Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Cell ; 37(1): 9-53, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985645

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system (CNS), characterized by demyelination and axonal loss. It is induced by attack of autoreactive lymphocytes on the myelin sheath and endogenous remyelination failure, eventually leading to accumulation of neurological disability. Disease-modifying agents can successfully address inflammatory relapses, but have low efficacy in progressive forms of MS, and cannot stop the progressive neurodegenerative process. Thus, the stem cell replacement therapy approach, which aims to overcome CNS cell loss and remyelination failure, is considered a promising alternative treatment. Although the mechanisms behind the beneficial effects of stem cell transplantation are not yet fully understood, neurotrophic support, immunomodulation, and cell replacement appear to play an important role, leading to a multifaceted fight against the pathology of the disease. The present systematic review is focusing on the efficacy of stem cells to migrate at the lesion sites of the CNS and develop functional oligodendrocytes remyelinating axons. While most studies confirm the improvement of neurological deficits after the administration of different stem cell types, many critical issues need to be clarified before they can be efficiently introduced into clinical practice.


Asunto(s)
Esclerosis Múltiple , Enfermedades Neurodegenerativas , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Células Madre/fisiología , Oligodendroglía/patología , Oligodendroglía/fisiología
2.
Stem Cell Reports ; 18(9): 1827-1840, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37541259

RESUMEN

Adherens junctions (AJs) provide adhesive properties through cadherins and associated cytoplasmic catenins and participate in morphogenetic processes. We examined AJs formed between ISL1+ cardiovascular progenitor cells during differentiation of embryonic stem cells (ESCs) in vitro and in mouse embryogenesis in vivo. We found that, in addition to N-CADHERIN, a percentage of ISL1+ cells transiently formed vascular endothelial (VE)-CADHERIN-mediated AJs during in vitro differentiation on days 4 and 5, and the same pattern was observed in vivo. Fluorescence-activated cell sorting (FACS) analysis extended morphological data showing that VE-CADHERIN+/ISL1+ cells constitute a significant percentage of cardiac progenitors on days 4 and 5. The VE-CADHERIN+/ISL1+ cell population represented one-third of the emerging FLK1+/PDGFRa+ cardiac progenitor cells (CPCs) for a restricted time window (days 4-6). Ablation of VE-CADHERIN during ESC differentiation results in severe inhibition of cardiac differentiation. Disruption of all classic cadherins in the VE-CADHERIN+ population via a cadherin dominant-negative mutant's expression resulted in a dramatic decrease in the ISL1+ population and inhibition of cardiac differentiation.


Asunto(s)
Antígenos CD , Cadherinas , Corazón , Animales , Ratones , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Corazón/embriología
3.
Int J Dev Biol ; 66(1-2-3): 77-83, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34881790

RESUMEN

Vascular Endothelial cadherin, a type II classical cadherin, is the major cadherin molecule participating in homotypic cell-cell adhesion structures between endothelial cells. It associates with cytoplasmic and membrane cytoskeletal elements to form endothelial adherens junctions (AJs), pivotal in regulating endothelial barrier function in the adult. VE-cadherin-mediated AJs are also involved in signaling via direct or indirect associations with receptors. The generation of mutant animals, especially mice and zebrafish, revealed many details concerning the role of VE-cadherin-mediated AJs in cardiovascular development. In general, VE-cadherin knockout (KO) in mice is embryonic lethal due to severe cardiovascular defects, and major signaling pathways as well as vascular formation cues were discovered in developing endothelium. However, there is little information regarding AJs formation and their components in cardiovascular progenitors. We have characterized in detail the activation pattern of mouse VE-cadherin promoter (Pvec) in a mouse embryonic stem cells (ESCs) differentiation system in vitro. Surprisingly, we found that it is activated transiently in cardiac progenitors that belong to the second heart field. Based on Pvec activation, we isolated this population in vitro and found that it can self-renew by induction of the Wnt/ß-catenin pathway. Next, we successfully established cell culture conditions that allowed self-renewal of this population that consists of endothelial and cardiac progenitors. Transplantation in rat hearts showed that they can survive and differentiate to cardiomyocytes and endothelial cells. Although further characterization is needed, these cells can be used in cell-based therapies as well as in drug screening.


Asunto(s)
Uniones Adherentes , Células Endoteliales , Uniones Adherentes/metabolismo , Animales , Antígenos CD , Cadherinas/genética , Cadherinas/metabolismo , Células Cultivadas , Endotelio Vascular/metabolismo , Ratones , Ratas , Células Madre/metabolismo , Pez Cebra/metabolismo
4.
J Pept Sci ; 27(11): e3359, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34131994

RESUMEN

The generation of cell-penetrating peptides as cargo-delivery systems has produced an immense number of studies owing to the importance of these systems as tools to deliver molecules into the cells, as well as due to the interest to shed light into a yet unclear mechanism of the entrance of these peptides into the cells. However, many cell-penetrating peptides might present drawbacks due to causing cellular toxicity, or due to being entrapped in endosomes, or as a result of their degradation before they meet their target. In this work, a cargo transporting molecule, the Cell Penetrating Sequential Oligopeptide Carrier (CPSOC), formed by the repetitive -Lys-Aib-Cys- moiety, was tested for its ability to penetrate the cell membrane and transport the conjugated peptides into the cells. The cysteine residue anchors bioactive molecules through a stable thioether bond. The lysine supplies the positive charge to the construct, whereas the α-amino isobutyric acid is well known to induce helicoid conformation to the peptide backbone and protects from enzymatic degradation. The present study demonstrates that CPSOC penetrates the membrane transporting the conjugated cargo into the cell. When we tested CPSOC-conjugated peptides carrying critical domains of Cdc42, a small GTPase implicated in exocytosis, the internalized peptides were found to be functional because they inhibited exocytosis of von Willebrand factor from endothelial Weibel-Palade bodies a trafficking event depending on the Cdc42 protein. The data suggest that the carrier can deliver efficiently functional peptides into the cells, and thus, it can be used as a multiple-cargo transporting molecule.


Asunto(s)
Péptidos de Penetración Celular , Cuerpos de Weibel-Palade , Exocitosis , Factor de von Willebrand
5.
Viruses ; 13(4)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805449

RESUMEN

The Covid-19 pandemic has required nonpharmaceutical interventions, primarily physical distancing, personal hygiene and face mask use, to limit community transmission, irrespective of seasons. In fact, the seasonality attributes of this pandemic remain one of its biggest unknowns. Early studies based on past experience from respiratory diseases focused on temperature or humidity, with disappointing results. Our hypothesis that ultraviolet (UV) radiation levels might be a factor and a more appropriate parameter has emerged as an alternative to assess seasonality and exploit it for public health policies. Using geographical, socioeconomic and epidemiological criteria, we selected twelve North-equatorial-South countries with similar characteristics. We then obtained UV levels, mobility and Covid-19 daily incidence rates for nearly the entire 2020. Using machine learning, we demonstrated that UV radiation strongly associated with incidence rates, more so than mobility did, indicating that UV is a key seasonality indicator for Covid-19, irrespective of the initial conditions of the epidemic. Our findings can inform the implementation of public health emergency measures, partly based on seasons in the Northern and Southern Hemispheres, as the pandemic unfolds into 2021.


Asunto(s)
COVID-19/epidemiología , COVID-19/virología , SARS-CoV-2/efectos de la radiación , Humanos , Incidencia , Aprendizaje Automático , Pandemias , SARS-CoV-2/fisiología , Estaciones del Año , Temperatura , Rayos Ultravioleta , Tiempo (Meteorología)
6.
Oncol Rep ; 44(1): 126-138, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32377731

RESUMEN

Retrotransposons copy their sequences via an RNA intermediate, followed by reverse transcription into cDNA and random insertion, into a new genomic locus. New retrotransposon copies may lead to cell transformation and/or tumorigenesis through insertional mutagenesis. Methylation is a major defense mechanism against retrotransposon RNA expression and retrotransposition in differentiated cells, whereas stem cells are relatively hypo­methylated. Epithelial­to­mesenchymal transition (EMT), which transforms normal epithelial cells into mesenchymal­like cells, also contributes to tumor progression and tumor metastasis. Cancer stem cells (CSCs), a fraction of undifferentiated tumor­initiating cancer cells, are reciprocally related to EMT. In the present study, the outcome of long terminal repeat (LTR)­Viral­Like 30 (VL30) retrotransposition was examined in mouse mammary stem­like/progenitor HC11 epithelial cells. The transfection of HC11 cells with a VL30 retrotransposon, engineered with an EGFP­based retrotransposition cassette, elicited a higher retrotransposition frequency in comparison to differentiated J3B1A and C127 mouse mammary cells. Fluorescence microscopy and PCR analysis confirmed the specificity of retrotransposition events. The differentiated retrotransposition­positive cells retained their epithelial morphology, while the respective HC11 cells acquired mesenchymal features associated with the loss of E­cadherin, the induction of N­cadherin, and fibronectin and vimentin protein expression, as well as an increased transforming growth factor (TGF)­ß1, Slug, Snail­1 and Twist mRNA expression. In addition, they were characterized by cell proliferation in low serum, and the acquisition of CSC­like properties indicated by mammosphere formation under anchorage­independent conditions. Mammospheres exhibited an increased Nanog and Oct4 mRNA expression and a CD44+/CD24­/low antigenic phenotype, as well as self­renewal and differentiation capacity, forming mammary acini­like structures. DNA sequencing analysis of retrotransposition­positive HC11 cells revealed retrotransposed VL30 copies integrated at the vicinity of EMT­, cancer type­ and breast cancer­related genes. The inoculation of these cells into Balb/c mice produced cytokeratin­positive tumors containing pancytokeratin­positive cells, indicative of cell invasion features. On the whole, the findings of the present study demonstrate, for the first time, to the best of our knowledge, that stem­like epithelial HC11 cells are amenable to VL30 retrotransposition associated with the induction of EMT and CSC generation, leading to tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Mamarias Experimentales/patología , Células Madre Neoplásicas/metabolismo , Retroelementos , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Transfección
7.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L352-63, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27233997

RESUMEN

Increased pulmonary vascular resistance in pulmonary hypertension (PH) is caused by vasoconstriction and obstruction of small pulmonary arteries by proliferating vascular cells. In analogy to cancer, subsets of proliferating cells may be derived from endothelial cells transitioning into a mesenchymal phenotype. To understand phenotypic shifts transpiring within endothelial cells in PH, we injected rats with alkaloid monocrotaline to induce PH and measured lung tissue levels of endothelial-specific protein and critical differentiation marker vascular endothelial (VE)-cadherin. VE-cadherin expression by immonoblotting declined significantly 24 h and 15 days postinjection to rebound to baseline at 30 days. There was a concomitant increase in transcriptional repressors Snail and Slug, along with a reduction in VE-cadherin mRNA. Mesenchymal markers α-smooth muscle actin and vimentin were upregulated by immunohistochemistry and immunoblotting, and α-smooth muscle actin was colocalized with endothelial marker platelet endothelial cell adhesion molecule-1 by confocal microscopy. Apoptosis was limited in this model, especially in the 24-h time point. In addition, monocrotaline resulted in activation of protein kinase B/Akt, endothelial nitric oxide synthase (eNOS), nuclear factor (NF)-κB, and increased lung tissue nitrotyrosine staining. To understand the etiological relationship between nitrosative stress and VE-cadherin suppression, we incubated cultured rat lung endothelial cells with endothelin-1, a vasoconstrictor and pro-proliferative agent in pulmonary arterial hypertension. This resulted in activation of eNOS, NF-κB, and Akt, in addition to induction of Snail, downregulation of VE-cadherin, and synthesis of vimentin. These effects were blocked by eNOS inhibitor N(ω)-nitro-l-arginine methyl ester. We propose that transcriptional repression of VE-cadherin by nitrosative stress is involved in endothelial-mesenchymal transdifferentiation in experimental PH.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/fisiología , Hipertensión Pulmonar/metabolismo , Animales , Antígenos CD/genética , Apoptosis , Cadherinas/genética , Transdiferenciación Celular , Células Cultivadas , Regulación hacia Abajo , Endotelina-1/fisiología , Endotelio Vascular/patología , Activación Enzimática , Silenciador del Gen , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/patología , Pulmón/patología , Monocrotalina , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas Wistar , Transcripción Genética
8.
Stem Cells Int ; 2016: 8305624, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28101109

RESUMEN

Embryonic Stem (ES) or induced Pluripotent Stem (iPS) cells are important sources for cardiomyocyte generation, targeted for regenerative therapies. Several in vitro protocols are currently utilized for their differentiation, but the value of cell-based approaches remains unclear. Here, we characterized a cardiovascular progenitor population derived during ES differentiation, after selection based on VE-cadherin promoter (Pvec) activity. ESCs were genetically modified with an episomal vector, allowing the expression of puromycin resistance gene, under Pvec activity. Puromycin-surviving cells displayed cardiac and endothelial progenitor cells characteristics. Expansion and self-renewal of this cardiac and endothelial dual-progenitor population (CEDP) were achieved by Wnt/ß-catenin pathway activation. CEDPs express early cardiac developmental stage-specific markers but not markers of differentiated cardiomyocytes. Similarly, CEDPs express endothelial markers. However, CEDPs can undergo differentiation predominantly to cTnT+ (~47%) and VE-cadherin+ (~28%) cells. Transplantation of CEDPs in the left heart ventricle of adult rats showed that CEDPs-derived cells survive and differentiate in vivo for at least 14 days after transplantation. A novel, dual-progenitor population was isolated during ESCs differentiation, based on Pvec activity. This lineage can self-renew, permitting its maintenance as a source of cardiovascular progenitor cells and constitutes a useful source for regenerative approaches.

9.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L363-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18515405

RESUMEN

Activation of the Rho GTPase Cdc42 has been shown in endothelial cell monolayers to prevent disassembly of interendothelial junctions and the increase in endothelial permeability. Here, we addressed the in vivo role of Cdc42 activity in mediating endothelial barrier protection in lungs by generating mice expressing the dominant active mutant V12Cdc42 protein in vascular endothelial cells targeted via the VE-cadherin promoter. These mice developed normally and exhibited constitutively active GTP-bound Cdc42. The increase in lung vascular permeability and gain in tissue water content in response to intraperitoneal lipopolysaccharide challenge (7 mg/kg) were markedly attenuated in the transgenic mice. To address the basis of the protective effect, we observed that expression of V12Cdc42 mutant in endothelial monolayers reduced the decrease in transendothelial electrical resistance, a measure of opening of interendothelial junctions, thus indicating that Cdc42 activity preserved junctional integrity. RhoA activity in V12Cdc42-expressing endothelial monolayers was reduced compared with untransfected cells, suggesting that activated Cdc42 functions by counteracting the canonical RhoA-mediated mechanism of endothelial hyperpermeability. Therefore, Cdc42 activity of microvessel endothelial cells is a critical determinant of junctional barrier restrictiveness and may represent a means of therapeutically modulating increased lung vascular permeability and edema formation.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Mutación Missense , Edema Pulmonar/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Antígenos CD/genética , Barrera Alveolocapilar/patología , Cadherinas/genética , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Impedancia Eléctrica , Células Endoteliales/patología , Genes Dominantes/genética , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Edema Pulmonar/inducido químicamente , Edema Pulmonar/genética , Edema Pulmonar/patología , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
10.
Anticancer Res ; 28(2A): 843-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18507027

RESUMEN

BACKGROUND: Colorectal tumors arise from unregulated cell proliferation of the intestinal epithelium through a multistep process the first step usually being premalignant adenomas. Familial adenomatous polyposis patients carry a germ line mutation in the APC gene leading to the development of thousands of polyps, which, if left untreated, lead to cancer. The goal of this study was the establishment of conditions for the culture of epithelial cells composing an adenomatic structure. MATERIALS AND METHODS: All colorectal specimens were obtained from FAP patients within 1-2 hours of surgery. Cells were cultured by standard procedures. Characterization was carried out by immunostaining with pancytokeratin, vimentin and desmoplakin antibodies. RESULTS: A culture protocol that gave rise to epithelial cell growth with high efficiency and efficacy was established. Successful subculturing of the cell sheets took place only when dispase prepared in Ca2+ and Mg2+ free medium, was used to digest polyp tissue taken from FAP patients. By using immunostaining these cells were characterized as epithelial. CONCLUSION: The protocol we developed here provides a means of preparing cell cultures from human colorectal adenomas, which aid in the research of the transition from adenoma to carcinoma.


Asunto(s)
Adenoma/patología , Poliposis Adenomatosa del Colon/patología , Técnicas de Cultivo de Célula/métodos , Células Epiteliales/patología , Pólipos del Colon/patología , Neoplasias Colorrectales/patología , Humanos , Fenotipo
11.
Am J Physiol Lung Cell Mol Physiol ; 291(4): L764-71, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16782751

RESUMEN

Lung inflammatory disease is characterized by increased polymorphonuclear leukocyte (PMN) infiltration and vascular permeability. PMN infiltration into tissue involves signaling between endothelial cells and migrating PMNs, which leads to alterations in the organization of adherens junctions (AJs). We addressed the possible role of the protein constituents of AJs, endothelium-specific vascular-endothelial (VE)-cadherin, in the migration of PMNs. Studies were made using VE-cadherin mutant constructs lacking the extracellular domain (DeltaEXD) or, additionally, lacking the COOH-terminus beta-catenin-binding domain (DeltaEXDDeltabeta). Either construct was transduced in pulmonary microvessel endothelia of mice using cationic liposome-encapuslated cDNA constructs injected intravenously. Optimal expression of constructs was seen by Western blot analysis within 24 h. Vessel wall liquid permeability measured as the lung microvessel capillary filtration coefficient increased threefold in DeltaEXD-transduced lungs, indicating patency of interendothelial junctions, whereas the control DeltaEXDDeltabeta construct was ineffective. To study lung tissue PMN recruitment, we challenged mice intraperitoneally with LPS (3 mg/kg) for 6 h and measured PMN numbers by bronchoalveolar lavage and their accumulation morphometrically in lung tissue. DeltaEXD expression markedly reduced the PMN sequestration and migration seen in nontransfected (control wild type) or DeltaEXDDeltabeta-transfected (negative control) mice challenged with LPS. In addition, DeltaEXD transfection suppressed LPS-induced activation of NF-kappaB and consequent ICAM-1 expression. These results suggest that disassembly of VE-cadherin junctions serves as a negative signal for limiting transendothelial PMN migration secondary to decreased ICAM-1 expression in the mouse model of LPS-induced sepsis.


Asunto(s)
Antígenos CD/fisiología , Cadherinas/fisiología , Endotelio Vascular/metabolismo , Pulmón/fisiología , Infiltración Neutrófila/fisiología , Uniones Adherentes/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Técnicas In Vitro , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Microcirculación , Mutación , FN-kappa B/metabolismo , Estructura Terciaria de Proteína , Transfección
12.
Circ Res ; 98(1): 73-80, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16322481

RESUMEN

The endothelial adherens junctions (AJs) consist of trans-oligomers of membrane spanning vascular endothelial (VE)-cadherin proteins, which bind beta-catenin through their cytoplasmic domain. beta-Catenin in turn binds alpha-catenin and connects the AJ complex with the actin cytoskeleton. We addressed the in vivo effects of loss of VE-cadherin interactions on lung vascular endothelial permeability and the role of specific Rho GTPase effectors in regulating the increase in permeability induced by AJ destabilization. We used cationic liposomes encapsulating the mutant of VE-cadherin lacking the extracellular domain (DeltaEXD) to interfere with AJ assembly in mouse lung endothelial cells. We observed that lung vascular permeability (quantified as microvessel filtration coefficient [K(f,c)]) was increased 5-fold in lungs expressing DeltaEXD. This did not occur to the same degree on expression of the VE-cadherin mutant, DeltaEXDDeltabeta, lacking the beta-catenin-binding site. The increased vascular permeability was the result of destabilization of VE-cadherin homotypic interaction induced by a shift in the binding of beta-catenin from wild-type VE-cadherin to the expressed DeltaEXD mutant. Because DeltaEXD expression in endothelial cells activated the Rho GTPase Cdc42, we addressed its role in the mechanism of increased endothelial permeability induced by AJ destabilization. Coexpression of dominant-negative Cdc42 (N17Cdc42) prevented the increase in K(f,c) induced by DeltaEXD. This was attributed to inhibition of the association of alpha-catenin with the DeltaEXD-beta-catenin complex. The results demonstrate that Cdc42 regulates AJ permeability by controlling the binding of alpha-catenin with beta-catenin and the consequent interaction of the VE-cadherin/catenin complex with the actin cytoskeleton.


Asunto(s)
Cadherinas/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , alfa Catenina/metabolismo , Proteína de Unión al GTP cdc42/fisiología , Antígenos CD , Células Cultivadas , Citoesqueleto/fisiología , Humanos
13.
Circ Res ; 94(2): 159-66, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14656933

RESUMEN

The mechanisms involved in the restoration of endothelial cell junctions subsequent to barrier disruption remain unclear. It is known that formation of adherens junctions (AJs) affects cytoskeletal actin arrangement and that Rho GTPases regulate the state of actin polymerization. In the present study, we examined the role of the Rho GTPases, Rho, Rac, and Cdc42 in the reannealing of AJs. We studied the response to thrombin, which increases endothelial permeability through disassembly of AJs, followed by recovery of barrier function through junctional reannealing within 2 hours. Cdc42 was activated late, at approximately 1 hour after thrombin exposure, concurrent with its translocation from the cytoplasm to the membrane. Activation and translocation of Cdc42 preceded the reformation of AJs. Expression of the dnCdc42 mutant (N17Cdc42) significantly delayed the reformation of the VE-cadherin-containing AJs and restoration of endothelial barrier function. We also studied the lung microcirculation to address the in vivo relevance of Cdc42 signaling in barrier restoration. N17Cdc42 expression in the mouse lung endothelium markedly attenuated the endothelial barrier recovery after the permeability increase induced by activation of the thrombin receptor protease-activated receptor-1. These findings demonstrate the critical function of Cdc42 in restoring AJ-dependent, endothelial cell homotypic adhesion and barrier function. The delayed activation of Cdc42 represents a negative-feedback mechanism that signals AJ reassembly after the increase in endothelial permeability induced by inflammatory mediators such as thrombin.


Asunto(s)
Uniones Adherentes/fisiología , Endotelio Vascular/citología , Proteína de Unión al GTP cdc42/fisiología , Animales , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Endotelio/citología , Retroalimentación Fisiológica , Humanos , Pulmón/citología , Masculino , Ratones , Microcirculación , Oligopéptidos/farmacología , Transporte de Proteínas , Receptor PAR-1/efectos de los fármacos , Receptor PAR-1/fisiología , Proteínas Recombinantes de Fusión/fisiología , Organismos Libres de Patógenos Específicos , Trombina/farmacología , Transfección , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/fisiología
14.
Am J Physiol Lung Cell Mol Physiol ; 285(2): L434-42, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12740216

RESUMEN

The adherens junction is a multiprotein complex consisting of the transmembrane vascular endothelial cadherin (VEC) and cytoplasmic catenins (p120, beta-catenin, plakoglobin, alpha-catenin) responsible for the maintenance of endothelial barrier function. Junctional disassembly and modifications in cadherin/catenin complex lead to increased paracellular permeability of the endothelial barrier. However, the mechanisms of junctional disassembly remain unclear. In this study, we used the proinflammatory mediator thrombin to compromise the barrier function and test the hypothesis that phosphorylation-induced alterations of VEC, beta-catenin, and p120 regulate junction disassembly and mediate the increased endothelial permeability response. The study showed that thrombin induced dephosphorylation of VEC, which is coupled to disassembly of cell-cell contacts, but VEC remained in aggregates at the plasma membrane. The cytoplasmic catenins dissociated from the VEC cytoplasmic domain in thin membrane projections formed in interendothelial gaps. We also showed that thrombin induced dephosphorylation of beta-catenin and phosphorylation of p120. Thrombin-induced interendothelial gap formation and increased endothelial permeability were blocked by protein kinase C inhibition using chelerythrine and Gö-6976 but not by LY-379196. Chelerythrine also prevented thrombin-induced phosphorylation changes of the cadherin/catenin complex. Thus the present study links posttranslational modifications of VEC, beta-catenin, and p120 to the mechanism of thrombin-induced increase in endothelial permeability.


Asunto(s)
Cadherinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endotelio Vascular/fisiología , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Arteria Pulmonar/fisiología , Trombina/farmacología , Transactivadores/metabolismo , Alcaloides , Antígenos CD , Benzofenantridinas , Carbazoles/farmacología , Cateninas , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/farmacología , Cinética , Mesilatos/farmacología , Fenantridinas/farmacología , Procesamiento Proteico-Postraduccional , Pirroles/farmacología , beta Catenina , Catenina delta
15.
J Biol Chem ; 278(18): 16230-6, 2003 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-12595527

RESUMEN

The cytoplasmic domain of cadherins and the associated catenins link the cytoskeleton with signal transduction pathways. To study the signaling function of non-junctional VE-cadherin, which can form during the loss VE-cadherin homotypic adhesion, wild type VE-cadherin or VE-cadherin cytoplasmic domain (DeltaEXD) was expressed in sub-confluent endothelial cells. We observed that Cdc42 was activated in transfected cells and that these cells also developed Cdc42-dependent >70-microm-long plasma membrane protrusions. The formation of these structures required actin polymerization, and they developed specifically in endothelial cells as compared with epithelial cells. Expression of the VE-cadherin cytoplasmic domain lacking the beta-catenin binding site also induced Cdc42 activation; thus, its activation cannot be ascribed to beta-catenin binding. However, these cells were not able to form the protrusions. These results suggest that the cytoplasmic domain of non-junctional VE-cadherin can serve as a scaffold involved in Cdc42 activation at the endothelial plasma membrane. beta-Catenin and the associated alpha-catenin may serve as support sites for actin polymerization, leading to formation of long plasma membrane protrusions. Thus, non-junctional VE-cadherin actively participates in inside-out signaling at the plasma membrane, leading to the development of endothelial membrane protrusions.


Asunto(s)
Cadherinas/fisiología , Membrana Celular/metabolismo , Endotelio Vascular/citología , Proteína de Unión al GTP cdc42/fisiología , Antígenos CD , Cateninas , Moléculas de Adhesión Celular/fisiología , Células Cultivadas , Proteínas del Citoesqueleto/fisiología , Endotelio Vascular/ultraestructura , Humanos , Fosfoproteínas/fisiología , Transactivadores/fisiología , beta Catenina , Catenina delta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...