Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Genesis ; 57(10): e23330, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31386299

RESUMO

Yes-associated protein (YAP), a key effector of the Hippo signaling pathway, is expressed in the nucleus of spermatogonia in mice, suggesting a potential role in spermatogenesis. Here, we report the generation of a conditional knockout mouse model (Yapflox/flox ; Ddx4cre/+ ) that specifically inactivates Yap in the germ cells. The inactivation of Yap in spermatogonia was found to be highly efficient in this model. The loss of Yap in the germ cells had no observable effect on spermatogenesis in vivo. Histological examination of the testes showed no structural differences between mutant animals and age-matched Yapflox/flox controls, nor was any differences detected in gonadosomatic index, expression of germ cell markers or sperm counts. Cluster-forming assay using undifferentiated spermatogonia, including spermatogonial stem cells (SSCs), also showed that YAP is dispensable for SSC cluster formation in vitro. However, an increase in the expression of spermatogenesis and oogenesis basic helix-loop-helix 1 (Sohlh1) and neurogenin 3 (Ngn3) was observed in clusters derived from Yapflox/flox ; Ddx4cre/+ animals. Taken together, these results suggest that YAP fine-tunes the expression of genes associated with spermatogonial fate commitment, but that its loss is not sufficient to alter spermatogenesis in vivo.


Assuntos
Proteínas Proto-Oncogênicas c-yes/fisiologia , Espermatogênese/fisiologia , Animais , Células Cultivadas , RNA Helicases DEAD-box/genética , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-yes/genética , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/fisiologia
3.
Life Sci ; 219: 129-135, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30639391

RESUMO

AIMS: Aberrantly expressed miRNAs are demonstrated to be involved in the development of congenital heart disease (CHD). miR-9 was proposed to be upregulated in cardiac tissues from CHD cases. However, the role of miR-9 in hypoxia-induced cardiomyocytes and the potential mechanism are far from being addressed. MAIN METHODS: qRT-PCR and western blot analysis were performed to detect miR-9 and Yes-associated protein 1 (Yap1) expressions in hypoxic H9c2 cells. CCK-8, flow cytometry analysis, caspase-3/7 activity assay were applied to evaluate cell proliferation, apoptosis, and caspase-3/7 activity, respectively. The interaction between miR-9 and Yap1 was explored by luciferase reporter assay, qRT-PCR and western blot. KEY FINDINGS: miR-9 was upregulated and Yap1 was downregulated in H9c2 cells in response to hypoxia in a time-dependent manner. Knockdown of miR-9 promoted cell proliferation, and inhibited apoptosis and caspase-3/7 activity in hypoxic H9c2 cells, while miR-9 overexpression exerted the opposite effects on hypoxic H9c2 cells. In addition, Yap1 was a direct target of miR-9 in H9c2 cells. Yap1 knockdown suppressed cell proliferation and promoted apoptosis in hypoxia-exposed H9c2 cells. Yap1 knockdown attenuated the effect of anti-miR-9 on cell proliferation and apoptosis in hypoxia-exposed H9c2 cells. SIGNIFICANCE: miR-9 knockdown inhibited hypoxia-induced cardiomyocyte apoptosis by targeting Yap1. Our study provided a novel insight into the mechanism of the adaptation of cardiomyocytes to chronic hypoxia.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose , Hipóxia/fisiopatologia , MicroRNAs/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Animais , Western Blotting , Caspase 3/metabolismo , Caspase 7/metabolismo , Proliferação de Células , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Hipóxia/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real
4.
Plast Reconstr Surg ; 139(4): 888e-899e, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28350663

RESUMO

BACKGROUND: Noninvasive external volume expansion device has been applied to stimulate nonsurgical breast enlargement in clinical settings. Although previous results demonstrate the capacity of external volume expansion to increase the number of adipocytes, this strategy alone is insufficient to reconstruct soft-tissue defects or increase breast mass. The authors combined a minimally invasive tissue dissection method with external volume expansion to generate large volumes of adipose tissue. METHOD: In vitro, various densities of adipose-derived stem cells were prepared to evaluate relations between cell contacts and cell proliferation. In vivo, dorsal adipose tissue of rabbits was thoroughly dissected and the external volume expansion device was applied to maintain the released state. External volume expansion without tissue dissection served as the control. RESULTS: In the dissection group, the generated adipose tissue volume was much larger than that in the control group at all time points. A larger number of proliferating cells appeared in the dissection samples than in the control samples at the early stage after tissue dissection. At low cell density, adipose-derived stem cells displayed an increasing proliferation rate compared to high cell density. Protein expression analysis revealed that cell proliferation was mediated by a similar mechanism both in vivo and in vitro, involving the release of cell contact inhibition and Hippo/Yes-associated protein pathway activation. CONCLUSIONS: Adipose tissue dissection releases cell-to-cell contacts and induces adipose-derived stem cell proliferation. Preexpanded adipose-derived stem cells undergo adipogenesis under the adipogenic environment created by external volume expansion, leading to better adipose regeneration compared with the control.


Assuntos
Tecido Adiposo/cirurgia , Expansão de Tecido , Adipócitos , Adipogenia , Tecido Adiposo/citologia , Animais , Proliferação de Células , Células Cultivadas , Dissecação , Feminino , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Coelhos , Transdução de Sinais
5.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L880-8, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26919896

RESUMO

Endothelial cells are essential for normal lung function: they sense and respond to circulating factors and hemodynamic alterations. In inflammatory lung diseases such as acute respiratory distress syndrome, endothelial cell apoptosis is an inciting event in pathogenesis and a prominent pathological feature. Endothelial cell apoptosis is mediated by circulating inflammatory factors, which bind to receptors on the cell surface, activating signal transduction pathways, leading to caspase-3-mediated apoptosis. We hypothesized that yes and src have differential effects on caspase-3 activation in human pulmonary microvascular endothelial cells (hPMVEC) due to differential downstream signaling effects. To test this hypothesis, hPMVEC were treated with siRNA against src (siRNAsrc), siRNA against yes (siRNAyes), or their respective scramble controls. After recovery, the hPMVEC were treated with cytomix (LPS, IL-1ß, TNF-α, and IFN-γ). Treatment with cytomix induced activation of the extracellular signal-regulated kinase (ERK) pathway and caspase-3-mediated apoptosis. Treatment with siRNAsrc blunted cytomix-induced ERK activation and enhanced cleaved caspase-3 levels, while treatment with siRNAyes enhanced cytomix-induced ERK activation and attenuated levels of cleaved caspase-3. Inhibition of the ERK pathway using U0126 enhanced cytomix-induced caspase-3 activity. Treatment of hPMVEC with cytomix induced Akt activation, which was inhibited by siRNAsrc. Inhibition of the phosphatidylinositol 3-kinase/Akt pathway using LY294002 prevented cytomix-induced ERK activation and augmented cytomix-induced caspase-3 cleavage. Together, our data demonstrate that, in hPMVEC, yes activation blunts the ERK cascade in response to cytomix, resulting in greater apoptosis, while cytomix-induced src activation induces the phosphatidylinositol 3-kinase pathway, which leads to activation of Akt and ERK and attenuation of apoptosis.


Assuntos
Apoptose , Células Endoteliais/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Quinases da Família src/fisiologia , Caspase 3/metabolismo , Sobrevivência Celular , Células Cultivadas , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Humanos , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases , Microvasos/enzimologia , Microvasos/imunologia , Síndrome do Desconforto Respiratório/enzimologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia
6.
Am J Physiol Endocrinol Metab ; 307(7): E553-62, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25117412

RESUMO

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. However, it undergoes cyclic restructuring during the epithelial cycle of spermatogenesis in which the "old" BTB located above the preleptotene spermatocytes being transported across the immunological barrier is "disassembled," whereas the "new" BTB found behind these germ cells is rapidly "reassembled," i.e., mediated by endocytic vesicle-mediated protein trafficking events. Thus, the immunological barrier is maintained when preleptotene spermatocytes connected in clones via intercellular bridges are transported across the BTB. Yet the underlying mechanism(s) in particular the involving regulatory molecules that coordinate these events remains unknown. We hypothesized that c-Src and c-Yes might work in contrasting roles in endocytic vesicle-mediated trafficking, serving as molecular switches, to effectively disassemble and reassemble the old and the new BTB, respectively, to facilitate preleptotene spermatocyte transport across the BTB. Following siRNA-mediated specific knockdown of c-Src or c-Yes in Sertoli cells, we utilized biochemical assays to assess the changes in protein endocytosis, recycling, degradation and phagocytosis. c-Yes was found to promote endocytosed integral membrane BTB proteins to the pathway of transcytosis and recycling so that internalized proteins could be effectively used to assemble new BTB from the disassembling old BTB, whereas c-Src promotes endocytosed Sertoli cell BTB proteins to endosome-mediated protein degradation for the degeneration of the old BTB. By using fluorescence beads mimicking apoptotic germ cells, Sertoli cells were found to engulf beads via c-Src-mediated phagocytosis. A hypothetical model that serves as the framework for future investigation is thus proposed.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas Proto-Oncogênicas c-yes/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Células de Sertoli/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Endocitose/fisiologia , Técnicas de Silenciamento de Genes , Genes src/genética , Masculino , Proteínas de Membrana/metabolismo , Fagocitose/fisiologia , Proteínas Proto-Oncogênicas c-yes/genética , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
7.
Am J Physiol Endocrinol Metab ; 304(2): E145-59, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23169788

RESUMO

During spermatogenesis, extensive restructuring takes place at the cell-cell interface since developing germ cells migrate progressively from the basal to the adluminal compartment of the seminiferous epithelium. Since germ cells per se are not motile cells, their movement relies almost exclusively on the Sertoli cell. Nonetheless, extensive exchanges in signaling take place between these cells in the seminiferous epithelium. c-Yes, a nonreceptor protein tyrosine kinase belonging to the Src family kinases (SFKs) and a crucial signaling protein, was recently shown to be upregulated at the Sertoli cell-cell interface at the blood-testis barrier (BTB) at stages VIII-IX of the seminiferous epithelial cycle of spermatogenesis. It was also highly expressed at the Sertoli cell-spermatid interface known as apical ectoplasmic specialization (apical ES) at stage V to early stage VIII of the epithelial cycle during spermiogenesis. Herein, it was shown that the knockdown of c-Yes by RNAi in vitro and in vivo affected both Sertoli cell adhesion at the BTB and spermatid adhesion at the apical ES, causing a disruption of the Sertoli cell tight junction-permeability barrier function, germ cell loss from the seminiferous epithelium, and also a loss of spermatid polarity. These effects were shown to be mediated by changes in distribution and/or localization of adhesion proteins at the BTB (e.g., occludin, N-cadherin) and at the apical ES (e.g., nectin-3) and possibly the result of changes in the underlying actin filaments at the BTB and the apical ES. These findings implicate that c-Yes is a likely target of male contraceptive research.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas Proto-Oncogênicas c-yes/fisiologia , Epitélio Seminífero/citologia , Epitélio Seminífero/fisiologia , Animais , Barreira Hematotesticular/fisiologia , Caderinas/metabolismo , Adesão Celular/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Polaridade Celular/genética , Células Cultivadas , Masculino , Ocludina/metabolismo , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas c-yes/metabolismo , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Células de Sertoli/fisiologia , Transdução de Sinais/genética , Espermatogênese/genética , Distribuição Tecidual/genética
8.
Arterioscler Thromb Vasc Biol ; 32(10): 2444-51, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22904271

RESUMO

OBJECTIVE: Endothelium dysfunction is an initiating factor in atherosclerosis. A disintegrin and metalloproteinase 15 (ADAM 15) is a multidomain metalloprotease recently identified as a regulator of endothelial permeability. However, whether and how ADAM15 contributes to atherosclerosis remains unknown. METHODS AND RESULTS: Genetic ablation of ADAM15 in apolipoprotein E-deficient mice led to a significant reduction in aortic atherosclerotic lesion size (by 52%), plaque macrophage infiltration (by 69%), and smooth muscle cell deposition (by 82%). In vitro studies implicated endothelial-derived ADAM15 in barrier dysfunction and monocyte transmigration across mouse aortic and human umbilical vein endothelial cell monolayers. This role of ADAM15 depended on intact functioning of the cytoplasmic domain, as evidenced in experiments with site-directed mutagenesis targeting the metalloprotease active site (E349A), the disintegrin domain (Arginine-Glycine-Aspartic acid→Threonine-Aspartic acid-Aspartic acid), or the cytoplasmic tail. Further investigations revealed that ADAM15-induced barrier dysfunction was concomitant with dissociation of endothelial adherens junctions (vascular endothelial [VE]-cadherin/γ-catenin), an effect that was sensitive to Src family kinase inhibition. Through small interfering RNA-mediated knockdown of distinct Src family kinase members, c-Src and c-Yes were identified as important mediators of these junctional effects of ADAM15. CONCLUSIONS: These results suggest that endothelial cell-derived ADAM15, signaling through c-Src and c-Yes, contributes to atherosclerotic lesion development by disrupting adherens junction integrity and promoting monocyte transmigration.


Assuntos
Proteínas ADAM/fisiologia , Aterosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Proteínas de Membrana/fisiologia , Transdução de Sinais/fisiologia , Quinases da Família src/fisiologia , Proteínas ADAM/efeitos dos fármacos , Proteínas ADAM/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Proteína Tirosina Quinase CSK , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/patologia , Humanos , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Monócitos/fisiologia , Proteínas Proto-Oncogênicas c-yes/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas c-yes/fisiologia , RNA Interferente Pequeno/farmacologia , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/genética
9.
Hepatology ; 56(3): 1097-107, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22886419

RESUMO

UNLABELLED: Human chronic cholestatic liver diseases are characterized by cholangiocyte proliferation, hepatocyte injury, and fibrosis. Yes-associated protein (YAP), the effector of the Hippo tumor-suppressor pathway, has been shown to play a critical role in promoting cholangiocyte and hepatocyte proliferation and survival during embryonic liver development and hepatocellular carcinogenesis. Therefore, the aim of this study was to examine whether YAP participates in the regenerative response after cholestatic injury. First, we examined human liver tissue from patients with chronic cholestasis. We found more-active nuclear YAP in the bile ductular reactions of primary sclerosing cholangitis and primary biliary cirrhosis patient liver samples. Next, we used the murine bile duct ligation (BDL) model to induce cholestatic liver injury. We found significant changes in YAP activity after BDL in wild-type mice. The function of YAP in the hepatic response after BDL was further evaluated with liver-specific Yap conditional deletion in mice. Ablating Yap in the mouse liver not only compromised bile duct proliferation, but also enhanced hepatocyte necrosis and suppressed hepatocyte proliferation after BDL. Furthermore, primary hepatocytes and cholangiocytes isolated from Yap-deficient livers showed reduced proliferation in response to epidermal growth factor in vitro. Finally, we demonstrated that YAP likely mediates its biological effects through the modulation of Survivin expression. CONCLUSION: Our data suggest that YAP promotes cholangiocyte and hepatocyte proliferation and prevents parenchymal damage after cholestatic injury in mice and thus may mediate the response to cholestasis-induced human liver disease.


Assuntos
Ductos Biliares/citologia , Colestase/complicações , Hepatócitos/fisiologia , Regeneração Hepática , Proteínas Proto-Oncogênicas c-yes/fisiologia , Animais , Humanos , Ligadura , Masculino , Camundongos
11.
PLoS One ; 6(2): e17237, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21390316

RESUMO

c-Yes, a member of the Src tyrosine kinase family, is found highly activated in colon carcinoma but its importance relative to c-Src has remained unclear. Here we show that, in HT29 colon carcinoma cells, silencing of c-Yes, but not of c-Src, selectively leads to an increase of cell clustering associated with a localisation of ß-catenin at cell membranes and a reduction of expression of ß-catenin target genes. c-Yes silencing induced an increase in apoptosis, inhibition of growth in soft-agar and in mouse xenografts, inhibition of cell migration and loss of the capacity to generate liver metastases in mice. Re-introduction of c-Yes, but not c -Src, restores transforming properties of c-Yes depleted cells. Moreover, we found that c-Yes kinase activity is required for its role in ß-catenin localisation and growth in soft agar, whereas kinase activity is dispensable for its role in cell migration. We conclude that c-Yes regulates specific oncogenic signalling pathways important for colon cancer progression that is not shared with c-Src.


Assuntos
Carcinoma/patologia , Transformação Celular Neoplásica/genética , Neoplasias do Colo/patologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Animais , Carcinoma/genética , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Células HCT116 , Células HT29 , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Especificidade de Órgãos/genética , Proteínas Proto-Oncogênicas c-yes/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transplante Heterólogo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética , Quinases da Família src/fisiologia
12.
J Biol Chem ; 284(44): 30433-40, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19748888

RESUMO

Therapeutic strategies such as using channel blockers and reducing culture temperature have been used to rescue some long QT-associated voltage-gated potassium Kv trafficking defective mutant channels. A hyperpolarization-activated cyclic nucleotide-gated HCN4 pacemaker channel mutant (D553N) has been recently found in a patient associated with cardiac arrhythmias including long QT. D553N showed the defective trafficking to the cell surface, leading to little ionic current expression (loss-of-function). We show in this report that enhanced tyrosine phosphorylation mediated by Src, Fyn, and Yes kinases was able to restore the surface expression of D553N for normal current expression. Src or Yes, but not Fyn, significantly increased the current density and surface expression of D553N. Fyn accelerated the activation kinetics of the rescued D553N. Co-expression of D553N with Yes exhibited the slowest activation kinetics of D553N. Src, Fyn, and Yes significantly enhanced the tyrosine phosphorylation of D553N. A combination of Src, Fyn, and Yes rescued the current expression and the gating of D553N comparable with those of wild-type HCN4. In conclusion, we demonstrate a novel mechanism using three endogenous Src kinases to rescue a trafficking defective HCN4 mutant channel (D553N) by enhancing the tyrosine phosphorylation of the mutant channel protein.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Proteínas Musculares/genética , Proteínas Tirosina Quinases/fisiologia , Linhagem Celular , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Proteínas Musculares/metabolismo , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Fosforilação , Canais de Potássio , Transporte Proteico , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Quinases da Família src/fisiologia
13.
PLoS Genet ; 3(12): e225, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18159945

RESUMO

Shp2 is a cytoplasmic protein-tyrosine phosphatase that is essential for normal development. Activating and inactivating mutations have been identified in humans to cause the related Noonan and LEOPARD syndromes, respectively. The cell biological cause of these syndromes remains to be determined. We have used the zebrafish to assess the role of Shp2 in early development. Here, we report that morpholino-mediated knockdown of Shp2 in zebrafish resulted in defects during gastrulation. Cell tracing experiments demonstrated that Shp2 knockdown induced defects in convergence and extension cell movements. In situ hybridization using a panel of markers indicated that cell fate was not affected by Shp2 knock down. The Shp2 knockdown-induced defects were rescued by active Fyn and Yes and by active RhoA. We generated mutants of Shp2 with mutations that were identified in human patients with Noonan or LEOPARD Syndrome and established that Noonan Shp2 was activated and LEOPARD Shp2 lacked catalytic protein-tyrosine phosphatase activity. Expression of Noonan or LEOPARD mutant Shp2 in zebrafish embryos induced convergence and extension cell movement defects without affecting cell fate. Moreover, these embryos displayed craniofacial and cardiac defects, reminiscent of human symptoms. Noonan and LEOPARD mutant Shp2s were not additive nor synergistic, consistent with the mutant Shp2s having activating and inactivating roles in the same signaling pathway. Our results demonstrate that Shp2 is required for normal convergence and extension cell movements during gastrulation and that Src family kinases and RhoA were downstream of Shp2. Expression of Noonan or LEOPARD Shp2 phenocopied the craniofacial and cardiac defects of human patients. The finding that defective Shp2 signaling induced cell movement defects as early as gastrulation may have implications for the monitoring and diagnosis of Noonan and LEOPARD syndrome.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Diferenciação Celular , Movimento Celular , Modelos Animais de Doenças , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Marcação de Genes , Humanos , Síndrome LEOPARD/enzimologia , Síndrome LEOPARD/genética , Mutação , Síndrome de Noonan/enzimologia , Síndrome de Noonan/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Transdução de Sinais , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
14.
Neoplasia ; 9(9): 745-54, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17898870

RESUMO

Increases in the levels and/or activity of nonreceptor tyrosine kinases c-Src and c-Yes are often associated with colorectal carcinogenesis. The physiological consequences of increased c-Yes activity during the early and late stages of tumorigenesis, in addition to the degree of redundancy between c-Yes and c-Src in colorectal cancer cells, remain elusive. To study the consequences of increases in c-Yes levels and activity in later stages of colorectal carcinogenesis, we developed human colorectal cancer cell lines in which c-Yes levels and activity can be inducibly increased by a tightly controlled expression of wild-type c-Yes or by constitutively active mutants of c-Yes, c-YesY537F, and c-Yes Delta t6aa. c-Yes induction resulted in increased cell motility but did not promote proliferation either in vitro or in vivo. These results suggest that in later stages of colorectal carcinogenesis, elevations in c-Yes levels/activity may promote cancer spread and metastasis rather than tumor growth.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Adesão Celular/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Doxiciclina/farmacologia , Indução Enzimática/efeitos dos fármacos , Feminino , Humanos , Camundongos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Recombinantes de Fusão/fisiologia , Quinases da Família src/fisiologia
15.
Oncogene ; 26(49): 7028-37, 2007 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17486066

RESUMO

The receptor-type protein tyrosine phosphatase epsilon (RPTPepsilon) activates c-Src in mammary tumor cells induced in vivo by Neu. Tumor cells lacking RPTPepsilon exhibit reduced c-Src activity, appear less transformed morphologically and proliferate slower in vitro and in vivo. Expression of Src rescues most of these phenotypes, indicating that c-Src activity is important for maintaining the transformed phenotype. However, the molecular mechanisms that control activation of c-Src by RPTPepsilon are unknown. We show that Neu induces phosphorylation of RPTPepsilon exclusively at its C-terminal Y695, and that this phosphorylation is required for activation of c-Src by RPTPepsilon. Phosphorylation of RPTPepsilon does not affect its activity toward another substrate, the voltage-gated potassium channel Kv2.1, suggesting that phosphorylation directs RPTPepsilon activity toward c-Src. Phosphorylation of RPTPepsilon reduces its dimerization at the cell membrane, although this does not affect its activity significantly. RPTPepsilon is subject to strong auto- and trans-dephosphorylation, suggesting that dephosphorylation limits the activation of c-Src downstream of Neu. We conclude that an Neu-RPTPepsilon-Src signaling pathway exists in mammary tumor cells, in which phosphorylation of RPTPepsilon by Neu directs RPTPepsilon to activate c-Src. Reversible phosphorylation of RPTPepsilon at Y695 may thus function as a 'molecular switch', which affects the substrate specificity of the phosphatase.


Assuntos
Neoplasias Mamárias Experimentais/etiologia , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Receptor ErbB-2/genética , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/metabolismo , Animais , Feminino , Imunofluorescência , Imunoprecipitação , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fragmentos de Peptídeos/imunologia , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas c-yes/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/genética
16.
Growth Factors ; 23(4): 263-72, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16338789

RESUMO

Transmembrane receptors link the extracellular environment to the internal control elements of the cell. This signaling influences cell division, differentiation, survival, motility, adhesion, spreading and vesicular transport. Central to this signaling is the Src family of nonreceptor tyrosine kinases. The most studied kinase of this nine member family, c-Src, shares a similar structure, as well as a similar expression pattern to that of another Src family protein, c-Yes. Despite high conservation in sequence, molecular studies demonstrate that the functional domains of these kinases can contribute to specificity in signaling. At the cellular level, analysis of tight junction formation also serves as a model to differentiate c-Yes and c-Src signaling. Results suggest that c-Yes promotes formation of the tight junction by phosphorylating occludin, while c-Src signaling downregulates occludin formation in a Raf-1 dependent manner. In addition, pp62c-Yes knockout mice exhibit a specific physiological function phenotype that is distinct from c-src-/- mice. In these studies, c-yes-/- mice exhibit decreased transcytosis of pIgA from the blood to the bile, while c-src-/- mice exhibit deficits in osteoclasts function and bone resorption. Of particular interest in this review are receptor signals that specifically influence the actions of c-Yes. Growth factors that influence many Src family proteins include the PDGF-R, CSF-1 receptor and others. Since these receptors interact with various Src-family kinases, it is predicted that specific signaling is generated by differential recruitment to the cell membrane and/or differentiated interactions with substrates and binding partners. This review provides an overview of c-Yes interactions with specific receptor signaling pathways and how this interaction potentially influences the known physiological roles of c-Yes.


Assuntos
Proteínas Proto-Oncogênicas c-yes/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais , Animais , Humanos , Proteínas Proto-Oncogênicas c-yes/química , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Citocinas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia
17.
Angiogenesis ; 8(4): 315-26, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16400523

RESUMO

Widely coexpressed Src family kinase (SFK) members Src, Fyn and Yes are involved in various cellular events, often acting downstream of receptor tyrosine kinases, such as vascular endothelial growth factor (VEGF) receptors. They are well known for their functional redundancy; any unique features remain largely undefined. Utilizing RNA interference, we have selectively knocked down Src, Fyn and Yes in human retinal microvascular endothelial cells (HRMECs). Cells with single SFK knockdown showed that all three kinases were required for VEGF mitogenic signaling. VEGF-induced cell migration was significantly increased in Fyn-deficient cells and decreased in Yes-deficient cells. Selective interference of Fyn, but not Src or Yes, impaired VEGF-induced tube formation in HRMECs. Cells in which all three SFKs were targeted showed significant inhibition of all three cellular events. In addition, interference of Src, Fyn and Yes did not affect the anti-apoptotic effect of VEGF in HRMECs, as determined by DNA fragmentation analysis. These results provide direct evidence that Src, Fyn and Yes maintain distinct properties in the regulation of VEGF-mediated endothelial cell events.


Assuntos
Células Endoteliais/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Microvasos/citologia , Microvasos/fisiologia , Mitógenos/fisiologia , Mitose/fisiologia , Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-yes/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Proteínas Proto-Oncogênicas pp60(c-src)/genética , RNA Interferente Pequeno/fisiologia , Vasos Retinianos/citologia , Vasos Retinianos/fisiologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...