Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Cell Sci ; 126(Pt 9): 1952-61, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23447675

RESUMO

Amyloid ß peptides (Aß1-40 and Aß1-42) cause cerebral degeneration by impairing the activity of angiogenic factors and inducing apoptosis and senescence in the endothelium. Amyloid peptides are known to induce oxidative stress. Impairment of mitochondrial aldehyde dehydrogenase 2 (ALDH2) following oxidative stress, results in accumulation of toxic aldehydes, particularly 4-hydroxynoneal (4-HNE). We sought to determine the role of mitochondrial ALDH2 in Aß-related impairment of angiogenesis. We hypothesized that by increasing the detoxification activity of ALDH2 we would reduce Aß-driven endothelial injuries and restore angiogenesis. We used a selective ALDH2 activator, Alda-1, assessing its ability to repair mitochondrial dysfunction in the endothelium. Treatment of human endothelial cells with Aß1-40 (5-50 µM) induced loss of mitochondrial membrane potential, increased cytochrome c release and ROS accumulation. These events were associated with 4-HNE accumulation and decrease in ALDH2 activity (40%), and resulted in disassembly of endothelial junctions, as evidenced by ß-catenin phosphorylation, disorganization of adherens and tight junctions, and by disruption of pseudocapillary formation. Alda-1 (10-40 µM) abolished Aß-induced 4-HNE accumulation, apoptosis and vascular leakiness, fully restoring the pro-angiogenic endothelial phenotype and responses to FGF-2. Our data document that mitochondrial ALDH2 in the endothelium is a target for the vascular effect of Aß, including loss of barrier function and angiogenesis. ALDH2 activation, by restoring mitochondrial functions in the endothelium, prevents Aß-induced dysfunction and anti-angiogenic effects. Thus, agents activating ALDH2 may reduce endothelial injuries including those occurring in cerebral amyloid angiopathy, preserving the angiogenic potential of the endothelium.


Assuntos
Aldeído Desidrogenase/metabolismo , Peptídeos beta-Amiloides/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Mitocondriais/metabolismo , Neovascularização Fisiológica , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Aldeído Desidrogenase/genética , Aldeído-Desidrogenase Mitocondrial , Peptídeos beta-Amiloides/metabolismo , Benzamidas/farmacologia , Benzodioxóis/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Proteínas Mitocondriais/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Espécies Reativas de Oxigênio/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
2.
J Pharmacol Exp Ther ; 351(3): 500-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25238748

RESUMO

At the cardiovascular level, nitric oxide (NO) controls smooth muscle functions, maintains vascular integrity, and exerts an antihypertensive effect. Metal-nonoates are a recently discovered class of NO donors, with NO release modulated through the complexation of the N-aminoethylpiperazine N-diazeniumdiolate ligand to metal ions, and thus representing a significant innovation with respect to the drugs traditionally used. In this study, we characterized the vascular protective effects of the most effective compound of this class, Ni(PipNONO)Cl, compared with the commercial N-diazeniumdiolate group derivate, diethylenetriamine/nitric oxide (DETA/NO). Ni(PipNONO)Cl induced a concentration-dependent relaxation of precontracted rat aortic rings. The ED50 was 0.67 µM, compared with 4.3 µM obtained with DETA/NO. When tested on cultured microvascular endothelial cells, Ni(PipNONO)Cl exerted a protective effect on the endothelium, promoting cell proliferation and survival in the picomolar range. The administration of Ni(PipNONO)Cl to vascular smooth muscle cells reduced the cell number, promoting their apoptosis at a high concentration (10 µM). Inhibition of smooth muscle cell migration, a hallmark of atherosclerosis, was accompanied by cytoskeletal rearrangement and loss of lamellipodia. When added to isolated platelets, Ni(PipNONO)Cl significantly reduced ADP-induced aggregation. Since atherosclerosis is accompanied by an inflammatory environment, cultured endothelial cells were exposed to interleukin (IL)-1ß. In the presence of IL-1ß, Ni(PipNONO)Cl inhibited cyclooxygenase-2 and inducible nitric oxide synthase upregulation, and reduced endothelial permeability and the platelet and monocyte adhesion markers CD31 and CD40 at the plasma membrane. Overall, these data indicate that Ni(PipNONO)Cl exerts vascular protective effects relevant for vascular dysfunction and prevention of atherosclerosis and thrombosis.


Assuntos
Cardiotônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Cardiotônicos/química , Proliferação de Células/fisiologia , Relação Dose-Resposta a Droga , Endotélio Vascular/fisiologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Masculino , Músculo Liso Vascular/fisiologia , Doadores de Óxido Nítrico/química , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
3.
FASEB J ; 24(7): 2385-95, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20207941

RESUMO

Cerebral amyloid angiopathy (CAA) caused by amyloid beta (Abeta) deposition around brain microvessels results in vascular degenerative changes. Antiangiogenic Abeta properties are known to contribute to the compromised cerebrovascular architecture. Here we hypothesize that Abeta peptides impair angiogenesis by causing endothelial cells to enter senescence at an early stage of vascular development. Wild-type (WT) Abeta and its mutated variant E22Q peptide, endowed with marked vascular tropism, were used in this study. In vivo, in zebrafish embryos, the WT or E22Q peptides reduced embryo survival with an IC(50) of 6.1 and 4.7 microM, respectively. The 2.5 microM concentration, showing minimal toxicity, was chosen. Alkaline phosphatase staining revealed disorganized vessel patterning, narrowing, and reduced branching of vessels. Beta-galactosidase staining and the cyclin-dependent kinase inhibitor p21 expression, indicative of senescence, were increased. In vitro, WT and E22Q reduced endothelial cell survival with an IC(50) of 12.3 and 8.8 microM, respectively. The 5 microM concentration, devoid of acute effects on the endothelium, was applied chronically to long-term cultured human umbilical vein endothelial cells (HUVECs). We observed reduced cumulative population doubling, which coincided with beta-galactosidase accumulation, down-regulation of telomerase reverse-transcriptase mRNA expression, decreased telomerase activity, and p21 activation. Senescent HUVECs showed marked angiogenesis impairment, as Abeta treatment reduced tube sprouting. The endothelial injuries caused by the E22Q peptide were much more aggressive than those induced by the WT peptide. Premature Abeta-induced senescence of the endothelium, producing progressive alterations of microvessel morphology and functions, may represent one of the underlying mechanisms for sporadic or heritable CAA.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Senescência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Inibidores da Angiogênese , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Angiopatia Amiloide Cerebral , Embrião não Mamífero/efeitos dos fármacos , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Endotélio Vascular/patologia , Humanos , Mutação de Sentido Incorreto , Neovascularização Fisiológica/efeitos dos fármacos , Taxa de Sobrevida , Peixe-Zebra
4.
Exp Cell Res ; 315(3): 385-95, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19061884

RESUMO

Single point mutations of the amyloid precursor protein generate Abeta variants bearing amino acid substitutions at positions 21-23. These mutants are associated with distinct hereditary phenotypes of cerebral amyloid angiopathy, manifesting varying degrees of tropism for brain vessels, and impaired microvessel remodeling and angiogenesis. We examined the differential effects of E22Q (Dutch), and E22G (Arctic) variants in comparison to WT Abeta on brain endothelial cell proliferation, angiogenic phenotype expression triggered by fibroblast growth factor (FGF-2), pseudo-capillary sprouting, and induction of apoptosis. E22Q exhibited a potent anti-angiogenic profile in contrast to E22G, which had a much weaker effect. Investigations on the FGF-2 signaling pathway revealed the greatest differences among the peptides: E22Q and WT peptides suppressed FGF-2 expression while E22G had barely any effect. Phosphorylation of the FGF-2 receptor, FGFR-1, and the survival signal Akt were abolished by E22Q and WT peptides, but not by E22G. The biological dissimilar effect of the mutant and WT peptides on cerebral EC cannot be assigned to a particular Abeta structure, suggesting that the toxic effect of the Abeta assemblies goes beyond mere multimerization.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/genética , Apoptose , Células Cultivadas , Humanos , Microvasos/citologia , Mutação , Fragmentos de Peptídeos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
5.
FASEB J ; 21(10): 2418-30, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17384145

RESUMO

In squamous cell carcinoma, the levels of nitric oxide (NO) derived from inducible NO synthase (iNOS) and prostaglandin E2 (PGE2) derived from cyclooxygenase-2 (COX-2) originated from tumor cells or tumor-associated inflammatory cells have been reported to correlate with tumor growth, metastasis, and angiogenesis. The present study examined the role of the iNOS signaling pathway in PGE2-mediated tumor invasiveness and proliferation in squamous cell carcinoma, A431, and SCC-9 cells. Cell invasion and proliferation promoted by PGE2 were blocked by iNOS silencing RNA or iNOS/guanylate cyclase (GC) pharmacological inhibition. Consistently, iNOS-GC pathway inhibitors blocked mitogen-activated protein kinase-ERK1/2 phosphorylation, which was required to mediate PGE2 functions. In vivo, in A431 cells implanted in nude mice, GC inhibition also decreased the tumor proliferation index and ERK1/2 activation. PGE2 effects were confined to the selective stimulation of the EP2 receptor subtype, leading to epidermal growth factor receptor (EGFR) transactivation via protein kinase A (PKA) and c-Src activation. EP2-mediated ERK1/2 activation and cell functions were abolished by inhibitors of PKA, c-Src, and EGFR, as well as by inhibiting iNOS pathway. Silencing of iNOS also impaired EGFR-induced ERK1/2 phosphorylation. These results indicate that iNOS/GC signaling is a downstream player in the control of EP2/EGFR-mediated tumor cell proliferation and invasion.


Assuntos
Carcinoma de Células Escamosas/patologia , Divisão Celular/fisiologia , Receptores ErbB/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/fisiologia , Receptores de Prostaglandina E/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Imuno-Histoquímica , Proteína Quinase 3 Ativada por Mitógeno/genética , Invasividade Neoplásica , Óxido Nítrico Sintase Tipo II/genética , RNA Interferente Pequeno/genética , Receptores de Prostaglandina E Subtipo EP2 , Proteínas Recombinantes/metabolismo , Ativação Transcricional , Transfecção
6.
Bone ; 41(4): 523-34, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17681892

RESUMO

In mineralized tissue the process of angiogenesis is required for normal osteogenesis during bone repair and in reconstructive and substitutive surgery, for proper biomaterial/tissue integration. Nanotechnologies have been proposed to improve the compatibility of biomaterials for use in orthopaedic and reconstructive surgery (e.g., nanocrystals). The aim of this study was to determine the effect of nanostructured hydroxyapatite (HA) on angiogenesis. Microvascular endothelial cell survival, proliferation and migration, crucial events in the angiogenic process, were evaluated together with cytoskeleton and biochemical signalling markers. Induction of migration, metalloproteinase (MMP-2) and focal adhesion Kinase (FAK) activity documented the ability of HA nanocrystals to stimulate capillary endothelium toward an angiogenic phenotype. HA concentrations, ranging from 2 to 10 microg/ml, promoted endothelium survival and proliferation, preserved alphavbeta3-integrin localization, stimulated beta-actin reorganization and Akt phosphorylation (98% vs control). Immunoassays for key signalling pathways in angiogenesis (i.e., endothelial nitric oxide synthase (eNOS) and fibroblast growth factor-2 (FGF-2)) demonstrated that HA increased their expression. Moreover, quantitative RT-PCR and Western blotting analysis confirmed that HA nanocrystals exposure up-regulated FGF-2 mRNA by 6 fold and increased 18 kDa protein isoform by 40%. HA enhanced cell responsiveness to vascular endothelial growth factor (VEGF) in terms of NOS activity (1.5 fold over control), increasing the ability of microvascular endothelium to differentiate into capillary-like structures when grown in 3D fibrin gel. In conclusion our data document the proangiogenic properties of HA nanocrystals. This material stimulates endothelial cell functions and biochemical pathways to an extent similar to VEGF, and primes them to VEGF stimulation, leading to differentiation in pseudocapillary formations in 3D matrices.


Assuntos
Durapatita/química , Durapatita/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Nanopartículas/química , Regulação para Cima/efeitos dos fármacos , Animais , Bovinos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Endotélio/irrigação sanguínea , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Nanopartículas/ultraestrutura , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais
7.
J Biomed Mater Res A ; 76(3): 656-63, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16294324

RESUMO

To favor bone reconstitution with biomaterials endothelial cells should maintain proper functions to drive angiogenesis. To this aim nanocrystals of hydroxyapatite (HA) have been synthesized and characterized on endothelial cells. Microvascular endothelial cells have been exposed to stoichiometric HA nanocrystals. Cell morphology and organization of cytoskeletal proteins have been monitored by SEM analysis and immunofluorescence. Biochemical markers of physiological and pathological responses of endothelial cells, endothelial constitutive nitric oxide synthase, and cycloxygenase-2 (ecNOS and COX-2, respectively) have been measured by immunofluorescence. Crystallized HA sustained endothelial survival without any cytotoxic effect. At the observation with SEM, endothelial cell morphology was maintained in the presence of HA. The localization and organization of beta-actin documented the formation of stress fibers, indicating an activation of endothelial cells induced by HA nanocrystals. Immunohistochemistry for biochemical key signaling pathways in endothelium demonstrated that nanocrystals of HA maintained the expression of ecNOS and did not increase COX-2 expression. In conclusion, the present findings indicate that HA nanocrystals exhibit high biocompatibility for microvascular endothelium. In the presence of HA nanocrystals endothelial cells maintain biochemical markers of healthy endothelium. They do not acquire a proinflammatory or thrombogenic phenotype, but express markers of functioning endothelium that might contribute to angiogenesis.


Assuntos
Durapatita , Células Endoteliais/metabolismo , Nanoestruturas , Actinas/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular , Células Cultivadas , Ciclo-Oxigenase 2/biossíntese , Durapatita/efeitos adversos , Células Endoteliais/citologia , Humanos , Imuno-Histoquímica , Teste de Materiais/métodos , Proteínas de Membrana/biossíntese , Nanoestruturas/efeitos adversos , Óxido Nítrico Sintase Tipo III/biossíntese , Transdução de Sinais , Fibras de Estresse/metabolismo
8.
Cell Cycle ; 8(8): 1271-8, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19305142

RESUMO

In acute thromboembolic stroke, neurological damage is due to ischemia-induced apoptotic death of neuronal cells and the surrounding vascular network. Here, we demonstrate that the BH4 domain of the anti-apoptotic protein, Bcl-x(L), attached to the membrane transport peptide, TAT, reduces stroke injury after intracerebroventricular infusion into immature rats subjected to carotid artery ligation and additional exposure to hypoxia. The injected TAT-BH4 entered neuron bodies, maintained brain architecture, protected neuronal and endothelial cells from apoptosis and promoted neuronal stem cell recruitment. In vitro, TAT-BH4 enhanced the survival of endothelial cells exposed to H(2)O(2), increased neuronal differentiation, and induced axonal remodelling of adult neuronal stem cells. These findings indicate that TAT-BH4 administration protects against acute hypoxia/ischemia injury in the brain by preventing endothelial and neuron cell apoptosis and by inducing neuronal plasticity.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/prevenção & controle , Membrana Celular/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Inibidores de Caspase , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipóxia Encefálica/tratamento farmacológico , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/prevenção & controle , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
9.
J Biol Chem ; 283(4): 2139-46, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18042549

RESUMO

Prostaglandin E(2) (PGE(2)) behaves as a mitogen in epithelial tumor cells as well as in many other cell types. We investigated the actions of PGE(2) on microvascular endothelial cells (capillary venular endothelial cells) with the purpose of delineating the signaling pathway leading to the acquisition of the angiogenic phenotype and to new vessel formation. PGE(2) (100 nM) produced activation of the fibroblast growth factor receptor 1 (FGFR-1), as measured by its phosphorylation, but not of vascular endothelial growth factor receptor 2. PGE(2) stimulated the EP3 subtype receptor, as deduced by abrogation of EP3 Galpha(i) subunit activity through pertussis toxin. Consistent with this result, in human umbilical venular endothelial cells missing the EP3 receptor, PGE(2) did not phosphorylate FGFR-1. Upon binding to its receptor, PGE(2) initiated an autocrine/paracrine signaling cascade involving the intracellular activation of c-Src, activation of matrix metalloproteinase (predominantly MMP2), which in turn caused the mobilization of membrane-anchored fibroblast growth factor-2 (FGF-2). In fact, in cells unable to release FGF-2 the transfection with both FGFR-1 and EP3 did not result in FGFR-1 phosphorylation in response to PGE(2). Relevance for the FGF2-FGFR-1 system was highlighted by confocal analysis, showing receptor internalization after cell exposure to the prostanoid. ERK1/2 appeared to be the distal signal involved, its phosphorylation being sensitive to either cSrc inhibitor or FGFR-1 blocker. Finally, PGE(2) stimulated cell migration and capillary formation in aortic rings, which were severely reduced by inhibitors of signaling molecules or by receptor antagonist. In conclusion, this study provides evidence for the involvement of FGFR-1 through FGF2 in eliciting PGE(2) angiogenic responses. This signaling pattern is similar to the autocrine-paracrine mechanism which operates in endothelial cells to support neovascular growth.


Assuntos
Dinoprostona/farmacologia , Células Endoteliais/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neovascularização Fisiológica/fisiologia , Ocitócicos/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/fisiologia , Células CHO , Capilares/citologia , Capilares/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Cricetinae , Cricetulus , Dinoprostona/metabolismo , Células Endoteliais/citologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células Epiteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Genes src/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Ocitócicos/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/fisiologia , Fosforilação/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP3
10.
Int J Cancer ; 120(5): 995-1004, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17131343

RESUMO

Src tyrosine kinase family cooperates with activated growth factor receptors to regulate growth, invasion and metastasis. The authors examined the influence of a novel c-Src inhibitor, 1l, derived from 4-amino-substituted-pyrazolo-pyrimidines, on tumor angiogenesis and on the angiogenic output of squamous carcinoma cells, A431 and SCC-4. The effect of 1l was assessed on growth and microvessel density in A431 tumors and its effect compared with the established c-Src inhibitor PP-1. The effects of c-Src inhibition were investigated on vascular endothelial growth factor (VEGF) expression and activity in tumor cells grown in vivo and in vitro, as well as on VEGF mediated signaling and on endothelial cell functions. Nanomolar concentrations of 1l decreased tumor volume promoted by A431 implanted in nude mice, without affecting in vitro cell tumor survival. This effect was related to 1l inhibition of VEGF production, and secondary to an effect on tumor microvessel density. The rabbit cornea assay confirmed that 1l markedly decreased neovessel growth induced by VEGF. In cultured endothelial cells, 1l inhibited the VEGF-induced phosphorylation on tyr416 of c-Src, resulting in a reduced cell proliferation and invasion. Consistently, 1l dowregulated endothelial nitric oxide synthase, MAPK-extracellular receptor kinase 1-2 (ERK1-2) activity and matrix metalloproteinases (MMP-2/MMP-9), while the tissue inhibitors of metalloproteinases (TIMP2/TIMP-1) were upregulated. These results demonstrate that nM concentrations of c-Src kinase inhibitors (1l and PP-1), by reducing the production of VEGF released by tumor cell and its endothelial cell responses, have a highly selective antiangiogenesis effect, which might be useful in combination therapies.


Assuntos
Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/enzimologia , Neovascularização Patológica/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Carcinoma de Células Escamosas/patologia , Humanos , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Nus , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Neovascularização Patológica/patologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Coelhos , Transdução de Sinais/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Células Tumorais Cultivadas , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Pharmacol Exp Ther ; 319(2): 515-22, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16868034

RESUMO

The beneficial effect exerted by angiotensin-converting enzyme inhibitors (ACEI) on vascular endothelium has been attributed to restoration of endothelial cell survival properties and improvement of angiogenesis. Fibroblast growth factor (FGF)-2 is an angiogenic factor for the microvascular endothelium, which tonically promotes endothelial cell growth and survival through an autocrine/paracrine mechanism. Here, we formulate the hypothesis that FGF-2 might contribute to the prosurvival/proangiogenic effect of ACEI. We investigated zofenoprilat and, in selected experiments, lisinopril, as representatives of ACEI. These compounds induced formation of pseudocapillaries in vessel fragments isolated from porcine coronary and human umbilical arteries by increasing endothelial cell growth up to 5-fold. Angiogenesis was abolished by inhibitors of nitric-oxide synthase (NOS) pathway and by anti-FGF-2 antibodies. Consistently, in cultured coronary endothelial cells (CVECs), ACEI up-regulated endothelial NOS (eNOS) and FGF-2 and induced mitogen-activated protein kinase extracellular signal-regulated kinase 1/2 activation. The overexpression of eNOS/FGF-2 produced, at the functional level, enhanced cell proliferation and migration, the latter effect being dose-dependent and maximal at 0.1 microM zofenoprilat. The importance of FGF-2 for the acquisition of the angiogenic phenotype elicited by ACEI was clearly demonstrated by the impairment of endothelial functions following transfection of CVECs with small interference RNA for FGF-2. Moreover, FGF-2 silencing greatly affected the nuclear translocation of the FGF receptor (FGFR)-1, highlighting the autocrine mode of action of FGF-2. At the endothelial membrane level, zofenoprilat appeared to activate the bradykinin B1 receptor, a known stimulant of FGF-2 expression. In conclusion, we show that ACEI exert protective/proangiogenic effects in microvascular coronary endothelial cells by activating the endogenous FGF-2/FGFR-1 system.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Vasos Coronários/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Captopril/análogos & derivados , Captopril/farmacologia , Células Cultivadas , Células Endoteliais/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Receptor B1 da Bradicinina/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/fisiologia , Suínos
12.
Neurourol Urodyn ; 24(4): 389-96, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15605370

RESUMO

AIMS: To investigate the effect of anoxia/glucopenia and re-superfusion on intrinsic nerves in the mammalian urinary bladder. METHODS: Strips of detrusor smooth muscle were dissected from monkey and human urinary bladder and mounted for tension recording in organ baths superfused with Krebs solution. Human, monkey, and guinea-pig urinary bladders were treated to evaluate glycogen contents by a biochemical method. RESULTS: Detrusor strips from both monkeys and humans had to be exposed to anoxia-glucopenia for up to 2-2.5 hr to observe a progressive decline in the response to electrical field stimulation (EFS) of the intrinsic nerves, at variance with guinea-pig detrusor strips. In contrast, the response to direct activation of the smooth muscle with carbachol remained almost unaltered. Incubation of human and monkey detrusor strips with 2-deoxyglucose (2-DG) during 1 hr anoxia-glucopenia, however, caused a marked damage to the intrinsic nerves. The glycogen contents of both human detrusor specimens and monkey urinary bladders were 2.0- and 1.4-fold higher, respectively, than that found in guinea-pig urinary bladder; furthermore, untreated monkey detrusor sections showed a greater number of glycogen granules as compared to those subjected to anoxia-glucopenia and re-superfusion. In guinea-pig and in monkey detrusor sections glycogen granules were found in smooth muscle cells but not in neurons of intramural ganglia. CONCLUSIONS: A higher susceptibility of guinea-pig as compared to monkey and human nerves has been demonstrated; it is suggested that anaerobic glucose metabolism during anoxia-glucopenia is crucial for the functional recovery of detrusor intrinsic nerves from damage caused by anoxia-glucopenia and re-superfusion.


Assuntos
Glucose/metabolismo , Glucose/fisiologia , Hipóxia/fisiopatologia , Músculo Liso/inervação , Músculo Liso/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Bexiga Urinária/inervação , Bexiga Urinária/metabolismo , Anaerobiose , Animais , Antimetabólitos/farmacologia , Carbacol/farmacologia , Cebus , Desoxiglucose/farmacologia , Estimulação Elétrica , Gânglios Autônomos/efeitos dos fármacos , Glicogênio/metabolismo , Glicólise , Cobaias , Humanos , Técnicas In Vitro , Isquemia/metabolismo , Agonistas Muscarínicos/farmacologia , Estresse Oxidativo/fisiologia , Fluxo Sanguíneo Regional/fisiologia
13.
Microvasc Res ; 64(1): 47-55, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12074630

RESUMO

The microfibrils of anchoring filaments, a typical ultrastructural feature of initial lymphatic vessels, consist mainly of fibrillin and are similar to the microfibrils of elastic fibers. As we previously demonstrated, they radiate from focal adhesions of lymphatic endothelium to the perivascular elastic network. Although present in large blood vessels, fibrillin microfibrils have never been detected in blood capillaries. Here we report immunohistochemical evidence that cultured bovine aortic and lymphatic endothelial cells express fibrillin microfibrils. These microfibrils form an irregular web in lymphatic endothelial cells, whereas in blood vessel endothelial cells they are arranged in a honeycomb pattern. Cultured lymphatic and blood vessel endothelial cells also produce focal adhesion molecules: focal adhesion kinase, vinculin, talin, and cytoskeletal beta-actin. Our data suggest that anchoring filaments of initial lymphatic vessels in vivo may be produced by endothelium. Through their connection with focal adhesions, they may form a mechanical anchorage for the thin wall of initial lymphatic vessels and a transduction device for mechanical signals from the extracellular matrix into biochemical signals in endothelial cells. The complex anchoring filaments-focal adhesions may control the permeability of lymphatic endothelium and finely adjust lymph formation to the physiological conditions of the extracellular matrix. The different deposition of fibrillin microfibrils in blood vessel endothelial cells may be related to the necessity of withstanding shear forces. Thus, in our opinion, differences in fibrillin deposition imply a different role of fibrillin in blood vessel and lymphatic endothelium.


Assuntos
Actinas/biossíntese , Endotélio Vascular/citologia , Sistema Linfático/citologia , Proteínas dos Microfilamentos/biossíntese , Proteínas Tirosina Quinases/biossíntese , Talina/biossíntese , Vinculina/biossíntese , Animais , Bovinos , Células Cultivadas , Citoplasma/metabolismo , Fibrilinas , Proteína-Tirosina Quinases de Adesão Focal , Imuno-Histoquímica , Ducto Torácico/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA