27 research outputs found

    Study of molecular and cellular mechanisms involved in tne neuroprotective effects of Octadecaneuropeptide (ODN) in a murine model of Parkinson's disease

    No full text
    La maladie de Parkinson (MP) est un trouble neurodégénératif caractérisé par une perte progressive de neurones dopaminergiques (DA) de la substance noire pars compacta (SNpc). Différents mécanismes sont associés à la neuropathogénèse de la MP et en particulier le dysfonctionnement de la chaîne respiratoire mitochondriale, le stress oxydatif, l’apoptose et les processus neuro-inflammatoires. L'octadécaneuropeptide (ODN) est un peptide dérivé du diazepam-binding inhibitor (DBI) exprimé par les cellules astrogliales, qui exerce une action neuroprotectrice dans un modèle cellulaire in vitro de la MP. A ce jour, aucune étude in vivo n’a été réalisée, afin de déterminer si les données obtenues sur les modèles cellulaires in vitro peuvent être transposées in vivo. Le projet de cette thèse consiste ainsi à mettre en évidence l’action protectrice de l’ODN sur la survie des neurones DA de la SNpc dans un modèle murin de la MP et à rechercher les conséquences de l’invalidation du gène du précurseur de l’ODN (DBI) sur la vulnérabilité des neurones DA. Les résultats obtenus montrent qu’une seule injection intra-cérébroventriculaire d’une faible quantité d’ODN (10 ng), 1 h après la dernière administration systémique de 1-méthyl-4-phényl-1,2,3,6-tétrahydropyridine (MPTP) prévient significativement la perte des neurones DA dans la substance noire et la dégénérescence de leurs prolongements nerveux vers le striatum comme mesuré par des marquages et des mesures d’expression de la tyrosine hydroxylase. Cet effet neuroprotecteur de l’ODN est accompagné par une réduction du nombre d’astrocytes réactifs, une forte inhibition de l'expression de gènes pro-inflammatoires tels que les interleukines (IL) IL-1β et IL-6, et tumor necrosis factor-α. De plus, l'ODN bloque l'inhibition du gène anti-apoptotique Bcl-2 et la stimulation des gènes pro-apoptotiques Bax et caspase-3, induite par le MPTP dans la SNpc et le striatum. L'ODN réduit également l’accumulation d'espèces réactives de l'oxygène (ROS) et de produits d'oxydation lipidique dans les neurones DA. Par ailleurs, les souris knock-out DBI (DBI-/-) sont plus vulnérables que les animaux sauvages (DBI+/+) vis-à-vis de la neurotoxicité du MPTP. L’absence de production d’ODN endogène, chez les souris DBI-/- parkinsoniennes, augmente les dommages cellulaires induits par le MPTP, la réactivité gliale, les taux de ROS, l’expression de cytokines pro-inflammatoires et l'activité de la caspase-3 dans la région nigro-striée. L’ensemble de ces résultats montre que le gliopeptide ODN exerce un puissant effet neuroprotecteur contre la dégénérescence des neurones DA de la SNpc induite par le MPTP, chez la souris. Cette action protectrice met en jeu des mécanismes impliquant l’inhibition des processus neuro-inflammatoires, oxydatifs et apoptotiques. D’autre part, la déficience en ODN potentialise les effets délétères du MPTP, suggérant que ce peptide joue un rôle clé lors de la réponse à un stress cellulaire.Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of loss of dopaminergic (DA) neurons within the substantia nigra pars compacta (SNpc). Different mechanisms are associated with the neuropathogenesis of PD including dysfunction of the mitochondrial respiratory chain, oxidative stress, apoptosis and neuroinflammatory processes. Octadecaneuropeptide (ODN) is a diazepam-binding inhibitor (DBI)-derived peptide, expressed by astrocytes, which protects neurons against oxidative cell damages and apoptosis in an in vitro model of PD. Nevertheless, its protective action in vivo has never been investigated. Therefore, the aim of the project of this thesis was to investigate whether intracerebroventricular (i.c.v) injection of ODN could prevent DA neuron degeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, and to explore the vulnerability of ODN precursor knockout (DBI KO) mice to MPTP-induced neurotoxicity. The results show that a single i.c.v injection of 10 ng/μl ODN, 1 h after the last systemic administration of MPTP, prevents the reduction of the number of tyrosine hydroxylase (TH)-positive cell bodies and fibers in the SNpc and striatum, respectively. Immunofluorescence imaging, Western blot analysis and Q-PCR studies revealed that ODN totally abolished MPTP-induced decrease of TH positive cells, mRNA expression and protein levels. This neuroprotective effect of ODN is accompanied by a reduction in the number of reactive astrocytes, an inhibition of the expression of pro-inflammatory genes such as interleukins (IL) IL-1β and IL-6, and a decrease of tumor necrosis factor -α. In addition, ODN blocks the inhibition of the anti-apoptotic Bcl-2 gene and the stimulation of Bax and caspase-3 expression induced by MPTP in the SNpc and striatum. ODN also reduces the accumulation of reactive oxygen species (ROS) and lipid oxidation products in DA neurons. Furthermore, DBI-/- mice exhibited more vulnerability to MPTP than wild-type animals (DBI+/+). Thus, ODN KO mice are more sensitive to MPTP-induced inflammatory and oxidative brain damages, suggesting that the endogenous OD may also be neuroprotective. These results indicate that, based on its anti-oxidative, anti-inflammatory and anti-apoptotic effect, the gliopeptide ODN could lead to the development of effective therapeutic agents for the treatment of cerebral injuries involving oxidative neurodegeneration

    Etude des mécanismes cellulaires et moléculaires impliqués dans les effets neuroprotecteurs du gliopeptide OctaDecaNeuropeptide (ODN) dans un model murin de la Maladie de Parkinson

    No full text
    Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of loss of dopaminergic (DA) neurons within the substantia nigra pars compacta (SNpc). Different mechanisms are associated with the neuropathogenesis of PD including dysfunction of the mitochondrial respiratory chain, oxidative stress, apoptosis and neuroinflammatory processes. Octadecaneuropeptide (ODN) is a diazepam-binding inhibitor (DBI)-derived peptide, expressed by astrocytes, which protects neurons against oxidative cell damages and apoptosis in an in vitro model of PD. Nevertheless, its protective action in vivo has never been investigated. Therefore, the aim of the project of this thesis was to investigate whether intracerebroventricular (i.c.v) injection of ODN could prevent DA neuron degeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, and to explore the vulnerability of ODN precursor knockout (DBI KO) mice to MPTP-induced neurotoxicity. The results show that a single i.c.v injection of 10 ng/μl ODN, 1 h after the last systemic administration of MPTP, prevents the reduction of the number of tyrosine hydroxylase (TH)-positive cell bodies and fibers in the SNpc and striatum, respectively. Immunofluorescence imaging, Western blot analysis and Q-PCR studies revealed that ODN totally abolished MPTP-induced decrease of TH positive cells, mRNA expression and protein levels. This neuroprotective effect of ODN is accompanied by a reduction in the number of reactive astrocytes, an inhibition of the expression of pro-inflammatory genes such as interleukins (IL) IL-1β and IL-6, and a decrease of tumor necrosis factor -α. In addition, ODN blocks the inhibition of the anti-apoptotic Bcl-2 gene and the stimulation of Bax and caspase-3 expression induced by MPTP in the SNpc and striatum. ODN also reduces the accumulation of reactive oxygen species (ROS) and lipid oxidation products in DA neurons. Furthermore, DBI-/- mice exhibited more vulnerability to MPTP than wild-type animals (DBI+/+). Thus, ODN KO mice are more sensitive to MPTP-induced inflammatory and oxidative brain damages, suggesting that the endogenous OD may also be neuroprotective. These results indicate that, based on its anti-oxidative, anti-inflammatory and anti-apoptotic effect, the gliopeptide ODN could lead to the development of effective therapeutic agents for the treatment of cerebral injuries involving oxidative neurodegeneration.La maladie de Parkinson (MP) est un trouble neurodégénératif caractérisé par une perte progressive de neurones dopaminergiques (DA) de la substance noire pars compacta (SNpc). Différents mécanismes sont associés à la neuropathogénèse de la MP et en particulier le dysfonctionnement de la chaîne respiratoire mitochondriale, le stress oxydatif, l’apoptose et les processus neuro-inflammatoires. L'octadécaneuropeptide (ODN) est un peptide dérivé du diazepam-binding inhibitor (DBI) exprimé par les cellules astrogliales, qui exerce une action neuroprotectrice dans un modèle cellulaire in vitro de la MP. A ce jour, aucune étude in vivo n’a été réalisée, afin de déterminer si les données obtenues sur les modèles cellulaires in vitro peuvent être transposées in vivo. Le projet de cette thèse consiste ainsi à mettre en évidence l’action protectrice de l’ODN sur la survie des neurones DA de la SNpc dans un modèle murin de la MP et à rechercher les conséquences de l’invalidation du gène du précurseur de l’ODN (DBI) sur la vulnérabilité des neurones DA. Les résultats obtenus montrent qu’une seule injection intra-cérébroventriculaire d’une faible quantité d’ODN (10 ng), 1 h après la dernière administration systémique de 1-méthyl-4-phényl-1,2,3,6-tétrahydropyridine (MPTP) prévient significativement la perte des neurones DA dans la substance noire et la dégénérescence de leurs prolongements nerveux vers le striatum comme mesuré par des marquages et des mesures d’expression de la tyrosine hydroxylase. Cet effet neuroprotecteur de l’ODN est accompagné par une réduction du nombre d’astrocytes réactifs, une forte inhibition de l'expression de gènes pro-inflammatoires tels que les interleukines (IL) IL-1β et IL-6, et tumor necrosis factor-α. De plus, l'ODN bloque l'inhibition du gène anti-apoptotique Bcl-2 et la stimulation des gènes pro-apoptotiques Bax et caspase-3, induite par le MPTP dans la SNpc et le striatum. L'ODN réduit également l’accumulation d'espèces réactives de l'oxygène (ROS) et de produits d'oxydation lipidique dans les neurones DA. Par ailleurs, les souris knock-out DBI (DBI-/-) sont plus vulnérables que les animaux sauvages (DBI+/+) vis-à-vis de la neurotoxicité du MPTP. L’absence de production d’ODN endogène, chez les souris DBI-/- parkinsoniennes, augmente les dommages cellulaires induits par le MPTP, la réactivité gliale, les taux de ROS, l’expression de cytokines pro-inflammatoires et l'activité de la caspase-3 dans la région nigro-striée. L’ensemble de ces résultats montre que le gliopeptide ODN exerce un puissant effet neuroprotecteur contre la dégénérescence des neurones DA de la SNpc induite par le MPTP, chez la souris. Cette action protectrice met en jeu des mécanismes impliquant l’inhibition des processus neuro-inflammatoires, oxydatifs et apoptotiques. D’autre part, la déficience en ODN potentialise les effets délétères du MPTP, suggérant que ce peptide joue un rôle clé lors de la réponse à un stress cellulaire

    Neuroprotection with the Endozepine Octadecaneuropeptide, ODN

    No full text
    International audienceThe term endozepines designates a family of astroglia-secreted proteins including the diazepambinding inhibitor (DBI) and its processing products, which have been originally isolated and characterized as endogenous ligands of benzodiazepine receptors. It is now clearly established that the octadecaneuropeptide ODN (DBI33-50), acting through the central-type benzodiazepine receptor or a metabotropic receptor, exerts important functions such as proconflict behavior, induction of anxiety, inhibition of pentobarbital-provoked sleep, decrease of water consumption and reduction of food intake. To mediate its effects, ODN regulates both glial cell and neuronal activities by acting on neurosteroid biosynthesis and/or neuropeptide expression. In addition, ODN stimulates astrocyte proliferation and protects both neurons and astrocytes from oxidative stress-induced cell death. The antiapoptotic effect of ODN on neural cells is mediated through activation of the ODN metabotropic receptor positively coupled to PKA, PKC and MAPK/ERK transduction pathways, which ultimately reduces the pro-apoptotic gene Bax and stimulates Bcl-2 expressions, and inhibits intracellular reactive oxygen species accumulation. The imbalance in favor of Bcl2 promotes mitochondria functions and blocks in turn caspases activation while at the same time, ODN also activates the endogenous antioxidant system i.e. glutathione biosynthesis, and expression and activities of antioxidant enzymes. In cultured astrocytes, DBI expression is up-regulated during moderate oxidative stress, and authentic ODN production is increased, suggesting that ODN may act as a paracrine factor protecting neighboring neurons. Taken together, the remarkable effect of ODN on the apoptotic cascade suggests that innovative ODN derivatives could potentially be useful for treatment of cerebral injuries involving oxidative stress and neurodegeneration

    Involvement of endogenous antioxidant systems in the protective activity of pituitary adenylate cyclase-activating polypeptide against hydrogen peroxide-induced oxidative damages in cultured rat astrocytes.

    No full text
    International audienceAstroglial cells possess an array of cellular defense mechanisms, including superoxide dismutase (SOD) and catalase antioxidant enzymes, to prevent damages caused by oxidative stress. Nevertheless, astroglial cell viability and functionality can be affected by significant oxidative stress. We have previously shown that pituitary adenylate cyclase-activating polypeptide (PACAP) is a potent glioprotective agent that prevents hydrogen peroxide (H2 O2 )-induced apoptosis in cultured astrocytes. The purpose of this study was to investigate the potential protective effect of PACAP against oxidative-generated alteration of astrocytic antioxidant systems. Incubation of cells with subnanomolar concentrations of PACAP inhibited H2 O2 -evoked reactive oxygen species accumulation, mitochondrial respiratory burst, and caspase-3 mRNA level increase. PACAP also stimulated SOD and catalase activities in a concentration-dependent manner, and counteracted the inhibitory effect of H2 O2 on the activity of these two antioxidant enzymes. The protective action of PACAP against H2 O2 -evoked inhibition of antioxidant systems in astrocytes was protein kinase A, PKC, and MAP-kinase dependent. In the presence of H2 O2 , the SOD blocker NaCN and the catalase inhibitor 3-aminotriazole, both suppressed the protective effects of PACAP on SOD and catalase activities, mitochondrial function, and cell survival. Taken together, these results indicate that the anti-apoptotic effect of PACAP on astroglial cells can account for the activation of endogenous antioxidant enzymes and reduction in respiration rate, thus preserving mitochondrial integrity and preventing caspase-3 expression provoked by oxidative stress. Considering its powerful anti-apoptotic and anti-oxidative properties, the PACAPergic signaling system should thus be considered for the development of new therapeutical approaches to cure various pathologies involving oxidative neurodegeneration. We propose the following cascade for the glioprotective action of Pituitary adenylate cyclase-activating polypeptide (PACAP) against H2 O2 -induced astrocyte damages and cell apoptosis in cultured rat astrocytes. PACAP, through activation of its receptor, PAC1-R, and the protein kinase A (PKA), protein kinase C (PKC), and MAP-kinases signaling pathways, prevents accumulation of ROS and inhibition of SOD and catalase activities. This allows the preservation of mitochondrial membrane integrity and the reduction in caspase-3 activation induced by H2 O2 . These data may lead to the development of new strategies for cerebral injury treatment. Cat, catalase; Cyt. C, cytochrome C; SOD, superoxide dismutase

    Pituitary adenylate cyclase-activating polypeptide protects astroglial cells against oxidative stress-induced apoptosis

    No full text
    International audienceOxidative stress, associated with a variety of disorders including neurodegenerative diseases, results from accumulation of reactive oxygen species (ROS). Oxidative stress is not only responsible for neuron apoptosis, but can also provoke astroglial cell death. Numerous studies indicate that pituitary adenylate cyclase-activating polypeptide (PACAP) promotes neuron survival, but nothing is known regarding the action of PACAP on astroglial cell survival. Thus, the purpose of the present study was to investigate the potential glioprotective effect of PACAP on H(2)O(2)-induced astrocyte death. Pre-treatment of cultured rat astrocytes with nanomolar concentrations of PACAP prevented cell death provoked by H(2)O(2) (300 ÎĽM), whereas vasoactive intestinal polypeptide was devoid of protective activity. The effect of PACAP on astroglial cell survival was abolished by the type 1 PACAP receptor antagonist, PACAP6-38. The protective action of PACAP was blocked by the protein kinase A inhibitor H89, the protein kinase C inhibitor chelerythrine and the mitogen-activated protein (MAP)-kinase kinase (MEK) inhibitor U0126. PACAP stimulated glutathione formation, and blocked H(2)O(2)-evoked ROS accumulation and glutathione content reduction. In addition, PACAP prevented the decrease of mitochondrial activity and caspase 3 activation induced by H(2)O(2). Taken together, these data indicate for the first time that PACAP, acting through type 1 PACAP receptor, exerts a potent protective effect against oxidative stress-induced astrocyte death. The anti-apoptotic activity of PACAP on astrocytes is mediated through the protein kinase A, protein kinase C and MAPK transduction pathways, and can be accounted for by inhibition of ROS-induced mitochondrial dysfunctions and caspase 3 activation
    corecore