Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Life Sci Alliance ; 6(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36410791

RESUMEN

Modulation of integrin function is required in many physiological and pathological settings, such as angiogenesis and cancer. Integrin allosteric changes, clustering, and trafficking cooperate to regulate cell adhesion and motility on extracellular matrix proteins via mechanisms that are partly defined. By exploiting four monoclonal antibodies recognizing distinct conformational epitopes, we show that in endothelial cells (ECs), the extracellular ßI domain, but not the hybrid or I-EGF2 domain of active ß1 integrins, promotes their FAK-regulated clustering into tensin 1-containing fibrillar adhesions and impairs their endocytosis. In this regard, the ßI domain-dependent clustering of active ß1 integrins is necessary to favor fibronectin-elicited directional EC motility, which cannot be effectively promoted by ß1 integrin conformational activation alone.


Asunto(s)
Células Endoteliales , Integrina beta1 , Integrina beta1/metabolismo , Células Endoteliales/metabolismo , Adhesión Celular/fisiología , Integrinas , Análisis por Conglomerados
3.
Nat Commun ; 13(1): 5191, 2022 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-36057632

RESUMEN

Epithelial-mesenchymal transition (EMT) is a complex and pivotal process involved in organogenesis and is related to several pathological processes, including cancer and fibrosis. During heart development, EMT mediates the conversion of epicardial cells into vascular smooth muscle cells and cardiac interstitial fibroblasts. Here, we show that the oncogenic transcription factor EB (TFEB) is a key regulator of EMT in epicardial cells and that its genetic overexpression in mouse epicardium is lethal due to heart defects linked to impaired EMT. TFEB specifically orchestrates the EMT-promoting function of transforming growth factor (TGF) ß, and this effect results from activated transcription of thymine-guanine-interacting factor (TGIF)1, a TGFß/Smad pathway repressor. The Tgif1 promoter is activated by TFEB, and in vitro and in vivo findings demonstrate its increased expression when Tfeb is overexpressed. Furthermore, Tfeb overexpression in vitro prevents TGFß-induced EMT, and this effect is abolished by Tgif1 silencing. Tfeb loss of function, similar to that of Tgif1, sensitizes cells to TGFß, inducing an EMT response to low doses of TGFß. Together, our findings reveal an unexpected function of TFEB in regulating EMT, which might provide insights into injured heart repair and control of cancer progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Factor de Crecimiento Transformador beta , Animales , Células Cultivadas , Transición Epitelial-Mesenquimal/fisiología , Ratones , Organogénesis , Pericardio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
J Neurosci ; 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35953295

RESUMEN

The N-Methyl-D-aspartate receptors (NMDAR) are key players in both physiological and pathological synaptic plasticity because of their involvement in many aspects of neuronal transmission as well as learning and memory. The contribution in these events of different types of GluN2A-interacting proteins is still unclear. The p140Cap scaffold protein acts as a hub for postsynaptic complexes relevant to psychiatric and neurological disorders and regulates synaptic functions like the stabilization of mature dendritic spine, memory consolidation, long-term potentiation, and depression. Here we demonstrate that p140Cap directly binds the GluN2A subunit of NMDAR and modulates GluN2A-associated molecular network. Indeed, in p140Cap knockout male mice, GluN2A is less associated with PSD95 both in ex vivo synaptosomes and in cultured hippocampal neurons and p140Cap expression in knockout neurons can rescue GluN2A and PSD95 colocalization. p140Cap is crucial in the recruitment of GluN2A-containing NMDARs and, consequently, in regulating NMDARs intrinsic properties. p140Cap is associated to synaptic lipid-raft (LR) and to soluble postsynaptic membranes and GluN2A and PSD95 are less recruited into synaptic LR of p140Cap knockout male mice. g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in LR in an activity-dependent fashion. In the synaptic compartment p140Cap influences the association between GluN2A and PSD95 and modulates GluN2A enrichment into LR. Overall, such increase in these membrane domains rich in signalling molecules results in improved signal transduction efficiency.SIGNIFICANT STATEMENTHere we originally show that the adaptor protein p140Cap directly binds the GluN2A subunit of NMDAR and modulates the GluN2A-associated molecular network. Moreover, we show for the first time that p140Cap also associates to synaptic lipid rafts and controls the selective recruitment of GluN2A and PSD95 to this specific compartment. Finally, g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in lipid rafts in an activity-dependent fashion. Overall, our findings provide the molecular and functional dissection of p140Cap as a new active member of a highly dynamic synaptic network involved in memory consolidation, LTP and LTD that are known to be altered in neurological and psychiatric disorders.

5.
Matrix Biol ; 112: 62-71, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35961423

RESUMEN

In metazoans, cell adhesion to the extracellular matrix (ECM) drives the development, functioning, and repair of different tissues, organs, and systems. Disruption or dysregulation of cell-to-ECM adhesion promote the initiation and progression of several diseases, such as bleeding, immune disorders and cancer. Integrins are major ECM transmembrane receptors, whose function depends on both allosteric changes and exo-endocytic traffic, which carries them to and from the plasma membrane. In apico-basally polarized cells, asymmetric adhesion to the ECM is maintained by continuous targeting of the plasma membrane by vesicles coming from the trans Golgi network and carrying ECM proteins. Active integrin-bound ECM is indeed endocytosed and replaced by the exocytosis of fresh ECM. Such vesicular traffic is finely driven by the teamwork of microtubules (MTs) and their associated kinesin and dynein motors. Here, we review the main cytoskeletal actors involved in the control of the spatiotemporal distribution of active integrins and their ECM ligands, highlighting the key role of the synchronous (ant)agonistic cooperation between MT motors transporting vesicular cargoes, in the same or in opposite direction, in the regulation of traffic logistics, and the establishment of epithelial and endothelial cell polarity.


Asunto(s)
Dineínas , Cinesinas , Adhesión Celular , Dineínas/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Integrinas/genética , Integrinas/metabolismo , Ligandos
6.
Nat Commun ; 13(1): 4188, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35858913

RESUMEN

The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.


Asunto(s)
Neuropilina-1 , Triptófano-ARNt Ligasa , Uniones Adherentes/metabolismo , Animales , Antígenos CD , Cadherinas/genética , Permeabilidad Capilar , Células Endoteliales/metabolismo , Histamina , Ligandos , Ratones , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteómica , Triptófano-ARNt Ligasa/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Angiogenesis ; 25(4): 471-492, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35545719

RESUMEN

The dynamic integrin-mediated adhesion of endothelial cells (ECs) to the surrounding ECM is fundamental for angiogenesis both in physiological and pathological conditions, such as embryonic development and cancer progression. The dynamics of EC-to-ECM adhesions relies on the regulation of the conformational activation and trafficking of integrins. Here, we reveal that oncogenic transcription factor EB (TFEB), a known regulator of lysosomal biogenesis and metabolism, also controls a transcriptional program that influences the turnover of ECM adhesions in ECs by regulating cholesterol metabolism. We show that TFEB favors ECM adhesion turnover by promoting the transcription of genes that drive the synthesis of cholesterol, which promotes the aggregation of caveolin-1, and the caveolin-dependent endocytosis of integrin ß1. These findings suggest that TFEB might represent a novel target for the pharmacological control of pathological angiogenesis and bring new insights in the mechanism sustaining TFEB control of endocytosis.


Asunto(s)
Células Endoteliales , Integrinas , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Caveolina 1/metabolismo , Adhesión Celular/genética , Colesterol , Células Endoteliales/metabolismo , Humanos , Integrina beta1/metabolismo , Integrinas/metabolismo , Neovascularización Patológica/metabolismo
8.
J Cell Biol ; 220(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34581723

RESUMEN

Dynamic modulation of endothelial cell-to-cell and cell-to-extracellular matrix (ECM) adhesion is essential for blood vessel patterning and functioning. Yet the molecular mechanisms involved in this process have not been completely deciphered. We identify the adhesion G protein-coupled receptor (ADGR) Latrophilin 2 (LPHN2) as a novel determinant of endothelial cell (EC) adhesion and barrier function. In cultured ECs, endogenous LPHN2 localizes at ECM contacts, signals through cAMP/Rap1, and inhibits focal adhesion (FA) formation and nuclear localization of YAP/TAZ transcriptional regulators, while promoting tight junction (TJ) assembly. ECs also express an endogenous LPHN2 ligand, fibronectin leucine-rich transmembrane 2 (FLRT2), that prevents ECM-elicited EC behaviors in an LPHN2-dependent manner. Vascular ECs of lphn2a knock-out zebrafish embryos become abnormally stretched, display a hyperactive YAP/TAZ pathway, and lack proper intercellular TJs. Consistently, blood vessels are hyperpermeable, and intravascularly injected cancer cells extravasate more easily in lphn2a null animals. Thus, LPHN2 ligands, such as FLRT2, may be therapeutically exploited to interfere with cancer metastatic dissemination.


Asunto(s)
Permeabilidad Capilar/fisiología , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Modificados Genéticamente , Células COS , Línea Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Transducción de Señal/fisiología , Transactivadores/metabolismo , Pez Cebra
9.
Fac Rev ; 10: 45, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34131655

RESUMEN

Integrin-mediated adhesion of cells to the extracellular matrix (ECM) is crucial for the physiological development and functioning of tissues but is pathologically disrupted in cancer. Indeed, abnormal regulation of integrin receptors and ECM ligands allows cancer cells to break down tissue borders, breach into blood and lymphatic vessels, and survive traveling in suspension through body fluids or residing in metabolically or pharmacologically hostile environments. Different molecular and cellular mechanisms responsible for the modulation of integrin adhesive function or mechanochemical signaling are altered and participate in cancer. Cancer development and progression are also bolstered by dysfunctionalities of integrin-mediated ECM adhesion occurring both in tumor cells and in elements of the surrounding tumor microenvironment, such as vascular cells, cancer-associated fibroblasts, and immune cells. Mounting evidence suggests that integrin inhibitors may be effectively exploited to overcome resistance to standard-of-care anti-cancer therapies.

10.
Cancer Lett ; 510: 13-23, 2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-33862151

RESUMEN

An interactive crosstalk between tumor and stroma cells is essential for metastatic melanoma progression. We evidenced that ESDN/DCBLD2/CLCP1 plays a crucial role in endothelial cells during the spread of melanoma. Precisely, increased extravasation and metastasis formation were revealed in ESDN-null mice injected with melanoma cells, even if the primary tumor growth, vessel permeability, and angiogenesis were not enhanced. Interestingly, improved adhesion of melanoma cells to ESDN-depleted endothelial cells was observed, due to the presence of higher levels of E-selectin transcripts/proteins in ESDN-defective cells. In accordance with these results, anticorrelation was observed between ESDN and E-selectin in human endothelial cells. Most importantly, our data revealed that cimetidine, an E-selectin inhibitor, was able to block cell adhesion, extravasation, and metastasis formation in ESDN-null mice, underlying a major role of ESDN in E-selectin transcription upregulation, which according to our data, may presumably be linked to STAT3. Based on our results, we propose a protective role for ESDN during the spread of melanoma and reveal its therapeutic potential.


Asunto(s)
Selectina E/antagonistas & inhibidores , Células Endoteliales/metabolismo , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Selectina E/biosíntesis , Selectina E/metabolismo , Humanos , Melanoma/genética , Melanoma/patología , Ratones , Microambiente Tumoral
11.
Phys Rev Lett ; 126(8): 088101, 2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33709726

RESUMEN

We introduce a simple physical picture to explain the process of molecular sorting, whereby specific proteins are concentrated and distilled into submicrometric lipid vesicles in eukaryotic cells. To this purpose, we formulate a model based on the coupling of spontaneous molecular aggregation with vesicle nucleation. Its implications are studied by means of a phenomenological theory describing the diffusion of molecules toward multiple sorting centers that grow due to molecule absorption and are extracted when they reach a sufficiently large size. The predictions of the theory are compared with numerical simulations of a lattice-gas realization of the model and with experimental observations. The efficiency of the distillation process is found to be optimal for intermediate aggregation rates, where the density of sorted molecules is minimal and the process obeys simple scaling laws. Quantitative measures of endocytic sorting performed in primary endothelial cells are compatible with the hypothesis that these optimal conditions are realized in living cells.


Asunto(s)
Células Eucariotas/metabolismo , Lípidos de la Membrana/metabolismo , Modelos Biológicos , Proteínas/metabolismo , Difusión , Vesículas Transportadoras/metabolismo
12.
Methods Mol Biol ; 2217: 301-311, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33215388

RESUMEN

In endothelial cells (ECs), the onset of apicobasal polarity is primarily regulated by the interaction of integrins with the surrounding extracellular matrix (ECM). ECs secrete and polymerize fibronectin (FN), a unique, permissive substrate that allows for vascular morphogenesis and lumen formation. We previously identified a signaling pathway that, under the control of the adhesion site adaptor protein PPFIA1, integrates the polarized secretion of freshly synthesized FN with the recycling of conformationally active α5ß1 integrin, the main FN receptor in ECs. To characterize the functional role of PPFIA1-dependent signaling in ECs, we set up a Transwell-based assay to quantify the polarized secretion of ECM proteins. To this aim, we allowed ECs to form a confluent monolayer on the Transwell membrane and checked its integrity by measuring transendothelial electric resistance and controlling the stability of tight junctions over time by fluorescent confocal microscope analysis. Finally, we quantified apical and basolateral FN secretion in control and PPFIA1-silenced EC culture medium by western blot analysis coupled to spike-in normalization.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/genética , Integrina alfa5beta1/genética , Uniones Estrechas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transporte Biológico , Polaridad Celular , Cámaras de Difusión de Cultivos , Células Endoteliales/ultraestructura , Matriz Extracelular/ultraestructura , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Integrina alfa5beta1/metabolismo , Microscopía Fluorescente/métodos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Uniones Estrechas/ultraestructura
13.
Curr Biol ; 30(11): R652-R654, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32516616

RESUMEN

In endothelial cells, trafficking of active α5ß1 integrins and polarized fibronectin secretion are important for vascular morphogenesis. A new study unveils how the endothelial small GTPase RHOJ, by repressing trafficking of active α5ß1 integrins, controls fibronectin polymerization and in vivo angiogenesis.


Asunto(s)
Fibronectinas , Proteínas de Unión al GTP Monoméricas , Células Endoteliales/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Morfogénesis
14.
EMBO J ; 39(6): e102513, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32073171

RESUMEN

Various species of non-coding RNAs (ncRNAs) are enriched in specific subcellular compartments, but the mechanisms orchestrating their localization and their local functions remain largely unknown. We investigated both aspects using the elongating retinal ganglion cell axon and its tip, the growth cone, as models. We reveal that specific endogenous precursor microRNAs (pre-miRNAs) are actively trafficked to distal axons by hitchhiking primarily on late endosomes/lysosomes. Upon exposure to the axon guidance cue semaphorin 3A (Sema3A), pre-miRNAs are processed specifically within axons into newly generated miRNAs, one of which, in turn, silences the basal translation of tubulin beta 3 class III (TUBB3), but not amyloid beta precursor protein (APP). At the organismal level, these mature miRNAs are required for growth cone steering and a fully functional visual system. Overall, our results uncover a novel mode of ncRNA transport from one cytosolic compartment to another within polarized cells. They also reveal that newly generated miRNAs are critical components of a ncRNA-based signaling pathway that transduces environmental signals into the structural remodeling of subcellular compartments.


Asunto(s)
MicroARNs/genética , ARN no Traducido/genética , Transducción de Señal , Animales , Axones/fisiología , Transporte Biológico , Endosomas/metabolismo , Femenino , Conos de Crecimiento/fisiología , Ratones Endogámicos C57BL , Precursores del ARN/genética , Células Ganglionares de la Retina/fisiología , Xenopus laevis
15.
Biochem Soc Trans ; 48(1): 83-93, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32065228

RESUMEN

Spatiotemporal control of integrin-mediated cell adhesion to the extracellular matrix (ECM) is critical for physiological and pathological events in multicellular organisms, such as embryonic development, angiogenesis, platelet aggregation, leukocytes extravasation, and cancer cell metastatic dissemination. Regulation of integrin adhesive function and signaling relies on the modulation of both conformation and traffic. Indeed, integrins exist in a dynamic equilibrium between a bent/closed (inactive) and an extended/open (active) conformation, respectively endowed with low and high affinity for ECM ligands. Increasing evidence proves that, differently to what hypothesized in the past, detachment from the ECM and conformational inactivation are not mandatory for integrin to get endocytosed and trafficked. Specific transmembrane and cytosolic proteins involved in the control of ECM proteolytic fragment-bound active integrin internalization and recycling exist. In the complex masterplan that governs cell behavior, active integrin traffic is key to the turnover of ECM polymers and adhesion sites, the polarized secretion of endogenous ECM proteins and modifying enzymes, the propagation of motility and survival endosomal signals, and the control of cell metabolism.


Asunto(s)
Endocitosis/fisiología , Integrinas/química , Integrinas/metabolismo , Transporte de Proteínas/fisiología , Animales , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Movimiento Celular , Endosomas/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Ligandos , Conformación Proteica
16.
Int J Mol Sci ; 20(23)2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31783481

RESUMEN

Cadherins are a major family of cell-cell adhesive receptors, which are implicated in development, tissue homeostasis, and cancer. Here, we show a novel mechanism of post-translational regulation of E-cadherin in cancer cells by an intramembrane protease of the Rhomboid family, RHBDL2, which leads to the shedding of E-cadherin extracellular domain. In addition, our data indicate that RHBDL2 mediates a similar activity on VE-cadherin, which is selectively expressed by endothelial cells. We show that RHBDL2 promotes cell migration, which is consistent with its ability to interfere with the functional role of cadherins as negative regulators of motility; moreover, the two players appear to lie in the same functional pathway. Importantly, we show that RHBDL2 expression is induced by the inflammatory chemokine TNFα. The E-cadherin extracellular domain is known to be released by metalloproteases (MMPs); however, here, we provide evidence of a novel MMP-independent, TNFα inducible, E-cadherin processing mechanism that is mediated by RHBDL2. Thus, the intramembrane protease RHBDL2 is a novel regulator of cadherins promoting cell motility.


Asunto(s)
Cadherinas/metabolismo , Metaloproteasas/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Quimiocinas/metabolismo , Chlorocebus aethiops , Perros , Células HEK293 , Humanos , Inflamación/metabolismo , Células de Riñón Canino Madin Darby , Células PC-3 , Serina Proteasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Sci Transl Med ; 10(442)2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29794061

RESUMEN

Vascular normalizing strategies, aimed at ameliorating blood vessel perfusion and lessening tissue hypoxia, are treatments that may improve the outcome of cancer patients. Secreted class 3 semaphorins (SEMA3), which are thought to directly bind neuropilin (NRP) co-receptors that, in turn, associate with and elicit plexin (PLXN) receptor signaling, are effective normalizing agents of the cancer vasculature. Yet, SEMA3A was also reported to trigger adverse side effects via NRP1. We rationally designed and generated a safe, parenterally deliverable, and NRP1-independent SEMA3A point mutant isoform that, unlike its wild-type counterpart, binds PLXNA4 with nanomolar affinity and has much greater biochemical and biological activities in cultured endothelial cells. In vivo, when parenterally administered in mouse models of pancreatic cancer, the NRP1-independent SEMA3A point mutant successfully normalized the vasculature, inhibited tumor growth, curbed metastatic dissemination, and effectively improved the supply and anticancer activity of chemotherapy. Mutant SEMA3A also inhibited retinal neovascularization in a mouse model of age-related macular degeneration. In summary, mutant SEMA3A is a vascular normalizing agent that can be exploited to treat cancer and, potentially, other diseases characterized by pathological angiogenesis.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Proteínas Mutantes/metabolismo , Neuropilina-1/metabolismo , Semaforina-3A/agonistas , Animales , Antineoplásicos/uso terapéutico , Permeabilidad Capilar/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/patología , Simulación por Computador , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Ratones Transgénicos , Proteínas Mutantes/química , Neoplasias/irrigación sanguínea , Neoplasias/patología , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/efectos de los fármacos , Semaforina-3A/química
18.
Arterioscler Thromb Vasc Biol ; 37(9): 1710-1721, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28729362

RESUMEN

OBJECTIVE: Molecular pathways governing blood vessel patterning are vital to vertebrate development. Because of their ability to counteract proangiogenic factors, antiangiogenic secreted Sema3 (class 3 semaphorins) control embryonic vascular morphogenesis. However, if and how Sema3 may play a role in the control of extraembryonic vascular development is presently unknown. APPROACH AND RESULTS: By characterizing genetically modified mice, here, we show that surprisingly Sema3F acts instead as a selective extraembryonic, but not intraembryonic proangiogenic cue. Both in vivo and in vitro, in visceral yolk sac epithelial cells, Sema3F signals to inhibit the phosphorylation-dependent degradation of Myc, a transcription factor that drives the expression of proangiogenic genes, such as the microRNA cluster 17/92. In Sema3f-null yolk sacs, the transcription of Myc-regulated microRNA 17/92 cluster members is impaired, and the synthesis of Myc and microRNA 17/92 foremost antiangiogenic target Thbs1 (thrombospondin 1) is increased, whereas Vegf (vascular endothelial growth factor) signaling is inhibited in yolk sac endothelial cells. Consistently, exogenous recombinant Sema3F inhibits the phosphorylation-dependent degradation of Myc and the synthesis of Thbs1 in mouse F9 teratocarcinoma stem cells that were in vitro differentiated in visceral yolk sac epithelial cells. Sema3f-/- mice placentas are also highly anemic and abnormally vascularized. CONCLUSIONS: Sema3F functions as an unconventional Sema3 that promotes extraembryonic angiogenesis by inhibiting the Myc-regulated synthesis of Thbs1 in visceral yolk sac epithelial cells.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Placenta/irrigación sanguínea , Saco Vitelino/irrigación sanguínea , Animales , Línea Celular Tumoral , Células Madre de Carcinoma Embrionario/metabolismo , Células Endoteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Fosforilación , Embarazo , Proteolisis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Cell Biol ; 216(7): 2107-2130, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28550110

RESUMEN

Endothelial cell adhesion and migration are critical steps of the angiogenic process, whose dysfunction is associated with tumor growth and metastasis. The TRPM8 channel has recently been proposed to play a protective role in prostate cancer by impairing cell motility. However, the mechanisms by which it could influence vascular behavior are unknown. Here, we reveal a novel non-channel function for TRPM8 that unexpectedly acts as a Rap1 GTPase inhibitor, thereby inhibiting endothelial cell motility, independently of pore function. TRPM8 retains Rap1 intracellularly through direct protein-protein interaction, thus preventing its cytoplasm-plasma membrane trafficking. In turn, this mechanism impairs the activation of a major inside-out signaling pathway that triggers the conformational activation of integrin and, consequently, cell adhesion, migration, in vitro endothelial tube formation, and spheroid sprouting. Our results bring to light a novel, pore-independent molecular mechanism by which endogenous TRPM8 expression inhibits Rap1 GTPase and thus plays a critical role in the behavior of vascular endothelial cells by inhibiting migration.


Asunto(s)
Movimiento Celular , Células Endoteliales/enzimología , Neovascularización Fisiológica , Canales Catiónicos TRPM/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Adhesión Celular , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Integrina beta1/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Microscopía por Video , Modelos Cardiovasculares , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Transducción de Señal , Canales Catiónicos TRPM/genética , Factores de Tiempo , Transfección , Proteínas de Unión al GTP rap1/genética
20.
Nat Commun ; 7: 13546, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876801

RESUMEN

Basolateral polymerization of cellular fibronectin (FN) into a meshwork drives endothelial cell (EC) polarity and vascular remodelling. However, mechanisms coordinating α5ß1 integrin-mediated extracellular FN endocytosis and exocytosis of newly synthesized FN remain elusive. Here we show that, on Rab21-elicited internalization, FN-bound/active α5ß1 is recycled to the EC surface. We identify a pathway, comprising the regulators of post-Golgi carrier formation PI4KB and AP-1A, the small GTPase Rab11B, the surface tyrosine phosphatase receptor PTPRF and its adaptor PPFIA1, which we propose acts as a funnel combining FN secretion and recycling of active α5ß1 integrin from the trans-Golgi network (TGN) to the EC surface, thus allowing FN fibrillogenesis. In this framework, PPFIA1 interacts with active α5ß1 integrin and localizes close to EC adhesions where post-Golgi carriers are targeted. We show that PPFIA1 is required for FN polymerization-dependent vascular morphogenesis, both in vitro and in the developing zebrafish embryo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fibronectinas/metabolismo , Regulación de la Expresión Génica/fisiología , Integrina alfa5beta1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Embrión no Mamífero , Fibronectinas/genética , Aparato de Golgi/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa5beta1/genética , Neovascularización Fisiológica/fisiología , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA