Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
Biol Open ; 1(8): 723-30, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23213465

RESUMO

Focal adhesion kinase (FAK) is critically positioned to integrate signals from the extracellular matrix and cellular adhesion. It is essential for normal vascular development and has been implicated in a wide range of cellular functions including the regulation of cell proliferation, migration, differentiation, and survival. It is currently being actively targeted therapeutically using different approaches. We have used human endothelial cells as a model system to compare the effects of inhibiting FAK through several different approaches including dominant negatives, kinase inhibitors and shRNA. We find that manipulations of FAK signaling that result in inhibition of FAK 397 phosphorylation inhibit proliferation and migration. However, abolition of FAK expression using stable (shRNA) or transient (siRNA) approaches does not interfere with these cellular functions. The ability to regulate cell proliferation by FAK manipulation is correlated with the activation status of Rac, an essential signal for the regulation of cyclin-dependent kinase inhibitors. The knockdown of FAK, while not affecting cellular proliferation or migration, dramatically interferes with vascular morphogenesis and survival, mirroring in vivo findings. We propose a novel model of FAK signaling whereby one of the multifunctional roles of FAK as a signaling protein includes FAK as a phospho-regulated repressor of Rac activation, with important implications on interpretation of research experiments and therapeutic development.

4.
J Exp Clin Cancer Res ; 27: 61, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18983664

RESUMO

BACKGROUND: Endostatin and anastellin, fragments of collagen type XVIII and fibronectin, respectively, belong to a family of endogenous inhibitors of angiogenesis which inhibit tumor growth and metastasis in a number of mouse models of human cancer. The mechanism of action of these inhibitors is not well understood, but they have great potential usefulness as non-toxic long-term therapy for cancer treatment. METHODS: In this study, we compare the anti-angiogenic properties of endostatin and anastellin using cell proliferation and transwell migration assays. RESULTS: Anastellin but not endostatin completely inhibited human dermal microvessel endothelial cell proliferation in response to serum stimulation. Both anastellin and endostatin additively inhibited endothelial cell migration in response to VEGF. Anastellin but not endostatin lowered basal levels of active ERK. CONCLUSION: These data indicate that anastellin and endostatin exert their anti-angiogenic effects by modulating distinct steps in the angiogenic pathway and suggest that matrix-derived inhibitors of angiogenesis may exhibit higher efficacy when used in combination.


Assuntos
Inibidores da Angiogênese/farmacologia , Endostatinas/farmacologia , Fibronectinas/farmacologia , Neovascularização Patológica/metabolismo , Fragmentos de Peptídeos/farmacologia , Proliferação de Células , Células Endoteliais/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases
5.
Cancer Res ; 68(18): 7371-9, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794124

RESUMO

Integrins, the major receptors for cell adhesion to the extracellular matrix, play important roles during tumor progression. However, it is still unclear whether genetic lesions that occur during carcinoma development can lead to altered integrin function, and how changes in integrin function contribute to subsequent carcinoma progression. Loss-of-function mutations in p53 and activating mutations in H-Ras, which immortalize and transform epithelial cells, respectively, are common causal events in squamous cell carcinoma (SCC). Phenotypes resulting from these two genetic lesions promote SCC progression and are, therefore, potential targets for anticancer therapies. We developed a model system of keratinocyte transformation that has allowed us to investigate the individual roles of p53 mutation and oncogenic Ras mutation in the acquisition of integrin alpha3beta1-regulated phenotypes that promote SCC progression. Using this model, we show that keratinocyte immortalization by p53-null mutation causes a switch in alpha3beta1 function that induces matrix metalloproteinase (MMP)-9 gene expression in tumorigenic cells. This acquired alpha3beta1-dependent regulation of MMP-9 was maintained during subsequent transformation by oncogenic Ras, and it promoted invasion of tumorigenic keratinocytes. Our results show that loss of p53 function leads to changes in integrin-mediated gene regulation that occur during SCC progression and play a critical role in tumor cell invasion.


Assuntos
Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/patologia , Integrina alfa3beta1/metabolismo , Queratinócitos/patologia , Metaloproteinase 9 da Matriz/biossíntese , Animais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/genética , Linhagem Celular Transformada , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Expressão Gênica , Humanos , Queratinócitos/enzimologia , Queratinócitos/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
6.
J Invest Dermatol ; 127(1): 31-40, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16917494

RESUMO

Upon epidermal wounding, keratinocytes at the wound edge become activated, deposit newly synthesized laminin-5 into the extracellular matrix, and migrate into the wound bed. The interaction between integrin alpha3beta1 and laminin-5 is essential for establishment of a stable, leading lamellipodium and persistent keratinocyte migration. We previously showed that integrin alpha3beta1 activates the Rho family GTPase Rac1 and regulates Rac1-dependent formation of polarized, leading lamellipodia in migrating keratinocytes. In the present study, we explored the role of focal adhesion kinase (FAK) and src signaling in this process. We show that overexpression of the FAK inhibitor FAK-related non-kinase or of the FAK(Y397F) auto-phosphorylation mutant, induced abnormal, non-polarized spreading of keratinocytes on laminin-5. Integrin alpha3beta1 was required for full FAK auto-phosphorylation at Y397, and subsequent src kinase-dependent phosphorylation of FAK at residues Y861 and Y925, sites responsible for promoting signal transduction downstream of FAK, indicating that alpha3beta1 regulates the coordination of FAK/src signal transduction. Inhibiting either src kinase activity or FAK signaling interfered with alpha3beta1-mediated Rac1 activation and polarized cell spreading. These findings reveal a novel pathway in migratory keratinocytes wherein alpha3beta1-laminin-5 interactions regulate src kinase signaling through FAK, promoting Rac1 activation and polarized lamellipodium extension.


Assuntos
Moléculas de Adesão Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Integrina alfa3beta1/fisiologia , Queratinócitos/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Quinases da Família src/fisiologia , Animais , Adesão Celular , Polaridade Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Camundongos , Fosforilação , Pirimidinas/farmacologia , Transdução de Sinais , Calinina
7.
J Biol Chem ; 280(22): 20995-1003, 2005 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15790570

RESUMO

Pathological conditions such as hypertension and hyperglycemia as well as abrasions following balloon angioplasty all lead to endothelial dysfunction that impacts disease morbidity. These conditions are associated with the elaboration of a variety of cytokines and increases in p38 activity in endothelial cells. However, the relationship between enhanced p38 activity and endothelial cell function remains poorly understood. To investigate the effect of enhanced p38 MAPK activity on endothelial cell function, we expressed an activated mutant of MEK6 (MEK6E), an upstream regulator of p38. Expression of MEK6E activated p38 and resulted in phosphorylation of its downstream substrate, heat shock protein 27 (Hsp27). Activation of p38 was not sufficient to induce apoptosis; however, it did induce p38-dependent cell cycle arrest. MEK6E expression was sufficient to inhibit ERK phosphorylation triggered by growth factors and integrin engagement. MAPK phosphatase-1 (MKP-1) expression was increased upon p38 activation, and expression of a "substrate-trapping" MKP-1 was sufficient to restore ERK activity. Activation of p38 was sufficient to induce cell migration, which was accompanied by alterations in actin architecture characterized by enhanced lamellipodia. Co-expression of a mutant form of Hsp27, lacking all three phosphorylation sites, reversed MEK6E-induced cell migration and altered the cytoskeletal changes induced by p38 activation. Collectively, these results suggest that cellular decisions regarding migration and proliferation are influenced by p38 activity and that prolonged activation of p38 may result in an anti-angiogenic phenotype that contributes to endothelial dysfunction.


Assuntos
Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Actinas/química , Actinas/metabolismo , Apoptose , Sítios de Ligação , Western Blotting , Proteínas de Ciclo Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Citoesqueleto/metabolismo , Fosfatase 1 de Especificidade Dupla , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Imediatamente Precoces , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , MAP Quinase Quinase 6/metabolismo , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Chaperonas Moleculares , Mutação , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica , Fosfoproteínas Fosfatases , Fosforilação , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases , Fatores de Tempo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
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
9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...