endothelial cells. Using endothelial cells isolated from TRPM2 −/− mice or after small interfering RNA depletion of TRPM2, we demonstrated that TRPM2-activated Ca2+ signaling was required for cellular Src kinase–induced phosphorylation of vascular endothelial-cadherin at Y658 and Y731, the crucial sites involved in vascular endothelial-cadherin internalization in response to VEGF. VEGF-induced reactive oxygen species generation activated TRPM2-induced Ca2+ entry, whereas the reactive oxygen species–insensitive TRPM2 mutant (C1008→A) showed impaired Ca2+ entry. Endothelial cells depleted of TRPM2 also displayed significantly perturbed migratory phenotype and impaired activation of cellular Src in response to VEGF. TRPM2 −/− mice reconstituted with wild-type myeloid cells demonstrated aberrant angiogenesis and neovascularization in the hindlimb ischemia model as compared with wild-type mice. Conclusions— VEGF-induced angiogenesis and postischemic neovascularization in mice required reactive oxygen species generation in endothelial cells and resultant TRPM2 activation. Thus, our findings provide novel insight into the role of TRPM2 in mechanism of angiogenesis and ischemic neovascularization.


Archive | 1996

Activation of Cardiac Adenosine Triphosphate-Sensitive K + Channels: An Obligatory Pathway for the Cardioprotection Afforded by Ischemic and Pharmacological Preconditioning

Icilio Cavero; Claudie Hecquet; Yasmine Djellas; Véronique Gollot-Robert; Michel Mestre

The role of ATP-sensitive K+ channels (KATP) in protecting the myocardium from ischemic damage was explored. In the in vitro paced (2 Hz) guinea pig right-ventricular wall, which was perfused through the cannulated right coronary artery, ischemia/reperfusion injury was produced by suspending perfusion of oxygenated Tyrode solution for 30 min and then reinstalling it for 60 min. Aprikalim (AP), a K+ channel opener (0.1 μM), and 90-s ischemia (PC, preconditioning stimulus) were applied singly or together (AP + PC) before starting the 30-min ischemic period. The effects of glibenclamide (1 μM) alone or preceding AP + PC were also assessed. The parameters measured were resting (RT) and developed (DT) tension, and the duration of the action potential at 50% repolarization (APD50). In control, AP-, or PC-pretreated preparations, ischemia produced a similarly rapid decline of DT, no substantial elevation of RT, and a progressive shortening of APD50. After 60-min reflow, DT remained depressed by 55%, RT increased by 45% (in the AP group, only 18%) and APD50 fully recovered its preischemia values. Glibenclamide reduced APD50 shortening without affecting the kinetics of contractility loss during ischemia. At the end of reperfusion, glibenclamide-pretreated preparations recovered their contractility slightly less than control preparations. The application of both AP and 90-s PC did not influence the effects of ischemia but reduced significantly (by ~30%) the postischemic depression of DT, as well as the increase in RT, at the end of reperfusion. Thus, combining a concentration of AP and a duration of ischemic stress, which delivered singly do not curtail ischemia/reperfusion injury, improved substantially the recovery of contractile function. This salutary effect was negated by glibenclamide. Thus, activation of ATP-sensitive K+ channels mediates this protection independently of an acceleration of contractile failure at the onset of ischemia. In conclusion, the opening of ATP-sensitive K+ channels may constitute a natural mechanism by which the myocardium, if it is preconditioned by ischemia or by pharmacological agents, can be protected against the deleterious consequence of a prolonged ischemic insult.


Circulation Research | 2013

Cooperative Interaction of trp Melastatin Channel TRPM2 with its Splice Variant TRPM2-S is Essential for Endothelial Cell Apoptosis

Claudie Hecquet; Min Zhang; Manish Mittal; Stephen M. Vogel; Anke Di; Xiaopei Gao; Marcelo G. Bonini; Asrar B. Malik

Rationale: Oxidants generated by activated endothelial cells are known to induce apoptosis, a pathogenic feature of vascular injury and inflammation from multiple pathogeneses. The melastatin-family transient receptor potential 2 (TRPM2) channel is an oxidant-sensitive Ca2+ permeable channel implicated in mediating apoptosis; however, the mechanisms of gating of the supranormal Ca2+ influx required for initiating of apoptosis are not understood. Objective: Here, we addressed the role of TRPM2 and its interaction with the short splice variant TRPM2 short variant (TRPM2-S) in mediating the Ca2+ entry burst required for induction of endothelial cell apoptosis. Methods and Results: We observed that TRPM2-S was basally associated with TRPM2 in the endothelial plasmalemma, and this interaction functioned to suppress TRPM2-dependent Ca2+ gating constitutively. Reactive oxygen species production in endothelial cells or directly applying reactive oxygen species induced protein kinase C-&agr; activation and phosphorylation of TRPM2 at Ser 39. This in turn stimulated a large entry of Ca2+ and activated the apoptosis pathway. A similar TRPM2-dependent endothelial apoptosis mechanism was seen in intact vessels. The protein kinase C-&agr;–activated phosphoswitch opened the TRPM2 channel to allow large Ca2+ influx by releasing TRPM2-S inhibition of TRPM2, which in turn activated caspase-3 and cleaved the caspase substrate poly(ADP-ribose) polymerase. Conclusions: Here, we describe a fundamental mechanism by which activation of the trp superfamily TRPM2 channel induces apoptosis of endothelial cells. The signaling mechanism involves reactive oxygen species–induced protein kinase C-&agr; activation resulting in phosphorylation of TRPM2-S that allows enhanced TRPM2-mediated gating of Ca2+ and activation of the apoptosis program. Strategies aimed at preventing the uncoupling of TRPM2-S from TRPM2 and subsequent Ca2+ gating during oxidative stress may mitigate endothelial apoptosis and its consequences in mediating vascular injury and inflammation.

Collaboration


Dive into the Claudie Hecquet's collaboration.

Top Co-Authors

Avatar

Asrar B. Malik

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Stephen M. Vogel

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Anke Di

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Ervin G. Erdös

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Manish Mittal

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Xiaopei Gao

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Fulong Tan

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Gias U. Ahmmed

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Takeshi Kawamura

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Xiao Pei Gao

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge