Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Semin Thromb Hemost ; 45(6): 576-592, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31096305

RESUMO

Similarities between solid tumor stroma generation, wound healing, chronic inflammation, and associated inflammatory diseases have prompted interest from the time of Virchow. However, it was not until the 1970s that these entities were shown to share important molecular mechanisms. Foundational to all of them is the initiating role of vascular endothelial growth factor (VEGF-A) in increasing vascular permeability to plasma and plasma proteins. Extravasated plasma activates the tissue factor clotting pathway, leading to extravascular deposition of a fibrin gel. Fibrin serves initially as a provisional stroma that provides a favorable substrate for the attachment and migration of tumor cells, as well as host fibroblasts, endothelial, and inflammatory cells. Fibrin and its degradation products have proangiogenic activity with important roles in the generation of new blood vessels and connective tissue stroma. Over time, fibrin is degraded and replaced by vascular and subsequently by dense, relatively avascular collagenous connective tissue, the end-product referred to as desmoplasia in tumors and scar in healed wounds. Fibrin and the mature stroma that replaces it provide a diffusion barrier to chemotherapy and a structural barrier that inflammatory cells must cross to reach tumor cells. Plasma solutes of varying size cross the endothelial cells lining capillaries and venules of normal tissues and "mother" vessels of tumors and wounds by different anatomical pathways. VEGF-A levels fall back to normal as wounds heal but remain perpetually elevated in solid tumors. Thus, tumors may heal centrally but continually initiate new healing activity as they grow and invade surrounding normal tissues.


Assuntos
Permeabilidade Capilar/fisiologia , Fibrina/metabolismo , Inflamação/metabolismo , Neoplasias/metabolismo , Humanos , Trombose , Cicatrização
2.
Am J Pathol ; 187(10): 2337-2347, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28736316

RESUMO

Tumors induce their heterogeneous vasculature by secreting vascular endothelial growth factor (VEGF)-A. Anti-VEGF/VEGF receptor (VEGFR) drugs treat cancer, but the underlying mechanisms remain unclear. An adenovirus expressing VEGF-A (Ad-VEGF-A164) replicates the tumor vasculature in mice without tumor cells. Mother vessels (MV) are the first angiogenic vessel type to form in tumors and after Ad-VEGF-A164. Multiday treatments with a VEGF trap reverted MV back to normal microvessels. We now show that, within hours, a single dose of several anti-VEGF drugs collapsed MV to form glomeruloid microvascular proliferations (GMP), accompanied by only modest endothelial cell death. GMP, common in many human cancers but of uncertain origin, served as an intermediary step in MV reversion to normal microvessels. The vasodisruptive drug combretastatin CA4 also targeted MV selectively but acted differently, extensively killing MV endothelium. Antivascular changes were quantified with a novel Evans blue dye assay that measured vascular volumes. As in tumors, Ad-VEGF-A164 strikingly increased endothelial nitric oxide synthase (eNOS) expression. The eNOS inhibitor N(G)-Nitro-l-arginine methyl ester mimicked anti-VEGF/VEGFR drugs, rapidly collapsing MV to GMP. Inhibition of eNOS reduces synthesis of its vasodilatory product, nitric oxide, leading to arterial contraction. Patients and mice receiving anti-VEGF/VEGFR drugs develop hypertension, reflecting systemic arterial contraction. Together, anti-VEGF/VEGFR drugs act in part by inhibiting eNOS, causing vasocontraction, MV collapse to GMP, and subsequent reversion of GMP to normal microvessels, all without extensive vascular killing.


Assuntos
Inibidores da Angiogênese/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adenoviridae/metabolismo , Animais , Bibenzilas/farmacologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Hipertensão/patologia , Camundongos Endogâmicos C57BL , Camundongos Nus , Microvasos/efeitos dos fármacos , Microvasos/patologia , Modelos Biológicos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
J Pathol ; 241(3): 362-374, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27859259

RESUMO

Anti-angiogenic therapies have shown limited efficacy in the clinical management of metastatic disease, including lung metastases. Moreover, the mechanisms via which tumours resist anti-angiogenic therapies are poorly understood. Importantly, rather than utilizing angiogenesis, some metastases may instead incorporate pre-existing vessels from surrounding tissue (vessel co-option). As anti-angiogenic therapies were designed to target only new blood vessel growth, vessel co-option has been proposed as a mechanism that could drive resistance to anti-angiogenic therapy. However, vessel co-option has not been extensively studied in lung metastases, and its potential to mediate resistance to anti-angiogenic therapy in lung metastases is not established. Here, we examined the mechanism of tumour vascularization in 164 human lung metastasis specimens (composed of breast, colorectal and renal cancer lung metastasis cases). We identified four distinct histopathological growth patterns (HGPs) of lung metastasis (alveolar, interstitial, perivascular cuffing, and pushing), each of which vascularized via a different mechanism. In the alveolar HGP, cancer cells invaded the alveolar air spaces, facilitating the co-option of alveolar capillaries. In the interstitial HGP, cancer cells invaded the alveolar walls to co-opt alveolar capillaries. In the perivascular cuffing HGP, cancer cells grew by co-opting larger vessels of the lung. Only in the pushing HGP did the tumours vascularize by angiogenesis. Importantly, vessel co-option occurred with high frequency, being present in >80% of the cases examined. Moreover, we provide evidence that vessel co-option mediates resistance to the anti-angiogenic drug sunitinib in preclinical lung metastasis models. Assuming that our interpretation of the data is correct, we conclude that vessel co-option in lung metastases occurs through at least three distinct mechanisms, that vessel co-option occurs frequently in lung metastases, and that vessel co-option could mediate resistance to anti-angiogenic therapy in lung metastases. Novel therapies designed to target both angiogenesis and vessel co-option are therefore warranted. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Capilares/efeitos dos fármacos , Humanos , Imunoterapia/métodos , Indóis/uso terapêutico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Modelos Biológicos , Pirróis/uso terapêutico , Sunitinibe
4.
Biochem Biophys Res Commun ; 465(3): 338-43, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26241677

RESUMO

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane-associated glycoprotein that is highly and selectively expressed on the plasma membranes of tumor cells, cultured endothelial cells, and, in vivo, on tumor-associated endothelium. Immunofluorescence microscopy also demonstrated TM4SF1 in cytoplasm and, tentatively, within nuclei. With monoclonal antibody 8G4, and the finer resolution afforded by immuno-nanogold transmission electron microscopy, we now demonstrate TM4SF1 in uncoated cytoplasmic vesicles, nuclear pores and nucleoplasm. Because of its prominent surface location on tumor cells and tumor-associated endothelium, TM4SF1 has potential as a dual therapeutic target using an antibody drug conjugate (ADC) approach. For ADC to be successful, antibodies reacting with cell surface antigens must be internalized for delivery of associated toxins to intracellular targets. We now report that 8G4 is efficiently taken up into cultured endothelial cells by uncoated vesicles in a dynamin-dependent, clathrin-independent manner. It is then transported along microtubules through the cytoplasm and passes through nuclear pores into the nucleus. These findings validate TM4SF1 as an attractive candidate for cancer therapy with antibody-bound toxins that have the capacity to react with either cytoplasmic or nuclear targets in tumor cells or tumor-associated vascular endothelium.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Superfície/imunologia , Clatrina/imunologia , Dinaminas/imunologia , Células Endoteliais/imunologia , Proteínas de Neoplasias/imunologia , Frações Subcelulares/imunologia , Células Cultivadas , Células Endoteliais/ultraestrutura , Humanos
5.
Blood ; 121(11): 2154-64, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23315169

RESUMO

Angiogenesis plays an important role in cancer and in many other human diseases. Vascular endothelial growth factor-A (VEGF-A), the best known angiogenic factor, was originally discovered as a potent vascular permeability factor (VPF), suggesting that other vascular permeabilizing agents, such as histamine and serotonin, might also have angiogenic activity. We recently demonstrated that, like VEGF-A, histamine and serotonin up-regulate the orphan nuclear receptor and transcription factor TR3 (mouse homolog Nur77) and that TR3/Nur77 is essential for their vascular permeabilizing activities. We now report that histamine and serotonin are also angiogenic factors that, at low micromolar concentrations, induce endothelial cell proliferation, migration and tube formation in vitro, and angiogenesis in vivo. All of these responses are mediated through specific histamine and serotonin receptors, are independent of VEGF-A, and are directly dependent on TR3/Nur77. Initially, the angiogenic response closely resembled that induced by VEGF-A, with generation of "mother" vessels. However, after ~10 days, mother vessels began to regress as histamine and serotonin, unlike VEGF-A, up-regulated the potent angiogenesis inhibitor thrombospondin-1, thereby triggering a negative feedback loop. Thus, histamine and serotonin induce an angiogenic response that fits the time scale of acute inflammation.


Assuntos
Histamina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Serotonina/farmacologia , Trombospondina 1/fisiologia , Animais , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Neovascularização Fisiológica/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Trombospondina 1/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
Angiogenesis ; 17(4): 897-907, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24986520

RESUMO

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane glycoprotein that regulates cell motility and proliferation. TM4SF1 is an attractive cancer target because of its high expression in both tumor cells and on the vascular endothelial cells lining tumor blood vessels. We generated mouse monoclonal antibodies against human TM4SF1 in order to evaluate their therapeutic potential; 13 of the antibodies we generated reacted with extracellular loop-2 (EL2), TM4SF1's larger extracellular, lumen-facing domain. However, none of these antibodies reacted with mouse TM4SF1, likely because the EL2 of mouse TM4SF1 differs significantly from that of its human counterpart. Therefore, to test our antibodies in vivo, we employed an established model of engineered human vessels in which human endothelial colony-forming cells (ECFC) and human mesenchymal stem cells (MSC) are incorporated into Matrigel plugs that are implanted subcutaneously in immunodeficient nude mice. We modified the original protocol by (1) preculturing human ECFC on laminin, fibronectin, and collagen-coated plates, and (2) increasing the ECFC/MSC ratio. These modifications significantly increased the human vascular network in Matrigel implants. Two injections of one of our anti-TM4SF1 EL2 monoclonal antibodies, 8G4, effectively eliminated the human vascular component present in these plugs; they also abrogated human PC3 prostate cancer cells that were incorporated into the ECFC/MSC Matrigel mix. Together, these studies provide a mouse model for assessing tumor xenografts that are supplied by a human vascular network and demonstrate that anti-TM4SF1 antibodies such as 8G4 hold promise for cancer therapy.


Assuntos
Antígenos de Superfície/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Neovascularização Patológica , Engenharia Tecidual/métodos , Transcrição Gênica
7.
Blood ; 120(11): 2167-73, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22674805

RESUMO

VEGF induces vascular permeability (VP) in ischemic diseases and cancer, leading to many pathophysiological consequences. The molecular mechanisms by which VEGF acts to induce hyperpermeability are poorly understood and in vivo models that easily facilitate real-time, genetic studies of VP do not exist. In the present study, we report a heat-inducible VEGF transgenic zebrafish (Danio rerio) model through which VP can be monitored in real time. Using this approach with morpholino-mediated gene knock-down and knockout mice, we describe a novel role of phospholipase Cß3 as a negative regulator of VEGF-mediated VP by regulating intracellular Ca2+ release. Our results suggest an important effect of PLCß3 on VP and provide a new model with which to identify genetic regulators of VP crucial to several disease processes.


Assuntos
Permeabilidade Capilar , Endotélio Vascular/metabolismo , Fosfolipase C beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Geneticamente Modificados , Sinalização do Cálcio/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Embrião não Mamífero , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Resposta ao Choque Térmico , Ensaios de Triagem em Larga Escala , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Knockout , Morfolinos/farmacologia , Fosfolipase C beta/antagonistas & inibidores , Fosfolipase C beta/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Cancer Cell ; 10(2): 159-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904613

RESUMO

Endothelial cells in growing tumors express activated Akt, which when modeled by transgenic endothelial expression of myrAkt1 was sufficient to recapitulate the abnormal structural and functional features of tumor blood vessels in nontumor tissues. Sustained endothelial Akt activation caused increased blood vessel size and generalized edema from chronic vascular permeability, while acute permeability in response to VEGF-A was unaffected. These changes were reversible, demonstrating an ongoing requirement for Akt signaling for the maintenance of these phenotypes. Furthermore, rapamycin inhibited endothelial Akt signaling, vascular changes from myrAkt1, tumor growth, and tumor vascular permeability. Akt signaling in the tumor vascular stroma was sensitive to rapamycin, suggesting that rapamycin may affect tumor growth in part by acting as a vascular Akt inhibitor.


Assuntos
Células Endoteliais/patologia , Endotélio Vascular/patologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Animais , Permeabilidade Capilar , Células Cultivadas , Edema/metabolismo , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/fisiologia
9.
Proc Natl Acad Sci U S A ; 108(29): 12066-71, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21730126

RESUMO

Low-level basal vascular permeability (BVP) provides nutrients to normal tissues, and increased vascular permeability is characteristic of inflammation and cancer. We recently reported that VEGF-A, a potent vascular permeabilizing and angiogenic factor, exerts much of its angiogenic activity by up-regulating expression of TR3/Nur77, an orphan nuclear transcription factor, in vascular endothelial cells (EC). To determine whether TR3/Nur77 had a more general role in regulating vascular permeability, we found that histamine, serotonin, and platelet-activating factor, small molecule vascular permeabilizing agents, also increased TR3/Nur77 expression acutely in EC. BVP and the acute vascular hyperpermeability (AVH) induced by these vascular permeabilizing factors were greatly decreased in Nur77(-/-) mice, and both BVP and AVH correlated with Nur77 expression levels in several different mouse strains. BVP and AVH were enhanced in transgenic mice in which Nur77 was selectively overexpressed in vascular EC, whereas both were suppressed in mice overexpressing dominant-negative Nur77. Chronic vascular hyperpermeability (CVH) was induced long before the onset of angiogenesis in a modified, in vivo Matrigel assay that included PT67 cells packaging retroviruses expressing Nur77-sense, whereas inclusion of cells packaging viruses expressing Nur77-antisense prevented VEGF-A-induced CVH. TR3/Nur77 modulated vascular permeability by increasing endothelial nitric-oxide synthase expression and by downregulating several EC junction proteins that maintain vascular homeostasis. Both functions required TR3/Nur77 transcriptional activity. Taking these data together, TR3/Nur77 is up-regulated by several vascular permeabilizing agents and has critical roles in mediating BVP, AVH, and CVH.


Assuntos
Permeabilidade Capilar/fisiologia , Regulação da Expressão Gênica/fisiologia , Junções Intercelulares/fisiologia , Microvasos/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Análise de Variância , Animais , Western Blotting , Linhagem Celular , Colágeno , Primers do DNA/genética , Combinação de Medicamentos , Imuno-Histoquímica , Laminina , Camundongos , Camundongos Transgênicos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Reação em Cadeia da Polimerase , Proteoglicanas
10.
J Exp Med ; 204(6): 1431-40, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17535974

RESUMO

Lymphatic vessel growth, or lymphangiogenesis, is regulated by vascular endothelial growth factor-C (VEGF-C) and -D via VEGF receptor 3 (VEGFR-3). Recent studies suggest that VEGF, which does not bind to VEGFR-3, can also induce lymphangiogenesis through unknown mechanisms. To dissect the receptor pathway that triggers VEGFR-3-independent lymphangiogenesis, we used both transgenic and adenoviral overexpression of placenta growth factor (PlGF) and VEGF-E, which are specific activators of VEGFR-1 and -2, respectively. Unlike PlGF, VEGF-E induced circumferential lymphatic vessel hyperplasia, but essentially no new vessel sprouting, when transduced into mouse skin via adenoviral vectors. This effect was not inhibited by blocking VEGF-C and -D. Postnatal lymphatic hyperplasia, without increased density of lymphatic vessels, was also detected in transgenic mice expressing VEGF-E in the skin, but not in mice expressing PlGF. Surprisingly, VEGF-E induced lymphatic hyperplasia postnatally, and it did not rescue the loss of lymphatic vessels in transgenic embryos where VEGF-C and VEGF-D were blocked. Our data suggests that VEGFR-2 signals promote lymphatic vessel enlargement, but unlike in the blood vessels, are not involved in vessel sprouting to generate new lymphatic vessels in vivo.


Assuntos
Linfangiogênese/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoviridae , Animais , Western Blotting , Bromodesoxiuridina , Primers do DNA , Vetores Genéticos/genética , Camundongos , Camundongos Transgênicos , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Cancer Cell ; 7(3): 251-61, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15766663

RESUMO

Angiogenesis inhibitors, such as TNP-470 and the nontoxic HPMA copolymer-TNP-470 (caplostatin), are emerging as a class of anticancer drugs. We report that TNP-470 and caplostatin inhibit vascular hyperpermeability of tumor blood vessels as well as that induced in mouse skin by different mediators. Treatment with TNP-470 or angiostatin for 3 days was sufficient to reduce permeability of tumor blood vessels, delayed-type hypersensitivity, and pulmonary edema induced by IL-2. TNP-470 also inhibited VPF/VEGF-induced phosphorylation of VEGFR-2, calcium influx, and RhoA activation in endothelial cells. These results identify an activity of TNP-470, that of inhibiting vessel hyperpermeability. This activity likely contributes to TNP-470's antiangiogenic effect and suggests that caplostatin can be used in the treatment of cancer and inflammation.


Assuntos
Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Capilares/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Sesquiterpenos/química , Sesquiterpenos/farmacologia , Angiostatinas/farmacologia , Animais , Cálcio/metabolismo , Capilares/metabolismo , Capilares/ultraestrutura , Permeabilidade Capilar/fisiologia , Movimento Celular/efeitos dos fármacos , Cicloexanos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Feminino , Hipersensibilidade Tardia , Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , O-(Cloroacetilcarbamoil)fumagilol , Edema Pulmonar/induzido quimicamente , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Pele/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
J Exp Med ; 203(3): 719-29, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16520388

RESUMO

Vascular endothelial growth factor (VEGF)-A has essential roles in vasculogenesis and angiogenesis, but the downstream steps and mechanisms by which human VEGF-A acts are incompletely understood. We report here that human VEGF-A exerts much of its angiogenic activity by up-regulating the expression of TR3 (mouse homologue Nur77), an immediate-early response gene and orphan nuclear receptor transcription factor previously implicated in tumor cell, lymphocyte, and neuronal growth and apoptosis. Overexpression of TR3 in human umbilical vein endothelial cells (HUVECs) resulted in VEGF-A-independent proliferation, survival, and induction of several cell cycle genes, whereas expression of antisense TR3 abrogated the response to VEGF-A in these assays and also inhibited tube formation. Nur77 was highly expressed in several types of VEGF-A-dependent pathological angiogenesis in vivo. Also, using a novel endothelial cell-selective retroviral targeting system, overexpression of Nur77 DNA potently induced angiogenesis in the absence of exogenous VEGF-A, whereas Nur77 antisense strongly inhibited VEGF-A-induced angiogenesis. B16F1 melanoma growth and angiogenesis were greatly inhibited in Nur77-/- mice. Mechanistic studies with TR3/Nur77 mutants revealed that TR3/Nur77 exerted most of its effects on cultured HUVECs and its pro-angiogenic effects in vivo, through its transactivation and DNA binding domains (i.e., through transcriptional activity).


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Feminino , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Estrutura Terciária de Proteína/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Retroviridae , Fatores de Transcrição/genética , Transdução Genética/métodos , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
13.
Angiogenesis ; 14(3): 345-54, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21626280

RESUMO

Transmembrane-4-L-six-family-1 (TM4SF1) is a tetraspanin-like membrane protein that is highly and selectively expressed by cultured endothelial cells (EC) and, in vivo, by EC lining angiogenic tumor blood vessels. TM4SF1 is necessary for the formation of unusually long (up to a 50 µm), thin (~100-300 nm wide), F-actin-poor EC cell projections that we term 'nanopodia'. Immunostaining of nanopodia at both the light and electron microsopic levels localized TM4SF1 in a regularly spaced, banded pattern, forming TM4FS1-enriched domains. Live cell imaging of GFP-transduced HUVEC demonstrated that EC project nanopodia as they migrate and interact with neighboring cells. When TM4SF1 mRNA levels in EC were increased from the normal ~90 mRNA copies/cell to ~400 copies/cell through adenoviral transduction, EC projected more and longer nanopodia from the entire cell circumference but were unable to polarize or migrate effectively. When fibroblasts, which normally express TM4SF1 at ~5 copies/cell, were transduced to express TM4SF1 at EC-like levels, they formed typical TM4SF1-banded nanopodia, and broadened, EC-like lamellipodia. Mass-spectrometry demonstrated that TM4SF1 interacted with myosin-10 and ß-actin, proteins involved in filopodia formation and cell migration. In summary, TM4SF1, like genuine tetraspanins, serves as a molecular organizer that interacts with membrane and cytoskeleton-associated proteins and uniquely initiates the formation of nanopodia and facilitates cell polarization and migration.


Assuntos
Antígenos de Superfície/metabolismo , Estruturas da Membrana Celular/metabolismo , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Pseudópodes/metabolismo , Antígenos de Superfície/genética , Estruturas da Membrana Celular/genética , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Fibroblastos/citologia , Humanos , Proteínas de Neoplasias/genética , Pseudópodes/genética
14.
J Surg Oncol ; 103(6): 468-74, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21480238

RESUMO

Tumor blood vessels are heterogeneous, of at least six distinct types, are induced primarily by vascular endothelial growth factor-A (VEGF-A), and provide a potentially useful therapeutic target. Breast cancer is characterized by changes in the microenvironment that result in altered tensional homeostasis. Also, breast cancers arise as the result of epigenetic as well as genetic changes. Tumor blood vessel pericytes result, in part, from bone marrow precursor cells, and VEGF is a negative regulator of glioblastoma tumor cell invasion.


Assuntos
Progressão da Doença , Terapia de Alvo Molecular , Neoplasias/irrigação sanguínea , Neoplasias/fisiopatologia , Neovascularização Patológica/fisiopatologia , Microambiente Tumoral , Inibidores da Angiogênese/uso terapêutico , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/fisiopatologia , Feminino , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/fisiopatologia , Neoplasias/genética , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
15.
Nat Med ; 8(8): 831-40, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12091877

RESUMO

The therapeutic potential of placental growth factor (PlGF) and its receptor Flt1 in angiogenesis is poorly understood. Here, we report that PlGF stimulated angiogenesis and collateral growth in ischemic heart and limb with at least a comparable efficiency to vascular endothelial growth factor (VEGF). An antibody against Flt1 suppressed neovascularization in tumors and ischemic retina, and angiogenesis and inflammatory joint destruction in autoimmune arthritis. Anti-Flt1 also reduced atherosclerotic plaque growth and vulnerability, but the atheroprotective effect was not attributable to reduced plaque neovascularization. Inhibition of VEGF receptor Flk1 did not affect arthritis or atherosclerosis, indicating that inhibition of Flk1-driven angiogenesis alone was not sufficient to halt disease progression. The anti-inflammatory effects of anti-Flt1 were attributable to reduced mobilization of bone marrow-derived myeloid progenitors into the peripheral blood; impaired infiltration of Flt1-expressing leukocytes in inflamed tissues; and defective activation of myeloid cells. Thus, PlGF and Flt1 constitute potential candidates for therapeutic modulation of angiogenesis and inflammation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Arteriosclerose/tratamento farmacológico , Artrite Experimental/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica , Proteínas da Gravidez/farmacologia , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Arteriosclerose/imunologia , Arteriosclerose/patologia , Artrite Experimental/patologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/farmacologia , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/tratamento farmacológico , Isquemia/patologia , Articulações/patologia , Linfocinas/farmacologia , Camundongos , Camundongos Nus , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/imunologia , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Curr Opin Hematol ; 17(3): 225-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20375889

RESUMO

PURPOSE OF REVIEW: The blood vasculature supplies tissues with nutrients, clears waste products, and carries and directs leukocytes to inflammatory sites. To accomplish these functions, microvessels regulate the extravasation of small molecules, plasma proteins and inflammatory cells. The mechanisms responsible for these events have been the subject of intense investigation and, often, dispute. RECENT FINDINGS: Recent progress has contributed to a better understanding of the mechanisms by which microvessels of different types and in different vascular beds regulate the passage of small and large molecules and cells. Roles are shown for the glycocalyx, caveolae, perictyes, sphingosine-1-phosphate, and newly discovered signaling pathways. SUMMARY: Vascular permeability is important for maintaining homeostasis and is greatly increased in acute and chronic inflammation, wound healing, and growing tumors. New work has contributed importantly to the mechanisms responsible for regulating permeability.


Assuntos
Proteínas Sanguíneas/metabolismo , Vasos Sanguíneos/fisiologia , Permeabilidade Capilar/fisiologia , Permeabilidade da Membrana Celular , Animais , Humanos , Transdução de Sinais
17.
Front Cell Dev Biol ; 9: 660609, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33834026

RESUMO

It is widely believed that vascular endothelial growth factor (VEGF) induces angiogenesis by its direct mitogenic and motogenic actions on vascular endothelial cells. However, these activities are only detected when endothelial cells are cultured at very low (0.1%) serum concentrations and would not be expected to take place at the much higher serum levels found in angiogenic sites in vivo. This conundrum can be resolved by recalling VEGF's original function, that of an extremely potent vascular permeability factor (VPF). In vivo VPF/VEGF increases microvascular permeability such that whole plasma leaks into the tissues where it undergoes clotting by tissue factor that is expressed on tumor and host connective tissue cells to deposit fibrin and generate serum. By providing tissue support and by reprogramming the gene expression patterns of cells locally, fibrin and serum can together account for the formation of vascular connective tissue stroma. In sum, by increasing vascular permeability, VPF/VEGF triggers the "wound healing response," setting in motion a fundamental pathophysiological process that induces the mature stroma that is found not only in healing wounds but also in solid tumors and chronic inflammatory diseases. Once initiated by increased vascular permeability, this response may be difficult to impede, perhaps contributing to the limited success of anti-VEGF therapies in treating cancer.

18.
J Exp Med ; 196(11): 1497-506, 2002 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-12461084

RESUMO

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF, VEGF-A) is a multifunctional cytokine with important roles in pathological angiogenesis. Using an adenoviral vector engineered to express murine VEGF-A(164), we previously investigated the steps and mechanisms by which this cytokine induced the formation of new blood vessels in adult immunodeficient mice and demonstrated that the newly formed blood vessels closely resembled those found in VEGF-A-expressing tumors. We now report that, in addition to inducing angiogenesis, VEGF-A(164) also induces a strong lymphangiogenic response. This finding was unanticipated because lymphangiogenesis has been thought to be mediated by other members of the VPF/VEGF family, namely, VEGF-C and VEGF-D. The new "giant" lymphatics generated by VEGF-A(164) were structurally and functionally abnormal: greatly enlarged with incompetent valves, sluggish flow, and delayed lymph clearance. They closely resembled the large lymphatics found in lymphangiomas/lymphatic malformations, perhaps implicating VEGF-A in the pathogenesis of these lesions. Whereas the angiogenic response was maintained only as long as VEGF-A was expressed, giant lymphatics, once formed, became VEGF-A independent and persisted indefinitely, long after VEGF-A expression ceased. These findings raise the possibility that similar, abnormal lymphatics develop in other pathologies in which VEGF-A is overexpressed, e.g., malignant tumors and chronic inflammation.


Assuntos
Indutores da Angiogênese/fisiologia , Fatores de Crescimento Endotelial/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Sistema Linfático/patologia , Linfocinas/fisiologia , Neovascularização Patológica/etiologia , Adenoviridae/genética , Animais , Feminino , Metástase Linfática , Sistema Linfático/fisiologia , Linfoma/etiologia , Camundongos , Timidina/metabolismo , Fator A de Crescimento do Endotélio Vascular , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Fatores de Crescimento do Endotélio Vascular
19.
Am J Pathol ; 175(4): 1768-76, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19729487

RESUMO

Caveolin-1, the signature protein of endothelial cell caveolae, has many important functions in vascular cells. Caveolae are thought to be the transcellular pathway by which plasma proteins cross normal capillary endothelium, but, unexpectedly, cav-1(-/-) mice, which lack caveolae, have increased permeability to plasma albumin. The acute increase in vascular permeability induced by agents such as vascular endothelial growth factor (VEGF)-A occurs through venules, not capillaries, and particularly through the vesiculo-vacuolar organelle (VVO), a unique structure composed of numerous interconnecting vesicles and vacuoles that together span the venular endothelium from lumen to ablumen. Furthermore, the hyperpermeable blood vessels found in pathological angiogenesis, mother vessels, are derived from venules. The present experiments made use of cav-1(-/-) mice to investigate the relationship between caveolae and VVOs and the roles of caveolin-1 in VVO structure in the acute vascular hyperpermeability induced by VEGF-A and in pathological angiogenesis and associated chronic vascular hyperpermeability. We found that VVOs expressed caveolin-1 variably but, in contrast to caveolae, were present in normal numbers and with apparently unaltered structure in cav-1(-/-) mice. Nonetheless, VEGF-A-induced hyperpermeability was strikingly reduced in cav-1(-/-) mice, as was pathological angiogenesis and associated chronic vascular hyperpermeability, whether induced by VEGF-A(164) or by a tumor. Thus, caveolin-1 is not necessary for VVO structure but may have important roles in regulating VVO function in acute vascular hyperpermeability and angiogenesis.


Assuntos
Permeabilidade Capilar/fisiologia , Caveolina 1/deficiência , Neovascularização Patológica/fisiopatologia , Adenoviridae , Animais , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Pele/irrigação sanguínea , Pele/patologia , Pele/ultraestrutura , Vacúolos/metabolismo , Vacúolos/ultraestrutura , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Semin Thromb Hemost ; 36(3): 321-31, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20490982

RESUMO

The blood vessels supplying tumors are strikingly heterogeneous and differ from their normal counterparts with respect to organization, structure, and function. Six distinctly different tumor vessel types have been identified, and much has been learned about the steps and mechanisms by which they form. Four of the six vessel types (mother vessels, capillaries, glomeruloid microvascular proliferations, and vascular malformations) develop from preexisting normal venules and capillaries by angiogenesis. The two remaining vessel types (feeder arteries and draining veins) develop from arterio-venogenesis, a parallel, poorly understood process that involves the remodeling of preexisting arteries and veins. All six of these tumor vessel types can be induced to form sequentially in normal mouse tissues by an adenoviral vector expressing vascular endothelial growth factor (VEGF)-A164. Current antiangiogenic cancer therapies directed at VEGF-A or its receptors have been of only limited benefit to cancer patients, perhaps because they target only the endothelial cells of the tumor blood vessel subset that requires exogenous VEGF-A for maintenance. A goal of future work is to identify therapeutic targets on tumor blood vessel endothelial cells that have lost this requirement.


Assuntos
Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/patologia , Inibidores da Angiogênese/uso terapêutico , Animais , Humanos , Fenótipo , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA