Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Circulation ; 127(4): 424-34, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23357661

RESUMEN

BACKGROUND: There is an unmet need for proangiogenic therapeutic molecules for the treatment of tissue ischemia in cardiovascular diseases. However, major inducers of angiogenesis such as vascular endothelial growth factor (VEGF/VEGF-A) have side effects that limit their therapeutic utility in vivo, especially at high concentrations. Angiopoietin-1 has been considered to be a blood vessel stabilization factor that can inhibit the intrinsic property of VEGF to promote vessel leakiness. In this study, we have designed and tested the angiogenic properties of chimeric molecules consisting of receptor-binding parts of VEGF and angiopoietin-1. We aimed at combining the activities of both factors into 1 molecule for easy delivery and expression in target tissues. METHODS AND RESULTS: The VEGF-angiopoietin-1 (VA1) chimeric protein bound to both VEGF receptor-2 and Tie2 and induced the activation of both receptors. Detailed analysis of VA1 versus VEGF revealed differences in the kinetics of VEGF receptor-2 activation and endocytosis, downstream kinase activation, and VE-cadherin internalization. The delivery of a VA1 transgene into mouse skeletal muscle led to increased blood flow and enhanced angiogenesis. VA1 was also very efficient in rescuing ischemic limb perfusion. However, VA1 induced less plasma protein leakage and myeloid inflammatory cell recruitment than VEGF. Furthermore, angioma-like structures associated with VEGF expression were not observed with VA1. CONCLUSIONS: The VEGF-angiopoietin-1 chimera is a potent angiogenic factor that triggers a novel mode of VEGF receptor-2 activation, promoting less vessel leakiness, less tissue inflammation, and better perfusion in ischemic muscle than VEGF. These properties of VA1 make it an attractive therapeutic tool.


Asunto(s)
Angiopoyetina 1/farmacología , Terapia Genética/métodos , Isquemia/tratamiento farmacológico , Neovascularización Fisiológica/fisiología , Proteínas Recombinantes de Fusión/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Adenoviridae/genética , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Permeabilidad Capilar/fisiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isquemia/genética , Leucemia Mieloide , Ratones , Ratones Endogámicos , Músculo Esquelético/irrigación sanguínea , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor TIE-2 , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
2.
Circ Res ; 111(4): 426-36, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22723300

RESUMEN

RATIONALE: Lymphatic vasculature plays important roles in tissue fluid homeostasis maintenance and in the pathology of human diseases. Yet, the molecular mechanisms that control lymphatic vessel maturation remain largely unknown. OBJECTIVE: We analyzed the gene expression profiles of ex vivo isolated lymphatic endothelial cells to identify novel lymphatic vessel expressed genes and we investigated the role of semaphorin 3A (Sema3A) and neuropilin-1 (Nrp-1) in lymphatic vessel maturation and function. METHODS AND RESULTS: Lymphatic and blood vascular endothelial cells from mouse intestine were isolated using fluorescence-activated cell sorting, and transcriptional profiling was performed. We found that the axonal guidance molecules Sema3A and Sema3D were highly expressed by lymphatic vessels. Importantly, we found that the semaphorin receptor Nrp-1 is expressed on the perivascular cells of the collecting lymphatic vessels. Treatment of mice in utero (E12.5-E16.5) with an antibody that blocks Sema3A binding to Nrp-1 but not with an antibody that blocks VEGF-A binding to Nrp-1 resulted in a complex phenotype of impaired lymphatic vessel function, enhanced perivascular cell coverage, and abnormal lymphatic vessel and valve morphology. CONCLUSIONS: Together, these results reveal an unanticipated role of Sema3A-Nrp-1 signaling in the maturation of the lymphatic vascular network likely via regulating the perivascular cell coverage of the vessels thus affecting lymphatic vessel function and lymphatic valve development.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/metabolismo , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Transducción de Señal , Animales , Anticuerpos Neutralizantes/administración & dosificación , Linaje de la Célula , Movimiento Celular , Separación Celular/métodos , Células Cultivadas , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Edad Gestacional , Humanos , Vasos Linfáticos/embriología , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Neuropilina-1/genética , Neuropilina-1/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Pericitos/metabolismo , Semaforina-3A/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Angiogenesis ; 16(3): 525-40, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23325334

RESUMEN

Preclinical vascular research has been hindered by a lack of methods that can sensitively image and quantify vascular perfusion and leakage in vivo. In this study, we have developed dynamic near-infrared imaging methods to repeatedly visualize and quantify vascular leakage in mouse skin in vivo, and we have applied these methods to transgenic mice with overexpression of vascular endothelial growth factors VEGF-A or -C. Near-infrared dye conjugates were developed to identify a suitable vascular tracer that had a prolonged circulation lifetime and slow leakage into normal tissue after intravenous injection. Dynamic simultaneous imaging of ear skin and a large blood vessel in the leg enabled determination of the intravascular signal (blood volume fraction) from the tissue signal shortly after injection and quantifications of vascular leakage into the extravascular tissue over time. This method allowed for the sensitive detection of increased blood vascularity and leakage rates in K14-VEGF-A transgenic mice and also reliably measured inflammation-induced changes of vascularity and leakage over time in the same mice. Measurements after injection of recombinant VEGF-A surprisingly revealed increased blood vascular leakage and lymphatic clearance in K14-VEGF-C transgenic mice which have an expanded cutaneous lymphatic vessel network, potentially indicating unanticipated effects of lymphatic drainage on vascular leakage. Increased vascular leakage was also detected in subcutaneous tumors, confirming that the method can also be applied to deeper tissues. This new imaging method might facilitate longitudinal investigations of the in vivo effects of drug candidates, including angiogenesis inhibitors, in preclinical disease models.


Asunto(s)
Síndrome de Fuga Capilar/diagnóstico , Síndrome de Fuga Capilar/patología , Diagnóstico por Imagen/métodos , Rayos Infrarrojos , Piel/patología , Análisis de Varianza , Animales , Permeabilidad Capilar/fisiología , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Dimetilsulfóxido , Femenino , Indoles/farmacocinética , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Polietilenglicoles , Espectrofotometría Ultravioleta , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo
4.
Blood ; 116(20): 4376-84, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20716773

RESUMEN

Podoplanin is a small transmembrane protein required for development and function of the lymphatic vascular system. To investigate the effects of interfering with its function, we produced an Fc fusion protein of its ectodomain. We found that podoplanin-Fc inhibited several functions of cultured lymphatic endothelial cells and also specifically suppressed lymphatic vessel growth, but not blood vessel growth, in mouse embryoid bodies in vitro and in mouse corneas in vivo. Using a keratin 14 expression cassette, we created transgenic mice that overexpressed podoplanin-Fc in the skin. No obvious outward phenotype was identified in these mice, but surprisingly, podoplanin-Fc-although produced specifically in the skin-entered the blood circulation and induced disseminated intravascular coagulation, characterized by microthrombi in most organs and by thrombocytopenia, occasionally leading to fatal hemorrhage. These findings reveal an important role of podoplanin in lymphatic vessel formation and indicate the potential of podoplanin-Fc as an inhibitor of lymphangiogenesis. These results also demonstrate the ability of podoplanin to induce platelet aggregation in vivo, which likely represents a major function of lymphatic endothelium. Finally, keratin 14 podoplanin-Fc mice represent a novel genetic animal model of disseminated intravascular coagulation.


Asunto(s)
Coagulación Intravascular Diseminada/fisiopatología , Linfangiogénesis/fisiología , Vasos Linfáticos/patología , Glicoproteínas de Membrana/metabolismo , Receptores Fc/metabolismo , Piel/metabolismo , Piel/patología , Animales , Coagulación Sanguínea , Plaquetas/metabolismo , Adhesión Celular , Línea Celular , Movimiento Celular , Córnea/patología , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/metabolismo , Coagulación Intravascular Diseminada/patología , Cuerpos Embrioides/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Queratina-14/metabolismo , Vasos Linfáticos/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Fisiológica , Activación Plaquetaria , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo
5.
Dev Dyn ; 240(8): 1989-2001, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21761482

RESUMEN

Classical cadherins are important cell adhesion molecules specifying and separating brain nuclei and developmental compartments. Cadherin-22 (Cdh22) belongs to type II subfamily of classical cadherins, and is expressed at the midbrain-hindbrain boundary during early embryogenesis. In Fgfr1 mutant mouse embryos, which have a disturbed midbrain-hindbrain border, Cdh22 is down-regulated. Here, we studied expression of Cdh22 in developing mouse brain in more detail and compared it to expression of related family members. This revealed both complementary and overlapping patterns of Cdh22, Cdh11, Cdh8, and Cdh6 expression in distinct regions of the forebrain and midbrain. We used a mutated allele of Cdh22 to study its function in brain development. Loss of Cdh22 caused reduced postnatal viability. Despite strong Cdh22 expression in the developing brain, we did not observe defects in compartmentalization or abnormalities in the midbrain and forebrain nuclei in Cdh22 mutants. This may be explained by functional redundancy between type II cadherins.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/embriología , Encéfalo/metabolismo , Cadherinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Cadherinas/genética , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Extremidades/anatomía & histología , Extremidades/embriología , Genotipo , Masculino , Ratones , Morfogénesis/fisiología , Mutación , Tasa de Supervivencia , Testículo/anatomía & histología , Testículo/embriología , Testículo/metabolismo
6.
Circ Res ; 104(11): 1302-12, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19443835

RESUMEN

The therapeutic potential of vascular endothelial growth factor (VEGF)-C and VEGF-D in skeletal muscle has been of considerable interest as these factors have both angiogenic and lymphangiogenic activities. Previous studies have mainly used adenoviral gene delivery for short-term expression of VEGF-C and VEGF-D in pig, rabbit, and mouse skeletal muscles. Here we have used the activated mature forms of VEGF-C and VEGF-D expressed via recombinant adeno-associated virus (rAAV), which provides stable, long-lasting transgene expression in various tissues including skeletal muscle. Mouse tibialis anterior muscle was transduced with rAAV encoding human or mouse VEGF-C or VEGF-D. Two weeks later, immunohistochemical analysis showed increased numbers of both blood and lymph vessels, and Doppler ultrasound analysis indicated increased blood vessel perfusion. The lymphatic vessels further increased at the 4-week time point were functional, as shown by FITC-lectin uptake and transport. Furthermore, receptor activation and arteriogenic activity were increased by an alanine substitution mutant of human VEGF-C (C137A) having an increased dimer stability and by a chimeric CAC growth factor that contained the VEGF receptor-binding domain flanked by VEGF-C propeptides, but only the latter promoted significantly more blood vessel perfusion when compared to the other growth factors studied. We conclude that long-term expression of VEGF-C and VEGF-D in skeletal muscle results in the generation of new functional blood and lymphatic vessels. The therapeutic value of intramuscular lymph vessels in draining tissue edema and lymphedema can now be evaluated using this model system.


Asunto(s)
Vasos Sanguíneos/fisiología , Corazón/fisiología , Vasos Linfáticos/fisiología , Músculo Esquelético/fisiología , Factor B de Crecimiento Endotelial Vascular/fisiología , Factor C de Crecimiento Endotelial Vascular/fisiología , Animales , Dimerización , Estabilidad de Medicamentos , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/irrigación sanguínea , Mutación , Polimorfismo de Nucleótido Simple , Proteínas Recombinantes/metabolismo , Factor B de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
7.
Mol Metab ; 4(2): 93-105, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25685697

RESUMEN

OBJECTIVE: Elevated serum levels of the lymphangiogenic factors VEGF-C and -D have been observed in obese individuals but their relevance for the metabolic syndrome has remained unknown. METHODS: K14-VEGFR-3-Ig (sR3) mice that constitutively express soluble-VEGFR-3-Ig in the skin, scavenging VEGF-C and -D, and wildtype (WT) mice were fed either chow or high-fat diet for 20 weeks. To assess the effect of VEGFR-3 blockage on adipose tissue growth and insulin sensitivity, we evaluated weight gain, adipocyte size and hepatic lipid accumulation. These results were complemented with insulin tolerance tests, FACS analysis of adipose tissue macrophages, in vitro 3T3-L1 differentiation assays and in vivo blocking antibody treatment experiments. RESULTS: We show here that sR3 mice are protected from obesity-induced insulin resistance and hepatic lipid accumulation. This protection is associated with enhanced subcutaneous adipose tissue hyperplasia and an increased number of alternatively-activated (M2) macrophages in adipose tissue. We also show that VEGF-C and -D are chemotactic for murine macrophages and that this effect is mediated by VEGFR-3, which is upregulated on M1 polarized macrophages. Systemic antibody blockage of VEGFR-3 in db/db mice reduces adipose tissue macrophage infiltration and hepatic lipid accumulation, and improves insulin sensitivity. CONCLUSIONS: These results reveal an unanticipated role of the lymphangiogenic factors VEGF-C and -D in the mediation of metabolic syndrome-associated adipose tissue inflammation. Blockage of these lymphangiogenic factors might constitute a new therapeutic strategy for the prevention of obesity-associated insulin resistance.

8.
Cancer Res ; 73(14): 4212-21, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695550

RESUMEN

VEGF-C and VEGF-D were identified as lymphangiogenic growth factors and later shown to promote tumor metastasis, but their effects on carcinogenesis are poorly understood. Here, we have studied the effects of VEGF-C and VEGF-D on tumor development in the murine multistep chemical carcinogenesis model of squamous cell carcinoma by using a soluble VEGF-C/VEGF-D inhibitor. After topical treatment with a tumor initiator and repeated tumor promoter applications, transgenic mice expressing a soluble VEGF-C/VEGF-D receptor (sVEGFR-3) in the skin developed significantly fewer squamous cell tumors with a delayed onset when compared with wild-type mice or mice expressing sVEGFR-3 lacking the ligand-binding site. Epidermal proliferation was reduced in the carcinogen-treated transgenic skin, whereas epidermal keratinocyte proliferation in vitro was not affected by VEGF-C or VEGF-D, indicating indirect effects of sVEGFR-3 expression. Importantly, transgenic mouse skin was less sensitive to tumor promoter-induced inflammation, with reduced angiogenesis and blood vessel leakage. Cutaneous leukocytes, especially macrophages, were reduced in transgenic skin without major changes in macrophage polarization or blood monocyte numbers. Several macrophage-associated cytokines were also reduced in transgenic papillomas, although the dermal macrophages themselves did not express VEGFR-3. These findings indicate that VEGF-C/VEGF-D are involved in shaping the inflammatory tumor microenvironment that regulates early tumor progression. Our results support the use of VEGF-C/VEGF-D-blocking agents not only to inhibit metastatic progression, but also during the early stages of tumor growth.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Piel/efectos de los fármacos , Factor C de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor D de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Carcinógenos , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Epidermis/patología , Femenino , Inflamación/metabolismo , Inflamación/patología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/patología , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor D de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
J Biol Chem ; 284(23): 16037-48, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19366703

RESUMEN

Members of the vascular endothelial growth factor (VEGF) family play a pivotal role in angiogenesis and lymphangiogenesis. They are potential therapeutics to induce blood vessel formation in myocardium and skeletal muscle, when normal blood flow is compromised. Most members of the VEGF/platelet derived growth factor protein superfamily exist as covalently bound antiparallel dimers. However, the mature form of VEGF-D (VEGF-D(DeltaNDeltaC)) is predominantly a non-covalent dimer even though the cysteine residues (Cys-44 and Cys-53) forming the intersubunit disulfide bridges in the other members of the VEGF family are also conserved in VEGF-D. Moreover, VEGF-D bears an additional cysteine residue (Cys-25) at the subunit interface. Guided by our model of VEGF-D(DeltaNDeltaC), the cysteines at the subunit interface were mutated to study the effect of these residues on the structural and functional properties of VEGF-D(DeltaNDeltaC). The conserved cysteines Cys-44 and Cys-53 were found to be essential for the function of VEGF-D(DeltaNDeltaC). More importantly, the substitution of the Cys-25 at the dimer interface by various amino acids improved the activity of the recombinant VEGF-D(DeltaNDeltaC) and increased the dimer to monomer ratio. Specifically, substitutions to hydrophobic amino acids Ile, Leu, and Val, equivalent to those found in other VEGFs, most favorably affected the activity of the recombinant VEGF-D(DeltaNDeltaC). The increased activity of these mutants was mainly due to stabilization of the protein. This study enables us to better understand the structural determinants controlling the biological activity of VEGF-D. The novel variants of VEGF-D(DeltaNDeltaC) described here are potential agents for therapeutic applications, where induction of vascular formation is required.


Asunto(s)
Factor D de Crecimiento Endotelial Vascular/genética , Animales , Sitios de Unión , Velocidad del Flujo Sanguíneo , Circulación Coronaria , Variación Genética , Corazón/fisiología , Homeostasis , Humanos , Modelos Moleculares , Músculo Esquelético/irrigación sanguínea , Miocardio/metabolismo , Neovascularización Fisiológica , Conformación Proteica , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Relación Estructura-Actividad , Porcinos , Factor D de Crecimiento Endotelial Vascular/química , Factor D de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA