Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
2.
Neurotherapeutics ; 20(6): 1682-1691, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37823970

RÉSUMÉ

Neurological disorders represent some of the most challenging therapeutic areas for successful drug approvals. The escalating global burden of death and disability for such diseases represents a significant worldwide public health challenge, and the rate of failure of new therapies for chronic progressive disorders of the nervous system is higher relative to other non-neurological conditions. However, progress is emerging rapidly in advancing the drug development landscape in both rare and common neurodegenerative diseases. In October 2022, the Critical Path Institute (C-Path) and the US Food and Drug Administration (FDA) organized a Neuroscience Annual Workshop convening representatives from the drug development industry, academia, the patient community, government agencies, and regulatory agencies regarding the future development of tools and therapies for neurological disorders. This workshop focused on five chronic progressive diseases: Alzheimer's disease, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, and inherited ataxias. This special conference report reviews the key points discussed during the three-day dynamic workshop, including shared learnings, and recommendations that promise to catalyze future advancement of novel therapies and drug development tools.


Sujet(s)
Maladie de Huntington , Myopathie de Duchenne , Maladies du système nerveux , Maladie de Parkinson , Humains , Maladies du système nerveux/traitement médicamenteux , Maladie de Parkinson/traitement médicamenteux , Développement de médicament
3.
Lancet Neurol ; 21(7): 632-644, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35716693

RÉSUMÉ

The current research paradigm for Huntington's disease is based on participants with overt clinical phenotypes and does not address its pathophysiology nor the biomarker changes that can precede by decades the functional decline. We have generated a new research framework to standardise clinical research and enable interventional studies earlier in the disease course. The Huntington's Disease Integrated Staging System (HD-ISS) comprises a biological research definition and evidence-based staging centred on biological, clinical, and functional assessments. We used a formal consensus method that involved representatives from academia, industry, and non-profit organisations. The HD-ISS characterises individuals for research purposes from birth, starting at Stage 0 (ie, individuals with the Huntington's disease genetic mutation without any detectable pathological change) by using a genetic definition of Huntington's disease. Huntington's disease progression is then marked by measurable indicators of underlying pathophysiology (Stage 1), a detectable clinical phenotype (Stage 2), and then decline in function (Stage 3). Individuals can be precisely classified into stages based on thresholds of stage-specific landmark assessments. We also demonstrated the internal validity of this system. The adoption of the HD-ISS could facilitate the design of clinical trials targeting populations before clinical motor diagnosis and enable data standardisation across ongoing and future studies.


Sujet(s)
Maladie de Huntington , Évolution de la maladie , Humains , Maladie de Huntington/diagnostic , Maladie de Huntington/génétique , Études longitudinales , Phénotype
4.
J Huntingtons Dis ; 11(2): 97-104, 2022.
Article de Anglais | MEDLINE | ID: mdl-35466945

RÉSUMÉ

Huntington's disease (HD) is a devastating neurodegenerative disorder that urgently needs disease-modifying therapeutics. To this end, collaboration to standardize clinical research practices in the field and drive progress in addressing drug development challenges is paramount. At a meeting in 2017 organized by CHDI Foundation and the Critical Path Institute, stakeholders across the pharmaceutical industry, academia, regulatory agencies, and patient advocacy groups discussed the need for and potential impact of a consortium dedicated to HD regulatory science. Consequently, the Huntington's Disease Regulatory Science Consortium (HD-RSC) was formed, a precompetitive consortium that is dedicated to building a regulatory strategy to expedite the approval of HD therapeutics.


Sujet(s)
Maladie de Huntington , Humains , Protéine huntingtine , Maladie de Huntington/traitement médicamenteux
5.
Ther Innov Regul Sci ; 55(3): 591-600, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33398663

RÉSUMÉ

INTRODUCTION: Patient-level data sharing has the potential to significantly impact the lives of patients by optimizing and improving the medical product development process. In the product development setting, successful data sharing is defined as data sharing that is actionable and facilitates decision making during the development and review of medical products. This often occurs through the creation of new product development tools or methodologies, such as novel clinical trial design and enrichment strategies, predictive pre-clinical and clinical models, clinical trial simulation tools, biomarkers, and clinical outcomes assessments, and more. METHODS: To be successful, extensive partnerships must be established between all relevant stakeholders, including industry, academia, research institutes and societies, patient-advocacy groups, and governmental agencies, and a neutral third-party convening organization that can provide a pre-competitive space for data sharing to occur. CONCLUSIONS: Data sharing focused on identified regulatory deliverables that improve the medical product development process encounters significant challenges that are not seen with data sharing aimed at advancing clinical decision making and requires the commitment of all stakeholders. Regulatory data sharing challenges and solutions, as well as multiple examples of previous successful data sharing initiatives are presented and discussed in the context of medical product development.


Sujet(s)
Agences gouvernementales , Diffusion de l'information , Collecte de données , Humains
6.
Mol Psychiatry ; 26(6): 1808-1831, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-32071385

RÉSUMÉ

Maternal immune activation (MIA) disrupts the central innate immune system during a critical neurodevelopmental period. Microglia are primary innate immune cells in the brain although their direct influence on the MIA phenotype is largely unknown. Here we show that MIA alters microglial gene expression with upregulation of cellular protrusion/neuritogenic pathways, concurrently causing repetitive behavior, social deficits, and synaptic dysfunction to layer V intrinsically bursting pyramidal neurons in the prefrontal cortex of mice. MIA increases plastic dendritic spines of the intrinsically bursting neurons and their interaction with hyper-ramified microglia. Treating MIA offspring by colony stimulating factor 1 receptor inhibitors induces depletion and repopulation of microglia, and corrects protein expression of the newly identified MIA-associated neuritogenic molecules in microglia, which coalesces with correction of MIA-associated synaptic, neurophysiological, and behavioral abnormalities. Our study demonstrates that maternal immune insults perturb microglial phenotypes and influence neuronal functions throughout adulthood, and reveals a potent effect of colony stimulating factor 1 receptor inhibitors on the correction of MIA-associated microglial, synaptic, and neurobehavioral dysfunctions.


Sujet(s)
Microglie , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Comportement animal , Encéphale , Modèles animaux de maladie humaine , Femelle , Inflammation , Facteur de stimulation des colonies de macrophages , Souris , Neurones , Grossesse , Récepteur de facteur de croissance granulocyte-macrophage
7.
Clin Transl Sci ; 14(1): 214-221, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32702147

RÉSUMÉ

Interest in drug development for rare diseases has expanded dramatically since the Orphan Drug Act was passed in 1983, with 40% of new drug approvals in 2019 targeting orphan indications. However, limited quantitative understanding of natural history and disease progression hinders progress and increases the risks associated with rare disease drug development. Use of international data standards can assist in data harmonization and enable data exchange, integration into larger datasets, and a quantitative understanding of disease natural history. The US Food and Drug Administration (FDA) requires the use of Clinical Data Interchange Consortium (CDISC) Standards in new drug submissions to help the agency efficiently and effectively receive, process, review, and archive submissions, as well as to help integrate data to answer research questions. Such databases have been at the core of biomarker qualification efforts and fit-for-purpose models endorsed by the regulators. We describe the development of CDISC therapeutic area user guides for Duchenne muscular dystrophy and Huntington's disease through Critical Path Institute consortia. These guides describe formalized data structures and controlled terminology to map and integrate data from different sources. This will result in increased standardization of data collection and allow integration and comparison of data from multiple studies. Integration of multiple data sets enables a quantitative understanding of disease progression, which can help overcome common challenges in clinical trial design in these and other rare diseases. Ultimately, clinical data standardization will lead to a faster path to regulatory approval of urgently needed new therapies for patients.


Sujet(s)
Développement de médicament/normes , Échange d'informations de santé/normes , Maladie de Huntington/traitement médicamenteux , Myopathie de Duchenne/traitement médicamenteux , Maladies rares/traitement médicamenteux , Recherche biomédicale/normes , Bases de données factuelles/normes , Agrément de médicaments , Humains , Médicament orphelin/normes , États-Unis , Food and Drug Administration (USA)/normes
8.
Nat Commun ; 8(1): 1659, 2017 11 21.
Article de Anglais | MEDLINE | ID: mdl-29162813

RÉSUMÉ

The role of Arc in synaptic plasticity and memory consolidation has been investigated for many years with recent evidence that defects in the expression or activity of this immediate-early gene may also contribute to the pathophysiology of brain disorders including schizophrenia and fragile X syndrome. These results bring forward the concept that reversing Arc abnormalities could provide an avenue to improve cognitive or neurological impairments in different disease contexts, but how to achieve this therapeutic objective has remained elusive. Here, we present results from a chemogenomic screen that probed a mechanistically diverse library of small molecules for modulators of BDNF-induced Arc expression in primary cortical neurons. This effort identified compounds with a range of influences on Arc, including promoting its acetylation-a previously uncharacterized post-translational modification of this protein. Together, our data provide insights into the control of Arc that could be targeted to harness neuroplasticity for clinical applications.


Sujet(s)
Protéines du cytosquelette/composition chimique , Protéines du cytosquelette/métabolisme , Lysine/métabolisme , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/métabolisme , Acétylation , Motifs d'acides aminés , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Protéines du cytosquelette/génétique , Mâle , Souris , Souris de lignée C57BL , Protéines de tissu nerveux/génétique , Neurones/composition chimique , Neurones/métabolisme , Stabilité protéique
9.
Epilepsia ; 57(2): 222-32, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26659482

RÉSUMÉ

OBJECTIVE: We investigated the role of chloride homeostasis in seizure progression and development of pharmacoresistant status epilepticus (SE) by pharmacologically targeting the Na-K-Cl cotransporter (NKCC1) with bumetanide. We also investigated the ability of bumetanide to restore the efficacy of diazepam following SE. METHODS: Kainic acid (KA)-induced SE in vivo and 0-Mg(2+) -induced seizure-like events (SLEs) in vitro were monitored using electroencephalography (EEG) recordings in freely moving adult male mice and extracellular field potential recordings in acute entorhinal cortex-hippocampus slices, respectively. The ability of bumetanide to decrease epileptiform activity and prevent the development of pharmacoresistance to diazepam following SE was evaluated. RESULTS: Bumetanide treatment significantly reduced KA-induced ictal activity in vivo and SLEs in vitro. In addition, bumetanide restored the efficacy of diazepam in decreasing ictal activity following SE in both the in vivo and in vitro models. SIGNIFICANCE: Our data demonstrate an anticonvulsant effect of bumetanide on KA-induced seizures in adult mice, suggesting a role for chloride plasticity in seizure progression. These data also demonstrate that the erosion of inhibition during seizure progression could underlie the development of pharmacoresistant SE and implicate a role for chloride plasticity in this process.


Sujet(s)
Anticonvulsivants/pharmacologie , Bumétanide/pharmacologie , Diazépam/pharmacologie , Épilepsie pharmacorésistante/physiopathologie , Cortex entorhinal/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Crises épileptiques/physiopathologie , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , État de mal épileptique/physiopathologie , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/physiopathologie , Évolution de la maladie , Électroencéphalographie , Cortex entorhinal/physiopathologie , Agonistes des acides aminés excitateurs/toxicité , Hippocampe/physiopathologie , Techniques in vitro , Acide kaïnique/toxicité , Mâle , Souris , Souris de lignée C57BL , Techniques de patch-clamp , Crises épileptiques/induit chimiquement
10.
J Neurosci ; 35(21): 8291-6, 2015 May 27.
Article de Anglais | MEDLINE | ID: mdl-26019342

RÉSUMÉ

GABA(A) receptors form Cl(-) permeable channels that mediate the majority of fast synaptic inhibition in the brain. The K(+)/Cl(-) cotransporter KCC2 is the main mechanism by which neurons establish low intracellular Cl(-) levels, which is thought to enable GABAergic inhibitory control of neuronal activity. However, the widely used KCC2 inhibitor furosemide is nonselective with antiseizure efficacy in slices and in vivo, leading to a conflicting scheme of how KCC2 influences GABAergic control of neuronal synchronization. Here we used the selective KCC2 inhibitor VU0463271 [N-cyclopropyl-N-(4-methyl-2-thiazolyl)-2-[(6-phenyl-3-pyridazinyl)thio]acetamide] to investigate the influence of KCC2 function. Application of VU0463271 caused a reversible depolarizing shift in E(GABA) values and increased spiking of cultured hippocampal neurons. Application of VU0463271 to mouse hippocampal slices under low-Mg(2+) conditions induced unremitting recurrent epileptiform discharges. Finally, microinfusion of VU0463271 alone directly into the mouse dorsal hippocampus rapidly caused epileptiform discharges. Our findings indicated that KCC2 function was a critical inhibitory factor ex vivo and in vivo.


Sujet(s)
Hippocampe/physiologie , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Symporteurs/antagonistes et inhibiteurs , Symporteurs/physiologie , Animaux , Animaux nouveau-nés , Cellules cultivées , Cellules HEK293 , Hippocampe/effets des médicaments et des substances chimiques , Humains , Potentiels de membrane/effets des médicaments et des substances chimiques , Potentiels de membrane/physiologie , Souris , Souris de lignée C57BL , Techniques de culture d'organes , Rats , Rat Sprague-Dawley , Transmission synaptique/effets des médicaments et des substances chimiques , Transmission synaptique/physiologie ,
11.
Neuropharmacology ; 88: 103-9, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25229715

RÉSUMÉ

A robust increase in the functional expression of the neuronal K-Cl cotransporter KCC2 during CNS development is necessary for the emergence of hyperpolarizing ionotropic GABAergic transmission. BDNF-TrkB signaling has been implicated in the developmental up-regulation of KCC2 and, in mature animals, in fast activity-dependent down-regulation of KCC2 function following seizures and trauma. In contrast to the decrease in KCC2 expression observed in the adult hippocampus following trauma, seizures in the neonate trigger a TrkB-dependent up-regulation of neuronal Cl(-) extrusion capacity associated with enhanced surface expression of KCC2. Here, we show that this effect is transient, and impaired in the hippocampus of Bdnf(-/-) mice. Notably, however, a complete absence of BDNF does not compromise the increase in KCC2 protein or K-Cl transport functionality during neuronal development. Furthermore, we present data indicating that the functional up-regulation of KCC2 by neonatal seizures is temporally limited by calpain activity.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/déficit , Facteur neurotrophique dérivé du cerveau/physiologie , Hippocampe/physiopathologie , Crises épileptiques/physiopathologie , Symporteurs/métabolisme , Animaux , Animaux nouveau-nés , Technique de Western , Facteur neurotrophique dérivé du cerveau/génétique , Calpain/métabolisme , Chlorures/métabolisme , Modèles animaux de maladie humaine , Hippocampe/effets des médicaments et des substances chimiques , Acide kaïnique , Souris knockout , Neurones/effets des médicaments et des substances chimiques , Neurones/physiologie , État de mal épileptique/physiopathologie , Techniques de culture de tissus , Régulation positive ,
12.
J Neurosci ; 33(7): 2973-83, 2013 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-23407955

RÉSUMÉ

The cellular form of prion protein PrP(C) is highly expressed in the brain, where it can be converted into its abnormally folded isoform PrP(Sc) to cause neurodegenerative diseases. Its predominant synaptic localization suggests a crucial role in synaptic signaling. Interestingly, PrP(C) is developmentally regulated and its high expression in the immature brain could be instrumental in regulating neurogenesis and cell proliferation. Here, PrP(C)-deficient (Prnp(0/0)) mice were used to assess whether the prion protein is involved in synaptic plasticity processes in the neonatal hippocampus. To this aim, calcium transients associated with giant depolarizing potentials, a hallmark of developmental networks, were transiently paired with mossy fiber activation in such a way that the two events were coincident. While this procedure caused long-term potentiation (LTP) in wild-type (WT) animals, it caused long-term depression (LTD) in Prnp(0/0) mice. Induction of LTP was postsynaptic and required the activation of cAMP-dependent protein kinase A (PKA) signaling. The induction of LTD was presynaptic and relied on G-protein-coupled GluK1 receptor and protein lipase C. In addition, at emerging CA3-CA1 synapses in WT mice, but not in Prnp(0/0) mice, pairing Schaffer collateral stimulation with depolarization of CA1 principal cells induced LTP, known to be PKA dependent. Postsynaptic infusion of a constitutively active isoform of PKA catalytic subunit Cα into CA1 and CA3 principal cells in the hippocampus of Prnp(0/0) mice caused a persistent synaptic facilitation that was occluded by subsequent pairing. These data suggest that PrP(C) plays a crucial role in regulating via PKA synaptic plasticity in the developing hippocampus.


Sujet(s)
Cyclic AMP-Dependent Protein Kinases/physiologie , Hippocampe/physiologie , Plasticité neuronale/physiologie , Protéines PrPC/physiologie , Synapses/physiologie , Animaux , Région CA1 de l'hippocampe/physiologie , Région CA3 de l'hippocampe/physiologie , AMP cyclique/physiologie , Interprétation statistique de données , Phénomènes électrophysiologiques , Protéines G/physiologie , Hippocampe/croissance et développement , Potentialisation à long terme/physiologie , Souris , Souris knockout , Réseau nerveux/physiologie , Techniques de patch-clamp , Cellules pyramidales/physiologie , Récepteurs kaïnate/physiologie , Transmission synaptique/physiologie , Acide gamma-amino-butyrique/physiologie
13.
Sci Rep ; 2: 285, 2012.
Article de Anglais | MEDLINE | ID: mdl-22368777

RÉSUMÉ

Early in postnatal life, mossy fibres (MF), the axons of granule cells in the dentate gyrus, release GABA which is depolarizing and excitatory. Synaptic currents undergo spike-time dependent long-term depression (STD-LTD) regardless of the temporal order of stimulation (pre versus post and viceversa). Here we show that at P3 but not at P21, STD-LTD, induced by negative pairing, is mediated by endocannabinoids mobilized from the postsynaptic cell during spiking-induced membrane depolarization. By diffusing backward, endocannabinoids activate cannabinoid type-1 (CB1) receptors probably expressed on MF. Thus, STD-LTD was prevented by CB1 receptor antagonists and was absent in CB1-KO mice. Consistent with these data, in situ hybridization experiments revealed detectable level of CB1 mRNA in the granule cell layer at P3 but not at P21. These results indicate that CB1 receptors are transiently expressed on immature MF terminals where they counteract the enhanced neuronal excitability induced by the excitatory action of GABA.

14.
Adv Exp Med Biol ; 717: 11-26, 2011.
Article de Anglais | MEDLINE | ID: mdl-21713663

RÉSUMÉ

Kainate receptors (KARs) are glutamate-gated ion channels assembled from various combinations of GluK1-GluK5 subunits with different physiological and pharmacological properties. In the hippocampus, KARs expressed at postsynaptic sites mediate a small component of excitatory postsynaptic currents while at presynaptic sites they exert a powerful control on transmitter release at both excitatory and inhibitory connections. KARs are developmentally regulated and play a key role in several developmental processes including neuronal migration, differentiation and synapse formation. Interestingly, they can signal through a canonical ionotropic pathway but also through a noncanonical modality involving pertussis toxin-sensitive G proteins and downstream signaling molecules.In this Chapter some of our recent data concerning the functional role of presynaptic KARs in regulation of transmitter release from immature mossy fiber terminals and in synaptic plasticity processes will be reviewed. Early in postnatal development, MFs release into their targeted neurons mainly GABA which is depolarizing and excitatory. Endogenous activation of GluK1 KARs localized on MF terminals by glutamate present in the extracellular space down regulates GABA release, leading sometimes to synapse silencing. The depressant effect of GluK1 on MF responses is mediated by a metabotropic process, sensitive to pertussis toxin and phospholipase C (PLC) along the transduction pathway downstream to G protein activation. Blocking PLC with the selective antagonist U73122, unmasks the potentiating effect of GluK1 on MF-evoked GABAergic currents, which probably depend on the ionotropic type of action of these receptors.In addition, GluK1 KARs dynamically regulate the direction of spike-time dependent plasticity, a particular form of Hebbian type of learning which consists in bidirectional modifications in synaptic strength according to the temporal order of pre and postsynaptic spiking. At immature MF-CA3 synapses pairing MF stimulation with postsynaptic spiking and vice versa induces long term depression of MF-evoked GABAergic currents. In the case of positive pairing synaptic depression can be switched into spike-time dependent potentiation by blocking GluK1 KARs with UBP 302. The depressant action exerted by GluK1 KARs on MF responses would prevent the excessive activation of the CA3 associative network by the excitatory action of GABA early in postnatal development.


Sujet(s)
Hippocampe/métabolisme , Récepteurs kaïnate/métabolisme , Acide gamma-amino-butyrique/métabolisme , Animaux , Régulation négative , Hippocampe/croissance et développement , Humains
15.
J Neurosci ; 31(1): 34-45, 2011 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-21209187

RÉSUMÉ

GABA depolarizes immature neurons because of a high [Cl(-)](i) and orchestrates giant depolarizing potential (GDP) generation. Zilberter and coworkers (Rheims et al., 2009; Holmgren et al., 2010) showed recently that the ketone body metabolite DL-3-hydroxybutyrate (DL-BHB) (4 mM), lactate (4 mM), or pyruvate (5 mM) shifted GABA actions to hyperpolarizing, suggesting that the depolarizing effects of GABA are attributable to inadequate energy supply when glucose is the sole energy source. We now report that, in rat pups (postnatal days 4-7), plasma D-BHB, lactate, and pyruvate levels are 0.9, 1.5, and 0.12 mM, respectively. Then, we show that DL-BHB (4 mM) and pyruvate (200 µM) do not affect (i) the driving force for GABA(A) receptor-mediated currents (DF(GABA)) in cell-attached single-channel recordings, (2) the resting membrane potential and reversal potential of synaptic GABA(A) receptor-mediated responses in perforated patch recordings, (3) the action potentials triggered by focal GABA applications, or (4) the GDPs determined with electrophysiological recordings and dynamic two-photon calcium imaging. Only very high nonphysiological concentrations of pyruvate (5 mM) reduced DF(GABA) and blocked GDPs. Therefore, DL-BHB does not alter GABA signals even at the high concentrations used by Zilberter and colleagues, whereas pyruvate requires exceedingly high nonphysiological concentrations to exert an effect. There is no need to alter conventional glucose enriched artificial CSF to investigate GABA signals in the developing brain.


Sujet(s)
Potentiels d'action/effets des médicaments et des substances chimiques , Corps cétoniques/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/physiologie , Acide pyruvique/métabolisme , Acide gamma-amino-butyrique/pharmacologie , Acide 3-hydroxy-butyrique/sang , Acide 3-hydroxy-butyrique/pharmacologie , Potentiels d'action/physiologie , Animaux , Animaux nouveau-nés/sang , Bicuculline/pharmacologie , Encéphale/cytologie , Encéphale/croissance et développement , Bumétanide/pharmacologie , Calcium/métabolisme , Relation dose-effet des médicaments , Antagonistes des acides aminés excitateurs/pharmacologie , Femelle , Antagonistes du récepteur GABA-A/pharmacologie , Techniques in vitro , Acide lactique/sang , Mâle , Techniques de patch-clamp/méthodes , Acide pyruvique/pharmacologie , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs du symport chlorure potassium sodium/pharmacologie
16.
J Neurosci ; 30(5): 1750-9, 2010 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-20130184

RÉSUMÉ

Presynaptic kainate receptors regulate synaptic transmission in several brain areas but are not known to have this action at immature mossy fiber (MF) terminals, which during the first week of postnatal life release GABA, which exerts into targeted cells a depolarizing and excitatory action. Here, we report that, during the first week of postnatal life, endogenous activation of GluK1 receptors by glutamate present in the extracellular space severely depresses MF-mediated GABAergic currents [GABA(A)-mediated postsynaptic currents (GPSCs)]. Activation of GluK1 receptors was prevented by treating the slices with enzymatic glutamate scavengers that enhanced the clearance of glutamate from the extracellular space. The depressant effect of GluK1 on MF-GPSCs was mediated by a metabotropic process sensitive to pertussis toxin. In the presence of U73122 (1-[6-[[(17b)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione), a selective inhibitor of phospholipase C, along the transduction pathway downstream to G-protein, GluK1 activation increased the probability of GABA release, thus unveiling the ionotropic action of this receptor. In line with this type of action, we found that GluK1 enhanced MF excitability by directly depolarizing MF terminals via calcium-permeable cation channels. Furthermore, GluK1 dynamically regulated the direction of spike time-dependent plasticity occurring by pairing MF stimulation with postsynaptic spiking and switched spike time-dependent potentiation into depression. The GluK1-induced depression of MF-GPSCs would prevent excessive activation of the CA3 associative network by the excitatory action of GABA and the emergence of seizures in the immature brain.


Sujet(s)
Hippocampe/croissance et développement , Fibres moussues de l'hippocampe/métabolisme , Récepteurs kaïnate/métabolisme , Acide gamma-amino-butyrique/métabolisme , Potentiels d'action , Animaux , Régulation négative , Activation enzymatique , Acide glutamique/métabolisme , Plasticité neuronale/physiologie , Techniques de patch-clamp , Cellules pyramidales/cytologie , Cellules pyramidales/métabolisme , Rats , Rat Wistar
17.
Article de Anglais | MEDLINE | ID: mdl-21423487

RÉSUMÉ

In this review some of the recent work carried out in our laboratory concerning the functional role of GABAergic signalling at immature mossy fibres (MF)-CA3 principal cell synapses has been highlighted. While in adulthood MF, the axons of dentate gyrus granule cells release onto CA3 principal cells and interneurons glutamate, early in postnatal life they release GABA, which exerts into targeted cells a depolarizing and excitatory action. We found that GABA(A)-mediated postsynaptic currents (MF-GPSCs) exhibited a very low probability of release, were sensitive to L-AP4, a group III metabotropic glutamate receptor agonist, and revealed short-term frequency-dependent facilitation. Moreover, MF-GPSCs were down regulated by presynaptic GABA(B) and kainate receptors, activated by spillover of GABA from MF terminals and by glutamate present in the extracellular medium, respectively. Activation of these receptors contributed to the low release probability and in some cases to synapses silencing. By pairing calcium transients, associated with network-driven giant depolarizing potentials or GDPs (a hallmark of developmental networks thought to represent a primordial form of synchrony between neurons), generated by the synergistic action of glutamate and GABA with MF activation increased the probability of GABA release and caused the conversion of silent synapses into conductive ones suggesting that GDPs act as coincident detector signals for enhancing synaptic efficacy. Finally, to compare the relative strength of CA3 pyramidal cell output in relation to their MF glutamatergic or GABAergic inputs in adulthood or in postnatal development, respectively, a realistic model was constructed taking into account different biophysical properties of these synapses.

18.
J Neurosci ; 29(8): 2637-47, 2009 Feb 25.
Article de Anglais | MEDLINE | ID: mdl-19244539

RÉSUMÉ

In the adult rat hippocampus, the axons of granule cells in the dentate gyrus, the mossy fibers (MF), form excitatory glutamatergic synapses with CA3 principal cells. In neonates, MF release into their targets mainly GABA, which at this developmental stage is depolarizing. Here we tested the hypothesis that, at immature MF-CA3 synapses, correlated presynaptic [single fiber-evoked GABA(A)-mediated postsynaptic potentials (GPSPs)] and postsynaptic activity (back propagating action potentials) may exert a critical control on synaptic efficacy. This form of plasticity, called spike-timing-dependent plasticity (STDP), is a Hebbian type form of learning extensively studied at the level of glutamatergic synapses. Depending on the relative timing, pairing postsynaptic spiking and single MF-GPSPs induced bidirectional changes in synaptic efficacy. In case of positive pairing, spike-timing-dependent-long-term potentiation (STD-LTP) was associated with a persistent increase in GPSP slope and in the probability of cell firing. The transduction pathway involved a rise of calcium in the postsynaptic cell and the combined activity of cAMP-dependent PKA (protein kinase A) and brain-derived neurotrophic factor (BDNF). Retrograde signaling via BDNF and presynaptic TrkB receptors led to a persistent increase in GABA release. In "presynaptically" silent neurons, the enhanced probability of GABA release induced by the pairing protocol, unsilenced these synapses. Shifting E(GABA) from the depolarizing to the hyperpolarizing direction with bumetanide failed to modify synaptic strength. Thus, STD-LTP of GPSPs provides a reliable way to convey information from granule cells to the CA3 associative network at a time when glutamatergic synapses are still poorly developed.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Fibres moussues de l'hippocampe/physiologie , Synapses/physiologie , Transmission synaptique/physiologie , Acide gamma-amino-butyrique/métabolisme , Potentiels d'action/effets des médicaments et des substances chimiques , Potentiels d'action/physiologie , Animaux , Animaux nouveau-nés , Bumétanide/pharmacologie , Calcium/métabolisme , Stimulation électrique/méthodes , Antienzymes/pharmacologie , Hippocampe/cytologie , Techniques in vitro , Neurones/effets des médicaments et des substances chimiques , Neurones/physiologie , Techniques de patch-clamp/méthodes , Rats , Rat Wistar , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Synapses/effets des médicaments et des substances chimiques , Potentiels synaptiques/physiologie , Transmission synaptique/effets des médicaments et des substances chimiques , Facteurs temps
19.
Proc Natl Acad Sci U S A ; 104(32): 13176-81, 2007 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-17656555

RÉSUMÉ

At early developmental stages, correlated neuronal activity is thought to exert a critical control on functional and structural refinement of synaptic connections. In the hippocampus, between postnatal day 2 (P2) and P6, network-driven giant depolarizing potentials (GDPs) are generated by the synergistic action of glutamate and GABA, which is depolarizing and excitatory. Here the rising phase of GDPs was used to trigger Schaffer collateral stimulation in such a way that synchronized network activity was coincident with presynaptic activation of afferent input. This procedure produced a persistent increase in spontaneous and evoked alpha-amino-3-hydroxy-5-methyl-4-isoxadepropionic acid-mediated glutamatergic currents, an effect that required calcium influx through postsynaptic L-type calcium channels. No potentiation was observed when a delay of 3 sec was introduced between GDPs and afferent stimulation. Pairing-induced potentiation was prevented by scavengers of endogenous BDNF or tropomyosin-related kinase receptor B (TrkB) receptor antagonists. Blocking TrkB receptors in the postsynaptic cell did not prevent the effects of pairing, suggesting that BDNF, possibly secreted from the postsynaptic cell during GDPs, acts on TrkB receptors localized on presynaptic neurons. Application of exogenous BDNF mimicked the effects of pairing on synaptic transmission. In addition, pairing-induced synaptic potentiation was blocked by ERK inhibitors, suggesting that BDNF activates the MAPK/ERK cascade, which may lead to transcriptional regulation and new protein synthesis in the postsynaptic neuron. These results support the hypothesis that, during a critical period of postnatal development, GABAA-mediated GDPs are instrumental in tuning excitatory synaptic connections and provide insights into the molecular mechanisms involved in this process.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/pharmacologie , Extracellular Signal-Regulated MAP Kinases/physiologie , Hippocampe/physiologie , Système de signalisation des MAP kinases/physiologie , Synapses/physiologie , Animaux , Calcium/métabolisme , Potentiels post-synaptiques excitateurs , Potentialisation à long terme , Rats , Rat Wistar , Récepteurs du N-méthyl-D-aspartate/physiologie
20.
Genes Cells ; 11(9): 1071-83, 2006 Sep.
Article de Anglais | MEDLINE | ID: mdl-16923126

RÉSUMÉ

Sustained contraction of cells depends on sustained Rho-associated kinase (Rho-kinase) activation. We developed a computational model of the Rho-kinase pathway to understand the systems characteristics. Thrombin-dependent in vivo transient responses of Rho activation and Ca2+ increase could be reproduced in silico. Low and high thrombin stimulation induced transient and sustained phosphorylation, respectively, of myosin light chain (MLC) and myosin phosphatase targeting subunit 1 (MYPT1) in vivo. The transient phosphorylation of MLC and MYPT1 could be reproduced in silico, but their sustained phosphorylation could not. This discrepancy between in vivo and in silico in the sustained responses downstream of Rho-kinase indicates that a missing pathway(s) may be responsible for the sustained Rho-kinase activation. We found, experimentally, that the sustained phosphorylation of MLC and MYPT1 exhibit all-or-none responses. Bromoenol lactone, a specific inhibitor of Ca2+ -independent phospholipase A2 (iPLA2), inhibited sustained phosphorylation of MLC and MYPT1, which indicates that sustained Rho-kinase activation requires iPLA2 activity. Thus, the systems analysis of the Rho-kinase pathway identified a novel iPLA2-dependent mechanism of the sustained Rho-kinase activation, which exhibits an all-or-none response.


Sujet(s)
Calcium/métabolisme , Simulation numérique , Protéines et peptides de signalisation intracellulaire/métabolisme , Phospholipases A/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Cellules cultivées , Relation dose-effet des médicaments , Cellules endothéliales/cytologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Activation enzymatique/effets des médicaments et des substances chimiques , Group IV phospholipases A2 , Humains , Modèles biologiques , Chaînes légères de myosine/métabolisme , Myosin-light-chain phosphatase/métabolisme , Phospholipases A2 , Phosphorylation/effets des médicaments et des substances chimiques , Thrombine/pharmacologie , Protéines G rho/métabolisme , rho-Associated Kinases
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...