Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Cachexia Sarcopenia Muscle ; 9(3): 514-529, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29722201

RESUMEN

BACKGROUND: Cancer cachexia increases morbidity and mortality, and blocking of activin receptor ligands has improved survival in experimental cancer. However, the underlying mechanisms have not yet been fully uncovered. METHODS: The effects of blocking activin receptor type 2 (ACVR2) ligands on both muscle and non-muscle tissues were investigated in a preclinical model of cancer cachexia using a recombinant soluble ACVR2B (sACVR2B-Fc). Treatment with sACVR2B-Fc was applied either only before the tumour formation or with continued treatment both before and after tumour formation. The potential roles of muscle and non-muscle tissues in cancer cachexia were investigated in order to understand the possible mechanisms of improved survival mediated by ACVR2 ligand blocking. RESULTS: Blocking of ACVR2 ligands improved survival in tumour-bearing mice only when the mice were treated both before and after the tumour formation. This occurred without effects on tumour growth, production of pro-inflammatory cytokines or the level of physical activity. ACVR2 ligand blocking was associated with increased muscle (limb and diaphragm) mass and attenuation of both hepatic protein synthesis and splenomegaly. Especially, the effects on the liver and the spleen were observed independent of the treatment protocol. The prevention of splenomegaly by sACVR2B-Fc was not explained by decreased markers of myeloid-derived suppressor cells. Decreased tibialis anterior, diaphragm, and heart protein synthesis were observed in cachectic mice. This was associated with decreased mechanistic target of rapamycin (mTOR) colocalization with late-endosomes/lysosomes, which correlated with cachexia and reduced muscle protein synthesis. CONCLUSIONS: The prolonged survival with continued ACVR2 ligand blocking could potentially be attributed in part to the maintenance of limb and respiratory muscle mass, but many observed non-muscle effects suggest that the effect may be more complex than previously thought. Our novel finding showing decreased mTOR localization in skeletal muscle with lysosomes/late-endosomes in cancer opens up new research questions and possible treatment options for cachexia.


Asunto(s)
Receptores de Activinas Tipo II/farmacología , Caquexia/metabolismo , Hígado/efectos de los fármacos , Proteínas Recombinantes/farmacología , Bazo/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Receptores de Activinas Tipo II/uso terapéutico , Animales , Biomarcadores , Caquexia/tratamiento farmacológico , Caquexia/etiología , Caquexia/patología , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación , Estimación de Kaplan-Meier , Hígado/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/mortalidad , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Bazo/inmunología , Bazo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Neoplasia ; 18(7): 436-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27435926

RESUMEN

Vascular endothelial growth factor D (VEGF-D) promotes the lymph node metastasis of cancer by inducing the growth of lymphatic vasculature, but its specific roles in tumorigenesis have not been elucidated. We monitored the effects of VEGF-D in cutaneous squamous cell carcinoma (cSCC) by subjecting transgenic mice overexpressing VEGF-D in the skin (K14-mVEGF-D) and VEGF-D knockout mice to a chemical skin carcinogenesis protocol involving 7,12-dimethylbenz[a]anthracene and 12-O-tetradecanoylphorbol-13-acetate treatments. In K14-mVEGF-D mice, tumor lymphangiogenesis was significantly increased and the frequency of lymph node metastasis was elevated in comparison with controls. Most notably, the papillomas regressed more often in K14-mVEGF-D mice than in littermate controls, resulting in a delay in tumor incidence and a remarkable reduction in the total tumor number. Skin tumor growth and metastasis were not obviously affected in the absence of VEGF-D; however, the knockout mice showed a trend for reduced lymphangiogenesis in skin tumors and in the untreated skin. Interestingly, K14-mVEGF-D mice showed an altered immune response in skin tumors. This consisted of the reduced accumulation of macrophages, mast cells, and CD4(+) T-cells and an increase of cytotoxic CD8(+) T-cells. Cytokine profiling by flow cytometry and quantitative real time PCR revealed that elevated VEGF-D expression results in an attenuated Th2 response and promotes M1/Th1 and Th17 polarization in the early stage of skin carcinogenesis, leading to an anti-tumoral immune environment and the regression of primary tumors. Our data suggest that VEGF-D may be beneficial in early-stage tumors since it suppresses the pro-tumorigenic inflammation, while at later stages VEGF-D-induced tumor lymphatics provide a route for metastasis.


Asunto(s)
Carcinógenos/toxicidad , Carcinoma de Células Escamosas/patología , Ganglios Linfáticos/irrigación sanguínea , Linfangiogénesis/efectos de los fármacos , Neoplasias Cutáneas/patología , Factor D de Crecimiento Endotelial Vascular/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Escamosas/inducido químicamente , Citometría de Flujo , Metástasis Linfática , Vasos Linfáticos/patología , Recuento de Linfocitos , Macrófagos/inmunología , Mastocitos/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Acetato de Tetradecanoilforbol/toxicidad
3.
Proc Natl Acad Sci U S A ; 112(42): 13063-8, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26430242

RESUMEN

Cardiac hypertrophy accompanies many forms of heart disease, including ischemic disease, hypertension, heart failure, and valvular disease, and it is a strong predictor of increased cardiovascular morbidity and mortality. Deletion of bone marrow kinase in chromosome X (Bmx), an arterial nonreceptor tyrosine kinase, has been shown to inhibit cardiac hypertrophy in mice. This finding raised the possibility of therapeutic use of Bmx tyrosine kinase inhibitors, which we have addressed here by analyzing cardiac hypertrophy in gene-targeted mice deficient in Bmx tyrosine kinase activity. We found that angiotensin II (Ang II)-induced cardiac hypertrophy is significantly reduced in mice deficient in Bmx and in mice with inactivated Bmx tyrosine kinase compared with WT mice. Genome-wide transcriptomic profiling showed that Bmx inactivation suppresses myocardial expression of genes related to Ang II-induced inflammatory and extracellular matrix responses whereas expression of RNAs encoding mitochondrial proteins after Ang II administration was maintained in Bmx-inactivated hearts. Very little or no Bmx mRNA was expressed in human cardiomyocytes whereas human cardiac endothelial cells expressed abundant amounts. Ang II stimulation of endothelial cells increased Bmx phosphorylation, and Bmx gene silencing inhibited downstream STAT3 signaling, which has been implicated in cardiac hypertrophy. Furthermore, activation of the mechanistic target of rapamycin complex 1 pathway by Ang II treatment was decreased in the Bmx-deficient hearts. Our results demonstrate that inhibition of the cross-talk between endothelial cells and cardiomyocytes by Bmx inactivation suppresses Ang II-induced signals for cardiac hypertrophy. These results suggest that the endothelial Bmx tyrosine kinase could provide a target to attenuate the development of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/enzimología , Endotelio Vascular/enzimología , Proteínas Tirosina Quinasas/metabolismo , Angiotensina II/farmacología , Animales , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/enzimología , Transducción de Señal
4.
J Clin Pathol ; 68(8): 614-21, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26021331

RESUMEN

AIMS: To build and evaluate an automated method for assessing tumour viability in histological tissue samples using texture features and supervised learning. METHODS: H&E-stained sections (n=56) of human non-small cell lung adenocarcinoma xenografts were digitised with a whole-slide scanner. A novel image analysis method based on local binary patterns and a support vector machine classifier was trained with a set of sample regions (n=177) extracted from the whole-slide images and tested with another set of images (n=494). The extracted regions, or single-tissue entity images, were chosen to represent as pure as possible examples of three morphological tissue entities: viable tumour tissue, non-viable tumour tissue and mouse host tissue. RESULTS: An agreement of 94.5% (area under the curve=0.995, kappa=0.90) was achieved to classify the single-tissue entity images in the test set (n=494) into the viable tumour and non-viable tumour tissue categories. The algorithm assigned 250 of the 252 non-viable and 219 of the 242 of viable sample regions to the correct categories, respectively. This corresponds to a sensitivity of 90.5% and specificity of 99.2%. CONCLUSIONS: The proposed image analysis-based tumour viability assessment resulted in a high agreement with expert annotations. By providing extraction of detailed information of the tumour microenvironment, the automated method can be used in preclinical research settings. The method could also have implications in cancer diagnostics, cancer outcome prognostics and prediction.


Asunto(s)
Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Interpretación de Imagen Asistida por Computador/métodos , Neoplasias Pulmonares/patología , Coloración y Etiquetado/métodos , Adenocarcinoma del Pulmón , Algoritmos , Animales , Área Bajo la Curva , Inteligencia Artificial , Automatización de Laboratorios , Línea Celular Tumoral , Supervivencia Celular , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Valor Predictivo de las Pruebas , Curva ROC , Reproducibilidad de los Resultados , Microambiente Tumoral
5.
Cancer Res ; 75(10): 2083-94, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808867

RESUMEN

Lymphatic invasion and accumulation of continuous collagen bundles around tumor cells are associated with poor melanoma prognosis, but the underlying mechanisms and molecular determinants have remained unclear. We show here that a copy-number gain or overexpression of the membrane-type matrix metalloproteinase MMP16 (MT3-MMP) is associated with poor clinical outcome, collagen bundle assembly around tumor cell nests, and lymphatic invasion. In cultured WM852 melanoma cells derived from human melanoma metastasis, silencing of MMP16 resulted in cell-surface accumulation of the MMP16 substrate MMP14 (MT1-MMP) as well as L1CAM cell adhesion molecule, identified here as a novel MMP16 substrate. When limiting the activities of these trans-membrane protein substrates toward pericellular collagen degradation, cell junction disassembly, and blood endothelial transmigration, MMP16 supported nodular-type growth of adhesive collagen-surrounded melanoma cell nests, coincidentally steering cell collectives into lymphatic vessels. These results uncover a novel mechanism in melanoma pathogenesis, whereby restricted collagen infiltration and limited mesenchymal invasion are unexpectedly associated with the properties of the most aggressive tumors, revealing MMP16 as a putative indicator of adverse melanoma prognosis.


Asunto(s)
Colágeno/metabolismo , Metaloproteinasa 16 de la Matriz/fisiología , Melanoma/enzimología , Neoplasias Cutáneas/enzimología , Animales , Células COS , Adhesión Celular , Chlorocebus aethiops , Matriz Extracelular/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Estimación de Kaplan-Meier , Ganglios Linfáticos/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Melanoma/mortalidad , Melanoma/secundario , Metalotioneína 3 , Ratones Endogámicos ICR , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Proteolisis , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología
6.
Cell Rep ; 8(6): 1943-1956, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25242330

RESUMEN

Colorectal cancer (CRC) initiation and growth is often attributed to stem cells, yet little is known about the regulation of these cells. We show here that a subpopulation of Prox1-transcription-factor-expressing cells have stem cell activity in intestinal adenomas, but not in the normal intestine. Using in vivo models and 3D ex vivo organoid cultures of mouse adenomas and human CRC, we found that Prox1 deletion reduced the number of stem cells and cell proliferation and decreased intestinal tumor growth via induction of annexin A1 and reduction of the actin-binding protein filamin A, which has been implicated as a prognostic marker in CRC. Loss of Prox1 also decreased autophagy and the survival of hypoxic tumor cells in tumor transplants. Thus, Prox1 is essential for the expansion of the stem cell pool in intestinal adenomas and CRC without being critical for the normal functions of the gut.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Anexina A1/antagonistas & inhibidores , Anexina A1/genética , Anexina A1/metabolismo , Autofagia , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Filaminas/antagonistas & inhibidores , Filaminas/genética , Filaminas/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Isquemia/patología , Isquemia/prevención & control , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Trasplante Heterólogo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
7.
Genes Dev ; 28(14): 1592-603, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25030698

RESUMEN

Primitive lymphatic vessels are remodeled into functionally specialized initial and collecting lymphatics during development. Lymphatic endothelial cell (LEC) junctions in initial lymphatics transform from a zipper-like to a button-like pattern during collecting vessel development, but what regulates this process is largely unknown. Angiopoietin 2 (Ang2) deficiency leads to abnormal lymphatic vessels. Here we found that an ANG2-blocking antibody inhibited embryonic lymphangiogenesis, whereas endothelium-specific ANG2 overexpression induced lymphatic hyperplasia. ANG2 inhibition blocked VE-cadherin phosphorylation at tyrosine residue 685 and the concomitant formation of button-like junctions in initial lymphatics. The defective junctions were associated with impaired lymph uptake. In collecting lymphatics, adherens junctions were disrupted, and the vessels leaked upon ANG2 blockade or gene deletion. ANG2 inhibition also suppressed the onset of lymphatic valve formation and subsequent valve maturation. These data identify ANG2 as the first essential regulator of the functionally important interendothelial cell-cell junctions that form during lymphatic development.


Asunto(s)
Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , Uniones Intercelulares/fisiología , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/genética , Animales , Cadherinas/metabolismo , Embrión de Mamíferos , Células Endoteliales/citología , Eliminación de Gen , Linfangiogénesis/fisiología , Tejido Linfoide/embriología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Fosforilación
8.
Circulation ; 129(19): 1962-71, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24552833

RESUMEN

BACKGROUND: Hennekam lymphangiectasia-lymphedema syndrome (Online Mendelian Inheritance in Man 235510) is a rare autosomal recessive disease, which is associated with mutations in the CCBE1 gene. Because of the striking phenotypic similarity of embryos lacking either the Ccbe1 gene or the lymphangiogenic growth factor Vegfc gene, we searched for collagen- and calcium-binding epidermal growth factor domains 1 (CCBE1) interactions with the vascular endothelial growth factor-C (VEGF-C) growth factor signaling pathway, which is critical in embryonic and adult lymphangiogenesis. METHODS AND RESULTS: By analyzing VEGF-C produced by CCBE1-transfected cells, we found that, whereas CCBE1 itself does not process VEGF-C, it promotes proteolytic cleavage of the otherwise poorly active 29/31-kDa form of VEGF-C by the A disintegrin and metalloprotease with thrombospondin motifs-3 protease, resulting in the mature 21/23-kDa form of VEGF-C, which induces increased VEGF-C receptor signaling. Adeno-associated viral vector-mediated transduction of CCBE1 into mouse skeletal muscle enhanced lymphangiogenesis and angiogenesis induced by adeno-associated viral vector-VEGF-C. CONCLUSIONS: These results identify A disintegrin and metalloprotease with thrombospondin motifs-3 as a VEGF-C-activating protease and reveal a novel type of regulation of a vascular growth factor by a protein that enhances its proteolytic cleavage and activation. The results suggest that CCBE1 is a potential therapeutic tool for the modulation of lymphangiogenesis and angiogenesis in a variety of diseases that involve the lymphatic system, such as lymphedema or lymphatic metastasis.


Asunto(s)
Proteínas ADAM/metabolismo , Proteínas de Unión al Calcio/metabolismo , Linfangiogénesis/fisiología , Procolágeno N-Endopeptidasa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Proteínas ADAMTS , Adenoviridae/genética , Animales , Proteínas de Unión al Calcio/genética , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos , Modelos Animales , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neovascularización Fisiológica/fisiología , Transfección , Proteínas Supresoras de Tumor/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
J Clin Invest ; 124(2): 824-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24430181

RESUMEN

The endothelial Tie1 receptor is ligand-less, but interacts with the Tie2 receptor for angiopoietins (Angpt). Angpt2 is expressed in tumor blood vessels, and its blockade inhibits tumor angiogenesis. Here we found that Tie1 deletion from the endothelium of adult mice inhibits tumor angiogenesis and growth by decreasing endothelial cell survival in tumor vessels, without affecting normal vasculature. Treatment with VEGF or VEGFR-2 blocking antibodies similarly reduced tumor angiogenesis and growth; however, no additive inhibition was obtained by targeting both Tie1 and VEGF/VEGFR-2. In contrast, treatment of Tie1-deficient mice with a soluble form of the extracellular domain of Tie2, which blocks Angpt activity, resulted in additive inhibition of tumor growth. Notably, Tie1 deletion decreased sprouting angiogenesis and increased Notch pathway activity in the postnatal retinal vasculature, while pharmacological Notch suppression in the absence of Tie1 promoted retinal hypervasularization. Moreover, substantial additive inhibition of the retinal vascular front migration was observed when Angpt2 blocking antibodies were administered to Tie1-deficient pups. Thus, Tie1 regulates tumor angiogenesis, postnatal sprouting angiogenesis, and endothelial cell survival, which are controlled by VEGF, Angpt, and Notch signals. Our results suggest that targeting Tie1 in combination with Angpt/Tie2 has the potential to improve antiangiogenic therapy.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Células Endoteliales/citología , Endotelio Vascular/metabolismo , Eliminación de Gen , Receptor TIE-1/genética , Vasos Retinianos/patología , Inhibidores de la Angiogénesis/química , Angiopoyetina 1/metabolismo , Animales , Apoptosis , Supervivencia Celular , Homocigoto , Ligandos , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Neovascularización Patológica , Fenotipo , Receptor TIE-1/fisiología , Receptor TIE-2/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Sci Signal ; 6(282): ra52, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23821770

RESUMEN

Vascular endothelial growth factors (VEGFs) regulate blood and lymphatic vessel development through VEGF receptors (VEGFRs). The VEGFR immunoglobulin homology domain 2 (D2) is critical for ligand binding, and D3 provides additional interaction sites. VEGF-B and placenta growth factor (PlGF) bind to VEGFR-1 with high affinity, but only PlGF is angiogenic in most tissues. We show that VEGF-B, unlike other VEGFs, did not require D3 interactions for high-affinity binding. VEGF-B with a PlGF-derived L1 loop (B-L1P) stimulated VEGFR-1 activity, whereas PlGF with a VEGF-B-derived L1 loop (P-L1B) did not. Unlike P-L1B and VEGF-B, B-L1P and PlGF were also angiogenic in mouse skeletal muscle. Furthermore, B-L1P also bound to VEGFR-2 and activated downstream signaling. These results establish a role for L1-mediated D3 interactions in VEGFR activation in endothelial cells and indicate that VEGF-B is a high-affinity VEGFR-1 ligand that, unlike PlGF, cannot efficiently induce signaling downstream of VEGFR-1.


Asunto(s)
Proteínas Gestacionales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Unión Competitiva , Western Blotting , Línea Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Células 3T3 NIH , Fosforilación , Factor de Crecimiento Placentario , Proteínas Gestacionales/química , Proteínas Gestacionales/genética , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/genética , Factor B de Crecimiento Endotelial Vascular/química , Factor B de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
12.
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
13.
Cancer Res ; 72(14): 3512-21, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22593188

RESUMEN

Bmx, [corrected] also known as Etk, is a member of the Tec family of nonreceptor tyrosine kinases. Bmx is expressed mainly in arterial endothelia and in myeloid hematopoietic cells. Bmx regulates ischemia-mediated arteriogenesis and lymphangiogenesis, but its role in tumor angiogenesis is not known. In this study, we characterized the function of Bmx in tumor growth using both Bmx knockout and transgenic mice. Isogenic colon, lung, and melanoma tumor xenotransplants showed reductions in growth and tumor angiogenesis in Bmx gene-deleted ((-/-)) mice, whereas developmental angiogenesis was not affected. In addition, growth of transgenic pancreatic islet carcinomas and intestinal adenomas was also slower in Bmx(-/-) mice. Knockout mice showed high levels of Bmx expression in endothelial cells of tumor-associated and peritumoral arteries. Moreover, endothelial cells lacking Bmx showed impaired phosphorylation of extracellular signal-regulated kinase (Erk) upon VEGF stimulation, indicating that Bmx contributes to the transduction of vascular endothelial growth factor signals. In transgenic mice overexpressing Bmx in epidermal keratinocytes, tumors induced by a two-stage chemical skin carcinogenesis treatment showed increased growth and angiogenesis. Our findings therefore indicate that Bmx activity contributes to tumor angiogenesis and growth.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Neoplasias del Colon/irrigación sanguínea , Melanoma Experimental/irrigación sanguínea , Neovascularización Patológica/genética , Proteínas Tirosina Quinasas/genética , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Femenino , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Ratones Transgénicos
14.
J Natl Cancer Inst ; 104(6): 461-75, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22343031

RESUMEN

BACKGROUND: Angiopoietin-2 (Ang2), a ligand for endothelial TEK (Tie2) tyrosine kinase receptor, is induced in hypoxic endothelial cells of tumors, where it promotes tumor angiogenesis and growth. However, the effects of Ang2 on tumor lymphangiogenesis and metastasis are poorly characterized. METHODS: We addressed the effect of Ang2 on tumor progression and metastasis using systemic Ang2 overexpression in mice carrying tumor xenografts, endothelium-specific overexpression of Ang2 in VEC-tTA/Tet-OS-Ang2 transgenic mice implanted with isogenic tumors, and administration of Ang2-blocking antibodies to tumor-bearing immunodeficient mice. Fisher's exact test was used for analysis of metastasis occurrence, and repeated measures one-way analysis of variance was used for the analysis of primary tumor growth curves. Unpaired t test was used for all other analyses. All statistical tests were two-sided. RESULTS: Adenoviral expression of Ang2 increased lymph node and lung metastasis in tumor xenografts. The metastatic burden in the lungs was increased in transgenic mice in which Ang2 expression was induced specifically in the vascular endothelium (tumor burden per grid, VEC-tTA/Tet-OS-Ang2 mice [n = 5] vs control mice [n = 4]: 45.23 vs 12.26 mm(2), difference = 32.67 mm(2), 95% confidence interval = 31.87 to 34.07, P < .001). Ang2-blocking antibodies reduced lymph node and lung metastasis, as well as tumor lymphangiogenesis, and decreased tumor cell homing to the lungs after intravenous injection. In the lung metastases, Ang2 overexpression decreased endothelial integrity, whereas the Ang2-blocking antibodies improved endothelial cell-cell junctions and basement membrane contacts of metastasis-associated lung capillaries. At the cellular level, the Ang2-blocking antibodies induced the internalization of Ang2-Tie2 receptor complexes from endothelial cell-cell junctions in endothelial-tumor cell cocultures. CONCLUSION: Our results indicate that blocking Ang2 inhibits metastatic dissemination in part by enhancing the integrity of endothelial cell-cell junctions.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/metabolismo , Endotelio Vascular/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Animales , Anticuerpos Bloqueadores/metabolismo , Anticuerpos Bloqueadores/farmacología , Hipoxia de la Célula , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/patología , Femenino , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Uniones Intercelulares/patología , Neoplasias Pulmonares/secundario , Linfangiogénesis , Metástasis Linfática , Melanoma/irrigación sanguínea , Melanoma/secundario , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Neovascularización Patológica/tratamiento farmacológico , Receptor TIE-2/metabolismo , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Trasplante Heterólogo , Regulación hacia Arriba
15.
Blood ; 118(4): 1154-62, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21566091

RESUMEN

Notch signaling plays a central role in cell-fate determination, and its role in lateral inhibition in angiogenic sprouting is well established. However, the role of Notch signaling in lymphangiogenesis, the growth of lymphatic vessels, is poorly understood. Here we demonstrate Notch pathway activity in lymphatic endothelial cells (LECs), as well as induction of delta-like ligand 4 (Dll4) and Notch target genes on stimulation with VEGF or VEGF-C. Suppression of Notch signaling by a soluble form of Dll4 (Dll4-Fc) synergized with VEGF in inducing LEC sprouting in 3-dimensional (3D) fibrin gel assays. Expression of Dll4-Fc in adult mouse ears promoted lymphangiogenesis, which was augmented by coexpressing VEGF. Lymphangiogenesis triggered by Notch inhibition was suppressed by a monoclonal VEGFR-2 Ab as well as soluble VEGF and VEGF-C/VEGF-D ligand traps. LECs transduced with Dll4 preferentially adopted the tip cell position over nontransduced cells in 3D sprouting assays, suggesting an analogous role for Dll4/Notch in lymphatic and blood vessel sprouting. These results indicate that the Notch pathway controls lymphatic endothelial quiescence, and explain why LECs are poorly responsive to VEGF compared with VEGF-C. Understanding the role of the Notch pathway in lymphangiogenesis provides further insight for the therapeutic manipulation of the lymphatic vessels.


Asunto(s)
Linfangiogénesis/fisiología , Vasos Linfáticos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Western Blotting , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Surg Oncol ; 103(6): 484-8, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21480240

RESUMEN

Tumor-associated neovascularization allows tumor cells to express their critical growth advantage, whereas lymphatic invasion is crucial for the metastatic process. Various growth factors stimulate blood and lymphatic neovascularization and modulate vessel permeability in tumors. The first anti-angiogenic drugs are already in routine use, and new anti-vascular therapeutics are evaluated in clinical trials. Conversely, pro-lymphangiogenic therapy could be implemented to treat cancer survivors suffering from secondary lymphedema.


Asunto(s)
Linfangiogénesis , Neoplasias/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Inhibidores de la Angiogénesis/uso terapéutico , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Sci Transl Med ; 3(69): 69ra11, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21307301

RESUMEN

The dissemination of tumor cells to sites far from the primary tumor (metastasis) is the principal cause of death in cancer patients. Tumor-associated lymphatic vessels are a key conduit for metastatic tumor cells, which typically first colonize the lymph nodes. Although the primary tumor and affected lymph nodes can be removed during surgery, tumor cells inside lymphatic vessels are left behind. Here, we show that in-transit tumor cells inside lymphatic vessels in mice bearing mouse melanomas or human lung tumors give rise to metastases. Using photodynamic therapy with the benzoporphyrin derivative verteporfin, we selectively destroyed lymphatic vessels in mice and pigs. Destruction of tumor-associated lymphatic vessels also eradicated intralymphatic tumor cells and prevented metastasis of mouse melanoma cells and subsequent relapse. Photodynamic therapy, when combined with anti-lymphangiogenic therapy, prevented further tumor invasion of lymphatic vessels. These findings highlight the potential of targeting in-transit tumor cells in patients.


Asunto(s)
Metástasis Linfática/prevención & control , Vasos Linfáticos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Animales , Humanos , Melanoma/tratamiento farmacológico , Ratones , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/uso terapéutico , Porcinos , Verteporfina
18.
Cancer Cell ; 18(6): 630-40, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21130043

RESUMEN

Antibodies that block vascular endothelial growth factor (VEGF) have become an integral part of antiangiogenic tumor therapy, and antibodies targeting other VEGFs and receptors (VEGFRs) are in clinical trials. Typically receptor-blocking antibodies are targeted to the VEGFR ligand-binding site. Here we describe a monoclonal antibody that inhibits VEGFR-3 homodimer and VEGFR-3/VEGFR-2 heterodimer formation, signal transduction, as well as ligand-induced migration and sprouting of microvascular endothelial cells. Importantly, we show that combined use of antibodies blocking ligand binding and receptor dimerization improves VEGFR inhibition and results in stronger inhibition of endothelial sprouting and vascular network formation in vivo. These results suggest that receptor dimerization inhibitors could be used to enhance antiangiogenic activity of antibodies blocking ligand binding in tumor therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Multimerización de Proteína , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Células Cultivadas , Humanos , Morfogénesis , Receptor ErbB-2/química , Factor C de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Circulation ; 122(17): 1725-33, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20937974

RESUMEN

BACKGROUND: Vascular endothelial growth factor-B (VEGF-B) binds to VEGF receptor-1 and neuropilin-1 and is abundantly expressed in the heart, skeletal muscle, and brown fat. The biological function of VEGF-B is incompletely understood. METHODS AND RESULTS: Unlike placenta growth factor, which binds to the same receptors, adeno-associated viral delivery of VEGF-B to mouse skeletal or heart muscle induced very little angiogenesis, vascular permeability, or inflammation. As previously reported for the VEGF-B(167) isoform, transgenic mice and rats expressing both isoforms of VEGF-B in the myocardium developed cardiac hypertrophy yet maintained systolic function. Deletion of the VEGF receptor-1 tyrosine kinase domain or the arterial endothelial Bmx tyrosine kinase inhibited hypertrophy, whereas loss of VEGF-B interaction with neuropilin-1 had no effect. Surprisingly, in rats, the heart-specific VEGF-B transgene induced impressive growth of the epicardial coronary vessels and their branches, with large arteries also seen deep inside the subendocardial myocardium. However, VEGF-B, unlike other VEGF family members, did not induce significant capillary angiogenesis, increased permeability, or inflammatory cell recruitment. CONCLUSIONS: VEGF-B appears to be a coronary growth factor in rats but not in mice. The signals for the VEGF-B-induced cardiac hypertrophy are mediated at least in part via the endothelium. Because cardiomyocyte damage in myocardial ischemia begins in the subendocardial myocardium, the VEGF-B-induced increased arterial supply to this area could have therapeutic potential in ischemic heart disease.


Asunto(s)
Permeabilidad Capilar/fisiología , Vasos Coronarios/crecimiento & desarrollo , Inflamación/fisiopatología , Neovascularización Fisiológica/fisiología , Factor B de Crecimiento Endotelial Vascular/fisiología , Adenoviridae/genética , Animales , Cardiomegalia/fisiopatología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Modelos Animales , Músculo Esquelético/irrigación sanguínea , Miocardio , Neuropilina-1/fisiología , Ratas , Ratas Transgénicas , Ratas Wistar , Factor B de Crecimiento Endotelial Vascular/genética
20.
Cancer Res ; 69(11): 4656-64, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19487284

RESUMEN

The angiopoietin-1 (Ang1)/Tie2 signaling pathway is known to play an important role in the regulation of vascular maturation and maintenance of vessel integrity. In this study, we have investigated the effect of systemic Tie2 activation or inhibition on tumor growth and metastasis. We found that treatment with Ang1 delivered via an adenoviral vector promoted s.c. implanted tumor metastasis to the lungs. Ang1 treatment did not significantly increase vascular density in the tumors but induced enlargement of blood vessels in both the tumor and normal tissues, which increased tumor cell dissemination into the blood circulation. Ang1 also enhanced the formation of metastatic foci in the lungs when tumor cells were injected into the circulation via the tail vein. The effect of Ang1 on metastasis was validated by a simultaneous treatment with a soluble form of Tie2 (sTie2), which led to the suppression of Ang1-induced increase of tumor metastasis. Furthermore, using a highly metastatic tumor model, we confirmed that systemic treatment with sTie2 suppressed tumor metastasis to the lungs and lymph nodes, whereas tumor-associated angiogenesis and lymphangiogenesis were not significantly affected. This suggests that the Ang1/Tie2 signals contribute to tumor progression by increasing vascular entry and exit of tumor cells to facilitate tumor dissemination and establishment of metastases.


Asunto(s)
Angiopoyetina 1/genética , Movimiento Celular/genética , Endotelio Vascular/patología , Metástasis de la Neoplasia/genética , Células Neoplásicas Circulantes/patología , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Angiopoyetina 1/fisiología , Animales , Permeabilidad Capilar/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/efectos de los fármacos , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/fisiología , Células Tumorales Cultivadas , Regulación hacia Arriba/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...