Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Genes Dev ; 28(14): 1592-603, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25030698

RESUMO

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.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , Junções Intercelulares/fisiologia , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/genética , Animais , Caderinas/metabolismo , Embrião de Mamíferos , Células Endoteliais/citologia , Deleção de Genes , Linfangiogênese/fisiologia , Tecido Linfoide/embriologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
2.
Proc Natl Acad Sci U S A ; 112(42): 13063-8, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26430242

RESUMO

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.


Assuntos
Cardiomegalia/enzimologia , Endotélio Vascular/enzimologia , Proteínas Tirosina Quinases/metabolismo , Angiotensina II/farmacologia , Animais , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Transdução de Sinais
3.
Circulation ; 129(19): 1962-71, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24552833

RESUMO

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.


Assuntos
Proteínas ADAM/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linfangiogênese/fisiologia , Pró-Colágeno N-Endopeptidase/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Proteínas ADAMTS , Adenoviridae/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Animais , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Neovascularização Fisiológica/fisiologia , Transfecção , Proteínas Supressoras de Tumor/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Circulation ; 127(4): 424-34, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23357661

RESUMO

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.


Assuntos
Angiopoietina-1/farmacologia , Terapia Genética/métodos , Isquemia/tratamento farmacológico , Neovascularização Fisiológica/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adenoviridae/genética , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Permeabilidade Capilar/fisiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Células HEK293 , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana , Humanos , Isquemia/genética , Leucemia Mieloide , Camundongos , Camundongos Endogâmicos , Músculo Esquelético/irrigação sanguínea , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Blood ; 118(4): 1154-62, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21566091

RESUMO

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.


Assuntos
Linfangiogênese/fisiologia , Vasos Linfáticos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Células Endoteliais/metabolismo , Imunofluorescência , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Circulation ; 122(17): 1725-33, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20937974

RESUMO

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.


Assuntos
Permeabilidade Capilar/fisiologia , Vasos Coronários/crescimento & desenvolvimento , Inflamação/fisiopatologia , Neovascularização Fisiológica/fisiologia , Fator B de Crescimento do Endotélio Vascular/fisiologia , Adenoviridae/genética , Animais , Cardiomegalia/fisiopatologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Modelos Animais , Músculo Esquelético/irrigação sanguínea , Miocárdio , Neuropilina-1/fisiologia , Ratos , Ratos Transgênicos , Ratos Wistar , Fator B de Crescimento do Endotélio Vascular/genética
7.
J Surg Oncol ; 103(6): 484-8, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21480240

RESUMO

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.


Assuntos
Linfangiogênese , Neoplasias/irrigação sanguínea , Neovascularização Patológica/fisiopatologia , Inibidores da Angiogênese/uso terapêutico , Humanos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Circ Res ; 103(12): 1359-62, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18988895

RESUMO

Bmx nonreceptor tyrosine kinase has an established role in endothelial and lymphocyte signaling; however, its role in the heart is unknown. To determine whether Bmx participates in cardiac growth, we subjected mice deficient in the molecule (Bmx knockout mice) to transverse aortic constriction (TAC). In comparison with wild-type mice, which progressively developed massive hypertrophy following TAC, Bmx knockout mice were resistant to TAC-induced cardiac growth at the organ and cell level. Loss of Bmx preserved cardiac ejection fraction and decreased mortality following TAC. These findings are the first to demonstrate a necessary role for the Tec family of tyrosine kinases in the heart and reveal a novel regulator (Bmx) of pressure overload-induced hypertrophic growth.


Assuntos
Pressão Sanguínea/fisiologia , Cardiomegalia/metabolismo , Cardiomegalia/prevenção & controle , Proteínas Tirosina Quinases/deficiência , Animais , Pressão Sanguínea/genética , Cardiomegalia/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia
9.
Cancer Res ; 67(2): 593-9, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234768

RESUMO

Vascular endothelial growth factor receptor 3 (VEGFR-3) binds VEGF-C and VEGF-D and is essential for the development of the lymphatic vasculature. Experimental tumors that overexpress VEGFR-3 ligands induce lymphatic vessel sprouting and enlargement and show enhanced metastasis to regional lymph nodes and beyond, whereas a soluble form of VEGFR-3 that blocks receptor signaling inhibits these changes and metastasis. Because VEGFR-3 is also essential for the early blood vessel development in embryos and is up-regulated in tumor angiogenesis, we wanted to determine if an antibody targeting the receptor that interferes with VEGFR-3 ligand binding can inhibit primary tumor growth. Our results show that antibody interference with VEGFR-3 function can inhibit the growth of several human tumor xenografts in immunocompromised mice. Immunohistochemical analysis showed that the blood vessel density of anti-VEGFR-3-treated tumors was significantly decreased and hypoxic and necrotic tumor tissue was increased when compared with tumors treated with control antibody, indicating that blocking of the VEGFR-3 pathway inhibits angiogenesis in these tumors. As expected, the anti-VEGFR-3-treated tumors also lacked lymphatic vessels. These results suggest that the VEGFR-3 pathway contributes to tumor angiogenesis and that effective inhibition of tumor progression may require the inhibition of multiple angiogenic targets.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Processos de Crescimento Celular , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Neovascularização Patológica/terapia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Cachexia Sarcopenia Muscle ; 9(3): 514-529, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29722201

RESUMO

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.


Assuntos
Receptores de Activinas Tipo II/farmacologia , Caquexia/metabolismo , Fígado/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Baço/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Receptores de Activinas Tipo II/uso terapêutico , Animais , Biomarcadores , Caquexia/tratamento farmacológico , Caquexia/etiologia , Caquexia/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação , Estimativa de Kaplan-Meier , Fígado/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/uso terapêutico , Baço/imunologia , Baço/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Res ; 65(11): 4739-46, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930292

RESUMO

Lymphangiogenic growth factors vascular endothelial growth factor (VEGF)-C and VEGF-D have been shown to promote lymphatic metastasis by inducing tumor-associated lymphangiogenesis. In this study, we have investigated how tumor cells gain access into lymphatic vessels and at what stage tumor cells initiate metastasis. We show that VEGF-C produced by tumor cells induced extensive lymphatic sprouting towards the tumor cells as well as dilation of the draining lymphatic vessels, suggesting an active role of lymphatic endothelial cells in lymphatic metastasis. A significant increase in lymphatic vessel growth occurred between 2 and 3 weeks after tumor xenotransplantation, and lymph node metastasis occurred at the same stage. These processes were blocked dose-dependently by inhibition of VEGF receptor 3 (VEGFR-3) signaling by systemic delivery of a soluble VEGFR-3-immunoglobulin (Ig) fusion protein via adenoviral or adeno-associated viral vectors. However, VEGFR-3-Ig did not suppress lymph node metastasis when the treatment was started at a later stage after the tumor cells had already spread out, suggesting that tumor cell entry into lymphatic vessels is a key step during tumor dissemination via the lymphatics. Whereas lymphangiogenesis and lymph node metastasis were significantly inhibited by VEGFR-3-Ig, some tumor cells were still detected in the lymph nodes in some of the treated mice. This indicates that complete blockade of lymphatic metastasis may require the targeting of both tumor lymphangiogenesis and tumor cell invasion.


Assuntos
Endotélio Linfático/patologia , Neoplasias Pulmonares/patologia , Linfonodos/patologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Imunoglobulinas/administração & dosagem , Imunoglobulinas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Metástase Linfática , Camundongos , Camundongos SCID , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/administração & dosagem , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Neoplasia ; 18(7): 436-46, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27435926

RESUMO

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.


Assuntos
Carcinógenos/toxicidade , Carcinoma de Células Escamosas/patologia , Linfonodos/irrigação sanguínea , Linfangiogênese/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Fator D de Crescimento do Endotélio Vascular/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Escamosas/induzido quimicamente , Citometria de Fluxo , Metástase Linfática , Vasos Linfáticos/patologia , Contagem de Linfócitos , Macrófagos/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente , Acetato de Tetradecanoilforbol/toxicidade
14.
J Clin Pathol ; 68(8): 614-21, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26021331

RESUMO

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.


Assuntos
Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Interpretação de Imagem Assistida por Computador/métodos , Neoplasias Pulmonares/patologia , Coloração e Rotulagem/métodos , Adenocarcinoma de Pulmão , Algoritmos , Animais , Área Sob a Curva , Inteligência Artificial , Automação Laboratorial , Linhagem Celular Tumoral , Sobrevivência Celular , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Valor Preditivo dos Testes , Curva ROC , Reprodutibilidade dos Testes , Microambiente Tumoral
15.
Cancer Res ; 75(10): 2083-94, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25808867

RESUMO

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.


Assuntos
Colágeno/metabolismo , Metaloproteinase 16 da Matriz/fisiologia , Melanoma/enzimologia , Neoplasias Cutâneas/enzimologia , Animais , Células COS , Adesão Celular , Chlorocebus aethiops , Matriz Extracelular/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Estimativa de Kaplan-Meier , Linfonodos/patologia , Metaloproteinase 14 da Matriz/metabolismo , Melanoma/mortalidade , Melanoma/secundário , Metalotioneína 3 , Camundongos Endogâmicos ICR , Camundongos SCID , Invasividade Neoplásica , Transplante de Neoplasias , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Proteólise , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia
16.
Cell Rep ; 8(6): 1943-1956, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25242330

RESUMO

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.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Anexina A1/antagonistas & inibidores , Anexina A1/genética , Anexina A1/metabolismo , Autofagia , Técnicas de Cultura de Células , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Filaminas/antagonistas & inibidores , Filaminas/genética , Filaminas/metabolismo , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Isquemia/patologia , Isquemia/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transplante Heterólogo , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Regulação para Cima
17.
J Clin Invest ; 124(2): 824-34, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24430181

RESUMO

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.


Assuntos
Angiopoietina-1/antagonistas & inibidores , Células Endoteliais/citologia , Endotélio Vascular/metabolismo , Deleção de Genes , Receptor de TIE-1/genética , Vasos Retinianos/patologia , Inibidores da Angiogênese/química , Angiopoietina-1/metabolismo , Animais , Apoptose , Sobrevivência Celular , Homozigoto , Ligantes , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Neovascularização Patológica , Fenótipo , Receptor de TIE-1/fisiologia , Receptor TIE-2/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
Sci Signal ; 6(282): ra52, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23821770

RESUMO

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.


Assuntos
Proteínas da Gravidez/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Ligação Competitiva , Western Blotting , Linhagem Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Ligantes , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Células NIH 3T3 , Fosforilação , Fator de Crescimento Placentário , Proteínas da Gravidez/química , Proteínas da Gravidez/genética , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/genética , Fator B de Crescimento do Endotélio Vascular/química , Fator B de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
19.
Cancer Res ; 72(14): 3512-21, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22593188

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Lewis/irrigação sanguínea , Neoplasias do Colo/irrigação sanguínea , Melanoma Experimental/irrigação sanguínea , Neovascularização Patológica/genética , Proteínas Tirosina Quinases/genética , Animais , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Feminino , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos
20.
J Natl Cancer Inst ; 104(6): 461-75, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22343031

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/metabolismo , Endotélio Vascular/efeitos dos fármacos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Neoplasias Pulmonares/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , Animais , Anticorpos Bloqueadores/metabolismo , Anticorpos Bloqueadores/farmacologia , Hipóxia Celular , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/patologia , Feminino , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Junções Intercelulares/patologia , Neoplasias Pulmonares/secundário , Linfangiogênese , Metástase Linfática , Melanoma/irrigação sanguínea , Melanoma/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Neovascularização Patológica/tratamento farmacológico , Receptor TIE-2/metabolismo , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Transplante Heterólogo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA