977 resultados para Receptor Neurons
Resumo:
Brain derived neurotrophic factor (BDNF) is a member of the family of neurotrophins and binds to the tropomyosin-related kinase B (TrkB) receptor. Like other neurotrophic factors, BDNF is involved in the development and differentiation of neurons. Recently, studies have suggested important roles for BDNF in the regulation of energy homeostasis. The paraventricular nucleus (PVN) is critical for normal energy balance contains high levels of both BDNF and TrkB mRNA. Studies have shown that microinjections of BDNF into the PVN increase energy expenditure, suggesting BDNF plays a role in energy homeostasis through direct actions in this hypothalamic nucleus. We used male Sprague-Dawley rats to perform whole-cell current-clamp experiments from PVN neurons in slice preparation. BDNF was bath applied at a concentration of 2nM and caused depolarizations in 54% of neurons (n = 25; mean change in membrane potential: 8.9 ± 1.2 mV), hyperpolarizations in 23% (n = 11; mean change in membrane potential: -6.7 ± 1.4 mV), while the remaining cells tested were unaffected. Previous studies showing effects of BDNF on γ-aminobutyric acid type A (GABAA) mediated neurotransmission in PVN led us to examine if these BDNF-mediated changes in membrane potential were maintained in the presence of tetrodotoxin (TTX) sodium channel blocker (N = 9; 56% depolarized, 22% hyperpolarized, 22% non-responders) and bicuculline (GABAA antagonist) (N = 12; 42% depolarized, 17% hyperpolarized, 41% non-responders), supporting the conclusion that these effects on membrane potential were postsynaptic. We also evaluated the effects of BDNF on these neurons across varying physiologically relevant extracellular glucose concentrations. At 10 mM 23% (n = 11; mean: -6.7 ± 1.4 mV) of PVN neurons hyperpolarized in response to BDNF treatment, whereas at 0.2 mM glucose, 71% showed hyperpolarizing effects (n = 12; mean: -6.3 ± 2.8 mV). Our findings reveal that BDNF has direct impacts on PVN neurons and that these neurons are capable of integrating multiple sources of metabolically relevant input. Our analysis regarding glucose concentrations and their effects on these neurons’ response to other metabolic signals emphasizes the importance of using physiologically relevant conditions for study of central pathways involved in the regulation of energy homeostasis.
Resumo:
The transient receptor potential melastatin 8 (TRPM8) channel has been characterized as a cold and menthol receptor expressed in a subpopulation of sensory neurons but was recently identified in other tissues, including the respiratory tract, urinary system, and vasculature. Thus TRPM8 may play multiple functional roles, likely to be in a tissue- and activation state-dependent manner. We examined the TRPM8 channel presence in large arteries from rats and the functional consequences of their activation. We also aimed to examine whether these channels contribute to control of conscious human skin blood flow. TRPM8 mRNA and protein were detected in rat tail, femoral and mesenteric arteries, and thoracic aorta. This was confirmed in single isolated vascular myocytes by immunocytochemistry. Isometric contraction studies on endothelium-denuded relaxed rat vessels found small contractions on application of the TRPM8-specific agonist menthol (300 microM). However, both menthol and another agonist icilin (50 microM) caused relaxation of vessels precontracted with KCl (60 mM) or the alpha-adrenoceptor agonist phenylephrine (2 microM) and a reduction in sympathetic nerve-mediated contraction. These effects were antagonized by bromoenol lactone treatment, suggesting the involvement of Ca(2+)-independent phospholipase A(2) activation in TRPM8-mediated vasodilatation. In thoracic aorta with intact endothelium, menthol-induced inhibition of KCl-induced contraction was enhanced. This was unaltered by preincubation with either N(omega)-nitro-l-arginine methyl ester (l-NAME; 100 nM), a nitric oxide synthase inhibitor, or the ACh receptor antagonist atropine (1 microM). Application of menthol (3% solution, topical application) to skin caused increased blood flow in conscious humans, as measured by laser Doppler fluximetry. Vasodilatation was markedly reduced or abolished by prior application of l-NAME (passive application, 10 mM) or atropine (iontophoretic application, 100 nM, 30 s at 70 microA). We conclude that TRPM8 channels are present in rat artery vascular smooth muscle and on activation cause vasoconstriction or vasodilatation, dependent on previous vasomotor tone. TRPM8 channels may also contribute to human cutaneous vasculature control, likely with the involvement of additional neuronal mechanisms.
Resumo:
Acetaminophen [N-acetyl-p-aminophenol (APAP)] is the most common antipyretic/analgesic medicine worldwide. If APAP is overdosed, its metabolite, N-acetyl-p-benzo-quinoneimine (NAPQI), causes liver damage. However, epidemiological evidence has associated previous use of therapeutic APAP doses with the risk of chronic obstructive pulmonary disease (COPD) and asthma. The transient receptor potential ankyrin-1 (TRPA1) channel is expressed by peptidergic primary sensory neurons. Because NAPQI, like other TRPA1 activators, is an electrophilic molecule, we hypothesized that APAP, via NAPQI, stimulates TRPA1, thus causing airway neurogenic inflammation. NAPQI selectively excites human recombinant and native (neuroblastoma cells) TRPA1. TRPA1 activation by NAPQI releases proinflammatory neuropeptides (substance P and calcitonin gene-related peptide) from sensory nerve terminals in rodent airways, thereby causing neurogenic edema and neutrophilia. Single or repeated administration of therapeutic (15-60 mg/kg) APAP doses to mice produces detectable levels of NAPQI in the lung, and increases neutrophil numbers, myeloperoxidase activity, and cytokine and chemokine levels in the airways or skin. Inflammatory responses evoked by NAPQI and APAP are abated by TRPA1 antagonism or are absent in TRPA1-deficient mice. This novel pathway, distinguished from the tissue-damaging effect of NAPQI, may contribute to the risk of COPD and asthma associated with therapeutic APAP use.-Nassini, R., Materazzi, S., Andre, E., Sartiani, L., Aldini, G., Trevisani, M., Carnini, C., Massi, D., Pedretti, P., Carini, M., Cerbai, E., Preti, D., Villetti, G., Civelli, M., Trevisan, G., Azzari, C., Stokesberry, S., Sadofsky, L., McGarvey, L., Patacchini, R., Geppetti, P. Acetaminophen, via its reactive metabolite N-acetyl-p-benzo-quinoneimine and transient receptor potential ankyrin-1 stimulation causes neurogenic inflammation in the airways and other tissues in rodents. FASEB J. 24, 4904-4916 (2010). www.fasebj.org
Resumo:
One important mechanism of membrane ion channels regulation involves their non-functional isoforms generated by alternative splicing. However, knowledge of such isoforms for the members of transient receptor potential (TRP) superfamily of ion channels remains quite limited. This study focuses on TRPM member, TRPM8, which functions as a cold receptor in sensory neurons, but is also expressed in tissues not exposed to ambient temperatures, as well as in cancer tissues. We report the cloning from prostate cancer cells of new short-splice variants of TRPM8, termed short TRPM8a (sM8a) and short TRPM8ß (sM8ß). Our results show that both variants are in a closed configuration with the C-terminal tail of the full-size TRPM8 chan-nel, resulting in stabilization of its closed state and thus reducing both its cold sensitivity and its activity. Our findings, therefore, uncover a new mode of the regulation of TRPM8 channel by its splice variants.
Resumo:
Introduction: Transient receptor potential (TRP) channels are widely, but not uniformly, distributed in tissues. To date the dominant focus of attention has been on TRP expression and functionality in neurons. However, their expression and activation in selected non-neuronal cells suggest TRPs have a potential role in coordinating cross-talk during the inflammatory process. Fibroblasts comprise the major cell type in the dental pulp and play an important role in pulpal inflammation. Objectives: The aim of this study was to investigate the expression and functionality of the TRP channels TRPA1, TRPM8, TRPV4 and TRPV1 in human dental pulp fibroblasts. Methods: Dental pulp fibroblasts were derived by explant culture of pulps removed from extracted healthy teeth. Fibroblasts were cultured in DMEM supplemented with 10% FCS, 100U/ml penicillin and 100µg/ml streptomycin. Protein expression of TRP channels was investigated by SDS- polyacrylamide gel electrophoresis and Western blotting of cell lysates from fibroblast cells in culture. TRPA1, TRPM8, TRPV4 and TRPV1 expression was determined by specific antibodies, detected using appropriate anti-species antibodies and chemiluminescence. Functionality of TRP channels was determined by Ca2+ microfluorimetry. Cells were grown on cover slips and incubated with Fura 2AM prior to stimulation with icilin (TRPA1 agonist), menthol (TRPM8 agonist), 4 alpha-phorbol 12,13-didecanoate (4alphaPDD) (TRPV4 agonist) or capsaicin (TRPV1 agonist). Emitted fluorescence (F340/F380) was used to determine intracellular [Ca2+] levels. Results: Fibroblast expression of TRPA1, TRPM8, TRPV4 and TRPV1 was confirmed at the protein level by Western blotting. Increased intracellular [Ca2+] levels in response to icillin, methanol, 4alphaPDD and capsacin, indicated functional expression of TRPA1, TRPM8, TRPV4 and TRPV respectively. Conclusions: The presence and functionality of TRP channels on dental pulp fibroblasts suggests a potential role for these cells in the pulpal neurogenic inflammatory response. (Supported by a research grant from the Royal College of Surgeons of Edinburgh).
Resumo:
Tese de mestrado, Neurociências, Faculdade de Medicina, Universidade de Lisboa, 2016
Resumo:
The neuronal-specific cholesterol 24S-hydroxylase (CYP46A1) is important for brain cholesterol elimination. Cyp46a1 null mice exhibit severe deficiencies in learning and hippocampal long-term potentiation, suggested to be caused by a decrease in isoprenoid intermediates of the mevalonate pathway. Conversely, transgenic mice overexpressing CYP46A1 show an improved cognitive function. These results raised the question of whether CYP46A1 expression can modulate the activity of proteins that are crucial for neuronal function, namely of isoprenylated small guanosine triphosphate-binding proteins (sGTPases). Our results show that CYP46A1 overexpression in SH-SY5Y neuroblastoma cells and in primary cultures of rat cortical neurons leads to an increase in 3-hydroxy-3-methyl-glutaryl-CoA reductase activity and to an overall increase in membrane levels of RhoA, Rac1, Cdc42 and Rab8. This increase is accompanied by a specific increase in RhoA activation. Interestingly, treatment with lovastatin or a geranylgeranyltransferase-I inhibitor abolished the CYP46A1 effect. The CYP46A1-mediated increase in sGTPases membrane abundance was confirmed in vivo, in membrane fractions obtained from transgenic mice overexpressing this enzyme. Moreover, CYP46A1 overexpression leads to a decrease in the liver X receptor (LXR) transcriptional activity and in the mRNA levels of ATP-binding cassette transporter 1, sub-family A, member 1 and apolipoprotein E. This effect was abolished by inhibition of prenylation or by co-transfection of a RhoA dominant-negative mutant. Our results suggest a novel regulatory axis in neurons; under conditions of membrane cholesterol reduction by increased CYP46A1 expression, neurons increase isoprenoid synthesis and sGTPase prenylation. This leads to a reduction in LXR activity, and consequently to a decrease in the expression of LXR target genes.
Resumo:
Summary : The hypothalamus represents less than 1 % of the total volume of the brain tissue, yet it plays a crucial role in endocrine regulations. Puberty is defined as a process leading to physical, sexual and psychosocial maturation. The hypothalamus is central to this process, via the activation of GnRH neurons. Pulsatile GnRH secretion, minimal during childhood, increases with the onset of puberty. The primary function of GnRH is to regulate the growth, development and function of testes in boys and ovaries in girls, by stimulating the pituitary gland secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Several factors contribute to the timing of puberty, including sex and ethnicity, genetics, dietary intake and energy expenditure. Kisspeptins constitute a family of small peptides arising from the proteolytic cleavage of metastin, a peptide with 54 amino acids initially purified from human placenta. These kisspeptins were the subject of much attention following their discovery because of their antimetastatic properties, but it was more recently that their determining role in the reproductive function was demonstrated. It was shown that kisspeptins are ligands of a receptor, GPR54, whose natural inactivating mutation in humans, or knockout in the mouse, lead to infertility. GnRH neurons play a pivotal role in the central regulation of fertility. Kisspeptin greatly increases GnRH release and GnRH neuron firing activity, but the neurobiological mechanisms for these actions are unknown. Gprotein-coupled receptor 54, the receptor for kisspeptin, is expressed by GnRH neurons as well as other hypothalamic neurons, suggesting that both direct and indirect effects are possible. In the first part of my thesis, we investigated a possible connection between the acceleration of sexual development induced by leptin and hypothalamic metastin neurons. However, the data generated by our preliminary experiments confirmed that the commercially available antibodies are non-specific. This finding constituted a major drawback for our studies, which relied heavily upon the neuroanatomical study of the hypothalamic metastinergic pathways to elucidate their sensitivity to exogenous leptin. Therefore, we decided to postpone any further in vivo experiment until a better antibody becomes available, and focused on in vitro studies to better understand the mechanisms of action of kisspeptins in the modulation of the activity of GnRH neurons. We used two GnRH-expressing neuronal cell lines to investigate the cellular and molecular mechanisms of action of metastin in GnRH neurons. We demonstrated that kisspeptin induces an early activation of the MAP kinase intracellular signaling pathway in both cell lines, whereas the SAP/JNK or the Akt pathways were unaffected. Moreover, we found an increase in GnRH mRNA levels after 6h of metastin stimulation. Thus, we can conclude that kisspeptin regulates GnRH neurons both at the secretion and the gene expression levels. The MAPK pathway is the major pathway activated by metastin in GnRH expressing neurons. Taken together, these data provide the first mechanism of action of kisspeptin on GnRH neurons. Résumé : L'hypothalamus est une zone située au centre du cerveau, dont il représente moins de 1 du volume total. La puberté est la période de transition entre l'enfance et l'age adulte, qui s'accompagne de transformations somatiques, psychologiques, métaboliques et hormonales conduisant à la possibilité de procréer. La fonction principale de la GnRH est la régulation de la croissance, du développement et de la fonction des testicules chez les hommes, et des ovaires chez les femmes en stimulant la sécrétion de l'hormone lutéinisante (LH) et de l'hormone folliculostimulante (FSH) par la glande hypophysaire. Plusieurs facteurs contribuent au déclanchement de la puberté, y compris le sexe et l'appartenance ethnique, la génétique, l'apport alimentaire et la dépense énergétique. Les Kisspeptines constituent une famille de peptides résultant de la dissociation proteolytique de la métastine, un peptide de 54 acides aminés initialement purifié à partir de placenta humain. Ces kisspeptines ont fait l'objet de beaucoup d'attention à la suite de leur découverte en raison de leurs propriétés anti-metastatiques, et c'est plus récemment que leur rôle déterminant dans la fonction reproductive a été démontré. Les kisspeptines sont des ligands du récepteur GPR54, dont la mutation inactivatrice chez l'homme, ou le knockout chez la souris, conduisent à l'infertilité par hypogonadisme hypogonadotrope. Les neurones à GnRH jouent un rôle central dans le règlement des fonctions reproductrices et la kisspeptine stimule l'activité des neurones à GnRH et la libération de GnRH par ces neurones. Toutefois, les mécanismes neurobiologiques de ces actions ne sont pas connus. Dans la première partie de ma thèse, nous avons étudié le lien potentiel entre l'accélération du développement sexuel induite par la leptine et les neurones hypothalamiques à metastine. Les données générées dans cette première série d'expériences ont malheureusement confirmé que les anticorps anti-metastine disponibles dans le commerce sont aspécifiques. Ceci a constitué un inconvénient majeur pour nos études, qui devaient fortement s'appuyer sur l' étude neuroanatomique des neurones hypothalamiques à metastine pour évaluer leur sensibilité à la leptine exogène. Nous avons donc décidé de focaliser nos travaux sur une étude in vitro des mécanismes d'action de la kisspeptine pour moduler l'activité des neurones à GnRH. Nous avons utilisé deux lignées de cellules neuronales exprimant la GnRH pour étudier les mécanismes d'action cellulaires et moléculaires de la metastine dans des neurones. Nous avons ainsi pu démontrer que la kisspeptine induit une activation précoce de la voie f de signalisation de la MAP kinase dans les deux lignées cellulaires, alors que nous n'avons observé aucune activation de la voie de signalisation de la P13 Kinase et de la SAP/JNK. Nous avons en outre démontré une augmentation de l'expression de la GnRH par la stimulation avec la Kisspeptine. L'ensemble de ces données contribue à élucider le mécanisme d'action avec lequel la kisspeptine agit dans les neurones à GnRH, en démontrant un effet sur l'expression génique de la GnRH. Nous pouvons également conclure que la voie de la MAPK est la voie principale activée par la metastine dans les neurones exprimant la GnRH.
Resumo:
NMDA receptors (NMDARs) mediate ischemic brain damage, for which interactions between the C termini of NR2 subunits and PDZ domain proteins within the NMDAR signaling complex (NSC) are emerging therapeutic targets. However, expression of NMDARs in a non-neuronal context, lacking many NSC components, can still induce cell death. Moreover, it is unclear whether targeting the NSC will impair NMDAR-dependent prosurvival and plasticity signaling. We show that the NMDAR can promote death signaling independently of the NR2 PDZ ligand, when expressed in non-neuronal cells lacking PSD-95 and neuronal nitric oxide synthase (nNOS), key PDZ proteins that mediate neuronal NMDAR excitotoxicity. However, in a non-neuronal context, the NMDAR promotes cell death solely via c-Jun N-terminal protein kinase (JNK), whereas NMDAR-dependent cortical neuronal death is promoted by both JNK and p38. NMDAR-dependent pro-death signaling via p38 relies on neuronal context, although death signaling by JNK, triggered by mitochondrial reactive oxygen species production, does not. NMDAR-dependent p38 activation in neurons is triggered by submembranous Ca(2+), and is disrupted by NOS inhibitors and also a peptide mimicking the NR2B PDZ ligand (TAT-NR2B9c). TAT-NR2B9c reduced excitotoxic neuronal death and p38-mediated ischemic damage, without impairing an NMDAR-dependent plasticity model or prosurvival signaling to CREB or Akt. TAT-NR2B9c did not inhibit JNK activation, and synergized with JNK inhibitors to ameliorate severe excitotoxic neuronal loss in vitro and ischemic cortical damage in vivo. Thus, NMDAR-activated signals comprise pro-death pathways with differing requirements for PDZ protein interactions. These signals are amenable to selective inhibition, while sparing synaptic plasticity and prosurvival signaling.
Resumo:
Adult neurogenesis is regulated by the neurogenic niche, through mechanisms that remain poorly defined. Here, we investigated whether niche-constituting astrocytes influence the maturation of adult-born hippocampal neurons using two independent transgenic approaches to block vesicular release from astrocytes. In these models, adult-born neurons but not mature neurons showed reduced glutamatergic synaptic input and dendritic spine density that was accompanied with lower functional integration and cell survival. By taking advantage of the mosaic expression of transgenes in astrocytes, we found that spine density was reduced exclusively in segments intersecting blocked astrocytes, revealing an extrinsic, local control of spine formation. Defects in NMDA receptor (NMDAR)-mediated synaptic transmission and dendrite maturation were partially restored by exogenous D-serine, whose extracellular level was decreased in transgenic models. Together, these results reveal a critical role for adult astrocytes in local dendritic spine maturation, which is necessary for the NMDAR-dependent functional integration of newborn neurons.
Resumo:
Retinoic acid, a derivative of vitamin A, is known to play diverse roles in development and regeneration. Previous research in the mollusc Lymnaea stagnalis has shown that a gradient of all-trans retinoic acid attracts the growth cones of cultured neurons. The present study investigates the sub-cellular mechanisms within the growth cones of Lymnaea pedal A neurons which mediate the attractive response to a gradient of alltrans retinoic acid. In this study, the mechanism of growth cone turning is shown to be local, as neurites mechanically isolated from their cell body retain the capacity to turn towards an exogenous gradient of all-trans retinoic acid. The turning response is dependent on the initiation of protein synthesis and calcium influx, but does not appear to involve signaling through protein kinase C (PKC). The retinoid X receptor (RXR), which classically functions as a transcription factor, was also shown to be involved in the turning response, functioning locally through a non-genomic pathway. These data show, for the first time in any species, that all-trans retinoic acid's chemotropic action involves a local mechanism involving non-genomic signaling through the RXR. As retinoic acid is known to playa role in regeneration, understanding the mechanisms underlying retinoic acid signaling may lead to further advances in regenerative neuroscience.
Resumo:
La leptine circule en proportion de la masse graisseuse du corps et la transduction de son signal à travers la forme longue de son récepteur via un certain nombre de voies neurales , y compris MAPK, PI3-K ,AMPK et JAK2 - STAT3 . Il faut noter que STAT3 constitue une voie clée au récepteur de la leptine par laquelle la leptine module l'expression des gènes impliqués dans la régulation du bilan énergétique. La plupart des recherches ont porté sur la fonction du récepteur de la leptine au sein de l' hypothalamus, en particulier la fonction du récepteur de la leptine dans le noyau arqué. Toutefois, les récepteurs de la leptine sont également exprimés sur les neurones dopaminergiques de l'aire tégmentale ventrale et la leptine agit sur cette région du cerveau pour influencer la prise alimentaire, la motivation, la locomotion, l'anxiété et la transmission de la dopamine. De plus, la leptine active la STAT3 dans les dopaminergiques et GABAergiques populations neuronales. Bien que ces résultats contribuent à notre compréhension des multiples actions de la leptine dans le système nerveux central, il reste à résoudre les cellules et la signalisation du récepteur de la leptine qui sont responsables des effets neurocomportementaux de la leptine dans le mésencéphale. Visant à déterminer la contribution de la voie de signalisation STAT3 dans les neurones dopaminergiques du mésencéphale, nous avons généré une lignée de souris knockout conditionnel dans lequel l'activation du gène de STAT3 sur son résidu tyrosine 705 ( Tyr 705 ) est absent spécifiquement dans les neurones dopaminergiques. Avec l'utilisation de ce modèle de souris génétique, nous avons évalué l'impact de l'ablation de la signalisation STAT3 dans les neurones dopaminergiques sur un certain nombre de fonctions liées à la dopamine, y compris l'alimentation, la locomotion, les comportements liés à la récompense, l'émotion et la libération de dopamine dans le noyau accumbens. Fait intéressant, nous avons observé un dimorphisme sexuel dans le phénotype des souris STAT3DAT-KO. L'activation de la voie de signalisation STAT3 dans les neurones dopaminergiques est responsable de l'action de la leptine dans la réduction de la locomotion, récompense liée à l'activité physique, et de l'augmentation de la libération et de la disponibilité de la dopamine chez les souris mâles. Cependant, il ne module pas le comportement émotionnel. D'autre part, les souris femelles STAT3DAT-KO augmentent les niveaux d'anxiété et les niveaux plasmatiques de corticostérone, sans provoquer de changements de la dépression. Cependant, la perte d'activation de STAT3 dans les neurones dopaminergiques ne module pas le comportement locomoteur chez les souris femelles. Notamment, les actions de la leptine dans le mésencéphale pour influencer le comportement alimentaire ne sont pas médiées par l'activation de STAT3 dans les neurones dopaminergiques, considérant que les souris mâles et femelles ont un comportement alimentaire normal. Nos résultats démontrent que la voie de signalisation STAT3 dans les neurones dopaminergiques est responsable des effets anxiolytiques de la leptine, et soutient l'hypothèse que la leptine communique l'état d'énergie du corps (i.e. la relation entre la dépense et les apports énergétiques) pour les régions mésolimbiques pour atténuer les effets de motivation et de récompense de plusieurs comportements qui servent à réhabiliter ou à épuiser les réserves d'énergie. En outre, ce travail souligne l'importance d'étudier la modulation de la signalisation de la leptine dans différente types de cellules, afin d'identifier les voies de signalisation et les mécanismes cellulaires impliqués dans les différentes fonctions neuro-comportementales de la leptine.
Resumo:
L’aire tegmentaire ventrale (VTA) contient une forte densité de terminaisons neurotensinergiques ainsi que des récepteurs à la surface des neurones dopaminergiques et non-dopaminergiques. Le VTA a été impliqué dans des maladies comme la schizophrénie, les psychoses et l’abus de substance. Les drogues d’abus sont connues pour induire le phénomène de sensibilisation - un processus de facilitation par lequel l’exposition à un stimulus produit une réponse augmentée lors de l’exposition subséquente au même stimulus. La sensibilisation se développe dans le VTA et implique mécanismes dopaminergiques et glutamatergiques. Il a été montré que les antagonistes neurotensinergiques bloquaient le développement de la sensibilisation et certains mécanismes de récompense et ces effets pourraient être médiés indirectement par une modulation de la neurotransmission glutamatergique. Cependant, on connaît peu les mécanismes de modulation de la transmission glutamatergique par la neurotensine (NT) dans le VTA. Le but de la présente thèse était d’étudier la modulation neurotensinergique de la neurotransmission glutamatergique dans les neurones dopaminergiques et non-dopaminergiques du VTA. Pour ce faire, nous avons utilisé la technique du patch clamp dans la cellule entière dans des tranches horizontales du VTA pour étudier les effets de différents agonistes et antagonistes neurotensinergiques. Les neurones ont été identifié comme Ih+ (présumés dopaminergiques) ou Ih- (présumés non-dopaminergiques) selon qu’ils exprimaient ou non un courant cationique activé par l’hyperpolarisation (Ih). Des techniques d’immunocytochimie ont été utilisées pour marquer les neurones et vérifier leur localisation dans le VTA. Dans une première étude nous avons trouvé que la neurotensine indigène (NT1-13) ou son fragment C-terminal, NT8-13, induisait une augmentation comparable des courants postsynaptiques excitateurs glutamatergiques (CPSEs) dans les neurones Ih+ ou Ih- du VTA. L'augmentation induite dans les neurones Ih+ par la NT8-13 a été bloquée par le SR48692, un antagoniste des récepteurs NTS1, et par le SR142948A, un antagoniste des récepteurs NTS1 et NTS2, suggérant que l'augmentation était médiée par l’activation des récepteurs NTS1. Dans les neurones Ih- l'augmentation n’a été bloquée que par le SR142948A indiquant une implication des récepteurs NTS2. Dans une deuxième étude, nous avons testé les effets de la D-Tyr[11]NT (un analogue neurotensinergique ayant différentes affinités de liaison pour les sous-types de récepteurs neurotensinergiques) sur les CPSEs glutamatergiques dans les neurones Ih+ et Ih- en parallèle avec une série d’expériences comportementales utilisant un paradigme de préférence de place conditionnée (PPC) menée dans le laboratoire de Pierre-Paul Rompré. Nous avons constaté que la D-Tyr[11]NT induisaient une inhibition dépendante de la dose dans les neurones Ih+ médiée par l'activation de récepteurs NTS2. En revanche, la D-Tyr[11]NT a produit une augmentation des CPSEs glutamatergiques médiée par des récepteurs NTS1 dans les neurones Ih-. Les résultats des expériences comportementales ont montré que des microinjections bilatérales de D-Tyr[11]NT dans le VTA induisait une PPC bloquée uniquement par la co-injection de SR142948A et SR48692, indiquant un rôle pour les deux types de récepteurs, NTS1 et NTS2. Cette étude nous a permis de conclure que i) la D-Tyr[11]NT agit dans le VTA via des récepteurs NTS1 et NTS2 pour induire un effet de récompense et ii) que cet effet est dû, au moins en partie, à une augmentation de la neurotransmission glutamatergique dans les neurones non-dopaminergiques (Ih-). Dans une troisième étude nous nous sommes intéressés aux effets de la D-Tyr[11]NT sur les réponses isolées médiées par les récepteurs N-méthyl-D-aspartate (NMDA) et acide α-amino-3- hydroxy-5-méthyl-4-isoxazolepropionique (AMPA) dans les neurones du VTA. Nous avons constaté que dans les neurones Ih+ l’amplitude des CPSEs NMDA et AMPA étaient atténuées de la même manière par la D-Tyr[11] NT. Cette modulation des réponses était médiée par les récepteurs NTS1 et NTS2. Au contraire, dans les neurones Ih-, l’amplitude des réponses NMDA et AMPA étaient augmentées en présence de D-Tyr[11]NT et ces effets dépendaient de l’activation des récepteurs NTS1 localisés sur les terminaisons glutamatergiques. Ces résultats fournissent une preuve supplémentaire que le NT exerce une modulation bidirectionnelle sur la neurotransmission glutamatergique dans les neurones du VTA et met en évidence un nouveau type de modulation peptidergique des neurones non-dopaminergiques qui pourrait être impliqué dans la sensibilisation. En conclusion, la modulation neurotensinergique de la neurotransmission glutamatergique dans les neurones dopaminergiques et non-dopaminergiques du VTA se fait en sens opposé soit, respectivement, par une inhibition ou par une excitation. De plus, ces effets sont médiés par différents types de récepteurs neurotensinergiques. En outre, nos études mettent en évidence une modulation peptidergique de la neurotransmission glutamatergique dans le VTA qui pourrait jouer un rôle important dans les mécanismes de lutte contre la toxicomanie.
Resumo:
In the present study, the effects of 5-HT, GABA and Bone Marrow Cells infused intranigrally to substantia nigra individually and in combinations on unilateral rotenone infused Parkinsonism induced rats. Scatchard analysis of DA, DA D1 and D2 receptors in the corpus striatum, cerebral cortex, cerebellum, brain stem and hippocampus showed a significant increase in the Brain regions of rotenone infused rat compared to control. Real Time PCR amplification of DA D1, D2, Bax and ubiquitin carboxy-terminal hydrolase were up regulated in the brain regions of rotenone infused rats compared to control. Gene expression studies of -Synuclien, cGMP and Cyclic AMP response element-binding protein showed a significant down regulation in Rotenone infused rats compared to control. Behavioural studies were carried out to confirm the biochemical and molecular studies.Our study demonstrated that BMC administration alone cannot reverse the above said molecular changes occurring in PD rat. 5-HT and GABA acting through their specific receptors in combination with bone marrow cells play a crucial role in the functional recovery of PD rats. 5-HT, GABA and Bone marrow cells treated PD rats showed significant reversal to control in DA receptor binding and gene expression. 5-HT and GABA have co-mitogenic property. Proliferation and differentiation of cells re-establishing the connections in Parkinson's disease facilitates the functional recovery. Thus, it is evident that 5-HT and GABA along with BMC to rotenone infused rats renders protection against oxidative, related motor and cognitive deficits which makes them clinically significant for cellbased therapy. The BMC transformed to neurons when co-transplanted with 5-HT and GABA which was confirmed with PKH2GL and nestin. These newly formed neurons have functional significance in the therapeutic recovery of Parkinson’s disease.
Resumo:
Parkinson's disease is a chronic progressive neurodegenerative movement disorder characterized by a profound and selective loss of nigrostriatal dopaminergic neurons. Our findings demonstrated that glutamatergic system is impaired during PD. The evaluations of these damages have important implications in understanding the molecular mechanism underlying motor, cognitive and memory deficits in PD. Our results showed a significant increase of glutamate content in the brain regions of 6- OHDA infused rat compared to control. This increased glutamate content caused an increase in glutamatergic and NMDA receptors function. Glutamate receptor subtypes- NMDAR1, NMDA2B and mGluR5 have differential regulatory role in different brain regions during PD. The second messenger studies confirmed that the changes in the receptor levels alter the IP3, cAMP and cGMP content. The alteration in the second messengers level increased the expression of pro-apoptotic factors - Bax and TNF-α, intercellular protein - α-synuclein and reduced the expression of transcription factor - CREB. These neurofunctional variations are the key contributors to motor and cognitive abnormalities associated with PD. Nestin and GFAP expression study confirmed that 5-HT and GABA induced the differentiation and proliferation of the BMC to neurons and glial cells in the SNpc of rats. We also observed that activated astrocytes are playing a crucial role in the proliferation of transplanted BMC which makes them significant for stem cell-based therapy. Our molecular and behavioural results showed that 5-HT and GABA along with BMC potentiates a restorative effect by reversing the alterations in glutamate receptor binding, gene expression and behaviour abnormality that occur during PD. The therapeutic significance in Parkinson’s disease is of prominence.