Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 6(28): 25356-67, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26327325

RESUMEN

Triple-negative breast cancer (TNBC) is a high medical need disease with limited treatment options. CD8+ T cell-mediated immunotherapy may represent an attractive approach to address TNBC. The objectives of this study were to assess the expression of CXorf61 in TNBCs and healthy tissues and to evaluate its capability to induce T cell responses. We show by transcriptional profiling of a broad comprehensive set of normal human tissue that CXorf61 expression is strictly restricted to testis. 53% of TNBC patients express this antigen in at least 30% of their tumor cells. In CXorf61-negative breast cancer cell lines CXorf61 expression is activated by treatment with the hypomethylating agent 5-aza-2'-deoxycytidine. By vaccination of HLA-A*02-transgenic mice with CXorf61 encoding RNA we obtained high frequencies of CXorf61-specific T cells. Cloning and characterization of T cell receptors (TCRs) from responding T cells resulted in the identification of the two HLA-A*0201-restricted T cell epitopes CXorf6166-74 and CXorf6179-87. Furthermore, by in vitro priming of human CD8+ T cells derived from a healthy donor recognizing CXorf6166-74 we were able to induce a strong antigen-specific immune response and clone a human TCR recognizing this epitope. In summary, our data confirms this antigen as promising target for T cell based therapies.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Neoplasias de la Mama Triple Negativas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Clonación Molecular , Técnicas de Cocultivo , Metilación de ADN , Mapeo Epitopo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Esquemas de Inmunización , Células K562 , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Transgénicos , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transfección , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
2.
Mol Cancer Ther ; 12(10): 2226-36, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23924947

RESUMEN

Epigenetic alterations are a hallmark of cancer that govern the silencing of genes. Up to now, 5-azacytidine (5-aza-CR, Vidaza) and 5-aza-2'-deoxycytidine (5-aza-dC, Dacogen) are the only clinically approved DNA methyltransferase inhibitors (DNMTi). Current effort tries to exploit DNMTi application beyond acute leukemia or myelodysplastic syndrome, especially to solid tumors. Although both drugs only differ by a minimal structural difference, they trigger distinct molecular mechanisms that are highly relevant for a rational choice of new combination therapies. Therefore, we investigated cell death pathways in vitro in human hepatoma, colon, renal, and lung cancer cells and in vivo in chorioallantoic membrane and xenograft models. Real-time cancer cell monitoring and cytokine profiling revealed a profoundly distinct response pattern to both drugs. 5-aza-dC induced p53-dependent tumor cell senescence and a high number of DNA double-strand breaks. In contrast, 5-aza-CR downregulated p53, induced caspase activation and apoptosis. These individual response patterns of tumor cells could be verified in vivo in chorioallantoic membrane assays and in a hepatoma xenograft model. Although 5-aza-CR and 5-aza-dC are viewed as drugs with similar therapeutic activity, they induce a diverse molecular response in tumor cells. These findings together with other reported differences enable and facilitate a rational design of new combination strategies to further exploit the epigenetic mode of action of these two drugs in different areas of clinical oncology.


Asunto(s)
Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena , Metilación de ADN/genética , Decitabina , Células Hep G2 , Humanos , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Anticancer Drugs ; 22(6): 494-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21566522

RESUMEN

Glioblastoma is the most common primary brain tumor with a dismal prognosis, highlighting the need for novel treatment strategies. Here, we provide the first evidence that the histone deacetylase inhibitor, MS275, sensitizes glioblastoma cells for chemotherapy-induced apoptosis. Pretreatment of glioblastoma cells with MS275 causes acetylation of histone H3 protein and significantly enhances doxorubicin-induced apoptosis. Calculation of combination index showed that MS275 and doxorubicin acted in a synergistic manner to trigger apoptosis. Furthermore, pre-exposure to MS275 significantly increases apoptosis in response to temozolomide, etoposide, and cisplatin. In contrast, treatment with MS275 before the addition of vincristine and taxol significantly reduces the induction of apoptosis. Analysis of cell cycle alterations showed that treatment with MS275 triggers G1 cell cycle arrest, which in turn renders cells less sensitive to the cytotoxic effects of mitotic inhibitors, such as vincristine and taxol. Thus, these findings show for the first time that the histone deacetylase inhibitor, MS275, represents a promising strategy to prime glioblastoma cells for chemotherapy-induced apoptosis in a drug-specific manner.


Asunto(s)
Antineoplásicos , Benzamidas/farmacología , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Piridinas/farmacología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Cisplatino/farmacología , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Doxorrubicina/farmacología , Sinergismo Farmacológico , Etopósido/farmacología , Histonas/metabolismo , Humanos , Microtúbulos/efectos de los fármacos , Temozolomida
4.
Gut ; 60(2): 156-65, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21106551

RESUMEN

BACKGROUND: The cytokine tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) has shown promising anticancer activity in early clinical settings by selectively inducing apoptosis in different tumour types. However, some tumour entities such as hepatocellular carcinoma (HCC) display an inherent resistance to TRAIL. A huge effort has been made to unravel strategies for a clinically applicable sensitisation of resistant cancer cells to TRAIL. Reversible epigenetic alterations such as DNA methylation play a major role in development, maintenance and resistance phenomena of tumour cells. Currently, several clinical trials are exploiting the potential of epigenetic drugs, such as 5-azacytidine (5-aza-CR) or 5-aza-2'-deoxycytidine (5-aza-dC) to break primary or secondary resistance phenomena of cancer cells. Therefore, 5-aza-CR and 5-aza-dC were investigated in the context of TRAIL resistance. METHODS: Alterations in proliferation, apoptosis, regulatory proteins and toxicity were investigated in TRAIL-resistant hepatoma, and also in renal, colon and pancreatic cancer cells as well as non-transformed human-derived primary hepatocytes, tissue slices isolated from human liver and non-malignant colon cells, all of which had been exposed to demethylating drugs and/or TRAIL. RESULTS: Within hours, 5-aza-CR but not 5-aza-dC sensitised in vitro cultured tumour cells to TRAIL, first by activating caspases, followed by a subsequent induction of apoptosis. This surprisingly rapid sensitisation was confirmed in vivo employing a chorioallantoic membrane assay. As a major mechanism, a 5-aza-CR-induced inhibition of cellular protein synthesis was found which led to a breakdown of tumour-protecting factors such as the antiapoptotic factor FLICE inhibitory protein (FLIP). Importantly, TRAIL and 5-aza-CR did not induce relevant toxicity or apoptosis in primary hepatocytes, liver slices from different human donors and in normal colon cells. CONCLUSIONS: Molecular evidence is provided for a novel 5-aza-CR-based translational approach enabling a twofold treatment of apoptosis-resistant tumour entities, not only by an epigenetic reversion of the malignancy-associated phenotype but also by an efficient resensitization to apoptosis-inducing substances such as TRAIL.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/farmacología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Carcinoma Hepatocelular/metabolismo , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/genética , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Humanos , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Células Tumorales Cultivadas
5.
J Cell Mol Med ; 13(10): 4239-56, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19725919

RESUMEN

NF-kappaB is activated by DNA-damaging anticancer drugs as part of the cellular stress response. However, the consequences of drug-induced NF-kappaB activation are still only partly understood. To investigate the impact of NF-kappaB on the cell's response to DNA damage, we engineered glioblastoma cells that stably express mutant IkappaBalpha superrepressor (IkappaBalpha-SR) to block NF-kappaB activation. Here, we identify a novel pro-apoptotic function of NF-kappaB in the DNA damage response in glioblastoma cells. Chemotherapeutic drugs that intercalate into DNA and inhibit topoisomerase II such as Doxorubicin, Daunorubicin and Mitoxantrone stimulate NF-kappaB DNA binding and transcriptional activity prior to induction of cell death. Importantly, specific inhibition of drug-induced NF-kappaB activation by IkappaBalpha-SR or RNA interference against p65 significantly reduces apoptosis upon treatment with Doxorubicin, Daunorubicin or Mitoxantrone. NF-kappaB exerts this pro-apoptotic function especially after pulse drug exposure as compared to continuous treatment indicating that the contribution of NF-kappaB becomes relevant during the recovery phase following the initial DNA damage. Mechanistic studies show that NF-kappaB inhibition does not alter Doxorubicin uptake and efflux or cell cycle alterations. Genetic silencing of p53 by RNA interference reveals that NF-kappaB promotes drug-induced apoptosis in a p53-independent manner. Intriguingly, drug-mediated NF-kappaB activation results in a significant increase in DNA damage prior to the induction of apoptosis. By demonstrating that NF-kappaB promotes DNA damage formation and apoptosis upon pulse treatment with DNA intercalators, our findings provide novel insights into the control of the DNA damage response by NF-kappaB in glioblastoma.


Asunto(s)
Apoptosis , Daño del ADN , FN-kappa B/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Sustancias Intercalantes/farmacología , FN-kappa B/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Cancer Res ; 66(20): 10016-23, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17047064

RESUMEN

Inactivation of caspase-8 because of aberrant gene methylation has been associated with amplification of the MYCN oncogene and aggressive disease in neuroblastoma, suggesting that caspase-8 may function as tumor suppressor. However, the prognostic effect of caspase-8 in neuroblastoma has remained obscure. Therefore, we investigated caspase-8 expression and its correlation with established prognostic markers and survival outcome in a large cohort of neuroblastoma patients. Here, we report that loss of caspase-8 protein expression occurs in the majority (75%) of neuroblastoma and is not restricted to advanced disease stages. Surprisingly, no correlation was observed between caspase-8 expression and MYCN amplification. Similarly, ectopic expression of MYCN or antisense-mediated down-regulation of MYCN had no effect on caspase-8 expression in neuroblastoma cell lines. In addition, caspase-8 expression did not correlate with other variables of high-risk disease (e.g., 1p36 aberrations, disease stage, age at diagnosis, or tumor histology). Most importantly, loss of caspase-8 protein had no effect on event-free or overall survival in the overall study population or in distinct subgroups of patients. By revealing no correlation between caspase-8 expression and MYCN amplification or other established variables of aggressive disease, our findings in a large cohort of neuroblastoma patients show that inactivation of caspase-8 is not a characteristic feature of aggressive neuroblastoma. Thus, our study provides novel insight into the biology of this tumor, which may have important clinical implications.


Asunto(s)
Caspasa 8/biosíntesis , Genes myc , Neuroblastoma/enzimología , Síndrome de Alstrom , Western Blotting , Caspasa 8/genética , Línea Celular Tumoral , Metilación de ADN , Progresión de la Enfermedad , Amplificación de Genes , Perfilación de la Expresión Génica , Humanos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética
7.
J Biol Chem ; 279(7): 5049-52, 2004 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-14676199

RESUMEN

Messenger RNAs are transported to the cytoplasm bound to several shuttling mRNA-binding proteins. Here, we present the characterization of Hrb1, a novel component of the transported ribonucleoprotein complex in Saccharomyces cerevisiae. The protein is similar to the other two serine/arginine (SR)-type proteins in yeast, Gbp2 and Npl3. Hrb1 is nuclear at steady state and its import is mediated by the karyopherin Mtr10. Hrb1 binds to poly(A)+ RNA in vivo and its binding is significantly increased in MTR10 mutants, suggesting a role for Mtr10 in dissociating Hrb1 from the mRNAs. Interestingly, by comparing the export requirements of all three SR proteins we find similarities but also striking differences. While the export of all three proteins is dependent on the export of mRNAs in general, as no transport is observed in mutants defective in transcription (rpb1-1) or mRNA export (mex67-5), we find specific requirements for components of the THO complex, involved in transcription elongation. While both Hrb1 and Gbp2 depend on Mft1 and Hpr1 for their nuclear export, Npl3 is exported independently of both proteins. These findings suggest that Hrb1 and Gbp2, but not Npl3, might be loaded onto the growing mRNA via the THO complex components Mtf1 and Hrp1.


Asunto(s)
Arginina/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Serina/química , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes , Ribonucleoproteínas Nucleares Heterogéneas/química , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Proteínas Luminiscentes/metabolismo , Proteínas Mitocondriales , Mutación , Proteínas Nucleares , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Plásmidos/metabolismo , Poli A/metabolismo , Proteínas de Unión a Poli(A) , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Factores de Transcripción/metabolismo , Transcripción Genética , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...