Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Natl Cancer Inst ; 109(1)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27707838

RESUMO

Background: Previously we identified a DNA damage response-deficient (DDRD) molecular subtype within breast cancer. A 44-gene assay identifying this subtype was validated as predicting benefit from DNA-damaging chemotherapy. This subtype was defined by interferon signaling. In this study, we address the mechanism of this immune response and its possible clinical significance. Methods: We used immunohistochemistry (IHC) to characterize immune infiltration in 184 breast cancer samples, of which 65 were within the DDRD subtype. Isogenic cell lines, which represent DDRD-positive and -negative, were used to study the effects of chemokine release on peripheral blood mononuclear cell (PBMC) migration and the mechanism of immune signaling activation. Finally, we studied the association between the DDRD subtype and expression of the immune-checkpoint protein PD-L1 as detected by IHC. All statistical tests were two-sided. Results: We found that DDRD breast tumors were associated with CD4+ and CD8+ lymphocytic infiltration (Fisher's exact test P < .001) and that DDRD cells expressed the chemokines CXCL10 and CCL5 3.5- to 11.9-fold more than DNA damage response-proficient cells (P < .01). Conditioned medium from DDRD cells statistically significantly attracted PBMCs when compared with medium from DNA damage response-proficient cells (P < .05), and this was dependent on CXCL10 and CCL5. DDRD cells demonstrated increased cytosolic DNA and constitutive activation of the viral response cGAS/STING/TBK1/IRF3 pathway. Importantly, this pathway was activated in a cell cycle-specific manner. Finally, we demonstrated that S-phase DNA damage activated expression of PD-L1 in a STING-dependent manner. Conclusions: We propose a novel mechanism of immune infiltration in DDRD tumors, independent of neoantigen production. Activation of this pathway and associated PD-L1 expression may explain the paradoxical lack of T-cell-mediated cytotoxicity observed in DDRD tumors. We provide a rationale for exploration of DDRD in the stratification of patients for immune checkpoint-based therapies.


Assuntos
Neoplasias da Mama/imunologia , Dano ao DNA/imunologia , DNA/análise , Imunidade Inata , Leucócitos Mononucleares/fisiologia , Linfócitos do Interstício Tumoral , Proteínas de Membrana/metabolismo , Antígeno B7-H1/metabolismo , Neoplasias da Mama/genética , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/metabolismo , Quimiotaxia/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Citosol/química , Feminino , Humanos , Imuno-Histoquímica , Fator Regulador 3 de Interferon/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fase S/genética , Transdução de Sinais
2.
Oncogene ; 33(6): 713-723, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23435429

RESUMO

BRCA1 mediates resistance to apoptosis in response to DNA-damaging agents, causing BRCA1 wild-type tumours to be significantly more resistant to DNA damage than their mutant counterparts. In this study, we demonstrate that following treatment with the DNA-damaging agents, etoposide or camptothecin, BRCA1 is required for the activation of nuclear factor-κB (NF-κB), and that BRCA1 and NF-κB cooperate to regulate the expression of the NF-κB antiapoptotic targets BCL2 and XIAP. We show that BRCA1 and the NF-κB subunit p65/RelA associate constitutively, whereas the p50 NF-κB subunit associates with BRCA1 only upon DNA damage treatment. Consistent with this BRCA1 and p65 are present constitutively on the promoters of BCL2 and XIAP, whereas p50 is recruited to these promoters only in damage treated cells. Importantly, we demonstrate that the recruitment of p50 onto the promoters of BCL2 and XIAP is dependent upon BRCA1, but independent of its NF-κB partner subunit p65. The functional relevance of NF-κB activation by BRCA1 in response to etoposide and camptothecin is demonstrated by the significantly reduced survival of BRCA1 wild-type cells upon NF-κB inhibition. This study identifies a novel BRCA1-p50 complex, and demonstrates for the first time that NF-κB is required for BRCA1-mediated resistance to DNA damage. It reveals a functional interdependence between BRCA1 and NF-κB, further elucidating the role played by NF-κB in mediating cellular resistance of BRCA1 wild-type tumours to DNA-damaging agents.


Assuntos
Proteína BRCA1/metabolismo , Camptotecina/farmacologia , Dano ao DNA , Etoposídeo/farmacologia , Subunidade p50 de NF-kappa B/metabolismo , NF-kappa B/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína BRCA1/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Células HEK293 , Humanos , NF-kappa B/genética , Subunidade p50 de NF-kappa B/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica , Transfecção
3.
Cancer Res ; 70(6): 2538-47, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20215511

RESUMO

We carried out a yeast two-hybrid screen using a BRCA1 bait composed of amino acids 1 to 1142 and identified BRD7 as a novel binding partner of BRCA1. This interaction was confirmed by coimmunoprecipitation of endogenous BRCA1 and BRD7 in T47D and HEK-293 cells. BRD7 is a bromodomain containing protein, which is a subunit of PBAF-specific Swi/Snf chromatin remodeling complexes. To determine the functional consequences of the BRCA1-BRD7 interaction, we investigated the role of BRD7 in BRCA1-dependent transcription using microarray-based expression profiling. We found that a variety of targets were coordinately regulated by BRCA1 and BRD7, such as estrogen receptor alpha (ERalpha). Depletion of BRD7 or BRCA1 in either T47D or MCF7 cells resulted in loss of expression of ERalpha at both the mRNA and protein level, and this loss of ERalpha was reflected in resistance to the antiestrogen drug fulvestrant. We show that BRD7 is present, along with BRCA1 and Oct-1, on the ESR1 promoter (the gene which encodes ERalpha). Depletion of BRD7 prevented the recruitment of BRCA1 and Oct-1 to the ESR1 promoter; however, it had no effect on the recruitment of the other Swi/Snf subunits BRG1, BAF155, and BAF57 or on RNA polymerase II recruitment. These results support a model whereby the regulation of ERalpha transcription by BRD7 is mediated by its recruitment of BRCA1 and Oct-1 to the ESR1 promoter.


Assuntos
Proteína BRCA1/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína BRCA1/genética , Neoplasias da Mama/tratamento farmacológico , Proteínas Cromossômicas não Histona/genética , Moduladores de Receptor Estrogênico/farmacologia , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Feminino , Perfilação da Expressão Gênica , Genes BRCA1 , Humanos , Imunoprecipitação , Fator 1 de Transcrição de Octâmero/genética , Fator 1 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transcrição Gênica , Transfecção
4.
J Exp Med ; 197(3): 343-51, 2003 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-12566418

RESUMO

Toll-like receptors (TLRs) are crucial in the innate immune response to pathogens, in that they recognize and respond to pathogen associated molecular patterns, which leads to activation of intracellular signaling pathways and altered gene expression. Vaccinia virus (VV), the poxvirus used to vaccinate against smallpox, encodes proteins that antagonize important components of host antiviral defense. Here we show that the VV protein A52R blocks the activation of the transcription factor nuclear factor kappa B (NF-kappa B) by multiple TLRs, including TLR3, a recently identified receptor for viral RNA. A52R associates with both interleukin 1 receptor-associated kinase 2 (IRAK2) and tumor necrosis factor receptor-associated factor 6 (TRAF6), two key proteins important in TLR signal transduction. Further, A52R could disrupt signaling complexes containing these proteins. A virus deletion mutant lacking the A52R gene was attenuated compared with wild-type and revertant controls in a murine intranasal model of infection. This study reveals a novel mechanism used by VV to suppress the host immunity. We demonstrate viral disabling of TLRs, providing further evidence for an important role for this family of receptors in the antiviral response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Drosophila , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/imunologia , Receptores de Superfície Celular/imunologia , Vaccinia virus/imunologia , Vaccinia virus/patogenicidade , Proteínas Virais/imunologia , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Feminino , Deleção de Genes , Genes Virais , Humanos , Tolerância Imunológica , Quinases Associadas a Receptores de Interleucina-1 , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Receptores de Superfície Celular/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais , Fator 6 Associado a Receptor de TNF , Receptor 3 Toll-Like , Receptores Toll-Like , Vaccinia virus/genética , Proteínas Virais/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA