Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 327
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Science ; 373(6554)2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34326210

RESUMEN

Aging is an established risk factor for vascular diseases, but vascular aging itself may contribute to the progressive deterioration of organ function. Here, we show in aged mice that vascular endothelial growth factor (VEGF) signaling insufficiency, which is caused by increased production of decoy receptors, may drive physiological aging across multiple organ systems. Increasing VEGF signaling prevented age-associated capillary loss, improved organ perfusion and function, and extended life span. Healthier aging was evidenced by favorable metabolism and body composition and amelioration of aging-associated pathologies including hepatic steatosis, sarcopenia, osteoporosis, "inflammaging" (age-related multiorgan chronic inflammation), and increased tumor burden. These results indicate that VEGF signaling insufficiency affects organ aging in mice and suggest that modulating this pathway may result in increased mammalian life span and improved overall health.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento Saludable , Longevidad , Factor A de Crecimiento Endotelial Vascular/metabolismo , Tejido Adiposo , Animales , Vasos Sanguíneos/fisiología , Composición Corporal , Distribución de la Grasa Corporal , Metabolismo de los Hidratos de Carbono , Carcinogénesis , Endotelio Vascular/metabolismo , Hígado Graso/patología , Femenino , Inflamación/prevención & control , Hígado/patología , Masculino , Ratones , Densidad Microvascular , Microvasos/fisiología , Osteoporosis/prevención & control , Consumo de Oxígeno , Sarcopenia/prevención & control , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/sangre
2.
Am J Transplant ; 16(4): 1160-72, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26689983

RESUMEN

Organ damage and innate immunity during heart transplantation may evoke adaptive immunity with serious consequences. Because lymphatic vessels bridge innate and adaptive immunity, they are critical in immune surveillance; however, their role in ischemia-reperfusion injury (IRI) in allotransplantation remains unknown. We investigated whether the lymphangiogenic VEGF-C/VEGFR3 pathway during cardiac allograft IRI regulates organ damage and subsequent interplay between innate and adaptive immunity. We found that cardiac allograft IRI, within hours, increased graft VEGF-C expression and lymphatic vessel activation in the form of increased lymphatic VEGFR3 and adhesion protein expression. Pharmacological VEGF-C/VEGFR3 stimulation resulted in early lymphatic activation and later increase in allograft inflammation. In contrast, pharmacological VEGF-C/VEGFR3 inhibition during cardiac allograft IRI decreased early lymphatic vessel activation with subsequent dampening of acute and chronic rejection. Genetic deletion of VEGFR3 specifically in the lymphatics of the transplanted heart recapitulated the survival effect achieved by pharmacological VEGF-C/VEGFR3 inhibition. Our results suggest that tissue damage rapidly changes lymphatic vessel phenotype, which, in turn, may shape the interplay of innate and adaptive immunity. Importantly, VEGF-C/VEGFR3 inhibition during solid organ transplant IRI could be used as lymphatic-targeted immunomodulatory therapy to prevent acute and chronic rejection.


Asunto(s)
Células Endoteliales/metabolismo , Rechazo de Injerto/metabolismo , Trasplante de Corazón , Daño por Reperfusión/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Aloinjertos , Animales , Células Endoteliales/patología , Rechazo de Injerto/patología , Supervivencia de Injerto/inmunología , Inflamación/metabolismo , Inflamación/patología , Linfangiogénesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Daño por Reperfusión/fisiopatología , Donantes de Tejidos , Factor C de Crecimiento Endotelial Vascular/metabolismo
3.
Am J Transplant ; 15(8): 2075-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25932532

RESUMEN

The major cause of death during the first year after heart transplantation is primary graft dysfunction due to preservation and ischemia-reperfusion injury (IRI). Angiopoietin-1 is a Tie2 receptor-binding paracrine growth factor with anti-inflammatory properties and indispensable roles in vascular development and stability. We used a stable variant of angiopoietin-1 (COMP-Ang1) to test whether ex vivo intracoronary treatment with a single dose of COMP-Ang1 in donor Dark Agouti rat heart subjected to 4-h cold ischemia would prevent microvascular dysfunction and inflammatory responses in the fully allogeneic recipient Wistar Furth rat. COMP-Ang1 reduced endothelial cell-cell junction disruption of the donor heart in transmission electron microscopy during 4-h cold ischemia, improved myocardial reflow, and reduced microvascular leakage and cardiomyocyte injury of transplanted allografts during IRI. Concurrently, the treatment reduced expression of danger signals, dendritic cell maturation markers, endothelial cell adhesion molecule VCAM-1 and RhoA/Rho-associated protein kinase activation and the influx of macrophages and neutrophils. Furthermore, COMP-Ang1 treatment provided sustained anti-inflammatory effects during acute rejection and prevented the development of cardiac fibrosis and allograft vasculopathy. These results suggest donor heart treatment with COMP-Ang1 having important clinical implications in the prevention of primary and subsequent long-term injury and dysfunction in cardiac allografts.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Corazón , Proteínas Recombinantes de Fusión/uso terapéutico , Daño por Reperfusión/prevención & control , Donantes de Tejidos , Animales , Ratas
4.
Sci Rep ; 4: 6238, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25168313

RESUMEN

Vascular endothelial growth factor-B (VEGF-B) has been implicated to play a significant role in coronary vessel growth and endothelial uptake and transport of fatty acids in heart and skeletal muscle. Additionally, recent studies have shown that Vegf-b deficiency protects from high-fat diet (HFD)-induced diabetes and insulin resistance. We compared the cardiac function and the effects of HFD on body composition and glucose metabolism in two available Vegf-b knockout (Vegf-b(-/-) strains) mouse strains side by side with their respective littermate controls. We found no differences in HFD-induced weight gain, glucose tolerance or insulin resistance between the Vegf-b(-/-) strains and their littermate control mice. Furthermore, there was no difference in basal cardiac function and cardiac expression of genes involved in glucose or fatty acid metabolism between the Vegf-b(-/-) strains and their littermate control mice. We conclude that VEGF-B is dispensable for normal cardiac function under unstressed conditions and for HFD-induced metabolic changes.


Asunto(s)
Enfermedades Cardiovasculares/genética , Dieta Alta en Grasa/efectos adversos , Factor B de Crecimiento Endotelial Vascular/genética , Animales , Enfermedades Cardiovasculares/etiología , Expresión Génica , Técnicas de Inactivación de Genes , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transcriptoma , Factor B de Crecimiento Endotelial Vascular/metabolismo
5.
Am J Transplant ; 14(5): 1096-108, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24708486

RESUMEN

Transplant ischemia-reperfusion injury (Tx-IRI) and allograft dysfunction remain as two of the major clinical challenges after heart transplantation. We investigated the role of angiopoietin-2 (Ang2) in Tx-IRI and rejection using fully MHC-mismatched rat cardiac allografts. We report that plasma levels of Ang2 were significantly enhanced in the human and rat recipients of cardiac allografts, but not in the rat recipients of syngrafts, during IRI. Ex vivo intracoronary treatment of rat cardiac allografts with anti-Ang2 antibody before 4-h cold preservation prevented microvascular dysfunction, endothelial cell (EC) adhesion molecule expression and leukocyte infiltration, myocardial injury and the development of cardiac fibrosis and allograft vasculopathy. Recipient preoperative and postoperative treatment with anti-Ang2 antibody produced otherwise similar effects without effecting microvascular dysfunction, and in additional experiments prolonged allograft survival. Recipient preoperative treatment alone failed to produce these effects. Moreover, ex vivo intracoronary treatment of allografts with recombinant Ang2 enhanced Tx-IRI and, in an add-back experiment, abolished the beneficial effect of the antibody. We demonstrate that neutralization of Ang2 prevents EC activation, leukocyte infiltration, Tx-IRI and the development of chronic rejection in rat cardiac allografts. Our results suggest that blocking Ang2 pathway is a novel, clinically feasible, T cell-independent strategy to protect cardiac allografts.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Rechazo de Injerto/prevención & control , Trasplante de Corazón , Complicaciones Posoperatorias/prevención & control , Daño por Reperfusión/prevención & control , Adulto , Anciano , Aloinjertos , Angiopoyetina 2/sangre , Angiopoyetina 2/inmunología , Animales , Muerte Encefálica , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Rechazo de Injerto/etiología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto , Cardiopatías/cirugía , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/metabolismo , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Adulto Joven
6.
Arterioscler Thromb Vasc Biol ; 33(2): 266-74, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23288156

RESUMEN

OBJECTIVE: The pathophysiology of lymphedema is incompletely understood. We asked how transcapillary fluid balance parameters and lymph flow are affected in a transgenic mouse model of primary lymphedema, which due to an inhibition of vascular endothelial growth factor receptor-3 (VEGFR-3) signaling lacks dermal lymphatics, and whether protein accumulation in the interstitium occurring in lymphedema results in inflammation. METHODS AND RESULTS: As estimated using a new optical-imaging technique, we found that this signaling defect resulted in lymph drainage in hind limb skin of K14-VEGFR-3-Ig mice that was 34% of the corresponding value in wild-type. The interstitial fluid pressure and tissue fluid volumes were significantly increased in the areas of visible swelling only, whereas the colloid osmotic pressure in plasma, and thus the colloid osmotic pressure gradient, was reduced compared to wild-type mice. An acute volume load resulted in an exaggerated interstitial fluid pressure response in transgenic mice. There was no accumulation of collagen or lipid in skin, suggesting that chronic edema presented in the K14-VEGFR-3-Ig mouse was not sufficient to induce changes in tissue composition. Proinflammatory cytokines (interleukin-2, interleukin-6, interleukin-12) in subcutaneous interstitial fluid and macrophage infiltration in skin of the paw were lower, whereas the monocyte/macrophage cell fraction in blood and spleen was higher in transgenic compared with wild-type mice. CONCLUSIONS: Our data suggest that a high interstitial protein concentration and longstanding edema is not sufficient to induce fibrosis and inflammation characteristic for the human condition and may have implications for our understanding of the pathophysiology of this condition.


Asunto(s)
Dermatitis Alérgica por Contacto/metabolismo , Líquido Extracelular/metabolismo , Vasos Linfáticos/metabolismo , Linfedema/metabolismo , Piel/metabolismo , Animales , Colágeno/metabolismo , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/patología , Dermatitis Alérgica por Contacto/fisiopatología , Modelos Animales de Enfermedad , Femenino , Fibrosis , Genotipo , Mediadores de Inflamación/metabolismo , Interleucina-12/metabolismo , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Vasos Linfáticos/fisiopatología , Linfedema/genética , Linfedema/inmunología , Linfedema/patología , Linfedema/fisiopatología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presión Osmótica , Permeabilidad , Fenotipo , Proteínas/metabolismo , Albúmina Sérica/metabolismo , Piel/inmunología , Piel/patología , Piel/fisiopatología , Factores de Tiempo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Desequilibrio Hidroelectrolítico/inmunología , Desequilibrio Hidroelectrolítico/metabolismo , Desequilibrio Hidroelectrolítico/patología
7.
J Virol ; 86(8): 4463-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301147

RESUMEN

Fibroblasts can be reprogrammed into induced pluripotent stem cells (iPSC) by ectopic expression of key transcription factors. Current methods for the generation of integration-free iPSC are limited by the low efficiency of iPSC generation and by challenges in reprogramming methodology. Recombinant adeno-associated virus (rAAV) is a potent gene delivery vehicle capable of efficient transduction of transgenic DNA into cells. rAAV stays mainly as an episome in nondividing cells, and the extent of integration is still poorly defined for various replicating cells. In this study, we aimed to induce iPSC from mouse and human fibroblasts by using rAAV vector-mediated transient delivery of reprogramming factors. We succeeded in deriving induced pluripotent stem cells from mouse but not human fibroblasts. Unexpectedly, the rAAV vector-mediated reprogramming led to frequent genomic integration of vector sequences during the reprogramming process, independent of the amount of virus used, and to persistent expression of reprogramming factors in generated iPSC clones. It thus appears that rAAV vectors are not compatible with the derivation of integration-free iPSC.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Transducción Genética , Integración Viral , Animales , Diferenciación Celular , Línea Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Br J Cancer ; 105(9): 1346-51, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21970873

RESUMEN

BACKGROUND: PROX1 is a specific target of the ß-catenin/TCF pathway in the intestinal epithelium. It acts as a regulator of progression from a benign to a highly dysplastic phenotype in colorectal tumours. However, the clinical significance of PROX1 expression is not known. METHODS: We studied the prognostic value of immunohistochemical expression of PROX1 in a series of 517 patients with colorectal cancer (CRC). RESULTS: The majority of the tumour samples expressed PROX1 (91%, 471 out of 517). High PROX1 expression was associated with a poor grade of tumour differentiation (P<0.0001). In the subgroup of patients with colon cancer, high PROX1 expression was associated with unfavourable colorectal cancer-specific survival (CCSS) as compared with low PROX1 expression (CCSS 47% vs 62%; P=0.045; RR 1.47). The association between high PROX1 and poor outcome was further strengthened in female colon cancer patients (CCSS 38% vs 63%; P=0.007; RR 2.02). Nonetheless, in multivariate survival analysis PROX1 expression was not retained as an independent prognostic factor. CONCLUSION: High PROX1 expression is associated with a poor grade of tumour differentiation, and, in colon cancer patients, also with less favourable patient outcome. Our results strengthen the previous preclinical observations that PROX1 has a role in tumour progression in CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/mortalidad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Factores de Transcripción/metabolismo
9.
Neuroscience ; 192: 550-63, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21767614

RESUMEN

Neurotrophic factors regulate the development and maintenance of the nervous system and protect and repair dopaminergic neurons in animal models of Parkinson's disease (PD). Vascular endothelial growth factors A (VEGF-A) and B have also neurotrophic effects on various types of neurons, including dopaminergic neurons. We examined the ability of the key lymphangiogenic factor VEGF-C to protect dopaminergic cells in vitro and in vivo. The study was initiated by a finding from microarray profiling of Neuro2A-20 cells which revealed up-regulation of VEGF-C by glial cell-line-derived neurotrophic factor (GDNF). Next, we observed that VEGF-C can rescue embryonic dopaminergic neurons and activate the mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) pathway in vivo. VEGF receptors 1-2 and co-receptors, neuropilins 1-2, were expressed both in mouse embryonic cultures and adult midbrains. In vivo, VEGF-C had a robust functional effect in the rat unilateral 6-hydroxydopamine (6-OHDA) model of PD and there was a small additive effect on the survival of tyrosine hydroxylase (TH)-positive cells with GDNF. The neuroprotective effect of VEGF-C is most likely due to a combination of direct and indirect neurotrophic effects because, VEGF-C, unlike GDNF, induced also angiogenesis in the striatum following 6-OHDA insult as it did in human umbilical vein endothelial cells (HUVEC). However, we detected activation of astroglia and microglia as well as blood-brain barrier disruption after intracerebral delivery of VEGF-C, raising a concern of its safe usage as a therapeutic molecule. Our results provide evidence of VEGF-C as a neurotrophic factor that influences the dopaminergic system through multiple mechanisms.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Western Blotting , Supervivencia Celular , Dopamina/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Ratones , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Cancer Gene Ther ; 18(2): 100-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20865022

RESUMEN

Despite optimal surgery and chemotherapy, the prognosis of ovarian cancer patients remains poor and new treatments are urgently needed. Solid tumors require the formation of new vessels for growth and metastasis. In the present study, we have used soluble vascular endothelial growth factor (sVEGF) receptors sVEGFR-1 and -3, soluble receptors Tie1 and Tie2 and their combinations in an ovarian cancer xenograft model. Human ovarian cancer cells were injected intraperitoneally into nude mice (n=42) and magnetic resonance imaging (MRI) was used for confirming tumors before gene delivery. Treatment with combined AdsVEGFR-1, AdsVEGFR-3 and AdsTie2 significantly decreased the size of the intraperitoneal tumors compared with the controls (AdLacZ; P=0.038) with significantly less microvessels and vascular area. Unexpectedly, treatment with combined AdsTie1 and AdsTie2 led to a dramatic shortening of the survival which was not observed in the groups receiving either of the soluble receptors alone (P=0.031). The only difference to other treatments was liver toxicity observed after the combined Tie receptor treatment. In conclusion, combined inhibition of VEGFR-1, VEGFR-3 and Tie2 pathways was safe and provided efficient therapy for ovarian cancer in mice.


Asunto(s)
Terapia Genética/métodos , Neoplasias Ováricas/terapia , Receptor TIE-2/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Ascitis/patología , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Femenino , Humanos , Linfangiogénesis/genética , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Med Genet ; 46(6): 399-404, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19289394

RESUMEN

BACKGROUND: Heterozygous mutations in VEGFR3 have been identified in some familial cases with dominantly inherited primary congenital lymphoedema, known as Nonne-Milroy disease. Recessive cases of primary lymphoedema with a genetic cause are not known, except for two families with syndromic hypotrichosis-lymphoedema-telangiectasia, with a SOX18 mutation. METHODS AND RESULTS: In this study, we present the first case of isolated primary congenital lymphoedema with recessive inheritance, caused by a homozygous mutation in VEGFR3. The novel mutation is a transition from alanine-to-threonine in amino acid 855, located in the ATP binding domain of the VEGFR3 receptor. Assessment of receptor function showed impaired ligand induced internalisation and ERK1/2 activity. Moreover, receptor phosphorylation was reduced, although less so than for a kinase-dead VEGFR3 mutation, which causes Nonne-Milroy disease. CONCLUSION: A hypomorphic VEGFR3 mutation, with moderate effect on receptor function, in a homozygous state can result in insufficient lymphatic functioning. Thus, in addition to Nonne-Milroy disease with dominant inheritance, VEGFR3 alterations can cause isolated recessive primary congenital lymphoedema. These data expand our understanding of the aetiology of congenital lymphoedema and suggest that large scale screening of VEGFR3 in all primary lymphoedema patients is necessary.


Asunto(s)
Genes Recesivos , Linfedema/congénito , Linfedema/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Linaje , Alineación de Secuencia , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Transplant Proc ; 38(10): 3271-3, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17175246

RESUMEN

BACKGROUND: In chronic rejection, parenchymal fibrosis and cardiac allograft vasculopathy (CAV) characterized by neointimal growth are the leading causes of graft loss for heart transplant recipients. During alloimmune responses a variety of cytokines, adhesion proteins, and growth factors, such as platelet-derived growth factor (PDGF), are up-regulated. The PDGF family (AA, AB, BB, CC, DD), which acts mainly on connective tissue cells, is considered to be a potent mitogenic and chemotactic factor for fibroblasts and vascular smooth muscle cells. In this study, we evaluated the effects of PDGF ligands in chronic rejection. METHODS: Heterotopic heart transplantations were performed between fully major histocompatability complex-mismatched Dark Agouti to Wistar Furth rats receiving cyclosporine immunosuppression. Allograft coronary arteries were perfused with a recombinant adeno-associated virus (AAV) encoding enhanced green fluorescence protein (EGFP) as a control gene or PDGF-A, -B, -C, -D. Allografts were harvested at 100 days for morphometric analysis of CAV and fibrosis. RESULTS: AAV-mediated transgene expression was detected by EGFP immunoreactivity across the graft section (at 100 days) in AAV EGFP-perfused allografts. AAV PDGF-A, -C, and -D perfusion resulted in accelerated CAV and fibrosis. In contrast, AAV PDGF-B perfusion did not induce arteriosclerotic changes or fibrosis in cardiac allografts. CONCLUSIONS: AAV PDGF-A, -C, and -D overexpression accelerated the development of chronic rejection, whereas PDGF-B did not. Our results suggested that more targeted therapy with monoclonal antibodies blocking the active sites of PDGF-A, -C, and -D may produce beneficial effects on heart transplant survival.


Asunto(s)
Enfermedad Coronaria/patología , Trasplante de Corazón/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Complicaciones Posoperatorias/patología , Animales , Dependovirus/genética , Fibrosis , Ratas , Ratas Endogámicas , Ratas Endogámicas WF , Trasplante Homólogo , Enfermedades Vasculares
13.
Gene Ther ; 12(5): 388-94, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15647771

RESUMEN

This study was performed to evaluate angiogenic responses of angiopoietin-1 (Ang1) in vivo after adenovirus-mediated gene transfer in the periadventitial space of the rabbit carotid arteries using a collar technique. Adenoviruses encoding LacZ and vascular endothelial growth factor (VEGF) receptor-1-Ig fusion protein (VEGF-R1-Ig) adenoviruses were used as controls. Increased neovessel formation was seen in adventitia of the Ang1 transduced arteries 7 days after the gene transfer. Neovessels in the Ang1 transduced arteries were large in size and well perfused. Ang1 binds to Tie2 (tyrosine kinase with immunoglobulin and epidermal growth factor homology domain) receptors, which were expressed in the endothelium of the neovessels. When VEGF-R1-Ig was used with Ang1, it resulted in a decrease in the number of neovessels, which implies that VEGF-A or some other VEGF-R1 ligand(s) play a crucial role in angiogenesis occurring in response to Ang1. There were no significant differences in the total number of capillaries in the adventitia of the VEGF-R1-Ig transduced arteries as compared to LacZ controls. Neointima formation was not increased in the Ang1 transduced arteries as compared to the controls. We conclude that in the periadventitial space Ang1 shows angiogenic activity and is a potentially useful factor for the induction of therapeutic vascular growth in vivo.


Asunto(s)
Angiopoyetina 1/genética , Arterias Carótidas , Terapia Genética/métodos , Neovascularización Fisiológica , Transducción Genética/métodos , Adenoviridae/genética , Animales , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Inmunoglobulinas/genética , Inyecciones , Conejos , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Transgenes , Túnica Íntima/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
15.
Mech Dev ; 109(1): 69-77, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11677054

RESUMEN

Several signalling pathways have been defined by studies of genes originally characterised in Drosophila. However, some mammalian signalling systems have so far escaped discovery in the fly. Here, we describe the identification and characterisation of fly homologs for the mammalian vascular endothelial growth factor/platelet derived growth factor (VEGF/PDGF) and the VEGF receptor. The Drosophila factor (DmVEGF-1) gene has two splice variants and is expressed during all stages, the signal distribution during embryogenesis being ubiquitous. The receptor (DmVEGFR) gene has several splice variants; the variations affecting only the extracellular domain. The most prominent form is expressed in cells of the embryonic haematopoietic cell lineage, starting in the mesodermal area of the head around stage 10 of embryogenesis. Expression persists in hemocytes as embryonic development proceeds and the cells migrate posteriorly. In a fly strain carrying a deletion uncovering the DmVEGFR gene, hemocytes are still present, but their migration is hampered and the hemocytes remain mainly in the anterior end close to their origin. These data suggest that the VEGF/PDGF signalling system may regulate the migration of the Drosophila embryonic haemocyte precursor cells.


Asunto(s)
Empalme Alternativo , Drosophila melanogaster/genética , Factores de Crecimiento Endotelial/genética , Expresión Génica , Genes de Insecto , Hemocitos/citología , Linfocinas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Drosophila melanogaster/embriología , Factores de Crecimiento Endotelial/clasificación , Humanos , Linfocinas/clasificación , Datos de Secuencia Molecular , Filogenia , Proteínas Tirosina Quinasas Receptoras/clasificación , Receptores de Factores de Crecimiento/clasificación , Receptores de Factores de Crecimiento Endotelial Vascular , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
16.
Proc Natl Acad Sci U S A ; 98(22): 12677-82, 2001 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-11592985

RESUMEN

Primary human lymphedema (Milroy's disease), characterized by a chronic and disfiguring swelling of the extremities, is associated with heterozygous inactivating missense mutations of the gene encoding vascular endothelial growth factor C/D receptor (VEGFR-3). Here, we describe a mouse model and a possible treatment for primary lymphedema. Like the human patients, the lymphedema (Chy) mice have an inactivating Vegfr3 mutation in their germ line, and swelling of the limbs because of hypoplastic cutaneous, but not visceral, lymphatic vessels. Neuropilin (NRP)-2 bound VEGF-C and was expressed in the visceral, but not in the cutaneous, lymphatic endothelia, suggesting that it may participate in the pathogenesis of lymphedema. By using virus-mediated VEGF-C gene therapy, we were able to generate functional lymphatic vessels in the lymphedema mice. Our results suggest that growth factor gene therapy is applicable to human lymphedema and provide a paradigm for other diseases associated with mutant receptors.


Asunto(s)
Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/genética , Terapia Genética , Linfedema/terapia , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Dependovirus/genética , Factores de Crecimiento Endotelial/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/análisis , Neuropilina-1 , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Factor C de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular
17.
EMBO J ; 20(17): 4762-73, 2001 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-11532940

RESUMEN

Vascular endothelial growth factor receptor-3 (VEGFR-3/Flt4) binds two known members of the VEGF ligand family, VEGF-C and VEGF-D, and has a critical function in the remodelling of the primary capillary vasculature of midgestation embryos. Later during development, VEGFR-3 regulates the growth and maintenance of the lymphatic vessels. In the present study, we have isolated and cultured stable lineages of blood vascular and lymphatic endothelial cells from human primary microvascular endothelium by using antibodies against the extracellular domain of VEGFR-3. We show that VEGFR-3 stimulation alone protects the lymphatic endothelial cells from serum deprivation-induced apoptosis and induces their growth and migration. At least some of these signals are transduced via a protein kinase C-dependent activation of the p42/p44 MAPK signalling cascade and via a wortmannin-sensitive induction of Akt phosphorylation. These results define the critical role of VEGF-C/VEGFR-3 signalling in the growth and survival of lymphatic endothelial cells. The culture of isolated lymphatic endothelial cells should now allow further studies of the molecular properties of these cells.


Asunto(s)
Apoptosis/fisiología , Factores de Crecimiento Endotelial/farmacología , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/fisiología , Endotelio/fisiología , Sistema Linfático/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Apoptosis/efectos de los fármacos , Técnicas Biosensibles , Capilares/embriología , Capilares/fisiología , División Celular , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Endotelio/citología , Endotelio Vascular/citología , Activación Enzimática , Humanos , Cinética , Sistema Linfático/citología , Microcirculación/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Piel/irrigación sanguínea , Venas Umbilicales , Factor C de Crecimiento Endotelial Vascular , Factor D de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular
18.
Cell Microbiol ; 3(9): 623-32, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11553014

RESUMEN

Bartonella henselae causes the vasculoproliferative disorders bacillary angiomatosis (BA) and bacillary peliosis (BP). The pathomechanisms of these tumorous proliferations are unknown. Our results suggest a novel bacterial two-step pathogenicity strategy, in which the pathogen triggers growth factor production for subsequent proliferation of its own host cells. In fact, B. henselae induces host cell production of the angiogenic factor vascular endothelial growth factor (VEGF), leading to proliferation of endothelial cells. The presence of B. henselae pili was associated with host cell VEGF production, as a Pil- mutant of B. henselae was unable to induce VEGF production. In turn, VEGF-stimulated endothelial cells promoted the growth of B. henselae. Immunohistochemistry for VEGF in specimens from patients with BA or BP revealed increased VEGF expression in vivo. These findings suggest a novel bacteria-dependent mechanism of tumour growth.


Asunto(s)
Bartonella henselae/patogenicidad , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/microbiología , Linfocinas/metabolismo , Angiomatosis Bacilar/metabolismo , Angiomatosis Bacilar/patología , Bartonella henselae/fisiología , División Celular , Línea Celular , Medios de Cultivo Condicionados , Endotelio Vascular/metabolismo , Fimbrias Bacterianas/metabolismo , Humanos , Inmunohistoquímica , Peliosis Hepática/metabolismo , Peliosis Hepática/patología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
19.
Am J Pathol ; 159(3): 893-903, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11549582

RESUMEN

Interactions of tumor cells with lymphatic vessels are of paramount importance for tumor progression, however, the underlying molecular mechanisms are poorly understood. Whereas enlarged lymphatic vessels are frequently observed at the periphery of malignant melanomas, it has remained unclear whether intratumoral lymphangiogenesis occurs within these tumors. Here, we demonstrate the presence of intratumoral lymphatics and enlargement of lymphatic vessels at the tumor periphery in vascular endothelial growth factor (VEGF)-C-overexpressing human melanomas transplanted onto nude mice. VEGF-C expression also resulted in enhanced tumor angiogenesis, indicating a coordinated regulation of lymphangiogenesis and angiogenesis in melanoma progression. The specific biological effects of VEGF-C were critically dependent on its proteolytic processing in vivo. Furthermore, VEGF-C induced chemotaxis of macrophages in vitro and in vivo, revealing a potential function of VEGF-C as an immunomodulator. Taken together, our results identify VEGF-C as multifunctional factor involved in regulating tumor lymphangiogenesis, angiogenesis, and immune response.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Sistema Linfático/patología , Melanoma/metabolismo , Melanoma/patología , Animales , División Celular/fisiología , Movimiento Celular/fisiología , Factores de Crecimiento Endotelial/fisiología , Humanos , Sistema Linfático/crecimiento & desarrollo , Macrófagos/fisiología , Melanoma/irrigación sanguínea , Melanoma/fisiopatología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/fisiopatología , Células Tumorales Cultivadas , Factor C de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA