Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 39(7): 1402-1418, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31242036

RESUMEN

Objective- Pathological neovascularization is crucial for progression and morbidity of serious diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. While mechanisms of ongoing pathological neovascularization have been extensively studied, the initiating pathological vascular remodeling (PVR) events, which precede neovascularization remains poorly understood. Here, we identify novel molecular and cellular mechanisms of preneovascular PVR, by using the adult choriocapillaris as a model. Approach and Results- Using hypoxia or forced overexpression of VEGF (vascular endothelial growth factor) in the subretinal space to induce PVR in zebrafish and rats respectively, and by analyzing choriocapillaris membranes adjacent to choroidal neovascular lesions from age-related macular degeneration patients, we show that the choriocapillaris undergo robust induction of vascular intussusception and permeability at preneovascular stages of PVR. This PVR response included endothelial cell proliferation, formation of endothelial luminal processes, extensive vesiculation and thickening of the endothelium, degradation of collagen fibers, and splitting of existing extravascular columns. RNA-sequencing established a role for endothelial tight junction disruption, cytoskeletal remodeling, vesicle- and cilium biogenesis in this process. Mechanistically, using genetic gain- and loss-of-function zebrafish models and analysis of primary human choriocapillaris endothelial cells, we determined that HIF (hypoxia-induced factor)-1α-VEGF-A-VEGFR2 signaling was important for hypoxia-induced PVR. Conclusions- Our findings reveal that PVR involving intussusception and splitting of extravascular columns, endothelial proliferation, vesiculation, fenestration, and thickening is induced before neovascularization, suggesting that identifying and targeting these processes may prevent development of advanced neovascular disease in the future. Visual Overview- An online visual overview is available for this article.


Asunto(s)
Neovascularización Patológica/etiología , Remodelación Vascular/fisiología , Adulto , Animales , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Degeneración Macular/etiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Pez Cebra
2.
Proc Natl Acad Sci U S A ; 113(38): E5618-27, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27608497

RESUMEN

Vascular pericytes, an important cellular component in the tumor microenvironment, are often associated with tumor vasculatures, and their functions in cancer invasion and metastasis are poorly understood. Here we show that PDGF-BB induces pericyte-fibroblast transition (PFT), which significantly contributes to tumor invasion and metastasis. Gain- and loss-of-function experiments demonstrate that PDGF-BB-PDGFRß signaling promotes PFT both in vitro and in in vivo tumors. Genome-wide expression analysis indicates that PDGF-BB-activated pericytes acquire mesenchymal progenitor features. Pharmacological inhibition and genetic deletion of PDGFRß ablate the PDGF-BB-induced PFT. Genetic tracing of pericytes with two independent mouse strains, TN-AP-CreERT2:R26R-tdTomato and NG2-CreERT2:R26R-tdTomato, shows that PFT cells gain stromal fibroblast and myofibroblast markers in tumors. Importantly, coimplantation of PFT cells with less-invasive tumor cells in mice markedly promotes tumor dissemination and invasion, leading to an increased number of circulating tumor cells and metastasis. Our findings reveal a mechanism of vascular pericytes in PDGF-BB-promoted cancer invasion and metastasis by inducing PFT, and thus targeting PFT may offer a new treatment option of cancer metastasis.


Asunto(s)
Carcinoma de Células Renales/genética , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Becaplermina , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Pericitos/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nat Commun ; 7: 12680, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27580750

RESUMEN

The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the 'off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Indoles/uso terapéutico , Hígado/irrigación sanguínea , Hígado/patología , Neoplasias/patología , Pirroles/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Femenino , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia/patología , Neoplasias/tratamiento farmacológico , Sunitinib , Factor A de Crecimiento Endotelial Vascular/genética
4.
Nat Commun ; 7: 11385, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27150562

RESUMEN

Signalling molecules and pathways that mediate crosstalk between various tumour cellular compartments in cancer metastasis remain largely unknown. We report a mechanism of the interaction between perivascular cells and tumour-associated macrophages (TAMs) in promoting metastasis through the IL-33-ST2-dependent pathway in xenograft mouse models of cancer. IL-33 is the highest upregulated gene through activation of SOX7 transcription factor in PDGF-BB-stimulated pericytes. Gain- and loss-of-function experiments validate that IL-33 promotes metastasis through recruitment of TAMs. Pharmacological inhibition of the IL-33-ST2 signalling by a soluble ST2 significantly inhibits TAMs and metastasis. Genetic deletion of host IL-33 in mice also blocks PDGF-BB-induced TAM recruitment and metastasis. These findings shed light on the role of tumour stroma in promoting metastasis and have therapeutic implications for cancer therapy.


Asunto(s)
Interleucina-33/metabolismo , Macrófagos/metabolismo , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Factores de Transcripción SOXF/metabolismo , Células del Estroma/metabolismo , Animales , Becaplermina , Línea Celular Tumoral , Femenino , Humanos , Interleucina-33/genética , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Factores de Transcripción SOXF/genética
5.
Nat Commun ; 7: 10824, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26926389

RESUMEN

Lymphangiogensis is involved in various pathological conditions, such as arthritis and cancer metastasis. Although many factors have been identified to stimulate lymphatic vessel growth, little is known about lymphangiogenesis inhibitors. Here we report that membrane type 1-matrix metalloproteinase (MT1-MMP) is an endogenous suppressor of lymphatic vessel growth. MT1-MMP-deficient mice exhibit spontaneous corneal lymphangiogenesis without concomitant changes in angiogenesis. Mice lacking MT1-MMP in either lymphatic endothelial cells or macrophages recapitulate corneal lymphangiogenic phenotypes observed in Mmp14(-/-) mice, suggesting that the spontaneous lymphangiogenesis is both lymphatic endothelial cells autonomous and macrophage associated. Mechanistically, MT1-MMP directly cleaves LYVE-1 on lymphatic endothelial cells to inhibit LYVE-1-mediated lymphangiogenic responses. In addition, MT1-MMP-mediated PI3Kδ signalling restrains the production of VEGF-C from prolymphangiogenic macrophages through repressing the activation of NF-κB signalling. Thus, we identify MT1-MMP as an endogenous inhibitor of physiological lymphangiogenesis.


Asunto(s)
Células Endoteliales/fisiología , Glicoproteínas/metabolismo , Linfangiogénesis/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Trasplante Óseo , Células Cultivadas , Córnea , Regulación de la Expresión Génica/fisiología , Glicoproteínas/genética , Macrófagos/fisiología , Metaloproteinasa 14 de la Matriz/genética , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Factor C de Crecimiento Endotelial Vascular/genética
6.
Oncoimmunology ; 4(3): e989777, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25949904

RESUMEN

Although inflammation and metastasis are two well-known hallmarks of malignant disease, the relationship between inflammation and lymphatic metastasis is an unexplored research area. We recently elucidated a sophisticated mechanism by which TNFα-induced tumor inflammation conscripts macrophage-mediated VEGF-C-VEGFR3 signaling in lymphangiogenesis and metastasis.

7.
Nat Commun ; 5: 4944, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25229256

RESUMEN

Inflammation and lymphangiogenesis are two cohesively coupled processes that promote tumour growth and invasion. Here we report that TNF-α markedly promotes tumour lymphangiogenesis and lymphatic metastasis. The TNF-α-TNFR1 signalling pathway directly stimulates lymphatic endothelial cell activity through a VEGFR3-independent mechanism. However, VEGFR3-induced lymphatic endothelial cell tips are a prerequisite for lymphatic vessel growth in vivo, and a VEGFR3 blockade completely ablates TNF-α-induced lymphangiogenesis. Moreover, TNF-α-TNFR1-activated inflammatory macrophages produce high levels of VEGF-C to coordinately activate VEGFR3. Genetic deletion of TNFR1 (Tnfr1(-/-)) in mice or depletion of tumour-associated macrophages (TAMs) virtually eliminates TNF-α-induced lymphangiogenesis and lymphatic metastasis. Gain-of-function experiments show that reconstitution of Tnfr1(+/+) macrophages in Tnfr1(-/-) mice largely restores tumour lymphangiogenesis and lymphatic metastasis. These findings shed mechanistic light on the intimate interplay between inflammation and lymphangiogenesis in cancer metastasis, and propose therapeutic intervention of lymphatic metastasis by targeting the TNF-α-TNFR1 pathway.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis , Metástasis Linfática , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Metástasis de la Neoplasia , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Células U937
8.
Sci Rep ; 3: 2053, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23792494

RESUMEN

The functional impact of amyloid peptides (Aßs) on the vascular system is less understood despite these pathologic peptides are substantially deposited in the brain vasculature of Alzheimer's patients. Here we show substantial accumulation of Aßs 40 and 42 in the brain arterioles of Alzheimer's patients and of transgenic Alzheimer's mice. Purified Aßs 1-40 and 1-42 exhibited vascular regression activity in the in vivo animal models and vessel density was reversely correlated with numbers and sizes of amyloid plaques in human patients. A significant high number of vascular cells underwent cellular apoptosis in the brain vasculature of Alzheimer's patients. VEGF significantly prevented Aß-induced endothelial apoptosis in vitro. Neuronal expression of VEGF in transgenic mice restored memory behavior of Alzheimer's. These findings provide conceptual implication of improvement of vascular functions as a novel therapeutic approach for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Apoptosis , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Humanos , Trastornos de la Memoria/patología , Ratones , Persona de Mediana Edad
9.
Proc Natl Acad Sci U S A ; 109(39): 15894-9, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22967508

RESUMEN

Interplay between various lymphangiogenic factors in promoting lymphangiogenesis and lymphatic metastasis remains poorly understood. Here we show that FGF-2 and VEGF-C, two lymphangiogenic factors, collaboratively promote angiogenesis and lymphangiogenesis in the tumor microenvironment, leading to widespread pulmonary and lymph-node metastases. Coimplantation of dual factors in the mouse cornea resulted in additive angiogenesis and lymphangiogenesis. At the molecular level, we showed that FGFR-1 expressed in lymphatic endothelial cells is a crucial receptor that mediates the FGF-2-induced lymphangiogenesis. Intriguingly, the VEGFR-3-mediated signaling was required for the lymphatic tip cell formation in both FGF-2- and VEGF-C-induced lymphangiogenesis. Consequently, a VEGFR-3-specific neutralizing antibody markedly inhibited FGF-2-induced lymphangiogenesis. Thus, the VEGFR-3-induced lymphatic endothelial cell tip cell formation is a prerequisite for FGF-2-stimulated lymphangiogenesis. In the tumor microenvironment, the reciprocal interplay between FGF-2 and VEGF-C collaboratively stimulated tumor growth, angiogenesis, intratumoral lymphangiogenesis, and metastasis. Thus, intervention and targeting of the FGF-2- and VEGF-C-induced angiogenic and lymphangiogenic synergism could be potentially important approaches for cancer therapy and prevention of metastasis.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Linfoma/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Microambiente Tumoral , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Transformada , Línea Celular Tumoral , Células Endoteliales/metabolismo , Células Endoteliales/patología , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Linfoma/genética , Linfoma/patología , Linfoma/terapia , Ratones , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Transducción de Señal/genética , Trasplante Heterólogo , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Cell Cycle ; 11(15): 2793-8, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22801544

RESUMEN

MT1-MMP is a membrane-tethered enzyme capable of remodeling extracellular matrix. MT1-MMP-deficient mice exhibit systematic defects during development, especially in craniofacial development characterized by retarded calvarial bone formation. Recently, we identified MT1-MMP as a critical positive modulator of FGF signaling during intramembranous ossification. MT1-MMP cleaves ADAM9 to protect FGFR2 from ectodomain shedding. Depletion of ADAM9 in MT1-MMP-deficient mice significantly rescued the calvarial defects via restoring FGF signaling. Interestingly, this regulatory mechanism seems to be highly tissue-specific, as defective FGF2-induced corneal angiogenesis in Mmp14-/- mice could not be rescued by removal of ADAM9. In addition, MT1-MMP also cleaves another ADAM family member, ADAM15. Our current findings not only present a novel regulatory mechanism for FGF signaling but also reveal a functional crosstalk between MMP and ADAM families. Better understanding of the interplay between ADAMs and MT1-MMP and its consequences for signaling pathways will provide new insights into therapeutic approaches for the management of developmental disorders and various diseases, such as cancer.


Asunto(s)
Proteínas ADAM/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Osteogénesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Huesos/metabolismo , Membrana Celular/enzimología , Neovascularización de la Córnea , Metaloproteinasa 14 de la Matriz/genética , Ratones , Transducción de Señal
11.
Nat Med ; 18(1): 100-10, 2011 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-22138754

RESUMEN

The platelet-derived growth factor (PDGF) signaling system contributes to tumor angiogenesis and vascular remodeling. Here we show in mouse tumor models that PDGF-BB induces erythropoietin (EPO) mRNA and protein expression by targeting stromal and perivascular cells that express PDGF receptor-ß (PDGFR-ß). Tumor-derived PDGF-BB promoted tumor growth, angiogenesis and extramedullary hematopoiesis at least in part through modulation of EPO expression. Moreover, adenoviral delivery of PDGF-BB to tumor-free mice increased both EPO production and erythropoiesis, as well as protecting from irradiation-induced anemia. At the molecular level, we show that the PDGF-BB-PDGFR-bß signaling system activates the EPO promoter, acting in part through transcriptional regulation by the transcription factor Atf3, possibly through its association with two additional transcription factors, c-Jun and Sp1. Our findings suggest that PDGF-BB-induced EPO promotes tumor growth through two mechanisms: first, paracrine stimulation of tumor angiogenesis by direct induction of endothelial cell proliferation, migration, sprouting and tube formation, and second, endocrine stimulation of extramedullary hematopoiesis leading to increased oxygen perfusion and protection against tumor-associated anemia.


Asunto(s)
Eritropoyetina/metabolismo , Hematopoyesis , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Anemia/genética , Animales , Becaplermina , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Eritropoyetina/genética , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neovascularización Patológica/genética , Proteínas Proto-Oncogénicas c-sis/genética , Transducción de Señal , Células del Estroma/metabolismo
12.
Circulation ; 124(9): 1059-69, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21824923

RESUMEN

BACKGROUND: Therapeutic angiogenesis is a promising approach for the treatment of cardiovascular diseases, including myocardial infarction and chronic heart failure. We aimed to improve proangiogenic therapies by identifying novel arteriogenic growth factor combinations, developing injectable delivery systems for spatiotemporally controlled growth factor release, and evaluating functional consequences of targeted intramyocardial growth factor delivery in chronic heart failure. METHODS AND RESULTS: First, we observed that fibroblast growth factor and hepatocyte growth factor synergistically stimulate vascular cell migration and proliferation in vitro. Using 2 in vivo angiogenesis assays (n=5 mice per group), we found that the growth factor combination results in a more potent and durable angiogenic response than either growth factor used alone. Furthermore, we determined that the molecular mechanisms involve potentiation of Akt and mitogen-activated protein kinase signal transduction pathways, as well as upregulation of angiogenic growth factor receptors. Next, we developed crosslinked albumin-alginate microcapsules that sequentially release fibroblast growth factor-2 and hepatocyte growth factor. Finally, in a rat model of chronic heart failure induced by coronary ligation (n=14 to 15 rats per group), we found that intramyocardial slow release of fibroblast growth factor-2 with hepatocyte growth factor potently stimulates angiogenesis and arteriogenesis and prevents cardiac hypertrophy and fibrosis, as determined by immunohistochemistry, leading to improved cardiac perfusion after 3 months, as shown by magnetic resonance imaging. These multiple beneficial effects resulted in reduced adverse cardiac remodeling and improved left ventricular function, as revealed by echocardiography. CONCLUSION: Our data showing the selective advantage of using fibroblast growth factor-2 together with hepatocyte growth factor suggest that this growth factor combination may constitute an efficient novel treatment for chronic heart failure.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Insuficiencia Cardíaca/prevención & control , Factor de Crecimiento de Hepatocito/administración & dosificación , Miocardio , Neovascularización Fisiológica/efectos de los fármacos , Animales , Cápsulas , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/prevención & control , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiopatología , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Ratas , Ratas Wistar
13.
Cell Cycle ; 9(10): 1882-5, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473029

RESUMEN

Cancer-associated retinopathy (CAR) is believed to be associated with autoimmune responses against retinal specific antigens.  However, CAR patients often show little evidence of immunological reactions at the cellular or molecular levels in their eyes. We have recently shown that tumor-derived VEGF and PlGF significantly remodel the retinal vasculature by ablation of pericytes and impair the blood-retinal barrier, leading to increased vascular leakage. Surprisingly, VEGFR1, but not VEGFR2, is the primary receptor that transduces signals in endothelial or mural cells to produce these vascular pathologies.  These results demonstrate that tumor-derived angiogenic factors significantly confer the development of CAR and anti-VEGF agents might be potentially used for the treatment of CAR.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/fisiopatología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Animales , Humanos , Modelos Biológicos , Factor de Crecimiento Placentario , Proteínas Gestacionales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(2): 856-61, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20080765

RESUMEN

VEGF coordinates complex regulation of cellular regeneration and interactions between endothelial and perivascular cells; dysfunction of the VEGF signaling system leads to retinopathy. Here, we show that systemic delivery of VEGF and placental growth factor (PlGF) by protein implantation, tumors, and adenoviral vectors ablates pericytes from the mature retinal vasculature through the VEGF receptor 1 (VEGFR1)-mediated signaling pathway, leading to increased vascular leakage. In contrast, we demonstrate VEGF receptor 2 (VEGFR2) is primarily expressed in nonvascular photoreceptors and ganglion cells. Moreover, blockade of VEGFR1 but not VEGFR2 significantly restores pericyte saturation in mature retinal vessels. Our findings link VEGF and PlGF to cancer-associated retinopathy, reveal the molecular mechanisms of VEGFR1 ligand-mediated retinopathy, and define VEGFR1 as an important target of antiangiogenic therapy for treatment of retinopathy.


Asunto(s)
Neoplasias/complicaciones , Pericitos/patología , Enfermedades de la Retina/epidemiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Placentario , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/fisiología , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , Retina/patología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/patología
15.
Proc Natl Acad Sci U S A ; 106(43): 18408-13, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19822749

RESUMEN

The blood and lymphatic vasculatures are structurally and functionally coupled in controlling tissue perfusion, extracellular interstitial fluids, and immune surveillance. Little is known, however, about the molecular mechanisms that underlie the regulation of bloodlymphatic vessel connections and lymphatic perfusion. Here we show in the adult zebrafish and glass catfish (Kryptopterus bicirrhis) that blood-lymphatic conduits directly connect arterial vessels to the lymphatic system. Under hypoxic conditions, arterial-lymphatic conduits (ALCs) became highly dilated and linearized by NO-induced vascular relaxation, which led to blood perfusion into the lymphatic system. NO blockage almost completely abrogated hypoxia-induced ALC relaxation and lymphatic perfusion. These findings uncover mechanisms underlying hypoxia-induced oxygen compensation by perfusion of existing lymphatics in fish. Our results might also imply that the hypoxia-induced NO pathway contributes to development of progression of pathologies, including promotion of lymphatic metastasis by modulating arterial-lymphatic conduits, in the mammalian system.


Asunto(s)
Arterias/metabolismo , Bagres/metabolismo , Hipoxia/metabolismo , Vasos Linfáticos/metabolismo , Óxido Nítrico/metabolismo , Pez Cebra/metabolismo , Envejecimiento , Animales , Perfusión
16.
Proc Natl Acad Sci U S A ; 106(41): 17505-10, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19805167

RESUMEN

Vascular functions of PlGF remain poorly understood and controversial. Here, we show that tumor cell-derived PlGF-1 and PlGF-2 displayed significant remodeling effects on the tumor vasculature, leading to a normalized vascular phenotype and improved functions against leakage. In two murine tumor models, that is, T241 fibrosarcoma and Lewis lung carcinoma, stable expression of PlGF-1 and PlGF-2 in tumor cells resulted in significant reduction of tumor microvascular density and branch formation. Markedly, the vasculature in PlGF-expressing tumors consisted of relatively large-diameter microvessels with substantial improvement of pericyte coverage. Similarly, PlGF-induced vascular normalization and remodeling were also observed in a spontaneous human choriocarcinoma that expressed endogenous PlGF. Our findings shed light on functions of PlGF as a vascular remodeling factor that normalizes the tumor vasculature and thus may have conceptual implications of cancer therapy.


Asunto(s)
Neoplasias Pulmonares/irrigación sanguínea , Proteínas Gestacionales/uso terapéutico , Animales , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Humanos , Ratones , Neovascularización Patológica/patología , Pericitos/citología , Pericitos/patología , Pericitos/fisiología , Factor de Crecimiento Placentario , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor C de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/uso terapéutico
17.
Cell Metab ; 9(1): 99-109, 2009 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-19117550

RESUMEN

The molecular mechanisms of angiogenesis in relation to adipose tissue metabolism remain poorly understood. Here, we show that exposure of mice to cold led to activation of angiogenesis in both white and brown adipose tissues. In the inguinal depot, cold exposure resulted in elevated expression levels of brown-fat-associated proteins, including uncoupling protein-1 (UCP1) and PGC-1alpha. Proangiogenic factors such as VEGF were upregulated, and endogenous angiogenesis inhibitors, including thrombospondin, were downregulated. In wild-type mice, the adipose tissues became hypoxic during cold exposure; in UCP1(-/-) mice, hypoxia did not occur, but, remarkably, the augmented angiogenesis was unaltered and was thus hypoxia independent. Intriguingly, VEGFR2 blockage abolished the cold-induced angiogenesis and significantly impaired nonshivering thermogenesis capacity. Unexpectedly, VEGFR1 blockage resulted in the opposite effects: increased adipose vascularity and nonshivering thermogenesis capacity. Our findings have conceptual implications concerning application of angiogenesis modulators for treatment of obesity and metabolic disorders.


Asunto(s)
Aclimatación , Tejido Adiposo Pardo/irrigación sanguínea , Tejido Adiposo Blanco/irrigación sanguínea , Frío , Neovascularización Fisiológica , Tejido Adiposo Pardo/anatomía & histología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/anatomía & histología , Tejido Adiposo Blanco/metabolismo , Animales , Hipoxia de la Célula , Perfilación de la Expresión Génica , Canales Iónicos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteína Desacopladora 1 , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
FASEB J ; 23(1): 153-63, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18827023

RESUMEN

Preclinical and clinical evaluations of individual proangiogenic/arteriogenic factors for the treatment of ischemic myocardium and skeletal muscle have produced unfulfilled promises. The establishment of functional and stable arterial vascular networks may require combinations of different angiogenic and arteriogenic factors. Using in vivo angiogenesis and ischemic hind-limb animal models, we have compared the angiogenic and therapeutic activities of fibroblast growth factor 2 (FGF-2) in combinations with PDGF-AA and PDGF-AB, two members of the platelet-derived growth factor (PDGF) family, with distinct receptor binding patterns. We show that both PDGF-AA/FGF-2 and PDGF-AB/FGF-2 in combinations synergistically induce angiogenesis in the mouse cornea. FGF-2 up-regulates PDGFR-alpha and -beta expression levels in the newly formed blood vessels. Interestingly, PDGF-AB/FGF-2, but not PDGF-AA/FGF-2, is able to stabilize the newly formed vasculature by recruiting pericytes, and an anti-PDGFR-beta neutralizing antibody significantly blocks PDGF-AB/FGF-2-induced vessel stability. These findings demonstrate that PDGFR-beta receptor is essential for vascular stability. Similarly, PDGF-AB/FGF-2 significantly induces stable collateral growth in the rat ischemic hind limb. The high number of collaterals induced by PDGF-AB/FGF-2 leads to dramatic improvement of the paw's skin perfusion. Immunohistochemical analysis of the treated skeletal muscles confirms that a combination of PDGF-AB and FGF-2 significantly induces arteriogenesis in the ischemic tissue. A combination of PDGF-AB and FGF-2 would be optimal proangiogenic agents for the treatment of ischemic diseases.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Circulación Colateral/efectos de los fármacos , Córnea/irrigación sanguínea , Córnea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Neovascularización Fisiológica/fisiología , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Ratas
19.
Proc Natl Acad Sci U S A ; 105(47): 18513-8, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19017793

RESUMEN

The underlying mechanism by which anti-VEGF agents prolong cancer patient survival is poorly understood. We show that in a mouse tumor model, VEGF systemically impairs functions of multiple organs including those in the hematopoietic and endocrine systems, leading to early death. Anti-VEGF antibody, bevacizumab, and anti-VEGF receptor 2 (VEGFR-2), but not anti-VEGFR-1, reversed VEGF-induced cancer-associated systemic syndrome (CASS) and prevented death in tumor-bearing mice. Surprisingly, VEGFR2 blockage improved survival by rescuing mice from CASS without significantly compromising tumor growth, suggesting that "off-tumor" VEGF targets are more sensitive than the tumor vasculature to anti-VEGF drugs. Similarly, VEGF-induced CASS occurred in a spontaneous breast cancer mouse model overexpressing neu. Clinically, VEGF expression and CASS severity positively correlated in various human cancers. These findings define novel therapeutic targets of anti-VEGF agents and provide mechanistic insights into the action of this new class of clinically available anti-VEGF cancer drugs.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Anemia/fisiopatología , Animales , Permeabilidad Capilar , Humanos , Inmunohistoquímica , Hígado/fisiopatología , Ratones , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/fisiopatología , Neoplasias Experimentales/prevención & control
20.
PLoS One ; 3(7): e2748, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18648503

RESUMEN

Mechanistic understanding and defining novel therapeutic targets of diabetic retinopathy and age-related macular degeneration (AMD) have been hampered by a lack of appropriate adult animal models. Here we describe a simple and highly reproducible adult fli-EGFP transgenic zebrafish model to study retinal angiogenesis. The retinal vasculature in the adult zebrafish is highly organized and hypoxia-induced neovascularization occurs in a predictable area of capillary plexuses. New retinal vessels and vascular sprouts can be accurately measured and quantified. Orally active anti-VEGF agents including sunitinib and ZM323881 effectively block hypoxia-induced retinal neovascularization. Intriguingly, blockage of the Notch signaling pathway by the inhibitor DAPT under hypoxia, results in a high density of arterial sprouting in all optical arteries. The Notch suppression-induced arterial sprouting is dependent on tissue hypoxia. However, in the presence of DAPT substantial endothelial tip cell formation was detected only in optic capillary plexuses under normoxia. These findings suggest that hypoxia shifts the vascular targets of Notch inhibitors. Our findings for the first time show a clinically relevant retinal angiogenesis model in adult zebrafish, which might serve as a platform for studying mechanisms of retinal angiogenesis, for defining novel therapeutic targets, and for screening of novel antiangiogenic drugs.


Asunto(s)
Hipoxia , Neovascularización Patológica , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/patología , Neovascularización Retiniana/patología , Inhibidores de la Angiogénesis/farmacología , Animales , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Indoles/farmacología , Microscopía Confocal , Pirroles/farmacología , Receptores Notch/metabolismo , Neovascularización Retiniana/metabolismo , Transducción de Señal , Sunitinib , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA