Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
PLoS One ; 8(10): e75356, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098377

RESUMO

Due to narrow therapeutic window of cancer therapeutic agents and the development of resistance against these agents, there is a need to discover novel agents to treat breast cancer. The antitumor activities of thymoquinone (TQ), a compound isolated from Nigella sativa oil, were investigated in breast carcinoma in vitro and in vivo. Cell responses after TQ treatment were assessed by using different assays including MTT assay, annexin V-propidium iodide staining, Mitosox staining and Western blot. The antitumor effect was studied by breast tumor xenograft mouse model, and the tumor tissues were examined by histology and immunohistochemistry. The level of anti-oxidant enzymes/molecules in mouse liver tissues was measured by commercial kits. Here, we show that TQ induced p38 phosphorylation and ROS production in breast cancer cells. These inductions were found to be responsible for TQ's anti-proliferative and pro-apoptotic effects. Moreover, TQ-induced ROS production regulated p38 phosphorylation but not vice versa. TQ treatment was found to suppress the tumor growth and this effect was further enhanced by combination with doxorubicin. TQ also inhibited the protein expression of anti-apoptotic genes, such as XIAP, survivin, Bcl-xL and Bcl-2, in breast cancer cells and breast tumor xenograft. Reduced Ki67 and increased TUNEL staining were observed in TQ-treated tumors. TQ was also found to increase the level of catalase, superoxide dismutase and glutathione in mouse liver tissues. Overall, our results demonstrated that the anti-proliferative and pro-apoptotic effects of TQ in breast cancer are mediated through p38 phosphorylation via ROS generation.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Neoplasias da Mama/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Inativação Gênica , Humanos , Imidazóis/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
2.
Nature ; 496(7446): 518-22, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23467095

RESUMO

There has been a marked increase in the incidence of autoimmune diseases in the past half-century. Although the underlying genetic basis of this class of diseases has recently been elucidated, implicating predominantly immune-response genes, changes in environmental factors must ultimately be driving this increase. The newly identified population of interleukin (IL)-17-producing CD4(+) helper T cells (TH17 cells) has a pivotal role in autoimmune diseases. Pathogenic IL-23-dependent TH17 cells have been shown to be critical for the development of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, and genetic risk factors associated with multiple sclerosis are related to the IL-23-TH17 pathway. However, little is known about the environmental factors that directly influence TH17 cells. Here we show that increased salt (sodium chloride, NaCl) concentrations found locally under physiological conditions in vivo markedly boost the induction of murine and human TH17 cells. High-salt conditions activate the p38/MAPK pathway involving nuclear factor of activated T cells 5 (NFAT5; also called TONEBP) and serum/glucocorticoid-regulated kinase 1 (SGK1) during cytokine-induced TH17 polarization. Gene silencing or chemical inhibition of p38/MAPK, NFAT5 or SGK1 abrogates the high-salt-induced TH17 cell development. The TH17 cells generated under high-salt conditions display a highly pathogenic and stable phenotype characterized by the upregulation of the pro-inflammatory cytokines GM-CSF, TNF-α and IL-2. Moreover, mice fed with a high-salt diet develop a more severe form of EAE, in line with augmented central nervous system infiltrating and peripherally induced antigen-specific TH17 cells. Thus, increased dietary salt intake might represent an environmental risk factor for the development of autoimmune diseases through the induction of pathogenic TH17 cells.


Assuntos
Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Cloreto de Sódio na Dieta/farmacologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Animais , Células Cultivadas , Encefalomielite Autoimune Experimental/patologia , Inativação Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Proteínas Imediatamente Precoces/metabolismo , Interleucina-2/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Células Th17/patologia , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Radiat Res ; 178(6): 556-67, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23088768

RESUMO

Vascular endothelium is a key compartment involved in the development of normal tissue toxicity associated with cancer radiation therapy, i.e., acute inflammation and late fibrosis. Radiation-induced endothelial cell activation has been extensively studied, and activated endothelial cells are characterized by increased expression of inflammatory mediators and adhesion molecules, and activation of the coagulation and thrombosis pathways. However, little is known about the role of vascular endothelium interaction with resident immune cells, such as mast cells on its response to irradiation. Here, we report that endothelial exposure to mast cell conditioned medium and irradiation induces a synergistic expression of many inflammatory genes including interleukin-6 and interleukin-8, CXCL2 and E-selectin. This synergy is blocked by the histamine H1 receptor antagonist mepyramine and partially mimicked by exogenous histamine addition before irradiation. Using pharmacological and molecular inhibition approaches, we show the p38α MAP kinase and p65 (NF-κB) dependence of the synergy. Moreover, our data show a link between both pathways, with p65 (NF-κB) being downstream of p38. These data highlight the possible exacerbation of the radiation-induced endothelial inflammatory response by its interactions with immune cells. It also suggest that p38α MAP kinase and p65 (NF-κB) inhibition in vascular endothelium may limit excessive tissue inflammation induced by radiation therapy, and thereby limit the associated acute and late tissue damage.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos da radiação , Inflamação/genética , Mastócitos/citologia , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inativação Gênica , Histamina/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Inflamação/imunologia , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
4.
Oncogene ; 30(30): 3317-27, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21383696

RESUMO

The largest energy consumer in the cell is the ribosome biogenesis whose aberrancy elicits various diseases in humans. It has been recently revealed that p53 induction, along with cell cycle arrest, is related with abnormal ribosome biogenesis, but the exact mechanism still remains unknown. In this study, we have found that aberrant ribosome biogenesis activates two parallel cellular pathways, c-Myc and ASK1/p38, which result in p53 induction and G1 arrest. The c-Myc stabilizes p53 by rpL11-mediated HDM2 inhibition, and ASK1/p38 activates p53 by phosphorylation on serine 15 and 33. Our studies demonstrate the relationship between these two pathways and p53 induction. The changes caused by impaired ribosomal stress, such as p53 induction and G1 arrest, were completely disappeared by inhibition of either pathway. These findings suggest a monitoring mechanism of c-Myc and ASK1/p38 against abnormal ribosome biogenesis through controlling the stability and activity of p53 protein.


Assuntos
Fase G1 , MAP Quinase Quinase Quinase 5/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ribossomos/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Fase G1/genética , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Fosforilação/genética , Processamento de Proteína Pós-Traducional/genética , Proteínas Ribossômicas/deficiência , Proteínas Ribossômicas/genética , Ribossomos/genética , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Br J Pharmacol ; 162(5): 1143-55, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21077851

RESUMO

BACKGROUND AND PURPOSE: Dietary intake of ω-3 polyunsaturated fatty acids (ω-3 PUFAs) like eicosapentaenoic acid (EPA) decreases cancer risk, while arachidonic acid and other ω-6 PUFAs increase risk, but the underlying mechanisms are unclear. Cytochrome P450 (CYP)-derived epoxides contribute to enhanced tumourigenesis due to ω-6 PUFA intake. Thus, ω-6 arachidonic acid epoxides (EETs) inhibit apoptosis and stimulate proliferation by up-regulating cyclin D1 expression in cells. The present study evaluated the corresponding ω-3 PUFA epoxides and assessed their role in the regulation of cell proliferation. EXPERIMENTAL APPROACH: Four chemically stable EPA epoxides (formed at the 8,9-, 11,12-, 14,15- and 17,18-olefinic bonds) were synthesized and tested against growth-related signalling pathways in brain microvascular endothelial bEND.3 cells. Cell cycle distribution was determined by flow cytometry and cyclin gene expression by immunoblotting and real-time PCR. The role of the p38 mitogen-activated protein (MAP) kinase in cyclin D1 dysregulation was assessed using specific inhibitors and dominant-negative expression plasmids. KEY RESULTS: The ω-3 17,18-epoxide of EPA decreased cell proliferation, interrupted the cell cycle in S-phase and down-regulated the cyclin D1/cyclin-dependent kinase (CDK)-4 complex, whereas the 8,9-, 11,12- and 14,15-epoxides were either inactive or enhanced proliferation. Cyclin D1 down-regulation by 17,18-epoxy-EPA was mediated by activation of the growth-suppressing p38 MAP kinase, but the alternate EPA-epoxides were inactive. CONCLUSIONS AND IMPLICATIONS: The present findings suggest that the epoxide formed by CYP enzymes at the ω-3 olefinic bond may contribute to the beneficial effects of ω-3 PUFA by down-regulating cyclin D1 and suppressing cell proliferation.


Assuntos
Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Ácido Eicosapentaenoico/análogos & derivados , Células Endoteliais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Regulação para Baixo/efeitos dos fármacos , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/farmacologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , Compostos de Epóxi/química , Compostos de Epóxi/farmacologia , Camundongos , Neoplasias/etiologia , Neoplasias/prevenção & controle , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
6.
FEBS Lett ; 584(23): 4711-6, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21050851

RESUMO

p53 plays a fundamental role in the maintenance of genome integrity after DNA damage, deciding whether cells repair and live, or die. However, the rules that govern its choice are largely undiscovered. Here we show that the functional relationship between p38 and p53 is crucial in defining the cell fate after DNA damage. Upon low dose ultraviolet (UV) radiation, p38 and p53 protect the cells from apoptosis separately. Conversely, they function together to favor apoptosis upon high dose UV exposure. Taken together, a UV-induced, dose-dependent interaction between p38 and p53 acts as a switch to determine cell fate.


Assuntos
Apoptose/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Técnicas de Inativação de Genes , Humanos , Camundongos , Células NIH 3T3 , Fosforilação/efeitos da radiação , Ligação Proteica/efeitos da radiação , Serina , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
7.
Circ Res ; 104(4): 455-65, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19122179

RESUMO

The molecular events linking lipid accumulation in atherosclerotic plaques to complications such as aneurysm formation and plaque disruption are poorly understood. BALB/c-Apoe(-/-) mice bearing a null mutation in the Npc1 gene display prominent medial erosion and atherothrombosis, whereas their macrophages accumulate free cholesterol in late endosomes and show increased cathepsin K (Ctsk) expression. We now show increased cathepsin K immunostaining and increased cysteinyl proteinase activity using near infrared fluorescence imaging over proximal aortas of Apoe(-/-), Npc1(-/-) mice. In mechanistic studies, cholesterol loading of macrophage plasma membranes (cyclodextrin-cholesterol) or endosomal system (AcLDL+U18666A or Npc1 null mutation) activated Toll-like receptor (TLR) signaling, leading to sustained phosphorylation of p38 mitogen-activated protein kinase and induction of p38 targets, including Ctsk, S100a8, Mmp8, and Mmp14. Studies in macrophages from knockout mice showed major roles for TLR4, following plasma membrane cholesterol loading, and for TLR3, after late endosomal loading. TLR signaling via p38 led to phosphorylation and activation of the transcription factor Microphthalmia transcription factor, acting at E-box elements in the Ctsk promoter. These studies suggest that free cholesterol enrichment of either plasma or endosomal membranes in macrophages leads to activation of signaling via various TLRs, prolonged p38 mitogen-activated protein kinase activation, and induction of Mmps, Ctsk, and S100a8, potentially contributing to plaque complications.


Assuntos
Catepsinas/biossíntese , Membrana Celular/metabolismo , Colesterol/metabolismo , Endossomos/metabolismo , Macrófagos/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Aorta/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Calgranulina A , Catepsina K , Membrana Celular/enzimologia , Membrana Celular/imunologia , Células Cultivadas , Elementos E-Box , Endossomos/enzimologia , Endossomos/imunologia , Indução Enzimática , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/enzimologia , Macrófagos/imunologia , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 8 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteína C1 de Niemann-Pick , Fosforilação , Regiões Promotoras Genéticas , Proteínas/genética , Proteínas/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Proteínas S100/metabolismo , Fatores de Tempo , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas rab de Ligação ao GTP/metabolismo
8.
BMC Immunol ; 9: 52, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18796140

RESUMO

BACKGROUND: Streptococcus pneumoniae is a human pathogenic bacteria and a major cause of severe invasive diseases, including pneumonia, bacteremia, and meningitis. Infections with S. pneumoniae evoke a strong inflammatory response, which plays a major role in the pathogenesis of pneumococcal disease. RESULTS: In this study, we have examined how S. pneumoniae affects expression of the inflammatory cytokine tumor necrosis factor (TNF) alpha, and the molecular mechanisms involved. Secretion of TNF-alpha was strongly induced by S. pneumoniae, which was able to stabilize TNF-alpha mRNA through a mechanism dependent on the viability of the bacteria as well as the adenylate uridylate-rich elements in the 3'untranslated region of TNF-alpha mRNA. The ability of S. pneumoniae to stabilize TNF-alpha mRNA was dependent on the mitogen-activated protein kinase (MAPK) p38 whereas inhibition of Toll-like receptor signaling via MyD88 did not affect S. pneumoniae-induced mRNA stabilization. P38 was activated through a pathway involving the upstream kinase transforming growth factor-activated kinase 1 and MAPK kinase 3. CONCLUSION: Thus, S. pneumoniae stabilizes TNF-alpha mRNA through a pathway dependent on p38 but independent of Toll-like receptors. Production of TNF-alpha may contribute significantly to the inflammatory response raised during pneumococcal infection.


Assuntos
Ativação de Macrófagos/genética , Macrófagos Peritoneais/metabolismo , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/metabolismo , Transdução de Sinais/imunologia , Streptococcus pneumoniae/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , MAP Quinase Quinase 3/genética , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Estabilidade de RNA/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/genética , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
9.
EMBO Rep ; 9(10): 1048-54, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18704119

RESUMO

Activation of c-Jun amino-terminal kinase (JNK) facilitates tumour necrosis factor (TNF)-induced cell death. The p38 mitogen-activated protein kinase pathway is induced by TNF stimulation, but it has not been implicated in TNF-induced cell death. Here, we show that hepatocyte-specific ablation of p38alpha in mice results in excessive activation of JNK in the liver after in vivo challenge with bacterial lipopolysaccharide (LPS). Despite increased JNK activity, p38alpha-deficient hepatocytes were not sensitive to LPS/TNF toxicity showing that JNK activation was not sufficient to mediate TNF-induced liver damage. By contrast, LPS injection caused liver failure in mice lacking both p38alpha and IkappaB kinase 2 (IKK2) in hepatocytes. Therefore, when combined with partial nuclear factor-kappaB inhibition, p38alpha deficiency sensitizes the liver to cytokine-induced damage. Collectively, these results reveal a new function of p38alpha in collaborating with IKK2 to protect the liver from LPS/TNF-induced failure by controlling JNK activation.


Assuntos
Endotoxinas/toxicidade , Quinase I-kappa B/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Falência Hepática/enzimologia , Falência Hepática/prevenção & controle , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Endotoxinas/antagonistas & inibidores , Ativação Enzimática/fisiologia , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Falência Hepática/induzido quimicamente , Falência Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
10.
Thromb Haemost ; 98(5): 1118-26, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18000619

RESUMO

Despite the success of the anti-coagulant protease protein C (PC) in treating septic shock in humans, the signaling pathways used are still unclear. To explore the effects of treatment with PC zymogen and its activated form aPC in a setting of sepsis, we employed a piglet model of endotoxic shock. In the aPC group, we observed a 65%-90% reduction in plasma TNF-alpha levels and a concomitant clinical improvement. Unexpectedly, administration of aPC also resulted in stabilization of the plasma pH above 7.2. Moreover, phosphorylated p38 mitogen-activated protein kinase (p38MAPK) was virtually absent in the livers of those piglets receiving aPC. In cultured human umbilical vein endothelial cells, we observed that nanomolar concentrations of PC and aPC inhibited the phosphorylation of p38MAPK. Furthermore, we showed that the regulation of the pro-apoptotic cell cycle regulator p53 by PC and aPC is dependent on the reduction of p38MAPK activation. The transduction of these effects involves all three receptors associated with protein C signaling, namely endothelial protein C receptor, protease-activated receptor 1, and sphingosine 1-phosphate receptor 1. Ultimately, this study elucidates novel signaling pathways regulated by protein C and emphasises the pivotal importance of its multiple modes of action beyond anticoagulation. APC's clinical success may, in part, be due to p38MAPK inhibition.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Proteína C/farmacologia , Choque Séptico/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/citologia , Humanos , Fígado/química , Fosforilação/efeitos dos fármacos , Proteína C/administração & dosagem , Proteína C/uso terapêutico , Suínos , Proteína Supressora de Tumor p53 , Veias Umbilicais/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Breast Cancer Res Treat ; 101(3): 269-78, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16897432

RESUMO

Wild-type p53-induced phosphatase (Wip1 or PPM1D) is a serine/threonine protein phosphatase expressed under various stress conditions, which selectively inactivates p38 MAPK. The finding that this gene is amplified in association with frequent gain of 17q21-24 in breast cancers supports its role as a driver oncogene. However, the pathogenetic mechanism of the wip1 gene expression in breast carcinogenesis remains to be elucidated. In this study, we examine Wip1 mRNA and protein expression in 20 breast cancer tissues and six cell lines. We additionally investigate the relationship among Wip1, active p38 MAPK, p53, and p16 proteins. In our experiments, Wip1 mRNA was significantly upregulated in 7 of 20 (35%) invasive breast cancer samples. Overexpression of Wip1 was inversely correlated with that of active (phosphor-) p38 MAPK (P = 0.007). Furthermore, Wip1-overexpressing tumors exhibited no or low levels of p16, which normally accumulates upon p38 MAPK activation (P = 0.057). Loss of p16 expression was not associated with hypermethylation of its promoter or loss of heterozygosity on 9p21. Among the 135 primary breast carcinomas further examined, a significant association was found between the Wip1 overexpression and negative staining for p53 (P value = 0.057), indicating that the tumors are wild-type for p53. This is first report showing that Wip1 overexpression abrogates the homeostatic balance maintained through the p38-p53-Wip1 pathway, and contributes to malignant progression by inactivating wild-type p53 and p38 MAPK as well as decreasing p16 protein levels in human breast tissues.


Assuntos
Neoplasias da Mama/genética , Carcinoma/genética , Genes p16/fisiologia , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/genética , Proteínas Supressoras de Tumor/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Biomarcadores Tumorais , Linhagem Celular , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Genes p53/fisiologia , Humanos , Imuno-Histoquímica , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Osteossarcoma/genética , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 2C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo , Neoplasias do Colo do Útero/genética , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
12.
J Immunol ; 174(3): 1239-44, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15661878

RESUMO

Signals mediated by the p38alpha MAPK have been implicated in many processes required for the development and effector functions of innate and adaptive immune responses. As mice deficient in p38alpha exhibit embryonic lethality, most analyses of p38alpha function in lymphocytes have relied on the use of pharmacologic inhibitors and dominant-negative or constitutively active transgenes. In this study, we have generated a panel of low passage p38alpha(+/+), p38alpha(+/-), and p38alpha(-/-) embryonic stem (ES) cells through the intercrossing of p38alpha(+/-) mice. These ES cells were used to generate chimeric mice by RAG-deficient blastocyst complementation, with the lymphocytes in these mice being derived entirely from the ES cells. Surprisingly, B and T cell development were indistinguishable when comparing chimeric mice generated with p38alpha(+/+), p38alpha(+/-), and p38alpha(-/-) ES cell lines. Moreover, proliferation of p38alpha(-/-) B and T cells in response to Ag receptor and non-Ag receptor stimuli was intact. Thus, p38alpha is not an essential component of signaling pathways required for robust B and T lymphocyte developmental, nor is p38alpha essential for the proliferation of mature B and T cells.


Assuntos
Proliferação de Células , Linfopoese , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Animais , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/enzimologia , Subpopulações de Linfócitos B/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular , Cruzamentos Genéticos , Embrião de Mamíferos , Feminino , Citometria de Fluxo , Triagem de Portadores Genéticos , Homozigoto , Imunofenotipagem , Isoenzimas/biossíntese , Isoenzimas/deficiência , Isoenzimas/fisiologia , Linfopoese/genética , Linfopoese/imunologia , Camundongos , Baço/citologia , Baço/enzimologia , Baço/imunologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Células-Tronco/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA