Network


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

Hotspot


Dive into the research topics where David Hamel is active.

Publication


Featured researches published by David Hamel.


Nature Medicine | 2008

The succinate receptor GPR91 in neurons has a major role in retinal angiogenesis

Przemyslaw Sapieha; Mirna Sirinyan; David Hamel; Karine Zaniolo; Jean Sébastien Joyal; Jang Hyeon Cho; Jean Claude Honoré; Elsa Kermorvant-Duchemin; Daya R. Varma; Sophie Tremblay; Martin Leduc; Lenka Rihakova; Pierre Hardy; William H. Klein; Xiuqian Mu; Orval Mamer; Pierre Lachapelle; Adriana Di Polo; Christian M. Beauséjour; Gregor Andelfinger; Grant A. Mitchell; Florian Sennlaub; Sylvain Chemtob

Vascularization is essential for tissue development and in restoration of tissue integrity after an ischemic injury. In studies of vascularization, the focus has largely been placed on vascular endothelial growth factor (VEGF), yet other factors may also orchestrate this process. Here we show that succinate accumulates in the hypoxic retina of rodents and, via its cognate receptor G protein–coupled receptor-91 (GPR91), is a potent mediator of vessel growth in the settings of both normal retinal development and proliferative ischemic retinopathy. The effects of GPR91 are mediated by retinal ganglion neurons (RGCs), which, in response to increased succinate levels, regulate the production of numerous angiogenic factors including VEGF. Accordingly, succinate did not have proangiogenic effects in RGC-deficient rats. Our observations show a pathway of metabolite signaling where succinate, acting through GPR91, governs retinal angiogenesis and show the propensity of RGCs to act as sensors of ischemic stress. These findings provide a new therapeutic target for modulating revascularization.


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.


The International Journal of Biochemistry & Cell Biology | 2010

Proliferative retinopathies: angiogenesis that blinds.

Przemyslaw Sapieha; David Hamel; Zhuo Shao; José Carlos Rivera; Karine Zaniolo; Jean Sébastien Joyal; Sylvain Chemtob

Proliferative ischemic retinopathies such as proliferative diabetic retinopathy (PDR), retinopathy of prematurity (ROP) and those stemming from retinal vein occlusion are the leading causes of blindness in the working age and pediatric populations of industrialized countries. They present major financial burdens for health care systems and account for significant loss of productivity. These pathologies are characterized by excessive pre-retinal blood vessel growth that can ultimately lead to a fibrous scar formation and culminate in retinal detachment. This abnormal and disproportionate hyper-vascularization is a compensatory mechanism to overcome an earlier phase of microvessel degeneration and reinstate metabolic equilibrium to the hypoxic retina. To date, the treatment modalities to counter these diseases largely rely on invasive and moderately efficient surgical interventions. In this review, we discuss the current views on retinal vaso-obliteration, neovascularization and available treatments and present future strategies to tackle these diseases.


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.


Nature Medicine | 2014

Subcellular localization of coagulation factor II receptor-like 1 in neurons governs angiogenesis

Jean Sébastien Joyal; Satra Nim; Tang Zhu; Nicholas Sitaras; José Carlos Rivera; Zhuo Shao; Przemyslaw Sapieha; David Hamel; Melanie Sanchez; Karine Zaniolo; Manon St-Louis; Johanne Ouellette; Martín Montoya-Zavala; Alexandra Zabeida; Emilie Picard; Pierre Hardy; Vikrant K. Bhosle; Daya R. Varma; Christian M. Beauséjour; Christelle Boileau; William H. Klein; Morley D. Hollenberg; Alfredo Ribeiro-da-Silva; Gregor Andelfinger; Sylvain Chemtob

Neurons have an important role in retinal vascular development. Here we show that the G protein–coupled receptor (GPCR) coagulation factor II receptor-like 1 (F2rl1, previously known as Par2) is abundant in retinal ganglion cells and is associated with new blood vessel formation during retinal development and in ischemic retinopathy. After stimulation, F2rl1 in retinal ganglion cells translocates from the plasma membrane to the cell nucleus using a microtubule-dependent shuttle that requires sorting nexin 11 (Snx11). At the nucleus, F2rl1 facilitates recruitment of the transcription factor Sp1 to trigger Vegfa expression and, in turn, neovascularization. In contrast, classical plasma membrane activation of F2rl1 leads to the expression of distinct genes, including Ang1, that are involved in vessel maturation. Mutant versions of F2rl1 that prevent nuclear relocalization but not plasma membrane activation interfere with Vegfa but not Ang1 expression. Complementary angiogenic factors are therefore regulated by the subcellular localization of a receptor (F2rl1) that governs angiogenesis. These findings may have implications for the selectivity of drug actions based on the subcellular distribution of their targets.


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 | 2012

Role of receptor-mediated endocytosis in the antiangiogenic effects of human T lymphoblastic cell-derived microparticles

Chun Yang; Wei Xiong; Qian Qiu; Zhuo Shao; David Hamel; Houda Tahiri; Gregoire Leclair; Pierre Lachapelle; Sylvain Chemtob; Pierre Hardy

Microparticles possess therapeutic potential regarding angiogenesis. We have demonstrated the contribution of apoptotic human CEM T lymphocyte-derived microparticles (LMPs) as inhibitors of angiogenic responses in animal models of inflammation and tumor growth. In the present study, we characterized the antivascular endothelial growth factor (VEGF) effects of LMPs on pathological angiogenesis in an animal model of oxygen-induced retinopathy and explored the role of receptor-mediated endocytosis in the effects of LMPs on human retinal endothelial cells (HRECs). LMPs dramatically inhibited cell growth of HRECs, suppressed VEGF-induced cell migration in vitro experiments, and attenuated VEGF-induced retinal vascular leakage in vivo. Intravitreal injections of fluorescently labeled LMPs revealed accumulation of LMPs in retinal tissue, with more than 60% reductions of the vascular density in retinas of rats with oxygen-induced neovascularization. LMP uptake experiments demonstrated that the interaction between LMPs and HRECs is dependent on temperature. In addition, endocytosis is partially dependent on extracellular calcium. RNAi-mediated knockdown of low-density lipoprotein receptor (LDLR) reduced the uptake of LMPs and attenuated the inhibitory effects of LMPs on VEGF-A protein expression and HRECs cell growth. Intravitreal injection of lentivirus-mediated RNA interference reduced LDLR protein expression in retina by 53% and significantly blocked the antiangiogenic effects of LMPs on pathological vascularization. In summary, the potent antiangiogenic LMPs lead to a significant reduction of pathological retinal angiogenesis through modulation of VEGF signaling, whereas LDLR-mediated endocytosis plays a partial, but pivotal, role in the uptake of LMPs in HRECs.


Viral Immunology | 2011

Herpes Simplex Virus Type 1-induced FasL Expression in Human Monocytic Cells and Its Implications for Cell Death, Viral Replication, and Immune Evasion

Alexandre Iannello; Olfa Debbeche; Raoudha El Arabi; Suzanne Samarani; David Hamel; Flore Rozenberg; Nikolaus Heveker; Ali Ahmad

Herpes simplex virus type 1 (HSV-1) is a ubiquitously occurring pathogen that infects humans early in childhood. The virus persists as a latent infection in dorsal root ganglia, especially of the trigeminal nerve, and frequently becomes reactivated in humans under conditions of stress. Monocytic cells constitute an important component of the innate and adaptive immune responses. We show here for the first time that HSV-1 stimulates human FasL promoter and induces de novo expression of FasL on the surface of human monocytic cells, including monocytes and macrophages. This virus-induced FasL expression causes death of monocytic cells growing in suspension, but not in monolayers (e.g., macrophages). The addition of a broad-spectrum caspase inhibitor, as well as anti-FasL antibodies, reduced cell death but increased viral replication in the virus-infected cell cultures. We also show here for the first time that the virus-induced de novo expression of FasL on the cell surface acts as an immune evasion mechanism by causing the death of interacting human CD4+ T cells, CD8+ T cells, and natural killer (NK) cells. Our study provides novel insights on FasL expression and cell death in HSV-infected human monocytic cells and their impact on interacting immune cells.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2009

VRQ397 (CRAVKY): a novel noncompetitive V2 receptor antagonist

Rihakova L; Christiane Quiniou; Hamdan Ff; Kaul R; Sonia Brault; Xin Hou; Isabelle Lahaie; Sapieha P; David Hamel; Zhuo Shao; Pierre Hardy; Jean-Sebastien Joyal; Nedev H; Duhamel F; Beauregard K; Nikolaus Heveker; Saragovi Hu; Guillon G; Bouvier M; William D. Lubell; Sylvain Chemtob

Vasopressin type 2 receptor (V2R) exhibits mostly important properties for hydroosmotic equilibrium and, to a lesser extent, on vasomotricity. Drugs currently acting on this receptor are analogs of the natural neuropeptide, arginine vasopressin (AVP), and hence are competitive ligands. Peptides that reproduce specific sequences of a given receptor have lately been reported to interfere with its action, and if such molecules arise from regions remote from the binding site they would be anticipated to exhibit noncompetitive antagonism, but this has yet to be shown for V2R. Six peptides reproducing juxtamembranous regions of V2R were designed and screened; the most effective peptide, cravky (labeled VRQ397), was characterized. VRQ397 was potent (IC(50) = 0.69 +/- 0.25 nM) and fully effective in inhibiting V2R-dependent physiological function, specifically desmopressin-L-desamino-8-arginine-vasopressin (DDAVP)-induced cremasteric vasorelaxation; this physiological functional assay was utilized to avoid overlooking interference of specific signaling events. A dose-response profile revealed a noncompetitive property of VRQ397; correspondingly, VRQ397 bound specifically to V2R-expressing cells could not displace its natural ligand, AVP, but modulated AVP binding kinetics (dissociation rate). Specificity of VRQ397 was further confirmed by its inability to bind to homologous V1 and oxytocin receptors and its inefficacy to alter responses to stimulation of these receptors. VRQ397 exhibited pharmacological permissiveness on V2R-induced signals, as it inhibited DDAVP-induced PGI(2) generation but not that of cAMP or recruitment of beta-arrestin2. Consistent with in vitro and ex vivo effects as a V2R antagonist, VRQ397 displayed anticipated in vivo aquaretic efficacy. We hereby describe the discovery of a first potent noncompetitive antagonist of V2R, which exhibits functional selectivity, in line with properties of a negative allosteric modulator.

Collaboration


Dive into the David Hamel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pierre Hardy

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karine Zaniolo

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Tang Zhu

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge