Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 629, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789481

RESUMEN

Integrin trafficking to and from membrane adhesions is a crucial mechanism that dictates many aspects of a cell's behaviour, including motility, polarisation, and invasion. In endothelial cells (ECs), the intracellular traffic of α5 integrin is regulated by both neuropilin 1 (NRP1) and neuropilin 2 (NRP2), yet the redundancies in function between these co-receptors remain unclear. Moreover, the endocytic complexes that participate in NRP-directed traffic remain poorly annotated. Here we identify an important role for the GTPase-activating protein p120RasGAP in ECs, promoting the recycling of α5 integrin from early endosomes. Mechanistically, p120RasGAP enables transit of endocytosed α5 integrin-NRP1-NRP2 complexes to Rab11+ recycling endosomes, promoting cell polarisation and fibronectin (FN) fibrillogenesis. Silencing of both NRP receptors, or p120RasGAP, resulted in the accumulation of α5 integrin in early endosomes, a loss of α5 integrin from surface adhesions, and attenuated EC polarisation. Endothelial-specific deletion of both NRP1 and NRP2 in the postnatal retina recapitulated our in vitro findings, severely impairing FN fibrillogenesis and polarised sprouting. Our data assign an essential role for p120RasGAP during integrin traffic in ECs and support a hypothesis that NRP receptors co-traffic internalised cargoes. Importantly, we utilise comparative proteomics analyses to isolate a comprehensive map of NRP1-dependent and NRP2-dependent α5 integrin interactions in ECs.


Asunto(s)
Endosomas , Células Endoteliales , Fibronectinas , Integrina alfa5 , Neuropilina-1 , Neuropilina-2 , Proteómica , Proteína Activadora de GTPasa p120 , Neuropilina-1/metabolismo , Neuropilina-1/genética , Humanos , Integrina alfa5/metabolismo , Integrina alfa5/genética , Endosomas/metabolismo , Proteómica/métodos , Neuropilina-2/metabolismo , Neuropilina-2/genética , Animales , Fibronectinas/metabolismo , Células Endoteliales/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Proteína Activadora de GTPasa p120/genética , Transporte de Proteínas , Ratones , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Integrinas
2.
Methods Mol Biol ; 2475: 133-141, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35451754

RESUMEN

The ability to study the role of specific genes in endothelial cell biology is made possible by our ability to modulate their expression through siRNA or knockout technologies. However, many in vitro protocols, particularly those of a biochemical nature, require large numbers of endothelial cells. These types of analyses are encumbered by the need to repeatedly produce and characterize primary endothelial cell cultures and can be greatly facilitated by the use of immortalized microvascular endothelial cells. However, we have found that the manipulation of gene expression in these cells is not always straight forward. Here we describe how we alter gene expression in polyoma middle T antigen immortalized microvascular endothelial cells isolated from wild-type and genetically modified mice to study the role of cell adhesion molecules in downstream assays.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Ratones , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Cancer Res Commun ; 2(12): 1626-1640, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36970722

RESUMEN

Neuropilin (NRP) expression is highly correlated with poor outcome in multiple cancer subtypes. As known coreceptors for VEGFRs, core drivers of angiogenesis, past investigations have alluded to their functional roles in facilitating tumorigenesis by promoting invasive vessel growth. Despite this, it remains unclear as to whether NRP1 and NRP2 act in a synergistic manner to enhance pathologic angiogenesis. Here we demonstrate, using NRP1 ECKO , NRP2 ECKO , and NRP1/NRP2 ECKO mouse models, that maximum inhibition of primary tumor development and angiogenesis is achieved when both endothelial NRP1 and NRP2 are targeted simultaneously. Metastasis and secondary site angiogenesis were also significantly inhibited in NRP1/NRP2 ECKO animals. Mechanistic studies revealed that codepleting NRP1 and NRP2 in mouse-microvascular endothelial cells stimulates rapid shuttling of VEGFR-2 to Rab7+ endosomes for proteosomal degradation. Our results highlight the importance of targeting both NRP1 and NRP2 to modulate tumor angiogenesis. Significance: The findings presented in this study demonstrate that tumor angiogenesis and growth can be arrested completely by cotargeting endothelial NRP1 and NRP2. We provide new insight into the mechanisms of action regulating NRP-dependent tumor angiogenesis and signpost a novel approach to halt tumor progression.


Asunto(s)
Neoplasias , Neuropilina-1 , Animales , Ratones , Neuropilina-1/genética , Neuropilina-2/genética , Células Endoteliales/metabolismo , Neovascularización Patológica/genética , Neoplasias/genética
4.
FASEB J ; 35(8): e21679, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34314542

RESUMEN

The ability to form a variety of cell-matrix connections is crucial for angiogenesis to take place. Without stable anchorage to the extracellular matrix (ECM), endothelial cells (ECs) are unable to sense, integrate and disseminate growth factor stimulated responses that drive growth of a vascular bed. Neuropilin-2 (NRP2) is a widely expressed membrane-bound multifunctional non-tyrosine kinase receptor, which has previously been implicated in influencing cell adhesion and migration by interacting with α5-integrin and regulating adhesion turnover. α5-integrin, and its ECM ligand fibronectin (FN) are both known to be upregulated during the formation of neo-vasculature. Despite being descriptively annotated as a candidate biomarker for aggressive cancer phenotypes, the EC-specific roles for NRP2 during developmental and pathological angiogenesis remain unexplored. The data reported here support a model whereby NRP2 actively promotes EC adhesion and migration by regulating dynamic cytoskeletal remodeling and by stimulating Rab11-dependent recycling of α5-integrin-p-FAK complexes to newly assembling adhesion sites. Furthermore, temporal depletion of EC-NRP2 in vivo impairs primary tumor growth by disrupting vessel formation. We also demonstrate that EC-NRP2 is required for normal postnatal retinal vascular development, specifically by regulating cell-matrix adhesion. Upon loss of endothelial NRP2, vascular outgrowth from the optic nerve during superficial plexus formation is disrupted, likely due to reduced FAK phosphorylation within sprouting tip cells.


Asunto(s)
Actinas/metabolismo , Células Endoteliales , Integrina alfa5/metabolismo , Pulmón/irrigación sanguínea , Neovascularización Patológica/metabolismo , Neuropilina-2/fisiología , Animales , Adhesión Celular , Línea Celular Tumoral , Células Endoteliales/metabolismo , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Front Cell Dev Biol ; 8: 395, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32528960

RESUMEN

Angiogenesis relies on the ability of endothelial cells (ECs) to migrate over the extracellular matrix via integrin receptors to respond to an angiogenic stimulus. Of the two neuropilin (NRP) orthologs to be identified, both have been reported to be expressed on normal blood and lymphatic ECs, and to play roles in the formation of blood and lymphatic vascular networks during angiogenesis. Whilst the role of NRP1 and its interactions with integrins during angiogenesis has been widely studied, the role of NRP2 in ECs is poorly understood. Here we demonstrate that NRP2 promotes Rac-1 mediated EC adhesion and migration over fibronectin (FN) matrices in a mechanistically distinct fashion to NRP1, showing no dependence on ß3 integrin (ITGB3) expression, or VEGF stimulation. Furthermore, we highlight evidence of a regulatory crosstalk between NRP2 and α5 integrin (ITGA5) in ECs, with NRP2 depletion eliciting an upregulation of ITGA5 expression and disruptions in ITGA5 cellular organization. Finally, we propose a mechanism whereby NRP2 promotes ITGA5 recycling in ECs; NRP2 depleted ECs were found to exhibit reduced levels of total ITGA5 subunit recycling compared to wild-type (WT) ECs. Our findings expose NRP2 as a novel angiogenic player by promoting ITGA5-mediated EC adhesion and migration on FN.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...