Network


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

Hotspot


Dive into the research topics where Baraa Noueihed is active.

Publication


Featured researches published by Baraa Noueihed.


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.


Journal of Immunology | 2015

Novel Noncompetitive IL-1 Receptor-Biased Ligand Prevents Infection- and Inflammation-Induced Preterm Birth.

Mathieu Nadeau-Vallée; Christiane Quiniou; Julia Palacios; Xin Hou; Atefeh Erfani; Ankush Madaan; Melanie Sanchez; Kelycia Leimert; Amarilys Boudreault; François Duhamel; José Carlos Rivera; Tang Zhu; Baraa Noueihed; Sarah A. Robertson; Xin Ni; David M. Olson; William D. Lubell; Sylvie Girard; Sylvain Chemtob

Preterm birth (PTB) is firmly linked to inflammation regardless of the presence of infection. Proinflammatory cytokines, including IL-1β, are produced in gestational tissues and can locally upregulate uterine activation proteins. Premature activation of the uterus by inflammation may lead to PTB, and IL-1 has been identified as a key inducer of this condition. However, all currently available IL-1 inhibitors are large molecules that exhibit competitive antagonism properties by inhibiting all IL-1R signaling, including transcription factor NF-κB, which conveys important physiological roles. We hereby demonstrate the efficacy of a small noncompetitive (all-d peptide) IL-1R–biased ligand, termed rytvela (labeled 101.10) in delaying IL-1β–, TLR2-, and TLR4-induced PTB in mice. The 101.10 acts without significant inhibition of NF-κB, and instead selectively inhibits IL-1R downstream stress-associated protein kinases/transcription factor c-jun and Rho GTPase/Rho-associated coiled-coil–containing protein kinase signaling pathways. The 101.10 is effective at decreasing proinflammatory and/or prolabor genes in myometrium tissue and circulating leukocytes in all PTB models independently of NF-κB, undermining NF-κB role in preterm labor. In this work, biased signaling modulation of IL-1R by 101.10 uncovers a novel strategy to prevent PTB without inhibiting NF-κB.


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.


Oxidative Medicine and Cellular Longevity | 2017

Ischemic Retinopathies: Oxidative Stress and Inflammation

José Carlos Rivera; Rabah Dabouz; Baraa Noueihed; Samy Omri; Houda Tahiri; Sylvain Chemtob

Ischemic retinopathies (IRs), such as retinopathy of prematurity (ROP), diabetic retinopathy (DR), and (in many cases) age-related macular degeneration (AMD), are ocular disorders characterized by an initial phase of microvascular changes that results in ischemia, followed by a second phase of abnormal neovascularization that may culminate into retinal detachment and blindness. IRs are complex retinal conditions in which several factors play a key role during the development of the different pathological stages of the disease. Increasing evidence reveals that oxidative stress and inflammatory processes are important contributors to the pathogenesis of IRs. Despite the beneficial effects of the photocoagulation and anti-VEGF therapy during neovascularization phase, the need to identify novel targets to prevent initial phases of these ocular pathologies is still needed. In this review, we provide an update on the involvement of oxidative stress and inflammation in the progression of IRs and address some therapeutic interventions by using antioxidants and anti-inflammatory agents.


Development | 2018

miR-126-5p promotes retinal endothelial cell survival through SetD5 regulation in neurons

Gaëlle Villain; Loïc Poissonnier; Baraa Noueihed; Gaëlle Bonfils; José Carlos Rivera; Sylvain Chemtob; Fabrice Soncin; Virginie Mattot

ABSTRACT MicroRNAs are key regulators of angiogenesis, as illustrated by the vascular defects observed in miR-126-deficient animals. The miR-126 duplex gives rise to two mature microRNAs (miR-126-3p and -5p). The vascular defects in these mutant animals were attributed to the loss of miR-126-3p but the role of miR-126-5p during normal angiogenesis in vivo remains unknown. Here, we show that miR-126-5p is expressed in endothelial cells but also by retinal ganglion cells (RGCs) of the mouse postnatal retina and participates in protecting endothelial cells from apoptosis during the establishment of the retinal vasculature. miR-126-5p negatively controls class 3 semaphorin protein (Sema3A) in RGCs through the repression of SetD5, an uncharacterized member of the methyltransferase family of proteins. In vitro, SetD5 controls Sema3A expression independently of its SET domain and co-immunoprecipitates with BRD2, a bromodomain protein that recruits transcription regulators onto the chromatin. Both SetD5 and BRD2 bind to the transcription start site and to upstream promoter regions of the Sema3a locus and BRD2 is necessary for the regulation of Sema3A expression by SetD5. Thus, neuronally expressed miR-126-5p regulates angiogenesis by protecting endothelial cells of the developing retinal vasculature from apoptosis. Summary: This study reports for the first time that, through repression of SetD5, neuronal miR-126-5p regulates angiogenesis in developing mouse retina by protecting endothelial cells from Sema3A-mediated apoptosis.


Investigative Ophthalmology & Visual Science | 2015

BIBF1120 (Vargatef) Inhibits Preretinal Neovascularization and Enhances Normal Vascularization in a Model of Vasoproliferative Retinopathy.

José Carlos Rivera; Baraa Noueihed; Samy Omri; J. Barrueco; Frank Hilberg; Sylvain Chemtob

PURPOSE This study evaluated the effects of BIBF1120, a novel triple angiokinase inhibitor against pathological retinal neovascularization. METHODS BIBF1120 effect on development of the normal retinal vasculature was evaluated in Sprague-Dawley rat pups. Two models of ischemic oxygen-induced retinopathy (OIR) and the aortic ring assay were used to assess the antiangiogenic effects of BIBF1120. In the vaso-obliteration model (VO), rat pups were exposed to 80% O2 from postnatal day (P) 5 to P10. In the preretinal neovascularization (NV) model, rat pups were exposed to cycling O2 (50% and 10%) from P1 to P14, followed by room air until P18. Animals were intravitreally or orally treated with BIBF1120. Retinal vasculature, VO, and NV were evaluated in retinal flat mounts. Retinal expression of VEGF, Delta-like ligand 4 (Dll4), Netrin-1, Ephrin-B2, and EphB4 was analyzed by quantitative PCR and Western blot analysis. RESULTS BIBF1120 interfered with normal retinal vascular development and microvessel branching in the aortic assay. However, in VO model BIBF1120 did not accrue VO. On the contrary, in the NV model BIBF1120 accelerated normal retinal vascularization and robustly diminished preretinal neovascularization compared to vehicle (by ~80%). The expression levels of VEGF negative regulator Dll4 and repulsive cues EphrinB2 and EphB4 mRNA in the retina of vehicle-treated OIR animals were markedly increased compared to normoxia, but were normalized by BIBF1120. CONCLUSIONS Data reveal efficacy of BIBF1120 on preretinal neovascularization and, of greater interest, on acceleration of normal vascularization, consistent with interference of major repulsive cues expressed in the retina during OIR. Accordingly, BIBF1120 appears to exhibit preferable properties compared to anti-VEGF therapies for the treatment of ischemic retinopathies.


Journal of Neuroinflammation | 2017

Tetrahydrobiopterin (BH4) deficiency is associated with augmented inflammation and microvascular degeneration in the retina

José Carlos Rivera; Baraa Noueihed; Ankush Madaan; Isabelle Lahaie; Jingyi Pan; Jaques Belik; Sylvain Chemtob


Annals of Eye Science | 2018

AB028. Mesenchymal stem cells repair retinal vascular damage in retinopathy of prematurity mouse model

Baraa Noueihed; José Carlos Rivera; Sylvain Chemtob


Annals of Eye Science | 2018

AB035. Lactate receptor GPR81 modulates epigenetic modification in the subretina

Xiaojuan Yang; Raphael Rouget; Tang Zhu; Christiane Quiniou; Shasha Lv; Suna Jung; François Duhamel; Houda Tahiri; Samy Omri; Baraa Noueihed; Xin Hou; José Carlos Rivera; Mathieu Nadeau-Vallée; Pierre Lachapelle; Sylvain Chemtob


Investigative Ophthalmology & Visual Science | 2015

Tetrahydrobiopterin (BH4) deficiency is associated with microglial activation and microvascular degeneration in the eye.

José Carlos Rivera; Baraa Noueihed; Isabelle Lehaie; Jingyi Pan; Jaques Belik; Sylvain Chemtob

Collaboration


Dive into the Baraa Noueihed's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Hamel

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samy Omri

Hôpital Maisonneuve-Rosemont

View shared research outputs
Top Co-Authors

Avatar

Xin Hou

Université de Montréal

View shared research outputs
Researchain Logo
Decentralizing Knowledge