Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nature ; 492(7428): 252-5, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23143332

RESUMEN

The innate immune response is essential for combating infectious disease. Macrophages and other cells respond to infection by releasing cytokines, such as interleukin-1ß (IL-1ß), which in turn activate a well-described, myeloid-differentiation factor 88 (MYD88)-mediated, nuclear factor-κB (NF-κB)-dependent transcriptional pathway that results in inflammatory-cell activation and recruitment. Endothelial cells, which usually serve as a barrier to the movement of inflammatory cells out of the blood and into tissue, are also critical mediators of the inflammatory response. Paradoxically, the cytokines vital to a successful immune defence also have disruptive effects on endothelial cell-cell interactions and can trigger degradation of barrier function and dissociation of tissue architecture. The mechanism of this barrier dissolution and its relationship to the canonical NF-κB pathway remain poorly defined. Here we show that the direct, immediate and disruptive effects of IL-1ß on endothelial stability in a human in vitro cell model are NF-κB independent and are instead the result of signalling through the small GTPase ADP-ribosylation factor 6 (ARF6) and its activator ARF nucleotide binding site opener (ARNO; also known as CYTH2). Moreover, we show that ARNO binds directly to the adaptor protein MYD88, and thus propose MYD88-ARNO-ARF6 as a proximal IL-1ß signalling pathway distinct from that mediated by NF-κB. Finally, we show that SecinH3, an inhibitor of ARF guanine nucleotide-exchange factors such as ARNO, enhances vascular stability and significantly improves outcomes in animal models of inflammatory arthritis and acute inflammation.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina/metabolismo , Factor 6 de Ribosilación del ADP , Adyuvantes Inmunológicos/farmacología , Animales , Artritis/patología , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Purinas/farmacología , Transducción de Señal , Tiofenos/farmacología
2.
Proc Natl Acad Sci U S A ; 107(23): 10520-5, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20498081

RESUMEN

Formation of the vascular system within organs requires the balanced action of numerous positive and negative factors secreted by stromal and epithelial cells. Here, we used a genetic approach to determine the role of SLITs in regulating the growth and organization of blood vessels in the mammary gland. We demonstrate that vascularization of the gland is not affected by loss of Slit expression in the epithelial compartment. Instead, we identify a stromal source of SLIT, mural cells encircling blood vessels, and show that loss of Slit in the stroma leads to elevated blood vessel density and complexity. We examine candidate SLIT receptors, Robo1 and Robo4, and find that increased vessel angiogenesis is phenocopied by loss of endothelial-specific Robo4, as long as it is combined with the presence of an angiogenic stimulus such as preneoplasia or pregnancy. In contrast, loss of Robo1 does not affect blood vessel growth. The enhanced growth of blood vessels in Robo4(-/-) endothelium is due to activation of vascular endothelial growth factor (VEGF)-R2 signaling through the Src and FAK kinases. Thus, our studies present a genetic dissection of SLIT/ROBO signaling during organ development. We identify a stromal, rather than epithelial, source of SLITs that inhibits blood vessel growth by signaling through endothelial ROBO4 to down-regulate VEGF/VEGFR2 signaling.


Asunto(s)
Glándulas Mamarias Animales/irrigación sanguínea , Glándulas Mamarias Animales/metabolismo , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Receptores de Superficie Celular , Receptores Inmunológicos/deficiencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Roundabout
3.
Oncogene ; 42(35): 2629-2640, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37500798

RESUMEN

Preventing or effectively treating metastatic uveal melanoma (UM) is critical because it occurs in about half of patients and confers a very poor prognosis. There is emerging evidence that hepatocyte growth factor (HGF) and insulin-like growth factor 1 (IGF-1) promote metastasis and contribute to the striking metastatic hepatotropism observed in UM metastasis. However, the molecular mechanisms by which HGF and IGF-1 promote UM liver metastasis have not been elucidated. ASAP1, which acts as an effector for the small GTPase ARF6, is highly expressed in the subset of uveal melanomas most likely to metastasize. Here, we found that HGF and IGF-1 hyperactivate ARF6, leading to its interaction with ASAP1, which then acts as an effector to induce nuclear localization and transcriptional activity of NFAT1. Inhibition of any component of this pathway impairs cellular invasiveness. Additionally, knocking down ASAP1 or inhibiting NFAT signaling reduces metastasis in a xenograft mouse model of UM. The discovery of this signaling pathway represents not only an advancement in our understanding of the biology of uveal melanoma metastasis but also identifies a novel pathway that could be targeted to treat or prevent metastatic uveal melanoma.


Asunto(s)
Melanoma , Neoplasias de la Úvea , Humanos , Animales , Ratones , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Melanoma/patología , Neoplasias de la Úvea/metabolismo , Modelos Animales de Enfermedad , Proteínas Adaptadoras Transductoras de Señales/metabolismo
4.
Neuron ; 110(19): 3106-3120.e7, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35961320

RESUMEN

Breakdown of the blood-central nervous system barrier (BCNSB) is a hallmark of many neuroinflammatory disorders, such as multiple sclerosis (MS). Using a mouse model of MS, experimental autoimmune encephalomyelitis (EAE), we show that endothelial-to-mesenchymal transition (EndoMT) occurs in the CNS before the onset of clinical symptoms and plays a major role in the breakdown of BCNSB function. EndoMT can be induced by an IL-1ß-stimulated signaling pathway in which activation of the small GTPase ADP ribosylation factor 6 (ARF6) leads to crosstalk with the activin receptor-like kinase (ALK)-SMAD1/5 pathway. Inhibiting the activation of ARF6 both prevents and reverses EndoMT, stabilizes BCNSB function, reduces demyelination, and attenuates symptoms even after the establishment of severe EAE, without immunocompromising the host. Pan-inhibition of ALKs also reduces disease severity in the EAE model. Therefore, multiple components of the IL-1ß-ARF6-ALK-SMAD1/5 pathway could be targeted for the treatment of a variety of neuroinflammatory disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Proteínas de Unión al GTP Monoméricas , Esclerosis Múltiple , Receptores de Activinas/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al GTP Monoméricas/metabolismo , Enfermedades Neuroinflamatorias , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal
5.
PLoS Genet ; 4(5): e1000073, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18483557

RESUMEN

In eutherian mammals, embryonic growth and survival is dependent on the formation of the placenta, an organ that facilitates the efficient exchange of oxygen, nutrients, and metabolic waste between the maternal and fetal blood supplies. Key to the placenta's function is the formation of its vascular labyrinth, a series of finely branched vessels whose molecular ontogeny remains largely undefined. In this report, we demonstrate that HOXA13 plays an essential role in labyrinth vessel formation. In the absence of HOXA13 function, placental endothelial cell morphology is altered, causing a loss in vessel wall integrity, edema of the embryonic blood vessels, and mid-gestational lethality. Microarray analysis of wild-type and mutant placentas revealed significant changes in endothelial gene expression profiles. Notably, pro-vascular genes, including Tie2 and Foxf1, exhibited reduced expression in the mutant endothelia, which also exhibited elevated expression of genes normally expressed in lymphatic or sinusoidal endothelia. ChIP analysis of HOXA13-DNA complexes in the placenta confirmed that HOXA13 binds the Tie2 and Foxf1 promoters in vivo. In vitro, HOXA13 binds sequences present in the Tie2 and Foxf1 promoters with high affinity (K(d) = 27-42 nM) and HOXA13 can use these bound promoter regions to direct gene expression. Taken together, these findings demonstrate that HOXA13 directly regulates Tie2 and Foxf1 in the placental labyrinth endothelia, providing a functional explanation for the mid-gestational lethality exhibited by Hoxa13 mutant embryos as well as a novel transcriptional program necessary for the specification of the labyrinth vascular endothelia.


Asunto(s)
Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Placenta/irrigación sanguínea , Animales , Secuencia de Bases , Endotelio Vascular/embriología , Endotelio Vascular/crecimiento & desarrollo , Factores de Transcripción Forkhead/genética , Genes Reporteros , Proteínas de Homeodominio/genética , Homocigoto , Técnicas In Vitro , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neovascularización Fisiológica , Análisis de Secuencia por Matrices de Oligonucleótidos , Placenta/embriología , Placenta/metabolismo , Regiones Promotoras Genéticas , Receptor TIE-2/genética , Trofoblastos/fisiología
6.
Cancer Res ; 79(11): 2892-2908, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31048499

RESUMEN

Melanoma has an unusual capacity to spread in early-stage disease, prompting aggressive clinical intervention in very thin primary tumors. Despite these proactive efforts, patients with low-risk, low-stage disease can still develop metastasis, indicating the presence of permissive cues for distant spread. Here, we show that constitutive activation of the small GTPase ARF6 (ARF6Q67L) is sufficient to accelerate metastasis in mice with BRAFV600E/Cdkn2aNULL melanoma at a similar incidence and severity to Pten loss, a major driver of PI3K activation and melanoma metastasis. ARF6Q67L promoted spontaneous metastasis from significantly smaller primary tumors than PTENNULL, implying an enhanced ability of ARF6-GTP to drive distant spread. ARF6 activation increased lung colonization from circulating melanoma cells, suggesting that the prometastatic function of ARF6 extends to late steps in metastasis. Unexpectedly, ARF6Q67L tumors showed upregulation of Pik3r1 expression, which encodes the p85 regulatory subunit of PI3K. Tumor cells expressing ARF6Q67L displayed increased PI3K protein levels and activity, enhanced PI3K distribution to cellular protrusions, and increased AKT activation in invadopodia. ARF6 is necessary and sufficient for activation of both PI3K and AKT, and PI3K and AKT are necessary for ARF6-mediated invasion. We provide evidence for aberrant ARF6 activation in human melanoma samples, which is associated with reduced survival. Our work reveals a previously unknown ARF6-PI3K-AKT proinvasive pathway, it demonstrates a critical role for ARF6 in multiple steps of the metastatic cascade, and it illuminates how melanoma cells can acquire an early metastatic phenotype in patients. SIGNIFICANCE: These findings reveal a prometastatic role for ARF6 independent of tumor growth, which may help explain how melanoma spreads distantly from thin, early-stage primary tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2892/F1.large.jpg.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Melanoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Cutáneas/patología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Guanosina Trifosfato/metabolismo , Humanos , Neoplasias Pulmonares/secundario , Melanoma/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Mutantes , Ratones SCID , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/metabolismo
8.
J Clin Invest ; 127(12): 4569-4582, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29058688

RESUMEN

The devastating sequelae of diabetes mellitus include microvascular permeability, which results in retinopathy. Despite clinical and scientific advances, there remains a need for new approaches to treat retinopathy. Here, we have presented a possible treatment strategy, whereby targeting the small GTPase ARF6 alters VEGFR2 trafficking and reverses signs of pathology in 4 animal models that represent features of diabetic retinopathy and in a fifth model of ocular pathological angiogenesis. Specifically, we determined that the same signaling pathway utilizes distinct GEFs to sequentially activate ARF6, and these GEFs exert distinct but complementary effects on VEGFR2 trafficking and signal transduction. ARF6 activation was independently regulated by 2 different ARF GEFs - ARNO and GEP100. Interaction between VEGFR2 and ARNO activated ARF6 and stimulated VEGFR2 internalization, whereas a VEGFR2 interaction with GEP100 activated ARF6 to promote VEGFR2 recycling via coreceptor binding. Intervening in either pathway inhibited VEGFR2 signal output. Finally, using a combination of in vitro, cellular, genetic, and pharmacologic techniques, we demonstrated that ARF6 is pivotal in VEGFR2 trafficking and that targeting ARF6-mediated VEGFR2 trafficking has potential as a therapeutic approach for retinal vascular diseases such as diabetic retinopathy.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Retinopatía Diabética/metabolismo , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Línea Celular , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Transporte de Proteínas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
9.
Nat Cell Biol ; 19(9): 1027-1036, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28812582

RESUMEN

Most differentiated cells convert glucose to pyruvate in the cytosol through glycolysis, followed by pyruvate oxidation in the mitochondria. These processes are linked by the mitochondrial pyruvate carrier (MPC), which is required for efficient mitochondrial pyruvate uptake. In contrast, proliferative cells, including many cancer and stem cells, perform glycolysis robustly but limit fractional mitochondrial pyruvate oxidation. We sought to understand the role this transition from glycolysis to pyruvate oxidation plays in stem cell maintenance and differentiation. Loss of the MPC in Lgr5-EGFP-positive stem cells, or treatment of intestinal organoids with an MPC inhibitor, increases proliferation and expands the stem cell compartment. Similarly, genetic deletion of the MPC in Drosophila intestinal stem cells also increases proliferation, whereas MPC overexpression suppresses stem cell proliferation. These data demonstrate that limiting mitochondrial pyruvate metabolism is necessary and sufficient to maintain the proliferation of intestinal stem cells.


Asunto(s)
Proliferación Celular , Drosophila melanogaster/metabolismo , Glucólisis , Mucosa Intestinal/metabolismo , Mitocondrias/metabolismo , Ácido Pirúvico/metabolismo , Células Madre/metabolismo , Acrilatos/farmacología , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Genotipo , Humanos , Intestinos/citología , Intestinos/efectos de los fármacos , Ácido Láctico/metabolismo , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Transportadores de Ácidos Monocarboxílicos , Fenotipo , Interferencia de ARN , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/efectos de los fármacos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
10.
Cancer Cell ; 29(6): 889-904, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27265506

RESUMEN

Activating mutations in Gαq proteins, which form the α subunit of certain heterotrimeric G proteins, drive uveal melanoma oncogenesis by triggering multiple downstream signaling pathways, including PLC/PKC, Rho/Rac, and YAP. Here we show that the small GTPase ARF6 acts as a proximal node of oncogenic Gαq signaling to induce all of these downstream pathways as well as ß-catenin signaling. ARF6 activates these diverse pathways through a common mechanism: the trafficking of GNAQ and ß-catenin from the plasma membrane to cytoplasmic vesicles and the nucleus, respectively. Blocking ARF6 with a small-molecule inhibitor reduces uveal melanoma cell proliferation and tumorigenesis in a mouse model, confirming the functional relevance of this pathway and suggesting a therapeutic strategy for Gα-mediated diseases.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Melanoma/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Neoplasias de la Úvea/tratamiento farmacológico , beta Catenina/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/antagonistas & inhibidores , Factores de Ribosilacion-ADP/genética , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Citoplasma/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Ratones , Trasplante de Neoplasias , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo
11.
Sci Signal ; 6(265): ra14, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23462101

RESUMEN

ß-Catenin has a dual function in cells: fortifying cadherin-based adhesion at the plasma membrane and activating transcription in the nucleus. We found that in melanoma cells, WNT5A stimulated the disruption of N-cadherin and ß-catenin complexes by activating the guanosine triphosphatase adenosine diphosphate ribosylation factor 6 (ARF6). Binding of WNT5A to the Frizzled 4-LRP6 (low-density lipoprotein receptor-related protein 6) receptor complex activated ARF6, which liberated ß-catenin from N-cadherin, thus increasing the pool of free ß-catenin, enhancing ß-catenin-mediated transcription, and stimulating invasion. In contrast to WNT5A, the guidance cue SLIT2 and its receptor ROBO1 inhibited ARF6 activation and, accordingly, stabilized the interaction of N-cadherin with ß-catenin and reduced transcription and invasion. Thus, ARF6 integrated competing signals in melanoma cells, thereby enabling plasticity in the response to external cues. Moreover, small-molecule inhibition of ARF6 stabilized adherens junctions, blocked ß-catenin signaling and invasiveness of melanoma cells in culture, and reduced spontaneous pulmonary metastasis in mice, suggesting that targeting ARF6 may provide a means of inhibiting WNT/ß-catenin signaling in cancer.


Asunto(s)
Factores de Ribosilacion-ADP/fisiología , Melanoma/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/fisiología , Activación Transcripcional/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Factor 6 de Ribosilación del ADP , Silenciador del Gen , Humanos , Transducción de Señal , Proteína Wnt-5a , beta Catenina/metabolismo
12.
Ann Biomed Eng ; 39(1): 337-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20737290

RESUMEN

Elastin is an essential component of arteries which provides structural integrity and instructs smooth muscle cells to adopt a quiescent state. Despite interaction of endothelial cells with elastin in the internal elastic lamina, the potential for exploiting this interaction therapeutically has not been explored in detail. In this study, we show that tropoelastin (a precursor of elastin) stimulates endothelial cell migration and adhesion more than smooth muscle cells. The biological activity of tropoelastin on endothelial cells is contained in the VGVAPG domain and in the carboxy-terminal 17-amino acids. We show that the effects of the carboxy-terminal 17 amino acids, but not those of VGVAPG, are mediated by integrin α(V)ß(3). We demonstrate that tropoelastin covalently linked to stainless steel disks promotes adhesion of endothelial progenitor cells and endothelial cells to the metal surfaces. The adherent cells on the tropoelastin-coated metal surfaces form monolayers that can withstand and respond to arterial shear stress. Because of the unique effects of tropoelastin on endothelial and smooth muscle cells, coating intravascular devices with tropoelastin may stimulate their endothelialization, inhibit smooth muscle hyperplasia, and improve device performance.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Tropoelastina/administración & dosificación , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos
13.
Sci Transl Med ; 2(23): 23ra19, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20375003

RESUMEN

The innate immune system provides a first line of defense against invading pathogens by releasing multiple inflammatory cytokines, such as interleukin-1beta and tumor necrosis factor-alpha, which directly combat the infectious agent and recruit additional immune responses. This exuberant cytokine release paradoxically injures the host by triggering leakage from capillaries, tissue edema, organ failure, and shock. Current medical therapies target individual pathogens with antimicrobial agents or directly either blunt or boost the host's immune system. We explored a third approach: activating with the soluble ligand Slit an endothelium-specific, Robo4-dependent signaling pathway that strengthens the vascular barrier, diminishing deleterious aspects of the host's response to the pathogen-induced cytokine storm. This approach reduced vascular permeability in the lung and other organs and increased survival in animal models of bacterial endotoxin exposure, polymicrobial sepsis, and H5N1 influenza. Thus, enhancing the resilience of the host vascular system to the host's innate immune response may provide a therapeutic strategy for treating multiple infectious agents.


Asunto(s)
Citocinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Sepsis/inmunología , Transducción de Señal , Animales , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Cateninas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/fisiología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/patología , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Sepsis/complicaciones , Sepsis/patología , Transducción de Señal/efectos de los fármacos , Catenina delta
14.
Nat Cell Biol ; 11(11): 1325-31, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19855388

RESUMEN

Slit-Roundabout (Robo) signalling has a well-understood role in axon guidance. Unlike in the nervous system, however, Slit-dependent activation of an endothelial-specific Robo, Robo4, does not initiate a guidance program. Instead, Robo4 maintains the barrier function of the mature vascular network by inhibiting neovascular tuft formation and endothelial hyperpermeability induced by pro-angiogenic factors. In this study, we used cell biological and biochemical techniques to elucidate the molecular mechanism underlying the maintenance of vascular stability by Robo4. Here, we demonstrate that Robo4 mediates Slit2-dependent suppression of cellular protrusive activity through direct interaction with the intracellular adaptor protein paxillin and its paralogue, Hic-5. Formation of a Robo4-paxillin complex at the cell surface blocks activation of the small GTPase Arf6 and, consequently, Rac by recruitment of Arf-GAPs (ADP-ribosylation factor- directed GTPase-activating proteins) such as GIT1. Consistent with these in vitro studies, inhibition of Arf6 activity in vivo phenocopies Robo4 activation by reducing pathologic angiogenesis in choroidal and retinal vascular disease and VEGF-165 (vascular endothelial growth factor-165)-induced retinal hyperpermeability. These data reveal that a Slit2-Robo4-paxillin-GIT1 network inhibits the cellular protrusive activity underlying neovascularization and vascular leak, and identify a new therapeutic target for ameliorating diseases involving the vascular system.


Asunto(s)
Factores de Ribosilacion-ADP/antagonistas & inhibidores , Vasos Sanguíneos/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Factor 6 de Ribosilación del ADP , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Línea Celular , Movimiento Celular , Cricetinae , Humanos , Ratones
15.
Development ; 135(4): 659-67, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18223200

RESUMEN

There is emerging evidence that the canonical neural guidance factor netrin can also direct the growth of blood vessels. We deleted the gene encoding UNC5B, a receptor for the netrin family of guidance molecules, specifically within the embryonic endothelium of mice. The result is a profound structural and functional deficiency in the arterioles of the placental labyrinth, which leads first to flow reversal in the umbilical artery and ultimately to embryonic death. As this is the only detectable site of vascular abnormality in the mutant embryos, and because the phenotype cannot be rescued by a wild-type trophectoderm, we propose that UNC5B-mediated signaling is a specific and autonomous component of fetal-placental angiogenesis. Disruption of UNC5B represents a unique example of a mutation that acts solely within the fetal-placental vasculature and one that faithfully recapitulates the structural and physiological characteristics of clinical uteroplacental insufficiency. This pro-angiogenic, but spatially restricted requirement for UNC5B is not unique to murine development, as the knock-down of the Unc5b ortholog in zebrafish similarly results in the specific and highly penetrant absence of the parachordal vessel, the precursor to the lymphatic system.


Asunto(s)
Vasos Sanguíneos/embriología , Neovascularización Fisiológica , Receptores de Superficie Celular/metabolismo , Animales , Arteriolas/anomalías , Arteriolas/patología , Vasos Sanguíneos/metabolismo , Pérdida del Embrión , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/patología , Embrión no Mamífero/citología , Endotelio/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipoxia , Ratones , Ratones Endogámicos C57BL , Receptores de Netrina , Especificidad de Órganos , Fenotipo , Placenta/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Flujo Sanguíneo Regional , Transducción de Señal , Cordón Umbilical/irrigación sanguínea , Pez Cebra/embriología
16.
Science ; 313(5787): 640-4, 2006 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16809490

RESUMEN

Axonal guidance and vascular patterning share several guidance cues, including proteins in the netrin family. We demonstrate that netrins stimulate proliferation, migration, and tube formation of human endothelial cells in vitro and that this stimulation is independent of known netrin receptors. Suppression of netrin1a messenger RNA in zebrafish inhibits vascular sprouting, implying a proangiogenic role for netrins during vertebrate development. We also show that netrins accelerate neovascularization in an in vivo model of ischemia and that they reverse neuropathy and vasculopathy in a diabetic murine model. We propose that the attractive vascular and neural guidance functions of netrins offer a unique therapeutic potential.


Asunto(s)
Células Endoteliales/fisiología , Neovascularización Fisiológica , Factores de Crecimiento Nervioso/fisiología , Proteínas Supresoras de Tumor/fisiología , Inductores de la Angiogénesis , Animales , Línea Celular , Movimiento Celular , Quimiotaxis , ADN Complementario , Angiopatías Diabéticas/terapia , Neuropatías Diabéticas/terapia , Embrión no Mamífero , Endotelio Vascular/citología , Terapia Genética , Humanos , Isquemia/tratamiento farmacológico , Ratones , Músculo Esquelético/irrigación sanguínea , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/farmacología , Receptores de Netrina , Netrina-1 , Netrinas , Conducción Nerviosa , Receptores de Superficie Celular/fisiología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Pez Cebra
17.
Dev Biol ; 261(1): 235-50, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12941632

RESUMEN

Several characteristic morphological and functional differences distinguish arteries from veins. It was thought that hemodynamic forces shaped these differences; however, increasing evidence suggests that morphogenetic programs play a central role in blood vessel differentiation. Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia characterized by the inappropriate fusion of arterioles with venules. The genes implicated in this disease, ALK1 and endoglin, may be involved in defining the fundamental boundaries between arteries and veins. We previously showed that mice lacking Alk1 lost structural, molecular, and functional distinctions between arteries and veins. Here, we report that mice lacking endoglin develop arterial-venous malformations and fail to confine intraembryonic hematopoiesis to arteries. In contrast to Alk1 mutants, endoglin mutants do not show profound vessel dilation or downregulation of arterial ephrinB2. Finally, our data indicate that a failure in cardiac cushion formation observed in both strains may be secondary to the peripheral vasculature defect. The phenotypic similarities, yet reduced severity, implicates endoglin as an accessory coreceptor that specifically modulates Alk1 signaling. We propose that endoglin and Alk1 are necessary for the maintenance of distinct arterial-venous vascular beds and that attenuation of the Alk1 signaling pathway is the precipitating event in the etiology of HHT.


Asunto(s)
Receptores de Activinas Tipo I/fisiología , Vasos Sanguíneos/embriología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Molécula 1 de Adhesión Celular Vascular/fisiología , Receptores de Activinas Tipo I/deficiencia , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II , Animales , Antígenos CD , Arterias/embriología , Malformaciones Arteriovenosas/embriología , Malformaciones Arteriovenosas/genética , Secuencia de Bases , ADN/genética , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/genética , Endoglina , Efrina-B2/genética , Hematopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores de Superficie Celular , Telangiectasia Hemorrágica Hereditaria/embriología , Telangiectasia Hemorrágica Hereditaria/genética , Molécula 1 de Adhesión Celular Vascular/genética , Venas/embriología
18.
Proc Natl Acad Sci U S A ; 101(46): 16210-5, 2004 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-15520390

RESUMEN

Blood vessels and nerves often follow parallel trajectories, suggesting that distal targets use common cues that induce vascularization and innervation. Netrins are secreted by the floor plate and attract commissural axons toward the midline of the neural tube. Here, we show that Netrin-1 is also a potent vascular mitogen. Netrin-1 stimulates proliferation, induces migration, and promotes adhesion of endothelial cells and vascular smooth muscle cells with a specific activity comparable to vascular endothelial growth factor and platelet-derived growth factor. Our evidence indicates that the netrin receptor, Neogenin, mediates netrin signaling in vascular smooth muscle cells, but suggests that an unidentified receptor mediates the proangiogenic effects of Netrin-1 on endothelial cells. Netrin-1 also stimulates angiogenesis in vivo and augments the response to vascular endothelial growth factor. Thus, we demonstrate that Netrin-1 is a secreted neural guidance cue with the unique ability to attract both blood vessels and axons, and suggest that other cues may also function as vascular endothelial growth factors.


Asunto(s)
Proteínas Angiogénicas/fisiología , Axones/fisiología , Factores de Crecimiento Nervioso/fisiología , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/farmacología , Animales , Secuencia de Bases , Bovinos , ADN/genética , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Técnicas In Vitro , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/farmacología , Netrina-1 , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteínas Supresoras de Tumor , Factor A de Crecimiento Endotelial Vascular/fisiología
19.
Dev Biol ; 261(1): 251-67, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12941633

RESUMEN

Guidance and patterning of axons are orchestrated by cell-surface receptors and ligands that provide directional cues. Interactions between the Robo receptor and Slit ligand families of proteins initiate signaling cascades that repel axonal outgrowth. Although the vascular and nervous systems grow as parallel networks, the mechanisms by which the vascular endothelial cells are guided to their appropriate positions remain obscure. We have identified a putative Robo homologue, Robo4, based on its differential expression in mutant mice with defects in vascular sprouting. In contrast to known neuronal Robo family members, the arrangement of the extracellular domains of Robo4 diverges significantly from that of all other Robo family members. Moreover, Robo4 is specifically expressed in the vascular endothelium during murine embryonic development. We show that Robo4 binds Slit and inhibits cellular migration in a heterologous expression system, analogous to the role of known Robo receptors in the nervous system. Immunoprecipitation studies indicate that Robo4 binds to Mena, a known effector of Robo-Slit signaling. Finally, we show that Robo4 is the only Robo family member expressed in primary endothelial cells and that application of Slit inhibits their migration. These data demonstrate that Robo4 is a bona fide member of the Robo family and may provide a repulsive cue to migrating endothelial cells during vascular development.


Asunto(s)
Endotelio Vascular/embriología , Receptores Inmunológicos/fisiología , Receptores de Activinas Tipo I/deficiencia , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/fisiología , Receptores de Activinas Tipo II , Secuencia de Aminoácidos , Animales , Línea Celular , Movimiento Celular , Mapeo Cromosómico , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Homología de Secuencia de Aminoácido , Transducción de Señal , Pez Cebra , Proteínas Roundabout
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA