Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Circ Res ; 127(6): 707-723, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32527198

RESUMEN

RATIONALE: How endothelial cells (ECs) migrate and form an immature vascular plexus has been extensively studied. Yet, mechanisms underlying vascular remodeling remain poorly established. A better understanding of these processes may lead to the design of novel therapeutic strategies complementary to current angiogenesis inhibitors. OBJECTIVE: Starting from our previous observations that PP2A (protein phosphatase 2) regulates the HIF (hypoxia-inducible factor)/PHD-2 (prolyl hydroxylase 2)-constituted oxygen machinery, we hypothesized that this axis could play an important role during blood vessel formation, tissue perfusion, and oxygen restoration. METHODS AND RESULTS: We show that the PP2A regulatory subunit B55α is at the crossroad between vessel pruning and vessel maturation. Blood vessels with high B55α counter cell stress conditions and thrive for stabilization and maturation. When B55α is inhibited, ECs cannot cope with cell stress and undergo apoptosis, leading to massive pruning of nascent blood vessels. Mechanistically, we found that the B55α/PP2A complex restrains PHD-2 activity, promoting EC survival in a HIF-dependent manner, and furthermore dephosphorylates p38, altogether protecting ECs against cell stress occurring, for example, during the onset of blood flow. In tumors, EC-specific B55α deficiency induces pruning of immature-like tumor blood vessels resulting in delayed tumor growth and metastasis, without affecting nonpathological vessels. Consistently, systemic administration of a pan-PP2A inhibitor disrupts vascular network formation and tumor progression in vivo without additional effects on B55α-deficient vessels. CONCLUSIONS: Our data underline a unique role of the B55α/PP2A phosphatase complex in vessel remodeling and suggest the use of PP2A-inhibitors as potent antiangiogenic drugs targeting specifically nascent blood vessels with a mode-of-action complementary to VEGF-R (vascular endothelial growth factor receptor)-targeted therapies. Graphical Abstract: A graphical abstract is available for this article.


Asunto(s)
Apoptosis , Neoplasias de la Mama/enzimología , Carcinoma Pulmonar de Lewis/enzimología , Células Endoteliales/enzimología , Neovascularización Patológica , Proteína Fosfatasa 2/metabolismo , Remodelación Vascular , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Proc Natl Acad Sci U S A ; 114(40): 10725-10730, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923935

RESUMEN

Retinoid X receptor (RXR) regulates several key functions in myeloid cells, including inflammatory responses, phagocytosis, chemokine secretion, and proangiogenic activity. Its importance, however, in tumor-associated myeloid cells is unknown. In this study, we demonstrate that deletion of RXR in myeloid cells enhances lung metastasis formation while not affecting primary tumor growth. We show that RXR deficiency leads to transcriptomic changes in the lung myeloid compartment characterized by increased expression of prometastatic genes, including important determinants of premetastatic niche formation. Accordingly, RXR-deficient myeloid cells are more efficient in promoting cancer cell migration and invasion. Our results suggest that the repressive activity of RXR on prometastatic genes is mediated primarily through direct DNA binding of the receptor along with nuclear receptor corepressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressors and is largely unresponsive to ligand activation. In addition, we found that expression and transcriptional activity of RXRα is down-modulated in peripheral blood mononuclear cells of patients with lung cancer, particularly in advanced and metastatic disease. Overall, our results identify RXR as a regulator in the myeloid cell-assisted metastatic process and establish lipid-sensing nuclear receptors in the microenvironmental regulation of tumor progression.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Células Mieloides/patología , Receptores X Retinoide/fisiología , Transcripción Genética , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Células Cultivadas , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 38(4): 854-869, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29449332

RESUMEN

OBJECTIVE: Endothelial upregulation of adhesion molecules serves to recruit leukocytes to inflammatory sites and appears to be promoted by NOTCH1; however, current models based on interactions between active NOTCH1 and NF-κB components cannot explain the transcriptional selectivity exerted by NOTCH1 in this context. APPROACH AND RESULTS: Observing that Cre/Lox-induced conditional mutations of endothelial Notch modulated inflammation in murine contact hypersensitivity, we found that IL (interleukin)-1ß stimulation induced rapid recruitment of RELA (v-rel avian reticuloendotheliosis viral oncogene homolog A) to genomic sites occupied by NOTCH1-RBPJ (recombination signal-binding protein for immunoglobulin kappa J region) and that NOTCH1 knockdown reduced histone H3K27 acetylation at a subset of NF-κB-directed inflammatory enhancers. CONCLUSIONS: Our findings reveal that NOTCH1 signaling supports the expression of a subset of inflammatory genes at the enhancer level and demonstrate how key signaling pathways converge on chromatin to coordinate the transition to an infla mmatory endothelial phenotype.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Histonas/metabolismo , Inflamación/prevención & control , Interleucina-1beta/farmacología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/metabolismo , Acetilación , Animales , Apendicitis/metabolismo , Apendicitis/patología , Células Cultivadas , Dermatitis por Contacto/genética , Dermatitis por Contacto/metabolismo , Dermatitis por Contacto/patología , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
4.
Development ; 140(14): 3051-61, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23785053

RESUMEN

Blood vessels form a hierarchically organized network of arteries, capillaries and veins, which develops through a series of growth, pruning and maturation processes. In contrast to the rapidly increasing insight into the processes controlling vascular growth and, in particular, endothelial sprouting and proliferation, the conversion of immature vessels into a fully functional, quiescent vasculature remains little understood. Here we used inducible, cell type-specific genetic approaches to show that endothelial Notch signaling is crucial for the remodeling of veins and the perivenous capillary plexus, which occurs after the completion of the initial angiogenic growth phase in the retina of adolescent mice. Mutant vessels showed ectopic proliferation and sprouting, defective recruitment of supporting mural cells, and failed to downregulate the expression of VEGF receptors. Surprisingly, by contrast Notch was dispensable in the endothelium of remodeling postnatal arteries. Taken together, our results identify key processes contributing to vessel remodeling, maturation and the acquisition of a quiescent phenotype in the final stage of developmental angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Receptores Notch/metabolismo , Vasos Retinianos/citología , Transducción de Señal , Animales , Proliferación Celular , Regulación hacia Abajo , Marcación de Gen , Ratones , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Arteria Retiniana/citología , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/metabolismo
5.
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
6.
Cancer Immunol Res ; 9(3): 309-323, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33361087

RESUMEN

IL1ß is a central mediator of inflammation. Secretion of IL1ß typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1ß in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1ß in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain-like protein in the host were dispensable for the release of intratumoral bioactive IL1ß. Inflammasome-independent IL1ß release promoted systemic neutrophil expansion and fostered accumulation of T-cell-suppressive neutrophils in the tumor. Moreover, IL1ß was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1ß allowed intratumoral accumulation of CD8+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1ß-deficient mice, demonstrating a crucial role for CD8+ T-cell-macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1ß through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors.


Asunto(s)
Interleucina-1beta/metabolismo , Neoplasias/inmunología , Neutrófilos/inmunología , Escape del Tumor , Microambiente Tumoral/inmunología , Animales , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Noqueados , Neoplasias/patología , Neutrófilos/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Linfocitos T Citotóxicos/inmunología , Macrófagos Asociados a Tumores/inmunología
7.
EMBO Mol Med ; 12(10): e11210, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32885605

RESUMEN

Glutamine synthetase (GS) generates glutamine from glutamate and controls the release of inflammatory mediators. In macrophages, GS activity, driven by IL10, associates to the acquisition of M2-like functions. Conditional deletion of GS in macrophages inhibits metastasis by boosting the formation of anti-tumor, M1-like, tumor-associated macrophages (TAMs). From this basis, we evaluated the pharmacological potential of GS inhibitors in targeting metastasis, identifying glufosinate as a specific human GS inhibitor. Glufosinate was tested in both cultured macrophages and on mice bearing metastatic lung, skin and breast cancer. We found that glufosinate rewires macrophages toward an M1-like phenotype both at the primary tumor and metastatic site, countering immunosuppression and promoting vessel sprouting. This was also accompanied to a reduction in cancer cell intravasation and extravasation, leading to synchronous and metachronous metastasis growth inhibition, but no effects on primary tumor growth. Glufosinate treatment was well-tolerated, without liver and brain toxicity, nor hematopoietic defects. These results identify GS as a druggable enzyme to rewire macrophage functions and highlight the potential of targeting metabolic checkpoints in macrophages to treat cancer metastasis.


Asunto(s)
Neoplasias de la Mama , Macrófagos , Aminobutiratos , Animales , Femenino , Humanos , Mediadores de Inflamación , Ratones
8.
Cell Metab ; 30(1): 16-18, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269424

RESUMEN

Cancer cells are highly heterogeneous, and their features markedly vary within different areas of the tumor microenvironment. In this issue, Kumar et al. (2019) identified perivascular tumor cells, derived from mouse glioblastoma xenografts, as the fraction that displays the highest mTOR-dependent anabolic metabolism, aggressiveness, and resistance to therapy.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Xenoinjertos , Ratones , Trasplante Heterólogo , Microambiente Tumoral
9.
Cell Metab ; 30(5): 917-936.e10, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31447322

RESUMEN

Among mammary tumor-infiltrating immune cells, the highest expression of podoplanin (PDPN) is found in a subset of tumor-associated macrophages (TAMs). We hereby demonstrate that PDPN is involved in the attachment of this TAM subset to lymphatic endothelial cells (LECs). Mechanistically, the binding of PDPN to LEC-derived galectin 8 (GAL8) in a glycosylation-dependent manner promotes the activation of pro-migratory integrin ß1. When proximal to lymphatics, PDPN-expressing macrophages (PoEMs) stimulate local matrix remodeling and promote vessel growth and lymphoinvasion. Anti-integrin ß1 blockade, macrophage-specific Pdpn knockout, or GAL8 inhibition impairs TAM adhesion to LECs, restraining lymphangiogenesis and reducing lymphatic cancer spread. In breast cancer patients, association of PoEMs with tumor lymphatic vessels correlates with incidences of lymph node and distant organ metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ganglios Linfáticos/patología , Linfangiogénesis/genética , Metástasis Linfática/genética , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Vasos Linfáticos/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad
10.
Cell Rep ; 21(10): 2842-2854, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29212030

RESUMEN

Although it is well established that tumor-associated macrophages take part in each step of cancer progression, less is known about the distinct role of the so-called metastasis-associated macrophages (MAMs) at the metastatic site. Previous studies reported that Caveolin-1 (Cav1) has both tumor-promoting and tumor-suppressive functions. However, the role of Cav1 in bone-marrow-derived cells is unknown. Here, we describe Cav1 as an anti-metastatic regulator in mouse models of lung and breast cancer pulmonary metastasis. Among all the recruited inflammatory cell populations, we show that MAMs uniquely express abundant levels of Cav1. Using clodronate depletion of macrophages, we demonstrate that macrophage Cav1 signaling is critical for metastasis and not for primary tumor growth. In particular, Cav1 inhibition does not affect MAM recruitment to the metastatic site but, in turn, favors angiogenesis. We describe a mechanism by which Cav1 in MAMs specifically restrains vascular endothelial growth factor A/vascular endothelial growth factor receptor 1 (VEGF-A/VEGFR1) signaling and its downstream effectors, matrix metallopeptidase 9 (MMP9) and colony-stimulating factor 1 (CSF1).


Asunto(s)
Caveolina 1/metabolismo , Macrófagos/metabolismo , Animales , Caveolina 1/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neovascularización Patológica/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Cell Rep ; 18(7): 1699-1712, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199842

RESUMEN

Oxygen-dependent HIF1α hydroxylation and degradation are strictly controlled by PHD2. In hypoxia, HIF1α partly escapes degradation because of low oxygen availability. Here, we show that PHD2 is phosphorylated on serine 125 (S125) by the mechanistic target of rapamycin (mTOR) downstream kinase P70S6K and that this phosphorylation increases its ability to degrade HIF1α. mTOR blockade in hypoxia by REDD1 restrains P70S6K and unleashes PP2A phosphatase activity. Through its regulatory subunit B55α, PP2A directly dephosphorylates PHD2 on S125, resulting in a further reduction of PHD2 activity that ultimately boosts HIF1α accumulation. These events promote autophagy-mediated cell survival in colorectal cancer (CRC) cells. B55α knockdown blocks neoplastic growth of CRC cells in vitro and in vivo in a PHD2-dependent manner. In patients, CRC tissue expresses higher levels of REDD1, B55α, and HIF1α but has lower phospho-S125 PHD2 compared with a healthy colon. Our data disclose a mechanism of PHD2 regulation that involves the mTOR and PP2A pathways and controls tumor growth.


Asunto(s)
Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Neoplasias Colorrectales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Proteína Fosfatasa 2/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/fisiología , Células HEK293 , Células HT29 , Humanos , Fosforilación/fisiología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/fisiología
12.
Nat Commun ; 8(1): 2210, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263363

RESUMEN

VEGFR-2/Notch signalling regulates angiogenesis in part by driving the remodelling of endothelial cell junctions and by inducing cell migration. Here, we show that VEGF-induced polarized cell elongation increases cell perimeter and decreases the relative VE-cadherin concentration at junctions, triggering polarized formation of actin-driven junction-associated intermittent lamellipodia (JAIL) under control of the WASP/WAVE/ARP2/3 complex. JAIL allow formation of new VE-cadherin adhesion sites that are critical for cell migration and monolayer integrity. Whereas at the leading edge of the cell, large JAIL drive cell migration with supportive contraction, lateral junctions show small JAIL that allow relative cell movement. VEGFR-2 activation initiates cell elongation through dephosphorylation of junctional myosin light chain II, which leads to a local loss of tension to induce JAIL-mediated junctional remodelling. These events require both microtubules and polarized Rac activity. Together, we propose a model where polarized JAIL formation drives directed cell migration and junctional remodelling during sprouting angiogenesis.


Asunto(s)
Actinas/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína 2 Relacionada con la Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Proteína 3 Relacionada con la Actina/metabolismo , Actinas/efectos de los fármacos , Antígenos CD/efectos de los fármacos , Cadherinas/efectos de los fármacos , Miosinas Cardíacas/metabolismo , Adhesión Celular , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Modelos Cardiovasculares , Cadenas Ligeras de Miosina/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Seudópodos/fisiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Remodelación Vascular , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rac/metabolismo
13.
Nat Cell Biol ; 19(8): 915-927, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28714968

RESUMEN

Endothelial sprouting and proliferation are tightly coordinated processes mediating the formation of new blood vessels during physiological and pathological angiogenesis. Endothelial tip cells lead sprouts and are thought to suppress tip-like behaviour in adjacent stalk endothelial cells by activating Notch. Here, we show with genetic experiments in postnatal mice that the level of active Notch signalling is more important than the direct Dll4-mediated cell-cell communication between endothelial cells. We identify endothelial expression of VEGF-A and of the chemokine receptor CXCR4 as key processes controlling Notch-dependent vessel growth. Surprisingly, genetic experiments targeting endothelial tip cells in vivo reveal that they retain their function without Dll4 and are also not replaced by adjacent, Dll4-positive cells. Instead, activation of Notch directs tip-derived endothelial cells into developing arteries and thereby establishes that Dll4-Notch signalling couples sprouting angiogenesis and artery formation.


Asunto(s)
Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Receptor Notch1/metabolismo , Arteria Retiniana/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Genotipo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptor Notch1/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Arteria Retiniana/citología , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Trends Mol Med ; 22(2): 85-87, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26774932

RESUMEN

Targeting the aberrant, nonfunctional tumor vasculature is one of the most promising approaches of new anticancer therapies. Here, we discuss a recent publication and a novel mechanism by which tumor-associated macrophages can actively induce vessel normalization and junction stability, and improve the distribution of therapeutic drugs into a tumor.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Humanos
15.
Nat Commun ; 6: 6429, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25752958

RESUMEN

Angiogenic blood vessel growth requires several distinct but integrated cellular activities. Endothelial cell sprouting and proliferation lead to the expansion of the vasculature and give rise to a highly branched, immature plexus, which is subsequently reorganized into a mature and stable network. Although it is known that integrin-mediated cell-matrix interactions are indispensable for embryonic angiogenesis, little is known about the function of integrins in different steps of vascular morphogenesis. Here, by investigating the integrin ß1-subunit with inducible and endothelial-specific gene targeting in the postnatal mouse retina, we show that ß1 integrin promotes endothelial sprouting but is a negative regulator of proliferation. In maturing vessels, integrin ß1 is indispensable for proper localization of VE-cadherin and thereby cell-cell junction integrity. The sum of our findings establishes that integrin ß1 has critical functions in the growing and maturing vasculature, and is required for the formation of stable, non-leaky blood vessels.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio/crecimiento & desarrollo , Integrina beta1/metabolismo , Uniones Intercelulares/fisiología , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Vasos Retinianos/crecimiento & desarrollo , Animales , Western Blotting , Encéfalo/anatomía & histología , Proliferación Celular , Endotelio/metabolismo , Marcación de Gen , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Inmunoprecipitación , Uniones Intercelulares/metabolismo , Ratones , Microscopía Electrónica , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Vasos Retinianos/ultraestructura
16.
J Exp Med ; 211(2): 281-95, 2014 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-24446488

RESUMEN

Cytochrome P450 (CYP) epoxygenases generate bioactive lipid epoxides which can be further metabolized to supposedly less active diols by the soluble epoxide hydrolase (sEH). As the role of epoxides and diols in angiogenesis is unclear, we compared retinal vasculature development in wild-type and sEH(-/-) mice. Deletion of the sEH significantly delayed angiogenesis, tip cell, and filopodia formation, a phenomenon associated with activation of the Notch signaling pathway. In the retina, sEH was localized in Müller glia cells, and Müller cell-specific sEH deletion reproduced the sEH(-/-) retinal phenotype. Lipid profiling revealed that sEH deletion decreased retinal and Müller cell levels of 19,20-dihydroxydocosapentaenoic acid (DHDP), a diol of docosahexenoic acid (DHA). 19,20-DHDP suppressed endothelial Notch signaling in vitro via inhibition of the γ-secretase and the redistribution of presenilin 1 from lipid rafts. Moreover, 19,20-DHDP, but not the parent epoxide, was able to rescue the defective angiogenesis in sEH(-/-) mice as well as in animals lacking the Fbxw7 ubiquitin ligase, which demonstrate strong basal activity of the Notch signaling cascade. These studies demonstrate that retinal angiogenesis is regulated by a novel form of neuroretina-vascular interaction involving the sEH-dependent generation of a diol of DHA in Müller cells.


Asunto(s)
Células Ependimogliales/metabolismo , Epóxido Hidrolasas/metabolismo , Ácidos Grasos Insaturados/biosíntesis , Neovascularización Fisiológica , Receptores Notch/metabolismo , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Epóxido Hidrolasas/deficiencia , Epóxido Hidrolasas/genética , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vasos Retinianos/citología , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
17.
PLoS One ; 7(7): e41116, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848434

RESUMEN

Notch signaling controls fundamental aspects of angiogenic blood vessel growth including the selection of sprouting tip cells, endothelial proliferation and arterial differentiation. The E3 ubiquitin ligase Fbxw7 is part of the SCF protein complex responsible for the polyubiquitination and thereby proteasomal degradation of substrates such as Notch, c-Myc and c-Jun. Here, we show that Fbxw7 is a critical regulator of angiogenesis in the mouse retina and the zebrafish embryonic trunk, which we attribute to its role in the degradation of active Notch. Growth of retinal blood vessel was impaired and the Notch ligand Dll4, which is also a Notch target, upregulated in inducible and endothelial cell-specific Fbxw7(iECKO) mutant mice. The stability of the cleaved and active Notch intracellular domain was increased after siRNA knockdown of the E3 ligase in cultured human endothelial cells. Injection of fbxw7 morpholinos interfered with the sprouting of zebrafish intersegmental vessels (ISVs). Arguing strongly that Notch and not other Fbxw7 substrates are primarily responsible for these phenotypes, the genetic inactivation of Notch pathway components reversed the impaired ISV growth in the zebrafish embryo as well as sprouting and proliferation in the mouse retina. Our findings establish that Fbxw7 is a potent positive regulator of angiogenesis that limits the activity of Notch in the endothelium of the growing vasculature.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/fisiología , Receptores Notch/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones , Ratones Transgénicos , Morfolinos/farmacología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Notch/genética , Retina/citología , Retina/metabolismo , Complejos de Ubiquitina-Proteína Ligasa , Ubiquitina-Proteína Ligasas/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA