Network


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

Hotspot


Dive into the research topics where Jean-Claude Honoré is active.

Publication


Featured researches published by Jean-Claude Honoré.


Blood | 2011

Ischemic neurons prevent vascular regeneration of neural tissue by secreting semaphorin 3A

Jean-Sebastien Joyal; Nicholas Sitaras; François Binet; José Carlos Rivera; Andreas Stahl; Karine Zaniolo; Zhuo Shao; Anna Polosa; Tang Zhu; David Hamel; Mikheil Djavari; Dario Kunik; Jean-Claude Honoré; Emilie Picard; Alexandra Zabeida; Daya R. Varma; Gilles R.X. Hickson; Joseph A. Mancini; Michael Klagsbrun; Santiago Costantino; Christian M. Beauséjour; Pierre Lachapelle; Lois E. H. Smith; Sylvain Chemtob; Przemyslaw Sapieha

The failure of blood vessels to revascularize ischemic neural tissue represents a significant challenge for vascular biology. Examples include proliferative retinopathies (PRs) such as retinopathy of prematurity and proliferative diabetic retinopathy, which are the leading causes of blindness in children and working-age adults. PRs are characterized by initial microvascular degeneration, followed by a compensatory albeit pathologic hypervascularization mounted by the hypoxic retina attempting to reinstate metabolic equilibrium. Paradoxically, this secondary revascularization fails to grow into the most ischemic regions of the retina. Instead, the new vessels are misdirected toward the vitreous, suggesting that vasorepulsive forces operate in the avascular hypoxic retina. In the present study, we demonstrate that the neuronal guidance cue semaphorin 3A (Sema3A) is secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine IL-1β. Sema3A contributes to vascular decay and later forms a chemical barrier that repels neo-vessels toward the vitreous. Conversely, silencing Sema3A expression enhances normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function. Overcoming the chemical barrier (Sema3A) released by ischemic neurons accelerates the vascular regeneration of neural tissues, which restores metabolic supply and improves retinal function. Our findings may be applicable to other neurovascular ischemic conditions such as stroke.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Microglia and Interleukin-1β in Ischemic Retinopathy Elicit Microvascular Degeneration Through Neuronal Semaphorin-3A

José Carlos Rivera; Nicholas Sitaras; Baraa Noueihed; David Hamel; Ankush Madaan; Tianwei (Ellen) Zhou; Jean-Claude Honoré; Christiane Quiniou; Jean-Sebastien Joyal; Pierre Hardy; Florian Sennlaub; William D. Lubell; Sylvain Chemtob

Objective—Proinflammatory cytokines contribute to the development of retinal vasculopathies. However, the role of these factors and the mechanisms by which they elicit their effects in retina are not known. We investigated whether activated microglia during early stages of ischemic retinopathy produces excessive interleukin-1&bgr; (IL-1&bgr;), which elicits retinal microvascular degeneration not directly but rather by triggering the release of the proapoptotic/repulsive factor semaphorin-3A (Sema3A) from neurons. Approach and Results—Sprague Dawley rats subjected to retinopathy induced by hyperoxia (80% O2; O2-induced retinopathy) exhibited retinal vaso-obliteration associated with microglial activation, NLRP3 upregulation, and IL-1&bgr; and Sema3A release; IL-1&bgr; was mostly generated by microglia. Intraperitoneal administration of IL-1 receptor antagonists (Kineret, or rytvela [101.10]) decreased these effects and enhanced retinal revascularization; knockdown of Sema3A resulted in microvessel preservation and, conversely, administration of IL-1&bgr; caused vaso-obliteration. In vitro, IL-1&bgr; derived from activated primary microglial cells, cultured under hyperoxia, stimulated the release of Sema3A in retinal ganglion cells-5, which in turn induced apoptosis of microvascular endothelium; antagonism of IL-1 receptor decreased microglial activation and on retinal ganglion cells-5 abolished the release of Sema3A inhibiting ensuing endothelial cell apoptosis. IL-1&bgr; was not directly cytotoxic to endothelial cells. Conclusions—Our findings suggest that in the early stages of O2-induced retinopathy, retinal microglia are activated to produce IL-1&bgr;, which sustains the activation of microglia and induces microvascular injury through the release of Sema3A from adjacent neurons. Interference with IL-1 receptor or Sema3A actions preserves the microvascular bed in ischemic retinopathies and, consequently, decreases ensued pathological preretinal neovascularization.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

G-Protein–Coupled Receptor 91 and Succinate Are Key Contributors in Neonatal Postcerebral Hypoxia-Ischemia Recovery

David Hamel; Melanie Sanchez; François Duhamel; Olivier Roy; Jean-Claude Honoré; Baraa Noueihed; Tianwei (Ellen) Zhou; Mathieu Nadeau-Vallée; Xin Hou; Jean-Claude Lavoie; Grant A. Mitchell; Orval Mamer; Sylvain Chemtob

Objective—Prompt post–hypoxia-ischemia (HI) revascularization has been suggested to improve outcome in adults and newborn subjects. Other than hypoxia-inducible factor, sensors of metabolic demand remain largely unknown. During HI, anaerobic respiration is arrested resulting in accumulation of carbohydrate metabolic intermediates. As such succinate readily increases, exerting its biological effects via a specific receptor, G-protein–coupled receptor (GPR) 91. We postulate that succinate/GPR91 enhances post-HI vascularization and reduces infarct size in a model of newborn HI brain injury. Approach and Results—The Rice–Vannucci model of neonatal HI was used. Succinate was measured by mass spectrometry, and microvascular density was evaluated by quantification of lectin-stained cryosection. Gene expression was evaluated by real-time polymerase chain reaction. Succinate levels rapidly increased in the penumbral region of brain infarcts. GPR91 was foremost localized not only in neurons but also in astrocytes. Microvascular density increased at 96 hours after injury in wild-type animals; it was diminished in GPR91-null mice leading to an increased infarct size. Stimulation with succinate led to an increase in growth factors implicated in angiogenesis only in wild-type mice. To explain the mode of action of succinate/GPR91, we investigated the role of prostaglandin E2–prostaglandin E receptor 4, previously proposed in neural angiogenesis. Succinate-induced vascular endothelial growth factor expression was abrogated by a cyclooxygenase inhibitor and a selective prostaglandin E receptor 4 antagonist. This antagonist also abolished succinate-induced neovascularization. Conclusions—We uncover a dominant metabolic sensor responsible for post-HI neurovascular adaptation, notably succinate/GPR91, acting via prostaglandin E2–prostaglandin E receptor 4 to govern expression of major angiogenic factors. We propose that pharmacological intervention targeting GPR91 could improve post-HI brain recovery.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Fatty acid receptor Gpr40 mediates neuromicrovascular degeneration induced by transarachidonic acids in rodents.

Jean-Claude Honoré; Amna Kooli; David Hamel; Thierry Alquier; José-Carlos Rivera; Christiane Quiniou; Xin Hou; Elsa Kermorvant-Duchemin; Pierre Hardy; Vincent Poitout; Sylvain Chemtob

Objective—Nitro-oxidative stress exerts a significant role in the genesis of hypoxic-ischemic (HI) brain injury. We previously reported that the &ohgr;-6 long chain fatty acids, transarachidonic acids (TAAs), which are nitrative stress-induced nonenzymatically generated arachidonic acid derivatives, trigger selective microvascular endothelial cell death in neonatal neural tissue. The primary molecular target of TAAs remains unidentified. GPR40 is a G protein–coupled receptor activated by long chain fatty acids, including &ohgr;-6; it is highly expressed in brain, but its functions in this tissue are largely unknown. We hypothesized that TAAs play a significant role in neonatal HI-induced cerebral microvascular degeneration through GPR40 activation. Approach and Results—Within 24 hours of a HI insult to postnatal day 7 rat pups, a cerebral infarct and a 40% decrease in cerebrovascular density was observed. These effects were associated with an increase in nitrative stress markers (3-nitrotyrosine immunoreactivity and TAA levels) and were reduced by treatment with nitric oxide synthase inhibitor. GPR40 was expressed in rat pup brain microvasculature. In vitro, in GPR40-expressing human embryonic kidney (HEK)-293 cells, [14C]-14E-AA (radiolabeled TAA) bound specifically, and TAA induced calcium transients, extracellular signal–regulated kinase 1/2 phosphorylation, and proapoptotic thrombospondin-1 expression. In vivo, intracerebroventricular injection of TAAs triggered thrombospondin-1 expression and cerebral microvascular degeneration in wild-type mice, but not in GPR40-null congeners. Additionally, HI-induced neurovascular degeneration and cerebral infarct were decreased in GPR40-null mice. Conclusions—GPR40 emerges as the first identified G protein–coupled receptor conveying actions of nonenzymatically generated nitro-oxidative products, specifically TAAs, and is involved in (neonatal) HI encephalopathy.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2010

Sustained hypercapnia induces cerebral microvascular degeneration in the immature brain through induction of nitrative stress.

Jean-Claude Honoré; Amna Kooli; Xin Hou; David Hamel; José Carlos Rivera; Emilie Picard; Pierre Hardy; Sophie Tremblay; Daya R. Varma; Robert P. Jankov; Joseph A. Mancini; Michael Balazy; Sylvain Chemtob

Hypercapnia is regularly observed in chronic lung disease, such as bronchopulmonary dysplasia in preterm infants. Hypercapnia results in increased nitric oxide synthase activity and in vitro formation of nitrates. Neural vasculature of the immature subject is particularly sensitive to nitrative stress. We investigated whether exposure to clinically relevant sustained high CO(2) causes microvascular degeneration in the newborn brain by inducing nitrative stress, and whether this microvascular degeneration has an impact on brain growth. Newborn rat pups were exposed to 10% CO(2) as inspired gas (Pa(CO(2)) = 60-70 mmHg) starting within 24 h of birth until postnatal day 7 (P7). Brains were notably collected at different time points to measure vascular density, determine brain cortical nitrite/nitrate, and trans-arachidonic acids (TAAs; products of nitration) levels as effectors of vessel damage. Chronic exposure of rat pups to high CO(2) (Pa(CO(2)) approximately 65 mmHg) induced a 20% loss in cerebrovascular density at P3 and a 15% decrease in brain mass at P7; at P30, brain mass remained lower in CO(2)-exposed animals. Within 24 h of exposure to CO(2), brain eNOS expression and production of nitrite/nitrate doubled, lipid nitration products (TAAs) increased, and protein nitration (3-nitrotyrosine immunoreactivity) was also coincidently augmented on brain microvessels (lectin positive). Intracerebroventricular injection of TAAs (10 microM) replicated cerebrovascular degeneration. Treatment of rat pups with NOS inhibitor (L-N(omega)-nitroarginine methyl ester) or a peroxynitrite decomposition catalyst (FeTPPS) prevented hypercapnia-induced microvascular degeneration and preserved brain mass. Cytotoxic effects of high CO(2) were reproduced in vitro/ex vivo on cultured endothelial cells and sprouting microvessels. In summary, hypercapnia at values frequently observed in preterm infants with chronic lung disease results in increased nitrative stress, which leads to cerebral cortical microvascular degeneration and curtails brain growth.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2013

Restoration of renal function by a novel prostaglandin EP4 receptor-derived peptide in models of acute renal failure

Martin Leduc; Xin Hou; David Hamel; Melanie Sanchez; Christiane Quiniou; Jean-Claude Honoré; Olivier Roy; Ankush Madaan; William D. Lubell; Daya R. Varma; Joseph A. Mancini; François Duhamel; Krishna G. Peri; Vincent Pichette; Nikolaus Heveker; Sylvain Chemtob

Acute renal failure (ARF) is a serious medical complication characterized by an abrupt and sustained decline in renal function. Despite significant advances in supportive care, there is currently no effective treatment to restore renal function. PGE(2) is a lipid hormone mediator abundantly produced in the kidney, where it acts locally to regulate renal function; several studies suggest that modulating EP(4) receptor activity could improve renal function following kidney injury. An optimized peptidomimetic ligand of EP(4) receptor, THG213.29, was tested for its efficacy to improve renal function (glomerular filtration rate, renal plasma flow, and urine output) and histological changes in a model of ARF induced by either cisplatin or renal artery occlusion in Sprague-Dawley rats. THG213.29 modulated PGE(2)-binding dissociation kinetics, indicative of an allosteric binding mode. Consistently, THG213.29 antagonized EP(4)-mediated relaxation of piglet saphenous vein rings, partially inhibited EP(4)-mediated cAMP production, but did not affect Gα(i) activation or β-arrestin recruitment. In vivo, THG213.29 significantly improved renal function and histological changes in cisplatin- and renal artery occlusion-induced ARF models. THG213.29 increased mRNA expression of heme-oxygenase 1, Bcl2, and FGF-2 in renal cortex; correspondingly, in EP(4)-transfected HEK293 cells, THG213.29 augmented FGF-2 and abrogated EP(4)-dependent overexpression of inflammatory IL-6 and of apoptotic death domain-associated protein and BCL2-associated agonist of cell death. Our results demonstrate that THG213.29 represents a novel class of diuretic agent with noncompetitive allosteric modulator effects on EP(4) receptor, resulting in improved renal function and integrity following acute renal failure.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2011

Cortactin activation by FVIIa/tissue factor and PAR2 promotes endothelial cell migration

Tang Zhu; Joseph A. Mancini; Przemyslaw Sapieha; Chun Yang; Jean-Sebastien Joyal; Jean-Claude Honoré; Martin Leduc; Karine Zaniolo; Pierre Hardy; Zhuo Shao; Li Fan; Xin Hou; Georges-Etienne Rivard; Sylvain Chemtob

Cellular migration is a complex process that requires the polymerization of actin filaments to drive cellular extension. Smooth muscle and cancer cell migration has been shown to be affected by coagulation factors, notably the factor VII (FVIIa) and tissue factor (TF) complex. The present studies delineated mediators involved with the process of FVIIa/TF-induced cell migration and utilized a simple, precise, and reproducible, migration assay. Both FVIIa and protease-activated receptor-2 (PAR2)-activating peptide, SLIGRL, increased the migration rate of porcine cerebral microvascular endothelial cells (pCMVECs) overexpressing human TF. Ras homolog gene family member A (RhoA) and cortactin were upregulated during the process; expression of HIF, actin polymerization nuclear diaphanous-related formin-1 and -2 (Dia1, and Dia2) were unaffected. Gene silencing by shRNA to PAR2, RhoA, and cortactin attenuated this gene upregulation and migration induced by FVIIa/TF. Utilizing immunocellular localization, we demonstrate that during FVIIa/TF and PAR2 activation, cortactin molecules translocate from the cytoplasm to the cell periphery and assist in lamellipodia formation of pCMVECs. Overall, we demonstrate a novel regulation and role for cortactin in FVIIa/TF-mediated endothelial cell migration that occurs through a PAR2 and RhoA dependent mechanism.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2007

Lysophosphatidic acid induces endothelial cell death by modulating the redox environment

Sonia Brault; Fernand Gobeil; Audrey Fortier; Jean-Claude Honoré; Jean-Sebastien Joyal; Przemyslaw Sapieha; Amna Kooli; Elodie Martin; Pierre Hardy; Alfredo Ribeiro-da-Silva; Krishna G. Peri; Pierre Lachapelle; Daya R. Varma; Sylvain Chemtob


Free Radical Biology and Medicine | 2008

Trans-Arachidonic acids induce a heme oxygenase-dependent vasorelaxation of cerebral microvasculature

Amna Kooli; Elsa Kermorvant-Duchemin; Florian Sennlaub; Michela BossolascoM. Bossolasco; Xin Hou; Jean-Claude Honoré; Phyllis A. Dennery; Przemyslaw Sapieha; Daya R. Varma; Pierre Lachapelle; Tang Zhu; Sophie Tremblay; Pierre Hardy; Kavita Jain; Michael Balazy; Sylvain Chemtob


Investigative Ophthalmology & Visual Science | 2012

A Novel Allosteric Modulator of the IL-1 Receptor Prevents the Development of Oxygen-Induced Retinopathy

José Carlos Rivera; Nicolas Sitaras; David Hamel; Ankush Madaan; Jean-Claude Honoré; Baraa Noueihed; Martine Blais; Christian Quiniou; Przemyslaw Sapieha; Sylvain Chemtob

Collaboration


Dive into the Jean-Claude Honoré's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Hamel

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Pierre Hardy

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Xin Hou

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge