Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
2.
EMBO Mol Med ; 15(3): e16629, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36692026

RESUMO

Metastatic melanoma patients carrying a BRAFV600 mutation can be treated with a combination of BRAF and MEK inhibitors (BRAFi/MEKi), but innate and acquired resistance invariably occurs. Predicting patient response to targeted therapies is crucial to guide clinical decision. We describe here the development of a highly efficient patient-derived xenograft model adapted to patient melanoma biopsies, using the avian embryo as a host (AVI-PDXTM ). In this in vivo paradigm, we depict a fast and reproducible tumor engraftment of patient samples within the embryonic skin, preserving key molecular and phenotypic features. We show that sensitivity and resistance to BRAFi/MEKi can be reliably modeled in these AVI-PDXTM , as well as synergies with other drugs. We further provide proof-of-concept that the AVI-PDXTM models the diversity of responses of melanoma patients to BRAFi/MEKi, within days, hence positioning it as a valuable tool for the design of personalized medicine assays and for the evaluation of novel combination strategies.


Assuntos
Melanoma , Proteínas Proto-Oncogênicas B-raf , Animais , Humanos , Proteínas Proto-Oncogênicas B-raf/genética , Melanoma/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinases de Proteína Quinase Ativadas por Mitógeno , Mutação , Modelos Animais de Doenças
3.
Cell ; 185(21): 3931-3949.e26, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36240740

RESUMO

Neural migration is a critical step during brain development that requires the interactions of cell-surface guidance receptors. Cancer cells often hijack these mechanisms to disseminate. Here, we reveal crystal structures of Uncoordinated-5 receptor D (Unc5D) in complex with morphogen receptor glypican-3 (GPC3), forming an octameric glycoprotein complex. In the complex, four Unc5D molecules pack into an antiparallel bundle, flanked by four GPC3 molecules. Central glycan-glycan interactions are formed by N-linked glycans emanating from GPC3 (N241 in human) and C-mannosylated tryptophans of the Unc5D thrombospondin-like domains. MD simulations, mass spectrometry and structure-based mutants validate the crystallographic data. Anti-GPC3 nanobodies enhance or weaken Unc5-GPC3 binding and, together with mutant proteins, show that Unc5/GPC3 guide migrating pyramidal neurons in the mouse cortex, and cancer cells in an embryonic xenograft neuroblastoma model. The results demonstrate a conserved structural mechanism of cell guidance, where finely balanced Unc5-GPC3 interactions regulate cell migration.


Assuntos
Movimento Celular , Glipicanas/química , Receptores de Netrina/química , Animais , Glipicanas/metabolismo , Humanos , Camundongos , Proteínas Mutantes , Receptores de Netrina/metabolismo , Receptores de Superfície Celular/metabolismo , Anticorpos de Domínio Único , Trombospondinas
4.
Nat Commun ; 13(1): 2549, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538114

RESUMO

Embryonic malignant transformation is concomitant to organogenesis, often affecting multipotent and migratory progenitors. While lineage relationships between malignant cells and their physiological counterparts are extensively investigated, the contribution of exogenous embryonic signals is not fully known. Neuroblastoma (NB) is a childhood malignancy of the peripheral nervous system arising from the embryonic trunk neural crest (NC) and characterized by heterogeneous and interconvertible tumor cell identities. Here, using experimental models mimicking the embryonic context coupled to proteomic and transcriptomic analyses, we show that signals released by embryonic sympathetic ganglia, including Olfactomedin-1, induce NB cells to shift from a noradrenergic to mesenchymal identity, and to activate a gene program promoting NB metastatic onset and dissemination. From this gene program, we extract a core signature specifically shared by metastatic cancers with NC origin. This reveals non-cell autonomous embryonic contributions regulating the plasticity of NB identities and setting pro-dissemination gene programs common to NC-derived cancers.


Assuntos
Crista Neural , Neuroblastoma , Diferenciação Celular/genética , Criança , Sinais (Psicologia) , Humanos , Neuroblastoma/genética , Neuroblastoma/patologia , Proteômica
5.
iScience ; 24(12): 103423, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34849474

RESUMO

Lack of preclinical patient-derived xenograft cancer models in which to conduct large-scale molecular studies seriously impairs the development of effective personalized therapies. We report here an in vivo concept consisting of implanting human tumor cells in targeted tissues of an avian embryo, delivering therapeutics, evaluating their efficacy by measuring tumors using light sheet confocal microscopy, and conducting large-scale RNA-seq analysis to characterize therapeutic-induced changes in gene expression. The model was established to recapitulate triple-negative breast cancer (TNBC) and validated using TNBC standards of care and an investigational therapeutic agent.

6.
Front Mol Neurosci ; 12: 52, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30881286

RESUMO

In the developing organism, complex molecular programs orchestrate the generation of cells in adequate numbers, drive them to migrate along the correct pathways towards appropriate territories, eliminate superfluous cells, and induce terminal differentiation of survivors into the appropriate cell-types. Despite strict controls constraining developmental processes, malignancies can emerge in still immature organisms. This is the case of neuroblastoma (NB), a highly heterogeneous disease, predominantly affecting children before the age of 5 years. Highly metastatic forms represent half of the cases and are diagnosed when disseminated foci are detectable. NB arise from a transient population of embryonic cells, the neural crest (NC), and especially NC committed to the establishment of the sympatho-adrenal tissues. The NC is generated at the dorsal edge of the neural tube (NT) of the vertebrate embryo, under the action of NC specifier gene programs. NC cells (NCCs) undergo an epithelial to mesenchymal transition, and engage on a remarkable journey in the developing embryo, contributing to a plethora of cell-types and tissues. Various NCC sub-populations and derived lineages adopt specific migratory behaviors, moving individually as well as collectively, exploiting the different embryonic substrates they encounter along their path. Here we discuss how the specific features of NCC in development are re-iterated during NB metastatic behaviors.

7.
Cancer Cell ; 32(4): 427-443.e8, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017055

RESUMO

Neuroblastoma (NB) is a childhood cancer arising from sympatho-adrenal neural crest cells. Disseminated forms have high frequency of multiple tumoral foci whose etiology remains unknown; NB embryonic origin limits investigations in patients and current models. We developed an avian embryonic model driving human NB tumorigenesis in tissues homologous to patients. We found that aggressive NBs display a metastatic mode, secondary dissemination via peripheral nerves and aorta. Through tumor transcriptional profiling, we found that NB dissemination is induced by the shutdown of a pro-cohesion autocrine signal, SEMA3C, which constrains the tumoral mass. Lowering SEMA3C levels shifts the balance toward detachment, triggering NB cells to collectively evade the tumor. Together with patient cohort analysis, this identifies a microenvironment-driven pro-metastatic switch for NB.


Assuntos
Neuroblastoma/secundário , Microambiente Tumoral , Adolescente , Adulto , Animais , Adesão Celular , Embrião de Galinha , Criança , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Estadiamento de Neoplasias , Proteínas do Tecido Nervoso/fisiologia , Neuroblastoma/etiologia , Neuroblastoma/patologia , Receptores de Superfície Celular/fisiologia , Semaforinas/genética , Semaforinas/fisiologia
8.
Acta Neuropathol Commun ; 5(1): 55, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28709447

RESUMO

Neurofilament heavy chain (NEFH) gene was recently identified to cause autosomal dominant axonal Charcot-Marie-Tooth disease (CMT2cc). However, the clinical spectrum of this condition and the physio-pathological pathway remain to be delineated. We report 12 patients from two French families with axonal dominantly inherited form of CMT caused by two new mutations in the NEFH gene. A remarkable feature was the early involvement of proximal muscles of the lower limbs associated with pyramidal signs in some patients. Nerve conduction velocity studies indicated a predominantly motor axonal neuropathy. Unique deletions of two nucleotides causing frameshifts near the end of the NEFH coding sequence were identified: in family 1, c.3008_3009del (p.Lys1003Argfs*59), and in family 2 c.3043_3044del (p.Lys1015Glyfs*47). Both frameshifts lead to 40 additional amino acids translation encoding a cryptic amyloidogenic element. Consistently, we show that these mutations cause protein aggregation which are recognised by the autophagic pathway in motoneurons and triggered caspase 3 activation leading to apoptosis in neuroblastoma cells. Using electroporation of chick embryo spinal cord, we confirm that NEFH mutants form aggregates in vivo and trigger apoptosis of spinal cord neurons. Thus, our results provide a physiological explanation for the overlap between CMT and amyotrophic lateral sclerosis (ALS) clinical features in affected patients.


Assuntos
Apoptose/fisiologia , Doença de Charcot-Marie-Tooth/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Agregação Patológica de Proteínas/metabolismo , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Doença de Charcot-Marie-Tooth/genética , Embrião de Galinha , Família , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Proteínas de Neurofilamentos/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Adulto Jovem
9.
Int J Mol Sci ; 18(4)2017 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-28338624

RESUMO

Membrane microdomains or "lipid rafts" have emerged as essential functional modules of the cell, critical for the regulation of growth factor receptor-mediated responses. Herein we describe the dichotomy between caveolin-1 and caveolin-2, structural and regulatory components of microdomains, in modulating proliferation and differentiation. Caveolin-2 potentiates while caveolin-1 inhibits nerve growth factor (NGF) signaling and subsequent cell differentiation. Caveolin-2 does not appear to impair NGF receptor trafficking but elicits prolonged and stronger activation of MAPK (mitogen-activated protein kinase), Rsk2 (ribosomal protein S6 kinase 2), and CREB (cAMP response element binding protein). In contrast, caveolin-1 does not alter initiation of the NGF signaling pathway activation; rather, it acts, at least in part, by sequestering the cognate receptors, TrkA and p75NTR, at the plasma membrane, together with the phosphorylated form of the downstream effector Rsk2, which ultimately prevents CREB phosphorylation. The non-phosphorylatable caveolin-1 serine 80 mutant (S80V), no longer inhibits TrkA trafficking or subsequent CREB phosphorylation. MC192, a monoclonal antibody towards p75NTR that does not block NGF binding, prevents exit of both NGF receptors (TrkA and p75NTR) from lipid rafts. The results presented herein underline the role of caveolin and receptor signaling complex interplay in the context of neuronal development and tumorigenesis.


Assuntos
Caveolina 1/metabolismo , Núcleo Celular/metabolismo , Microdomínios da Membrana/metabolismo , Fator de Crescimento Neural/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos Monoclonais/imunologia , Proteína de Ligação a CREB/metabolismo , Caveolina 1/antagonistas & inibidores , Caveolina 1/genética , Caveolina 2/antagonistas & inibidores , Caveolina 2/genética , Caveolina 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Camundongos , Proteínas do Tecido Nervoso , Células PC12 , Fosforilação/efeitos dos fármacos , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/química , Receptor trkA/imunologia , Receptor trkA/metabolismo , Receptores de Fatores de Crescimento , Receptores de Fator de Crescimento Neural/química , Receptores de Fator de Crescimento Neural/imunologia , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo
10.
Development ; 143(12): 2183-93, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27122172

RESUMO

In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.


Assuntos
Axônios/metabolismo , Efrina-B3/metabolismo , Receptor EphB2/metabolismo , Transdução de Sinais , Medula Espinal/metabolismo , Quinase Syk/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Endocitose , Cones de Crescimento/metabolismo , Camundongos Knockout , Fosforilação
11.
Neuron ; 70(5): 966-78, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21658588

RESUMO

Growing axons are guided to their targets by attractive and repulsive cues. In the developing spinal cord, Netrin-1 and Shh guide commissural axons toward the midline. However, the combined inhibition of their activity in commissural axon turning assays does not completely abrogate turning toward floor plate tissue, suggesting that additional guidance cues are present. Here we show that the prototypic angiogenic factor VEGF is secreted by the floor plate and is a chemoattractant for commissural axons in vitro and in vivo. Inactivation of Vegf in the floor plate or of its receptor Flk1 in commissural neurons causes axon guidance defects, whereas Flk1 blockade inhibits turning of axons to VEGF in vitro. Similar to Shh and Netrin-1, VEGF-mediated commissural axon guidance requires the activity of Src family kinases. Our results identify VEGF and Flk1 as a novel ligand/receptor pair controlling commissural axon guidance.


Assuntos
Axônios/fisiologia , Quimiotaxia/fisiologia , Quiasma Óptico/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Quimiotaxia/genética , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática/métodos , Glicosídeo Hidrolases/metabolismo , Cones de Crescimento/metabolismo , Proteínas Hedgehog/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteínas Supressoras de Tumor/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Proteína Wnt1/genética
12.
Mol Cell Biol ; 28(7): 2314-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18212043

RESUMO

The chemotropic guidance cue netrin-1 promotes neurite outgrowth through its receptor Deleted in Colorectal Cancer (DCC) via activation of Rac1. The guanine nucleotide exchange factor (GEF) linking netrin-1/DCC to Rac1 activation has not yet been identified. Here, we show that the RhoGEF Trio mediates Rac1 activation in netrin-1 signaling. We found that Trio interacts with the netrin-1 receptor DCC in mouse embryonic brains and that netrin-1-induced Rac1 activation in brain is impaired in the absence of Trio. Trio(-/-) cortical neurons fail to extend neurites in response to netrin-1, while they are able to respond to glutamate. Accordingly, netrin-1-induced commissural axon outgrowth is reduced in Trio(-/-) spinal cord explants, and the guidance of commissural axons toward the floor plate is affected by the absence of Trio. The anterior commissure is absent in Trio-null embryos, and netrin-1/DCC-dependent axonal projections that form the internal capsule and the corpus callosum are defective in the mutants. Taken together, these findings establish Trio as a GEF that mediates netrin-1 signaling in axon outgrowth and guidance through its ability to activate Rac1.


Assuntos
Cones de Crescimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fatores de Crescimento Neural/fisiologia , Neuropeptídeos/fisiologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Encéfalo/anormalidades , Encéfalo/citologia , Encéfalo/embriologia , Células COS , Linhagem Celular , Células Cultivadas/citologia , Chlorocebus aethiops , Receptor DCC , Ativação Enzimática , Feminino , Cones de Crescimento/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Netrina-1 , Proteínas Oncogênicas/fisiologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Medula Espinal/citologia , Medula Espinal/embriologia , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP
13.
J Cell Sci ; 118(Pt 8): 1687-92, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15811950

RESUMO

During development, axons migrate long distances in responses to attractive or repulsive signals that are detected by their growth cones. One of these signals is mediated by netrin-1, a diffusible laminin-related molecule that both attracts and repels growth cones via interaction with its receptor DCC (deleted in colorectal cancer). Here we show that DCC in both commissural neurons and immortalized cells, is partially associated with cholesterol- and sphingolipid-enriched membrane domains named lipid rafts. This localization of DCC in lipid rafts is mediated by the palmitoylation within its transmembrane region. Moreover, this raft localization of DCC is required for netrin-1-induced DCC-dependent ERK activation, and netrin-1-mediated axon outgrowth requires lipid raft integrity. Thus, the presence of axon guidance-related receptors in lipid rafts appears to be a crucial pre-requisite for growth cone response to chemo-attractive or repulsive cues.


Assuntos
Moléculas de Adesão Celular/metabolismo , Quimiotaxia/fisiologia , Cones de Crescimento/metabolismo , Microdomínios da Membrana/metabolismo , Fatores de Crescimento Neural/metabolismo , Medula Espinal/embriologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Comunicação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Sinais (Psicologia) , Receptor DCC , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Lipídeos de Membrana/metabolismo , Netrina-1 , Ácido Palmítico/metabolismo , Ratos , Receptores de Superfície Celular , Transdução de Sinais/fisiologia
14.
Neoplasia ; 7(2): 180-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15802023

RESUMO

Previously, we demonstrated that loss of SEMA3F, a secreted semaphorin encoded in 3p21.3, is associated with higher stages in lung cancer and primary tumor cells studied with anti-vascular endothelial growth factor (VEGF) and SEMA3F antibodies. In vitro, SEMA3F inhibits cell spreading; this activity is opposed by VEGF. These results suggest that VEGF and SEMA3F compete for binding to their common neuropilin receptor. In the present report, we investigated the attractive/repulsive effects of SEMA3F on cell migration when cells were grown in a three-dimensional system and exposed to a SEMA3F gradient. In addition, we adapted the neurobiologic stripe assay to analyze the migration of tumor cells in response to SEMA3F. In the motile breast cancer cell line C100, which expresses both neuropilin-1 (NRP1) and neuropilin-2 (NRP2) receptors, SEMA3F had a repulsive effect, which was blocked by anti-NRP2 antibody. In less motile MCF7 cells, which express only NRP1, SEMA3F inhibited cell contacts with loss of membrane-associated E-cadherin and beta-catenin without motility induction. Cell spreading and proliferation were reduced. These results support the concept that in a first step during tumorigenesis, normal tissues expressing SEMA3F would try to prevent tumor cells from spreading and attaching to the stroma for further implantation.


Assuntos
Neoplasias da Mama/patologia , Caderinas/química , Adesão Celular , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Membrana Celular/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Rim/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Transativadores/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina
15.
Mol Cell Neurosci ; 26(1): 89-100, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15121181

RESUMO

During axon navigation, Semaphorin3A-induced growth cone retraction is correlated with endocytosis. Although its function remains elusive, we showed previously that the cell adhesion molecule of the immunoglobulin super family L1 associates with Neuropilin-1 (NP-1) the Sema3A-binding subunit of the receptor complex and is required for Sema3A to elicit axonal repulsive responses. We report here that upon Sema3A binding to NP-1, L1 and NP-1 are co-internalized through a clathrin-dependent mechanism mediated by L1. We show that in COS7 cells, L1/NP-1 endocytosis is correlated with a cell contraction similar to that observed with the Plexin (Plex)/NP-1 or Plex/NP1/L1 complexes. In neuronal cultures, a L1-mimetic peptide able to switch Sema3A repulsive responses to attraction blocks both endocytosis and growth cone collapse. Similarly, in the COS7 cell model, peptide application prevents both the Sema3-induced L1/NP-1 internalization and cell collapse. These studies demonstrate that the L1/NP-1 complex is able to confer a biological response to Sema3A with L1 mediating receptor internalization following ligand activation. They also reveal that endocytosis controlled by L1/NP-1 cis and trans interactions is pivotal in Sema3A-mediated axon guidance.


Assuntos
Endocitose/fisiologia , Cones de Crescimento/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Animais , Animais Recém-Nascidos , Células COS , Moléculas de Adesão Celular/metabolismo , Comunicação Celular/fisiologia , Células Cultivadas , Clatrina/metabolismo , Sinais (Psicologia) , Cones de Crescimento/ultraestrutura , Ligantes , Substâncias Macromoleculares , Camundongos , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Peptídeos/farmacologia , Ligação Proteica/fisiologia
16.
Mol Cell Neurosci ; 24(2): 409-18, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14572462

RESUMO

SEMA3F is a secreted semaphorin that affects axon and cell guidance in the developing nervous system, and is also thought to have anti-tumor activity. Two spliced forms of SEMA3F have been identified that differ by the insertion of 31 amino acids in the sema domain. Here, we investigated the bioactivity of these isoforms and show, using coculture and binding assays, that they share common axonal chemorepulsive properties and binding to neuropilin receptors. SEMA3F isoforms were also found to regulate endothelial cell morphology by remodeling lamellipodial protrusions. Although Sema3F expression globally decreased during mouse development, we noted an enrichment of the longest isoform at postnatal stages in some territories such as the brainstem and spinal cord. These results indicate that although functionally redundant in cell culture assays, Sema3F spliced forms are characterized in vivo by a temporal and regional specific regulation during maturation of the nervous system.


Assuntos
Processamento Alternativo , Hipocampo/metabolismo , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Animais Recém-Nascidos , Células COS , Chlorocebus aethiops , Regulação da Expressão Gênica/fisiologia , Hipocampo/crescimento & desenvolvimento , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo , Ligação Proteica , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Fatores de Tempo
17.
Rev. bras. biol ; 56(supl.1,pt.1): 21-31, Dec. 1996. ilus
Artigo em Inglês | LILACS | ID: lil-196828

RESUMO

The mammalian cerebral cortex is organized in layers and columns, which are reflected in the local intrinsic connections and in the projections to and from the cortex. It is well established that the development of the columnar architecture is under the influence of neuronal activity, but little is known about the mechanisms that control the laminar specificity of cortical circuits. Here we review some recent studies which show that diffusible and membrane-associated molecules provide sufficient information to reconstruct layer-specific intrinsic and extrinsic cortical circuits under in vitro conditions.


Assuntos
Animais , Transporte Axonal/fisiologia , Mapeamento Encefálico , Córtex Cerebral/fisiologia , Técnicas In Vitro , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA