Agnieszka Dejda
Université de Montréal
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Agnieszka Dejda.
Journal of Molecular Neuroscience | 2008
Agnieszka Dejda; Valérie Jolivel; Steve Bourgault; Tommy Seaborn; Alain Fournier; Hubert Vaudry; David Vaudry
Programmed cell death, which is part of the normal development of the central nervous system, is also implicated in various neurodegenerative disorders. Cysteine-dependent aspartate-specific proteases (caspases) play a pivotal role in the cascade of events leading to apoptosis. Many factors that inhibit cell death have now been identified, but the underlying mechanisms are not fully understood. Pituitary adenylate cylase-activating polypeptide (PACAP) has been shown to exert neurotrophic activities during development and to prevent neuronal apoptosis induced by various insults such as ischemia. Most of the neuroprotective effects of PACAP are mediated through the PAC1 receptor. This receptor activates a transduction cascade of second messengers to stimulate Bcl-2 expression, which inhibits cytochrome c release and blocks the activation of caspases. The inhibitory effect of PACAP on the apoptotic cascade suggests that selective, stable, and potent PACAP derivatives could potentially be of therapeutic value for the treatment of post-traumatic and/or chronic neurodegenerative processes.
Current Medicinal Chemistry | 2009
Steve Bourgault; David Vaudry; Agnieszka Dejda; Ngoc-Duc Doan; H. Vaudry; Alain Fournier
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid peptide that was initially isolated from hypothalamus extracts on the basis of its ability to stimulate the production of cAMP in cultured pituitary cells. Recent studies have shown that PACAP exerts potent neuroprotective effects not only in vitro but also in in vivo models of Parkinsons disease, Huntingtons disease, traumatic brain injury and stroke. The protective effects of PACAP are based on its capacity to prevent neuronal apoptosis by acting directly on neurons or indirectly through the release of neuroprotective factors by astrocytes. These biological activities are mainly mediated through activation of the PAC1 receptor which is currently considered as a potential target for the treatment of neurodegenerative diseases. However, the use of native PACAP, the endogenous ligand of PAC1, as an efficient neuroprotective drug is actually limited by its rapid degradation. Moreover, injection of PACAP to human induces peripheral side effects which are mainly mediated through VPAC1 and VPAC2 receptors. Strategies to overcome these compromising conditions include the development of metabolically stable analogs of PACAP acting as selective agonists of the PAC1 receptor. This review presents an overview of the structure-activity relationships of PACAP and summarizes the molecular and conformational requirements for activation of PAC1 receptor. The applicability of PACAP analogs as therapeutic agents for treatment of neurodegenerative diseases is also discussed.
Cell Metabolism | 2013
Agustin Cerani; Nicolas Tetreault; Catherine Ménard; Eric Lapalme; Chintan Patel; Nicholas Sitaras; Felix Beaudoin; Dominique Leboeuf; Vincent De Guire; François Binet; Agnieszka Dejda; Flavio Rezende; Khalil Miloudi; Przemyslaw Sapieha
The deterioration of the inner blood-retinal barrier and consequent macular edema is a cardinal manifestation of diabetic retinopathy (DR) and the clinical feature most closely associated with loss of sight. We provide evidence from both human and animal studies for the critical role of the classical neuronal guidance cue, semaphorin 3A, in instigating pathological vascular permeability in diabetic retinas via its cognate receptor neuropilin-1. We reveal that semaphorin 3A is induced in early hyperglycemic phases of diabetes within the neuronal retina and precipitates initial breakdown of endothelial barrier function. We demonstrate, by a series of orthogonal approaches, that neutralization of semaphorin 3A efficiently prevents diabetes-induced retinal vascular leakage in a stage of the disease when vascular endothelial growth factor neutralization is inefficient. These observations were corroborated in Tg(Cre-Esr1)/Nrp1(flox/flox) conditional knockout mice. Our findings identify a therapeutic target for macular edema and provide further evidence for neurovascular crosstalk in the pathogenesis of DR.
Peptides | 2007
Olfa Masmoudi-Kouki; Pierrick Gandolfo; Hélène Castel; Jérôme Leprince; Alain Fournier; Agnieszka Dejda; Hubert Vaudry; Marie-Christine Tonon
Astrocytes represent at least 50% of the volume of the human brain. Besides their roles in various supportive functions, astrocytes are involved in the regulation of stem cell proliferation, synaptic plasticity and neuroprotection. Astrocytes also influence neuronal physiology by responding to neurotransmitters and neuropeptides and by releasing regulatory factors termed gliotransmitters. In particular, astrocytes express the PACAP-specific receptor PAC1-R and the PACAP/VIP mutual receptors VPAC1-R and VPAC2-R during development and/or in the adult. There is now clear evidence that PACAP and VIP modulate a number of astrocyte activities such as proliferation, plasticity, glycogen production, and biosynthesis of neurotrophic factors and gliotransmitters.
Peptides | 2011
Agnieszka Dejda; Tommy Seaborn; Steve Bourgault; Omar Touzani; Alain Fournier; Hubert Vaudry; David Vaudry
Pituitary adenylate cyclase-activating polypeptide (PACAP) shows potent protective effects in numerous models of neurological insults. However, the use of PACAP as a clinically efficient drug is limited by its poor metabolic stability. By combining identification of enzymatic cleavage sites with targeted chemical modifications, a metabolically stable and potent PACAP38 analog was recently developed. The neuroprotective activity of this novel compound was for the first time evaluated and compared to the native peptide using a rat model of middle cerebral artery occlusion (MCAO). Our results show that as low as picomolar doses of PACAP38 and its analog strongly reduce infarct volume and improve neurological impairment induced by stroke. In particular, these peptides inhibit the expression of Bcl-2-associated death promoter, caspase 3, macrophage inflammatory protein-1α, inducible nitric oxide synthase 2, tumor necrosis factor-α mRNAs, and increase extracellular signal-regulated kinase 2, B-cell CLL/lymphoma 2 and interleukin 6 mRNA levels. These results indicate that the neuroprotective effect of PACAP after MCAO is not only due to its ability to inhibit apoptosis but also to modulate the inflammatory response. The present study highlights the potential therapeutic efficacy of very low concentrations of PACAP or its metabolically stable derivative for the treatment of stroke.
Journal of Clinical Investigation | 2014
Agnieszka Dejda; Gaelle Mawambo; Agustin Cerani; Khalil Miloudi; Zhuo Shao; Jean-François Daudelin; Salix Boulet; Malika Oubaha; Felix Beaudoin; Naoufal Akla; Sullivan Henriques; Catherine Ménard; Andreas Stahl; Jean-Sébastien Delisle; Flavio Rezende; Nathalie Labrecque; Przemyslaw Sapieha
Immunological activity in the CNS is largely dependent on an innate immune response and is heightened in diseases, such as diabetic retinopathy, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimers disease. The molecular dynamics governing immune cell recruitment to sites of injury and disease in the CNS during sterile inflammation remain poorly defined. Here, we identified a subset of mononuclear phagocytes (MPs) that responds to local chemotactic cues that are conserved among central neurons, vessels, and immune cells. Patients suffering from late-stage proliferative diabetic retinopathy (PDR) had elevated vitreous semaphorin 3A (SEMA3A). Using a murine model, we found that SEMA3A acts as a potent attractant for neuropilin-1-positive (NRP-1-positive) MPs. These proangiogenic MPs were selectively recruited to sites of pathological neovascularization in response to locally produced SEMA3A as well as VEGF. NRP-1-positive MPs were essential for disease progression, as NRP-1-deficient MPs failed to enter the retina in a murine model of oxygen-induced retinopathy (OIR), a proxy for PDR. OIR mice with NRP-1-deficient MPs exhibited decreased vascular degeneration and diminished pathological preretinal neovascularization. Intravitreal administration of a NRP-1-derived trap effectively mimicked the therapeutic benefits observed in mice lacking NRP-1-expressing MPs. Our findings indicate that NRP-1 is an obligate receptor for MP chemotaxis, bridging neural ischemia to an innate immune response in neovascular retinal disease.
Biochemical Pharmacology | 2011
Ngoc-Duc Doan; Steve Bourgault; Agnieszka Dejda; Myriam Létourneau; Michel Detheux; David Vaudry; Hubert Vaudry; David Chatenet; Alain Fournier
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide that exerts a large array of actions in the central nervous system and periphery. Through the activation of PAC1 and VPAC1, PACAP is able to exert neuroprotective, as well as anti-inflammatory effects, two phenomena involved in the pathogenesis and the progression of neurodegenerative diseases. The aim of the current study was to provide insights into the molecular arrangement of the amino terminus of PACAP and to develop new potent and selective PAC1/VPAC1 agonists promoting neuronal survival. We have synthesized a series of PACAP derivatives and measured their binding affinity and their ability to induce intracellular calcium mobilization for each receptor, i.e. PAC1, VPAC1, and VPAC2. Ultimately, analogs with an improved pharmacological profile were evaluated in an in vitro model of neuronal loss. Results showed that introduction of a hydroxyproline or an alanine moiety, respectively, at position 2 or 7 generated derivatives without significant VPAC2 agonistic activity. Moreover, the structure-activity relationship study suggests the presence of common (Asx-turn like) and distinct (different N-capping type) secondary structures that might be responsible for receptor recognition, selectivity and activation. Finally, evaluation of the neuroprotective activity of [Ala(7)]PACAP27 and [Hyp(2)]PACAP27 demonstrated their ability to protect potently human dopaminergic SH-SY5Y neuroblasts against the toxicity of MPP(+), in pre- and co-treatment experiments. These new pharmacological and structural data should prove useful for the rational design of PACAP-derived compounds that could be putative therapeutic agents for the treatment of neurodegenerative diseases.
Science Translational Medicine | 2016
Malika Oubaha; Khalil Miloudi; Agnieszka Dejda; Vera Guber; Gaelle Mawambo; Marie-Anne Germain; Guillaume Bourdel; Natalija Popovic; Flavio Rezende; Randal J. Kaufman; Frédérick A. Mallette; Przemyslaw Sapieha
Senescent cells encourage abnormal blood vessels and retinal disease. Old but deadly Diabetics often lose their sight, a result of poor blood supply to the retina. Oubaha and colleagues investigated how this oxygen deprivation leads to disease. They found that retinal cells do not simply die when oxygen-starved, rather they become senescent—a state in which the cells secrete a cocktail of undesirable molecules, in mice and humans. These cytokines drive more senescence, encourage abnormal blood vessel formation, and block healthy regeneration. Treatment of mice with a diabetes drug or a senescence inhibitor reduced retinal disease, suggesting that interrupting this process could be therapeutically beneficial. Pathological angiogenesis is the hallmark of diseases such as cancer and retinopathies. Although tissue hypoxia and inflammation are recognized as central drivers of vessel growth, relatively little is known about the process that bridges the two. In a mouse model of ischemic retinopathy, we found that hypoxic regions of the retina showed only modest rates of apoptosis despite severely compromised metabolic supply. Using transcriptomic analysis and inducible loss-of-function genetics, we demonstrated that ischemic retinal cells instead engage the endoplasmic reticulum stress inositol-requiring enzyme 1α (IRE1α) pathway that, through its endoribonuclease activity, induces a state of senescence in which cells adopt a senescence-associated secretory phenotype (SASP). We also detected SASP-associated cytokines (plasminogen activator inhibitor 1, interleukin-6, interleukin-8, and vascular endothelial growth factor) in the vitreous humor of patients suffering from proliferative diabetic retinopathy. Therapeutic inhibition of the SASP through intravitreal delivery of metformin or interference with effectors of senescence (semaphorin 3A or IRE1α) in mice reduced destructive retinal neovascularization in vivo. We conclude that the SASP contributes to pathological vessel growth, with ischemic retinal cells becoming prematurely senescent and secreting inflammatory cytokines that drive paracrine senescence, exacerbate destructive angiogenesis, and hinder reparative vascular regeneration. Reversal of this process may be therapeutically beneficial.
Annals of the New York Academy of Sciences | 2006
Agnieszka Dejda; Marta Jozwiak-Bebenista; Jerzy Z. Nowak
Abstract: Pituitary adenylate cyclase‐activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), and peptide histidine‐isoleucine (PHI) are members of a superfamily of structurally related peptides widely distributed in the body and displaying pleiotropic biological activities. All these peptides are known to act via common receptors—VPAC1 and VPAC2. In addition, the effects of PACAP are mediated through its specific receptor named PAC1. The main signal transduction pathway of the mentioned receptors is adenylyl cyclase (AC)→cAMP system. PACAP and VIP may also signal through receptor‐linked phospholipase C (PLC)→IP3/DAG→PKC and phospholipase D (PLD)→phosphatidic acid (PA) pathways. In the present article, we have studied the effects of PACAP, VIP, and PHI (0.001–5000 nM) on the AC‐, PLC‐, and PLD‐driven signaling pathways in rat primary glial cell (astrocytes) cultures. All tested peptides dose‐dependently and strongly stimulated cyclic adenosine 3´,5´‐monophosphate (cAMP) production in this experimental model, displaying the following rank order of potency: PACAP >> VIP ≥ PHI. Their effects on PLC‐IP3/DAG were weaker, while only PACAP and VIP (0.1‐5 μM) significantly stimulated PLD activity. The obtained results showed that rat cerebral cortex‐derived astrocytes are responsive to PACAP, VIP and PHI/PHM and possess PAC1 and likely VPAC‐type receptors linked to activation of AC‐cAMP‐, PLC‐IP3/DAG‐, and PLD‐PA signaling systems.
Biochimie | 2011
Agnieszka Dejda; Steve Bourgault; Ngoc Duc Doan; Myriam Létourneau; Alain Couvineau; Hubert Vaudry; David Vaudry; Alain Fournier
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts many crucial biological functions through the interaction with its specific PAC1 receptor (PAC1-R), a class B G protein-coupled receptor (GPCR). To identify the binding sites of PACAP in the PAC1-R, three peptide derivatives containing a photoreactive p-benzoyl-phenylalanine (Bpa) residue were developed. These photosensitive PACAP analogs were fully biologically active and competent to displace radiolabeled Ac-PACAP27 from the PAC1-R. Subsequently, the (125)I-labeled photoprobes were used to anchor the PAC1-R expressed in Chinese hamster ovary cells. Photolabeling led to the formation of two protein complexes of 76 and 67 kDa, representing different glycosylated forms of the receptor. Proteinase and chemical cleavages of the peptide-receptor complexes revealed that (125)I[Bpa(0), Nle(17)]PACAP27, (125)I[Bpa(6), Nle(17)]PACAP27 and (125)I[Nle(17), Bpa(22)]PACAP27 covalently labeled the Ser(98) - Met(111) segment, the Ser(124) - Glu(125) dipeptide and the Ser(141) - Met(172) fragment, respectively. Taking into account the topology of the PAC1-R, these segments are mainly located within the extracellular N-terminal domain, indicating that this PAC1-R domain is the major binding site of PACAP27. The present study constitutes the first characterization of the binding domains of PACAP to its specific receptor and suggests heterogeneity within the binding mode of peptide ligands to class B GPCRs.