Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Catheter Cardiovasc Interv ; 96(1): 179-186, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31638343

RESUMEN

OBJECTIVES: This observational study was designed to analyze the safety and feasibility of percutaneous skin closure using a purse-string suture (PSS) after MitraClip procedures. METHODS: Forty-one consecutive patients with severe mitral regurgitation who underwent MitraClip implantation from February 2018 to January 2019 at our institution received a PSS after percutaneous mitral valve repair before withdrawal of the 24-French (Fr) sheath. Protamine was not administered after venous closure at procedure end. No compression therapy (e.g., compression bandage or pneumatic compression device) was used. Patients were on bed rest for 6 hrs prior to suture removal, which was accomplished 18-24 hrs after MitraClip implantation. We analyzed the occurrence of any vascular or thromboembolic complications during the hospital stay and until the 3-month follow-up. RESULTS: The primary endpoint-any access-related major complication-did not occur in any patients. None of the patients revealed a pseudoaneurysm or an arteriovenous fistula, a thromboembolic complication, or local stenosis related to the PSS closure. The secondary endpoint- minor access-site vascular complications (hematoma)- was documented in six (14.6%) patients. CONCLUSIONS: Venous access-site closure with a PSS without the need for protamine administration or compression therapy appears to be safe and feasible in patients undergoing MitraClip implantation with access via a 24-Fr sheath.


Asunto(s)
Cateterismo Cardíaco/instrumentación , Cateterismo Periférico , Vena Femoral/cirugía , Implantación de Prótesis de Válvulas Cardíacas/instrumentación , Hemorragia/prevención & control , Técnicas Hemostáticas , Insuficiencia de la Válvula Mitral/cirugía , Válvula Mitral/cirugía , Técnicas de Sutura , Anciano , Anciano de 80 o más Años , Cateterismo Cardíaco/efectos adversos , Cateterismo Periférico/efectos adversos , Femenino , Prótesis Valvulares Cardíacas , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Hemorragia/etiología , Técnicas Hemostáticas/efectos adversos , Humanos , Masculino , Válvula Mitral/diagnóstico por imagen , Válvula Mitral/fisiopatología , Insuficiencia de la Válvula Mitral/diagnóstico por imagen , Insuficiencia de la Válvula Mitral/fisiopatología , Punciones , Índice de Severidad de la Enfermedad , Técnicas de Sutura/efectos adversos , Resultado del Tratamiento
2.
Arterioscler Thromb Vasc Biol ; 38(5): 1170-1177, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29599141

RESUMEN

OBJECTIVE: Endothelial cells play important roles in tissue homeostasis and vascularization, a function that is impaired by aging. Here, we aim to decipher the role of the microenvironment underlying the impairment of endothelial cell functions by aging. APPROACH AND RESULTS: RNA sequencing of isolated cardiac endothelial cells derived from young and 18-month-old mouse hearts revealed that aging affects the endothelial expression of genes encoding extracellular matrix proteins, specifically the laminin ß1 (Lamb1) and laminin ß2 (Lamb2) chains. Whereas Lamb1 was upregulated, Lamb2 was decreased in endothelial cells in old mice compared with young controls. A similar change in expression patterns was observed after induction of acute myocardial infarction. Mimicking aging and injury conditions by plating endothelial cells on laminin ß1-containing laminin 411 matrix impaired endothelial cell adhesion, migration, and tube formation and augmented endothelial-to-mesenchymal transition and endothelial detachment compared with laminin 421, which contains the laminin ß2 chain. Because laminins can signal via integrin receptors, we determined the activation of ITGB1 (integrin ß1). Laminin 421 coating induced a higher activation of ITGB1 compared with laminin 411. siRNA-mediated silencing of ITGB1 reduced laminin ß2-dependent adhesion, suggesting that laminin ß2 more efficiently activates ITGB1. CONCLUSIONS: Mimicking age-related modulation of laminin ß1 versus ß2 chain expression changes the functional properties and phenotype of endothelial cells. The dysregulation of the extracellular matrix during vascular aging may contribute to age-associated impairment of organ function and fibrosis.


Asunto(s)
Envejecimiento/metabolismo , Células Endoteliales/metabolismo , Laminina/metabolismo , Neovascularización Fisiológica , Factores de Edad , Envejecimiento/genética , Animales , Adhesión Celular , Movimiento Celular , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Microambiente Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/metabolismo , Laminina/genética , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Fenotipo , Transducción de Señal
3.
Mol Psychiatry ; 20(7): 880-888, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25385367

RESUMEN

Inflammation in the central nervous system (CNS) and disruption of its immune privilege are major contributors to the pathogenesis of multiple sclerosis (MS) and of its rodent counterpart, experimental autoimmune encephalomyelitis (EAE). We have previously identified developmental endothelial locus-1 (Del-1) as an endogenous anti-inflammatory factor, which inhibits integrin-dependent leukocyte adhesion. Here we show that Del-1 contributes to the immune privilege status of the CNS. Intriguingly, Del-1 expression decreased in chronic-active MS lesions and in the inflamed CNS in the course of EAE. Del-1-deficiency was associated with increased EAE severity, accompanied by increased demyelination and axonal loss. As compared with control mice, Del-1(-/-) mice displayed enhanced disruption of the blood-brain barrier and increased infiltration of neutrophil granulocytes in the spinal cord in the course of EAE, accompanied by elevated levels of inflammatory cytokines, including interleukin-17 (IL-17). The augmented levels of IL-17 in Del-1-deficiency derived predominantly from infiltrated CD8(+) T cells. Increased EAE severity and neutrophil infiltration because of Del-1-deficiency was reversed in mice lacking both Del-1 and IL-17 receptor, indicating a crucial role for the IL-17/neutrophil inflammatory axis in EAE pathogenesis in Del-1(-/-) mice. Strikingly, systemic administration of Del-1-Fc ameliorated clinical relapse in relapsing-remitting EAE. Therefore, Del-1 is an endogenous homeostatic factor in the CNS protecting from neuroinflammation and demyelination. Our findings provide mechanistic underpinnings for the previous implication of Del-1 as a candidate MS susceptibility gene and suggest that Del-1-centered therapeutic approaches may be beneficial in neuroinflammatory and demyelinating disorders.


Asunto(s)
Axones/metabolismo , Barrera Hematoencefálica/metabolismo , Proteínas Portadoras/metabolismo , Vaina de Mielina/metabolismo , Neuroinmunomodulación/fisiología , Médula Espinal/metabolismo , Animales , Axones/efectos de los fármacos , Axones/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Proteínas de Unión al Calcio , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Proteínas Portadoras/genética , Moléculas de Adhesión Celular , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/patología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Péptidos y Proteínas de Señalización Intercelular , Interleucina-17/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Neuroinmunomodulación/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Índice de Severidad de la Enfermedad , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
4.
J Mol Cell Cardiol ; 88: 111-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26456066

RESUMEN

MicroRNAs are endogenously expressed small noncoding RNAs that regulate gene expression. Laminar blood flow induces atheroprotective gene expression in endothelial cells (ECs) in part by upregulating the transcription factor KLF2. Here, we identified KLF2- and flow-responsive miRs that affect gene expression in ECs. Bioinformatic assessment of mRNA expression patterns identified the miR-30-5p seed sequence to be highly enriched in mRNAs that are downregulated by KLF2. Indeed, KLF2 overexpression and shear stress stimulation in vitro and in vivo increased the expression of miR-30-5p family members. Furthermore, we identified angiopoietin 2 (Ang2) as a target of miR-30. MiR-30 overexpression reduces Ang2 levels, whereas miR-30 inhibition by LNA-antimiRs induces Ang2 expression. Consistently, miR-30 reduced basal and TNF-α-induced expression of the inflammatory cell­cell adhesion molecules E-selectin, ICAM1 and VCAM1, which was rescued by stimulation with exogenous Ang2. In summary, KLF2 and shear stress increase the expression of the miR-30-5p family which acts in an anti-inflammatory manner in ECs by impairing the expression of Ang2 and inflammatory cell­cell adhesion molecules. The upregulation of miR-30-5p family members may contribute to the atheroprotective effects of shear stress.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/genética , ARN Mensajero/genética , Estrés Mecánico , Proteínas de Transporte Vesicular/genética , Adenoviridae/genética , Secuencia de Bases , Biología Computacional , Selectina E/genética , Selectina E/metabolismo , Regulación de la Expresión Génica , Hemorreología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Lentivirus/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Transducción de Señal , Transducción Genética , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteínas de Transporte Vesicular/metabolismo
5.
Blood ; 121(15): 3041-50, 2013 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-23386126

RESUMEN

Angiogenesis, defined as blood vessel formation from a preexisting vasculature, is governed by multiple signal cascades including integrin receptors, in particular integrin αVß3. Here we identify the endothelial cell (EC)-secreted factor epidermal growth factor-like protein 7 (EGFL7) as a novel specific ligand of integrin αVß3, thus providing mechanistic insight into its proangiogenic actions in vitro and in vivo. Specifically, EGFL7 attaches to the extracellular matrix and by its interaction with integrin αVß3 increases the motility of EC, which allows EC to move on a sticky underground during vessel remodeling. We provide evidence that the deregulation of EGFL7 in zebrafish embryos leads to a severe integrin-dependent malformation of the caudal venous plexus, pointing toward the significance of EGFL7 in vessel development. In biopsy specimens of patients with neurologic diseases, vascular EGFL7 expression rose with increasing EC proliferation. Further, EGFL7 became upregulated in vessels of the stroke penumbra using a mouse model of reversible middle cerebral artery occlusion. Our data suggest that EGFL7 expression depends on the remodeling state of the existing vasculature rather than on the phenotype of neurologic disease analyzed. In sum, our work sheds a novel light on the molecular mechanism EGFL7 engages to govern physiological and pathological angiogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Integrina alfaVbeta3/metabolismo , Secuencias de Aminoácidos/genética , Animales , Proteínas de Unión al Calcio , Adhesión Celular/genética , Movimiento Celular/genética , Familia de Proteínas EGF , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/metabolismo , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/farmacología , Matriz Extracelular/metabolismo , Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Integrina alfaVbeta3/genética , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra
6.
Circ Res ; 112(6): 924-34, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23362312

RESUMEN

RATIONALE: Polarity proteins are involved in the apico-basal orientation of epithelial cells, but relatively little is known regarding their function in mesenchymal cells. OBJECTIVE: We hypothesized that polarity proteins also contribute to endothelial processes like angiogenesis. METHODS AND RESULTS: Screening of endothelial cells revealed high expression of the polarity protein Scribble (Scrib). On fibronectin-coated carriers Scrib siRNA (siScrib) blocked directed but not random migration of human umbilical vein endothelial cells and led to an increased number and disturbed orientation of cellular lamellipodia. Coimmunoprecipitation/mass spectrometry and glutathione S-transferase (GST) pulldown assays identified integrin α5 as a novel Scrib interacting protein. By total internal reflection fluorescence (TIRF) microscopy, Scrib and integrin α5 colocalize at the basal plasma membrane of endothelial cells. Western blot and fluorescence activated cell sorting (FACS) analysis revealed that silencing of Scrib reduced the protein amount and surface expression of integrin α5 whereas surface expression of integrin αV was unaffected. Moreover, in contrast to fibronectin, the ligand of integrin α5, directional migration on collagen mediated by collagen-binding integrins was unaffected by siScrib. Mechanistically, Scrib supported integrin α5 recycling and protein stability by blocking its interaction with Rab7a, its translocation into lysosomes, and its subsequent degradation by pepstatin-sensitive proteases. In siScrib-treated cells, reinduction of the wild-type protein but not of PSD95, Dlg, ZO-1 (PDZ), or leucine rich repeat domain mutants restored integrin α5 abundance and directional cell migration. The downregulation of Scrib function in Tg(kdrl:EGFP)(s843) transgenic zebrafish embryos delayed the angiogenesis of intersegmental vessels. CONCLUSIONS: Scrib is a novel regulator of integrin α5 turnover and sorting, which is required for oriented cell migration and sprouting angiogenesis.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Integrina alfa5/metabolismo , Proteínas de la Membrana/fisiología , Neovascularización Fisiológica/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Ensayos de Migración Celular , Movimiento Celular/efectos de los fármacos , Células Endoteliales/fisiología , Humanos , Integrina alfaV/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , ARN Interferente Pequeño/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores
7.
Basic Res Cardiol ; 109(2): 404, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24522833

RESUMEN

ß1-Integrins are essential for angiogenesis. The mechanisms regulating integrin function in endothelial cells (EC) and their contribution to angiogenesis remain elusive. Brag2 is a guanine nucleotide exchange factor for the small Arf-GTPases Arf5 and Arf6. The role of Brag2 in EC and angiogenesis and the underlying molecular mechanisms remain unclear. siRNA-mediated Brag2-silencing reduced EC angiogenic sprouting and migration. Brag2-siRNA transfection differentially affected α5ß1- and αVß3-integrin function: specifically, Brag2-silencing increased focal/fibrillar adhesions and adhesion on ß1-integrin ligands (fibronectin and collagen), while reducing the adhesion on the αVß3-integrin ligand, vitronectin. Consistent with these results, Brag2-silencing enhanced surface expression of α5ß1-integrin, while reducing surface expression of αVß3-integrin. Mechanistically, Brag2-mediated αVß3-integrin-recycling and ß1-integrin endocytosis and specifically of the active/matrix-bound α5ß1-integrin present in fibrillar/focal adhesions (FA), suggesting that Brag2 contributes to the disassembly of FA via ß1-integrin endocytosis. Arf5 and Arf6 are promoting downstream of Brag2 angiogenic sprouting, ß1-integrin endocytosis and the regulation of FA. In vivo silencing of the Brag2-orthologues in zebrafish embryos using morpholinos perturbed vascular development. Furthermore, in vivo intravitreal injection of plasmids containing Brag2-shRNA reduced pathological ischemia-induced retinal and choroidal neovascularization. These data reveal that Brag2 is essential for developmental and pathological angiogenesis by promoting EC sprouting through regulation of adhesion by mediating ß1-integrin internalization and link for the first time the process of ß1-integrin endocytosis with angiogenesis.


Asunto(s)
Adhesión Celular/fisiología , Factores de Intercambio de Guanina Nucleótido/genética , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Neovascularización Patológica/fisiopatología , Retinopatía de la Prematuridad/fisiopatología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Animales , Animales Modificados Genéticamente , Células COS , Movimiento Celular/fisiología , Chlorocebus aethiops , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/fisiopatología , Modelos Animales de Enfermedad , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , ARN Interferente Pequeño/genética , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
8.
Circ Res ; 111(7): 854-62, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22821930

RESUMEN

RATIONALE: Cell therapy is a promising option for the treatment of acute or chronic myocardial ischemia. The intracoronary infusion of cells imposes the potential risk of cell clotting, which may be prevented by the addition of anticoagulants. However, a comprehensive analysis of the effects of anticoagulants on the function of the cells is missing. OBJECTIVE: Here, we investigated the effects of heparin and the thrombin inhibitor bivalirudin on bone marrow-derived mononuclear cell (BMC) functional activity and homing capacity. METHODS AND RESULTS: Heparin, but not bivalirudin profoundly and dose-dependently inhibited basal and stromal cell-derived factor 1 (SDF-1)-induced BMC migration. Incubation of BMCs with 20 U/mL heparin for 30 minutes abrogated SDF-1-induced BMC invasion (16±8% of control; P<0.01), whereas no effects on apoptosis or colony formation were observed (80±33% and 100±44% of control, respectively). Pretreatment of BMCs with heparin significantly reduced the homing of the injected cells in a mouse ear-wound model (69±10% of control; P<0.05). In contrast, bivalirudin did not inhibit in vivo homing of BMCs. Mechanistically, heparin binds to both, the chemoattractant SDF-1 and its receptor, chemokine receptor 4 (CXCR4), blocking CXCR4 internalization as well as SDF-1/CXCR4 signaling after SDF-1 stimulation. CONCLUSIONS: Heparin blocks SDF-1/CXCR4 signaling by binding to the ligand as well as the receptor, thereby interfering with migration and homing of BMCs. In contrast, the thrombin inhibitor bivalirudin did not interfere with BMC homing or SDF-1/CXCR4 signaling. These findings suggest that bivalirudin but not heparin might be recommended as an anticoagulant for intracoronary infusion of BMCs for cell therapy after cardiac ischemia.


Asunto(s)
Células de la Médula Ósea/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Quimiocina CXCL12/efectos de los fármacos , Heparina/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Infarto del Miocardio/terapia , Receptores CXCR4/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Anticoagulantes/farmacología , Antitrombinas/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Quimiocina CXCL12/fisiología , Modelos Animales de Enfermedad , Femenino , Hirudinas/farmacología , Humanos , Técnicas In Vitro , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Ratones , Ratones Endogámicos , Fragmentos de Péptidos/farmacología , Receptores CXCR4/fisiología , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología
9.
Blood ; 118(18): 5050-9, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21868579

RESUMEN

In human inflammatory diseases, we identified endothelial angiopoietin-2 (Ang-2) expression to be strongly associated with inflammations mediated by myeloid cells but not lymphocytes. To identify the underlying mechanism, we made use of a transgenic mouse model with inducible endothelial cell-specific expression of Ang-2. In this model, in the absence of inflammatory stimuli, long-term expression of Ang-2 led to a time-dependent accumulation of myeloid cells in numerous organs, suggesting that Ang-2 is sufficient to recruit myeloid cells. In models of acute inflammation, such as delayed-type hypersensitivity and peritonitis, Ang-2 transgenic animals showed an increased responsiveness. Intravital fluorescence video microscopy revealed augmented cell adhesion as an underlying event. Consequently, we demonstrated that Ang-2 is able to induce strong monocyte adhesion under shear in vitro, which could be blocked by antibodies to ß2-integrin. Taken together, our results describe Ang-2 as a novel, endothelial-derived regulator of myeloid cell infiltration that modulates ß2-integrin-mediated adhesion in a paracrine manner.


Asunto(s)
Angiopoyetina 2/fisiología , Antígenos CD18/fisiología , Movimiento Celular/genética , Células Mieloides/fisiología , Adulto , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Antígenos CD18/genética , Antígenos CD18/metabolismo , Adhesión Celular/genética , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/fisiología , Células Mieloides/metabolismo , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
J Cell Mol Med ; 16(10): 2387-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22353471

RESUMEN

Homing of endothelial progenitor cells (EPCs) is crucial for neoangiogenesis, which might be negatively affected by hypoxia. We investigated the influence of hypoxia on fibronectin binding integrins for migration and cell-matrix-adhesion. AMP-activated kinase (AMPK) and integrin-linked kinase (ILK) were examined as possible effectors of hypoxia. Human EPCs were expanded on fibronectin (FN) and integrin expression was profiled by flow cytometry. Cell-matrix-adhesion- and migration-assays on FN were performed to examine the influence of hypoxia and AMPK-activation. Regulation of AMPK and ILK was shown by Western blot analysis. We demonstrate the presence of integrin ß(1), ß(2) and α(5) on EPCs. Adhesion to FN is reduced by blocking ß(1) and α(5) (49% and 2% of control, P < 0.05) whereas α(4)-blockade has no effect. Corresponding effects were shown for migration. Hypoxia and AMPK-activation decrease adhesion on FN. Although total AMPK-expression remains unchanged, phospho-AMPK increases eightfold. The EPCs require α(5) for adhesion on FN. Hypoxia and AMPK-activation decrease adhesion. As α(5) is the major adhesive factor for EPCs on FN, this suggests a link between AMPK and α(5)-integrins. We found novel evidence for a connection between hypoxia, AMPK-activity and integrin activity. This might affect the fate of EPCs in ischaemic tissue.


Asunto(s)
Adhesión Celular , Células Endoteliales/citología , Integrinas/metabolismo , Células Madre/citología , Proteínas Quinasas Activadas por AMP/metabolismo , Hipoxia de la Célula , Línea Celular , Movimiento Celular/fisiología , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Humanos , Trasplante de Células Madre de Sangre Periférica , Fosforilación
11.
J Biol Chem ; 286(39): 34335-45, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21795701

RESUMEN

The semaphorins are a family of secreted or membrane-bound proteins that are known to guide axons in the developing nervous system. Genetic evidence revealed that a class III semaphorin, semaphorin 3E (Sema3E), and its receptor Plexin-D1 also control the vascular patterning during development. At the molecular level, we have recently shown that Sema3E acts on Plexin-D1 expressed in endothelial cells, thus initiating a novel antiangiogenic signaling pathway that results in the retraction of filopodia in endothelial tip cells. Sema3E induces the rapid disassembly of integrin-mediated adhesive structures, thereby inhibiting endothelial cell adhesion to the extracellular matrix. This process requires the activation of small GTPase Arf6 (ADP-ribosylation factor 6), which regulates intracellular trafficking of ß1 integrin. However, the molecular mechanisms by which Sema3E-Plexin-D1 activates Arf6 remained to be identified. Here we show that GEP100 (guanine nucleotide exchange protein 100)/Brag2, a guanine nucleotide exchange factor for Arf6, mediates Sema3E-induced Arf6 activation in endothelial cells. We provide evidence that upon activation by Sema3E, Plexin-D1 recruits phosphatidylinositol-4-phosphate 5-kinase, and its enzymatic lipid product, phosphatidylinositol 4,5-bisphosphate, binds to the pleckstrin homology domain of GEP100. Phosphatidylinositol 4,5-bisphosphate binding to GEP100 enhances its guanine nucleotide exchange factor activity toward Arf6, thus resulting in the disassembly of integrin-mediated focal adhesions and endothelial cell collapse. Our present study reveals a novel phospholipid-regulated antiangiogenic signaling pathway whereby Sema3E activates Arf6 through Plexin-D1 and consequently controls integrin-mediated endothelial cell attachment to the extracellular matrix and migration.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Endoteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Semaforinas/metabolismo , Transducción de Señal/fisiología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Animales , Células COS , Adhesión Celular/fisiología , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular/fisiología , Chlorocebus aethiops , Células Endoteliales/citología , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Integrinas/genética , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Semaforinas/genética
12.
Nat Med ; 11(2): 206-13, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15665831

RESUMEN

Infusion of endothelial progenitor cells (EPC), but not of mature endothelial cells, promotes neovascularization after ischemia. We performed gene expression profiling of EPC and endothelial cells to identify genes that might be important for the neovascularization capacity of EPC. Notably, the protease cathepsin L (CathL) was highly expressed in EPC as opposed to endothelial cells and was essential for matrix degradation and invasion by EPC in vitro. CathL-deficient mice showed impaired functional recovery following hind limb ischemia, supporting the concept of a crucial role for CathL in postnatal neovascularization. Infused CathL-deficient progenitor cells neither homed to sites of ischemia nor augmented neovascularization. Forced expression of CathL in mature endothelial cells considerably enhanced their invasive activity and sufficed to confer their capacity for neovascularization in vivo. We concluded that CathL has a critical role in the integration of circulating EPC into ischemic tissue and is required for EPC-mediated neovascularization.


Asunto(s)
Catepsinas/metabolismo , Cisteína Endopeptidasas/metabolismo , Células Endoteliales/fisiología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Biomarcadores , Catepsina L , Catepsinas/genética , Cisteína Endopeptidasas/genética , Células Endoteliales/citología , Perfilación de la Expresión Génica , Miembro Posterior/irrigación sanguínea , Miembro Posterior/fisiología , Humanos , Isquemia/metabolismo , Masculino , Ratones , Ratones Desnudos , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/citología
13.
J Exp Med ; 201(11): 1825-35, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15928198

RESUMEN

The regulation of acetylation is central for the epigenetic control of lineage-specific gene expression and determines cell fate decisions. We provide evidence that the inhibition of histone deacetylases (HDACs) blocks the endothelial differentiation of adult progenitor cells. To define the mechanisms by which HDAC inhibition prevents endothelial differentiation, we determined the expression of homeobox transcription factors and demonstrated that HoxA9 expression is down-regulated by HDAC inhibitors. The causal involvement of HoxA9 in the endothelial differentiation of adult progenitor cells is supported by the finding that HoxA9 overexpression partially rescued the endothelial differentiation blockade induced by HDAC inhibitors. Knockdown and overexpression studies revealed that HoxA9 acts as a master switch to regulate the expression of prototypical endothelial-committed genes such as endothelial nitric oxide synthase, VEGF-R2, and VE-cadherin, and mediates the shear stress-induced maturation of endothelial cells. Consistently, HoxA9-deficient mice exhibited lower numbers of endothelial progenitor cells and showed an impaired postnatal neovascularization capacity after the induction of ischemia. Thus, HoxA9 is regulated by HDACs and is critical for postnatal neovascularization.


Asunto(s)
Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/fisiología , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/biosíntesis , Animales , Antígenos CD , Cadherinas/metabolismo , Células Cultivadas , Células Endoteliales/citología , Sangre Fetal/citología , Sangre Fetal/fisiología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Humanos , Isquemia/metabolismo , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Estrés Mecánico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
J Exp Med ; 201(1): 63-72, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15623573

RESUMEN

The mechanisms of homing of endothelial progenitor cells (EPCs) to sites of ischemia are unclear. Here, we demonstrate that ex vivo-expanded EPCs as well as murine hematopoietic Sca-1+/Lin- progenitor cells express beta2-integrins, which mediate the adhesion of EPCs to endothelial cell monolayers and their chemokine-induced transendothelial migration in vitro. In a murine model of hind limb ischemia, Sca-1+/Lin- hematopoietic progenitor cells from beta2-integrin-deficient mice are less capable of homing to sites of ischemia and of improving neovascularization. Preactivation of the beta2-integrins expressed on EPCs by activating antibodies augments the EPC-induced neovascularization in vivo. These results provide evidence for a novel function of beta2-integrins in postnatal vasculogenesis.


Asunto(s)
Antígenos CD18/metabolismo , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Isquemia/fisiopatología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Animales , Antígenos CD18/fisiología , Adhesión Celular/fisiología , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Citometría de Flujo , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Humanos , Leucocitos Mononucleares , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/metabolismo
15.
Blood ; 113(2): 488-97, 2009 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18805968

RESUMEN

Ras-associated protein 1 (Rap1), a small GTPase, attracted attention because of its involvement in several aspects of cell adhesion, including integrin- and cadherin-mediated adhesion. Yet, the role of Rap1 genes and of Rap1 effectors for angiogenesis has not been investigated. Human umbilical vein endothelial cells (HUVECs) express Rap1a and Rap1b mRNA. To determine the contribution of Rap1 activity for angiogenesis, we overexpressed Rap1GAP1, a GTPase-activating protein that inhibits Rap1 activity. Overexpression of Rap1GAP1 significantly blocked angiogenic sprouting and tube-forming activity of HUVECs as well as migration and integrin-dependent adhesion. Silencing of Rap1a, Rap1b, or both significantly blocked HUVECs sprouting under basal and basic fibroblast growth factor-stimulated conditions and reduced HUVEC migration and integrin-dependent adhesion. We found that Rap1a and Rap1b are essential for the conformational activation of beta(1)-integrins in endothelial cells. Furthermore, silencing of Rap1a and Rap1b prevented phosphorylation of tyrosine 397 in focal adhesion kinase (FAK) and vascular endothelial growth factor-induced Akt1-activation. Rap1a(-/-)-deficient and Rap1a(+/-) heterozygote mice displayed reduced neovascularization after hind limb ischemia compared with wild-type mice. Silencing of RAPL significantly blocked the Rap1-induced sprouting of HUVECs, suggesting that the angiogenic activity of Rap1 is partly mediated by RAPL. Our data demonstrate a critical role of Rap1 in the regulation of beta(1)-integrin affinity, adhesion, and migration in endothelial cells and in postnatal neovascularization.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/enzimología , Integrina beta1/metabolismo , Neovascularización Fisiológica/fisiología , Venas Umbilicales/enzimología , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Adhesión Celular/fisiología , Células Endoteliales/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Silenciador del Gen , Miembro Posterior/irrigación sanguínea , Miembro Posterior/enzimología , Humanos , Isquemia/enzimología , Ratones , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt , Venas Umbilicales/citología
16.
Nat Commun ; 12(1): 3352, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099640

RESUMEN

Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.


Asunto(s)
Plaquetas/metabolismo , Factor Plaquetario 4/metabolismo , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Inductores de la Angiogénesis , Animales , Activación de Complemento , Complemento C5a , Inflamación , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptores CXCR3/genética , Transducción de Señal
17.
Basic Res Cardiol ; 105(6): 703-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20652278

RESUMEN

Diabetes is characterized by a chronic stage of hyperglycemia associated with endothelial progenitor cell dysfunction and reduced neovascularization in response to tissue ischemia. The underlying mechanisms are not entirely clear. The bone marrow niches provide the essential microenvironment for maintenance of stem cell function in the bone marrow. A disturbed stem cell niche might lead to stem cell dysfunction, thereby, impairing progenitor cell-dependent vascular repair. Therefore, we investigated the effects of streptozotocin-induced diabetes on the bone marrow stem cell niches and stem cell function in mice. Here, we show that long-term diabetes induced a reduction in Lin⁻Sca-1(+)c-kit(+) hematopoietic progenitor cells and reduced the repopulation capacity in a competitive engraftment experiment. Consistently, the expression of Bmi1, which prevents hematopoietic progenitor cell senescence, was significantly reduced in diabetic bone marrow cells. To address the mechanism underlying the progenitor cell dysfunction, we analyzed the composition of the stem cell niche and the cytokine environment. Although the morphology of the vascular and endosteal niche was not affected by diabetes, diabetic mice showed a significant deterioration of cytokine expression patterns in the bone marrow. In summary, these data indicate that diabetes imposes a long-term effect on the stem cell niche and affects important hematopoietic progenitor cell functions in mice.


Asunto(s)
Células de la Médula Ósea/patología , Proliferación Celular , Citocinas/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Células Madre Hematopoyéticas/patología , Nicho de Células Madre , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/inmunología , Trasplante de Médula Ósea , Diferenciación Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/fisiopatología , Células Endoteliales/inmunología , Células Endoteliales/patología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Miembro Posterior , Isquemia/inmunología , Isquemia/patología , Isquemia/fisiopatología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Proteínas Nucleares/metabolismo , Osteoblastos/inmunología , Osteoblastos/patología , Osteoclastos/inmunología , Osteoclastos/patología , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Represoras/metabolismo , Factores de Tiempo
18.
Circ Res ; 102(8): 942-9, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18323525

RESUMEN

Endothelial progenitor cells (EPCs) and hematopoietic progenitor cells are recruited to ischemic regions, improving neovascularization. beta1 and beta2 integrins play a crucial role for progenitor cell homing to ischemic tissues. Integrin activity is regulated by chemokines and their respective G protein-coupled receptors. The phosphatidylinositol-3-kinase catalytic subunit gamma (PI3Kgamma) is the PI3K isoform that selectively transduces signals from G protein-coupled receptors. Here, we investigated the role of PI3Kgamma as a signaling intermediate in the chemokine-induced integrin-dependent homing functions of progenitor cells. A pharmacological PI3Kgamma inhibitor significantly reduced chemokine-induced chemotaxis and stromal cell-derived factor (SDF)1alpha-induced transmigration of human EPCs. Moreover, the PI3Kgamma inhibitor significantly reduced SDF1alpha-induced adhesion of EPCs to intercellular adhesion molecule-1 and human umbilical vein endothelial cell monolayers. These findings were corroborated with Lin(-) bone marrow-derived progenitor cells from PI3Kgamma-deficient mice that displayed reduced SDF1alpha-induced migration and intercellular adhesion molecule-1 adhesion as compared with wild-type cells. Pharmacological inhibition or genetic ablation of PI3Kgamma reduced SDF1alpha-induced integrin activation in human EPCs and in murine Lin(-) BM-derived progenitor cells, respectively. In vivo, the homing of PI3Kgamma-deficient Lin(-) progenitor cells to ischemic muscles after intravenous infusion in the model of hindlimb ischemia and their neovascularization-promoting capacity was reduced as compared with wild-type cells. In conclusion, PI3Kgamma is integral to the integrin-dependent homing of progenitor cells.


Asunto(s)
Quimiocina CXCL12/fisiología , Quimiotaxis , Isquemia/patología , Fosfatidilinositol 3-Quinasas/fisiología , Células Madre/citología , Animales , Bovinos , Adhesión Celular , Moléculas de Adhesión Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib , Humanos , Integrinas/metabolismo , Isoenzimas/deficiencia , Isoenzimas/fisiología , Ratones , Ratones Noqueados , Músculo Esquelético/patología , Fosfatidilinositol 3-Quinasas/deficiencia , Transducción de Señal , Venas Umbilicales/citología
19.
Arterioscler Thromb Vasc Biol ; 29(4): 571-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19122169

RESUMEN

OBJECTIVE: Endothelial progenitor cells (EPCs) comprise a heterogeneous population of cells, which improve therapeutic neovascularization after ischemia. The neovascularization-promoting potential of progenitor cells depends on survival and retention of the infused cells to the tissue. Caspases mediate apoptosis but are also involved in other critical biological processes. Therefore, we aimed to address the role of caspases in proangiogenic cells. METHODS AND RESULTS: The caspase-8 inhibitor zIETD abrogated the ex vivo formation of EPCs, inhibited EPC adhesion and migration, and reduced their capacity to improve neovascularization in vivo. Consistently, cells isolated from caspase-8-deficient mice exhibited a reduced capacity for enhancing neovascularization when transplanted into mice after hindlimb ischemia. Because inhibition of Caspase-8 reduced the adhesion and homing functions of EPCs, we further determined the surface expression of integrins and receptors involved in cell recruitment to ischemic tissues. Pharmacological inhibition of caspase-8 and genetic depletion of caspase-8 reduced the expression of the fibronectin receptor subunits alpha5 and beta1 and the SDF-1 receptor CXCR4. Moreover, we identified the E3 ubiquitin ligase Cbl-b, which negatively regulates integrin and receptor-mediated signaling, as a potential Caspase-8 substrate. CONCLUSIONS: In summary, our data demonstrate a novel apoptosis-unrelated role of caspase-8 in proangiogenic cells.


Asunto(s)
Caspasa 8/metabolismo , Células Endoteliales/enzimología , Isquemia/enzimología , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Células Madre/enzimología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Síndrome de Alstrom , Animales , Caspasa 8/genética , Inhibidores de Caspasas , Adhesión Celular , Movimiento Celular , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/trasplante , Miembro Posterior , Humanos , Integrina alfaV/metabolismo , Integrina beta1/metabolismo , Isquemia/fisiopatología , Isquemia/cirugía , Ratones , Ratones Noqueados , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , ARN Mensajero/metabolismo , Receptores CXCR4/metabolismo , Receptores de Fibronectina/metabolismo , Trasplante de Células Madre , Células Madre/efectos de los fármacos
20.
Thromb Haemost ; 102(2): 191-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19652868

RESUMEN

Leukocyte recruitment plays a major role in the immune response to infectious pathogens and during inflammatory and autoimmune disorders. The process of leukocyte extravasation from the blood into the inflamed tissue requires a complex cascade of adhesive events between the leukocytes and the endothelium including leukocyte rolling, adhesion and transendothelial migration. Leukocyte-endothelial interactions are mediated by tightly regulated binding interactions between adhesion receptors on both cells. In this regard, leukocyte adhesion onto the endothelium is governed by leukocyte integrins and their endothelial counter-receptors of the immunoglobulin superfamily. The present review will focus on novel aspects with respect to the modulation of the leukocyte adhesion cascade.


Asunto(s)
Adhesión Celular/fisiología , Leucocitos/fisiología , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Moléculas de Adhesión Celular , Movimiento Celular/fisiología , Humanos , Integrinas/fisiología , Péptidos y Proteínas de Señalización Intercelular , Rodamiento de Leucocito/fisiología , Leucocitos/inmunología , Ratones , Modelos Biológicos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA