Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34187890

RESUMO

N-methyl-D-aspartate (NMDA) receptors (NMDARs), a principal subtype of excitatory neurotransmitter receptor, are composed as tetrameric assemblies of two glycine-binding GluN1 subunits and two glutamate-binding GluN2 subunits. NMDARs can signal nonionotropically through binding of glycine alone to its cognate site on GluN1. A consequence of this signaling by glycine is that NMDARs are primed such that subsequent gating, produced by glycine and glutamate, drives receptor internalization. The GluN1 subunit contains eight alternatively spliced isoforms produced by including or excluding the N1 and the C1, C2, or C2' polypeptide cassettes. Whether GluN1 alternative splicing affects nonionotropic signaling by NMDARs is a major outstanding question. Here, we discovered that glycine priming of recombinant NMDARs critically depends on GluN1 isoforms lacking the N1 cassette; glycine priming is blocked in splice variants containing N1. On the other hand, the C-terminal cassettes-C1, C2, or C2'-each permit glycine signaling. In wild-type mice, we found glycine-induced nonionotropic signaling at synaptic NMDARs in CA1 hippocampal pyramidal neurons. This nonionotropic signaling by glycine to synaptic NMDARs was prevented in mice we engineered, such that GluN1 obligatorily contained N1. We discovered in wild-type mice that, in contrast to pyramidal neurons, synaptic NMDARs in CA1 inhibitory interneurons were resistant to glycine priming. But we recapitulated glycine priming in inhibitory interneurons in mice engineered such that GluN1 obligatorily lacked the N1 cassette. Our findings reveal a previously unsuspected molecular function for alternative splicing of GluN1 in controlling nonionotropic signaling of NMDARs by activating the glycine site.


Assuntos
Processamento Alternativo/genética , Glicina/metabolismo , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Dinaminas/metabolismo , Endocitose , Interneurônios/metabolismo , Ativação do Canal Iônico , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Células Piramidais/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/genética , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Sinapses/metabolismo
2.
Mol Pain ; 18: 17448069221076634, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35174761

RESUMO

T lymphocytes are increasingly implicated in pain signaling. A subset of T lymphocytes, termed TChAT, express the rate-limiting enzyme for acetylcholine (ACh) production, choline acetyltransferase (ChAT), and mediate numerous physiological functions. Given that cholinergic signaling has long been known to modulate pain processing and is the basis for several analgesics used clinically, we asked whether TChAT could be the intersection between T lymphocyte and cholinergic mediation of pain signaling. In this study, we used a mouse gene knockout strategy to ablate ChAT specifically from T lymphocytes and examined the development and expression of mechanical and thermal hypersensitivity in a spared nerve injury (SNI) mouse model of neuropathic pain. We found that mice with ChAT knockout in T cells (floxed Chat plus CD4-Cre recombinase) did not differ from control mice with intact ChAT (floxed Chat, but no Cre recombinase) in their expression of mechanical sensitivity before or after injury. Similarly, thermal sensitivity was unaffected after injury, with control mice expressing similar patterns of thermal preference to mice whose T cells do not express ChAT. Our experiments demonstrate that cholinergic signaling initiated by T lymphocytes neither dampens nor exacerbates the expression of mechanical or thermal sensitivity in neuropathic mice. Thus, while both cholinergic signaling and T lymphocytes have established roles in modulating pain phenotypes, it is not cholinergic signaling initiated by T lymphocytes that drive this. Our findings will help to narrow in on which aspects of T-cell modulation may prove useful as therapies.


Assuntos
Neuralgia , Linfócitos T , Acetilcolina/metabolismo , Animais , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Colinérgicos/metabolismo , Camundongos , Neuralgia/metabolismo , Linfócitos T/metabolismo
3.
J Neurosci ; 39(16): 3081-3093, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30796159

RESUMO

Neonatal hindpaw incision primes developing spinal nociceptive circuitry, resulting in enhanced hyperalgesia following reinjury in adulthood. Spinal microglia contribute to this persistent effect, and microglial inhibition at the time of adult reincision blocks the enhanced hyperalgesia. Here, we pharmacologically inhibited microglial function with systemic minocycline or intrathecal SB203580 at the time of neonatal incision and evaluated sex-dependent differences following adult reincision. Incision in adult male and female rats induced equivalent hyperalgesia and spinal dorsal horn expression of genes associated with microglial proliferation (Emr1) and transformation to a reactive phenotype (Irf8). In control adults with prior neonatal incision, the enhanced degree and duration of incision-induced hyperalgesia and spinal microglial responses to reincision were equivalent in males and females. However, microglial inhibition at the time of the neonatal incision revealed sex-dependent effects: the persistent mechanical and thermal hyperalgesia following reincision in adulthood was prevented in males but unaffected in females. Similarly, reincision induced Emr1 and Irf8 gene expression was downregulated in males, but not in females, following neonatal incision with minocycline. To evaluate the distribution of reincision hyperalgesia, prior neonatal incision was performed at different body sites. Hyperalgesia was maximal when the same paw was reincised, and was increased following prior incision at ipsilateral, but not contralateral, sites, supporting a segmentally restricted spinal mechanism. These data highlight the contribution of spinal microglial mechanisms to persistent effects of early-life injury in males, and sex-dependent differences in the ability of microglial inhibition to prevent the transition to a persistent pain state span developmental stages.SIGNIFICANCE STATEMENT Following the same surgery, some patients develop persistent pain. Contributory mechanisms are not fully understood, but early-life experience and sex/gender may influence the transition to chronic pain. Surgery and painful procedural interventions in vulnerable preterm neonates are associated with long-term alterations in somatosensory function and pain that differ in males and females. Surgical injury in neonatal rodents primes the developing nociceptive system and enhances reinjury response in adulthood. Neuroimmune interactions are critical mediators of persistent pain, but sex-dependent differences in spinal neuroglial signaling influence the efficacy of microglial inhibitors following adult injury. Neonatal microglial inhibition has beneficial long-term effects on reinjury response in adult males only, emphasizing the importance of evaluating sex-dependent differences at all ages in preclinical studies.


Assuntos
Hiperalgesia/fisiopatologia , Microglia/metabolismo , Dor/fisiopatologia , Medula Espinal/fisiopatologia , Animais , Inibidores Enzimáticos/farmacologia , Feminino , Hiperalgesia/metabolismo , Imidazóis/farmacologia , Fatores Reguladores de Interferon/metabolismo , Masculino , Microglia/efeitos dos fármacos , Minociclina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/metabolismo , Limiar da Dor/fisiologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/metabolismo , Fatores Sexuais , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
4.
Neuroimage ; 163: 220-230, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28882630

RESUMO

MRI is a powerful modality to detect neuroanatomical differences that result from mutations and treatments. Knowing which genes drive these differences is important in understanding etiology, but candidate genes are often difficult to identify. We tested whether spatial gene expression data from the Allen Brain Institute can be used to inform us about genes that cause neuroanatomical differences. For many single-gene-mutation mouse models, we found that affected neuroanatomy was not strongly associated with the spatial expression of the altered gene and there are specific caveats for each model. However, among models with significant neuroanatomical differences from their wildtype controls, the mutated genes had preferential spatial expression in affected neuroanatomy. In mice exposed to environmental enrichment, candidate genes could be identified by a genome-wide search for genes with preferential spatial expression in the altered neuroanatomical regions. These candidates have functions related to learning and plasticity. We demonstrate that spatial gene expression of single-genes is a poor predictor of altered neuroanatomy, but altered neuroanatomy can identify candidate genes responsible for neuroanatomical phenotypes.


Assuntos
Encéfalo/anatomia & histologia , Animais , Modelos Animais de Doenças , Estudos de Associação Genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Fenótipo
5.
J Neurosci ; 33(9): 4055-65, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447614

RESUMO

Invertebrate studies have highlighted a role for EH and SH3 domain Intersectin (Itsn) proteins in synaptic vesicle recycling and morphology. Mammals have two Itsn genes (Itsn1 and Itsn2), both of which can undergo alternative splicing to include DBL/PH and C2 domains not present in invertebrate Itsn proteins. To probe for specific and redundant functions of vertebrate Itsn genes, we generated Itsn1, Itsn2, and double mutant mice. While invertebrate mutants showed severe synaptic abnormalities, basal synaptic transmission and plasticity were unaffected at Schaffer CA1 synapses in mutant mice. Surprisingly, intercortical tracts-corpus callosum, ventral hippocampal, and anterior commissures-failed to cross the midline in mice lacking Itsn1, but not Itsn2. In contrast, tracts extending within hemispheres and those that decussate to more caudal brain segments appeared normal. Itsn1 mutant mice showed severe deficits in Morris water maze and contextual fear memory tasks, whereas mice lacking Itsn2 showed normal learning and memory. Thus, coincident with the acquisition of additional signaling domains, vertebrate Itsn1 has been functionally repurposed to also facilitate interhemispheric connectivity essential for high order cognitive functions.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Cognição/fisiologia , Corpo Caloso/fisiologia , Lateralidade Funcional/genética , 2-Amino-5-fosfonovalerato/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Análise de Variância , Animais , Biofísica , Mapeamento Encefálico , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Sinais (Psicologia) , Imagem de Tensor de Difusão , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Comportamento Exploratório/fisiologia , Medo , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/fisiologia , Hipocampo/citologia , Imageamento Tridimensional , Técnicas In Vitro , Deficiências da Aprendizagem/genética , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos , Mutação/genética , Fibras Nervosas/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Vertebrados/genética , Vertebrados/metabolismo
6.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230236, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-38853562

RESUMO

Alternative splicing of Grin1 exon 5 regulates induction of long-term potentiation (LTP) at Schaffer collateral-CA1 synapses: LTP in mice lacking the GluN1 exon 5-encoded N1 cassette (GluN1a mice) is significantly increased compared with that in mice compulsorily expressing this exon (GluN1b mice). The mechanism underlying this difference is unknown. Here, we report that blocking the non-receptor tyrosine kinase Src prevents induction of LTP in GluN1a mice but not in GluN1b. We find that activating Src enhances pharmacologically isolated synaptic N-methyl-d-aspartate receptor (NMDAR) currents in GluN1a mice but not in GluN1b. Moreover, we observe that Src activation increases the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor component of Schaffer collateral-evoked excitatory post-synaptic potentials in GluN1a mice, but this increase is prevented by blocking NMDARs. We conclude that at these synapses, NMDARs in GluN1a mice are subject to upregulation by Src that mediates induction of LTP, whereas NMDARs in GluN1b mice are not regulated by Src, leading to Src-resistance of LTP. Thus, we have uncovered that a key regulatory mechanism for synaptic potentiation is gated by differential splicing of exon 5 of Grin1. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Assuntos
Processamento Alternativo , Éxons , Potenciação de Longa Duração , Proteínas do Tecido Nervoso , Receptores de N-Metil-D-Aspartato , Quinases da Família src , Animais , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Camundongos , Quinases da Família src/metabolismo , Quinases da Família src/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Masculino , Sinapses/fisiologia , Sinapses/metabolismo , Camundongos Endogâmicos C57BL
7.
Dis Model Mech ; 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38881329

RESUMO

MECP2 duplication syndrome (MDS) is a neurodevelopmental disorder caused by tandem duplication of the MECP2 locus and its surrounding genes, including IRAK1. Current MDS mouse models involve transgenic expression of MECP2 only, limiting their applicability to the study of the disease. Herein, we show that an efficient and precise CRISPR/Cas9 fusion proximity-based approach can be utilized to generate an Irak1-Mecp2 tandem duplication mouse model ("Mecp2 Dup"). The Mecp2 Dup mouse model recapitulates the genomic landscape of human MDS by harbouring a 160 kb tandem duplication encompassing Mecp2 and Irak1, representing the minimal disease-causing duplication, and the neighbouring genes Opnmw1 and Tex28. The Mecp2 Dup model exhibits neuro-behavioral abnormalities, and an abnormal immune response to infection not previously observed in other mouse models, possibly owing to Irak1 overexpression. The Mecp2 Dup model thus provides a tool to investigate MDS disease mechanisms and develop potential therapies applicable to patients.

8.
Br J Pharmacol ; 180(21): 2822-2836, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37336547

RESUMO

BACKGROUND AND PURPOSE: Chronic pain is a devastating problem affecting one in five individuals around the globe, with neuropathic pain the most debilitating and poorly treated type of chronic pain. Advances in transcriptomics have contributed to cataloguing diverse cellular pathways and transcriptomic alterations in response to peripheral nerve injury but have focused on phenomenology and classifying transcriptomic responses. EXPERIMENTAL APPROACH: To identifying new types of pain-relieving agents, we compared transcriptional reprogramming changes in the dorsal spinal cord after peripheral nerve injury cross-sex and cross-species, and imputed commonalities, as well as differences in cellular pathways and gene regulation. KEY RESULTS: We identified 93 transcripts in the dorsal horn that were increased by peripheral nerve injury in male and female mice and rats. Following gene ontology and transcription factor analyses, we constructed a pain interactome for the proteins encoded by the differentially expressed genes, discovering new, conserved signalling nodes. We investigated the interactome with the Drug-Gene database to predict FDA-approved medications that may modulate key nodes within the network. The top hit from the analysis was fostamatinib, the molecular target of which is the non-receptor spleen associated tyrosine kinase (Syk), which our analysis had identified as a key node in the interactome. We found that intrathecally administrating the active metabolite of fostamatinib, R406 and another Syk inhibitor P505-15, significantly reversed pain hypersensitivity in both sexes. CONCLUSIONS AND IMPLICATIONS: Thus, we have identified and shown the efficacy of an agent that could not have been previously predicted to have analgesic properties.


Assuntos
Dor Crônica , Neuralgia , Traumatismos dos Nervos Periféricos , Feminino , Ratos , Camundongos , Masculino , Animais , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Traumatismos dos Nervos Periféricos/metabolismo , Dor Crônica/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/genética , Neuralgia/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Hiperalgesia/metabolismo
9.
Neuropharmacology ; 193: 108615, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051267

RESUMO

The Src family kinases (SFKs) are cytoplasmic non-receptor tyrosine kinases involved in multiple signalling pathways. In the central nervous system (CNS), SFKs are key regulators of N-methyl-d-aspartate receptor (NMDAR) function and major points of convergence for neuronal transduction pathways. Physiological upregulation of NMDAR activity by members of the SFKs, namely Src and Fyn, is crucial for induction of plasticity at Schaffer collateral-CA1 synapses of the hippocampus. Aberrant SFK regulation of NMDARs is implicated in several pathological conditions in the CNS including schizophrenia and pain hypersensitivity. Here, evidence is presented to highlight the current understanding of the intermolecular interactions of SFKs within the NMDAR macromolecular complex, the upstream regulators of SFK activity on NMDAR function and the role Src and Fyn have in synaptic plasticity and metaplasticity. The targeting of SFK protein-protein interactions is discussed as a potential therapeutic strategy to restore signalling activity underlying glutamatergic dysregulation in CNS disease pathophysiology.


Assuntos
Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Animais , Humanos , Camundongos , Plasticidade Neuronal/fisiologia , Dor/metabolismo , Ratos , Esquizofrenia/metabolismo , Transdução de Sinais , Sinapses/metabolismo
10.
Traffic ; 9(5): 742-54, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18298590

RESUMO

Intersectins (Itsn) are conserved EH and SH3 domain containing adaptor proteins. In Drosophila melanogaster, ITSN is required to regulate synaptic morphology, to facilitate efficient synaptic vesicle recycling and for viability. Here, we report our genetic analysis of Caenorhabditis elegans intersectin. In contrast to Drosophila, C. elegans itsn-1 protein null mutants are viable and display grossly normal locomotion and development. However, motor neurons in these mutants show a dramatic increase in large irregular vesicles and accumulate membrane-associated vesicles at putative endocytic hotspots, approximately 300 nm from the presynaptic density. This defect occurs precisely where endogenous ITSN-1 protein localizes in wild-type animals and is associated with a significant reduction in synaptic vesicle number and reduced frequency of endogenous synaptic events at neuromuscular junctions (NMJs). ITSN-1 forms a stable complex with EHS-1 (Eps15) and is expressed at reduced levels in ehs-1 mutants. Thus, ITSN-1 together with EHS-1, coordinate vesicle recycling at C. elegans NMJs. We also found that both itsn-1 and ehs-1 mutants show poor viability and growth in a Disabled (dab-1) null mutant background. These results show for the first time that intersectin and Eps15 proteins function in the same genetic pathway, and appear to function synergistically with the clathrin-coat-associated sorting protein, Disabled, for viability.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Junção Neuromuscular/fisiologia , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/anatomia & histologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Drosophila melanogaster , Endocitose , Deleção de Genes , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Técnicas de Patch-Clamp , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/ultraestrutura , Técnicas do Sistema de Duplo-Híbrido
11.
Mol Brain ; 13(1): 23, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32070387

RESUMO

N-methyl-D-aspartate receptors (NMDARs) are excitatory glutamatergic receptors that are fundamental for many neuronal processes, including synaptic plasticity. NMDARs are comprised of four subunits derived from heterogeneous subunit families, yielding a complex diversity in NMDAR form and function. The quadruply-liganded state of binding of two glutamate and two glycine molecules to the receptor drives channel gating, allowing for monovalent cation flux, Ca2+ entry and the initiation of Ca2+-dependent signalling. In addition to this ionotropic function, non-ionotropic signalling can be initiated through the exclusive binding of glycine or of glutamate to the NMDAR. This binding may trigger a transmembrane conformational change of the receptor, inducing intracellular protein-protein signalling between the cytoplasmic domain and secondary messengers. In this review, we outline signalling cascades that can be activated by NMDARs and propose that the receptor transduces signalling through three parallel streams: (i) signalling via both glycine and glutamate binding, (ii) signalling via glycine binding, and (iii) signalling via glutamate binding. This variety in signal transduction mechanisms and downstream signalling cascades complements the widespread prevalence and rich diversity of NMDAR activity throughout the central nervous system and in disease pathology.


Assuntos
Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Animais , Ácido Glutâmico/metabolismo , Glicina/metabolismo , Humanos , Modelos Biológicos
12.
Cell Rep ; 29(13): 4285-4294.e5, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875540

RESUMO

NMDA receptors (NMDARs) are critical for physiological synaptic plasticity, learning, and memory and for pathological plasticity and neuronal death. The GluN1 subunit is encoded by a single gene, GRIN1, with 8 splice variants, but whether the diversity generated by this splicing has physiological consequences remains enigmatic. Here, we generate mice lacking from the GluN1 exon 5-encoded N1 cassette (GluN1a mice) or compulsorily expressing this exon (GluN1b mice). Despite no differences in basal synaptic transmission, long-term potentiation in the hippocampus is significantly enhanced in GluN1a mice compared with that in GluN1b mice. Furthermore, GluN1a mice learn more quickly and have significantly better spatial memory performance than do GluN1b mice. In addition, in human iPSC-derived neurons in autism spectrum disorder NMDARs show characteristics of N1-lacking GluN1. Our findings indicate that alternative splicing of GluN1 is a mechanism for controlling physiological long-lasting synaptic potentiation, learning, and memory.


Assuntos
Processamento Alternativo , Transtorno do Espectro Autista/genética , Potenciação de Longa Duração/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animais , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Morte Celular/genética , Diferenciação Celular , Potenciais Pós-Sinápticos Excitadores/fisiologia , Éxons , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurônios/patologia , Cultura Primária de Células , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Receptores de N-Metil-D-Aspartato/deficiência , Memória Espacial/fisiologia , Sinapses/metabolismo , Transmissão Sináptica
13.
Brain Struct Funct ; 224(2): 811-827, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30511336

RESUMO

Cerebellar granule neurons are the most numerous neuronal subtype in the central nervous system. Within the developing cerebellum, these neurons are derived from a population of progenitor cells found within the external granule layer of the cerebellar anlage, namely the cerebellar granule neuron precursors (GNPs). The timely proliferation and differentiation of these precursor cells, which, in rodents occurs predominantly in the postnatal period, is tightly controlled to ensure the normal morphogenesis of the cerebellum. Despite this, our understanding of the factors mediating how GNP differentiation is controlled remains limited. Here, we reveal that the transcription factor nuclear factor I X (NFIX) plays an important role in this process. Mice lacking Nfix exhibit reduced numbers of GNPs during early postnatal development, but elevated numbers of these cells at postnatal day 15. Moreover, Nfix-/- GNPs exhibit increased proliferation when cultured in vitro, suggestive of a role for NFIX in promoting GNP differentiation. At a mechanistic level, profiling analyses using both ChIP-seq and RNA-seq identified the actin-associated factor intersectin 1 as a downstream target of NFIX during cerebellar development. In support of this, mice lacking intersectin 1 also displayed delayed GNP differentiation. Collectively, these findings highlight a key role for NFIX and intersectin 1 in the regulation of cerebellar development.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proliferação de Células/fisiologia , Cerebelo/citologia , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Knockout , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo
14.
Ageing Res Rev ; 40: 84-94, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28941639

RESUMO

The immune system is now considered a major factor in Alzheimer Disease (AD). This review seeks to demonstrate how various aspects of the immune system, both in the brain and peripherally, interact to contribute to AD. We highlight classical nervous system immune components, such as complement and microglia, as well as novel aspects of the peripheral immune system that can influence disease, such as monocytes and lymphocytes. By detailing the roles of various immune cells in AD, we summarize an emerging perspective for disease etiology and future therapeutic targets.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Imunidade Inata/imunologia , Imunoterapia/métodos , Doença de Alzheimer/etiologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Imunidade Inata/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/imunologia , Resultado do Tratamento
15.
Sci Rep ; 6: 23837, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-27040756

RESUMO

NMDA receptor (NMDAR)-mediated fast excitatory neurotransmission is implicated in a broad range of physiological and pathological processes in the mammalian central nervous system. The function and regulation of NMDARs have been extensively studied in neurons from rodents and other non-human species, and in recombinant expression systems. Here, we investigated human NMDARs in situ by using neurons produced by directed differentiation of human induced pluripotent stem cells (iPSCs). The resultant cells showed electrophysiological characteristics demonstrating that they are bona fide neurons. In particular, human iPSC-derived neurons expressed functional ligand-gated ion channels, including NMDARs, AMPA receptors, GABAA receptors, as well as glycine receptors. Pharmacological and electrophysiological properties of NMDAR-mediated currents indicated that these were dominated by receptors containing GluN2B subunits. The NMDAR currents were suppressed by genistein, a broad-spectrum tyrosine kinase inhibitor. The NMDAR currents were also inhibited by a Fyn-interfering peptide, Fyn(39-57), but not a Src-interfering peptide, Src(40-58). Together, these findings are the first evidence that tyrosine phosphorylation regulates the function of NMDARs in human iPSC-derived neurons. Our findings provide a basis for utilizing human iPSC-derived neurons in screening for drugs targeting NMDARs in neurological disorders.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Diferenciação Celular , Células Cultivadas , Genisteína/farmacologia , Humanos , Plasticidade Neuronal , Neurônios/metabolismo , Transmissão Sináptica/efeitos dos fármacos
16.
Cell Rep ; 17(10): 2753-2765, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27926876

RESUMO

In chronic pain states, the neurotrophin brain-derived neurotrophic factor (BDNF) transforms the output of lamina I spinal neurons by decreasing synaptic inhibition. Pain hypersensitivity also depends on N-methyl-D-aspartate receptors (NMDARs) and Src-family kinases, but the locus of NMDAR dysregulation remains unknown. Here, we show that NMDAR-mediated currents at lamina I synapses are potentiated in a peripheral nerve injury model of neuropathic pain. We find that BDNF mediates NMDAR potentiation through activation of TrkB and phosphorylation of the GluN2B subunit by the Src-family kinase Fyn. Surprisingly, we find that Cl--dependent disinhibition is necessary and sufficient to prime potentiation of synaptic NMDARs by BDNF. Thus, we propose that spinal pain amplification is mediated by a feedforward mechanism whereby loss of inhibition gates the increase in synaptic excitation within individual lamina I neurons. Given that neither disinhibition alone nor BDNF-TrkB signaling is sufficient to potentiate NMDARs, we have discovered a form of molecular coincidence detection in lamina I neurons.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Neuralgia/genética , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas Proto-Oncogênicas c-fyn/genética , Receptor trkB/genética , Receptores de N-Metil-D-Aspartato/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Neuralgia/metabolismo , Neuralgia/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/fisiopatologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Nervos Espinhais/metabolismo , Nervos Espinhais/fisiopatologia , Sinapses/genética , Sinapses/patologia , Quinases da Família src/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa