Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cell Death Dis ; 10(2): 145, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760700

RESUMO

Anaplastic Thyroid Cancer (ATC) is an aggressive malignancy with limited therapeutic options and dismal patient survival. We have previously shown MADD to be differentially overexpressed in multiple cancer histologies and to contribute to tumor cell growth and survival. Therefore, we targeted MADD by gene silencing, explored its effect on cellular proliferation and metastases and examined its therapeutic potential in an orthotopic ATC model in athymic nude mice. When compared to untreated control and scramble siRNA, MADD siRNA treatment inhibited the proliferative capacity of 8505C, C643 and HTH7 cells in vitro and 8505C-derived-orthotopic tumor growth in vivo. MADD ablation caused a significant reduction in cellular migration and invasion potential; clonogenic capacity; as well as, mitochondrial length and potential in vitro. This MADD siRNA-induced anti-migratory/invasive effect corresponded with inhibition of epithelial-mesenchymal transition (EMT) and Wnt signaling. Mechanistically, MADD siRNA inhibited TNFα induced activation of pERK, pGSK3ß and ß-catenin, suggesting that MADD knockdown might exert its anti-migratory/invasive effects, by blocking TNFα/ERK/GSK3ß axis. MADD siRNA can inhibit ß-catenin nuclear translocation and consequently, the expression of its target genes in ATC cells. In in vivo experiments, along with tumor regression, MADD siRNA treatment also decreased evidence of lung metastases. Immunohistochemically, MADD siRNA-treated tumor tissues exhibited a reduction in Ki67 and N-Cadherin expression, and an increase in E-Cadherin expression. In conclusion, we show the crucial role of MADD in ATC tumorigenesis and metastasis and its potential implications as a molecular target for ATC therapy.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/biossíntese , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Carcinoma Anaplásico da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Carcinoma Anaplásico da Tireoide/genética , Carcinoma Anaplásico da Tireoide/patologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Transfecção
2.
Radiat Res ; 186(4): 345-359, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27643878

RESUMO

Radiation-induced brain injury (RIBI) is the most common adverse effect that occurs after cranial radiation therapy (CRT). We have previously reported that CRT-induced release of pro-inflammatory cytokines in brain tissues and inhibition of neurogenesis in the hippocampus might be caused by microglial activation and may play an important role in RIBI. In this study we examined the role of p53-induced protein with a death domain (PIDD) in radiation-induced activation of BV-2 cells. BV-2 cells were transfected with antisense oligonucleotide control mRNA or antisense oligonucleotide-targeted PIDD mRNA and were sham or 16 Gy irradiated. The state of microglia and expression of pro-inflammatory cytokines were detected using real-time polymerase chain reaction, Western blotting, immunofluorescence and flow cytometry. Findings from this study suggest that silencing PIDD expression could inhibit microglial activation by downregulating the PIDD-C/NF-κß transcription pathway. PIDD acts as a critical switcher between the NF-κß transcription pathway and radiation-induced apoptosis. Given these findings, this study offers a potential novel approach to further combination treatment of RIBI.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Microglia/citologia , Microglia/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Ciclo Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Senescência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Regulação para Baixo/efeitos da radiação , Inativação Gênica , Mediadores da Inflamação/metabolismo , Camundongos , Microglia/metabolismo , NF-kappa B/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação/genética , Transdução de Sinais/efeitos da radiação
3.
Methods Mol Biol ; 1165: 197-222, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24839027

RESUMO

DEDD, a member of a family of death effector domain-containing proteins, plays crucial roles in mediating apoptosis, regulating cell cycle, and inhibiting cell mitosis. Our recent work demonstrates that DEDD is a novel tumor repressor, which impedes metastasis by reversing the epithelial-mesenchymal transition (EMT) process in breast and colon cancers. DEDD expression therefore may represent a prognostic marker and potential therapeutic target for the prevention and treatment of cancer metastasis. To reveal the anti-metastatic roles of DEDD in these cancer cells, a number of experiments, including immunohistochemistry, the establishment of stably overexpressing or silencing cancer cells, chemoinvasion assay, soft agar assay, protein degradation, and protein-protein interaction were used in our in vitro and in vivo studies. This chapter focuses on the details of these experiments to provide references for the researchers to investigate the function of a gene in the regulation of tumor metastasis.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Resistencia a Medicamentos Antineoplásicos , Técnicas de Silenciamento de Genes , Inativação Gênica , Gentamicinas/farmacologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Metástase Neoplásica , Prognóstico , Proteólise , Transfecção , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Técnicas do Sistema de Duplo-Híbrido
4.
PLoS One ; 8(2): e56817, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23457619

RESUMO

The Map kinase Activating Death Domain containing protein (MADD) isoform of the IG20 gene is over-expressed in different types of cancer tissues and cell lines and it functions as a negative regulator of apoptosis. Therefore, we speculated that MADD might be over-expressed in human breast cancer tissues and that MADD knock-down might synergize with chemotherapeutic or TRAIL-induced apoptosis of breast cancer cells. Analyses of breast tissue microarrays revealed over-expression of MADD in ductal and invasive carcinomas relative to benign tissues. MADD knockdown resulted in enhanced spontaneous apoptosis in human breast cancer cell lines. Moreover, MADD knockdown followed by treatment with TRAIL or doxorubicin resulted in increased cell death compared to either treatment alone. Enhanced cell death was found to be secondary to increased caspase-8 activation. These data indicate that strategies to decrease MADD expression or function in breast cancer may be utilized to increase tumor cell sensitivity to TRAIL and doxorubicin induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Doxorrubicina/farmacologia , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Sequência de Aminoácidos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Receptores de Morte Celular/metabolismo
5.
J Clin Invest ; 121(1): 318-27, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21135503

RESUMO

During intrauterine life, the mammalian embryo survives via its physical connection to the mother. The uterine decidua, which differentiates from stromal cells after implantation in a process known as decidualization, plays essential roles in supporting embryonic growth before establishment of the placenta. Here we show that female mice lacking death effector domain-containing protein (DEDD) are infertile owing to unsuccessful decidualization. In uteri of Dedd-/- mice, development of the decidual zone and the surrounding edema after embryonic implantation was defective. This was subsequently accompanied by disintegration of implantation site structure, leading to embryonic death before placentation. Polyploidization, a hallmark of mature decidual cells, was attenuated in DEDD-deficient cells during decidualization. Such inefficient decidualization appeared to be caused by decreased Akt levels, since polyploidization was restored in DEDD-deficient decidual cells by overexpression of Akt. In addition, we showed that DEDD associates with and stabilizes cyclin D3, an important element in polyploidization, and that overexpression of cyclin D3 in DEDD-deficient cells improved polyploidization. These results indicate that DEDD is indispensable for the establishment of an adequate uterine environment to support early pregnancy in mice.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/fisiologia , Decídua/fisiologia , Animais , Diferenciação Celular , Ciclina D3/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Decídua/citologia , Implantação do Embrião/genética , Implantação do Embrião/fisiologia , Perda do Embrião/genética , Perda do Embrião/patologia , Perda do Embrião/fisiopatologia , Feminino , Humanos , Infertilidade/genética , Infertilidade/patologia , Infertilidade/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placentação/genética , Placentação/fisiologia , Poliploidia , Gravidez , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Proc Natl Acad Sci U S A ; 104(7): 2289-94, 2007 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-17283331

RESUMO

Accumulating evidence has shown that many molecules, including some cyclin-dependent kinases (Cdks) and cyclins, as well as the death-effector domain (DED)-containing FADD, function for both apoptosis and cell cycle. Here we identified that DEDD, which also possesses the DED domain, acts as a novel inhibitor of the mitotic Cdk1/cyclin B1 complex. DEDD associates with mitotic Cdk1/cyclin B1 complexes via direct binding to cyclin B1 and reduces their function. In agreement, kinase activity of nuclear Cdk1/cyclin B1 in DEDD-null (DEDD-/-) embryonic fibroblasts is increased compared with that in DEDD+/+ cells, which results in accelerated mitotic progression, thus exhibiting a shortened G2/M stage. Interestingly, DEDD-/- cells also demonstrated decreased G1 duration, which perhaps enhanced the overall reduction in rRNA amounts and cell volume, primarily caused by the rapid termination of rRNA synthesis before cell division. Likewise, DEDD-/- mice show decreased body and organ weights relative to DEDD+/+ mice. Thus, DEDD is an impeder of cell mitosis, and its absence critically influences cell and body size via modulation of rRNA synthesis.


Assuntos
Proteína Quinase CDC2/antagonistas & inibidores , Ciclina B/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/fisiologia , Mitose/fisiologia , Animais , Tamanho Corporal , Proteína Quinase CDC2/metabolismo , Células Cultivadas , Ciclina B/metabolismo , Ciclina B1 , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Fibroblastos , Interfase/fisiologia , Camundongos , Camundongos Knockout , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Tamanho do Órgão , Ligação Proteica , RNA Ribossômico/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA