Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Cell Sci ; 133(11)2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32409567

RESUMO

During angiogenesis, endothelial cells engage components of the extracellular matrix through integrin-mediated adhesion. Endothelial expression of laminin-411 and laminin-511 is known to promote vessel stability. However, little is known about the contribution of these laminins to endothelial morphogenesis. We used two organotypic cell culture angiogenesis assays, in conjunction with RNAi approaches, to demonstrate that depletion of either the α4 chain of laminin-411 (LAMA4) or the α5 chain of laminin-511 (LAMA5) from endothelial cells inhibits sprouting and tube formation. Depletion of α6 (ITGA6) integrins resulted in similar phenotypes. Gene expression analysis indicated that loss of either laminin-511 or α6 integrins inhibited the expression of CXCR4, a gene previously associated with angiogenic endothelial cells. Pharmacological or RNAi-dependent inhibition of CXCR4 suppressed endothelial sprouting and morphogenesis. Importantly, expression of recombinant CXCR4 rescued endothelial morphogenesis when α6 integrin expression was inhibited. Additionally, the depletion of α6 integrins from established tubes resulted in the loss of tube integrity and laminin-511. Taken together, our results indicate that α6 integrins and laminin-511 can promote endothelial morphogenesis by regulating the expression of CXCR4 and suggest that the α6-dependent deposition of laminin-511 protects the integrity of established endothelial tubes.


Assuntos
Células Endoteliais , Laminina , Adesão Celular , Integrina alfa6/genética , Integrinas , Laminina/genética , Morfogênese/genética , Receptores CXCR4
2.
J Cell Sci ; 127(Pt 3): 534-45, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24284076

RESUMO

Cytokinesis is the final stage in cell division. Although integrins can regulate cytokinesis, the mechanisms involved are not fully understood. In this study, we demonstrate that integrin-regulated ERK (extracellular signal-related kinase) and RSK (p90 ribosomal S6 kinase) signaling promotes successful cytokinesis. Inhibiting the activation of ERK and RSK in CHO cells by a mutation in the integrin ß1 cytoplasmic tail or with pharmacological inhibitors results in the accumulation of cells with midbodies and the formation of binucleated cells. Activation of ERK and RSK signaling by the expression of constitutively active RAF1 suppresses the mutant phenotype in a RSK-dependent manner. Constitutively active RSK2 also restores cytokinesis inhibited by the mutant integrin. Importantly, the regulatory role of the RSK pathway is not specific to CHO cells. MCF-10A human mammary epithelial cells and HPNE human pancreatic ductal epithelial cells exhibit a similar dependence on RSK for successful cytokinesis. In addition, depriving mitotic MCF10A cells of integrin-mediated adhesion by incubating them in suspension suppressed ERK and RSK activation and resulted in a failure of cytokinesis. Furthermore, inhibition of RSK or integrins within the 3D context of a developing salivary gland organ explant also leads to an accumulation of epithelial cells with midbodies, suggesting a similar defect in cytokinesis. Interestingly, neither ERK nor RSK regulates cytokinesis in human fibroblasts, suggesting cell-type specificity. Taken together, our results identify the integrin-RSK signaling axis as an important regulator of cytokinesis in epithelial cells. We propose that the proper interaction of cells with their microenvironment through integrins contributes to the maintenance of genomic stability by promoting the successful completion of cytokinesis.


Assuntos
Células Epiteliais/citologia , Integrina beta1/genética , Integrinas/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Animais , Células CHO , Cricetinae , Cricetulus , Citocinese/genética , Células Epiteliais/metabolismo , Humanos , Integrina beta1/metabolismo , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais/genética
3.
J Biol Chem ; 287(4): 2520-30, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22117069

RESUMO

Microtubule nucleation is an essential step in the formation of the microtubule cytoskeleton. We recently showed that androgen and Src promote microtubule nucleation and γ-tubulin accumulation at the centrosome. Here, we explore the mechanisms by which androgen and Src regulate these processes and ask whether integrins play a role. We perturb integrin function by a tyrosine-to-alanine substitution in membrane-proximal NPIY motif in the integrin ß1 tail and show that this mutant substantially decreases microtubule nucleation and γ-tubulin accumulation at the centrosome. Because androgen stimulation promotes the interaction of the androgen receptor with Src, resulting in PI3K/AKT and MEK/ERK signaling, we asked whether these pathways are inhibited by the mutant integrin and whether they regulate microtubule nucleation. Our results indicate that the formation of the androgen receptor-Src complex and the activation of downstream pathways are significantly suppressed when cells are adhered by the mutant integrin. Inhibitor studies indicate that microtubule nucleation requires MEK/ERK but not PI3K/AKT signaling. Importantly, the expression of activated RAF-1 is sufficient to rescue microtubule nucleation inhibited by the mutant integrin by promoting the centrosomal accumulation of γ-tubulin. Our data define a novel paradigm of integrin signaling, where integrins regulate microtubule nucleation by promoting the formation of androgen receptor-Src signaling complexes to activate the MEK/ERK signaling pathway.


Assuntos
Centrossomo/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Integrina beta1/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Microtúbulos/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Integrina beta1/genética , MAP Quinase Quinase Quinases/genética , Microtúbulos/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
4.
Animal Model Exp Med ; 6(4): 362-368, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37682010

RESUMO

BACKGROUND: Genetic analysis in human patients has linked mutations in PIK3CA, the catalytic subunit of PI-3'Kinase, to sporadic incidences of vascular malformations. METHODS: We have developed a mouse model with inducible and endothelial-specific expression of PIK3CAH1047R , resulting in the development of vascular malformations. Systemic induction of this mutation in adult mice results in rapid lethality, limiting our ability to track and study these lesions; therefore, we developed a topical and local induction protocol using the active metabolite of tamoxifen, 4OH-T, on the ear skin of adults. RESULTS: This approach allows us to successfully model the human disease in a mature and established vascular bed and track the development of vascular malformations. To validate the utility of this model, we applied a topical rapamycin ointment, as rapamycin is therapeutically beneficial to patients in clinical trials. We found that the induced ear lesions showed significant attenuation after treatment, which was easily quantified. CONCLUSIONS: These data collectively provide evidence of a new model to study vascular malformations in adult tissues, which should be particularly useful in environments lacking specialized small-animal imaging facilities.


Assuntos
Sirolimo , Malformações Vasculares , Humanos , Adulto , Animais , Camundongos , Domínio Catalítico , Classe I de Fosfatidilinositol 3-Quinases/genética , Modelos Animais de Doenças , Sirolimo/farmacologia , Malformações Vasculares/genética
5.
J Biol Chem ; 285(10): 7045-55, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20048167

RESUMO

Activation of Src family kinases (SFK) and the subsequent phosphorylation of VE-cadherin have been proposed as major regulatory steps leading to increases in vascular permeability in response to inflammatory mediators and growth factors. To investigate Src signaling in the absence of parallel signaling pathways initiated by growth factors or inflammatory mediators, we activated Src and SFKs by expression of dominant negative Csk, expression of constitutively active Src, or knockdown of Csk. Activation of SFK by overexpression of dominant negative Csk induced VE-cadherin phosphorylation at tyrosines 658, 685, and 731. However, dominant negative Csk expression was unable to induce changes in the monolayer permeability. In contrast, expression of constitutively active Src decreased barrier function and promoted VE-cadherin phosphorylation on tyrosines 658 and 731, although the increase in VE-cadherin phosphorylation preceded the increase in permeability by 4-6 h. Csk knockdown induced VE-cadherin phosphorylation at sites 658 and 731 but did not induce a loss in barrier function. Co-immunoprecipitation and immunofluorescence studies suggest that phosphorylation of those sites did not impair VE-cadherin ability to bind p120 and beta-catenin or the ability of these proteins to localize at the plasma membrane. Taken together, our data show that Src-induced tyrosine phosphorylation of VE-cadherin is not sufficient to promote an increase in endothelial cell monolayer permeability and suggest that signaling leading to changes in vascular permeability in response to inflammatory mediators or growth factors may require VE-cadherin tyrosine phosphorylation concurrently with other signaling pathways to promote loss of barrier function.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular , Tirosina/metabolismo , Quinases da Família src/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Junções Aderentes/metabolismo , Animais , Antígenos CD/genética , Caderinas/genética , Células Cultivadas , Células Endoteliais/citologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Ativação Enzimática , Humanos , Fosforilação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Proteína p120 Ativadora de GTPase/metabolismo , Quinases da Família src/genética
6.
J Cell Physiol ; 226(3): 800-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20803566

RESUMO

Chronic inflammation is tightly linked to diseases associated with endothelial dysfunction including aberrant angiogenesis. To better understand the endothelial role in pro-inflammatory angiogenesis, we analyzed signaling pathways in continuously activated endothelial cells, which were either chronically exposed to soluble TNF or the reactive oxygen species (ROS) generating H2O2, or express active transmembrane TNF. Testing in an in vitro capillary sprout formation assay, continuous endothelial activation increased angiogenesis dependent on activation of p38 MAP kinase, NADPH oxidase, and matrix metalloproteinases (MMP). p38 MAP kinase- and MMP-9-dependent angiogenesis in our assay system may be part of a positive feed forward autocrine loop because continuously activated endothelial cells displayed up-regulated ROS production and subsequent endothelial TNF expression. The pro-angiogenic role of the p38 MAP kinase in continuously activated endothelial cells was in stark contrast to the anti-angiogenic activity of the p38 MAP kinase in unstimulated control endothelial cells. In vivo, using an experimental prostate tumor, pharmacological inhibition of p38 MAP kinase demonstrated a significant reduction in tumor growth and in vessel density, suggesting a pro-angiogenic role of the p38 MAP kinase in pathological angiogenesis in vivo. In conclusion, our results suggest that continuous activation of endothelial cells can cause a switch of the p38 MAP kinase from anti-angiogenic to pro-angiogenic activities in conditions which link oxidative stress and autocrine TNF production.


Assuntos
Inflamação/complicações , Inflamação/enzimologia , Neovascularização Patológica/complicações , Neovascularização Patológica/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neoplasias/patologia , Ratos , Solubilidade/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
7.
Mol Cell Biol ; 26(11): 4201-13, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16705171

RESUMO

Endothelial cell proliferation is a critical step in angiogenesis and requires a coordinated response to soluble growth factors and the extracellular matrix. As focal adhesion kinase (FAK) integrates signals from both adhesion events and growth factor stimulation, we investigated its role in endothelial cell proliferation. Expression of a dominant-negative FAK protein, FAK-related nonkinase (FRNK), impaired phosphorylation of FAK and blocked DNA synthesis in response to multiple angiogenic stimuli. These results coincided with elevated cyclin-dependent kinase inhibitors (CDKIs) p21/Cip and p27/Kip, as a consequence of impaired degradation. FRNK inhibited the expression of Skp2, an F-box protein that targets CDKIs, by inhibiting mitogen-induced mRNA. The FAK-regulated degradation of p27/Kip was Skp2 dependent, while levels of p21/Cip were regulated independent of Skp2. Skp2 is required for endothelial cell proliferation as a consequence of degrading p27. Finally, knockdown of both p21 and p27 in FRNK-expressing cells completely restored mitogen-induced endothelial cell proliferation. These data demonstrate a critical role for FAK in the regulation of CDKIs through two independent mechanisms: Skp2 dependent and Skp2 independent. They also provide important insights into the requirement of focal adhesion kinase for normal vascular development and reveal novel regulatory control points for angiogenesis.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Adesão Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p27/deficiência , Inibidor de Quinase Dependente de Ciclina p27/genética , Células Endoteliais/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Regulação Enzimológica da Expressão Gênica , Humanos , Paxilina/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
8.
Cell Signal ; 19(9): 1949-55, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17629454

RESUMO

We have recently demonstrated that endothelial beta(2) adrenergic receptors (beta(2)AR) regulate eNOS activity and consequently vascular tone, through means of PKB/AKT. In this work we explored the signal transduction pathway leading to AKT/eNOS activation in endothelial cells (EC). Using pharmacological and molecular inhibitors both in cultured EC cells and in ex vivo rat carotid preparations, we found that G(i) coupling of the beta(2)AR is needed for AKT activation and vasorelaxation. Since endothelial activation is sensitive to pertussis toxin but not to G(ibetagamma) inhibition by betaARKct, we conclude that G(alphai) mediates betaAR induced AKT activation. Downstream, betaAR signalling requires the soluble tyrosine kinase SRC, as both in cultured EC and rat carotid, the mutant dominant negative of SRC prevent beta(2)AR induced endothelial activation and vasodilation. In EC, G(alphai) directly interacts with SRC and this interaction leads to SRC activation and phosphorylation in a manner that is regulated by beta(2)AR stimulation. We propose a novel signal transduction pathway for beta(2)AR stimulation trough G(alphai) and SRC, leading to activation of AKT.


Assuntos
Células Endoteliais/enzimologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Adenoviridae , Animais , Células Endoteliais/citologia , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Ativação Enzimática , Técnicas In Vitro , Peptídeos/metabolismo , Ligação Proteica , Ratos , Ratos Endogâmicos WKY , Proteínas Recombinantes/metabolismo , Transdução Genética
9.
Physiol Rep ; 6(12): e13733, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29932504

RESUMO

Nitric oxide, produced by eNOS, plays critical roles in the regulation of vascular function and maintenance. Chronic PI3K signaling has recently been associated with vascular malformations. A well described substrate downstream of PI3K signaling is eNOS. Another critical downstream target of PI3K is the metabolic regulator, mTORc1. The relationship between mTORc1 and eNOS regulation, has not been determined. We generated cells with manipulated PI3K signaling by expressing the activating mutation, PIK3CAH1047R , or knocking down PTEN expression. We investigated eNOSS1177 phosphorylation, a major activating regulatory site, following mTORC1 inhibition. We also tested the sufficiency of mTORc1 activation to stimulate eNOSS1177 phosphorylation. Our data indicate mTORc1 activity is required for the phosphorylation of eNOSS1177 , even in the presence of robust AKT activation. Moreover, we found that expression of RHEB, which functions in the absence of AKT activation to activate mTORc1, is sufficient to phosphorylate this site. Our data indicate that mTORc1, rather than AKT, may be the critical determinant of eNOSS1177 phosphorylation. As mTORc1 is a central regulator of cellular metabolism, the finding that this regulatory complex can directly participate in the regulation of eNOS provides new insights into metabolic uncoupling and vascular disease that often accompanies diabetes, high fat diets, and aging.


Assuntos
Células Endoteliais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Ativação Enzimática/fisiologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/fisiologia , Proteína Enriquecida em Homólogo de Ras do Encéfalo/fisiologia
10.
Cell Rep ; 24(11): 2869-2882, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30208313

RESUMO

Cerebrovascular malformations (CVMs) affect approximately 3% of the population, risking hemorrhagic stroke, seizures, and neurological deficits. Recently Ras mutations have been identified in a majority of brain arterio-venous malformations. We generated an endothelial-specific, inducible HRASV12 mouse model, which results in dilated, proliferative blood vessels in the brain, blood-brain barrier breakdown, intracerebral hemorrhage, and rapid lethality. Organoid morphogenesis models revealed abnormal cessation of proliferation, abnormalities in expression of tip and stalk genes, and a failure to properly form elongating tubes. These defects were influenced by both hyperactive PI-3' kinase signaling and altered TGF-ß signaling. Several phenotypic changes predicted by the in vitro morphogenesis analysis were validated in the mouse model. These data provide a model of brain vascular malformations induced by mutant Ras and reveal insights into intersecting molecular mechanisms in the pathogenesis of brain vascular malformations.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Malformações Vasculares/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Feminino , Imunofluorescência , Imidazóis/farmacologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Pericitos/citologia , Pericitos/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas p21(ras)/genética , Quinoxalinas/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/metabolismo
11.
J Neurosci Methods ; 294: 1-6, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061345

RESUMO

BACKGROUND: Morphometric analyses of biological features have become increasingly common in recent years with such analyses being subject to a large degree of observer bias, variability, and time consumption. While commercial software packages exist to perform these analyses, they are expensive, require extensive user training, and are usually dependent on the observer tracing the morphology. NEW METHOD: To address these issues, we have developed a broadly applicable, no-cost ImageJ plugin we call 'BranchAnalysis2D/3D', to perform morphometric analyses of structures with branching morphologies, such as neuronal dendritic spines, vascular morphology, and primary cilia. RESULTS: Our BranchAnalysis2D/3D algorithm allows for rapid quantification of the length and thickness of branching morphologies, independent of user tracing, in both 2D and 3D data sets. COMPARISON WITH EXISTING METHODS: We validated the performance of BranchAnalysis2D/3D against pre-existing software packages using trained human observers and images from brain and retina. We found that the BranchAnalysis2D/3D algorithm outputs results similar to available software (i.e., Metamorph, AngioTool, Neurolucida), while allowing faster analysis times and unbiased quantification. CONCLUSIONS: BranchAnalysis2D/3D allows inexperienced observers to output results like a trained observer but more efficiently, thereby increasing the consistency, speed, and reliability of morphometric analyses.


Assuntos
Encéfalo/citologia , Imageamento Tridimensional/métodos , Microscopia Confocal/métodos , Neurônios/citologia , Software , Algoritmos , Animais , Camundongos , Variações Dependentes do Observador , Reprodutibilidade dos Testes , Retina/anatomia & histologia
12.
Cancer Res ; 65(1): 148-56, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15665290

RESUMO

The formation of a microvascular endothelium plays a critical role in the growth and metastasis of established tumors. The ability of a fragment from the first type III repeat of fibronectin (III(1C)), anastellin, to suppress tumor growth and metastasis in vivo has been reported to be related to its antiangiogenic properties, however, the mechanism of action of anastellin remains unknown. Utilizing cultures of human dermal microvascular endothelial cells, we provide evidence that anastellin inhibits signaling pathways which regulate the extracellular signal-regulated (ERK) mitogen-activated protein kinase pathway and subsequent expression of cell cycle regulatory proteins. Addition of anastellin to primary microvascular endothelial cells resulted in a complete inhibition of serum-dependent proliferation. Growth inhibition correlated with a decrease in serum-dependent expression of cyclin D1, cyclin A and the cyclin-dependent kinase, cdk4, key regulators of cell cycle progression through G(1) phase. Consistent with a block in G(1)-S transition, anastellin inhibited serum-dependent incorporation of [(3)H]-thymidine into S-phase nuclei. Addition of anastellin to serum-starved microvessel cells resulted in a time-dependent and dose-dependent decrease in basal levels of phosphorylated MEK/ERK and blocked serum-dependent activation of ERK. Adenoviral infection with Ad.DeltaB-Raf:ER, an inducible estrogen receptor-B-Raf fusion protein, restored levels of active ERK in anastellin-treated cells, rescued levels of cyclin D1, cyclin A, and cdk4, and rescued [(3)H]-thymidine incorporation. These data suggest that the antiangiogenic properties of anastellin observed in mouse models of human cancer may be due to its ability to block endothelial cell proliferation by modulating ERK signaling pathways and down-regulating cell cycle regulatory gene expression required for G(1)-S phase progression.


Assuntos
Endotélio Vascular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fibronectinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Adulto , Apoptose/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Humanos , Cinética , Microcirculação/fisiologia , Proteínas Recombinantes/farmacologia , Pele/irrigação sanguínea
13.
Cancer Res ; 65(15): 6493-7, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061624

RESUMO

Experimental evidence suggests that CXCR4, a Gi protein-coupled receptor for the ligand CXCL12/stromal cell-derived factor-1alpha (SDF-1alpha), plays a role in breast cancer metastasis. Transactivation of HER2-neu by G protein-coupled receptor activation has been reported as a ligand-independent mechanism of activating tyrosine kinase receptors. We found that SDF-1alpha transactivated HER2-neu in the breast cancer cell lines MDA-MB-361 and SKBR3, which express both CXCR4 and HER2-neu. AMD3100, a CXCR4 inhibitor, PKI 166, an epidermal growth factor receptor/HER2-neu tyrosine kinase inhibitor, and PP2, a Src kinase inhibitor, each blocked SDF-1alpha-induced HER2-neu phosphorylation. Blocking Src kinase, with PP2 or using a kinase-inactive Src construct, and inhibiting epidermal growth factor receptor/HER2-neu signaling with PKI 166 each inhibited SDF-1alpha-stimulated cell migration. We report a novel mechanism of HER2-neu transactivation through SDF-1alpha stimulation of CXCR4 that involves Src kinase activation.


Assuntos
Neoplasias da Mama/metabolismo , Quimiocinas CXC/fisiologia , Receptor ErbB-2/biossíntese , Quinases da Família src/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Ativação Enzimática , Humanos , Receptor ErbB-2/genética , Transdução de Sinais , Ativação Transcricional , Quinases da Família src/genética
14.
J Cell Biol ; 216(5): 1473-1488, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28416479

RESUMO

Development of wound therapies is hindered by poor understanding of combinatorial integrin function in the epidermis. In this study, we generated mice with epidermis-specific deletion of α3ß1, α9ß1, or both integrins as well as keratinocyte lines expressing these integrin combinations. Consistent with proangiogenic roles for α3ß1, α3-null keratinocytes showed reduced paracrine stimulation of endothelial cell migration and survival, and wounds of epidermis-specific α3 knockout mice displayed impaired angiogenesis. Interestingly, α9ß1 in keratinocytes suppressed α3ß1-mediated stimulation of endothelial cells, and wounds of epidermis-specific α9 knockout mice displayed delayed vascular normalization and reduced endothelial apoptosis, indicating that α9ß1 cross-suppresses α3ß1 proangiogenic functions. Moreover, α9ß1 inhibited α3ß1 signaling downstream of focal adhesion kinase (FAK) autoactivation at the point of Src-mediated phosphorylation of FAK Y861/Y925. Finally, α9ß1 cross-suppressed many α3ß1-dependent genes, including the gene that encodes MMP-9, which we implicated as a regulator of integrin-dependent cross talk to endothelial cells. Our findings identify a novel physiological context for combinatorial integrin signaling, laying the foundation for therapeutic strategies that manipulate α9ß1 and/or α3ß1 during wound healing.


Assuntos
Epiderme/metabolismo , Integrina alfa3beta1/antagonistas & inibidores , Integrinas/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Cicatrização , Animais , Apoptose , Movimento Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina alfa3beta1/deficiência , Integrina alfa3beta1/metabolismo , Integrinas/deficiência , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Ferimentos e Lesões/sangue
15.
Oncogene ; 24(21): 3459-71, 2005 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15735667

RESUMO

Mutations in BRAF, a component of extracellular signal-regulated kinases 1 and 2 (ERK) cascade, are frequent in melanoma. It is important to understand how BRAF mutations contribute to malignant traits including anchorage- and growth factor-independence. We have previously shown that efficient activation of ERK in normal human epidermal melanocytes (NHEM) requires both adhesion to the extracellular matrix and growth factors. Mutant V599E BRAF is sufficient to promote ERK activation independent of adhesion and growth factors. Here, we analysed regulation of G1 cell cycle events in NHEM and human melanoma cells. We show that S phase entry in NHEM requires both adhesion and growth factor signaling through the MEK-ERK pathway. This control correlates with induction of cyclin D1 and downregulation of p27Kip1, two key G1 cell cycle events. In melanoma cells expressing V599E BRAF, cyclin D1 was constitutively expressed independent of adhesion but dependent upon MEK activation and nuclear accumulation of ERK. Reduction of cyclin D1 levels by RNA interference inhibited S phase entry in melanoma cells. Importantly, expression of V599E BRAF in NHEM was sufficient to promote cyclin D1 promoter activity in the absence of adhesion. Additionally, p27Kip1 levels were downregulated in V599E BRAF-expressing melanoma cells and active BRAF was sufficient to downregulate p27Kip1 in serum-starved NHEM. Thus, adhesion-growth factor cooperation, leading to efficient activation of ERK, regulates cyclin D1 and p27Kip1 levels in human melanocytes and mutant BRAF overrides adhesion-growth factor control of these two G1 cell cycle proteins in melanomas. These findings provide important insight into how BRAF mutations contribute to aberrant human melanocyte proliferation.


Assuntos
Adesão Celular , Proteínas de Ciclo Celular/biossíntese , Ciclina D1/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , MAP Quinase Quinase Quinases/farmacologia , Melanoma/genética , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/farmacologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/biossíntese , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Análise Mutacional de DNA , Regulação para Baixo , Genes Supressores de Tumor , Humanos , Melanócitos , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
16.
Oncogene ; 23(6): 1275-82, 2004 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-14676843

RESUMO

Vascular endothelial growth factor (VEGF) induces activation of p38 mitogen-activated protein kinase (MAPK) in primary endothelial cells and may be critical for VEGF-induced angiogenesis. We investigated the molecular basis for p38 activation in response to VEGF. The expression of a C-terminal splice variant of FAK, FRNK, had no affect on VEGF-induced activation of p38; however, expression of a dominant-negative RAFTK/Pyk2 mutant led to a decrease in the activation of p38, but had no affect on extracellular signal-regulated kinase (ERK). Since calcium regulates RAFTK/Pyk2, we investigated its role in p38 activity. Preincubation with EGTA suppressed p38 activation, while calcium ionophore induced p38 activity. Inhibition of phospholipase C (PLC) resulted in complete inhibition of ERK, while having no affect on p38 activity. These data suggested a bifurcation in the regulation of MAPKs that occurs at the level of PLC and RAFTK/Pyk2 activation. Src family kinases interact with RAFTK/Pyk2. Inhibition of Src by either pharmacological or genetic means decreased p38 activity. Finally, we found that both Src and RAFTK/Pyk2 were essential for endothelial cell migration. These data identified a novel regulatory network involving extracellular calcium, RAFTK/Pyk2, Src and p38. This signaling network appears to be critical for VEGF-induced endothelial cell migration.


Assuntos
Quimiotaxia/fisiologia , Endotélio Vascular/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Quinases da Família src/metabolismo , Cálcio/fisiologia , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Meios de Cultura Livres de Soro , Ácido Egtázico/farmacologia , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática , Quinase 2 de Adesão Focal , Humanos , Neovascularização Fisiológica , Veias Umbilicais , Proteínas Quinases p38 Ativadas por Mitógeno
17.
Oncogene ; 23(1): 192-200, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14712224

RESUMO

Angiogenic factors alter endothelial cell phenotype to promote the formation of new blood vessels, a process critical for a number of normal and pathological conditions. Ras is required for the induction of the angiogenic phenotype in response to vascular endothelial growth factor (VEGF). However, VEGF generates many signals, several of which are not dependent upon Ras activation. Our current study investigates the sufficiency of Ras activation for driving angiogenic responses. An activated Ras(V12) mutant induces prominent membrane ruffling, branching morphogenesis on three-dimensional collagen, DNA synthesis, and cell migration in primary endothelial cells. An upregulation of PI3K/AKT, Erk, and Jnk signaling pathways accompany these phenotypic changes. The inhibition of Erk blocked cell proliferation, but only partially attenuated migration. Blocking PI3K had no effect on DNA synthesis, but caused a modest reduction in cell migration. Lastly, Jnk played a significant role in both the proliferation and migration response. These effects of Ras(V12) are not the result of increased autocrine secretion of VEGF. These data suggest that the acquisition of activating Ras mutations can lead to a proangiogenic conversion in the phenotype of primary endothelial cells. Furthermore, these data raise the possibility that chronic Ras activation in endothelial cells may be sufficient to promote angiogenesis and the development of vascular anomalies.


Assuntos
Células Endoteliais/patologia , Genes ras/fisiologia , Neovascularização Patológica/etiologia , Divisão Celular , Células Cultivadas , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fenótipo , Fosfatidilinositol 3-Quinases/fisiologia , Fator A de Crescimento do Endotélio Vascular/análise
18.
Clin Cancer Res ; 9(1): 424-32, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12538496

RESUMO

PURPOSE: Elevated manganese superoxide dismutase (Sod2) levels have been reported to be associated with an increased frequency of tumor invasion and metastasis in certain cancers, and the aim of this study is to examine the molecular mechanisms by which this occurs. EXPERIMENTAL DESIGN: Sod2 and catalase overexpressing HT-1080 fibrosarcoma cell lines were used to evaluate the H(2)O(2)-dependent regulation of matrix metalloproteinase (MMP)-1 promoter activity, mitogen-activated protein (MAP) kinase signaling, DNA-binding activity, and MMP mRNA levels. The invasive and metastatic potential of Sod2 overexpressing cells was characterized using subrenal capsular implantation or tail vein injection of tumor cells into nude mice, respectively. RESULTS: Our data reveal that Sod2 overexpression increases the DNA-binding activity of transcription factors critical for MMP expression but also enhances MMP-1 promoter activity via the Ras//MAP/extracellular signal-regulated kinase (MEK) signaling cascade. A single nucleotide polymorphism that creates an Ets site at position -1607 bp confers Sod2-dependent MMP-1 promoter activity. Sod2 overexpression also increases the mRNA levels of MMPs-2, -3, -7, -10, -9, -11 and enhances the metastatic potential of fibrosarcoma cells when implanted in immunodeficient mice. The Sod2-dependent increases in AP-1 and SP-1 DNA-binding activity, MMP-1 promoter activity, general MMP expression, and collagen degradation can be reversed by the hydrogen peroxide-detoxifying enzyme, catalase. CONCLUSION: MMPs play a critical role in the process of stromal invasion and metastasis, and these findings suggest that the association between increased Sod2 and poor prognosis in certain cancers may be attributed to elevated MMP production.


Assuntos
Peróxido de Hidrogênio/farmacologia , Metaloproteinases da Matriz/metabolismo , Superóxido Dismutase/metabolismo , Animais , Regulação para Baixo , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Células HeLa , Humanos , Luciferases/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Oxirredução , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas
20.
PLoS One ; 7(11): e49222, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23145129

RESUMO

Neurofibromatosis is a well known familial tumor syndrome, however these patients also suffer from a number of vascular anomalies. The loss of NFl from the endothelium is embryonically lethal in mouse developmental models, however little is known regarding the molecular regulation by NF1 in endothelium. We investigated the consequences of losing NF1 expression on the function of endothelial cells using shRNA. The loss of NF1 was sufficient to elevate levels of active Ras under non-stimulated conditions. These elevations in Ras activity were associated with activation of downstream signaling including activation of ERK, AKT and mTOR. Cells knocked down in NF1 expression exhibited no cellular senescence. Rather, they demonstrated augmented proliferation and autonomous entry into the cell cycle. These proliferative changes were accompanied by enhanced expression of cyclin D, phosphorylation of p27(KIP), and decreases in total p27(KIP) levels, even under growth factor free conditions. In addition, NF1-deficient cells failed to undergo normal branching morphogenesis in a co-culture assay, instead forming planar islands with few tubules and branches. We find the changes induced by the loss of NF1 could be mitigated by co-expression of the GAP-related domain of NF1 implicating Ras regulation in these effects. Using doxycycline-inducible shRNA, targeting NF1, we find that the morphogenic changes are reversible. Similarly, in fully differentiated and stable vascular-like structures, the silencing of NF1 results in the appearance of abnormal vascular structures. Finally, the proliferative changes and the abnormal vascular morphogenesis are normalized by low-dose rapamycin treatment. These data provide a detailed analysis of the molecular and functional consequences of NF1 loss in human endothelial cells. These insights may provide new approaches to therapeutically addressing vascular abnormalities in these patients while underscoring a critical role for normal Ras regulation in maintaining the health and function of the vasculature.


Assuntos
Proliferação de Células , Neurofibromina 1/genética , Ciclo Celular/genética , Senescência Celular/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Inativação Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Neovascularização Patológica/genética , Neurofibromina 1/metabolismo , Neurofibromina 1/fisiologia , Proteínas/fisiologia , Transdução de Sinais/genética , Serina-Treonina Quinases TOR , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA