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










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2475: 205-214, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35451759

RESUMO

In vitro assays of endothelial cell migration have led to critical insights into the mechanisms of angiogenesis. The transwell assay, or modified Boyden chamber assay was developed to investigate chemotaxis, which corresponds to the directional migration of cells in response to a chemoattractant gradient. It is a reliable and convenient assay that does not require expensive equipment.In the modified Boyden chamber assay, two compartments are separated with a porous membrane through which cells can migrate. The lower compartment contains the chemoattractant, creating a gradient by diffusing into the upper chamber containing the cells. Adherent cells will migrate through the membrane and remain on the lower side of the membrane, where they can finally be fixed, stained, and counted.


Assuntos
Células Endoteliais , Fator A de Crescimento do Endotélio Vascular , Movimento Celular , Fatores Quimiotáticos , Quimiotaxia/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
2.
J Cardiovasc Dev Dis ; 8(5)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062899

RESUMO

In zebrafish, the spatiotemporal development of the vascular system is well described due to its stereotypical nature. However, the cellular and molecular mechanisms orchestrating post-embryonic vascular development, the maintenance of vascular homeostasis, or how coronary vessels integrate into the growing heart are less well studied. In the context of cardiac regeneration, the central cellular mechanism by which the heart regenerates a fully functional myocardium relies on the proliferation of pre-existing cardiomyocytes; the epicardium and the endocardium are also known to play key roles in the regenerative process. Remarkably, revascularisation of the injured tissue occurs within a few hours after cardiac damage, thus generating a vascular network acting as a scaffold for the regenerating myocardium. The activation of the endocardium leads to the secretion of cytokines, further supporting the proliferation of the cardiomyocytes. Although epicardium, endocardium, and myocardium interact with each other to orchestrate heart development and regeneration, in this review, we focus on recent advances in the understanding of the development of the endocardium and the coronary vasculature in zebrafish as well as their pivotal roles in the heart regeneration process.

3.
Int J Mol Sci ; 21(14)2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32708258

RESUMO

Cardiovascular disease, the leading cause of death worldwide, is predominantly associated with atherosclerosis. Atherosclerosis is a chronic inflammatory disease characterised by the narrowing of large to medium-sized arteries due to a build-up of plaque. Atherosclerotic plaque is comprised of lipids, extracellular matrix, and several cell types, including endothelial, immune, and vascular smooth muscle cells. Such narrowing of the blood vessels can itself restrict blood flow to vital organs but most severe clinical complications, including heart attacks and strokes, occur when lesions rupture, triggering the blood to clot and obstructing blood flow further down the vascular tree. To circumvent such obstructions, percutaneous coronary intervention or bypass grafts are often required; however, re-occlusion of the treated artery frequently occurs. Neuropilins (NRPs), a multifunctional family of cell surface co-receptors, are expressed by endothelial, immune, and vascular smooth muscle cells and are regulators of numerous signalling pathways within the vasculature. Here, we review recent studies implicating NRP2 in the development of occlusive vascular diseases and discuss how NRP2 could be targeted for therapeutic intervention.


Assuntos
Aterosclerose/metabolismo , Doenças Cardiovasculares/metabolismo , Células Endoteliais/metabolismo , Miócitos de Músculo Liso/metabolismo , Neuropilina-2/metabolismo , Placa Aterosclerótica/metabolismo , Animais , Células Endoteliais/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Monócitos/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Neovascularização Patológica , Neuropilina-2/uso terapêutico , Placa Aterosclerótica/patologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/genética
4.
Adv Healthc Mater ; 9(17): e2000806, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32666663

RESUMO

Peripheral arterial disease and critical limb ischemia are common symptoms of cardiovascular disease. Vascular surgery is used to create a bypass around occluded blood vessels to improve blood flow to ischemic muscle, thus avoiding the need for amputation. Attempts to vascularize tissues by therapeutic angiogenesis using delivery of exogenous angiogenic agents are underwhelming. A material-based approach that provides an endogenous stimulus capable of promoting angiogenesis and increased tissue perfusion would provide a paradigm shift in treatment options available. It is reported here that microporous biodegradable films produced using thermally induced phase separation provide a localized biophysical stimulus of proangiogenic genes in vivo that is associated with increased blood vessel density and restoration of blood flow to ischemic tissue. These findings show, for the first time, that acellular, nonfunctionalized biodegradable biomaterials can provide an innovative, material-based approach for therapeutic angiogenesis to enhance tissue reperfusion in vivo.


Assuntos
Isquemia , Neovascularização Fisiológica , Animais , Materiais Biocompatíveis , Modelos Animais de Doenças , Membro Posterior , Isquemia/terapia , Neovascularização Patológica
5.
Development ; 146(13)2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31167777

RESUMO

Unlike adult mammals, zebrafish can regenerate their heart. A key mechanism for regeneration is the activation of the epicardium, leading to the establishment of a supporting scaffold for new cardiomyocytes, angiogenesis and cytokine secretion. Neuropilins are co-receptors that mediate signaling of kinase receptors for cytokines with crucial roles in zebrafish heart regeneration. We investigated the role of neuropilins in response to cardiac injury and heart regeneration. All four neuropilin isoforms (nrp1a, nrp1b, nrp2a and nrp2b) were upregulated by the activated epicardium and an nrp1a-knockout mutant showed a significant delay in heart regeneration and displayed persistent collagen deposition. The regenerating hearts of nrp1a mutants were less vascularized, and epicardial-derived cell migration and re-expression of the developmental gene wt1b was impaired. Moreover, cryoinjury-induced activation and migration of epicardial cells in heart explants were reduced in nrp1a mutants. These results identify a key role for Nrp1 in zebrafish heart regeneration, mediated through epicardial activation, migration and revascularization.


Assuntos
Coração/fisiologia , Neovascularização Fisiológica/genética , Neuropilina-1/fisiologia , Pericárdio/fisiologia , Regeneração/genética , Animais , Animais Geneticamente Modificados , Movimento Celular/genética , Células Cultivadas , Temperatura Baixa , Vasos Coronários/fisiologia , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/patologia , Traumatismos Cardíacos/fisiopatologia , Miócitos Cardíacos/fisiologia , Neuropilina-1/genética , Ratos , Peixe-Zebra/fisiologia
6.
Am J Physiol Cell Physiol ; 316(3): C424-C433, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649916

RESUMO

Neuropilin 1 (NRP1) is important for neuronal and cardiovascular development due to its role in conveying class 3 semaphorin and vascular endothelial growth factor signaling, respectively. NRP1 is expressed in smooth muscle cells (SMCs) and mediates their migration and proliferation in cell culture and is implicated in pathological SMC remodeling in vivo. To address the importance of Nrp1 for SMC function during development, we generated conditional inducible Nrp1 SMC-specific knockout mice. Induction of early postnatal SMC-specific Nrp1 knockout led to pulmonary hemorrhage associated with defects in alveogenesis and revealed a specific requirement for Nrp1 in myofibroblast recruitment to the alveolar septae and PDGF-AA-induced migration in vitro. Furthermore, SMC-specific Nrp1 knockout inhibited PDGF-BB-stimulated SMC outgrowth ex vivo in aortic ring assays and reduced pathological arterial neointima formation in vivo. In contrast, we observed little significant effect of SMC-specific Nrp1 knockout on neonatal retinal vascularization. Our results point to a requirement of Nrp1 in vascular smooth muscle and myofibroblast function in vivo, which may have relevance for postnatal lung development and for pathologies characterized by excessive SMC and/or myofibroblast proliferation.


Assuntos
Pulmão/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neuropilina-1/metabolismo , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/metabolismo , Neointima/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 38(8): 1845-1858, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29880492

RESUMO

Objective- NRP1(neuropilin-1) acts as a coreceptor for VEGF (vascular endothelial growth factor) with an essential role in angiogenesis. Recent findings suggest that posttranslational proteolytic cleavage of VEGF receptors may be an important mechanism for regulating angiogenesis, but the role of NRP1 proteolysis and the NRP1 species generated by cleavage in endothelial cells is not known. Here, we characterize NRP1 proteolytic cleavage in endothelial cells, determine the mechanism, and investigate the role of NRP1 cleavage in regulation of endothelial cell function. Approach and Results- NRP1 species comprising the carboxy (C)-terminal and transmembrane NRP1 domains but lacking the ligand-binding A and B regions are constitutively expressed in endothelial cells. Generation of these C-terminal domain NRP1 proteins is upregulated by phorbol ester and Ca2+ ionophore, and reduced by pharmacological inhibition of metalloproteinases, by small interfering RNA-mediated knockdown of 2 members of ADAM (a disintegrin and metalloproteinase) family, ADAMs 9 and 10, and by a specific ADAM10 inhibitor. Furthermore, VEGF upregulates expression of these NRP1 species in an ADAM9/10-dependent manner. Transduction of endothelial cells with adenoviral constructs expressing NRP1 C-terminal domain fragments inhibited VEGF-induced phosphorylation of VEGFR2 (VEGF receptor tyrosine kinase)/KDR (kinase domain insert receptor) and decreased VEGF-stimulated endothelial cell motility and angiogenesis in coculture and aortic ring sprouting assays. Conclusions- These findings identify novel NRP1 species in endothelial cells and demonstrate that regulation of NRP1 proteolysis via ADAMs 9 and 10 is a new regulatory pathway able to modulate VEGF angiogenic signaling.


Assuntos
Proteínas ADAMTS/metabolismo , Proteína ADAMTS9/metabolismo , Indutores da Angiogênese/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Neuropilina-1/metabolismo , Fragmentos de Peptídeos/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas ADAMTS/genética , Proteína ADAMTS9/genética , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Camundongos , Neuropilina-1/genética , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Transdução de Sinais/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Cells ; 7(3)2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29543760

RESUMO

Receptor tyrosine kinases (RTKs) are membrane-based sensors that enable rapid communication between cells and their environment. Evidence is now emerging that interdependent regulatory mechanisms, such as membrane trafficking, ubiquitination, proteolysis and gene expression, have substantial effects on RTK signal transduction and cellular responses. Different RTKs exhibit both basal and ligand-stimulated ubiquitination, linked to trafficking through different intracellular compartments including the secretory pathway, plasma membrane, endosomes and lysosomes. The ubiquitin ligase superfamily comprising the E1, E2 and E3 enzymes are increasingly implicated in this post-translational modification by adding mono- and polyubiquitin tags to RTKs. Conversely, removal of these ubiquitin tags by proteases called de-ubiquitinases (DUBs) enables RTK recycling for another round of ligand sensing and signal transduction. The endocytosis of basal and activated RTKs from the plasma membrane is closely linked to controlled proteolysis after trafficking and delivery to late endosomes and lysosomes. Proteolytic RTK fragments can also have the capacity to move to compartments such as the nucleus and regulate gene expression. Such mechanistic diversity now provides new opportunities for modulating RTK-regulated cellular responses in health and disease states.

9.
Cardiovasc Res ; 108(2): 288-98, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26410366

RESUMO

AIMS: Neuropilins 1 and 2 (NRP1 and NRP2) play crucial roles in endothelial cell migration contributing to angiogenesis and vascular development. Both NRPs are also expressed by cultured vascular smooth muscle cells (VSMCs) and are implicated in VSMC migration stimulated by PDGF-BB, but it is unknown whether NRPs are relevant for VSMC function in vivo. We investigated the role of NRPs in the rat carotid balloon injury model, in which endothelial denudation and arterial stretch induce neointimal hyperplasia involving VSMC migration and proliferation. METHODS AND RESULTS: NRP1 and NRP2 mRNAs and proteins increased significantly following arterial injury, and immunofluorescent staining revealed neointimal NRP expression. Down-regulation of NRP1 and NRP2 using shRNA significantly reduced neointimal hyperplasia following injury. Furthermore, inhibition of NRP1 by adenovirally overexpressing a loss-of-function NRP1 mutant lacking the cytoplasmic domain (ΔC) reduced neointimal hyperplasia, whereas wild-type (WT) NRP1 had no effect. NRP-targeted shRNAs impaired, while overexpression of NRP1 WT and NRP1 ΔC enhanced, arterial re-endothelialization 14 days after injury. Knockdown of either NRP1 or NRP2 inhibited PDGF-BB-induced rat VSMC migration, whereas knockdown of NRP2, but not NRP1, reduced proliferation of cultured rat VSMC and neointimal VSMC in vivo. NRP knockdown also reduced the phosphorylation of PDGFα and PDGFß receptors in rat VSMC, which mediate VSMC migration and proliferation. CONCLUSION: NRP1 and NRP2 play important roles in the regulation of neointimal hyperplasia in vivo by modulating VSMC migration (via NRP1 and NRP2) and proliferation (via NRP2), independently of the role of NRPs in re-endothelialization.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Neointima/metabolismo , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Angioplastia com Balão/efeitos adversos , Animais , Lesões das Artérias Carótidas/etiologia , Lesões das Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Hiperplasia , Masculino , Miócitos de Músculo Liso/metabolismo , Neointima/etiologia , Neointima/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos Sprague-Dawley , Regulação para Cima
10.
Methods Mol Biol ; 1332: 151-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26285752

RESUMO

In vitro assays of endothelial cell migration have led to critical insights into the mechanisms of angiogenesis. The transwell assay, or modified Boyden chamber assay was developed to investigate chemotaxis, which corresponds to the directional migration of cells in response to a chemoattractant gradient. It is a robust and easy-to-use assay that does not require expensive equipment. In the modified Boyden chamber assay, two compartments are separated with a porous membrane through which cells can migrate. The lower compartment contains the chemoattractant, creating a gradient by diffusing into the upper chamber containing the cells. The cells will migrate through the membrane and remain on the lower side of the membrane, where they can finally be fixed and counted.


Assuntos
Quimiotaxia , Células Endoteliais/fisiologia , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Humanos , Técnicas In Vitro
11.
Cells ; 3(2): 363-85, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24785348

RESUMO

Rab GTPases are implicated in endosome-to-plasma membrane recycling, but how such membrane traffic regulators control vascular endothelial growth factor receptor 2 (VEGFR2/KDR) dynamics and function are not well understood. Here, we evaluated two different recycling Rab GTPases, Rab4a and Rab11a, in regulating endothelial VEGFR2 trafficking and signalling with implications for endothelial cell migration, proliferation and angiogenesis. In primary endothelial cells, VEGFR2 displays co-localisation with Rab4a, but not Rab11a GTPase, on early endosomes. Expression of a guanosine diphosphate (GDP)-bound Rab4a S22N mutant caused increased VEGFR2 accumulation in endosomes. TfR and VEGFR2 exhibited differences in endosome-to-plasma membrane recycling in the presence of chloroquine. Depletion of Rab4a, but not Rab11a, levels stimulated VEGF-A-dependent intracellular signalling. However, depletion of either Rab4a or Rab11a levels inhibited VEGF-A-stimulated endothelial cell migration. Interestingly, depletion of Rab4a levels stimulated VEGF-A-regulated endothelial cell proliferation. Rab4a and Rab11a were also both required for endothelial tubulogenesis. Evaluation of a transgenic zebrafish model showed that both Rab4 and Rab11a are functionally required for blood vessel formation and animal viability. Rab-dependent endosome-to-plasma membrane recycling of VEGFR2 is important for intracellular signalling, cell migration and proliferation during angiogenesis.

12.
J Cell Sci ; 127(Pt 12): 2647-58, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24762811

RESUMO

DOK1 regulates platelet-derived growth factor (PDGF)-BB-stimulated glioma cell motility. Mechanisms regulating tumour cell motility are essential for invasion and metastasis. We report here that PDGF-BB-mediated glioma cell invasion and migration are dependent on the adaptor protein downstream of kinase 1 (DOK1). DOK1 is expressed in several glioma cell lines and in tumour biopsies from high-grade gliomas. DOK1 becomes tyrosine phosphorylated upon PDGF-BB stimulation of human glioma cells. Knockdown of DOK1 or expression of a DOK1 mutant (DOK1FF) containing Phe in place of Tyr at residues 362 and 398, resulted in inhibition of both the PDGF-BB-induced tyrosine phosphorylation of p130Cas (also known as BCAR1) and the activation of Rap1. DOK1 colocalises with tyrosine phosphorylated p130Cas at the cell membrane of PDGF-BB-treated cells. Expression of a non-tyrosine-phosphorylatable substrate domain mutant of p130Cas (p130Cas15F) inhibited PDGF-BB-mediated Rap1 activation. Knockdown of DOK1 and Rap1 inhibited PDGF-BB-induced chemotactic cell migration, and knockdown of DOK1 and Rap1 and expression of DOK1FF inhibited PDGF-mediated three-dimensional (3D) spheroid invasion. These data show a crucial role for DOK1 in the regulation of PDGF-BB-mediated tumour cell motility through a p130Cas-Rap1 signalling pathway. [Corrected]


Assuntos
Neoplasias Encefálicas/metabolismo , Proteína Substrato Associada a Crk/metabolismo , Proteínas de Ligação a DNA/fisiologia , Glioblastoma/metabolismo , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Proteínas de Ligação a RNA/fisiologia , Proteínas de Ligação a Telômeros/metabolismo , Becaplermina , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Quimiotaxia , Glioblastoma/patologia , Humanos , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Complexo Shelterina , Transdução de Sinais , Quinases da Família src/metabolismo
13.
Cell Signal ; 25(4): 766-77, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23277200

RESUMO

p130Cas/breast cancer anti-oestrogen resistance 1 (BCAR1) is a member of the Cas (Crk-associated substrate) family of adaptor proteins, which have emerged as key signalling nodes capable of interactions with multiple proteins, with important regulatory roles in normal and pathological cell function. The Cas family of proteins is characterised by the presence of multiple conserved motifs for protein-protein interactions, and by extensive tyrosine and serine phosphorylations. Recent studies show that p130Cas contributes to migration, cell cycle control and apoptosis. p130Cas is essential during early embryogenesis, with a critical role in cardiovascular development. Furthermore, p130Cas has been reported to be involved in the development and progression of several human cancers. p130Cas is able to perform roles in multiple processes due to its capacity to regulate a diverse array of signalling pathways, transducing signals from growth factor receptor tyrosine kinases, non-receptor tyrosine kinases, and integrins. In this review we summarise the current understanding of the structure, function, and regulation of p130Cas, and discuss the importance of p130Cas in both physiological and pathophysiological settings, with a focus on the cardiovascular system and cancer.


Assuntos
Proteína Substrato Associada a Crk/metabolismo , Transdução de Sinais , Sistema Cardiovascular/metabolismo , Proteína Substrato Associada a Crk/análise , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Mecanotransdução Celular , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação
14.
PLoS One ; 7(11): e48539, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139789

RESUMO

Vascular endothelial growth factor A (VEGF-A) binds to the VEGFR2 receptor tyrosine kinase, regulating endothelial function, vascular physiology and angiogenesis. However, the mechanism underlying VEGFR2 turnover and degradation in this response is unclear. Here, we tested a role for heat-shock proteins in regulating the presentation of VEGFR2 to a degradative pathway. Pharmacological inhibition of HSP90 stimulated VEGFR2 degradation in primary endothelial cells and blocked VEGF-A-stimulated intracellular signaling via VEGFR2. HSP90 inhibition stimulated the formation of a VEGFR2-HSP70 complex. Clathrin-mediated VEGFR2 endocytosis is required for this HSP-linked degradative pathway for targeting VEGFR2 to the endosome-lysosome system. HSP90 perturbation selectively inhibited VEGF-A-stimulated human endothelial cell migration in vitro. A mouse femoral artery model showed that HSP90 inhibition also blocked blood vessel repair in vivo consistent with decreased endothelial regeneration. Depletion of either HSP70 or HSP90 caused defects in blood vessel formation in a transgenic zebrafish model. We conclude that perturbation of the HSP70-HSP90 heat-shock protein axis stimulates degradation of endothelial VEGFR2 and modulates VEGF-A-stimulated intracellular signaling, endothelial cell migration, blood vessel development and repair.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Proteínas de Choque Térmico/metabolismo , Neovascularização Fisiológica , Proteólise , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , Animais , Artérias/efeitos dos fármacos , Artérias/fisiologia , Benzoquinonas/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Movimento Celular/efeitos dos fármacos , Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Lactamas Macrocíclicas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Peixe-Zebra
15.
Mol Biol Cell ; 22(15): 2766-76, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21653826

RESUMO

In endothelial cells, neuropilin-1 (NRP1) binds vascular endothelial growth factor (VEGF)-A and is thought to act as a coreceptor for kinase insert domain-containing receptor (KDR) by associating with KDR and enhancing VEGF signaling. Here we report mutations in the NRP1 b1 domain (Y297A and D320A), which result in complete loss of VEGF binding. Overexpression of Y297A and D320A NRP1 in human umbilical vein endothelial cells reduced high-affinity VEGF binding and migration toward a VEGF gradient, and markedly inhibited VEGF-induced angiogenesis in a coculture cell model. The Y297A NRP1 mutant also disrupted complexation between NRP1 and KDR and decreased VEGF-dependent phosphorylation of focal adhesion kinase at Tyr407, but had little effect on other signaling pathways. Y297A NRP1, however, heterodimerized with wild-type NRP1 and NRP2 indicating that nonbinding NRP1 mutants can act in a dominant-negative manner through formation of NRP1 dimers with reduced binding affinity for VEGF. These findings indicate that VEGF binding to NRP1 has specific effects on endothelial cell signaling and is important for endothelial cell migration and angiogenesis mediated via complex formation between NRP1 and KDR and increased signaling to focal adhesions. Identification of key residues essential for VEGF binding and biological functions provides the basis for a rational design of antagonists of VEGF binding to NRP1.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Neuropilina-1/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Sítios de Ligação , Adesão Celular , Movimento Celular , Técnicas de Cocultura , Dimerização , Células Endoteliais/citologia , Endotélio Vascular/citologia , Quinase 1 de Adesão Focal/genética , Humanos , Mutação , Neovascularização Fisiológica , Neuropilina-1/genética , Fosforilação , Plasmídeos , Ligação Proteica , Transdução de Sinais/fisiologia , Transfecção , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
16.
Biochem J ; 435(3): 609-18, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21306301

RESUMO

NRP1 (neuropilin-1) is a co-receptor for members of the VEGF (vascular endothelial growth factor) family in endothelial cells, but is increasingly implicated in signalling induced by other growth factors. NRP1 is expressed in VSMCs (vascular smooth muscle cells), but its function and the mechanisms involved are poorly understood. The present study aimed to determine the role of NRP1 in the migratory response of HCASMCs (human coronary artery smooth muscle cells) to PDGF (platelet-derived growth factor), and to identify the signalling mechanisms involved. NRP1 is highly expressed in HAoSMCs (human aortic smooth muscle cells) and HCASMCs, and modified in VSMCs by CS (chondroitin sulfate)-rich O-linked glycosylation at Ser612. HCASMC migration induced by PDGF-BB and PDGF-AA was inhibited by NRP1 siRNA (small interfering RNA), and by adenoviral overexpression of an NRP1 mutant lacking the intracellular domain (Ad.NRP1ΔC). NRP1 co-immunoprecipitated with PDGFRα (PDGF receptor α), and immunofluorescent staining indicated that NRP1 and PDGFRα co-localized in VSMCs. NRP1 siRNA also inhibited PDGF-induced PDGFRα activation. NRP1-specific siRNA, Ad.NRP1ΔC and removal of CS glycans using chondroitinase all inhibited PDGF-BB and -AA stimulation of tyrosine phosphorylation of the adapter protein, p130Cas (Cas is Crk-associated substrate), with little effect on other major signalling pathways, and p130Cas knockdown inhibited HCASMC migration. Chemotaxis and p130Cas phosphorylation induced by PDGF were inhibited by chondroitinase, and, additionally, adenoviral expression of a non-glycosylatable NRP1S612A mutant inhibited chemotaxis, but not p130Cas phosphorylation. These results indicate a role for NRP1 and NRP1 glycosylation in mediating PDGF-induced VSMC migration, possibly by acting as a co-receptor for PDGFRα and via selective mobilization of a novel p130Cas tyrosine phosphorylation pathway.


Assuntos
Proteína Substrato Associada a Crk/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Neuropilina-1/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Adenoviridae , Movimento Celular , Células Cultivadas , Quimiotaxia , Proteína Substrato Associada a Crk/genética , Células Endoteliais , Humanos , Mutação , Miócitos de Músculo Liso/citologia , Neuropilina-1/genética , Fator de Crescimento Derivado de Plaquetas/genética
17.
Mol Cell Biol ; 31(6): 1174-85, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21245381

RESUMO

Neuropilin-1 (NRP1) is a receptor for vascular endothelial growth factor (VEGF) and plays an important role in mediating cell motility. However, the NRP1 signaling pathways important for cell motility are poorly understood. Here we report that p130(Cas) tyrosine phosphorylation is stimulated by hepatocyte growth factor and platelet-derived growth factor in U87MG glioma cells and VEGF in endothelial cells and is dependent on NRP1 via its intracellular domain. In endothelial cells, NRP1 silencing reduced, but did not prevent, VEGF receptor 2 (VEGFR2) phosphorylation, while expression of a mutant form of NRP1 lacking the intracellular domain (NRP1ΔC) did not affect receptor phosphorylation in U87MG cells or human umbilical vein endothelial cells (HUVECs). In HUVECs, NRP1 was also required for VEGF-induced phosphorylation of proline-rich tyrosine kinase 2, which was necessary for p130(Cas) phosphorylation. Importantly, knockdown of NRP1 or p130(Cas) or expression of either NRP1ΔC or a non-tyrosine-phosphorylatable substrate domain mutant protein (p130(Cas15F)) was sufficient to inhibit growth factor-mediated migration of glioma and endothelial cells. These data demonstrate for the first time the importance of the NRP1 intracellular domain in mediating a specific signaling pathway downstream of several receptor tyrosine kinases and identify a critical role for a novel NRP1-p130(Cas) pathway in the regulation of chemotaxis.


Assuntos
Movimento Celular , Proteína Substrato Associada a Crk/metabolismo , Células Endoteliais/metabolismo , Glioma/metabolismo , Neuropilina-1/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proteína Substrato Associada a Crk/genética , Células Endoteliais/citologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Glioma/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Mutação , Neuropilina-1/genética , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Tirosina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
Biochem Soc Trans ; 37(Pt 6): 1171-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19909241

RESUMO

NRPs (neuropilins) are receptors for class 3 semaphorins, polypeptides essential for axonal guidance, and for members of the VEGF (vascular endothelial growth factor) family of angiogenic cytokines. While mutant mouse studies show that NRP1 is essential for neuronal and cardiovascular development, little is known concerning the molecular mechanisms through which NRPs mediate the functions of their ligands in different cell types. NRP1 forms complexes with its co-receptors and is required for optimal function, but NRPs lack a clearly defined signalling domain and the role of NRP1 in receptor signalling and the function of the NRP1 cytosolic domain are unclear. Growing evidence indicates, however, that NRP1 plays a selective role in signalling at least in part via its C-terminal domain and interaction with intracellular binding partners.


Assuntos
Neuropilinas/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Endoteliais/metabolismo , Humanos , Neurônios/metabolismo , Neuropilinas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
EMBO Rep ; 9(10): 983-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18704117

RESUMO

Neuropilin 1 (NRP1), a non-tyrosine kinase receptor for vascular endothelial growth factor and class 3 Semaphorins, is highly expressed in many human tumour cell lines, but its function is poorly understood. Here, we describe the expression of a new chondroitin sulphate-modified NRP1 (NRP1-CS) in human tumour cell lines. Expression of a non-modifiable NRP1 mutant (S612A) in U87MG human glioma cells results in enhanced invasion in three dimensions (3D), whereas wild-type NRP1 has no effect. Furthermore, the S612A NRP1 cells show a significant increase in p130Cas tyrosine phosphorylation compared with control and wild-type NRP1 cells. Silencing of p130Cas in S612A NRP1 cells resulted in a loss of increased invasive phenotype. Interestingly, p130Cas silencing does not inhibit basal 3D invasion, but leads to a mesenchymal to amoeboid transition. Biopsies from both low- and high-grade human gliomas show strong expression of NRP1, and little expression of NRP1-CS. Our data establish distinct roles for NRP1 and NRP1-CS in modulating a new NRP1-p130Cas signalling pathway contributing to glioblastoma cell invasion in 3D.


Assuntos
Sulfatos de Condroitina/fisiologia , Proteína Substrato Associada a Crk/fisiologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Neuropilina-1/genética , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Glioblastoma/genética , Humanos , Camundongos , Dados de Sequência Molecular , Invasividade Neoplásica , Neuropilina-1/biossíntese , Interferência de RNA , Ratos , Suínos
20.
Biochem J ; 411(2): 211-26, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18363553

RESUMO

NRPs (neuropilins) are co-receptors for class 3 semaphorins, polypeptides with key roles in axonal guidance, and for members of the VEGF (vascular endothelial growth factor) family of angiogenic cytokines. They lack a defined signalling role, but are thought to mediate functional responses as a result of complex formation with other receptors, such as plexins in the case of semaphorins and VEGF receptors (e.g. VEGFR2). Mutant mouse studies show that NRP1 is essential for neuronal and cardiovascular development, whereas NRP2 has a more restricted role in neuronal patterning and lymphangiogenesis, but recent findings indicate that NRPs may have additional biological roles in other physiological and disease-related settings. In particular, NRPs are highly expressed in diverse tumour cell lines and human neoplasms and have been implicated in tumour growth and vascularization in vivo. However, despite the wealth of information regarding the probable biological roles of these molecules, many aspects of the regulation of cellular function via NRPs remain uncertain, and little is known concerning the molecular mechanisms through which NRPs mediate the functions of their various ligands in different cell types.


Assuntos
Neuropilinas/química , Neuropilinas/metabolismo , Animais , Doença , Regulação da Expressão Gênica , Genoma/genética , Humanos , Ligantes , Neuropilinas/genética , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...