Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Cancer ; 144(7): 1685-1696, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30478958

RESUMEN

Here, we present a functional assay to detect the repair switch to the alternative PARP1-dependent end joining (PARP1-EJ) pathway and the associated susceptibility to PARPi-mediated radiosensitization in freshly collected tumor samples from prostate cancer (PCa) patients, thereby facilitating the selection of patients who should benefit from combined PARPi plus radiotherapy (RT) treatment. Our optimized ex-vivo approach sustains tumor slices for up to 15 days under culture conditions that maintain proliferation and oxygenation rates, as measured by EdU incorporation and pimonidazole staining, respectively. We present a robust system to analyze DSB repair using, for the first time in an ex vivo tumor slice setting, two DSB-markers simultaneously i.e. γH2AX and 53BP1. A computer-based processing method (i) controls variations in DNA content and slicing on the number of repair foci and (ii) measures the PARPi-mediated enhancement ratio on DSB foci numbers to ensure inter-patient-comparability. We validated this approach using a PC3 xenograft model with its previously described repair switch to PARP1-EJ. More importantly, we show that approximately 30% of the analyzed tumor tissue samples collected from PCa patients display a switch to PARP1-EJ, as indicated by the enhanced number of residual γH2AX/53BP1 foci exclusively after PARPi+RT. Furthermore, normal prostatic tissues show no repair switch to PARP1-EJ, indicating that this repair switch and its associated radiosensitizing effect is tumor-specific. Collectively, we present here a predictive assay for the switch to PARP1-EJ that enables individualization of anti-cancer treatment using a combination of RT and radiosensitizing anticancer agents such as PARPi in PCa.


Asunto(s)
Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Neoplasias de la Próstata/terapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/efectos de la radiación , Histonas/metabolismo , Humanos , Masculino , Ratones , Clasificación del Tumor , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Fármacos Sensibilizantes a Radiaciones/farmacología , Técnicas de Cultivo de Tejidos , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
3.
BMC Cancer ; 18(1): 1106, 2018 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-30419865

RESUMEN

BACKGROUND: RBM3 expression has been suggested as prognostic marker in several cancer types. The purpose of this study was to assess the prevalence and clinical significance of altered RBM3 expression in esophageal cancer. METHODS: RBM3 protein expression was measured by immunohistochemistry using tissue microarrays containing samples from 359 esophageal adenocarcinoma (EAC) and 254 esophageal squamous cell cancer (ESCC) patients with oncological follow-up data. RESULTS: While nuclear RBM3 expression was always high in benign esophageal epithelium, high RBM3 expression was only detectable in 66.4% of interpretable EACs and 59.3% of ESCCs. Decreased RBM3 expression was linked to a subset of EACs with advanced UICC stage and presence of distant metastasis (P = 0.0031 and P = 0.0024). In ESCC, decreased RBM3 expression was associated with advanced UICC stage, high tumor stage, and positive lymph node status (P = 0.0213, P = 0.0061, and P = 0.0192). However, RBM3 expression was largely unrelated to survival of patients with esophageal cancer (EAC: P = 0.212 and ESCC: P = 0.5992). CONCLUSIONS: In summary, the present study shows that decreased RBM3 expression is associated with unfavourable esophageal cancer phenotype, but not significantly linked to patient prognosis.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ARN/genética , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Neoplasias Esofágicas/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Evaluación del Resultado de la Atención al Paciente , Fenotipo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Prostate ; 77(15): 1528-1538, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28929505

RESUMEN

BACKGROUND: Glyoxalase 1 (GLO1) is an enzyme involved in removal of toxic byproducts accumulating during glycolysis from the cell. GLO1 is up regulated in many cancer types but its role in prostate cancer is largely unknown. METHODS: Here, we employed GLO1 immunohistochemistry on a tissue microarray including 11 152 tumors and an attached clinical and molecular database. RESULTS: Normal prostate epithelium was negative for GLO1, whereas 2059 (27.3%) of 7552 interpretable cancers showed cytoplasmic GLO1 staining, which was considered weak in 8.8%, moderate in 12.5%, and strong in 6.1% of tumors. Up regulation of GLO1 was significantly linked to high original Gleason grade, advanced pathological tumor stage and positive lymph node status (P < 0.0001 each). Comparison of GLO1 staining with several common genomic alterations of prostate cancers revealed a strong link between GLO1 up regulation and TMPRSS2:ERG fusion (P < 0.0001) and an ERG-independent association with PTEN deletion (P < 0.0001). GLO1 up regulation was strongly linked to early biochemical recurrence in univariate analysis (P < 0.0001) and predicted poor prognosis independent from most (except from nodal stage) established prognostic parameters in multivariate analysis (P ≤ 0.03). CONCLUSIONS: GLO1 upregulation is linked to aggressive prostate cancers characterized by ERG fusion and PTEN deletion. The strong and independent prognostic value makes it a promising candidate for routine diagnostic applications either alone or in combination with other markers.


Asunto(s)
Lactoilglutatión Liasa/biosíntesis , Neoplasias de la Próstata/enzimología , Anciano , Biomarcadores de Tumor/biosíntesis , Humanos , Inmunohistoquímica , Calicreínas/metabolismo , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/enzimología , Pronóstico , Antígeno Prostático Específico/metabolismo , Análisis de Matrices Tisulares
5.
Tumour Biol ; 39(10): 1010428317712166, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29022485

RESUMEN

Microtubules are multifunctional cytoskeletal proteins that are involved in crucial cellular roles including maintenance of cell shape, intracellular transport, meiosis, and mitosis. Class III beta-tubulin (ßIII-tubulin, also known as TUBB3) is a microtubule protein, normally expressed in cells of neuronal origin. Its expression was also reported in various other tumor types, such as several types of lung cancer, ovarian cancer, and esophageal cancer. TUBB3 is of clinical relevance as overexpression has been linked to poor response to microtubule-targeting anti-cancer drugs such as taxanes. To systematically investigate the epidemiology of TUBB3 expression in normal and neoplastic tissues, we used tissue microarrays for analyzing the immunohistochemically detectable expression of TUBB3 in 3911 tissue samples from 100 different tumor categories and 76 different normal tissue types. At least 1 tumor with weak expression could be found in 93 of 100 (93%) different tumor types, and all these 93 entities also had at least 1 tumor with strong positivity. In normal tissues, a particularly strong expression was found in neurons of the brain, endothelium of blood vessels, fibroblasts, spermatogenic cells, stroma cells, endocrine cells, and acidophilic cells of the pituitary gland. In tumors, strong TUBB3 expression was most frequently found in various brain tumors (85%-100%), lung cancer (35%-80%), pancreatic adenocarcinoma (50%), renal cell carcinoma (15%-80%), and malignant melanoma (77%). In summary, these results identify a broad spectrum of cancers that can at least sporadically express TUBB3. Testing of TUBB3 in cancer types eligible for taxane-based therapies could be helpful to identify patients who might best benefit from this treatment.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Tubulina (Proteína)/genética , Biomarcadores de Tumor/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/clasificación , Neoplasias/patología , Taxoides/uso terapéutico , Tubulina (Proteína)/biosíntesis
6.
BMC Cancer ; 17(1): 504, 2017 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747165

RESUMEN

BACKGROUND: Animal model experiments have suggested a role of the DNA repair protein ERCC1 (Excision Repair Cross-Complementation Group 1) in prostate cancer progression. METHODS: To better understand the impact of ERCC1 protein expression in human prostate cancer, a preexisting tissue microarray (TMA) containing more than 12,000 prostate cancer specimens was analyzed by immunohistochemistry and data were compared with tumor phenotype, PSA recurrence and several of the most common genomic alterations (TMPRSS2:ERG fusions: deletions of PTEN, 6q, 5q, 3p). RESULTS: ERCC1 staining was seen in 64.7% of 10,436 interpretable tissues and was considered weak in 37.1%, moderate in 22.6% and strong in 5% of tumors. High-level ERCC1 staining was linked to advanced pT stage, high Gleason grade, positive lymph nodes, high pre-operative serum PSA, and positive surgical margin status (p < 0.0001 each). High ERCC1 expression was strongly associated with an elevated risk of PSA recurrence (p < 0.0001). This was independent of established prognostic features. A subgroup analysis of cancers defined by comparable quantitative Gleason grades revealed that the prognostic impact was mostly driven by low-grade tumors with a Gleason 3 + 3 or 3 + 4 (Gleason 4: ≤5%). High ERCC1 expression was strongly associated with the presence of genomic alterations and expression levels increased with the number of deletions present in the tumor. These latter data suggest a functional relationship of ERCC1 expression with genomic instability. CONCLUSION: The results of our study demonstrate that expression of ERCC1 - a potential surrogate for genomic instability - is an independent prognostic marker in prostate cancer with particular importance in low-grade tumors.


Asunto(s)
Aberraciones Cromosómicas , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Neoplasias de la Próstata/metabolismo , Anciano , Proliferación Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Inestabilidad Genómica , Humanos , Calicreínas/sangre , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Clasificación del Tumor , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Modelos de Riesgos Proporcionales , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología
7.
Strahlenther Onkol ; 191(3): 248-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25245467

RESUMEN

INTRODUCTION: Radioprotective agents are of interest for application in radiotherapy for cancer and in public health medicine in the context of accidental radiation exposure. Methylproamine is the lead compound of a class of radioprotectors which act as DNA binding anti-oxidants, enabling the repair of transient radiation-induced oxidative DNA lesions. This study tested methylproamine for the radioprotection of both directly targeted and bystander cells. METHODS: T98G glioma cells were treated with 15 µM methylproamine and exposed to (137)Cs γ-ray/X-ray irradiation and He(2+) microbeam irradiation. Radioprotection of directly targeted cells and bystander cells was measured by clonogenic survival or γH2AX assay. RESULTS: Radioprotection of directly targeted T98G cells by methylproamine was observed for (137)Cs γ-rays and X-rays but not for He(2+) charged particle irradiation. The effect of methylproamine on the bystander cell population was tested for both X-ray irradiation and He(2+) ion microbeam irradiation. The X-ray bystander experiments were carried out by medium transfer from irradiated to non-irradiated cultures and three experimental designs were tested. Radioprotection was only observed when recipient cells were pretreated with the drug prior to exposure to the conditioned medium. In microbeam bystander experiments targeted and nontargeted cells were co-cultured with continuous methylproamine treatment during irradiation and postradiation incubation; radioprotection of bystander cells was observed. DISCUSSION AND CONCLUSION: Methylproamine protected targeted cells from DNA damage caused by γ-ray or X-ray radiation but not He(2+) ion radiation. Protection of bystander cells was independent of the type of radiation which the donor population received.


Asunto(s)
Bencimidazoles/farmacología , Efecto Espectador , Supervivencia Celular/efectos de la radiación , Protectores contra Radiación/farmacología , Línea Celular Tumoral , Humanos
8.
Mol Oncol ; 17(6): 1129-1147, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36694344

RESUMEN

The use of mutation analysis of homologous recombination repair (HRR) genes to estimate PARP-inhibition response may miss a larger proportion of responding patients. Here, we provide preclinical models for castration-resistant prostate cancer (CRPC) that can be used to functionally predict HRR defects. In vitro, CRPC LNCaP sublines revealed an HRR defect and enhanced sensitivity to olaparib and cisplatin due to impaired RAD51 expression and recruitment. Ex vivo-induced castration-resistant tumor slice cultures or tumor slice cultures derived directly from CRPC patients showed increased olaparib- or cisplatin-associated enhancement of residual radiation-induced γH2AX/53BP1 foci. We established patient-derived tumor organoids (PDOs) from CRPC patients. These PDOs are morphologically similar to their primary tumors and genetically clustered with prostate cancer but not with normal prostate or other tumor entities. Using these PDOs, we functionally confirmed the enhanced sensitivity of CRPC patients to olaparib and cisplatin. Moreover, olaparib but not cisplatin significantly decreased the migration rate in CRPC cells. Collectively, we present robust patient-derived preclinical models for CRPC that recapitulate the features of their primary tumors and enable individualized drug screening, allowing translation of treatment sensitivities into tailored clinical therapy recommendations.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Reparación del ADN por Recombinación , Reparación del ADN/genética , Cisplatino/farmacología , Cisplatino/uso terapéutico
9.
Nucleic Acids Res ; 38(2): 477-87, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906707

RESUMEN

The cohesin protein complex holds sister chromatids together after synthesis until mitosis. It also contributes to post-replicative DNA repair in yeast and higher eukaryotes and accumulates at sites of laser-induced damage in human cells. Our goal was to determine whether the cohesin subunits SMC1 and Rad21 contribute to DNA double-strand break repair in X-irradiated human cells in the G2 phase of the cell cycle. RNA interference-mediated depletion of SMC1 sensitized HeLa cells to X-rays. Repair of radiation-induced DNA double-strand breaks, measured by gammaH2AX/53BP1 foci analysis, was slower in SMC1- or Rad21-depleted cells than in controls in G2 but not in G1. Inhibition of the DNA damage kinase DNA-PK, but not ATM, further inhibited foci loss in cohesin-depleted cells in G2. SMC1 depletion had no effect on DNA single-strand break repair in either G1 or late S/G2. Rad21 and SMC1 were recruited to sites of X-ray-induced DNA damage in G2-phase cells, but not in G1, and only when DNA damage was concentrated in subnuclear stripes, generated by partially shielded ultrasoft X-rays. Our results suggest that the cohesin complex contributes to cell survival by promoting the repair of radiation-induced DNA double-strand breaks in G2-phase cells in an ATM-dependent pathway.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Cromatina/efectos de la radiación , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Aberraciones Cromosómicas , Roturas del ADN de Cadena Simple , Replicación del ADN , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Fase G2 , Células HeLa , Humanos , Mitosis , Proteínas Nucleares/antagonistas & inhibidores , Fosfoproteínas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Interferencia de ARN , Tolerancia a Radiación , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Cohesinas
10.
Mutat Res Rev Mutat Res ; 787: 108346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34083038

RESUMEN

DNA replication stress is a major source of DNA damage, including double-stranded breaks that promote DNA damage response (DDR) signaling. Inefficient repair of such lesions can affect genome integrity. During DNA replication different factors act on chromatin remodeling in a coordinated way. While recent studies have highlighted individual molecular mechanisms of interaction, less is known about the orchestration of chromatin changes under replication stress. In this review we attempt to explore the complex relationship between DNA replication stress, DDR and genome integrity in mammalian cells, taking into account the role of chromatin disposition as an important modulator of DNA repair. Recent data on chromatin restoration and epigenetic re-establishment after DNA replication stress are reviewed.


Asunto(s)
Daño del ADN/fisiología , Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Animales , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Daño del ADN/genética , Replicación del ADN/genética , Inestabilidad Genómica/genética , Humanos
11.
Trends Cancer ; 6(11): 974-984, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32517958

RESUMEN

Molecular-targeted therapies and treatment stratification based on molecular biomarkers have rapidly gained momentum in the therapeutic spectrum for patients with prostate cancer, particularly those with aggressive disease. DNA damage repair (DDR) pathways are commonly impaired in prostate cancer. Recent studies have detailed mechanisms interconnecting the DDR with the androgen receptor (AR) signaling pathway as well as its interplay with the immune response. The prominent role of DDR deficiency in prostate cancer development and treatment response encourages innovative strategies for the detection of DDR deficiency in individual tumors. In this review, we describe recent preclinical and early clinical data on the exploitation of DDR defects as predictive biomarkers and also as molecular therapeutic targets.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/genética , Quimioradioterapia/métodos , Reparación del ADN/efectos de los fármacos , Neoplasias de la Próstata/genética , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Reparación del ADN/efectos de la radiación , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Masculino , Terapia Molecular Dirigida/métodos , Mutación , Pronóstico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Resultado del Tratamiento
12.
Cancer Lett ; 493: 179-188, 2020 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-32891715

RESUMEN

Trifluridine/tipiracil (FTD/TPI; marketed as Lonsurf®) has shown clinically relevant activity after fluoropyrimidine failure in colorectal cancer and may thus be of increased efficacy compared with current standard capecitabine chemoradiation. Here we investigated the colorectal cancer cell lines HT29, HCT116, SW48 and Caco-2 to provide a preclinical rationale for FTD/TPI-based chemoradiation treatment. All lines incorporated similar amounts of FTD, irrespective of treatment concentration and duration, then arrested in S phase, showed persistent γH2AX induction and eventually underwent endoreplication, resulting in polyploidy. Clonogenic assays performed for four combined treatment schedules demonstrated additivity for treatments given within 6 h of each other. However, 24 h FTD/TPI treatment prior to irradiation caused 1.6-2.4 fold radiosensitisation. Combined in vivo treatment was well tolerated and caused a marked tumour growth delay, similar to capecitabine radiochemotherapy regimes. Prolonged S phase arrest, persistent γH2AX signalling, endoreplication and polyploidy may contribute to the cytotoxicity of FTD/TPI. The strong radiosensitising effect observed in vitro after prolonged treatment with FTD/TPI and equivalence with capecitabine-based chemoradiation in vivo support a daily fractionated combined regime of FTD/TPI and radiation in rectal cancer treatment. This is now being tested in a phase I/II clinical trial (NCT04177602).


Asunto(s)
Neoplasias Colorrectales/terapia , Histonas/metabolismo , Pirrolidinas/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Timina/administración & dosificación , Trifluridina/administración & dosificación , Animales , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Quimioradioterapia , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Combinación de Medicamentos , Endorreduplicación , Femenino , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones , Poliploidía , Pirrolidinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Timina/farmacología , Trifluridina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancers (Basel) ; 12(9)2020 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-32878283

RESUMEN

(1) Background: The combination of the first-generation antiandrogens and radiotherapy (RT) has been studied extensively in the clinical setting of prostate cancer (PCa). Here, we evaluated the potential radiosensitizing effect of the second-generation antiandrogens abiraterone acetate, apalutamide and enzalutamide. (2) Methods: Cell proliferation and agarose-colony forming assay were used to measure the effect on survival. Double strand break repair efficiency was monitored using immunofluorescence staining of γH2AX/53BP1. (3) Results: We report retrospectively a minor benefit for PCa patients received first-generation androgen blockers and RT compared to patients treated with RT alone. Combining either of the second-generation antiandrogens and 2Gy suppressed cell growth and increased doubling time significantly more than 2Gy alone, in both hormone-responsive LNCaP and castration-resistant C4-2B cells. These findings were recapitulated in resistant sub-clones to (i) hormone ablation (LNCaP-abl), (ii) abiraterone acetate (LNCaP-abi), (iii) apalutamide (LNCaP-ARN509), (iv) enzalutamide (C4-2B-ENZA), and in castration-resistant 22-RV1 cells. This radiosensitization effect was not observable using the first-generation antiandrogen bicalutamide. Inhibition of DNA DSB repair was found to contribute to the radiosensitization effect of second-generation antiandrogens, as demonstrated by a significant increase in residual γH2AX and 53BP1 foci numbers at 24h post-IR. DSB repair inhibition was further demonstrated in 22 patient-derived tumor slice cultures treated with abiraterone acetate before ex-vivo irradiation with 2Gy. (4) Conclusion: Together, these data show that second-generation antiandrogens can enhance radiosensitivity in PCa through DSB repair inhibition, regardless of their hormonal status. Translated into clinical practice, our results may help to find additional strategies to improve the effectiveness of RT in localized PCa, paving the way for a clinical trial.

14.
Mutat Res ; 672(2): 82-9, 2009 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-19049903

RESUMEN

Considerable controversy still exists as to whether electric and magnetic fields (MF) at extremely low frequencies are genotoxic to humans. The aim of this study was to test the ability of alternating magnetic fields to induce DNA and chromosomal damage in primary human fibroblasts. Single- and double-strand breaks were quantified using the alkaline comet assay and the gammaH2AX-foci assay, respectively. Chromosomal damage was assayed for unstable aberrations, sister chromatid exchange and micronuclei. Cells were exposed to switching fields - 5min on, 10min off - for 15h over the range 50-1000microT. Exposure to ionizing radiation was used as a positive-effect calibration. In this study two separate MF exposure systems were used. One was based on a custom-built solenoid coil system and the other on a commercial system almost identical to that used in previous studies by the EU REFLEX programme. With neither system could DNA damage or chromosomal damage be detected as a result of exposure of fibroblasts to switching MF. The sensitive gammaH2AX assay could also not detect significant DNA damage in the MF-exposed fibroblasts, although the minimum threshold for this assay was equivalent to an X-ray dose of 0.025Gy. Therefore, with comparable MF parameters employed, this study could not confirm previous studies reporting significant effects for both the alkaline and neutral comet assays and chromosomal aberration induction.


Asunto(s)
Aberraciones Cromosómicas/efectos de la radiación , Daño del ADN/efectos de la radiación , Campos Electromagnéticos/efectos adversos , Línea Celular , Ensayo Cometa , Humanos
15.
Cancer Res ; 67(12): 5872-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575156

RESUMEN

The accepted paradigm for radiation effects is that direct DNA damage via energy deposition is required to trigger the downstream biological consequences. The radiation-induced bystander effect is the ability of directly irradiated cells to interact with their nonirradiated neighbors, which can then show responses similar to those of the targeted cells. p53 binding protein 1 (53BP1) forms foci at DNA double-strand break sites and is an important sensor of DNA damage. This study used an ionizing radiation microbeam approach that allowed us to irradiate specifically the nucleus or cytoplasm of a cell and quantify response in irradiated and bystander cells by studying ionizing radiation-induced foci (IRIF) formation of 53BP1 protein. Our results show that targeting only the cytoplasm of a cell is capable of eliciting 53BP1 foci in both hit and bystander cells, independently of the dose or the number of cells targeted. Therefore, direct DNA damage is not required to trigger 53BP1 IRIF. The use of common reactive oxygen species and reactive nitrogen species (RNS) inhibitors prevent the formation of 53BP1 foci in hit and bystander cells. Treatment with filipin to disrupt membrane-dependent signaling does not prevent the cytoplasmic irradiation-induced 53BP1 foci in the irradiated cells, but it does prevent signaling to bystander cells. Active mitochondrial function is required for these responses because pseudo-rho(0) cells, which lack mitochondrial DNA, could not produce a bystander signal, although they could respond to a signal from normal rho+ cells.


Asunto(s)
Citoplasma/efectos de la radiación , Daño del ADN/efectos de la radiación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/efectos de la radiación , Mitocondrias/efectos de la radiación , Núcleo Celular/efectos de la radiación , ADN Mitocondrial/efectos de la radiación , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Mitocondrias/metabolismo , Reacción en Cadena de la Polimerasa , Transporte de Proteínas/efectos de la radiación , Radiación Ionizante , Proteína 1 de Unión al Supresor Tumoral P53
16.
Exp Mol Pathol ; 85(3): 189-95, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18775423

RESUMEN

In studying immune responses towards the poliovirus, data about T cell mediated immunity in the intestine as the main portal of viral entry in disease and vaccination is lacking. We treated two macaques with oral Polio vaccine and collected duodenal and colonic biopsy specimens. RNA isolation, reverse transcription, and polymerase chain reaction were performed for fragment analysis of the complementarity determining region 3 (CDR3) of the T cell receptor beta chain variable region (TCRBV), followed by subcloning and sequencing of expanded bands. In the colon, oligoclonal expansions of TCRBV16+ or TCRBV13+ intestinal T cells with conserved motifs of the hypervariable CDR3 were found. Flow cytometric analysis of mucosal T cells revealed that activated colonic T cells were mainly CD4(+). Our results indicate that there is a local activation of oligoclonal T cells in the colon after oral Polio vaccination (OPV) which involves selected TCRBV families and may occur within the CD4(+) T cell subset.


Asunto(s)
Colon/citología , Colon/inmunología , Macaca mulatta/inmunología , Vacuna Antipolio Oral/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Clonales , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/inmunología , Duodeno/citología , Duodeno/inmunología , Activación de Linfocitos/inmunología , Masculino , Datos de Secuencia Molecular
17.
Mutat Res Rev Mutat Res ; 778: 13-22, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30454679

RESUMEN

This review summarises our current understanding of the radiation-induced bystander/systemic effect (RIBE) as well as other types of intercellular reactions induced by malignant tumours, chemotherapy, photodynamic stress and the microbiome. On the basis of striking similarities between these different types of responses RIBE is proposed as a prototype model of a unifying genotoxic stress response system. The early bystander response is initiated by a mitochondria-dependent increase of reactive oxygen species (ROS) and triggers a complex intercellular signalling cascade leading to a sustained increase of cellular DNA damage. The susceptibility to this DNA damage-inducing signal depends on the genetic make-up of the recipient cell population where ATR/ATM- and FA/BRCA-dependent DNA damage response pathways are key players. Long distance bystander/systemic effects observed in in vivo-models are sustained by macrophage-mediated inflammation. Of clinical importance is the potential contribution of bystander DNA damage to an increased risk of malignancies. Defects in DNA damage repair pathways are frequently observed in tumours, which may affect their susceptibility to bystander DNA damage. The potential role for molecular targeted inhibitors in the therapeutic exploitation of bystander responses as well as their differential modulation of targeted and non-targeted effects is also discussed in this context.


Asunto(s)
Efecto Espectador/efectos de la radiación , Daño del ADN/efectos de la radiación , ADN/efectos de la radiación , Radiación , Daño del ADN/genética , Humanos , Macrófagos/efectos de la radiación , Especies Reactivas de Oxígeno/efectos de la radiación
18.
Mol Cancer Res ; 16(3): 496-507, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29330294

RESUMEN

Clinical options for systemic therapy of neuroendocrine tumors (NET) are limited. Development of new drugs requires suitable representative in vitro and in vivo model systems. So far, the unavailability of a human model with a well-differentiated phenotype and typical growth characteristics has impaired preclinical research in NET. Herein, we establish and characterize a lymph node-derived cell line (NT-3) from a male patient with well-differentiated pancreatic NET. Neuroendocrine differentiation and tumor biology was compared with existing NET cell lines BON and QGP-1. In vivo growth was assessed in a xenograft mouse model. The neuroendocrine identity of NT-3 was verified by expression of multiple NET-specific markers, which were highly expressed in NT-3 compared with BON and QGP-1. In addition, NT-3 expressed and secreted insulin. Until now, this well-differentiated phenotype is stable since 58 passages. The proliferative labeling index, measured by Ki-67, of 14.6% ± 1.0% in NT-3 is akin to the original tumor (15%-20%), and was lower than in BON (80.6% ± 3.3%) and QGP-1 (82.6% ± 1.0%). NT-3 highly expressed somatostatin receptors (SSTRs: 1, 2, 3, and 5). Upon subcutaneous transplantation of NT-3 cells, recipient mice developed tumors with an efficient tumor take rate (94%) and growth rate (139% ± 13%) by 4 weeks. Importantly, morphology and neuroendocrine marker expression of xenograft tumors resembled the original human tumor.Implications: High expression of somatostatin receptors and a well-differentiated phenotype as well as a slow growth rate qualify the new cell line as a relevant model to study neuroendocrine tumor biology and to develop new tumor treatments. Mol Cancer Res; 16(3); 496-507. ©2018 AACR.


Asunto(s)
Modelos Animales de Enfermedad , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Técnicas de Genotipaje/métodos , Xenoinjertos , Humanos , Masculino , Ratones , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética
20.
Ann Transl Med ; 4(24): 523, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28151534

RESUMEN

Despite considerable progress in (I) our understanding of the aetiopathology of head and neck cancer and (II) the precise delivery of radiotherapy, long-term survival rates for many patients with head and neck cancer remain disappointingly low. Over the past years, gold nanoparticles (NP) have emerged as promising radiation dose enhancers. In a recent study published in Nanoscale, Popovtzer et al. have used gold NP coated with an antibody against the epidermal growth factor receptor (EGFR) in an attempt to enhance radiation-induced tumour cell killing in a head and neck cancer xenograft model. They report a significant impact of the combined treatment with radiation and gold NP on tumour growth and suggest an involvement of apoptosis, inhibition of angiogenesis and diminished tissue repair. In this perspective, we illustrate the underlying radiobiophysical concepts and discuss some of the challenges associated with this and related nanoparticle-radiotherapy studies from a physics, chemistry, biology and therapy angle. We conclude that strong interdisciplinary collaborations spanning all these areas are crucially important to proceed towards effective cancer treatment with gold NP "from bench to bedside".

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA