Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 49(9): 1392-1401, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38438594

RESUMO

Cognitive impairment is a common phenotype of neurodevelopmental disorders, but how these deficits arise remains elusive. Determining the onset of discrete cognitive capabilities facilitates studies in probing mechanisms underlying their emergence. The present study analyzed the emergence of contextual fear memory persistence (7-day memory retention) and remote memory (30-day memory retention). There was a rapid transition from postnatal day (P) 20 to P21, in which memory persistence emerged in C57Bl/6 J male and female mice. Remote memory was present at P23, but expression was not robust compared to pubertal and adult mice. Previous studies reported that following deletion of the MET receptor tyrosine kinase (MET), there are fear memory deficits in adult mice and the timing of critical period plasticity is altered in the developing visual cortex, positioning MET as a regulator for onset of contextual fear memory. Sustaining Met past the normal window of peak cortical expression or deleting Met, however, did not alter the timing of emergence of persistence or remote memory capabilities during development. Fear memory in young adults, however, was disrupted. Remarkably, compared to homecage controls, the number of FOS-expressing infragranular neurons in medial prefrontal cortex (mPFC) did not increase from contextual memory formation recall of fear conditioning at P35 but exhibited enhanced activation at P90 in male and female mice. Additionally, MET-expressing neurons were preferentially recruited at P90 compared to P35 during fear memory expression. The studies demonstrate a developmental profile of contextual fear memory capabilities. Further, developmental disruption of Met leads to a delayed functional deficit that arises in young adulthood, correlated with an increase of mPFC neuron activation during fear memory recall.


Assuntos
Medo , Camundongos Endogâmicos C57BL , Animais , Medo/fisiologia , Feminino , Masculino , Camundongos , Memória/fisiologia , Memória de Longo Prazo/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo
2.
bioRxiv ; 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36778231

RESUMO

Cognitive impairment is a common phenotype of neurodevelopmental disorders, but how these deficits arise remains elusive. Determining the onset of discrete cognitive capabilities facilitates studies in probing mechanisms underlying their emergence. The present study analyzed the emergence of contextual fear memory persistence (7-day memory retention) and remote memory (30-day memory retention). There was a rapid transition from postnatal day (P) 20 to P21, in which memory persistence emerged in C57Bl/6J male and female mice. Remote memory was present at P23, but expression was not robust compared to pubertal and adult mice. Previous studies reported that following deletion of the MET receptor tyrosine kinase (MET), there are fear memory deficits in adult mice and the timing of critical period plasticity is altered in the developing visual cortex, positioning MET as a regulator for onset of contextual fear memory. Sustaining Met past the normal window of peak cortical expression or deleting Met, however, did not alter the timing of emergence of persistence or remote memory capabilities during development. Fear memory in young adults, however, was disrupted. Remarkably, compared to homecage controls, the number of FOS-expressing infragranular neurons in medial prefrontal cortex (mPFC) did not increase from contextual memory formation recall of fear conditioning at P35 but exhibited enhanced activation at P90 in male and female mice. Additionally, MET-expressing neurons were preferentially recruited at P90 compared to P35 during fear memory expression. The studies demonstrate a developmental profile of contextual fear memory capabilities. Further, developmental disruption of Met leads to a delayed functional deficit that arises in young adulthood, correlated with an increase of mPFC neuron activation during fear memory recall.

3.
Dev Neurosci ; 45(3): 126-138, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36882009

RESUMO

Alterations in the expression of genes encoding proteins involved in synapse formation, maturation, and function are a hallmark of many neurodevelopmental and psychiatric disorders. For example, there is reduced neocortical expression of the MET receptor tyrosine kinase (MET) transcript and protein in Autism Spectrum Disorder (ASD) and Rett syndrome. Preclinical in vivo and in vitro models manipulating MET signaling reveal that the receptor modulates excitatory synapse development and maturation in select forebrain circuits. The molecular adaptations underlying the altered synaptic development remain unknown. We performed a comparative mass spectrometry analysis of synaptosomes generated from the neocortex of wild type and Met null mice during the peak of synaptogenesis (postnatal day 14; data are available from ProteomeXchange with identifier PXD033204). The analyses revealed broad disruption of the developing synaptic proteome in the absence of MET, consistent with the localization of MET protein in pre- and postsynaptic compartments, including proteins associated with the neocortical synaptic MET interactome and those encoded by syndromic and ASD risk genes. In addition to an overrepresentation of altered proteins associated with the SNARE complex, multiple proteins in the ubiquitin-proteasome system and associated with the synaptic vesicle, as well as proteins that regulate actin filament organization and synaptic vesicle exocytosis/endocytosis, were disrupted. Taken together, the proteomic changes are consistent with structural and functional changes observed following alterations in MET signaling. We hypothesize that the molecular adaptations following Met deletion may reflect a general mechanism that produces circuit-specific molecular changes due to loss or reduction of synaptic signaling proteins.


Assuntos
Transtorno do Espectro Autista , Neocórtex , Camundongos , Animais , Sinaptossomos/metabolismo , Proteoma/metabolismo , Transtorno do Espectro Autista/genética , Proteômica/métodos , Sinapses/metabolismo
4.
J Comp Neurol ; 531(1): 132-148, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36201439

RESUMO

Met encodes a receptor tyrosine kinase (MET) that is expressed during development and regulates cortical synapse maturation. Conditional deletion of Met in the nervous system during embryonic development leads to deficits in adult contextual fear learning, a medial prefrontal cortex (mPFC)-dependent cognitive task. MET also regulates the timing of critical period plasticity for ocular dominance in primary visual cortex (V1). However, the underlying circuitry responsible remains unknown. Therefore, this study determines the broad expression patterns of MET throughout postnatal development in mPFC and V1 projection neurons (PNs), providing insight into similarities and differences in the neuronal subtypes and temporal patterns of MET expression between cortical areas. Using a transgenic mouse line that expresses green fluorescent protein (GFP) in Met+ neurons, immunofluorescence and confocal microscopy were performed to visualize MET-GFP+ cell bodies and PN subclass-specific protein markers. Analyses reveal that the MET expression is highly enriched in infragranular layers of mPFC, but in supragranular layers of V1. Interestingly, temporal regulation of the percentage of MET+ neurons across development not only differs between cortical regions but also is distinct between lamina within a cortical region. Further, MET is expressed predominantly in the subcerebral PN subclass in mPFC, but the intratelencephalic PN subclass in V1. The data suggest that MET signaling influences the development of distinct circuits in mPFC and V1 that underlie subcerebral and intracortical functional deficits following Met deletion, respectively.


Assuntos
Proteínas Proto-Oncogênicas c-met , Córtex Visual , Animais , Camundongos , Gravidez , Feminino , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Córtex Visual/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Medo/fisiologia , Camundongos Transgênicos
5.
Neurobiol Stress ; 13: 100251, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33344706

RESUMO

Exposure to early-life stress (ELS) increases risk for poor mental and physical health outcomes that emerge at different stages across the lifespan. Yet, how age interacts with ELS to impact the expression of specific phenotypes remains largely unknown. An established limited-bedding paradigm was used to induce ELS in mouse pups over the early postnatal period. Initial analyses focused on the hippocampus, based on documented sensitivity to ELS in humans and various animal models, and the large body of data reporting anatomical and physiological outcomes in this structure using this ELS paradigm. An unbiased discovery proteomics approach revealed distinct adaptations in the non-nuclear hippocampal proteome in male versus female offspring at two distinct developmental stages: juvenile and adult. Gene ontology and KEGG pathway analyses revealed significant enrichment in proteins associated with mitochondria and the oxidative phosphorylation (OXPHOS) pathway in response to ELS in female hippocampus only. To determine whether the protein adaptations to ELS reflected altered function, mitochondrial respiration (driven through complexes II-IV) and complex I activity were measured in isolated hippocampal mitochondria using a Seahorse X96 Flux analyzer and immunocapture ELISA, respectively. ELS had no effect on basal respiration in either sex at either age. In contrast, ELS increased OXPHOS capacity in juvenile males and females, and reduced OXPHOS capacity in adult females but not adult males. A similar pattern of ELS-induced changes was observed for complex I activity. These data suggest that initial adaptations in juvenile hippocampus due to ELS were not sustained in adults. Mitochondrial adaptations to ELS were also exhibited peripherally by liver. Overall, the temporal distinctions in mitochondrial responses to ELS show that ELS-generated adaptations and outcomes are complex over the lifespan. This may contribute to differences in the timing of appearance of mental and physical disturbances, as well as potential sex differences that influence only select outcomes.

6.
J Neurosci ; 37(45): 10783-10791, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118206

RESUMO

Adversity in early childhood exerts an enduring impact on mental and physical health, academic achievement, lifetime productivity, and the probability of interfacing with the criminal justice system. More science is needed to understand how the brain is affected by early life stress (ELS), which produces excessive activation of stress response systems broadly throughout the child's body (toxic stress). Our research examines the importance of sex, timing and type of stress exposure, and critical periods for intervention in various brain systems across species. Neglect (the absence of sensitive and responsive caregiving) or disrupted interaction with offspring induces robust, lasting consequences in mice, monkeys, and humans. Complementary assessment of internalizing disorders and brain imaging in children suggests that early adversity can interfere with white matter development in key brain regions, which may increase risk for emotional difficulties in the long term. Neural circuits that are most plastic during ELS exposure in monkeys sustain the greatest change in gene expression, offering a mechanism whereby stress timing might lead to markedly different long-term behaviors. Rodent models reveal that disrupted maternal-infant interactions yield metabolic and behavioral outcomes often differing by sex. Moreover, ELS may further accelerate or delay critical periods of development, which reflect GABA circuit maturation, BDNF, and circadian Clock genes. Such factors are associated with several mental disorders and may contribute to a premature closure of plastic windows for intervention following ELS. Together, complementary cross-species studies are elucidating principles of adaptation to adversity in early childhood with molecular, cellular, and whole organism resolution.


Assuntos
Maus-Tratos Infantis/psicologia , Deficiências do Desenvolvimento/etiologia , Meio Social , Adulto , Criança , Pré-Escolar , Deficiências do Desenvolvimento/psicologia , Cuidados no Lar de Adoção/psicologia , Humanos , Estresse Psicológico/psicologia
7.
Biol Psychiatry ; 81(5): 424-433, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27837921

RESUMO

People with autism spectrum disorder and other neurodevelopmental disorders (NDDs) are behaviorally and medically heterogeneous. The combination of polygenicity and gene pleiotropy-the influence of one gene on distinct phenotypes-raises questions of how specific genes and their protein products interact to contribute to NDDs. A preponderance of evidence supports developmental and pathophysiological roles for the MET receptor tyrosine kinase, a multifunctional receptor that mediates distinct biological responses depending upon cell context. MET influences neuron architecture and synapse maturation in the forebrain and regulates homeostasis in gastrointestinal and immune systems, both commonly disrupted in NDDs. Peak expression of synapse-enriched MET is conserved across rodent and primate forebrain, yet regional differences in primate neocortex are pronounced, with enrichment in circuits that participate in social information processing. A functional risk allele in the MET promoter, enriched in subgroups of children with autism spectrum disorder, reduces transcription and disrupts socially relevant neural circuits structurally and functionally. In mice, circuit-specific deletion of Met causes distinct atypical behaviors. MET activation increases dendritic complexity and nascent synapse number, but synapse maturation requires reductions in MET. MET mediates its specific biological effects through different intracellular signaling pathways and has a complex protein interactome that is enriched in autism spectrum disorder and other NDD candidates. The interactome is coregulated in developing human neocortex. We suggest that a gene as pleiotropic and highly regulated as MET, together with its interactome, is biologically relevant in normal and pathophysiological contexts, affecting central and peripheral phenotypes that contribute to NDD risk and clinical symptoms.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Encéfalo/metabolismo , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Transtorno do Espectro Autista/imunologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica , Humanos , Macaca mulatta , Camundongos , Transtornos do Neurodesenvolvimento/imunologia , Plasticidade Neuronal , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-met/imunologia , Transdução de Sinais , Sinapses/metabolismo
8.
eNeuro ; 3(4)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27595133

RESUMO

MET, a pleiotropic receptor tyrosine kinase implicated in autism risk, influences multiple neurodevelopmental processes. There is a knowledge gap, however, in the molecular mechanism through which MET mediates developmental events related to disorder risk. In the neocortex, MET is expressed transiently during periods of peak dendritic outgrowth and synaptogenesis, with expression enriched at developing synapses, consistent with demonstrated roles in dendritic morphogenesis, modulation of spine volume, and excitatory synapse development. In a recent coimmunoprecipitation/mass spectrometry screen, ß-catenin was identified as part of the MET interactome in developing neocortical synaptosomes. Here, we investigated the influence of the MET/ß-catenin complex in mouse neocortical synaptogenesis. Western blot analysis confirms that MET and ß-catenin coimmunoprecipitate, but N-cadherin is not associated with the MET complex. Following stimulation with hepatocyte growth factor (HGF), ß-catenin is phosphorylated at tyrosine(142) (Y142) and dissociates from MET, accompanied by an increase in ß-catenin/N-cadherin and MET/synapsin 1 protein complexes. In neocortical neurons in vitro, proximity ligation assays confirmed the close proximity of these proteins. Moreover, in neurons transfected with synaptophysin-GFP, HGF stimulation increases the density of synaptophysin/bassoon (a presynaptic marker) and synaptophysin/PSD-95 (a postsynaptic marker) clusters. Mutation of ß-catenin at Y142 disrupts the dissociation of the MET/ß-catenin complex and prevents the increase in clusters in response to HGF. The data demonstrate a new mechanism for the modulation of synapse formation, whereby MET activation induces an alignment of presynaptic and postsynaptic elements that are necessary for assembly and formation of functional synapses by subsets of neocortical neurons that express MET/ß-catenin complex.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Neocórtex/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Sinapses/metabolismo , beta Catenina/metabolismo , Animais , Western Blotting , Caderinas/metabolismo , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Feminino , Guanilato Quinases/metabolismo , Imunoprecipitação , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Neocórtex/citologia , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Sinaptofisina/metabolismo
9.
Biol Psychiatry ; 80(12): 933-942, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27086544

RESUMO

BACKGROUND: Atypical synapse development and plasticity are implicated in many neurodevelopmental disorders (NDDs). NDD-associated, high-confidence risk genes have been identified, yet little is known about functional relationships at the level of protein-protein interactions, which are the dominant molecular bases responsible for mediating circuit development. METHODS: Proteomics in three independent developing neocortical synaptosomal preparations identified putative interacting proteins of the ligand-activated MET receptor tyrosine kinase, an autism risk gene that mediates synapse development. The candidates were translated into interactome networks and analyzed bioinformatically. Additionally, three independent quantitative proximity ligation assays in cultured neurons and four independent immunoprecipitation analyses of synaptosomes validated protein interactions. RESULTS: Approximately 11% (8/72) of MET-interacting proteins, including SHANK3, SYNGAP1, and GRIN2B, are associated with NDDs. Proteins in the MET interactome were translated into a novel MET interactome network based on human protein-protein interaction databases. High-confidence genes from different NDD datasets that encode synaptosomal proteins were analyzed for being enriched in MET interactome proteins. This was found for autism but not schizophrenia, bipolar disorder, major depressive disorder, or attention-deficit/hyperactivity disorder. There is correlated gene expression between MET and its interactive partners in developing human temporal and visual neocortices but not with highly expressed genes that are not in the interactome. Proximity ligation assays and biochemical analyses demonstrate that MET-protein partner interactions are dynamically regulated by receptor activation. CONCLUSIONS: The results provide a novel molecular framework for deciphering the functional relations of key regulators of synaptogenesis that contribute to both typical cortical development and to NDDs.


Assuntos
Neocórtex/crescimento & desenvolvimento , Neocórtex/metabolismo , Transtornos do Neurodesenvolvimento/genética , Neurogênese/fisiologia , Mapas de Interação de Proteínas/genética , Proteômica/métodos , Proteínas Proto-Oncogênicas c-met/genética , Sinapses/fisiologia , Células Cultivadas , Humanos , Neurônios
10.
Dev Neurobiol ; 76(10): 1160-81, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26818605

RESUMO

Hepatocyte growth factor (HGF) activation of the MET receptor tyrosine kinase influences multiple neurodevelopmental processes. Evidence from human imaging and mouse models shows that, in the forebrain, disruptions in MET signaling alter circuit formation and function. One likely means of modulation is by controlling neuron maturation. Here, we examined the signaling mechanisms through which MET exerts developmental effects in the neocortex. In situ hybridization revealed that hgf is located near MET-expressing neurons, including deep neocortical layers and periventricular zones. Western blot analyses of neocortical crude membranes demonstrated that HGF-induced MET autophosphorylation peaks during synaptogenesis, with a striking reduction in activation between P14 and P17 just before pruning. In vitro analysis of postnatal neocortical neurons assessed the roles of intracellular signaling following MET activation. There is rapid, HGF-induced phosphorylation of MET, ERK1/2, and Akt that is accompanied by two major morphological changes: increases in total dendritic growth and synapse density. Selective inhibition of each signaling pathway altered only one of the two distinct events. MAPK/ERK pathway inhibition significantly reduced the HGF-induced increase in dendritic length, but had no effect on synapse density. In contrast, inhibition of the PI3K/Akt pathway reduced HGF-induced increases in synapse density, with no effect on dendritic length. The data reveal a key role for MET activation during the period of neocortical neuron growth and synaptogenesis, with distinct biological outcomes mediated via discrete MET-linked intracellular signaling pathways in the same neurons. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1160-1181, 2016.


Assuntos
Dendritos/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neocórtex/crescimento & desenvolvimento , Neocórtex/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Sinapses/metabolismo , Animais , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Dendritos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Imuno-Histoquímica , Hibridização In Situ , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Neocórtex/citologia , Neocórtex/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Fotomicrografia , RNA Mensageiro/metabolismo , Sinapses/efeitos dos fármacos
11.
J Comp Neurol ; 521(14): 3241-59, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23787772

RESUMO

MET, a replicated autism risk gene, encodes a pleiotropic receptor tyrosine kinase implicated in multiple cellular processes during development and following injury. Previous studies suggest that Met modulates excitatory synapse development in the neocortex and hippocampus, although the underlying mechanism is unknown. The peak of Met expression corresponds to the period of process outgrowth and synaptogenesis, with robust expression in hippocampal and neocortical neuropil. Resolving whether neuropil expression represents presynaptic, postsynaptic or glial localization provides insight into potential mechanisms of Met action. The subcellular distribution of Met was characterized using complementary ultrastructural, in situ proximity ligation assay (PLA), and biochemical approaches. At postnatal day (P) 7, immunoelectron microscopy revealed near-equivalent proportions of Met-immunoreactive pre- (axons and terminals) and postsynaptic (dendritic shafts and spines) profiles in the stratum radiatum in the hippocampal CA1 region. Staining was typically in elements in which the corresponding pre- or postsynaptic apposition was unlabeled. By P21, Met-immunoreactive presynaptic profiles predominated and ~20% of Met-expressing profiles were glial. A different distribution of Met-immunoreactive profiles was observed in layer V of somatosensory cortex: Met-labeled spines were rare and a smaller proportion of glial profiles expressed Met. Strikingly, Met-immunoreactive presynaptic profiles predominated over postsynaptic profiles as early as P7. PLA analysis of neurons in vitro and biochemical analysis of tissue subsynaptic fractions confirmed the localization of Met in specific synaptic subcompartments. The study demonstrates that Met is enriched at synapses during development and its activation may modulate synapse formation and stability through both pre- and postsynaptic mechanisms.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/crescimento & desenvolvimento , Neocórtex/crescimento & desenvolvimento , Receptores Proteína Tirosina Quinases/metabolismo , Sinapses/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Dendritos/metabolismo , Dendritos/ultraestrutura , Embrião de Mamíferos , Feminino , Hipocampo/citologia , Hipocampo/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Neocórtex/citologia , Neocórtex/embriologia , Neurópilo/metabolismo , Neurópilo/ultraestrutura , Gravidez , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/ultraestrutura , Frações Subcelulares/metabolismo , Sinapses/ultraestrutura
12.
J Neurodev Disord ; 3(3): 282-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21509596

RESUMO

The validity for assigning disorder risk to an autism spectrum disorder (ASD) candidate gene comes from convergent genetic, clinical, and developmental neurobiology data. Here, we review these lines of evidence from multiple human genetic studies, and non-human primate and mouse experiments that support the conclusion that the MET receptor tyrosine kinase (RTK) functions to influence synapse development in circuits relevant to certain core behavioral domains of ASD. There is association of both common functional alleles and rare copy number variants that impact levels of MET expression in the human cortex. The timing of Met expression is linked to axon terminal outgrowth and synaptogenesis in the developing rodent and primate forebrain, and both in vitro and in vivo studies implicate this RTK in dendritic branching, spine maturation, and excitatory connectivity in the neocortex. This impact can occur in a cell-nonautonomous fashion, emphasizing the unique role that Met plays in specific circuits relevant to ASD.

13.
Autism Res ; 4(1): 68-83, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21328570

RESUMO

Candidate risk genes for autism spectrum disorder (ASD) have been identified, but the challenge of determining their contribution to pathogenesis remains. We previously identified two ASD risk genes encoding the receptor tyrosine kinase MET and the urokinase plasminogen activator receptor (PLAUR), which is thought to modulate availability of the MET ligand. We also reported a role for Met signaling in cortical interneuron development in vitro and a reduction of these neurons in uPAR (mouse ortholog of PLAUR) null mice, suggesting that disruption of either gene impacts cortical development similarly. Here, we modify this conclusion, reporting that interneuron numbers are unchanged in the neocortex of Met(fx/fx) / Dlx5/6(cre) mice, in which Met is ablated from cells arising from the ventral telencephalon (VTel). Consistent with this, Met transcript is not detected in the VTel during interneuron genesis and migration; furthermore, during the postnatal period of interneuron maturation, Met is co-expressed in glutamatergic projection neurons, but not interneurons. Low levels of Met protein are expressed in the VTel at E12.5 and E14.5, likely reflecting the arrival of Met containing corticofugal axons. Met expression, however, is induced in E12.5 VTel cells after 2 days in vitro, perhaps underlying discrepancies between observations in vitro and in Met(fx/fx) / Dlx5/6(cre) mice. We suggest that, in vivo, Met impacts the development of cortical projection neurons, whereas uPAR influences interneuron maturation. An altered balance between excitation and inhibition has been postulated as a biological mechanism for ASD; this imbalance could arise from different risk genes differentially affecting either or both elements.


Assuntos
Córtex Cerebral/fisiopatologia , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/fisiopatologia , Proteínas Proto-Oncogênicas c-met/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Alelos , Animais , Contagem de Células , Movimento Celular/genética , Córtex Cerebral/patologia , Criança , Transtornos Globais do Desenvolvimento Infantil/patologia , Expressão Gênica/genética , Estudos de Associação Genética , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Hibridização In Situ , Técnicas In Vitro , Interneurônios/patologia , Interneurônios/fisiologia , Malformações do Desenvolvimento Cortical/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Transdução de Sinais/genética , Regulação para Cima/genética
14.
Neural Dev ; 5: 32, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21122108

RESUMO

BACKGROUND: In an effort to identify genes that specify the mammalian forebrain, we used a comparative approach to identify mouse homologs of transcription factors expressed in developing Caenorhabditis elegans GABAergic neurons. A cell-specific microarray profiling study revealed a set of transcription factors that are highly expressed in embryonic C. elegans GABAergic neurons. RESULTS: Bioinformatic analyses identified mouse protein homologs of these selected transcripts and their expression pattern was mapped in the mouse embryonic forebrain by in situ hybridization. A review of human homologs indicates several of these genes are potential candidates in neurodevelopmental disorders. CONCLUSIONS: Our comparative approach has revealed several novel candidates that may serve as future targets for studies of mammalian forebrain development.


Assuntos
Perfilação da Expressão Gênica , Neurogênese/genética , Neurônios/fisiologia , Prosencéfalo/fisiologia , Homologia de Sequência de Aminoácidos , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Separação Celular , Mapeamento Cromossômico , Citometria de Fluxo , Humanos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Ácido gama-Aminobutírico/metabolismo
15.
J Comp Neurol ; 518(21): 4463-78, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20853516

RESUMO

Human genetic findings and murine neuroanatomical expression mapping have intersected to implicate Met receptor tyrosine kinase signaling in the development of forebrain circuits controlling social and emotional behaviors that are atypical in autism-spectrum disorders (ASD). To clarify roles for Met signaling during forebrain circuit development in vivo, we generated mutant mice (Emx1(Cre)/Met(fx/fx)) with an Emx1-Cre-driven deletion of signaling-competent Met in dorsal pallially derived forebrain neurons. Morphometric analyses of Lucifer yellow-injected pyramidal neurons in postnatal day 40 anterior cingulate cortex (ACC) revealed no statistically significant changes in total dendritic length but a selective reduction in apical arbor length distal to the soma in Emx1(Cre)/Met(fx/fx) neurons relative to wild type, consistent with a decrease in the total tissue volume sampled by individual arbors in the cortex. The effects on dendritic structure appear to be circuit-selective, insofar as basal arbor length was increased in Emx1(Cre)/Met(fx/fx) layer 2/3 neurons. Spine number was not altered on the Emx1(Cre)/Met(fx/fx) pyramidal cell populations studied, but spine head volume was significantly increased (∼20%). Cell-nonautonomous, circuit-level influences of Met signaling on dendritic development were confirmed by studies of medium spiny neurons (MSN), which do not express Met but receive Met-expressing corticostriatal afferents during development. Emx1(Cre)/Met(fx/fx) MSN exhibited robust increases in total arbor length (∼20%). As in the neocortex, average spine head volume was also increased (∼12%). These data demonstrate that a developmental loss of presynaptic Met receptor signaling can affect postsynaptic morphogenesis and suggest a mechanism whereby attenuated Met signaling could disrupt both local and long-range connectivity within circuits relevant to ASD.


Assuntos
Dendritos/ultraestrutura , Espinhas Dendríticas/ultraestrutura , Proteínas Proto-Oncogênicas c-met/deficiência , Transdução de Sinais/fisiologia , Animais , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Dendritos/metabolismo , Espinhas Dendríticas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Células Piramidais/citologia , Células Piramidais/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura
16.
J Comp Neurol ; 513(5): 511-31, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19226509

RESUMO

The establishment of appropriate neural circuitry depends on the coordination of multiple developmental events across space and time. These events include proliferation, migration, differentiation, and survival-all of which can be mediated by hepatocyte growth factor (HGF) signaling through the Met receptor tyrosine kinase. We previously found a functional promoter variant of the MET gene to be associated with autism spectrum disorder, suggesting that forebrain circuits governing social and emotional function may be especially vulnerable to developmental disruptions in HGF/Met signaling. However, little is known about the spatiotemporal distribution of Met expression in the forebrain during the development of such circuits. To advance our understanding of the neurodevelopmental influences of Met activation, we employed complementary Western blotting, in situ hybridization, and immunohistochemistry to comprehensively map Met transcript and protein expression throughout perinatal and postnatal development of the mouse forebrain. Our studies reveal complex and dynamic spatiotemporal patterns of expression during this period. Spatially, Met transcript is localized primarily to specific populations of projection neurons within the neocortex and in structures of the limbic system, including the amygdala, hippocampus, and septum. Met protein appears to be principally located in axon tracts. Temporally, peak expression of transcript and protein occurs during the second postnatal week. This period is characterized by extensive neurite outgrowth and synaptogenesis, supporting a role for the receptor in these processes. Collectively, these data suggest that Met signaling may be necessary for the appropriate wiring of forebrain circuits, with particular relevance to the social and emotional dimensions of behavior.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , Prosencéfalo/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fatores Etários , Tonsila do Cerebelo/crescimento & desenvolvimento , Tonsila do Cerebelo/metabolismo , Animais , Animais Recém-Nascidos , Transtorno Autístico/genética , Western Blotting , Diferenciação Celular , Embrião de Mamíferos , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Sistema Límbico/crescimento & desenvolvimento , Sistema Límbico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Reação em Cadeia da Polimerase , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
17.
J Comp Neurol ; 489(4): 449-66, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16025458

RESUMO

We have previously shown that in adult mice with a null mutation in the urokinase-type plasminogen activator receptor (uPAR) gene, maintained on a C57BL/6J/129Sv background, there is a selective loss of GABAergic interneurons in anterior cingulate and parietal cortex, with the parvalbumin-expressing subpopulation preferentially affected. Here, we performed a more detailed anatomical analysis of uPAR(-/-) mutation on the congenic C57BL/6J background. With glutamic acid decarboxylase-67 and gamma-aminobutyric acid (GABA) immunostaining, there is a similar region-selective loss of cortical interneurons in the congenic uPAR(-/-) mice from the earliest age examined (P21). In contrast, the loss of parvalbumin-immunoreactive cells is observed only in adult cortex, and the extent of this loss is less than in the mixed background. Moreover, earlier in development, although there are normal numbers of parvalbumin cells in the uPAR(-/-) cortex, fewer cells coexpress GABA, suggesting that the parvalbumin subpopulation migrates appropriately to the cortex, but does not differentiate normally. Among the other forebrain regions examined, only the adult hippocampus shows a loss of GABAergic interneurons, although the somatostatin, rather than the parvalbumin, subpopulation contributes to this loss. The data suggest that uPAR function is necessary for the normal development of a subpopulation of GABAergic neurons in the telencephalon. It is likely that the late-onset parvalbumin phenotype is due to the effects of an altered local environment on selectively vulnerable neurons and that the extent of this loss is strain dependent. Thus, an interplay between complex genetic factors and the environment may influence the phenotypic impact of the uPAR mutation both pre- and postnatally.


Assuntos
Envelhecimento/metabolismo , Diferenciação Celular/genética , Interneurônios/metabolismo , Receptores de Superfície Celular/genética , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/metabolismo , Ácido gama-Aminobutírico/biossíntese , Envelhecimento/genética , Animais , Animais Recém-Nascidos , Contagem de Células , Morte Celular/fisiologia , Movimento Celular/fisiologia , Feminino , Glutamato Descarboxilase/metabolismo , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Imuno-Histoquímica , Interneurônios/citologia , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parvalbuminas/metabolismo , Fenótipo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Somatostatina/metabolismo , Especificidade da Espécie , Telencéfalo/citologia
18.
Trends Neurosci ; 27(7): 400-6, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15219739

RESUMO

Neurodevelopmental disorders typically have complex endophenotypes, which can include abnormalities in neuronal excitability, processing of complex information, as well as behaviors such as anxiety and social interactions. Converging experimental and clinical evidence suggests that altered interneuron development may underlie part of the pathophysiological process of such disorders. Consistent with this, mice with abnormal hepatocyte growth factor signaling exhibit disturbances in the development of specific interneuron subclasses that are paralleled by seizure activity and a complex behavioral phenotype. Mutations in molecules that regulate different aspects of interneuron development could provide the heterogeneity in genetic susceptibility that, when combined with environmental disturbances, results in a phenotypic spectrum that serves as the hallmark pathophysiology for autism, mental retardation, schizophrenia and other neurodevelopmental disorders.


Assuntos
Encefalopatias/patologia , Deficiências do Desenvolvimento/patologia , Interneurônios/patologia , Neocórtex/anormalidades , Neocórtex/patologia , Animais , Encefalopatias/fisiopatologia , Criança , Deficiências do Desenvolvimento/fisiopatologia , Humanos
19.
Mol Cell Neurosci ; 24(3): 725-40, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14664821

RESUMO

The limbic system-associated membrane protein (LAMP) is a glycosylphosphatidylinositol-anchored glycoprotein with three immunoglobulin (Ig) domains that can either enhance or inhibit neurite outgrowth depending upon the neuronal population examined. In the present study, we investigate the domains responsible for these activities. Domain deletion revealed that the N-terminal IgI domain is necessary and sufficient for the neurite-promoting activity observed in hippocampal neurons. In contrast, inhibition of neurite outgrowth in SCG neurons, which is mediated by heterophilic interactions, requires full-length LAMP, although selective inhibition of the second Ig domain, but not the first or third domains, prevented the inhibitory effect. This indicates that the IgII domain of LAMP harbors the neurite-inhibiting activity, but only in the context of the full-length configuration. Covasphere-binding analyses demonstrate IgI/IgI interactions, but no interaction between IgII and any other domain, consistent with the biological activities that each domain mediates. The data suggest that LAMP may serve as a bifunctional guidance molecule, with distinct structural domains contributing to the promotion and inhibition of neurite outgrowth.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Diferenciação Celular/imunologia , Sistema Nervoso/embriologia , Neuritos/metabolismo , Animais , Sítios de Ligação/imunologia , Células CHO , Moléculas de Adesão Celular Neuronais/genética , Cricetinae , Feminino , Proteínas Ligadas por GPI , Hipocampo/citologia , Hipocampo/embriologia , Hipocampo/metabolismo , Imunoglobulinas/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Neuritos/ultraestrutura , Gravidez , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Ratos , Ratos Sprague-Dawley , Deleção de Sequência , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/embriologia , Gânglio Cervical Superior/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...