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










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 39(48): 7114-7126, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33005016

RESUMO

Glioblastoma multiforme (GBM) is a highly proliferative and locally invasive cancer with poor prognosis and a high recurrence rate. Although anti-VEGF (vascular endothelial growth factor) therapy offers short-term benefit to GBM patients, this approach fails as the tumor develops into a more invasive and drug-resistant phenotype and ultimately recurs. Recently, both glioma stemlike cells (GSCs) and brain tumor-initiating cells (BTICs) have been implicated in GBM recurrence and its resistance to therapy. We observed that patient-derived GBM cells expressing shRNAs of VEGF or neuropilin-1 (NRP-1) attenuate cancer stem cell markers, inhibit the tumor-initiating cell's neurosphere-forming capacity, and migration. Furthermore, both VEGF and NRP-1 knockdown inhibit the growth of patient-derived GBM xenografts in both zebrafish and mouse models. Interestingly, NRP-1-depleted patient-derived GBM xenografts substantially prolonged survival in mice compared to that of VEGF depletion. Our results also demonstrate that NRP-1 ablation of patient-derived GBM cells improves the sensitivity of TMZ and enhances the overall survival of the respective tumor-bearing mice. This improved outcome may provide insight into the inhibition of GBM progression and effective treatment strategies by targeting NRP-1 in addition to chemotherapy and radiotherapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Neuropilina-1/deficiência , Neuropilina-1/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica , Glioblastoma/patologia , Humanos , Camundongos , Fenótipo , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
FASEB J ; 34(9): 11925-11943, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32686172

RESUMO

Cell-cycle regulatory proteins (p21Cip1 /p27Kip1 ) inhibit cyclin and cyclin-dependent kinase (CDK) complex that promotes fibrosis and hypertrophy. The present study examined the role of CDK blockers, p21Cip1 /p27Kip1 in the progression of renal fibrosis and dysfunction using Npr1 (encoding guanylyl cyclase/natriuretic peptide receptor-A, GC-A/NPRA) gene-knockout (0-copy; Npr1-/- ), 2-copy (Npr1+/+ ), and 4-copy (Npr1++/++ ) mice treated with GC inhibitor, A71915 and cGMP-dependent protein kinase (cGK) inhibitor, (Rp-8-Br-cGMPS). A significant decrease in renal cGMP levels and cGK activity was observed in 0-copy mice and A71915- and Rp-treated 2-copy and 4-copy mice compared with controls. An increased phosphorylation of Erk1/2, p38, p21Cip1 , and p27Kip1 occurred in 0-copy and A71915-treated 2-copy and 4-copy mice, while Rp treatment caused minimal changes than controls. Pro-inflammatory (TNF-α, IL-6) and pro-fibrotic (TGF-ß1) cytokines were significantly increased in plasma and kidneys of 0-copy and A71915-treated 2-copy mice, but to lesser extent in 4-copy mice. Progressive renal pathologies, including fibrosis, mesangial matrix expansion, and tubular hypertrophy were observed in 0-copy and A71915-treated 2-copy and 4-copy mice, but minimally occurred in Rp-treated mice compared with controls. These results indicate that Npr1 has pivotal roles in inhibiting renal fibrosis and hypertrophy and exerts protective effects involving cGMP/cGK axis by repressing CDK blockers p21Cip1 and p27Kip1 .


Assuntos
GMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Nefropatias/metabolismo , Túbulos Renais/metabolismo , Transdução de Sinais , Animais , GMP Cíclico/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Citocinas/genética , Citocinas/metabolismo , Fibrose , Nefropatias/genética , Nefropatias/patologia , Túbulos Renais/patologia , Camundongos , Camundongos Knockout , Neuropilina-1/deficiência , Neuropilina-1/metabolismo
3.
Am J Physiol Renal Physiol ; 313(6): F1232-F1242, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28835419

RESUMO

Proteinuria has been reported in cancer patients receiving agents that target the transmembrane receptor neuropilin-1 (Nrp1) suggesting potential adverse effects on glomerular function. Here we show that Nrp1 is highly expressed by mesangial cells and that genetic deletion of the Nrp1 gene from PDGF receptor-ß+ mesangial cells results in proteinuric disease and glomerulosclerosis, leading to renal failure and death within 6 wk of age in mice. The major defect is a failure of mesangial cell migration that is required to establish the mature glomerular tuft. In vitro data show that the potent chemotactic effect of PDGFB is lost in Nrp1-deficient mesangial cells. Biochemical analyses reveal that Nrp1 is required for PDGFB-dependent phosphorylation of p130 Crk-associated substrate (p130Cas), a large-scaffold molecule that is involved in motility of other cell types. In stark contrast, matrix adhesion and activation of ERK and Akt, which mediate proliferation of mesangial cells in response to PDGFB, are unaffected by the absence of Nrp1. Taken together, these results identify a critical cell-autonomous role for Nrp1 in the migratory behavior of mesangial cells and may help explain the renal effects that occur in patients receiving Nrp1-inhibitory drugs.


Assuntos
Movimento Celular , Glomerulonefrite/metabolismo , Células Mesangiais/metabolismo , Neuropilina-1/metabolismo , Proteinúria/metabolismo , Insuficiência Renal/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Proteína Substrato Associada a Crk/metabolismo , Progressão da Doença , Predisposição Genética para Doença , Taxa de Filtração Glomerular , Glomerulonefrite/genética , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Humanos , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/ultraestrutura , Camundongos Knockout , Neuropilina-1/deficiência , Neuropilina-1/genética , Fenótipo , Fosforilação , Proteinúria/genética , Proteinúria/patologia , Proteinúria/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Interferência de RNA , Insuficiência Renal/genética , Insuficiência Renal/patologia , Insuficiência Renal/fisiopatologia , Transdução de Sinais , Fatores de Tempo , Transfecção
4.
Kidney Int ; 91(4): 868-879, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27988210

RESUMO

Neuropilin1 (Nrp1) is a co-receptor best known to regulate the development of endothelial cells and is a target of anticancer therapies. However, its role in other vascular cells including pericytes is emergent. The kidney is an organ with high pericyte density and cancer patients develop severe proteinuria following administration of NRP1B-neutralizing antibody combined with bevacizumab. Therefore, we investigated whether Nrp1 regulates glomerular capillary integrity after completion of renal development using two mouse models; tamoxifen-inducible NG2Cre to delete Nrp1 specifically in pericytes and administration of Nrp1-neutralizing antibodies. Specific Nrp1 deletion in pericytes did not affect pericyte number but mutant mice developed hematuria with glomerular basement membrane defects. Despite foot process effacement, albuminuria was absent and expression of podocyte proteins remained unchanged upon Nrp1 deletion. Additionally, these mice displayed dilation of the afferent arteriole and glomerular capillaries leading to glomerular hyperfiltration. Nidogen-1 mRNA was downregulated and collagen4α3 mRNA was upregulated with no significant effect on the expression of other basement membrane genes in the mutant mice. These features were phenocopied by treating wild-type mice with Nrp1-neutralizing antibodies. Thus, our results reveal a postdevelopmental role of Nrp1 in renal pericytes as an important regulator of glomerular basement membrane integrity. Furthermore, our study offers novel mechanistic insights into renal side effects of Nrp1 targeting cancer therapies.


Assuntos
Membrana Basal Glomerular/metabolismo , Taxa de Filtração Glomerular , Glomérulos Renais/metabolismo , Neuropilina-1/metabolismo , Pericitos/metabolismo , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Animais , Anticorpos Neutralizantes/farmacologia , Arteríolas/metabolismo , Arteríolas/fisiopatologia , Autoantígenos/genética , Autoantígenos/metabolismo , Capilares/metabolismo , Capilares/fisiopatologia , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Regulação da Expressão Gênica , Genótipo , Membrana Basal Glomerular/efeitos dos fármacos , Membrana Basal Glomerular/fisiopatologia , Membrana Basal Glomerular/ultraestrutura , Hematúria/genética , Hematúria/metabolismo , Hematúria/fisiopatologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/fisiopatologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/deficiência , Neuropilina-1/genética , Pericitos/efeitos dos fármacos , Pericitos/ultraestrutura , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Vasodilatação
5.
J Immunol ; 197(10): 3927-3935, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27815442

RESUMO

To stimulate a productive T cell response, dendritic cells (DC) must undergo maturation characterized by heightened cell surface expression of MHC and costimulatory molecules as well as cytokine production. Conversely, the inhibition of DC maturation is a central mechanism of immune tolerance. The control of the DC maturation process relies on the integration of several cellular stimulatory or inhibitory signals. The soluble factors and their receptors controlling this central aspect of DC biology are incompletely characterized. We show that murine bone marrow-derived DC (BMDC) maturation induced by LPS, as opposed to polyinosinic:polycytidylic acid or cytosine-phosphate-guanine, is robustly inhibited by vascular endothelial growth factor (VEGF), a previously identified immunosuppressive cytokine. Using BMDC from wild type and conditional knockout mice, we show that neuropilin-1 (NRP-1), a known receptor of VEGF, is necessary to suppress LPS-dependent BMDC maturation. The absence of NRP-1 had no ostensible effects on the biology of BMDC in the absence of VEGF. However, NRP-1-deficient BMDC remained completely insensitive to the VEGF-dependent inhibition of BMDC maturation in culture. In the presence of VEGF, NRP-1 directly interacted with the LPS receptor TLR4 and suppressed downstream signaling through ERK and NF-κß, resulting in a sharp inhibition of MHC class II and costimulatory molecules (CD40, CD86) expression as well as proinflammatory cytokine production. Consequently, we identify NRP-1 as a target to optimize DC maturation within environments that are rich in VEGF, such as tumors.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Neuropilina-1/imunologia , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígeno B7-2/efeitos dos fármacos , Antígeno B7-2/genética , Células da Medula Óssea/imunologia , Células da Medula Óssea/fisiologia , Antígenos CD40/efeitos dos fármacos , Antígenos CD40/genética , Diferenciação Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Citocinas/genética , Células Dendríticas/imunologia , Genes MHC da Classe II/efeitos dos fármacos , Genes MHC da Classe II/genética , Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Subunidade p50 de NF-kappa B/fisiologia , Neuropilina-1/deficiência , Poli I-C/farmacologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Oncotarget ; 7(9): 9801-14, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26755653

RESUMO

Gliomas are the most commonly diagnosed primary tumors of the central nervous system (CNS). Median times of survival are dismal regardless of the treatment approach, underlying the need to develop more effective therapies. Modulation of the immune system is a promising strategy as innate and adaptive immunity play important roles in cancer progression. Glioma associated microglia and macrophages (GAMs) can comprise over 30% of the cells in glioma biopsies. Gliomas secrete cytokines that suppress the anti-tumorigenic properties of GAMs, causing them to secrete factors that support the tumor's spread and growth. Neuropilin 1 (Nrp1) is a transmembrane receptor that in mice both amplifies pro-angiogenic signaling in the tumor microenvironment and affects behavior of innate immune cells. Using a Cre-lox system, we generated mice that lack expression of Nrp1 in GAMs. We demonstrate, using an in vivo orthotopic glioma model, that tumors in mice with Nrp1-deficient GAMs exhibit less vascularity, grow at a slower pace, and are populated by increased numbers of anti-tumorigenic GAMs. Moreover, glioma survival times in mice with Nrp1-deficient GAMs were significantly longer. Treating wild-type mice with a small molecule inhibitor of Nrp1's b1 domain, EG00229, which we show here is selective for Nrp1 over Nrp2, yielded an identical outcome. Nrp1-deficient or EG00229-treated wild-type microglia exhibited a shift towards anti-tumorigenicity as evident by altered inflammatory marker profiles in vivo and decreased SMAD2/3 activation when conditioned in the presence of glioma-derived factors. These results provide support for the proposal that pharmacological inhibition of Nrp1 constitutes a potential strategy for suppressing glioma progression.


Assuntos
Antineoplásicos/uso terapêutico , Glioma/tratamento farmacológico , Macrófagos/imunologia , Microglia/patologia , Neuropilina-1 , Animais , Linhagem Celular Tumoral , Progressão da Doença , Glioma/mortalidade , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/deficiência , Neuropilina-1/genética , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
7.
Ann Rheum Dis ; 75(8): 1541-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26359450

RESUMO

OBJECTIVES: In systemic sclerosis (SSc), vascular involvement is characterised by vascular endothelial growth factor (VEGF)-A/VEGF receptor (VEGFR) system disturbances. Neuropilin-1 (NRP1), a receptor for both class-3 semaphorins (Sema3s) and VEGF-A, is required for optimal VEGF-A/VEGFR-2 signalling. Here, we investigated the possible involvement of Sema3A/NRP1 axis in SSc. METHODS: Circulating Sema3A and soluble NRP1 (sNRP1) were measured in patients with SSc and controls. NRP1 and Sema3A expression in skin biopsies was evaluated by immunofluorescence and western blotting. NRP1 expression was assessed in SSc and healthy dermal microvascular endothelial cells (SSc-MVECs and H-MVECs), and in SSc and control endothelial progenitor cell (EPC)-derived endothelial cells (ECs). The possible impact of transcription factor Friend leukaemia integration 1 (Fli1) deficiency on endothelial NRP1 expression was investigated by gene silencing. The binding of Fli1 to NRP1 gene promoter was evaluated using chromatin immunoprecipitation. Capillary morphogenesis was performed on Matrigel. RESULTS: Decreased sNRP1 levels in SSc were associated with active and late nailfold videocapillaroscopy patterns and digital ulcers. No difference in Sema3A was found between patients and controls. NRP1 was significantly decreased in SSc-MVECs both ex vivo and in vitro. NRP1 and Fli1 significantly decreased in H-MVECs challenged with SSc sera, while they were not different in SSc and control EPC-derived ECs. Fli1 occupied the NRP1 gene promoter and Fli1 gene silencing reduced NRP1 expression in H-MVECs. NRP1 gene silencing in H-MVECs resulted in a significantly impaired angiogenic capacity comparable to that of cells treated with SSc sera. CONCLUSION: In SSc, NRP1 deficiency may be an additional factor in the perturbed VEGF-A/VEGFR-2 system contributing to peripheral microvasculopathy and defective angiogenesis.


Assuntos
Neovascularização Patológica/metabolismo , Neuropilina-1/metabolismo , Doenças Vasculares Periféricas/metabolismo , Escleroderma Sistêmico/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Células Cultivadas , Regulação para Baixo/fisiologia , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Angioscopia Microscópica/métodos , Pessoa de Meia-Idade , Neuropilina-1/deficiência , Neuropilina-1/genética , Doenças Vasculares Periféricas/etiologia , Proteína Proto-Oncogênica c-fli-1/deficiência , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/patologia , Semaforina-3A/sangue , Pele/irrigação sanguínea , Pele/patologia
8.
Nature ; 526(7575): 710-4, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26503042

RESUMO

Selective neuronal loss is a hallmark of neurodegenerative diseases, which, counterintuitively, are often caused by mutations in widely expressed genes. Charcot-Marie-Tooth (CMT) diseases are the most common hereditary peripheral neuropathies, for which there are no effective therapies. A subtype of these diseases--CMT type 2D (CMT2D)--is caused by dominant mutations in GARS, encoding the ubiquitously expressed enzyme glycyl-transfer RNA (tRNA) synthetase (GlyRS). Despite the broad requirement of GlyRS for protein biosynthesis in all cells, mutations in this gene cause a selective degeneration of peripheral axons, leading to deficits in distal motor function. How mutations in GlyRS (GlyRS(CMT2D)) are linked to motor neuron vulnerability has remained elusive. Here we report that GlyRS(CMT2D) acquires a neomorphic binding activity that directly antagonizes an essential signalling pathway for motor neuron survival. We find that CMT2D mutations alter the conformation of GlyRS, enabling GlyRS(CMT2D) to bind the neuropilin 1 (Nrp1) receptor. This aberrant interaction competitively interferes with the binding of the cognate ligand vascular endothelial growth factor (VEGF) to Nrp1. Genetic reduction of Nrp1 in mice worsens CMT2D symptoms, whereas enhanced expression of VEGF improves motor function. These findings link the selective pathology of CMT2D to the neomorphic binding activity of GlyRS(CMT2D) that antagonizes the VEGF-Nrp1 interaction, and indicate that the VEGF-Nrp1 signalling axis is an actionable target for treating CMT2D.


Assuntos
Ligação Competitiva , Doença de Charcot-Marie-Tooth/metabolismo , Glicina-tRNA Ligase/metabolismo , Animais , Axônios/enzimologia , Axônios/metabolismo , Axônios/patologia , Linhagem Celular , Sobrevivência Celular , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Feminino , Glicina-tRNA Ligase/química , Glicina-tRNA Ligase/genética , Ligantes , Masculino , Camundongos , Modelos Moleculares , Neurônios Motores/enzimologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Destreza Motora/efeitos dos fármacos , Mutação/genética , Neuropilina-1/deficiência , Neuropilina-1/genética , Neuropilina-1/metabolismo , Ligação Proteica , Multimerização Proteica , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
9.
J Clin Invest ; 125(7): 2661-76, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26053665

RESUMO

In mammals, the outflow tract (OFT) of the developing heart septates into the base of the pulmonary artery and aorta to guide deoxygenated right ventricular blood into the lungs and oxygenated left ventricular blood into the systemic circulation. Accordingly, defective OFT septation is a life-threatening condition that can occur in both syndromic and nonsyndromic congenital heart disease. Even though studies of genetic mouse models have previously revealed a requirement for VEGF-A, the class 3 semaphorin SEMA3C, and their shared receptor neuropilin 1 (NRP1) in OFT development, the precise mechanism by which these proteins orchestrate OFT septation is not yet understood. Here, we have analyzed a complementary set of ligand-specific and tissue-specific mouse mutants to show that neural crest-derived SEMA3C activates NRP1 in the OFT endothelium. Explant assays combined with gene-expression studies and lineage tracing further demonstrated that this signaling pathway promotes an endothelial-to-mesenchymal transition that supplies cells to the endocardial cushions and repositions cardiac neural crest cells (NCCs) within the OFT, 2 processes that are essential for septal bridge formation. These findings elucidate a mechanism by which NCCs cooperate with endothelial cells in the developing OFT to enable the postnatal separation of the pulmonary and systemic circulation.


Assuntos
Septos Cardíacos/embriologia , Ventrículos do Coração/metabolismo , Crista Neural/metabolismo , Neuropilina-1/metabolismo , Semaforinas/metabolismo , Animais , Apoptose , Proliferação de Células , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Feminino , Septos Cardíacos/citologia , Septos Cardíacos/metabolismo , Ventrículos do Coração/embriologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Crista Neural/embriologia , Neuropilina-1/deficiência , Neuropilina-1/genética , Gravidez , Semaforinas/deficiência , Semaforinas/genética , Transdução de Sinais , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Cell Metab ; 18(4): 505-18, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24093675

RESUMO

The deterioration of the inner blood-retinal barrier and consequent macular edema is a cardinal manifestation of diabetic retinopathy (DR) and the clinical feature most closely associated with loss of sight. We provide evidence from both human and animal studies for the critical role of the classical neuronal guidance cue, semaphorin 3A, in instigating pathological vascular permeability in diabetic retinas via its cognate receptor neuropilin-1. We reveal that semaphorin 3A is induced in early hyperglycemic phases of diabetes within the neuronal retina and precipitates initial breakdown of endothelial barrier function. We demonstrate, by a series of orthogonal approaches, that neutralization of semaphorin 3A efficiently prevents diabetes-induced retinal vascular leakage in a stage of the disease when vascular endothelial growth factor neutralization is inefficient. These observations were corroborated in Tg(Cre-Esr1)/Nrp1(flox/flox) conditional knockout mice. Our findings identify a therapeutic target for macular edema and provide further evidence for neurovascular crosstalk in the pathogenesis of DR.


Assuntos
Retinopatia Diabética/metabolismo , Neurônios/metabolismo , Semaforina-3A/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/patologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Pessoa de Meia-Idade , Neuropilina-1/deficiência , Neuropilina-1/genética , Neuropilina-1/metabolismo , Permeabilidade/efeitos dos fármacos , RNA Mensageiro/metabolismo , Retina/metabolismo , Retina/fisiopatologia , Semaforina-3A/genética , Semaforina-3A/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
11.
Nature ; 501(7466): 252-6, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23913274

RESUMO

Regulatory T cells (Treg cells) have a crucial role in the immune system by preventing autoimmunity, limiting immunopathology, and maintaining immune homeostasis. However, they also represent a major barrier to effective anti-tumour immunity and sterilizing immunity to chronic viral infections. The transcription factor Foxp3 has a major role in the development and programming of Treg cells. The relative stability of Treg cells at inflammatory disease sites has been a highly contentious subject. There is considerable interest in identifying pathways that control the stability of Treg cells as many immune-mediated diseases are characterized by either exacerbated or limited Treg-cell function. Here we show that the immune-cell-expressed ligand semaphorin-4a (Sema4a) and the Treg-cell-expressed receptor neuropilin-1 (Nrp1) interact both in vitro, to potentiate Treg-cell function and survival, and in vivo, at inflammatory sites. Using mice with a Treg-cell-restricted deletion of Nrp1, we show that Nrp1 is dispensable for suppression of autoimmunity and maintenance of immune homeostasis, but is required by Treg cells to limit anti-tumour immune responses and to cure established inflammatory colitis. Sema4a ligation of Nrp1 restrained Akt phosphorylation cellularly and at the immunologic synapse by phosphatase and tensin homologue (PTEN), which increased nuclear localization of the transcription factor Foxo3a. The Nrp1-induced transcriptome promoted Treg-cell stability by enhancing quiescence and survival factors while inhibiting programs that promote differentiation. Importantly, this Nrp1-dependent molecular program is evident in intra-tumoral Treg cells. Our data support a model in which Treg-cell stability can be subverted in certain inflammatory sites, but is maintained by a Sema4a-Nrp1 axis, highlighting this pathway as a potential therapeutic target that could limit Treg-cell-mediated tumour-induced tolerance without inducing autoimmunity.


Assuntos
Neuropilina-1/metabolismo , Semaforinas/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Autoimunidade/imunologia , Sobrevivência Celular , Colite/imunologia , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Homeostase/imunologia , Humanos , Tolerância Imunológica/imunologia , Sinapses Imunológicas , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Neuropilina-1/deficiência , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/citologia , Serina-Treonina Quinases TOR/metabolismo
12.
PLoS One ; 8(4): e61151, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23577203

RESUMO

Besides CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs), other immunosuppressive T cells also participated in the regulation of immune tolerance. Reportedly, neuropilin-1 (Nrp1) might be one of the molecules by which regulatory cells exert their suppressive effects. Indeed, CD4(+)CD25(-)Nrp1(+) T cells exhibit potent suppressive function in autoimmune inflammatory responses. Here we investigated the specific role of CD4(+)CD25(-)Nrp1(+) T cells in the setting of the transplant immune response. Through MLR assays, we found that CD4(+)CD25(-)Nrp1(+) T cells suppressed the proliferation of naive CD4(+)CD25(-) T cells activated by allogeneic antigen-stimulation. Adoptive transfer of CD4(+)CD25(-)Nrp1(+) T cells synergized with rapamycin to induce long-term graft survival in fully MHC-mismatched murine heart transplantation, which was associated with decreased IFN-γ, IL-17 and increased IL-10, TGF-ß, Foxp3 and Nrp1 expression in the grafts. Importantly, our data indicated that CD4(+)CD25(-)Nrp1(+) T cell transfer augments the accumulation of Tregs in the recipient, and creates conditions that favored induction of hyporesponsiveness of the T effector cells. In conclusion, this translational study indicates the possible therapeutic potential of CD4(+)CD25(-)Nrp1(+) T cells in preventing allorejection. CD4(+)Nrp1(+) T cells might therefore be used in bulk as a population of immunosuppressive cells with more beneficial properties concerning ex vivo isolation as compared to Foxp3(+) Tregs.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/efeitos adversos , Imunocompetência , Subunidade alfa de Receptor de Interleucina-2/deficiência , Neuropilina-1/deficiência , Sirolimo/farmacologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Separação Celular , Citocinas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Camundongos , Linfócitos T Reguladores/imunologia
13.
J Exp Med ; 209(11): 2001-16, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23045606

RESUMO

Infiltration of Foxp3(+) regulatory T (T reg) cells is considered to be a critical step during tumor development and progression. T reg cells supposedly suppress locally an effective anti-tumor immune response within tumor tissues, although the precise mechanism by which T reg cells infiltrate the tumor is still unclear. We provide evidence that Neuropilin 1 (Nrp-1), highly expressed by Foxp3(+) T reg cells, regulates the immunological anti-tumor control by guiding T reg cells into the tumor in response to tumor-derived vascular endothelial growth factor (VEGF). We demonstrate for the first time that T cell-specific ablation of Nrp-1 expression results in a significant breakdown in tumor immune escape in various transplantation models and in a spontaneous, endogenously driven melanoma model associated with strongly reduced tumor growth and prolonged tumor-free survival. Strikingly, numbers of tumor-infiltrating Foxp3(+) T reg cells were significantly reduced accompanied by enhanced activation of CD8(+) T cells within tumors of T cell-specific Nrp-1-deficient mice. This phenotype can be reversed by adoptive transfer of Nrp-1(+) T reg cells from wild-type mice. Thus, our data strongly suggest that Nrp-1 acts as a key mediator of Foxp3(+) T reg cell infiltration into the tumor site resulting in a dampened anti-tumor immune response and enhanced tumor progression.


Assuntos
Melanoma Experimental/imunologia , Neuropilina-1/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/imunologia , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Ativação Linfocitária/imunologia , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neuropilina-1/deficiência , Neuropilina-1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Linfócitos T Reguladores/metabolismo , Carga Tumoral/genética , Carga Tumoral/imunologia , Evasão Tumoral/genética , Evasão Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
14.
Cancer Res ; 72(22): 5801-11, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22986738

RESUMO

Neuropilin-1 (NRP1) is a coreceptor for multiple extracellular ligands. NRP1 is widely expressed in cancer cells and in advanced human tumors; however, its functional relevance and signaling mechanisms are unclear. Here, we show that NRP1 expression controls viability and proliferation of different cancer cells, independent of its short intracellular tail. We found that the extracellular domain of NRP1 interacts with the EGF receptor (EGFR) and promotes its signaling cascade elicited upon EGF or TGF-α stimulation. Upon NRP1 silencing, the ability of ligand-bound EGFR to cluster on the cell surface, internalize, and activate the downstream AKT pathway is severely impaired. EGFR is frequently activated in human tumors due to overexpression, mutation, or sustained autocrine/paracrine stimulation. Here we show that NRP1-blocking antibodies and NRP1 silencing can counteract ligand-induced EGFR activation in cancer cells. Thus our findings unveil a novel molecular mechanism by which NRP1 can control EGFR signaling and tumor growth.


Assuntos
Receptores ErbB/metabolismo , Neuropilina-1/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Endocitose , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Neuropilina-1/deficiência , Neuropilina-1/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção , Transplante Heterólogo
15.
Dev Biol ; 369(2): 277-85, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22790009

RESUMO

The sympathetic nervous system (SNS) arises from neural crest (NC) cells during embryonic development and innervates the internal organs of vertebrates to modulate their stress response. NRP1 and NRP2 are receptors for guidance cues of the class 3 semaphorin (SEMA) family and are expressed in partially overlapping patterns in sympathetic NC cells and their progeny. By comparing the phenotypes of mice lacking NRP1 or its ligand SEMA3A with mice lacking NRP1 in the sympathetic versus vascular endothelial cell lineages, we demonstrate that SEMA3A signalling through NRP1 has multiple cell-autonomous roles in SNS development. These roles include neuronal cell body positioning, neuronal aggregation and axon guidance, first during sympathetic chain assembly and then to regulate the innervation of the heart and aorta. Loss of NRP2 or its ligand SEMA3F impaired sympathetic gangliogenesis more mildly than loss of SEMA3A/NRP1 signalling, but caused ectopic neurite extension along the embryonic aorta. The analysis of compound mutants lacking SEMA3A and SEMA3F or NRP1 and NRP2 in the SNS demonstrated that both signalling pathways cooperate to organise the SNS. We further show that abnormal sympathetic development in mice lacking NRP1 in the sympathetic lineage has functional consequences, as it causes sinus bradycardia, similar to mice lacking SEMA3A.


Assuntos
Neurogênese/fisiologia , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/metabolismo , Animais , Aorta/embriologia , Aorta/inervação , Aorta/metabolismo , Axônios/metabolismo , Linhagem da Célula , Feminino , Coração Fetal/embriologia , Coração Fetal/inervação , Coração Fetal/metabolismo , Gânglios Simpáticos/crescimento & desenvolvimento , Gânglios Simpáticos/metabolismo , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/embriologia , Crista Neural/metabolismo , Neuritos/metabolismo , Neurogênese/genética , Neuropilina-1/deficiência , Neuropilina-1/genética , Neuropilina-2/deficiência , Neuropilina-2/genética , Gravidez , Semaforina-3A/deficiência , Semaforina-3A/genética , Semaforina-3A/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/citologia
16.
Circ Res ; 111(4): 437-45, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22723296

RESUMO

RATIONALE: The lymphatic vasculature plays a major role in fluid homeostasis, absorption of dietary lipids, and immune surveillance. Fluid transport depends on the presence of intraluminal valves within lymphatic collectors. Defective formation of lymphatic valves leads to lymphedema, a progressive and debilitating condition for which curative treatments are currently unavailable. How lymphatic valve formation is regulated remains largely unknown. OBJECTIVE: We investigated if the repulsive axon guidance molecule Semaphorin3A (Sema3A) plays a role in lymphatic valve formation. METHODS AND RESULTS: We show that Sema3A mRNA is expressed in lymphatic vessels and that Sema3A protein binds to lymphatic valves expressing the Neuropilin-1 (Nrp1) and PlexinA1 receptors. Using mouse knockout models, we show that Sema3A is selectively required for lymphatic valve formation, via interaction with Nrp1 and PlexinA1. Sema3a(-/-) mice exhibit defects in lymphatic valve formation, which are not due to abnormal lymphatic patterning or sprouting, and mice carrying a mutation in the Sema3A binding site of Nrp1, or deficient for Plxna1, develop lymphatic valve defects similar to those seen in Sema3a(-/-) mice. CONCLUSIONS: Our data demonstrate an essential direct function of Sema3A-Nrp1-PlexinA1 signaling in lymphatic valve formation.


Assuntos
Vasos Linfáticos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-1/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforina-3A/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/administração & dosagem , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Vasos Linfáticos/embriologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfogênese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neuropilina-1/deficiência , Neuropilina-1/genética , Neuropilina-1/imunologia , Fenótipo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Semaforina-3A/deficiência , Semaforina-3A/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Neuron ; 70(5): 951-65, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21658587

RESUMO

During development, the axons of retinal ganglion cell (RGC) neurons must decide whether to cross or avoid the midline at the optic chiasm to project to targets on both sides of the brain. By combining genetic analyses with in vitro assays, we show that neuropilin 1 (NRP1) promotes contralateral RGC projection in mammals. Unexpectedly, the NRP1 ligand involved is not an axon guidance cue of the class 3 semaphorin family, but VEGF164, the neuropilin-binding isoform of the classical vascular growth factor VEGF-A. VEGF164 is expressed at the chiasm midline and is required for normal contralateral growth in vivo. In outgrowth and growth cone turning assays, VEGF164 acts directly on NRP1-expressing contralateral RGCs to provide growth-promoting and chemoattractive signals. These findings have identified a permissive midline signal for axons at the chiasm midline and provide in vivo evidence that VEGF-A is an essential axon guidance cue.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuropilina-1/metabolismo , Quiasma Óptico/fisiologia , Transdução de Sinais/fisiologia , Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores Etários , Aminoácidos/metabolismo , Animais , Axônios/efeitos dos fármacos , Embrião de Mamíferos , Lateralidade Funcional , Regulação da Expressão Gênica no Desenvolvimento/genética , Cones de Crescimento/fisiologia , Indóis/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropilina-1/deficiência , Neuropilina-1/genética , Neuropilina-2/deficiência , Quiasma Óptico/efeitos dos fármacos , Quiasma Óptico/embriologia , Técnicas de Cultura de Órgãos , Retina/citologia , Retina/efeitos dos fármacos , Retina/embriologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/fisiologia , Semaforina-3A/deficiência , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/genética
18.
Dev Dyn ; 230(2): 299-308, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15162508

RESUMO

The chick embryo is widely used for the study of vertebrate development, but a general, reliable loss-of-function strategy for the analysis of gene function is currently not available. By using small inhibitory hairpin RNA (siRNA) molecules generated by the mouse U6 promoter, we have applied an RNA interference approach to achieve quantitative knockdown of the neuropilin-1 (Nrp-1) receptor in chick embryos. Functional knockdown was evident in the abolition of Sema3A-induced growth cone collapse in Nrp-1-siRNA but not Nrp-2-siRNA-expressing dorsal root ganglion (DRG) neurons. Two nervous system defects in Nrp-1 mutant mice were phenocopied in embryos treated with Nrp-1 siRNA. First, DRG axons prematurely entered the dorsal horn and projected inappropriately. Second, targeted early migrating neural crest cells destined for the sympathetic chain arrested ectopically within ventral spinal nerve roots. Localized knockdown induced by specific siRNA constructs will allow rapid functional analysis of genes regulating chick neural development whilst circumventing embryonic lethal effects often associated with global gene knockout in the mouse.


Assuntos
Galinhas/genética , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Neuropilina-1/deficiência , Neuropilina-1/genética , RNA Interferente Pequeno/metabolismo , Animais , Aves , Diferenciação Celular , Células Cultivadas , Embrião de Galinha , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Gânglios Espinais/metabolismo , Deleção de Genes , Vetores Genéticos/genética , Técnicas In Vitro , Camundongos , Camundongos Knockout , Sistema Nervoso/citologia , Crista Neural/citologia , Crista Neural/metabolismo , Neuropilina-1/metabolismo , Conformação de Ácido Nucleico , Fenótipo , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Nuclear Pequeno/genética
19.
Mol Cell Neurosci ; 25(2): 217-27, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15019939

RESUMO

During the initial stages of development, the notochord provides repulsive signals for dorsal root ganglion (DRG) axons via semaphorin 3A/neuropilin-1, axonin-1/SC2, and other unknown repulsive molecules. The notochord is known to produce aggrecan, one of the chondroitin sulfate proteoglycans (CSPGs). We report here that adding aggrecan to the culture medium cannot only induce DRG growth cone collapse, but also inhibit DRG axonal growth. Using cocultures composed of tissues derived from chick embryos or neuropilin-1-deficient mice treated with chondroitinase ABC, we show the direct evidence that CSPGs are involved in notochord-derived repulsion for DRG axons. At later developmental stages, CSPGs are involved in perinotochordal sheath-derived axon repulsion, but not in notochord core-derived repulsion. We further demonstrate that TAG-1/axonin-1/SC2 is not involved in mediating repulsive activities by CSPGs, but is required for notochord core-derived axon repulsion. Thus, notochord-derived multiple axon repulsions act in a spatiotemporal-specific manner to shape the initial trajectories of DRG axons.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas da Matriz Extracelular , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cones de Crescimento/metabolismo , Fatores de Crescimento Neural/metabolismo , Notocorda/metabolismo , Agrecanas , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Embrião de Galinha , Condroitina ABC Liase/farmacologia , Técnicas de Cocultura , Contactina 2 , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Cones de Crescimento/ultraestrutura , Lectinas Tipo C , Camundongos , Camundongos Knockout , Neuropilina-1/deficiência , Neuropilina-1/genética , Técnicas de Cultura de Órgãos , Proteoglicanas/metabolismo , Proteoglicanas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...