Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Development ; 145(10)2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773646

RESUMEN

Despite the essential role of the lymphatic vasculature in tissue homeostasis and disease, knowledge of the organ-specific origins of lymphatic endothelial progenitor cells remains limited. The assumption that most murine embryonic lymphatic endothelial cells (LECs) are venous derived has recently been challenged. Here, we show that the embryonic dermal blood capillary plexus constitutes an additional, local source of LECs that contributes to the formation of the dermal lymphatic vascular network. We describe a novel mechanism whereby rare PROX1-positive endothelial cells exit the capillary plexus in a Ccbe1-dependent manner to establish discrete LEC clusters. As development proceeds, these clusters expand and further contribute to the growing lymphatic system. Lineage tracing and analyses of Gata2-deficient mice confirmed that these clusters are endothelial in origin. Furthermore, ectopic expression of Vegfc in the vasculature increased the number of PROX1-positive progenitors within the capillary bed. Our work reveals a novel source of lymphatic endothelial progenitors employed during construction of the dermal lymphatic vasculature and demonstrates that the blood vasculature is likely to remain an ongoing source of LECs during organogenesis, raising the question of whether a similar mechanism operates during pathological lymphangiogenesis.


Asunto(s)
Capilares/citología , Células Endoteliales/citología , Proteínas de Homeodominio/genética , Linfangiogénesis/fisiología , Vasos Linfáticos/embriología , Células Madre/citología , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de Unión al Calcio/genética , Factor de Transcripción GATA2/genética , Linfangiogénesis/genética , Vasos Linfáticos/citología , Ratones , Ratones Transgénicos , Factor C de Crecimiento Endotelial Vascular/biosíntesis , Factor C de Crecimiento Endotelial Vascular/genética
2.
Circ Res ; 120(8): 1263-1275, 2017 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-28179432

RESUMEN

RATIONALE: Lymphatic vessel formation and function constitutes a physiologically and pathophysiologically important process, but its genetic control is not well understood. OBJECTIVE: Here, we identify the secreted Polydom/Svep1 protein as essential for the formation of the lymphatic vasculature. We analyzed mutants in mice and zebrafish to gain insight into the role of Polydom/Svep1 in the lymphangiogenic process. METHODS AND RESULTS: Phenotypic analysis of zebrafish polydom/svep1 mutants showed a decrease in venous and lymphovenous sprouting, which leads to an increased number of intersegmental arteries. A reduced number of primordial lymphatic cells populated the horizontal myoseptum region but failed to migrate dorsally or ventrally, resulting in severe reduction of the lymphatic trunk vasculature. Corresponding mutants in the mouse Polydom/Svep1 gene showed normal egression of Prox-1+ cells from the cardinal vein at E10.5, but at E12.5, the tight association between the cardinal vein and lymphatic endothelial cells at the first lymphovenous contact site was abnormal. Furthermore, mesenteric lymphatic structures at E18.5 failed to undergo remodeling events in mutants and lacked lymphatic valves. In both fish and mouse embryos, the expression of the gene suggests a nonendothelial and noncell autonomous mechanism. CONCLUSIONS: Our data identify zebrafish and mouse Polydom/Svep1 as essential extracellular factors for lymphangiogenesis. Expression of the respective genes by mesenchymal cells in intimate proximity with venous and lymphatic endothelial cells is required for sprouting and migratory events in zebrafish and for remodeling events of the lymphatic intraluminal valves in mouse embryos.


Asunto(s)
Células Endoteliales/metabolismo , Evolución Molecular , Linfangiogénesis , Vasos Linfáticos/metabolismo , Proteínas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular , Comunicación Celular , Movimiento Celular , Células Endoteliales/patología , Endotelio Linfático/anomalías , Endotelio Linfático/metabolismo , Endotelio Linfático/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Genotipo , Vasos Linfáticos/anomalías , Vasos Linfáticos/fisiopatología , Mesodermo/metabolismo , Mutación , Fenotipo , Proteínas/genética , Transducción de Señal , Factores de Tiempo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Int J Mol Sci ; 19(8)2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30071673

RESUMEN

Lymphedema is characterized by chronic swelling of any body part caused by malfunctioning or obstruction in the lymphatic system. Primary lymphedema is often considered genetic in origin. VEGFC, which is a gene encoding the ligand for the vascular endothelial growth factor receptor 3 (VEGFR3/FLT4) and important for lymph vessel development during lymphangiogenesis, has been associated with a specific subtype of primary lymphedema. Through Sanger sequencing of a proband with bilateral congenital pedal edema resembling Milroy disease, we identified a novel mutation (NM_005429.2; c.361+5G>A) in VEGFC. The mutation induced skipping of exon 2 of VEGFC resulting in a frameshift and the introduction of a premature stop codon (p.Ala50ValfsTer18). The mutation leads to a loss of the entire VEGF-homology domain and the C-terminus. Expression of this Vegfc variant in the zebrafish floorplate showed that the splice-site variant significantly reduces the biological activity of the protein. Our findings confirm that the splice-site variant, c.361+5G>A, causes the primary lymphedema phenotype in the proband. We examine the mutations and clinical phenotypes of the previously reported cases to review the current knowledge in this area.


Asunto(s)
Artrogriposis/genética , Fisura del Paladar/genética , Pie Equinovaro/genética , Mutación del Sistema de Lectura , Deformidades Congénitas de la Mano/genética , Empalme del ARN/genética , Factor C de Crecimiento Endotelial Vascular/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Artrogriposis/metabolismo , Artrogriposis/patología , Preescolar , Fisura del Paladar/metabolismo , Fisura del Paladar/patología , Pie Equinovaro/metabolismo , Pie Equinovaro/patología , Femenino , Deformidades Congénitas de la Mano/metabolismo , Deformidades Congénitas de la Mano/patología , Humanos , Lactante , Recién Nacido , Masculino , Dominios Proteicos , Factor C de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
4.
Development ; 141(6): 1239-49, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24523457

RESUMEN

The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.


Asunto(s)
Linfangiogénesis/fisiología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , ADN/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Linfangiogénesis/genética , Sistema de Señalización de MAP Quinasas , Ratones , Datos de Secuencia Molecular , Mutación Puntual , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Circ Res ; 116(10): 1660-9, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25814692

RESUMEN

RATIONALE: Collagen- and calcium-binding EGF domain-containing protein 1 (CCBE1) is essential for lymphangiogenesis in vertebrates and has been associated with Hennekam syndrome. Recently, CCBE1 has emerged as a crucial regulator of vascular endothelial growth factor-C (VEGFC) signaling. OBJECTIVE: CCBE1 is a secreted protein characterized by 2 EGF domains and 2 collagen repeats. The functional role of the different CCBE1 protein domains is completely unknown. Here, we analyzed the functional role of the different CCBE1 domains in vivo and in vitro. METHODS AND RESULTS: We analyzed the functionality of several CCBE1 deletion mutants by generating knock-in mice expressing these mutants, by analyzing their ability to enhance Vegfc signaling in vivo in zebrafish, and by testing their ability to induce VEGFC processing in vitro. We found that deleting the collagen domains of CCBE1 has a much stronger effect on CCBE1 activity than deleting the EGF domains. First, although CCBE1ΔCollagen mice fully phenocopy CCBE1 knock-out mice, CCBE1ΔEGF knock-in embryos still form rudimentary lymphatics. Second, Ccbe1ΔEGF, but not Ccbe1ΔCollagen, could partially substitute for Ccbe1 to enhance Vegfc signaling in zebrafish. Third, CCBE1ΔEGF, similarly to CCBE1, but not CCBE1ΔCollagen could activate VEGFC processing in vitro. Furthermore, a Hennekam syndrome mutation within the collagen domain has a stronger effect than a Hennekam syndrome mutation within the EGF domain. CONCLUSIONS: We propose that the collagen domains of CCBE1 are crucial for the activation of VEGFC in vitro and in vivo. The EGF domains of CCBE1 are dispensable for regulation of VEGFC processing in vitro, however, they are necessary for full lymphangiogenic activity of CCBE1 in vivo.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Sitios de Unión , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Colágeno/metabolismo , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Enfermedades de los Genitales Masculinos/genética , Enfermedades de los Genitales Masculinos/metabolismo , Genotipo , Edad Gestacional , Células HEK293 , Humanos , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/metabolismo , Vasos Linfáticos/embriología , Linfedema/genética , Linfedema/metabolismo , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
6.
EMBO J ; 30(20): 4157-70, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21857650

RESUMEN

To determine whether leukocytes need to open endothelial cell contacts during extravasation, we decided to generate mice with strongly stabilized endothelial junctions. To this end, we replaced VE-cadherin genetically by a VE-cadherin-α-catenin fusion construct. Such mice were completely resistant to the induction of vascular leaks by VEGF or histamine. Neutrophil or lymphocyte recruitment into inflamed cremaster, lung and skin were strongly inhibited in these mice, documenting the importance of the junctional route in vivo. Surprisingly, lymphocyte homing into lymph nodes was not inhibited. VE-cadherin-α-catenin associated more intensely with the actin cytoskeleton as demonstrated by its membrane mobility and detergent extractability. Our results establish the junctional route as the main pathway for extravasating leukocytes in several, although not in all tissues. Furthermore, in these tissues, plasticity of the VE-cadherin-catenin complex is central for the leukocyte diapedesis mechanism.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar , Leucocitos/fisiología , Migración Transendotelial y Transepitelial , alfa Catenina/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Células Cultivadas , Endotelio Vascular/metabolismo , Femenino , Técnicas de Sustitución del Gen , Inflamación/genética , Inflamación/metabolismo , Uniones Intercelulares/metabolismo , Leucocitos/metabolismo , Pulmón/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Piel/metabolismo , alfa Catenina/genética
7.
Circ Res ; 112(6): 956-60, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23410910

RESUMEN

RATIONALE: Mutations in vascular endothelial growth factor (VEGF) receptor-3 (VEGFR3 or FLT4) cause Milroy disease, an autosomal dominant condition that presents with congenital lymphedema. Mutations in VEGFR3 are identified in only 70% of patients with classic Milroy disease, suggesting genetic heterogeneity. OBJECTIVE: To investigate the underlying cause in patients with clinical signs resembling Milroy disease in whom sequencing of the coding region of VEGFR3 did not reveal any pathogenic variation. METHODS AND RESULTS: Exome sequencing of 5 such patients was performed, and a novel frameshift variant, c.571_572insTT in VEGFC, a ligand for VEGFR3, was identified in 1 proband. The variant cosegregated with the affected status in the family. An assay to assess the biological function of VEGFC activity in vivo, by expressing human VEGFC in the zebrafish floorplate was established. Forced expression of wild-type human VEGFC in the floorplate of zebrafish embryos leads to excessive sprouting in neighboring vessels. However, when overexpressing the human c.571_572insTT variant in the floorplate, no sprouting of vessels was observed, indicating that the base changes have a marked effect on the activity of VEGFC. CONCLUSIONS: We propose that the mutation in VEGFC is causative for the Milroy disease-like phenotype seen in this family. This is the first time a mutation in one of the ligands of VEGFR3 has been reported to cause primary lymphedema.


Asunto(s)
Mutación del Sistema de Lectura/genética , Linfedema/genética , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Linfedema/congénito , Linfedema/patología , Masculino , Linaje , Fenotipo , Adulto Joven , Pez Cebra
8.
Elife ; 122023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37097004

RESUMEN

Multiple factors are required to form functional lymphatic vessels. Here, we uncover an essential role for the secreted protein Svep1 and the transmembrane receptor Tie1 during the development of subpopulations of the zebrafish facial lymphatic network. This specific aspect of the facial network forms independently of Vascular endothelial growth factor C (Vegfc) signalling, which otherwise is the most prominent signalling axis in all other lymphatic beds. Additionally, we find that multiple specific and newly uncovered phenotypic hallmarks of svep1 mutants are also present in tie1, but not in tie2 or vegfc mutants. These phenotypes are observed in the lymphatic vasculature of both head and trunk, as well as in the development of the dorsal longitudinal anastomotic vessel under reduced flow conditions. Therefore, our study demonstrates an important function for Tie1 signalling during lymphangiogenesis as well as blood vessel development in zebrafish. Furthermore, we show genetic interaction between svep1 and tie1 in vivo, during early steps of lymphangiogenesis, and demonstrate that zebrafish as well as human Svep1/SVEP1 protein bind to the respective Tie1/TIE1 receptors in vitro. Since compound heterozygous mutations for SVEP1 and TIE2 have recently been reported in human glaucoma patients, our data have clinical relevance in demonstrating a role for SVEP1 in TIE signalling in an in vivo setting.


Asunto(s)
Vasos Linfáticos , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Ligandos , Vasos Linfáticos/metabolismo , Linfangiogénesis/genética , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Receptor TIE-1/genética , Receptor TIE-1/metabolismo
9.
Curr Opin Hematol ; 17(3): 230-6, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20393283

RESUMEN

PURPOSE OF REVIEW: Proper control of endothelial cell contacts is the basis for maintenance of the vascular barrier function. Loss of this function leads to leak of fluid and protein from the vasculature and extensive leaks cause shock and death. The endothelial barrier also controls the entry of leukocytes into tissue and it is believed that leukocytes target endothelial cell contacts to reach sites of inflammation. RECENT FINDINGS: Within the last 2 years several new molecular players and molecular interactions have been identified that either help in stabilizing the endothelial contacts or mediate their opening if triggered by the appropriate stimuli. Novel signaling mechanisms have been identified that regulate endothelial cell contacts. Whether, how and to what extent the complex of the endothelial specific adhesion molecule vascular endothelial-cadherin and its associated catenins is involved in these processes will be a major focus of this article. SUMMARY: Endothelial cell contacts are regulated by a complex interplay between various receptors and signaling mediators that control the plasticity of the cytoskeleton and the function of junctional adhesion molecules. Knowing and understanding the essential players of this network will allow development of agents that could prevent breakdown of the vascular permeability barrier in shock or that could block leukocyte extravasation and thereby antagonize inflammation.


Asunto(s)
Cadherinas/metabolismo , Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Leucocitos/metabolismo , Animales , Adhesión Celular , Humanos , Transducción de Señal
10.
J Immunol ; 181(9): 5847-56, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941173

RESUMEN

Jakmip1 belongs to a family of three related genes encoding proteins rich in coiled-coils. Jakmip1 is expressed predominantly in neuronal and lymphoid cells and colocalizes with microtubules. We have studied the expression of Jakmip1 mRNA and protein in distinct subsets of human primary lymphocytes. Jakmip1 is absent in naive CD8(+) and CD4(+) T lymphocytes from peripheral blood but is highly expressed in Ag-experienced T cells. In cord blood T lymphocytes, induction of Jakmip1 occurs upon TCR/CD28 stimulation and parallels induction of effector proteins, such as granzyme B and perforin. Further analysis of CD8(+) and CD4(+) T cell subsets showed a higher expression of Jakmip1 in the effector CCR7(-) and CD27(-) T cell subpopulations. In a gene expression follow-up of the development of CMV-specific CD8(+) response, Jakmip1 emerged as one of the most highly up-regulated genes from primary infection to latent stage. To investigate the relationship between Jakmip1 and effector function, we monitored cytotoxicity of primary CD8(+) T cells silenced for Jakmip1 or transduced with the full-length protein or the N-terminal region. Our findings point to Jakmip1 being a novel effector memory gene restraining T cell-mediated cytotoxicity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/inmunología , Citotoxicidad Inmunológica , Inhibidores de Crecimiento/fisiología , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/fisiología , Muerte Celular/genética , Muerte Celular/inmunología , Diferenciación Celular/genética , Línea Celular , Citomegalovirus/inmunología , Citotoxicidad Inmunológica/genética , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/genética , Humanos , Memoria Inmunológica/genética , Antígenos Comunes de Leucocito/biosíntesis , Proteínas de Unión al ARN/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/virología
11.
Cell Rep ; 18(5): 1256-1269, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28147279

RESUMEN

Endothelial basement membranes constitute barriers to extravasating leukocytes during inflammation, a process where laminin isoforms define sites of leukocyte exit; however, how this occurs is poorly understood. In addition to a direct effect on leukocyte transmigration, we show that laminin 511 affects endothelial barrier function by stabilizing VE-cadherin at junctions and downregulating expression of CD99L2, correlating with reduced neutrophil extravasation. Binding of endothelial cells to laminin 511, but not laminin 411 or non-endothelial laminin 111, enhanced transendothelial cell electrical resistance (TEER) and inhibited neutrophil transmigration. Data suggest that endothelial adhesion to laminin 511 via ß1 and ß3 integrins mediates RhoA-induced VE-cadherin localization to cell-cell borders, and while CD99L2 downregulation requires integrin ß1, it is RhoA-independent. Our data demonstrate that molecular information provided by basement membrane laminin 511 affects leukocyte extravasation both directly and indirectly by modulating endothelial barrier properties.


Asunto(s)
Membrana Basal/metabolismo , Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Laminina/metabolismo , Leucocitos/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/fisiología
12.
Mol Cell Biol ; 34(9): 1634-48, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24567373

RESUMEN

We have recently shown that genetic replacement of VE-cadherin by a VE-cadherin-α-catenin fusion construct strongly impairs opening of endothelial cell contacts during leukocyte extravasation and induction of vascular permeability in adult mice. Here we show that this mutation leads to lethality at midgestation on a clean C57BL/6 background. Investigating the reasons for embryonic lethality, we observed a lack of fetal liver hematopoiesis and severe lymphedema but no detectable defects in blood vessel formation and remodeling. As for the hematopoiesis defect, VE-cadherin-α-catenin affected neither the generation of hematopoietic stem and progenitor cells (HSPCs) from hemogenic endothelium nor their differentiation into multiple hematopoietic lineages. Instead, HSPCs accumulated in the fetal circulation, suggesting that their entry into the fetal liver was blocked. Edema formation was caused by disturbed lymphatic vessel development. Lymphatic progenitor cells of VE-cadherin-α-catenin-expressing embryos were able to leave the cardinal vein and migrate to the site of the first lymphatic vessel formation, yet subsequently, these cells failed to form large lumenized lymphatic vessels. Thus, stabilizing endothelial cell contacts by a covalent link between VE-cadherin and α-catenin affects recruitment of hematopoietic progenitors into the fetal liver and the development of lymph but not blood vessels.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Embrión de Mamíferos/patología , Hematopoyesis , Hígado/embriología , Sistema Linfático/embriología , Mutación , Neovascularización Fisiológica , alfa Catenina/genética , Animales , Antígenos CD/metabolismo , Vasos Sanguíneos/crecimiento & desarrollo , Cadherinas/metabolismo , Edema/patología , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Linfa/citología , Ratones , Ratones Endogámicos C57BL , Células Madre/citología , Células Madre/metabolismo , alfa Catenina/metabolismo
13.
Tissue Barriers ; 1(1): e23805, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24665379

RESUMEN

The passage of leukocytes across the blood vessel wall is a fundamental event in the inflammatory response. During the last decades, there has been significant progress in understanding the molecular mechanisms involved in leukocyte transmigration. However, it is still a matter of debate whether leukocytes migrate paracellularly or transcellularly through an endothelial cell layer. We could recently show that a VE-cadherin-α-catenin fusion protein locks endothelial junctions in the skin and strongly reduces leukocyte diapedesis in lung, skin and cremaster, establishing the paracellular route as the major transmigration pathway in these tissues. However, the homing of naïve lymphocytes into lymph nodes and extravasation of neutrophils in the inflamed peritoneum were not affected by VE-cadherin-α-catenin. This unexpected heterogeneity of the diapedesis process in different tissues as well as the complexity and dynamics of the cadherin-catenin complex in regulating endothelial junctions will be discussed.

14.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22975327

RESUMEN

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Neovascularización Fisiológica , Receptores de Lisoesfingolípidos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Esfingosina-1-Fosfato , Pez Cebra
15.
J Exp Med ; 208(8): 1721-35, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21788407

RESUMEN

Neutrophil extravasation and the regulation of vascular permeability require dynamic actin rearrangements in the endothelium. In this study, we analyzed in vivo whether these processes require the function of the actin nucleation-promoting factor cortactin. Basal vascular permeability for high molecular weight substances was enhanced in cortactin-deficient mice. Despite this leakiness, neutrophil extravasation in the tumor necrosis factor-stimulated cremaster was inhibited by the loss of cortactin. The permeability defect was caused by reduced levels of activated Rap1 (Ras-related protein 1) in endothelial cells and could be rescued by activating Rap1 via the guanosine triphosphatase (GTPase) exchange factor EPAC (exchange protein directly activated by cAMP). The defect in neutrophil extravasation was caused by enhanced rolling velocity and reduced adhesion in postcapillary venules. Impaired rolling interactions were linked to contributions of ß(2)-integrin ligands, and firm adhesion was compromised by reduced ICAM-1 (intercellular adhesion molecule 1) clustering around neutrophils. A signaling process known to be critical for the formation of ICAM-1-enriched contact areas and for transendothelial migration, the ICAM-1-mediated activation of the GTPase RhoG was blocked in cortactin-deficient endothelial cells. Our results represent the first physiological evidence that cortactin is crucial for orchestrating the molecular events leading to proper endothelial barrier function and leukocyte recruitment in vivo.


Asunto(s)
Actinas/metabolismo , Permeabilidad Capilar/inmunología , Cortactina/deficiencia , Cortactina/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Adhesión Celular/fisiología , Cortactina/genética , Cortactina/metabolismo , Células Endoteliales/metabolismo , GTP Fosfohidrolasas/metabolismo , Genotipo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Fluorescente , Neutrófilos/metabolismo , Oligonucleótidos/genética , ARN Interferente Pequeño/genética , Venas Umbilicales/citología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rho
16.
J Exp Med ; 208(12): 2393-401, 2011 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-22025303

RESUMEN

We have recently shown that vascular endothelial protein tyrosine phosphatase (VE-PTP), an endothelial membrane protein, associates with VE-cadherin and is required for optimal VE-cadherin function and endothelial cell contact integrity. The dissociation of VE-PTP from VE-cadherin is triggered by vascular endothelial growth factor (VEGF) and by the binding of leukocytes to endothelial cells in vitro, suggesting that this dissociation is a prerequisite for the destabilization of endothelial cell contacts. Here, we show that VE-cadherin/VE-PTP dissociation also occurs in vivo in response to LPS stimulation of the lung or systemic VEGF stimulation. To show that this dissociation is indeed necessary in vivo for leukocyte extravasation and VEGF-induced vascular permeability, we generated knock-in mice expressing the fusion proteins VE-cadherin-FK 506 binding protein and VE-PTP-FRB* under the control of the endogenous VE-cadherin promoter, thus replacing endogenous VE-cadherin. The additional domains in both fusion proteins allow the heterodimeric complex to be stabilized by a chemical compound (rapalog). We found that intravenous application of the rapalog strongly inhibited VEGF-induced (skin) and LPS-induced (lung) vascular permeability and inhibited neutrophil extravasation in the IL-1ß inflamed cremaster and the LPS-inflamed lung. We conclude that the dissociation of VE-PTP from VE-cadherin is indeed required in vivo for the opening of endothelial cell contacts during induction of vascular permeability and leukocyte extravasation.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Células Endoteliales/fisiología , Leucocitos/fisiología , Neumonía/inmunología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Migración Transendotelial y Transepitelial/fisiología , Animales , Células COS , Chlorocebus aethiops , Cartilla de ADN/genética , Células Endoteliales/metabolismo , Técnicas de Sustitución del Gen , Immunoblotting , Inmunoprecipitación , Lipopolisacáridos , Pulmón/metabolismo , Ratones , Proteínas Recombinantes de Fusión/metabolismo , Tacrolimus/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA