Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 20(1): 1213, 2020 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-33302912

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults, with a median survival of approximately 15 months. Semaphorin 3A (Sema3A), known for its axon guidance and antiangiogenic properties, has been implicated in GBM growth. We hypothesized that Sema3A directly inhibits brain tumor stem cell (BTSC) proliferation and drives invasion via Neuropilin 1 (Nrp1) and Plexin A1 (PlxnA1) receptors. METHODS: GBM BTSC cell lines were assayed by immunostaining and PCR for levels of Semaphorin 3A (Sema3A) and its receptors Nrp1 and PlxnA1. Quantitative BrdU, cell cycle and propidium iodide labeling assays were performed following exogenous Sema3A treatment. Quantitative functional 2-D and 3-D invasion assays along with shRNA lentiviral knockdown of Nrp1 and PlxnA1 are also shown. In vivo flank studies comparing tumor growth of knockdown versus control BTSCs were performed. Statistics were performed using GraphPad Prism v7. RESULTS: Immunostaining and PCR analysis revealed that BTSCs highly express Sema3A and its receptors Nrp1 and PlxnA1, with expression of Nrp1 in the CD133 positive BTSCs, and absence in differentiated tumor cells. Treatment with exogenous Sema3A in quantitative BrdU, cell cycle, and propidium iodide labeling assays demonstrated that Sema3A significantly inhibited BTSC proliferation without inducing cell death. Quantitative functional 2-D and 3-D invasion assays showed that treatment with Sema3A resulted in increased invasion. Using shRNA lentiviruses, knockdown of either NRP1 or PlxnA1 receptors abrogated Sema3A antiproliferative and pro-invasive effects. Interestingly, loss of the receptors mimicked Sema3A effects, inhibiting BTSC proliferation and driving invasion. Furthermore, in vivo studies comparing tumor growth of knockdown and control infected BTSCs implanted into the flanks of nude mice confirmed the decrease in proliferation with receptor KD. CONCLUSIONS: These findings demonstrate the importance of Sema3A signaling in GBM BTSC proliferation and invasion, and its potential as a therapeutic target.


Assuntos
Neoplasias Encefálicas/patologia , Receptores ErbB/genética , Genes erbB-1 , Glioblastoma/patologia , Glioma/patologia , Proteínas de Neoplasias/fisiologia , Semaforina-3A/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/genética , Glioma/metabolismo , Xenoenxertos , Humanos , Lentivirus/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neuropilina-1/biossíntese , Neuropilina-1/genética , Neuropilina-1/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Organismos Livres de Patógenos Específicos
2.
Cell Tissue Res ; 381(2): 299-308, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32242249

RESUMO

Sympathetic nerves innervate most organs and regulate organ blood flow. Specifically, in the uterus, estradiol (E2) elicits rapid degeneration of sympathetic axons and stimulates the growth of blood vessels. Both physiological remodeling processes, critical for reproduction, have been extensively studied but as independent events and are still not fully understood. Here, we examine the neuropilin-1 (NRP1), a shared receptor for axon guidance and angiogenic factors. Systemic estradiol or vehicle were chronically injected to prepubertal rats and uterine and sympathetic chain sections immunostained for NRP1. Uterine semaphorin-3A mRNA was evaluated by in situ hybridization. Control sympathetic uterine-projecting neurons (1-month-old) expressed NRP1 in their somas but not in their intrauterine terminal axons. Estradiol did not affect NRP1 in the distal ganglia. However, at the entrance of the organ, some sympathetic NRP1-positive nerves were recognized. Vascular NRP1 was confined to intrauterine small-diameter vessels in both hormonal conditions. Although the overall pattern of NRP1-IR was not affected by E2 treatment, a subpopulation of infiltrated eosinophil leukocytes showed immunoreactivity for NRP1. Sema3A transcripts were detected in this cellular type as well. No NRP1-immunoreactive axons nor infiltrated eosinophils were visualized in other estrogenized pelvic organs. Together, these data suggest the involvement of NRP1/Sema3A signaling in the selective E2-induced uterine neurovascular remodeling. Our data support a model whereby NRP1 could coordinate E2-induced uterine neurovascular remodeling, acting as a positive regulator of growth when expressed in vessels and as a negative regulator of growth when expressed on axons.


Assuntos
Plasticidade Neuronal , Neuropilina-1/fisiologia , Semaforina-3A/fisiologia , Sistema Nervoso Simpático , Útero , Remodelação Vascular , Animais , Estradiol/farmacologia , Feminino , Ratos , Ratos Wistar , Útero/irrigação sanguínea , Útero/inervação
3.
Chin Med J (Engl) ; 133(3): 301-309, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31929360

RESUMO

BACKGROUND: Mesenchymal stem or stromal cells (MSCs) derived from the induced pluripotent stem cells (iPSCs) have uniform biological activity, which makes the clinical application of MSCs in bone repair possible. Culturing the iPSC-MSCs onto osteoconductive materials is a promising tissue engineering-based strategy in bone regeneration. The aim of this work was to evaluate the effects of semaphorin 3A (Sema3A) and hypoxia inducible factor 1 subunit alpha (HIF1α) co-overexpression on the survival and osteogenic differentiation of iPSC-MSCs. METHODS: Sema3A and HIF1α were linked together with the three (GGGGS; G, glycine; S, serine) peptide fragment, and their co-expression in iPSC-MSCs was mediated by a lentiviral vector. The fusion protein retained the immune reactivity for both Sema3A and HIF1α as determined with Western blotting. iPSC-MSCs were infected with overexpression lentivirus (oeLenti) as negative control, oeLenti-Sema3A, oeLenti-HIF1α or oeLenti-Sema3A-HIF1α lentiviruses. RESULTS: Sema3A overexpression alone promoted the osteogenic differentiation of iPSC-MSCs (the activity and/or expression of osteoblast markers, such as alkaline phosphatase, osteopontin, and osteocalcin, were upregulated), and suppressed cell survival. The Sema3A-HIF1α fusion protein showed a comparable osteoconductive effect to that of Sema3A without reducing cell survival. We further seeded iPSC-MSCs modified by SemaA-HIF1α overexpression onto hydroxyapatite (HA) scaffolds, and evaluated their growth and differentiation on this three-dimensional material. Additional data indicated that, as compared to iPSC-MSCs cultured in ordinary two-dimensional dishes, cells cultured in HA scaffolds grew (blank vs. HA scaffolds: 0.83 vs. 1.39 for survival) and differentiated better (blank vs. HA scaffolds: 11.29 vs. 16.62 for alkaline phosphatase activity). CONCLUSION: Modifying iPSC-MSCs with pro-osteogenic (Sema3A) and pro-survival (HIF1α) factors may represent a promising strategy to optimize tissue engineering-based strategy in bone repair.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Semaforina-3A/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/biossíntese , Engenharia Tecidual
4.
Development ; 146(21)2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690636

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons regulate puberty onset and sexual reproduction by secreting GnRH to activate and maintain the hypothalamic-pituitary-gonadal axis. During embryonic development, GnRH neurons migrate along olfactory and vomeronasal axons through the nose into the brain, where they project to the median eminence to release GnRH. The secreted glycoprotein SEMA3A binds its receptors neuropilin (NRP) 1 or NRP2 to position these axons for correct GnRH neuron migration, with an additional role for the NRP co-receptor PLXNA1. Accordingly, mutations in SEMA3A, NRP1, NRP2 and PLXNA1 have been linked to defective GnRH neuron development in mice and inherited GnRH deficiency in humans. Here, we show that only the combined loss of PLXNA1 and PLXNA3 phenocopied the full spectrum of nasal axon and GnRH neuron defects of SEMA3A knockout mice. Together with Plxna1, the human orthologue of Plxna3 should therefore be investigated as a candidate gene for inherited GnRH deficiency.


Assuntos
Axônios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Padronização Corporal , Encéfalo/fisiologia , Movimento Celular , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/genética , Neuropilina-1/fisiologia , Neuropilina-2/fisiologia , Nariz , Fenótipo , Receptores de Superfície Celular/genética , Semaforina-3A/fisiologia , Maturidade Sexual/genética , Transdução de Sinais
5.
Mol Nutr Food Res ; 63(19): e1801386, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31327173

RESUMO

SCOPE: Loci-specific increase in DNA methylation occurs in cancer and may underlie gene silencing. It is investigated whether dietary stilbenoids, resveratrol, and pterostilbene exert time-dependent effects on DNA methylation patterns and specifically methylation-silenced tumor suppressor genes in breast cancer cells. METHODS AND RESULTS: Following genome-wide DNA methylation analysis with Illumina-450K, changes characteristic of early and late response to stilbenoids are identified. Interestingly, often the same genes but at different CpG loci, the same gene families, or the same functional gene categories are affected. CpG loci that lose methylation in exposed cells correspond to genes functionally associated with cancer suppression. There is a group of genes, including SEMA3A, at which the magnitude of hypomethylation in response to stilbenoids rises with increasing invasive potential of cancer cells. Decreased DNA methylation at SEMA3A promoter and concomitant gene upregulation coincide with increased occupancy of active histone marks. Open chromatin upon exposure to stilbenoids may be linked to decreased DNMT3A binding followed by increased NF1C transcription factor occupancy. Sequestration of DNMT3A is possibly a result of stilbenoid-mediated increase in SALL3 expression, which was previously shown to bind and inhibit DNMT3A activity. CONCLUSIONS: The findings define mechanistic players in stilbenoid-mediated epigenetic reactivation of genes suppressing cancer.


Assuntos
Neoplasias da Mama/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética/efeitos dos fármacos , Fatores de Transcrição NFI/metabolismo , Semaforina-3A/genética , Estilbenos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos dos fármacos , DNA Metiltransferase 3A , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Invasividade Neoplásica/prevenção & controle , Regiões Promotoras Genéticas/genética , Resveratrol/farmacologia , Semaforina-3A/fisiologia , Fatores de Transcrição/genética
6.
J Mol Histol ; 50(5): 427-434, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31270650

RESUMO

The accurately establishment of the eruptive pathway is of vital importance. The mechanisms governing tooth eruption pathway remain little known. This study is to elucidate the roles of Semaphorin 3A (Sema 3A) in mouse tooth eruptive pathway. C57BL/6 mice (11-13 and 15-17 days after birth) were chosen to observe eruptive pathway of mouse lower first molar. Expressions of Sema 3A and its receptor neuropilin 1 and plexin A1 were detected. Osteoclasts were identified by TRAP staining. Co-localization of Sema 3A and osteoclast maker CD68 was detected by double immunofluorescence staining. Picrosirius red staining was applied to observe collagen fibers during mucosal penetration phase. In vitro, Bone marrow-derived macrophages (BMMs) were prepared from 4 week C57BL/6 mice to observe the effect of Sema 3A on the differentiation of BMMs into osteoclasts by TRAP staining. Expressions of Sema 3A was observed by immunofluorescence and western blotting. At osseous eruption phase, many TRAP-positive multi-nucleated cells were distributed around occlusal alveolar bone. The positive expressions of Sema 3A were observed in the multi-nucleated cells. Fluorescence double staining showed that Sema 3A and CD68 were co-expressed in osteoclasts. Its receptor neuropilin 1 and plexin A1 were also found in osteoclasts. In vitro, Sema3A negatively regulated osteoclast differentiation. At mucosal penetration, occlusal alveolar bone had been completely resorbed and collagen fires were gradually degraded for eruptive pathway. Similar positive expressions of Sema 3A and its receptor neuropilin 1 and plexin A1 were also found in the mucosal penetration pathway. Sema 3A gets involved in the establishment of mouse tooth eruptive pathway by modulating osteoclast activity. Sema3A should be considered as a novel nervous agent or a potential biomarker for mouse tooth eruptive pathway.


Assuntos
Osteoclastos/metabolismo , Semaforina-3A/fisiologia , Erupção Dentária/fisiologia , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Colágeno/metabolismo , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Dente Molar/fisiologia , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo
7.
Eur J Neurosci ; 50(1): 1741-1758, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30706560

RESUMO

The development and survival of spiral ganglion neurons (SGNs) are dependent on multiple trophic factors as well as membrane electrical activity. Semaphorins (Sema) constitute a family of membrane-associated and secreted proteins that have garnered significant attention as a potential SGN "navigator" during cochlea development. Previous studies using mutant mice demonstrated that Sema3A plays a role in the SGN pathfinding. The mechanisms, however, by which Sema3A shapes SGNs firing behavior are not known. In these studies, we found that Sema3A plays a novel role in regulating SGN resting membrane potential and excitability. Using dissociated SGN from pre-hearing (P3-P5) and post-hearing mice (P12-P15), we recorded membrane potentials using whole-cell patch clamp recording techniques in apical and basal SGN populations. Recombinant Sema3A was applied to examine the effects on intrinsic membrane properties and action potentials evoked by current injections. Apical and basal SGNs from newborn mice treated with recombinant Sema3A (100 ng/ml) displayed a higher resting membrane potential, higher threshold, decreased amplitude, and prolonged latency and duration of spikes. Although a similar phenomenon was observed in SGNs from post-hearing mice, the resting membrane potential was essentially indistinguishable before and after Sema3A exposure. Sema3A-mediated changes in membrane excitability were associated with a significant decrease in K+ and Ca2+ currents. Sema3A acts through linopirdine-sensitive K+ channels in apical, but not in the basal SGNs. Therefore, Sema3A induces differential effects in SGN membrane excitability that are dependent on age and location, and constitutes an additional early and novel effect of Sema3A SGNs in vitro.


Assuntos
Cóclea/fisiologia , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Semaforina-3A/fisiologia , Gânglio Espiral da Cóclea/fisiologia , Animais , Animais Recém-Nascidos , Cóclea/crescimento & desenvolvimento , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Proteínas Recombinantes
8.
Cell Metab ; 29(3): 627-637.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30661929

RESUMO

Osteocyte survival is key to bone homeostasis and is perturbed in menopause and aging. However, it remains unknown how osteocyte-mediated maintenance of the skeleton is regulated by the osteoprotective factor semaphorin 3A (Sema3A), a secreted protein that is known to reduce bone resorption and enhance bone formation. Here, we show that estrogen induces osteocyte expression of Sema3A, which acts on its receptor on osteocytes to promote their survival and maintain bone homeostasis. Postnatal global and conditional deletion of Sema3a in osteoblastic cells resulted in a severe osteoporotic phenotype marked by fewer osteocytes. This phenotype was recapitulated by osteocyte-specific deficiency of either Sema3A or its receptor component neuropilin-1 (Nrp1). A stimulator of soluble guanylate cyclase-cGMP signaling mimicked Sema3A action and ameliorated bone loss after ovariectomy. We further show that serum levels of SEMA3A decreased with age or after menopause in humans. Thus, we provide a mechanistic insight into the estrogen action and a promising therapeutic approach to protect against bone-related aging.


Assuntos
Envelhecimento/metabolismo , Estrogênios/metabolismo , Menopausa/metabolismo , Osteócitos/metabolismo , Semaforina-3A/fisiologia , Animais , Reabsorção Óssea/metabolismo , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neuropilina-1/metabolismo , Osteócitos/citologia , Osteogênese/fisiologia
9.
Brain Res ; 1701: 28-35, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30025975

RESUMO

BACKGROUND: Deep brain stimulation (DBS) of the anterior nucleus of the thalamus (ANT) has demonstrated antiepileptic efficacy, especially for mesial temporal lobe epilepsy (MTLE). Mossy fiber sprouting (MFS) is involved in the pathogenesis of MTLE, and Sema-3A and GAP-43 are pivotal regulators of MFS. This study investigated the effects of ANT-DBS on MFS and expression levels of Sema-3A and GAP-43 as a possible mechanism for seizure suppression. METHODS: Adult male Sprague-Dawley rats were randomly divided into four groups: (1) control (saline injection), (2) KA (kainic acid injection), (3) KA + Sham-DBS (electrode implantation without stimulation), and (4) KA + DBS (electrode implantation with stimulation). Video electroencephalography (EEG) was used to ensure model establishment and monitor seizure frequency, latency, and severity (Racine stage). Chronic ANT stimulation was conducted for 35 days in the KA + DBS group, and MFS compared to the other groups by quantitative Timm staining. Sema-3A and GAP-43 expression levels in the hippocampal formation were evaluated in all groups with western blot. RESULTS: The latency period was significantly prolonged and spontaneous seizure frequency reduced in the KA + DBS group compared to KA and KA + Sham-DBS groups. Staining scores for MFS in CA3 and dentate gyrus (DG) were significantly lower in the KA + DBS group. The KA + DBS group also exhibited decreased GAP-43 expression and increased Sema-3A expression compared to KA and KA + Sham-DBS groups. CONCLUSION: These results suggest that ANT-DBS extends the latent period following epileptogenic stimulation by impeding MFS through modulation of GAP-43 and Sema-3A expression.


Assuntos
Núcleos Anteriores do Tálamo/metabolismo , Epilepsia/patologia , Fibras Musgosas Hipocampais/efeitos dos fármacos , Animais , Núcleo Celular/patologia , Estimulação Encefálica Profunda/métodos , Giro Denteado/efeitos dos fármacos , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/metabolismo , Epilepsia do Lobo Temporal/patologia , Proteína GAP-43/metabolismo , Proteína GAP-43/fisiologia , Hipocampo/efeitos dos fármacos , Ácido Caínico/farmacologia , Masculino , Fibras Musgosas Hipocampais/patologia , Ratos , Ratos Sprague-Dawley , Convulsões/patologia , Semaforina-3A/metabolismo , Semaforina-3A/fisiologia
10.
Inflammopharmacology ; 26(3): 655-665, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29696565

RESUMO

Autoimmune diseases (ADs) are featured by the body's immune responses being directed against its own tissues, resulting in prolonged inflammation and subsequent tissue damage. Currently, the exact pathogenesis of ADs remains not fully elucidated. Semaphorin-3A (Sema3A), a secreted member of semaphorin family, is a potent immunoregulator during all immune response stages. Sema3A has wide expression, such as in bone, connective tissue, kidney, neurons, and cartilage. Sema3A can downregulate ADs by suppressing the over-activity of both T-cell and B-cell autoimmunity. Moreover, Sema3A shows the ability to enhance T-cell and B-cell regulatory properties that control ADs, including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and systemic sclerosis. However, it can also induce ADs when overexpressed. Together, these data strongly suggest that Sema3A plays a pivotal role in ADs, and it may be a promising treatment target for these diseases. In the present review, we focus on the immunological functions of Sema3A and summarize recent studies on the involvement of Sema3A in the pathogenesis of ADs; the discoveries obtained from recent findings may translate into novel therapeutic agent for ADs.


Assuntos
Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Semaforina-3A/fisiologia , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Neuropilina-1/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Neuron ; 97(4): 796-805.e5, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29398357

RESUMO

Elimination of redundant synapses formed early in development and strengthening of necessary connections are crucial for shaping functional neural circuits. Purkinje cells (PCs) in the neonatal cerebellum are innervated by multiple climbing fibers (CFs) with similar strengths. A single CF is strengthened whereas the other CFs are eliminated in each PC during postnatal development. The underlying mechanisms, particularly for the strengthening of single CFs, are poorly understood. Here we report that progranulin, a multi-functional growth factor implicated in the pathogenesis of frontotemporal dementia, strengthens developing CF synaptic inputs and counteracts their elimination from postnatal day 11 to 16. Progranulin derived from PCs acts retrogradely onto its putative receptor Sort1 on CFs. This effect is independent of semaphorin 3A, another retrograde signaling molecule that counteracts CF synapse elimination. We propose that progranulin-Sort1 signaling strengthens and maintains developing CF inputs, and may contribute to selection of single "winner" CFs that survive synapse elimination.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Cerebelo/crescimento & desenvolvimento , Dendritos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Plasticidade Neuronal , Células de Purkinje/fisiologia , Sinapses/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores , Feminino , Granulinas , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Progranulinas/fisiologia , Ratos Sprague-Dawley , Semaforina-3A/fisiologia , Transdução de Sinais
12.
J Exp Med ; 214(4): 1049-1064, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28289053

RESUMO

The vascular endothelial growth factor (VEGF) isoform VEGF165 stimulates vascular growth and hyperpermeability. Whereas blood vessel growth is essential to sustain organ health, chronic hyperpermeability causes damaging tissue edema. By combining in vivo and tissue culture models, we show here that VEGF165-induced vascular leakage requires both VEGFR2 and NRP1, including the VEGF164-binding site of NRP1 and the NRP1 cytoplasmic domain (NCD), but not the known NCD interactor GIPC1. In the VEGF165-bound receptor complex, the NCD promotes ABL kinase activation, which in turn is required to activate VEGFR2-recruited SRC family kinases (SFKs). These results elucidate the receptor complex and signaling hierarchy of downstream kinases that transduce the permeability response to VEGF165. In a mouse model with choroidal neovascularisation akin to age-related macular degeneration, NCD loss attenuated vessel leakage without affecting neovascularisation. These findings raise the possibility that targeting NRP1 or its NCD interactors may be a useful therapeutic strategy in neovascular disease to reduce VEGF165-induced edema without compromising vessel growth.


Assuntos
Permeabilidade Capilar , Neuropilina-1/fisiologia , Proteínas Proto-Oncogênicas c-abl/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Ativação Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Semaforina-3A/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
13.
Methods Mol Biol ; 1493: 171-183, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787850

RESUMO

The axonal growth cone is a specialized structure enabling axon extension and proper guidance to its target by sensing the extracellular environment. A growth cone collapse assay is a popular approach designed to characterize the inhibitory effect of secreted guidance cues in vitro. However, the actin cytoskeleton of the growth cone is very sensitive to various factors like physical impact, temperature, and acidity of environment that may also induce responses resembling those of guidance signals. Herein, we provide an easy and reproducible method to analyze growth cone sensitivity to the prototypic guidance molecule family class 3 semaphorin. This protocol is intended to present a systematic approach that is easy to apply to any soluble factors with a potential to impact axon elongation.


Assuntos
Axônios/fisiologia , Cones de Crescimento/fisiologia , Semaforina-3A/fisiologia , Citoesqueleto/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Neurônios/fisiologia , Reprodutibilidade dos Testes
14.
Methods Mol Biol ; 1493: 277-286, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787858

RESUMO

Neurons can endocytose soluble semaphorins to either initiate or interrupt signaling at the cell membrane. Depending on the cell type and even on the specific subcellular domain, the endocytic process will differ in intensity, speed, and modality, and will subsequently facilitate diverse actions of semaphorin molecules. Therefore, in order to understand the physiology of guidance cues like semaphorins it is important to visualize endocytic events with good spatial and temporal resolution. Here, we describe methods to visualize endocytosed Semaphorin3A (Sema3A) molecules and to characterize the rate and pathway of internalization in primary rat neuronal cultures using semiconductor quantum dot nanoparticles (Q-dots).


Assuntos
Endocitose/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Pontos Quânticos , Semaforina-3A/fisiologia , Animais , Células Cultivadas , Humanos , Microscopia Confocal , Ratos
15.
Methods Mol Biol ; 1493: 287-298, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787859

RESUMO

Semaphorin3A (Sema3A) guides axonal growth during neuronal network development. Accumulating evidence indicates that Sema3A-induced growth cone collapse and repulsion involve endocytic membrane trafficking in the growth cone. It is now possible to visualize endocytic processes in living cells using total internal reflection fluorescence microscopy (TIRFM), a powerful tool for imaging dynamic subcellular events at the plasma membrane. In this chapter, we describe a method for TIRFM observation and analysis of clathrin-mediated endocytosis in growth cones of chicken dorsal root ganglion neurons that receive an extracellular concentration gradient of Sema3A in a culture medium.


Assuntos
Axônios/fisiologia , Clatrina/fisiologia , Endocitose/fisiologia , Semaforina-3A/fisiologia , Animais , Embrião de Galinha , Gânglios Espinais/citologia , Cones de Crescimento/fisiologia , Microscopia de Fluorescência/métodos , Frações Subcelulares/metabolismo
16.
Methods Mol Biol ; 1493: 393-399, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787866

RESUMO

Podocytes are complex epithelial cells with foot processes that are essential for the integrity and function of the kidney glomerular filters. Podocyte foot processes linked by slit diaphragms constitute signaling platforms that tightly regulate the cell shape and the function of the filtration barrier. Semaphorin (Sema) 3A is a class 3 semaphorin secreted by podocytes that has autocrine and paracrine functions in the kidney. We have shown that Sema3A regulates podocyte shape and that excess Sema3A signaling induces glomerular disease and aggravates diabetic nephropathy. MICAL-1 is an actin-binding protein that mediates Sema3A signals in podocytes. This chapter describes the methods used to examine how Sema3A signaling regulates podocyte shape.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Forma Celular , Proteínas do Citoesqueleto/fisiologia , Proteínas com Domínio LIM/fisiologia , Podócitos/citologia , Semaforina-3A/fisiologia , Animais , Linhagem Celular , Linhagem Celular Transformada , Humanos , Camundongos , Proteínas dos Microfilamentos , Oxigenases de Função Mista
17.
J Neurosci ; 36(50): 12697-12706, 2016 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-27974617

RESUMO

The establishment of precise topographic maps during neural development is facilitated by the presorting of axons in the pathway before they reach their targets. In the vertebrate visual system, such topography is seen clearly in the optic tract (OT) and in the optic radiations. However, the molecular mechanisms involved in pretarget axon sorting are poorly understood. Here, we show in zebrafish that the RNA-binding protein Hermes, which is expressed exclusively in retinal ganglion cells (RGCs), is involved in this process. Using a RiboTag approach, we show that Hermes acts as a negative translational regulator of specific mRNAs in RGCs. One of these targets is the guidance cue receptor Neuropilin 1 (Nrp1), which is sensitive to the repellent cue Semaphorin 3A (Sema3A). Hermes knock-down leads to topographic missorting in the OT through the upregulation of Nrp1. Restoring Nrp1 to appropriate levels in Hermes-depleted embryos rescues this effect and corrects the axon-sorting defect in the OT. Our data indicate that axon sorting relies on Hermes-regulated translation of Nrp1. SIGNIFICANCE STATEMENT: An important mechanism governing the formation of the mature neural map is pretarget axon sorting within the sensory tract; however, the molecular mechanisms involved in this process remain largely unknown. The work presented here reveals a novel function for the RNA-binding protein Hermes in regulating the topographic sorting of retinal ganglion cell (RGC) axons in the optic tract and tectum. We find that Hermes negatively controls the translation of the guidance cue receptor Neuropilin-1 in RGCs, with Hermes knock-down resulting in aberrant growth cone cue sensitivity and axonal topographic misprojections. We characterize a novel RNA-based mechanism by which axons restrict their translatome developmentally to achieve proper targeting.


Assuntos
Axônios/fisiologia , Neuropilina-1/fisiologia , Proteínas de Ligação a RNA/fisiologia , Vias Visuais/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Embrião não Mamífero , Técnicas de Silenciamento de Genes , Cones de Crescimento , Neuropilina-1/genética , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas de Ligação a RNA/genética , Células Ganglionares da Retina/metabolismo , Semaforina-3A/genética , Semaforina-3A/fisiologia , Colículos Superiores/anatomia & histologia , Colículos Superiores/fisiologia , Proteínas de Xenopus/genética , Xenopus laevis , Peixe-Zebra
18.
Neuron ; 91(6): 1276-1291, 2016 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-27618676

RESUMO

Subcellular target recognition in the CNS is the culmination of a multiple-step program including axon guidance, target recognition, and synaptogenesis. In cerebellum, basket cells (BCs) innervate the soma and axon initial segment (AIS) of Purkinje cells (PCs) to form the pinceau synapse, but the underlying mechanisms remain incompletely understood. Here, we demonstrate that neuropilin-1 (NRP1), a Semaphorin receptor expressed in BCs, controls both axonal guidance and subcellular target recognition. We show that loss of Semaphorin 3A function or specific deletion of NRP1 in BCs alters the stereotyped organization of BC axon and impairs pinceau synapse formation. Further, we identified NRP1 as a trans-synaptic binding partner of the cell adhesion molecule neurofascin-186 (NF186) expressed in the PC AIS during pinceau synapse formation. These findings identify a dual function of NRP1 in both axon guidance and subcellular target recognition in the construction of GABAergic circuitry.


Assuntos
Orientação de Axônios/fisiologia , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Neurônios GABAérgicos/fisiologia , Neuropilina-1/fisiologia , Animais , Células CHO , Moléculas de Adesão Celular/metabolismo , Técnicas de Cocultura , Cricetulus , Humanos , Fatores de Crescimento Neural/metabolismo , Neurogênese/fisiologia , Células de Purkinje/fisiologia , Semaforina-3A/fisiologia , Sinapses/fisiologia
19.
J Dent Res ; 95(11): 1282-90, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27302880

RESUMO

In cases of pulp exposure due to deep dental caries or severe traumatic injuries, existing pulp-capping materials have a limited ability to reconstruct dentin-pulp complexes and can result in pulpectomy because of their low potentials to accelerate dental pulp cell activities, such as migration, proliferation, and differentiation. Therefore, the development of more effective therapeutic agents has been anticipated for direct pulp capping. Dental pulp tissues are enriched with dental pulp stem cells (DPSCs). Here, the authors investigated the effects of semaphorin 3A (Sema3A) on various functions of human DPSCs in vitro and reparative dentin formation in vivo in a rat dental pulp exposure model. Immunofluorescence staining revealed expression of Sema3A and its receptor Nrp1 (neuropilin 1) in rat dental pulp tissue and human DPSC clones. Sema3A induced cell migration, chemotaxis, proliferation, and odontoblastic differentiation of DPSC clones. In addition, Sema3A treatment of DPSC clones increased ß-catenin nuclear accumulation, upregulated expression of the FARP2 gene (FERM, RhoGEF, and pleckstrin domain protein 2), and activated Rac1 in DPSC clones. Furthermore, in the rat dental pulp exposure model, Sema3A promoted reparative dentin formation with dentin tubules and a well-aligned odontoblast-like cell layer at the dental pulp exposure site and with novel reparative dentin almost completely covering pulp tissue at 4 wk after direct pulp capping. These findings suggest that Sema3A could play an important role in dentin regeneration via canonical Wnt/ß-catenin signaling. Sema3A might be an alternative agent for direct pulp capping, which requires further study.


Assuntos
Polpa Dentária/citologia , Odontoblastos/citologia , Semaforina-3A/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/fisiologia , Dentina/crescimento & desenvolvimento , Feminino , Imunofluorescência , Humanos , Odontoblastos/efeitos dos fármacos , Odontoblastos/fisiologia , Ratos , Ratos Wistar , Semaforina-3A/fisiologia , Adulto Jovem , beta Catenina/metabolismo
20.
Adv Exp Med Biol ; 900: 1-25, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27003394

RESUMO

Satellite cells are the "currency" for the muscle growth that is critical to meat production in many species, as well as to phenotypic distinctions in development at the level of species or taxa, and for human muscle growth, function and regeneration. Careful research on the activation and behaviour of satellite cells, the stem cells in skeletal muscle, including cross-species comparisons, has potential to reveal the mechanisms underlying pathological conditions in animals and humans, and to anticipate implications of development, evolution and environmental change on muscle function and animal performance.


Assuntos
Fator de Crescimento de Hepatócito/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Fatores Etários , Animais , Movimento Celular , Citoesqueleto/fisiologia , Distroglicanas/fisiologia , Humanos , Desenvolvimento Muscular , Semaforina-3A/fisiologia , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...