Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 358
Filtrar
1.
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
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Dev Biol ; 419(2): 285-297, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27618756

RESUMO

Collective cell migration is the coordinated movement of cells, which organize tissues during morphogenesis, repair and some cancers. The motile cell membrane of the advancing front in collective cell migration is termed the Leading Edge. The embryonic development of the vertebrate and Drosophila hearts are both characterized by the coordinated medial migration of a bilateral cluster of mesodermal cells. In Drosophila, the cardioblasts form cohesive bilateral rows that migrate collectively as a unit towards the dorsal midline to form the dorsal vessel. We have characterized the collective cell migration of cardioblasts as an in vivo quantitative model to study the behaviour of the Leading Edge. We investigated whether guidance signalling through Slit and Netrin pathways plays a role in cell migration during heart development. Through time-lapse imaging and quantitative assessment of migratory behaviour of the cardioblasts in loss-of-function mutants, we demonstrate that both Slit and Netrin mediated signals are autonomously and concomitantly required to maximize migration velocity, filopodial and lamellipodial activities. Additionally, we show that another Slit and Netrin receptor, Dscam1, the role of which during heart development was previously unknown, is required for both normal migration of cardioblasts and luminal expansion. Leading edge behaviour analysis revealed a dosage dependent genetic interaction between Slit and Netrin receptors suggesting that downstream signalling through these receptors converge on a common output that increases leading edge activity of the cardioblasts. Finally, we found that guidance signalling maintains the balance between epithelial and mesenchymal characteristics of the migrating cardioblasts.


Assuntos
Movimento Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Coração/embriologia , Miócitos Cardíacos/citologia , Pseudópodes/fisiologia , Células-Tronco/citologia , Animais , Orientação de Axônios , Moléculas de Adesão Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Transição Epitelial-Mesenquimal , Proteínas Luminescentes/análise , Morfogênese , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Receptores de Netrina , Netrina-1 , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/fisiologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores Imunológicos/genética , Receptores Imunológicos/fisiologia , Transdução de Sinais , Imagem com Lapso de Tempo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Proteínas Roundabout
13.
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
14.
Nat Rev Immunol ; 6(6): 457-64, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16691243

RESUMO

The immune system must be highly regulated to obtain optimal immune responses for the elimination of pathogens without causing undue side effects. This tight regulation involves complex interactions between membrane proteins on leukocytes. Members of the signal-regulatory protein (SIRP) family, which are expressed mainly by myeloid cells, provide one example of these regulatory membrane proteins. There are three SIRP-family genes that encode proteins that have similar extracellular regions but different signalling potentials, and are therefore known as 'paired receptors'. In this Review, we describe recent studies defining the ligands of the SIRP-family members, with particular emphasis on relating the molecular interactions of these proteins to their role in immune-cell regulation.


Assuntos
Adjuvantes Imunológicos/fisiologia , Antígenos de Diferenciação/fisiologia , Glicoproteínas de Membrana/fisiologia , Família Multigênica , Moléculas de Adesão de Célula Nervosa/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores Imunológicos/fisiologia , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
15.
J Neurosci ; 33(2): 790-803, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23303955

RESUMO

The Neural cell adhesion molecule (NCAM) plays an important role in regulation of nervous system development. To expand our understanding of the molecular mechanisms via which NCAM influences differentiation of neurons, we used a yeast two-hybrid screening to search for new binding partners of NCAM and identified p21-activated kinase 1 (Pak1). We show that NCAM interacts with Pak1 in growth cones of neurons. The autophosphorylation and activity of Pak1 were enhanced when isolated growth cones were incubated with NCAM function triggering antibodies, which mimic the interaction between NCAM and its extracellular ligands. The association of Pak1 with cell membranes, the efficiency of Pak1 binding to its activators, and Pak1 activity were inhibited in brains of NCAM-deficient mice. NCAM-dependent Pak1 activation was abolished after lipid raft disruption, suggesting that NCAM promotes Pak1 activation in the lipid raft environment. Phosphorylation of the downstream Pak1 effectors LIMK1 and cofilin was reduced in growth cones from NCAM-deficient neurons, which was accompanied by decreased levels of filamentous actin and inhibited filopodium mobility in the growth cones. Dominant-negative Pak1 inhibited and constitutively active Pak1 enhanced the ability of neurons to increase neurite outgrowth in response to the extracellular ligands of NCAM. Our combined observations thus indicate that NCAM activates Pak1 to drive actin polymerization to promote neuronal differentiation.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/fisiologia , Actinas/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Química Encefálica , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , DNA/genética , Feminino , Cones de Crescimento/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Imunoprecipitação , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Neuritos/efeitos dos fármacos , Fosforilação , Ratos , Saccharomyces cerevisiae/genética , Transdução de Sinais/genética , Proteína cdc42 de Ligação ao GTP/genética , Quinases Ativadas por p21/genética
16.
Nat Rev Neurosci ; 9(1): 26-35, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18059411

RESUMO

Polysialic acid (PSA) is a cell-surface glycan with an enormous hydrated volume that serves to modulate the distance between cells. This regulation has direct effects on several cellular mechanisms that underlie the formation of the vertebrate nervous system, most conspicuously in the migration and differentiation of progenitor cells and the growth and targeting of axons. PSA is also involved in a number of plasticity-related responses in the adult CNS, including changes in circadian and hormonal patterns, adaptations to pain and stress, and aspects of learning and memory. The ability of PSA to increase the plasticity of neural cells is being exploited to improve the repair of adult CNS tissue.


Assuntos
Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Plasticidade Neuronal/fisiologia , Ácidos Siálicos/fisiologia , Vertebrados/embriologia , Vertebrados/crescimento & desenvolvimento , Animais , Axônios/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/fisiopatologia , Desenvolvimento Embrionário , Humanos , Aprendizagem/fisiologia , Memória/fisiologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/fisiologia , Ácidos Siálicos/metabolismo , Células-Tronco/citologia , Células-Tronco/fisiologia
17.
Stress ; 16(6): 647-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24010949

RESUMO

The neural cell adhesion molecule (NCAM) is a key regulator of brain plasticity. Substantial evidence indicates that NCAM is down-regulated by exposure to sustained stress and chronic stress seems to lead to increased aggression. In addition, constitutional NCAM deletion in mice has been shown to lead to increased intermale aggression and altered emotionality Forebrain-specific postnatal NCAM knockout was previously shown to impair cognitive function, particularly when animals were exposed to subchronic stress, but the effects on emotional and social behavior remain unclear. In this study, we investigated the potential interplay of a forebrain-specific postnatal NCAM deletion and exposure to different lengths of repeated stress (i.e. subchronic: 14 days; chronic: 29 days) on aggressive and emotional behavior. Our results show that postnatal deletion of NCAM in the forebrain leads to increased aggression and altered emotionality depending on the duration of stress, whereas conditional NCAM knockout has no basal impact on these behaviors. These findings support the involvement of NCAM in the regulation of emotional and aggressive behaviors, suggesting that diminished NCAM expression might be a critical vulnerability factor for the development of these behavioral alterations under repeated exposure to stress.


Assuntos
Agressão/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Prosencéfalo/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Ansiedade/metabolismo , Comportamento Exploratório/fisiologia , Masculino , Camundongos , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/deficiência
18.
Neurochem Res ; 38(6): 1163-73, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23494903

RESUMO

Neural cell adhesion molecules (NCAMs) are complexes of transmembranal proteins critical for cell-cell interactions. Initially recognized as key players in the orchestration of developmental processes involving cell migration, cell survival, axon guidance, and synaptic targeting, they have been shown to retain these functions in the mature adult brain, in relation to plastic processes and cognitive abilities. NCAMs are able to interact among themselves (homophilic binding) as well as with other molecules (heterophilic binding). Furthermore, they are the sole molecule of the central nervous system undergoing polysialylation. Most interestingly polysialylated and non-polysialylated NCAMs display opposite properties. The precise contributions each of these characteristics brings in the regulations of synaptic and cellular plasticity in relation to cognitive processes in the adult brain are not yet fully understood. With the aim of deciphering the specific involvement of each interaction, recent developments led to the generation of NCAM mimetic peptides that recapitulate identified binding properties of NCAM. The present review focuses on the information such advances have provided in the understanding of NCAM contribution to cognitive function.


Assuntos
Materiais Biomiméticos/farmacologia , Encéfalo/metabolismo , Cognição/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Adulto , Animais , Encéfalo/efeitos dos fármacos , Giro Denteado/metabolismo , Humanos , Aprendizagem/fisiologia , Memória/fisiologia , Neuritos/fisiologia , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Ácidos Siálicos/metabolismo , Sinapses/fisiologia
19.
Lab Invest ; 92(9): 1297-309, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22732936

RESUMO

Hepatocellular carcinoma (HCC) is a very angiogenic and malignant cancer. Conventional chemotherapy is poorly effective because of the abnormal structural organization of HCC-infiltrating vessels. In previous work, we demonstrated that HCC angiogenesis is driven by transforming growth factor beta-1(TGF-ß1)/CD105 axis, stimulating liver-derived microvascular endothelial cells (Ld-MECs) migration. As TGF-ß1 also affects mural cells (MCs) recruitment and maturation, we asked whether it may contribute to HCC-induced vascular abnormalities. HCC and adjacent non-neoplastic liver (nNL) biopsies obtained from 12 patients were analyzed by immunohistochemistry for angiogenic markers CD105, TGF-ß1, CD44 and vascular endothelial growth factor-a (VEGFa) and for MC markers NG2, α-smooth muscle actin (αSMA) and neural cell adhesion molecule (NCAM). The same markers were also investigated by immunocytochemistry on cultured HCC-derived stromal cells (HCC-StCs) and nNL-derived StCs (nNL-StCs) isolated from the same liver biopsies. Angiogenic factors released by StCs were analyzed by ELISA and the interaction between StCs and Ld-MECs by adhesion assay. Compared with nNL, HCC biopsies showed increased angiogenic markers and αSMA that was localized in vessels. By contrast, NG2 and NCAM were substantially localized in tumor cells but absent in vessels and stroma. Cultured HCC-StCs showed less expression of NG2, αSMA and NCAM. They also demonstrated a lower capacity to release angiogenic factors and adhered on Ld-MECs. HCC-StCs and nNL-StCs treated with TGF-ß1 or with of HepG2 (a human hepatoma cell line) derived conditioned medium (CM), down-modulated NCAM expression, whereas anti-NCAM antibodies significantly reduced the adhesion of StCs to Ld-MECs. By further blocking TGF-ß1 with anti-TGF-ß1 antibodies or with Ly-364947 (a specific inhibitor TGF-ß1-receptor) adhesion to Ld-MECs and NCAM expression respectively was partially restored. TGF-ß1 contributes to HCC-induced vascular alterations by affecting the interaction between HCC-StCs and Ld-MECs through a down-modulation of NCAM expression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Regulação para Baixo , Neoplasias Hepáticas/metabolismo , Microvasos/anormalidades , Moléculas de Adesão de Célula Nervosa/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Biomarcadores/metabolismo , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/patologia , Neovascularização Patológica
20.
Nat Rev Neurosci ; 8(12): 915-20, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18026165

RESUMO

Our understanding of how the enormously complex task of interconnecting millions of nerve cells is accomplished remains rudimentary. What molecular mechanisms control its exquisite specificity? Can we pinpoint single molecular interactions that might help to explain some of the specificity requirements that underlie neuronal wiring? A series of recent studies on the molecular diversity of the Drosophila melanogaster cell-surface receptor Down syndrome cell-adhesion molecule (Dscam) provide one exceptional example of a novel mechanistic model of neuronal-wiring specificity, progressing from structural studies of single protein-protein interactions to biochemical analysis in vitro and to an understanding of complex neuronal differentiation at the single-cell or tissue levels.


Assuntos
Proteínas de Drosophila/fisiologia , Rede Nervosa/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurônios/fisiologia , Animais , Moléculas de Adesão Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Rede Nervosa/química , Rede Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/química , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa