Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 358
Filtrar
1.
Curr Opin Genet Dev ; 68: 64-70, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33756113

RESUMO

Neurexins are central to trans-synaptic cell adhesion and signaling during synapse specification and maintenance. The past two decades of human genetics research have identified structural variations in the neurexin gene family, in particular NRXN1 copy number variants (CNVs), implicated in multiple neuropsychiatric and developmental disorders. The heterogeneity and reduced penetrance of NRXN1 deletions, in addition to the pleiotropic, circuit-specific functions of NRXN1, present substantial obstacles to understanding how compromised NRXN1 function predisposes individuals to neuropsychiatric disorders. Here, we provide an updated review of NRXN1 genetics in disease, followed by recently published work using both human induced pluripotent stem cell (iPSC) derived systems and animal models to understand the mechanisms of disease pathophysiology. Finally, we suggest our outlook on how the field should progress to improve our understanding of neurexin mediated disease pathogenesis. We believe that understanding how structural genetic variants in NRXN1 contribute to disease pathophysiology requires parallel approaches in iPSC and mouse model systems, each leveraging their unique strengths - analysis of genetic interactions and background effects in iPSCs and neural circuit and behavioral analysis in mice.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Variações do Número de Cópias de DNA , Transtornos Mentais/genética , Modelos Biológicos , Moléculas de Adesão de Célula Nervosa/fisiologia , Animais , Genótipo , Humanos , Camundongos , Mutação , Fenótipo
2.
J Biol Chem ; 296: 100372, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33548223

RESUMO

Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1-/- mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1ß) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1-/- mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1-/- mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Antígeno CD56/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Encéfalo/metabolismo , Antígeno CD56/fisiologia , Moléculas de Adesão Celular/metabolismo , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Ácidos Siálicos/metabolismo , Análise Espaço-Temporal , Sinapses/metabolismo
3.
Elife ; 92020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33355091

RESUMO

Synapse formation and regulation require signaling interactions between pre- and postsynaptic proteins, notably cell adhesion molecules (CAMs). It has been proposed that the functions of neuroligins (Nlgns), postsynaptic CAMs, rely on the formation of trans-synaptic complexes with neurexins (Nrxns), presynaptic CAMs. Nlgn3 is a unique Nlgn isoform that localizes at both excitatory and inhibitory synapses. However, Nlgn3 function mediated via Nrxn interactions is unknown. Here we demonstrate that Nlgn3 localizes at postsynaptic sites apposing vesicular glutamate transporter 3-expressing (VGT3+) inhibitory terminals and regulates VGT3+ inhibitory interneuron-mediated synaptic transmission in mouse organotypic slice cultures. Gene expression analysis of interneurons revealed that the αNrxn1+AS4 splice isoform is highly expressed in VGT3+ interneurons as compared with other interneurons. Most importantly, postsynaptic Nlgn3 requires presynaptic αNrxn1+AS4 expressed in VGT3+ interneurons to regulate inhibitory synaptic transmission. Our results indicate that specific Nlgn-Nrxn signaling generates distinct functional properties at synapses.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Neurônios GABAérgicos/fisiologia , Hipocampo/fisiologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Animais , Região CA1 Hipocampal/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sinapses/fisiologia
4.
J Neurosci ; 40(44): 8438-8462, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33037075

RESUMO

Neurexins (Nrxns) and LAR-RPTPs (leukocyte common antigen-related protein tyrosine phosphatases) are presynaptic adhesion proteins responsible for organizing presynaptic machineries through interactions with nonoverlapping extracellular ligands. Here, we report that two members of the LAR-RPTP family, PTPσ and PTPδ, are required for the presynaptogenic activity of Nrxns. Intriguingly, Nrxn1 and PTPσ require distinct sets of intracellular proteins for the assembly of specific presynaptic terminals. In addition, Nrxn1α showed robust heparan sulfate (HS)-dependent, high-affinity interactions with Ig domains of PTPσ that were regulated by the splicing status of PTPσ. Furthermore, Nrxn1α WT, but not a Nrxn1α mutant lacking HS moieties (Nrxn1α ΔHS), inhibited postsynapse-inducing activity of PTPσ at excitatory, but not inhibitory, synapses. Similarly, cis expression of Nrxn1α WT, but not Nrxn1α ΔHS, suppressed the PTPσ-mediated maintenance of excitatory postsynaptic specializations in mouse cultured hippocampal neurons. Lastly, genetics analyses using male or female Drosophila Dlar and Dnrx mutant larvae identified epistatic interactions that control synapse formation and synaptic transmission at neuromuscular junctions. Our results suggest a novel synaptogenesis model whereby different presynaptic adhesion molecules combine with distinct regulatory codes to orchestrate specific synaptic adhesion pathways.SIGNIFICANCE STATEMENT We provide evidence supporting the physical interactions of neurexins with leukocyte common-antigen related receptor tyrosine phosphatases (LAR-RPTPs). The availability of heparan sulfates and alternative splicing of LAR-RPTPs regulate the binding affinity of these interactions. A set of intracellular presynaptic proteins is involved in common for Nrxn- and LAR-RPTP-mediated presynaptic assembly. PTPσ triggers glutamatergic and GABAergic postsynaptic differentiation in an alternative splicing-dependent manner, whereas Nrxn1α induces GABAergic postsynaptic differentiation in an alternative splicing-independent manner. Strikingly, Nrxn1α inhibits the glutamatergic postsynapse-inducing activity of PTPσ, suggesting that PTPσ and Nrxn1α might control recruitment of a different pool of postsynaptic machinery. Drosophila orthologs of Nrxns and LAR-RPTPs mediate epistatic interactions in controlling synapse structure and strength at neuromuscular junctions, underscoring the physiological significance in vivo.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Antígenos Comuns de Leucócito/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Potenciais Pós-Sinápticos Excitadores/fisiologia , Espaço Extracelular/metabolismo , Feminino , Células HEK293 , Humanos , Larva , Masculino , Camundongos , Conformação Molecular , Moléculas de Adesão de Célula Nervosa/metabolismo , Gravidez , Terminações Pré-Sinápticas/metabolismo , Ratos , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Transmissão Sináptica/fisiologia
5.
J Neurosci ; 40(42): 8088-8102, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32973045

RESUMO

Emerging evidence supports roles for secreted extracellular matrix proteins in boosting synaptogenesis, synaptic transmission, and synaptic plasticity. SPARCL1 (also known as Hevin), a secreted non-neuronal protein, was reported to increase synaptogenesis by simultaneously binding to presynaptic neurexin-1α and to postsynaptic neuroligin-1B, thereby catalyzing formation of trans-synaptic neurexin/neuroligin complexes. However, neurexins and neuroligins do not themselves mediate synaptogenesis, raising the question of how SPARCL1 enhances synapse formation by binding to these molecules. Moreover, it remained unclear whether SPARCL1 acts on all synapses containing neurexins and neuroligins or only on a subset of synapses, and whether it enhances synaptic transmission in addition to boosting synaptogenesis or induces silent synapses. To explore these questions, we examined the synaptic effects of SPARCL1 and their dependence on neurexins and neuroligins. Using mixed neuronal and glial cultures from neonatal mouse cortex of both sexes, we show that SPARCL1 selectively increases excitatory but not inhibitory synapse numbers, enhances excitatory but not inhibitory synaptic transmission, and augments NMDAR-mediated synaptic responses more than AMPAR-mediated synaptic responses. None of these effects were mediated by SPARCL1-binding to neurexins or neuroligins. Neurons from triple neurexin-1/2/3 or from quadruple neuroligin-1/2/3/4 conditional KO mice that lacked all neurexins or all neuroligins were fully responsive to SPARCL1. Together, our results reveal that SPARCL1 selectively boosts excitatory but not inhibitory synaptogenesis and synaptic transmission by a novel mechanism that is independent of neurexins and neuroligins.SIGNIFICANCE STATEMENT Emerging evidence supports roles for extracellular matrix proteins in boosting synapse formation and function. Previous studies demonstrated that SPARCL1, a secreted non-neuronal protein, promotes synapse formation in rodent and human neurons. However, it remained unclear whether SPARCL1 acts on all or on only a subset of synapses, induces functional or largely inactive synapses, and generates synapses by bridging presynaptic neurexins and postsynaptic neuroligins. Here, we report that SPARCL1 selectively induces excitatory synapses, increases their efficacy, and enhances their NMDAR content. Moreover, using rigorous genetic manipulations, we show that SPARCL1 does not require neurexins and neuroligins for its activity. Thus, SPARCL1 selectively boosts excitatory synaptogenesis and synaptic transmission by a novel mechanism that is independent of neurexins and neuroligins.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sinapses/fisiologia , Animais , Córtex Cerebral/citologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Neuroglia/metabolismo , Neurônios/metabolismo , Cultura Primária de Células , Receptores de Superfície Celular , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia
6.
Pharmacol Res ; 160: 105186, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32898689

RESUMO

Neuroplastic alterations are the key processes involved in adaptation and rehabilitation after all neurological injuries and pathologies. Being the central contributor to the developmental and adult neuroplasticity, the polysialylated form of Neural Cell Adhesion Molecule (PSA-NCAM) may prove to be a potential target to facilitate repair/regeneration after CNS injury and disease. Over the years, several experimental approaches have been developed to exploit the therapeutic potential of PSA-NCAM. Broadly, the studies focused on cell-transplantation strategies to alter PSA-NCAM properties at the injury site, injection of peptide based as well as synthetic PSA mimetics directly into the injury site or the application of PSA containing hydrogels and scaffolds as biomaterials. A comprehensive understanding of the PSA-based experimental approaches, as well as their pros and cons, is urgently required for successful implementation of this molecule in therapeutics. The current review, therefore, has been designed to give the readers a thorough account of all the diverse roles of PSA in the adult nervous system and the recent progress that has been made in developing PSA-based therapeutic approaches for neuroregeneration.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Plasticidade Neuronal/fisiologia , Ácidos Siálicos/farmacologia , Animais , Humanos , Regeneração Nervosa/efeitos dos fármacos , Moléculas de Adesão de Célula Nervosa/genética
7.
J Biol Chem ; 295(27): 9244-9262, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32434929

RESUMO

Calsyntenin-3 (Clstn3) is a postsynaptic adhesion molecule that induces presynaptic differentiation via presynaptic neurexins (Nrxns), but whether Nrxns directly bind to Clstn3 has been a matter of debate. Here, using LC-MS/MS-based protein analysis, confocal microscopy, RNAscope assays, and electrophysiological recordings, we show that ß-Nrxns directly interact via their LNS domain with Clstn3 and Clstn3 cadherin domains. Expression of splice site 4 (SS4) insert-positive ß-Nrxn variants, but not insert-negative variants, reversed the impaired Clstn3 synaptogenic activity observed in Nrxn-deficient neurons. Consistently, Clstn3 selectively formed complexes with SS4-positive Nrxns in vivo Neuron-specific Clstn3 deletion caused significant reductions in number of excitatory synaptic inputs. Moreover, expression of Clstn3 cadherin domains in CA1 neurons of Clstn3 conditional knockout mice rescued structural deficits in excitatory synapses, especially within the stratum radiatum layer. Collectively, our results suggest that Clstn3 links to SS4-positive Nrxns to induce presynaptic differentiation and orchestrate excitatory synapse development in specific hippocampal neural circuits, including Schaffer collateral afferents.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Animais , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/fisiologia , Cromatografia Líquida/métodos , Hipocampo/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Sinapses/metabolismo , Espectrometria de Massas em Tandem/métodos
8.
Development ; 147(4)2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31988188

RESUMO

Dendrites develop elaborate morphologies in concert with surrounding glia, but the molecules that coordinate dendrite and glial morphogenesis are mostly unknown. C. elegans offers a powerful model for identifying such factors. Previous work in this system examined dendrites and glia that develop within epithelia, similar to mammalian sense organs. Here, we focus on the neurons BAG and URX, which are not part of an epithelium but instead form membranous attachments to a single glial cell at the nose, reminiscent of dendrite-glia contacts in the mammalian brain. We show that these dendrites develop by retrograde extension, in which the nascent dendrite endings anchor to the presumptive nose and then extend by stretching during embryo elongation. Using forward genetic screens, we find that dendrite development requires the adhesion protein SAX-7/L1CAM and the cytoplasmic protein GRDN-1/CCDC88C to anchor dendrite endings at the nose. SAX-7 acts in neurons and glia, while GRDN-1 acts in glia to non-autonomously promote dendrite extension. Thus, this work shows how glial factors can help to shape dendrites, and identifies a novel molecular mechanism for dendrite growth by retrograde extension.


Assuntos
Encéfalo/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Proteínas dos Microfilamentos/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Neuroglia/fisiologia , Alelos , Animais , Caenorhabditis elegans/fisiologia , Membrana Celular/fisiologia , Citoplasma/fisiologia , Dendritos/fisiologia , Epitélio/fisiologia , Neurogênese , Isoformas de Proteínas , Células Receptoras Sensoriais/fisiologia
9.
Brain Res Bull ; 155: 92-101, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31812781

RESUMO

The posterodorsal medial amygdala (MePD) has a high concentration of receptors for gonadal hormones, is a sexually dimorphic region and dynamically controls the reproductive behavior of both males and females. Neurotrophic factors can promote dendritic spine remodeling and change synaptic input strength in a region-specific manner. Here, we analyzed the gene and protein expression of brain-derived neurotrophic factor (BDNF), insulin-like growth factor-I (IGF-1), polysialylated neural cell adhesion molecule (PSA-NCAM) and Ephrin-A4 in the MePD of adult males and females in diestrus, proestrus and estrus using real-time qPCR and fluorescent immunohistochemistry. The first approach showed their amplification except for Igf1 and the latter revealed that BDNF, IGF-1, PSA-NCAM and Ephrin-A4 are expressed in the MePD of the adult rats. Protein expression of these neurotrophic factors showed no differences between groups. However, proestrus females displayed a higher number of labelled puncta than males for BDNF expression and diestrus females for IGF-1 expression. In conjunction, results indicate that IGF-1 might be released rather than synthetized in the MePD, and the expression of specific neurotrophic factors varies specifically during proestrus. The dynamic modulation of BDNF and IGF-1 during this cyclic phase is coincident with synaptic changes and spine density remodeling in the MePD, the disinhibition of gonadotrophin secretion for ovulation and the display of sexual behavior.


Assuntos
Complexo Nuclear Corticomedial/fisiologia , Ciclo Estral , Fatores de Crescimento Neural/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Efrina-A4/análise , Efrina-A4/fisiologia , Feminino , Expressão Gênica , Masculino , Moléculas de Adesão de Célula Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Ratos Wistar , Caracteres Sexuais
10.
PLoS Biol ; 17(12): e3000522, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31805038

RESUMO

In epithelia, tricellular vertices are emerging as important sites for the regulation of epithelial integrity and function. Compared to bicellular contacts, however, much less is known. In particular, resident proteins at tricellular vertices were identified only at occluding junctions, with none known at adherens junctions (AJs). In a previous study, we discovered that in Drosophila embryos, the adhesion molecule Sidekick (Sdk), well-known in invertebrates and vertebrates for its role in the visual system, localises at tricellular vertices at the level of AJs. Here, we survey a wide range of Drosophila epithelia and establish that Sdk is a resident protein at tricellular AJs (tAJs), the first of its kind. Clonal analysis showed that two cells, rather than three cells, contributing Sdk are sufficient for tAJ localisation. Super-resolution imaging using structured illumination reveals that Sdk proteins form string-like structures at vertices. Postulating that Sdk may have a role in epithelia where AJs are actively remodelled, we analysed the phenotype of sdk null mutant embryos during Drosophila axis extension using quantitative methods. We find that apical cell shapes are abnormal in sdk mutants, suggesting a defect in tissue remodelling during convergence and extension. Moreover, adhesion at apical vertices is compromised in rearranging cells, with apical tears in the cortex forming and persisting throughout axis extension, especially at the centres of rosettes. Finally, we show that polarised cell intercalation is decreased in sdk mutants. Mathematical modelling of the cell behaviours supports the notion that the T1 transitions of polarised cell intercalation are delayed in sdk mutants, in particular in rosettes. We propose that this delay, in combination with a change in the mechanical properties of the converging and extending tissue, causes the abnormal apical cell shapes in sdk mutant embryos.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Proteínas do Olho/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Junções Íntimas/fisiologia , Junções Aderentes/metabolismo , Animais , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Polaridade Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Epitélio/metabolismo , Proteínas do Olho/fisiologia , Proteínas de Membrana/metabolismo , Moléculas de Adesão de Célula Nervosa/fisiologia
11.
Cereb Cortex ; 29(4): 1439-1459, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522129

RESUMO

The neural cell adhesion molecule 2 (NCAM2) is encoded by a gene on chromosome 21 in humans. NCAM2 accumulates in synapses, but its role in regulation of synapse formation remains poorly understood. We demonstrate that an increase in NCAM2 levels results in increased instability of dendritic protrusions and reduced conversion of protrusions to dendritic spines in mouse cortical neurons. NCAM2 overexpression induces an increase in the frequency of submembrane Ca2+ spikes localized in individual dendritic protrusions and promotes propagation of submembrane Ca2+ spikes over segments of dendrites or the whole dendritic tree. NCAM2-dependent submembrane Ca2+ spikes are L-type voltage-gated Ca2+ channel-dependent, and their propagation but not initiation depends on the c-Src protein tyrosine kinase. Inhibition of initiation or propagation of NCAM2-dependent submembrane Ca2+ spikes reduces the NCAM2-dependent instability of dendritic protrusions. Synaptic boutons formed on dendrites of neurons with elevated NCAM2 expression are enriched in the protein marker of immature synapses GAP43, and the number of boutons with mature activity-dependent synaptic vesicle recycling is reduced. Our results indicate that synapse maturation is inhibited in NCAM2-overexpressing neurons and suggest that changes in NCAM2 levels and altered submembrane Ca2+ dynamics can cause defects in synapse maturation in Down syndrome and other brain disorders associated with abnormal NCAM2 expression.


Assuntos
Encéfalo/fisiologia , Proteína Tirosina Quinase CSK/fisiologia , Sinalização do Cálcio , Dendritos/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sinapses/fisiologia , Animais , Canais de Cálcio Tipo L/fisiologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Cultura Primária de Células
12.
Neuron ; 99(2): 329-344.e7, 2018 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-29983322

RESUMO

Pyramidal neurons express rich repertoires of leucine-rich repeat (LRR)-containing adhesion molecules with similar synaptogenic activity in culture. The in vivo relevance of this molecular diversity is unclear. We show that hippocampal CA1 pyramidal neurons express multiple synaptogenic LRR proteins that differentially distribute to the major excitatory inputs on their apical dendrites. At Schaffer collateral (SC) inputs, FLRT2, LRRTM1, and Slitrk1 are postsynaptically localized and differentially regulate synaptic structure and function. FLRT2 controls spine density, whereas LRRTM1 and Slitrk1 exert opposing effects on synaptic vesicle distribution at the active zone. All LRR proteins differentially affect synaptic transmission, and their combinatorial loss results in a cumulative phenotype. At temporoammonic (TA) inputs, LRRTM1 is absent; FLRT2 similarly controls functional synapse number, whereas Slitrk1 function diverges to regulate postsynaptic AMPA receptor density. Thus, LRR proteins differentially control synaptic architecture and function and act in input-specific combinations and a context-dependent manner to specify synaptic properties.


Assuntos
Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Técnicas de Cocultura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/ultraestrutura , Proteínas de Membrana/análise , Proteínas de Membrana/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso , Moléculas de Adesão de Célula Nervosa/análise , Moléculas de Adesão de Célula Nervosa/ultraestrutura , Ratos , Ratos Wistar , Sinapses/química , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia
13.
Mol Cells ; 41(5): 373-380, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29665671

RESUMO

Synapses and neural circuits form with exquisite specificity during brain development to allow the precise and appropriate flow of neural information. Although this property of synapses and neural circuits has been extensively investigated for more than a century, molecular mechanisms underlying this property are only recently being unveiled. Recent studies highlight several classes of cell-surface proteins as organizing hubs in building structural and functional architectures of specific synapses and neural circuits. In the present mini-review, we discuss recent findings on various synapse organizers that confer the distinct properties of specific synapse types and neural circuit architectures in mammalian brains, with a particular focus on the hippocampus and cerebellum.


Assuntos
Cerebelo/fisiologia , Hipocampo/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Adesão Celular , Cerebelo/citologia , Córtex Entorrinal/citologia , Córtex Entorrinal/fisiologia , Hipocampo/citologia , Humanos , Interneurônios/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Células de Purkinje/fisiologia
14.
Development ; 145(3)2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29361567

RESUMO

The assembly of functional neuronal circuits requires growth cones to extend in defined directions and recognize the correct synaptic partners. Homophilic adhesion between vertebrate Sidekick proteins promotes synapse formation between retinal neurons involved in visual motion detection. We show here that Drosophila Sidekick accumulates in specific synaptic layers of the developing motion detection circuit and is necessary for normal optomotor behavior. Sidekick is required in photoreceptors, but not in their target lamina neurons, to promote the alignment of lamina neurons into columns and subsequent sorting of photoreceptor axons into synaptic modules based on their precise spatial orientation. Sidekick is also localized to the dendrites of the direction-selective T4 and T5 cells, and is expressed in some of their presynaptic partners. In contrast to its vertebrate homologs, Sidekick is not essential for T4 and T5 to direct their dendrites to the appropriate layers or to receive synaptic contacts. These results illustrate a conserved requirement for Sidekick proteins in establishing visual motion detection circuits that is achieved through distinct cellular mechanisms in Drosophila and vertebrates.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/fisiologia , Proteínas do Olho/fisiologia , Percepção de Movimento/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas do Olho/genética , Feminino , Genes de Insetos , Masculino , Mutação , Moléculas de Adesão de Célula Nervosa/genética , Células Fotorreceptoras de Invertebrados/citologia , Sinapses/metabolismo , Vias Visuais/citologia , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/fisiologia
15.
Biochem Biophys Res Commun ; 494(1-2): 120-125, 2017 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-29050943

RESUMO

Pathogen avoidance behaviors are found throughout the animal kingdom and are important for animal's survival in nature. As a free-living nematode, C. elegans is exposed to a variety of microorganisms, including toxic or pathogenic bacteria, in soil. C. elegans can develop efficient avoidance responses to pathogenic bacteria to minimize the infection risk. However, the role of microRNAs (miRNAs) in pathogen avoidance in C. elegans remains unclear. In this report, we showed that the miRNA mir-67 was involved in a behavioral avoidance response to P. aeruginosa PA14. Exposure to P. aeruginosa PA14 induced the expression of mir-67 in worms. mir-67(n4899) mutants exhibited a reduced ability to avoid P. aeruginosa PA14. By combining quantitative proteomic analysis with miRNA target prediction algorithms, we identified SAX-7/L1CAM, which is transmembrane cell adhesion receptor molecule, as the target of mir-67. Silencing of sax-7 by RNAi on mir-67 mutants rescued avoidance behavioral. Our data demonstrate that the mir-67-SAX-7 pathway modulate the behavioral avoidance response to pathogens, thus providing a new perspective in the role of miRNAs in host-microbe interactions.


Assuntos
Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , MicroRNAs/genética , RNA de Helmintos/genética , Animais , Aprendizagem da Esquiva/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/fisiologia , Pseudomonas aeruginosa/patogenicidade , Transdução de Sinais
16.
J Neurosci ; 37(41): 9828-9843, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-28871037

RESUMO

The proper formation of synapses-specialized unitary structures formed between two neurons-is critical to mediating information flow in the brain. Synaptic cell adhesion molecules (CAMs) are thought to participate in the initiation of the synapse formation process. However, in vivo functional analysis demonstrates that most well known synaptic CAMs regulate synaptic maturation and plasticity rather than synapse formation, suggesting that either CAMs work synergistically in the process of forming synapses or more CAMs remain to be found. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters in co-cultures of human embryonic kidney 293 cells and hippocampal neurons cultured from newborn mice regardless of gender. PTPRO was enriched in the mouse brain and localized to postsynaptic sites at excitatory synapses. The overexpression of PTPRO in cultured hippocampal neurons increased the number of synapses and the frequency of miniature EPSCs (mEPSCs). The knock-down (KD) of PTPRO expression in cultured neurons by short hairpin RNA (shRNA) reduced the number of synapses and the frequencies of the mEPSCs. The effects of shRNA KD were rescued by expressing either full-length PTPRO or a truncated PTPRO lacking the cytoplasmic domain. Consistent with these results, the N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in the co-culture assay. Our data show that PTPRO is a synaptic CAM that serves as a potent initiator of the formation of excitatory synapses.SIGNIFICANCE STATEMENT The formation of synapses is critical for the brain to execute its function and synaptic cell adhesion molecules (CAMs) play essential roles in initiating the formation of synapses. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters. Using loss-of-function and gain-of-function approaches, we show that PTPRO promotes the formation of excitatory synapses. The N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in cultured hippocampal neurons and the co-culture assay. Together, our data show that PTPRO is a synaptic CAM that serves as a potent initiator of synapse formation.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/fisiologia , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Técnicas de Cocultura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas de Silenciamento de Genes , Células HEK293 , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Moléculas de Adesão de Célula Nervosa/genética , Técnicas de Patch-Clamp , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética
17.
Front Neural Circuits ; 11: 19, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28386219

RESUMO

The neural cell adhesion molecule (NCAM), has been shown to be an obligate regulator of synaptic stability and pruning during critical periods of cortical maturation. However, the functional consequences of NCAM deletion on the organization of inhibitory circuits in cortex are not known. In vesicular gamma-amino butyric acid (GABA) transporter (VGAT)-channelrhodopsin2 (ChR2)-enhanced yellow fluorescent protein (EYFP) transgenic mice, NCAM is expressed postnatally at perisomatic synaptic puncta of EYFP-labeled parvalbumin, somatostatin and calretinin-positive interneurons, and in the neuropil in the anterior cingulate cortex (ACC). To investigate how NCAM deletion affects the spatial organization of inhibitory inputs to pyramidal cells, we used laser scanning photostimulation in brain slices of VGAT-ChR2-EYFP transgenic mice crossed to either NCAM-null or wild type (WT) mice. Laser scanning photostimulation revealed that NCAM deletion increased the strength of close-in inhibitory connections to layer 2/3 pyramidal cells of the ACC. In addition, in NCAM-null mice, the intrinsic excitability of pyramidal cells increased, whereas the intrinsic excitability of GABAergic interneurons did not change. The increase in inhibitory tone onto pyramidal cells, and the increased pyramidal cell excitability in NCAM-null mice will alter the delicate coordination of excitation and inhibition (E/I coordination) in the ACC, and may be a factor contributing to circuit dysfunction in diseases such as schizophrenia and bipolar disorder, in which NCAM has been implicated.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Giro do Cíngulo/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Células Piramidais/fisiologia , Animais , Giro do Cíngulo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Microscopia Confocal , Inibição Neural/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/citologia
18.
Nat Neurosci ; 20(5): 690-699, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28346453

RESUMO

Epigenetic mechanisms regulate the formation, consolidation and reconsolidation of memories. However, the signaling path from neuronal activation to epigenetic modifications within the memory-related brain circuit remains unknown. We report that learning induces long-lasting histone modifications in hippocampal memory-activated neurons to regulate memory stability. Neuronal activity triggers a late-onset shift in Nrxn1 splice isoform choice at splicing site 4 by accumulating a repressive histone marker, H3K9me3, to modulate the splicing process. Activity-dependent phosphorylation of p66α via AMP-activated protein kinase recruits HDAC2 and Suv39h1 to establish repressive histone markers and changes the connectivity of the activated neurons. Removal of Suv39h1 abolished the activity-dependent shift in Nrxn1 splice isoform choice and reduced the stability of established memories. We uncover a cell-autonomous process for memory preservation in which memory-related neurons initiate a late-onset reduction of their rewiring capacities through activity-induced histone modifications.


Assuntos
Código das Histonas/fisiologia , Histonas/fisiologia , Memória/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Técnicas de Cocultura , Condicionamento Psicológico/fisiologia , Epigênese Genética , Feminino , Fatores de Transcrição GATA , Hipocampo/fisiologia , Histona Desacetilase 2/metabolismo , Histonas/metabolismo , Aprendizagem/fisiologia , Masculino , Metiltransferases/metabolismo , Camundongos , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Cultura Primária de Células , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/metabolismo
19.
Neurosci Res ; 116: 29-38, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27884699

RESUMO

Cell adhesion molecules (CAMs) play essential roles in the central nervous system, where some families are involved in synaptic development and function. These synaptic adhesion molecules (SAMs) are involved in the regulation of synaptic plasticity, and the formation of neuronal networks. Recent findings from studies examining the consequences of sleep loss suggest that these molecules are candidates to act in sleep regulation. This review highlights the experimental data that lead to the identification of SAMs as potential sleep regulators, and discusses results supporting that specific SAMs are involved in different aspects of sleep regulation. Further, some potential mechanisms by which SAMs may act to regulate sleep are outlined, and the proposition that these molecules may serve as molecular machinery in the two sleep regulatory processes, the circadian and homeostatic components, is presented. Together, the data argue that SAMs regulate the neuronal plasticity that underlies sleep and wakefulness.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Sono/fisiologia , Animais , Comunicação Celular , Ritmo Circadiano , Humanos , Moléculas de Adesão de Célula Nervosa/genética , Neuroglia/fisiologia , Plasticidade Neuronal , Neurônios/fisiologia , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/fisiopatologia , Sinapses/metabolismo , Vigília/fisiologia
20.
Development ; 143(22): 4224-4235, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707798

RESUMO

The formation of synaptic connections during nervous system development requires the precise control of dendrite growth and synapse formation. Although glial cell line-derived neurotrophic factor (GDNF) and its receptor GFRα1 are expressed in the forebrain, the role of this system in the hippocampus remains unclear. Here, we investigated the consequences of GFRα1 deficiency for the development of hippocampal connections. Analysis of conditional Gfra1 knockout mice shows a reduction in dendritic length and complexity, as well as a decrease in postsynaptic density specializations and in the synaptic localization of postsynaptic proteins in hippocampal neurons. Gain- and loss-of-function assays demonstrate that the GDNF-GFRα1 complex promotes dendritic growth and postsynaptic differentiation in cultured hippocampal neurons. Finally, in vitro assays revealed that GDNF-GFRα1-induced dendrite growth and spine formation are mediated by NCAM signaling. Taken together, our results indicate that the GDNF-GFRα1 complex is essential for proper hippocampal circuit development.


Assuntos
Dendritos/fisiologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Hipocampo/crescimento & desenvolvimento , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurogênese/genética , Plasticidade Neuronal/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Embrião de Mamíferos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Complexos Multiproteicos/fisiologia , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Neurônios/fisiologia , Ligação Proteica , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...