Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 35: 101387, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30703581

RESUMO

The molecular mechanisms involved in induced pluripotent stem cells (iPSCs) generation are poorly understood. The cell death machinery of apoptosis-inducing caspases have been shown to facilitate the process of iPSCs reprogramming. However, the effect of other cell death processes, such as programmed necrosis (necroptosis), on iPSCs induction has not been studied. In this study, we investigated the role of receptor-interacting protein kinase 3 (RIP3), an essential regulator of necroptosis, in reprogramming mouse embryonic fibroblast cells (MEFs) into iPSCs. RIP3 was found to be upregulated in iPSCs compared to MEFs. Deletion of RIP3 dramatically suppressed the reprogramming of iPSCs (~82%). RNA-seq analysis and qRT-PCR showed that RIP3 KO MEFs expressed lower levels of genes that control cell cycle progression and cell division and higher levels of extracellular matrix-regulating genes. The growth rate of RIP3 KO MEFs was significantly slower than WT MEFs. These findings can partially explain the inhibitory effects of RIP3 deletion on iPSCs generation and show for the first time that the necroptosis kinase RIP3 plays an important role in iPSC reprogramming. In contrast to RIP3, the kinase and scaffolding functions of RIPK1 appeared to have distinct effects on reprogramming.


Assuntos
Ciclo Celular , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Regulação para Cima , Animais , Apoptose , Camundongos , Camundongos Knockout , Necrose , Proteína Serina-Treonina Quinases de Interação com Receptores/genética
2.
Sci Rep ; 8(1): 461, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29323137

RESUMO

Contradictory data have been presented regarding the implication of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome in age-related macular degeneration (AMD), the leading cause of vision loss in the Western world. Recognizing that antibody specificity may explain this discrepancy and in line with recent National Institutes of Health (NIH) guidelines requiring authentication of key biological resources, the specificity of anti-NLRP3 antibodies was assessed to elucidate whether non-immune RPE cells express NLRP3. Using validated resources, NLRP3 was not detected in human primary or human established RPE cell lines under multiple inflammasome-priming conditions, including purported NLRP3 stimuli in RPE such as DICER1 deletion and Alu RNA transfection. Furthermore, NLRP3 was below detection limits in ex vivo macular RPE from AMD patients, as well as in human induced pluripotent stem cell (hiPSC)-derived RPE from patients with overactive NLRP3 syndrome (Chronic infantile neurologic cutaneous and articulate, CINCA syndrome). Evidence presented in this study provides new data regarding the interpretation of published results reporting NLRP3 expression and upregulation in RPE and addresses the role that this inflammasome plays in AMD pathogenesis.


Assuntos
Anticorpos/análise , Degeneração Macular/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Epitélio Pigmentado da Retina/metabolismo , Elementos Alu , Animais , Especificidade de Anticorpos , Linhagem Celular , RNA Helicases DEAD-box/genética , Modelos Animais de Doenças , Deleção de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Degeneração Macular/genética , Camundongos , Epitélio Pigmentado da Retina/citologia , Ribonuclease III/genética , Células THP-1
3.
J Ethnopharmacol ; 193: 397-407, 2016 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-27660013

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Hot aqueous extracts of the plant Barleria lupulina (BL) are used for treating inflammatory conditions and diabetic vascular complications. AIM OF THE STUDY: The goal was to identify active compounds in hot aqueous extracts of BL (HAE-BL) that are consistent with a role in reducing inflammation and reducing the vascular pathology associated with diabetes. In particular, we examined activation of the Nrf2 cell defense pathway because our initial findings indicated that HAE-BL activates Nrf2, and because Nrf2 is known to suppress inflammation. Activation of Nrf2 by HAE-BL has not been described previously. MATERIALS AND METHODS: Human endothelial cells, real-time PCR, western blotting, cytoskeletal analyses, and assay-guided fractionation with HPLC were used to identify specific compounds in HAE-BL that activate the Nrf2 cell defense pathway and reduce markers of inflammation in vitro. RESULTS: HAE-BL potently activated the Nrf2 cell defense pathway in endothelial cells consistent with its traditional use and reported success in reducing inflammation. Assay guided fractionation with HPLC identified three alkyl catechols: 4-ethylcatechol, 4-vinylcatechol, and 4-methylcatechol, that are each potent Nrf2 activators. In addition to activating Nrf2, HAE-BL and akyl catechols each profoundly improved organization of the endothelial cell actin cytoskeleton, reduced actin stress fibers, organized cell-cell junctions, and induced expression of mRNA encoding claudin-5 that is important for formation of endothelial tight junctions and reducing vascular leak. CONCLUSIONS: HAE-BL contains important alkyl catechols that potently activate the Nrf2 cell defense pathway, improve organization of the endothelial cell cytoskeleton, and organize tight cell junctions. All of these properties are consistent with a role in reducing inflammation and reducing vascular leak. Because activation of the Nrf2 cell defense pathway also prevents cancers, neuro-degeneration, age-related macular degeneration, and also reduces the severity of chronic obstructive pulmonary disorder and multiple sclerosis, HAE-BL warrants additional consideration for these other serious disorders.


Assuntos
Acanthaceae/química , Actinas/metabolismo , Anti-Inflamatórios/farmacologia , Células Endoteliais/efeitos dos fármacos , Inflamação/prevenção & controle , Microvasos/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Compostos Fitoquímicos/farmacologia , Extratos Vegetais/farmacologia , Fibras de Estresse/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Catecóis/isolamento & purificação , Catecóis/farmacologia , Células Cultivadas , Claudina-5/genética , Claudina-5/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Microvasos/metabolismo , Microvasos/patologia , Fator 2 Relacionado a NF-E2/genética , Compostos Fitoquímicos/química , Compostos Fitoquímicos/isolamento & purificação , Fitoterapia , Componentes Aéreos da Planta , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Transdução de Sinais , Fibras de Estresse/metabolismo , Fibras de Estresse/patologia , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Fatores de Tempo , Regulação para Cima
4.
Int J Oncol ; 45(6): 2311-24, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25215935

RESUMO

Recent studies suggest that the anti-diabetic drug metformin may reduce the risk of cancer and have anti-proliferative effects for some but not all cancers. In this study, we examined the effects of metformin on human retinoblastoma cell proliferation in vitro and in vivo. Two different human retinoblastoma cell lines (Y79, WERI) were treated with metformin in vitro and xenografts of Y79 cells were established in nu/nu immune-deficient mice and used to assess the effects of pharmacological levels of metformin in vivo. Metformin inhibited proliferation of the retinoblastoma cells in vitro. Similar to other studies, high concentrations of metformin (mM) blocked the cell cycle in G0­G1, indicated by a strong decrease of G1 cyclins, especially cyclin D, cyclin-dependent kinases (4 and 6), and flow cytometry assessment of the cell cycle. This was associated with activation of AMPK, inhibition of the mTOR pathways and autophagy marker LC3B. However, metformin failed to suppress growth of xenografted tumors of Y79 human retinoblastoma cells in nu/nu mice, even when treated with a maximally tolerated dose level achieved in human patients. In conclusion, suprapharmacological levels (mM) of metformin, well above those tolerated in vivo, inhibited the proliferation of retinoblastoma cells in vitro. However, physiological levels of metformin, such as seen in the clinical setting, did not affect the growth of retinoblastoma cells in vitro or in vivo. This suggests that the potential beneficial effects of metformin seen in epidemiological studies may be limited to specific tumor types or be related to indirect effects/mechanisms not observed under acute laboratory conditions.


Assuntos
Proliferação de Células/efeitos dos fármacos , Metformina/administração & dosagem , Retinoblastoma/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/biossíntese , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Retinoblastoma/genética , Retinoblastoma/patologia , Serina-Treonina Quinases TOR/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochim Biophys Acta ; 1812(4): 549-57, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20804843

RESUMO

In ischemic retinopathies, underlying hypoxia drives abnormal neovascularization that damages retina and causes blindness. The abnormal neovasculature is tortuous and leaky and fails to alleviate hypoxia, resulting in more pathological neovascularization and retinal damage. With an established model of ischemic retinopathy we found that calpain inhibitors, when administered in moderation, reduced architectural abnormalities, reduced vascular leakage, and most importantly reduced retinal hypoxia. Mechanistically, these calpain inhibitors improved stability and organization of the actin cytoskeleton in retinal endothelial cells undergoing capillary morphogenesis in vitro, and they similarly improved organization of actin cables within new blood vessels in vivo. Hypoxia induced calpain activity in retinal endothelial cells and severely disrupted the actin cytoskeleton, whereas calpain inhibitors preserved actin cables under hypoxic conditions. Collectively, these findings support the hypothesis that hyper-activation of calpains by hypoxia contributes to disruption of the retinal endothelial cell cytoskeleton, resulting in formation of neovessels that are defective both architecturally and functionally. Modest suppression of calpain activity with calpain inhibitors restores cytoskeletal architecture and promotes formation of a functional neovasculature, thereby reducing underlying hypoxia. In sharp contrast to "anti-angiogenesis" strategies that cannot restore normoxia and may aggravate hypoxia, the therapeutic strategy described here does not inhibit neovascularization. Instead, by improving the function of neovascularization to reduce underlying hypoxia, moderate calpain inhibition offers a method for alleviating retinal ischemia, thereby suggesting a new treatment paradigm based on improvement rather than inhibition of new blood vessel growth.


Assuntos
Calpaína/metabolismo , Inibidores de Cisteína Proteinase/uso terapêutico , Glicoproteínas/uso terapêutico , Hipóxia/tratamento farmacológico , Retina/patologia , Doenças Retinianas/tratamento farmacológico , Neovascularização Retiniana/tratamento farmacológico , Actinas/metabolismo , Animais , Calpaína/antagonistas & inibidores , Calpaína/química , Domínio Catalítico/efeitos dos fármacos , Linhagem Celular , Inibidores de Cisteína Proteinase/farmacologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Glicoproteínas/farmacologia , Humanos , Hipóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Retina/citologia , Retina/efeitos dos fármacos , Doenças Retinianas/patologia , Neovascularização Retiniana/patologia , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/patologia
6.
Blood ; 117(5): 1751-60, 2011 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-21030561

RESUMO

Architecturally defective, leaky blood vessels typify pathologic angiogenesis induced by vascular endothelial growth factor-A (VEGF-A). Such neovascular defects aggravate disease pathology and seriously compromise the therapeutic utility of VEGF. Endothelial cell (EC) transduction with active L61Rac1 strongly improved VEGF-driven angiogenesis in vivo as measured by increased neovascular density, enhanced lumen formation, and reduced vessel leakiness. Conversely, transduction with dominant-negative N17Rac1 strongly inhibited neovascularization. In vitro, active L61Rac1 promoted organization of cortical actin filaments and vascular cords and improved EC-EC junctions, indicating that improved cytoskeletal dynamics are important to the mechanism by which active L61Rac1 rectifies VEGF-driven angiogenesis. SEW2871, a sphingosine 1-phosphate receptor-1 agonist that activates Rac1 in ECs, improved cord formation and EC-EC junctions in vitro similarly to active L61Rac. Moreover, SEW2871 administration in vivo markedly improved VEGF neovessel architecture and reduced neovascular leak. Angiopoietin-1, a cytokine that "normalizes" VEGF neovessels in vivo, activated Rac1 and improved cord formation and EC-EC junctions in vitro comparably to active L61Rac1, and a specific Rac1 inhibitor blocked these effects. These studies distinguish augmentation of Rac1 activity as a means to rectify the pathologic angioarchitecture and dysfunctionality of VEGF neovessels, and they identify a rational pharmacologic strategy for improving VEGF angiogenesis.


Assuntos
Angiopoietina-1/metabolismo , Endotélio Vascular/metabolismo , Neovascularização Patológica , Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Células Cultivadas , Derme/citologia , Derme/metabolismo , Endotélio Vascular/citologia , Prepúcio do Pênis/citologia , Prepúcio do Pênis/metabolismo , Genes Dominantes , Humanos , Immunoblotting , Masculino , Camundongos , Neovascularização Fisiológica , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/genética
7.
Microvasc Res ; 81(1): 34-43, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20849862

RESUMO

Vascular endothelial growth factor-A (VEGF) typically induces abnormal angiogenesis in the adult, thereby aggravating disease pathology and limiting utility of VEGF for therapeutic angiogenesis. To identify strategies for rectifying defects in pathological VEGF neovessels, we investigated consequences of modulating the Rho GTPase Cdc42. In a mouse skin model of VEGF-driven pathological angiogenesis, transduction with active Cdc42 (L28Cdc42) markedly improved VEGF neovessels, as measured by increased lumen formation, enlarged vessel diameter, and enhanced perfusion of macromolecular tracers. Conversely, transduction with dominant negative Cdc42 (N17Cdc42) impaired endothelial cell (EC) assembly into lumenized blood vessels and reduced neovessel diameter and tracer perfusion. In vitro, active Cdc42 improved coordination between actin filaments and microtubules and enhanced formation of vascular cords, suggesting that active Cdc42 rectifies defects in angiogenesis by improving cytoskeletal dynamics and capillary morphogenesis. Analyses of Cdc42 signaling in microvascular ECs indicated that active Cdc42 also inhibits glycogen synthase kinase-3ß (GSK-3ß), a multi-functional serine/threonine protein kinase. Pharmacological inhibition of GSK-3ß improved vascular cord formation in vitro and promoted proper neovessel formation in vivo comparably to active Cdc42, thus linking GSK-3ß inhibition to the mechanism by which active Cdc42 rectifies pathological neovascularization. These studies identify activation of Cdc42 and inhibition of GSK-3ß as novel strategies for correcting abnormalities associated with VEGF-driven angiogenesis, and they suggest new approaches for achieving improved therapeutic neovascularization with VEGF.


Assuntos
Vasos Sanguíneos/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Vasos Sanguíneos/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Nus , Microtúbulos/metabolismo , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Fosforilação/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Tiadiazóis/administração & dosagem , Tiadiazóis/farmacologia , Transdução Genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Proteína cdc42 de Ligação ao GTP/genética
8.
PLoS One ; 5(10): e13612, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21049044

RESUMO

BACKGROUND: Successful neovascularization requires that sprouting endothelial cells (ECs) integrate to form new vascular networks. However, architecturally defective, poorly integrated vessels with blind ends are typical of pathological angiogenesis induced by vascular endothelial growth factor-A (VEGF), thereby limiting the utility of VEGF for therapeutic angiogenesis and aggravating ischemia-related pathologies. Here we investigated the possibility that over-exuberant calpain activity is responsible for aberrant VEGF neovessel architecture and integration. Calpains are a family of intracellular calcium-dependent, non-lysosomal cysteine proteases that regulate cellular functions through proteolysis of numerous substrates. METHODOLOGY/PRINCIPAL FINDINGS: In a mouse skin model of VEGF-driven angiogenesis, retroviral transduction with dominant-negative (DN) calpain-I promoted neovessel integration and lumen formation, reduced blind ends, and improved vascular perfusion. Moderate doses of calpain inhibitor-I improved VEGF-driven angiogenesis similarly to DN calpain-I. Conversely, retroviral transduction with wild-type (WT) calpain-I abolished neovessel integration and lumen formation. In vitro, moderate suppression of calpain activity with DN calpain-I or calpain inhibitor-I increased the microtubule-stabilizing protein tau in endothelial cells (ECs), increased the average length of microtubules, increased actin cable length, and increased the interconnectivity of vascular cords. Conversely, WT calpain-I diminished tau, collapsed microtubules, disrupted actin cables, and inhibited integration of cord networks. Consistent with the critical importance of microtubules for vascular network integration, the microtubule-stabilizing agent taxol supported vascular cord integration whereas microtubule dissolution with nocodazole collapsed cord networks. CONCLUSIONS/SIGNIFICANCE: These findings implicate VEGF-induction of calpain activity and impairment of cytoskeletal dynamics in the failure of VEGF-induced neovessels to form and integrate properly. Accordingly, calpain represents an important target for rectifying key vascular defects associated with pathological angiogenesis and for improving therapeutic angiogenesis with VEGF.


Assuntos
Calpaína/metabolismo , Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Calpaína/genética , Linhagem Celular , Genes Dominantes , Camundongos , Morfogênese , Mutação , Pele/irrigação sanguínea , Transdução Genética
9.
Angiogenesis ; 13(3): 269-77, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20809259

RESUMO

In ischemic retinopathies, unrelieved hypoxia induces the formation of architecturally abnormal, leaky blood vessels that damage retina and ultimately can cause blindness. Because these newly formed blood vessels are functionally defective, they fail to alleviate underlying hypoxia, resulting in more pathological neovascularization and more damage to retina. With an established model of ischemic retinopathy, we investigated inhibition of glycogen synthase kinase-3ß (GSK-3ß) as a means for improving the architecture and functionality of pathological blood vessels in retina. In vitro, hypoxia increased GSK-3ß activity in retinal endothelial cells, reduced ß-catenin, and correspondingly impaired integrity of cell/cell junctions. Conversely, GSK-3ß inhibitors restored ß-catenin, improved cell/cell junctions, and enhanced the formation of capillary cords in three-dimensional collagen matrix. In vivo, GSK-3ß inhibitors, at appropriately moderate doses, strongly reduced abnormal vascular tufts, reduced abnormal vascular leakage, and improved vascular coverage and perfusion during the proliferative phase of ischemia-driven retinal neovascularization. Most importantly, these improvements in neovasculature were accompanied by marked reduction in retinal hypoxia, relative to controls. Thus, GSK-3ß inhibitors offer a promising strategy for alleviating retinal hypoxia by correcting key vascular defects typically associated with ischemia-driven neovascularization.


Assuntos
Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hipóxia/tratamento farmacológico , Isquemia/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Retina/patologia , Neovascularização Retiniana/tratamento farmacológico , Doenças Vasculares/tratamento farmacológico , Animais , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Hipóxia/complicações , Hipóxia/patologia , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Isquemia/complicações , Isquemia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fluxo Sanguíneo Regional , Retina/efeitos dos fármacos , Retina/enzimologia , Retina/metabolismo , Neovascularização Retiniana/complicações , Neovascularização Retiniana/enzimologia , Neovascularização Retiniana/patologia , Doenças Vasculares/complicações , Doenças Vasculares/patologia , beta Catenina/metabolismo
10.
Mol Cancer Res ; 4(11): 811-20, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17114339

RESUMO

Down syndrome candidate region 1 (DSCR1) is one of more than 50 genes located in a region of chromosome 21 that has been implicated in Down syndrome. DSCR1 can be expressed as four isoforms, one of which, isoform 4 (DSCR1-4), has recently been found to be strongly induced by vascular endothelial growth factor A (VEGF-A(165)) and to provide a negative feedback loop that inhibits VEGF-A(165)-induced endothelial cell proliferation in vitro and angiogenesis in vivo. We report here that another DSCR1 isoform, DSCR1-1L, is also up-regulated by VEGF-A(165) in cultured endothelial cells and is strongly expressed in several types of pathologic angiogenesis in vivo. In contrast to DSCR1-4, the overexpression of DSCR1-1L induced the proliferation and activation of the transcription factor NFAT in cultured endothelial cells and promoted angiogenesis in Matrigel assays in vivo, even in the absence of VEGF-A. Similarly, small interfering RNAs specific for DSCR1-1L and DSCR1-4 had opposing inhibitory and stimulatory effects, respectively, on these same functions. DSCR1-4 is thought to inhibit angiogenesis by inactivating calcineurin, thereby preventing activation and nuclear translocation of NFAT, a key transcription factor. In contrast, DSCR1-1L, regulated by a different promoter than DSCR1-4, activates NFAT and its proangiogenic activity is inhibited by cyclosporin, an inhibitor of calcineurin. In sum, DSCR1-1L, unlike DSCR1-4, potently activates angiogenesis and could be an attractive target for antiangiogenesis therapy.


Assuntos
Calcineurina/metabolismo , Proteínas Musculares/fisiologia , Fatores de Transcrição NFATC/agonistas , Neovascularização Patológica/genética , Transporte Ativo do Núcleo Celular , Animais , Inibidores de Calcineurina , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Proteínas de Ligação a DNA , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Laminina/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Chaperonas Moleculares , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Fragmentos de Peptídeos/farmacologia , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Proteínas/antagonistas & inibidores , Proteínas/genética , Proteínas/fisiologia , Proteoglicanas/metabolismo , Interferência de RNA , RNA Longo não Codificante , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transfecção , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/farmacologia
11.
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
12.
Methods Mol Biol ; 294: 269-85, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15576918

RESUMO

Angiogenesis is a complex process involving the organization of proliferating endothelial cells into new blood vessels. Both in vivo models and in vitro models are important for investigating angiogenesis and for defining the involvement of specific molecules. This chapter describes a basic mouse model of vascular endothelial growth factor-driven angiogenesis in mouse skin together with a modified version of this model in which retrovirus-packaging cells are included as a means to efficiently achieve retroviral transduction in vivo. With this approach, the contributions of specific proteins to angiogenesis can be defined. In addition, we describe a model of capillary morphogenesis in vitro that uses microvascular endothelial cells transduced with retrovirus in culture. This in vitro model provides a complementary strategy for investigating the importance of specific molecules for angiogenesis.


Assuntos
Divisão Celular/fisiologia , Endotélio Vascular/citologia , Neovascularização Fisiológica/fisiologia , Pele/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Capilares/efeitos dos fármacos , Capilares/fisiologia , Endotélio Vascular/fisiologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Masculino , Mamíferos , Camundongos , Camundongos Nus , Microcirculação/efeitos dos fármacos , Microcirculação/fisiologia , Modelos Animais , Morfogênese , Neovascularização Fisiológica/efeitos dos fármacos
13.
J Biol Chem ; 279(21): 22377-86, 2004 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-14970227

RESUMO

Tumor necrosis factor (TNF) alpha-converting enzyme (TACE/ADAM-17) has diverse roles in the proteolytic processing of cell surface molecules and, due to its ability to process TNFalpha, is a validated therapeutic target for anti-inflammatory therapies. Unlike a number of other ADAM proteins, which interact with integrin receptors via their disintegrin domains, there is currently no evidence for an ADAM-17-integrin association. By analyzing the adhesion of a series of cell lines with recombinant fragments of the extracellular domain of ADAM-17, we now demonstrate a functional interaction between ADAM-17 and alpha(5)beta(1) integrin in a trans orientation. Because ADAM-17-mediated adhesion was sensitive to RGD peptides and EDTA, and the integrin-binding site within ADAM-17 was narrowed down to the disintegrin/cysteine-rich region, the two molecules appear to have a ligand-receptor relationship mediated by the alpha(5)beta(1) ligand binding pocket. Intriguingly, ADAM-17 and alpha(5)beta(1) were found to co-localize in both membrane ruffles and focal adhesions in HeLa cells. When confluent HeLa cell monolayers were wounded, ADAM-17 and alpha(5)beta(1) redistributed to the leading edge and co-localized, which is suggestive of a cis orientation. We postulate that the interaction of ADAM-17 with alpha(5)beta(1) may target or modulate its metalloproteolytic activity.


Assuntos
Integrina alfa5beta1/metabolismo , Metaloendopeptidases/metabolismo , Proteínas ADAM , Proteína ADAM17 , Animais , Anticorpos Monoclonais/química , Sítios de Ligação , Western Blotting , Células COS , Cátions , Adesão Celular , Linhagem Celular , Movimento Celular , Separação Celular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ácido Edético/química , Fibroblastos/metabolismo , Citometria de Fluxo , Células HeLa , Humanos , Inflamação , Ligantes , Microscopia de Fluorescência , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Pele/citologia , Cicatrização
14.
Proc Natl Acad Sci U S A ; 101(7): 1874-9, 2004 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-14769914

RESUMO

The mechanisms that control organization of endothelial cells (ECs) into new blood vessels are poorly understood. We hypothesized that the GTPase Rho, which regulates cytoskeletal architecture, is important for EC organization during neovascularization. To test this hypothesis, we designed a highly versatile mouse skin model that used vascular endothelial growth factor-expressing cells together with packaging cells producing retroviruses encoding RhoA GTPase mutants. In this animal model, dominant negative N19RhoA selectively impaired assembly of ECs into new blood vessels; and, in contrast, active V14RhoA stimulated ECs to form blood vessels with functional lumens. In vitro, dominant negative N19RhoA reduced EC actin stress fibers and prevented ECs from contracting and reorganizing into precapillary cords within collagen gels. In contrast, active V14RhoA promoted EC stress fiber formation, contractility, and organization into cords. Neither N19RhoA nor V14RhoA significantly affected EC proliferation or migration in vitro; and, similarly, neither mutant significantly affected EC density during angiogenesis in vivo. Thus, these studies identify a critical and selective role for Rho activity in regulating EC assembly into new blood vessels, and they identify both negative and positive manipulation of Rho activity, respectively, as strategies for suppressing or promoting the organizational stages of neovascularization.


Assuntos
Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Neovascularização Fisiológica , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Divisão Celular , Movimento Celular , Colágeno/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Modelos Animais , Mutação , Perfusão , Fibras de Estresse/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína rhoA de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...