Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pediatr Blood Cancer ; 63(9): 1522-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27149120

RESUMEN

BACKGROUND: Compared to X-ray radiation therapy, proton therapy (PT) reduces the radiation dose to organs at risk, which is expected to translate into fewer second cancers and less cardiac morbidity decades after treatment. The Children's Oncology Group high-risk pediatric Hodgkin lymphoma (PHL) protocol, AHOD1331, allows the use of PT, yet limited data exist on the use of PT in PHL. PROCEDURE: Between 2010 and 2014, 22 pediatric patients were treated with PT for PHL at our institution: 7 intermediate-risk patients, 11 high-risk patients, and 4 relapsed patients. The patients' age ranged from 6 to 18 years old. Median follow-up was 36 months. All patients received chemotherapy before PT. RESULTS: The 2-year and 3-year overall survival rates were both 94%, and the progression-free survival rate was 86%. Recurrences occurred in three high-risk patients: one isolated in-field cervical lymph node and two in-field and out-of-field. All recurrences occurred within 5 months of completing PT. No PT-related grade 3 or higher acute or late complications were observed. CONCLUSION: PT for PHL showed no short-term severe toxicity and yields similar short-term control to recently published large multi-institutional clinical trials.


Asunto(s)
Enfermedad de Hodgkin/radioterapia , Terapia de Protones , Adolescente , Niño , Femenino , Enfermedad de Hodgkin/diagnóstico por imagen , Enfermedad de Hodgkin/mortalidad , Humanos , Masculino , Terapia de Protones/efectos adversos , Planificación de la Radioterapia Asistida por Computador , Recurrencia , Tomografía Computarizada por Rayos X
2.
Eur Arch Otorhinolaryngol ; 273(3): 661-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25773487

RESUMEN

We evaluated the treatment of squamous cell carcinoma (SCC) of the nasal vestibule. Eighty-six patients were treated with radiotherapy (RT) and 13 patients received surgery and RT. The mean follow-up was: 9.7 years (range 4 months-35.9 years). The 5- and 10-year outcomes were: local control (LC), 88 and 82 %; local-regional control (LRC), 78 and 73 %; freedom from distant metastases (FFDM), 96 and 96 %; cause-specific survival (CSS), 91 and 86 %; and overall survival, 75 and 51 %. The 5- and 10-year LC rates for patients treated with RT were 94 and 89 % overall. A multivariate analysis was performed. Tumor size predicted LC, LRC, OS, and CSS. Overall stage predicted LRC. RT cures most patients with T1-T2 and favorable T4 SCCs with acceptable toxicity. RT and surgery result in improved likelihood of cure for patients with advanced T4 lesions.


Asunto(s)
Carcinoma de Células Escamosas , Cavidad Nasal/patología , Procedimientos Quírurgicos Nasales/métodos , Neoplasias Nasales , Radioterapia/métodos , Adulto , Anciano , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Neoplasias Nasales/patología , Neoplasias Nasales/radioterapia , Análisis de Supervivencia , Resultado del Tratamiento
3.
Blood ; 121(21): 4359-65, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23568489

RESUMEN

Chromosomal translocations are common contributors to malignancy, yet little is known about the precise molecular mechanisms by which they are generated. Sequencing translocation junctions in acute leukemias revealed that the translocations were likely mediated by a DNA double-strand break repair pathway termed nonhomologous end-joining (NHEJ). There are major 2 types of NHEJ: (1) the classical pathway initiated by the Ku complex, and (2) the alternative pathway initiated by poly ADP-ribose polymerase 1 (PARP1). Recent reports suggest that classical NHEJ repair components repress translocations, whereas alternative NHEJ components were required for translocations. The rate-limiting step for initiation of alternative NHEJ is the displacement of the Ku complex by PARP1. Therefore, we asked whether PARP1 inhibition could prevent chromosomal translocations in 3 translocation reporter systems. We found that 2 PARP1 inhibitors or repression of PARP1 protein expression strongly repressed chromosomal translocations, implying that PARP1 is essential for this process. Finally, PARP1 inhibition also reduced both ionizing radiation-generated and VP16-generated translocations in 2 cell lines. These data define PARP1 as a critical mediator of chromosomal translocations and raise the possibility that oncogenic translocations occurring after high-dose chemotherapy or radiation could be prevented by treatment with a clinically available PARP1 inhibitor.


Asunto(s)
Leucemia/genética , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/fisiología , Translocación Genética/genética , Translocación Genética/fisiología , Enfermedad Aguda , Células Cultivadas , Roturas del ADN de Doble Cadena , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Indoles/farmacología , Leucemia/tratamiento farmacológico , Leucemia/prevención & control , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , ARN Interferente Pequeño/genética , Translocación Genética/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 108(2): 540-5, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187428

RESUMEN

Given its significant role in the maintenance of genomic stability, histone methylation has been postulated to regulate DNA repair. Histone methylation mediates localization of 53BP1 to a DNA double-strand break (DSB) during homologous recombination repair, but a role in DSB repair by nonhomologous end-joining (NHEJ) has not been defined. By screening for histone methylation after DSB induction by ionizing radiation we found that generation of dimethyl histone H3 lysine 36 (H3K36me2) was the major event. Using a novel human cell system that rapidly generates a single defined DSB in the vast majority of cells, we found that the DNA repair protein Metnase (also SETMAR), which has a SET histone methylase domain, localized to an induced DSB and directly mediated the formation of H3K36me2 near the induced DSB. This dimethylation of H3K36 improved the association of early DNA repair components, including NBS1 and Ku70, with the induced DSB, and enhanced DSB repair. In addition, expression of JHDM1a (an H3K36me2 demethylase) or histone H3 in which K36 was mutated to A36 or R36 to prevent H3K36me2 formation decreased the association of early NHEJ repair components with an induced DSB and decreased DSB repair. Thus, these experiments define a histone methylation event that enhances DNA DSB repair by NHEJ.


Asunto(s)
Reparación del ADN , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Lisina/química , Antígenos Nucleares/química , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Metilación de ADN , Enzimas de Restricción del ADN/farmacología , Proteínas de Unión al ADN/química , Desoxirribonucleasas de Localización Especificada Tipo II/farmacología , Dimerización , N-Metiltransferasa de Histona-Lisina/química , Humanos , Autoantígeno Ku , Modelos Teóricos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Saccharomyces cerevisiae/farmacología , Factores de Tiempo
5.
Nucleic Acids Res ; 38(17): 5681-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20457750

RESUMEN

Metnase is a human protein with methylase (SET) and nuclease domains that is widely expressed, especially in proliferating tissues. Metnase promotes non-homologous end-joining (NHEJ), and knockdown causes mild hypersensitivity to ionizing radiation. Metnase also promotes plasmid and viral DNA integration, and topoisomerase IIα (TopoIIα)-dependent chromosome decatenation. NHEJ factors have been implicated in the replication stress response, and TopoIIα has been proposed to relax positive supercoils in front of replication forks. Here we show that Metnase promotes cell proliferation, but it does not alter cell cycle distributions, or replication fork progression. However, Metnase knockdown sensitizes cells to replication stress and confers a marked defect in restart of stalled replication forks. Metnase promotes resolution of phosphorylated histone H2AX, a marker of DNA double-strand breaks at collapsed forks, and it co-immunoprecipitates with PCNA and RAD9, a member of the PCNA-like RAD9-HUS1-RAD1 intra-S checkpoint complex. Metnase also promotes TopoIIα-mediated relaxation of positively supercoiled DNA. Metnase is not required for RAD51 focus formation after replication stress, but Metnase knockdown cells show increased RAD51 foci in the presence or absence of replication stress. These results establish Metnase as a key factor that promotes restart of stalled replication forks, and implicate Metnase in the repair of collapsed forks.


Asunto(s)
Reparación del ADN , Replicación del ADN , N-Metiltransferasa de Histona-Lisina/fisiología , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/aislamiento & purificación , Proliferación Celular , Supervivencia Celular , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Superhelicoidal/metabolismo , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/aislamiento & purificación , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Inmunoprecipitación , Antígeno Nuclear de Célula en Proliferación/aislamiento & purificación , Recombinasa Rad51/análisis
6.
Blood ; 114(9): 1852-8, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19458360

RESUMEN

After DNA replication, sister chromatids must be untangled, or decatenated, before mitosis so that chromatids do not tear during anaphase. Topoisomerase IIalpha (Topo IIalpha) is the major decatenating enzyme. Topo IIalpha inhibitors prevent decatenation, causing cells to arrest during mitosis. Here we report that acute myeloid leukemia cells fail to arrest at the mitotic decatenation checkpoint, and their progression through this checkpoint is regulated by the DNA repair component Metnase (also termed SETMAR). Metnase contains a SET histone methylase and transposase nuclease domain, and is a component of the nonhomologous end-joining DNA double-strand break repair pathway. Metnase interacts with Topo IIalpha and enhances its decatenation activity. Here we show that multiple types of acute leukemia cells have an attenuated mitotic arrest when decatenation is inhibited and that in an acute myeloid leukemia (AML) cell line this is mediated by Metnase. Of further importance, Metnase permits continued proliferation of these AML cells even in the presence of the clinical Topo IIalpha inhibitor VP-16. In vitro, purified Metnase prevents VP-16 inhibition of Topo IIalpha decatenation of tangled DNA. Thus, Metnase expression levels may predict AML resistance to Topo IIalpha inhibitors, and Metnase is a potential therapeutic target for small molecule interference.


Asunto(s)
Cromosomas/ultraestructura , N-Metiltransferasa de Histona-Lisina/fisiología , Leucemia Mieloide Aguda/genética , Antígenos de Neoplasias/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , ADN/efectos de los fármacos , Daño del ADN , Reparación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Etopósido/farmacología , Regulación Leucémica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Mitosis , Modelos Biológicos
7.
Nucleic Acids Res ; 36(18): 5822-31, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18790802

RESUMEN

Metnase is a human SET and transposase domain protein that methylates histone H3 and promotes DNA double-strand break repair. We now show that Metnase physically interacts and co-localizes with Topoisomerase IIalpha (Topo IIalpha), the key chromosome decatenating enzyme. Metnase promotes progression through decatenation and increases resistance to the Topo IIalpha inhibitors ICRF-193 and VP-16. Purified Metnase greatly enhanced Topo IIalpha decatenation of kinetoplast DNA to relaxed circular forms. Nuclear extracts containing Metnase decatenated kDNA more rapidly than those without Metnase, and neutralizing anti-sera against Metnase reversed that enhancement of decatenation. Metnase automethylates at K485, and the presence of a methyl donor blocked the enhancement of Topo IIalpha decatenation by Metnase, implying an internal regulatory inhibition. Thus, Metnase enhances Topo IIalpha decatenation, and this activity is repressed by automethylation. These results suggest that cancer cells could subvert Metnase to mediate clinically relevant resistance to Topo IIalpha inhibitors.


Asunto(s)
Antígenos de Neoplasias/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Encadenado/metabolismo , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Línea Celular , Cromosomas Humanos/metabolismo , ADN de Cinetoplasto/metabolismo , Humanos , Metafase , Metilación
8.
DNA Repair (Amst) ; 7(12): 1927-37, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18773976

RESUMEN

Transposase domain proteins mediate DNA movement from one location in the genome to another in lower organisms. However, in human cells such DNA mobility would be deleterious, and therefore the vast majority of transposase-related sequences in humans are pseudogenes. We recently isolated and characterized a SET and transposase domain protein termed Metnase that promotes DNA double-strand break (DSB) repair by non-homologous end-joining (NHEJ). Both the SET and transposase domain were required for its NHEJ activity. In this study we found that Metnase interacts with DNA Ligase IV, an important component of the classical NHEJ pathway. We investigated whether Metnase had structural requirements of the free DNA ends for NHEJ repair, and found that Metnase assists in joining all types of free DNA ends equally well. Metnase also prevents long deletions from processing of the free DNA ends, and improves the accuracy of NHEJ. Metnase levels correlate with the speed of disappearance of gamma-H2Ax sites after ionizing radiation. However, Metnase has little effect on homologous recombination repair of a single DSB. Altogether, these results fit a model where Metnase plays a role in the fate of free DNA ends during NHEJ repair of DSBs.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , ADN Ligasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Recombinación Genética , Factores de Transcripción/metabolismo , Transposasas/metabolismo , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/fisiología , Daño del ADN/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/genética , Reparación del ADN/fisiología , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN , Técnica del Anticuerpo Fluorescente , Chaperonas de Histonas , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Humanos , Inmunoprecipitación , Rayos Infrarrojos , Riñón/metabolismo , Factores de Transcripción/genética , Transposasas/genética
9.
Curr Opin Hematol ; 15(4): 338-45, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18536572

RESUMEN

PURPOSE OF REVIEW: This review highlights recent findings about the known DNA repair machinery, its impact on chromosomal translocation mechanisms and their relevance to leukemia in the clinic. RECENT FINDINGS: Chromosomal translocations regulate the behavior of leukemia. They not only predict outcome but they define therapy. There is a great deal of knowledge on the products of leukemic translocations, yet little is known about the mechanism by which those translocations occur. Given the large number of DNA double-strand breaks that occur during normal progression through the cell cycle, especially from V(D)J recombination, stalled replication forks or failed decatenation, it is surprising that leukemogenic translocations do not occur more frequently. Fortunately, hematopoietic cells have sophisticated repair mechanisms to suppress such translocations. When these defenses fail leukemia becomes far more common, as seen in inherited deficiencies of DNA repair. Analyzing translocation sequences in cellular and animal models, and in human leukemias, has yielded new insights into the mechanisms of leukemogenic translocations. SUMMARY: New data from animal models suggest a two hit origin of leukemic translocations, where there must be both a defect in DNA double-strand break repair and a subsequent failure of cell cycle arrest for leukemogenesis.


Asunto(s)
Leucemia/genética , Translocación Genética , Ciclo Celular , Reparación del ADN/genética , Humanos , Leucemia/etiología
10.
Mol Cell Biol ; 25(5): 1949-57, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15713648

RESUMEN

Homologous recombinational repair (HRR) of DNA damage is critical for maintaining genome stability and tumor suppression. RAD51 and BRCA2 colocalization in nuclear foci is a hallmark of HRR. BRCA2 has important roles in RAD51 focus formation and HRR of DNA double-strand breaks (DSBs). We previously reported that BCCIPalpha interacts with BRCA2. We show that a second isoform, BCCIPbeta, also interacts with BRCA2 and that this interaction occurs in a region shared by BCCIPalpha and BCCIPbeta. We further show that chromatin-bound BRCA2 colocalizes with BCCIP nuclear foci and that most radiation-induced RAD51 foci colocalize with BCCIP. Reducing BCCIPalpha by 90% or BCCIPbeta by 50% by RNA interference markedly reduces RAD51 and BRCA2 foci and reduces HRR of DSBs by 20- to 100-fold. Similarly, reducing BRCA2 by 50% reduces RAD51 and BCCIP foci. These data indicate that BCCIP is critical for BRCA2- and RAD51-dependent responses to DNA damage and HRR.


Asunto(s)
Proteína BRCA2/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Recombinación Genética/fisiología , Proteína BRCA2/análisis , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Daño del ADN , Proteínas de Unión al ADN/análisis , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/análisis , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Recombinasa Rad51
11.
Oxf Med Case Reports ; 2018(1): omx078, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29340160

RESUMEN

Castration-resistant prostate cancer is an incurable disease. To date, six agents-abiraterone, enzalutamide, docetaxel, cabazitaxel, radium-223 and sipuleucel-T- have shown clinical efficacy in phase III clinical trials, leading to their FDA approval. Patients are typically sequenced through most or all of these agents, and then eventually succumb to their disease. Development of new treatments remains an unmet need. We report a case of a patient who progressed on enzalutamide with a single enlarging metastatic lesion, was treated with ablative stereotactic body radiation therapy while maintaining the same systemic treatment, who then had durable complete remission. Our findings have important clinical implications and suggest novel clinical trials for this difficult to treat disease.

12.
Am J Clin Oncol ; 41(12): 1216-1219, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29746367

RESUMEN

OBJECTIVE: Patients with mucosal squamous cell carcinoma (SCC) of the head and neck almost always have a primary site in the base of tongue or tonsillar fossa. Lingual tonsillectomy has recently been advocated as part of the diagnostic evaluation as opposed to directed biopsies of the base of tongue and thought to possibly result in an increased likelihood or cure. The purpose of this project is to determine whether this is probable. MATERIALS AND METHODS: We reviewed the medical records of patients treated with primary radiotherapy (RT) between January 1983 and March 2013. The outcomes were compared following RT in consecutively treated patients with either T1-2 base of tongue or unknown primary (cancer of unknown primary) SCC with predominantly level 2 adenopathy. RESULTS: At 10 years, there were no clinically significant differences in the 2 groups, in local control, regional control, freedom from distant metastases, disease-specific, or cause-specific survival. Overall survival at 10 years was improved with T1-2 base of tongue cancers but not for those with T0 N3 disease. The reasons for this are unclear. CONCLUSIONS: Tongue base biopsy (or lingual tonsillectomy) likely increases the probability of identifying an unknown primary in the base of tongue, but it does not improve outcome following RT for patients with cancer of unknown primary SCC with predominantly level 2 adenopathy.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Neoplasias Primarias Desconocidas/patología , Tonsila Palatina/patología , Neoplasias Tonsilares/patología , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Carcinoma de Células Escamosas/cirugía , Femenino , Estudios de Seguimiento , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Primarias Desconocidas/cirugía , Tonsila Palatina/cirugía , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Neoplasias Tonsilares/cirugía , Tonsilectomía
13.
Oncol Lett ; 13(3): 1087-1094, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28454218

RESUMEN

Chemotherapy and targeted therapies are effective palliative options for numerous unresectable or metastatic cancers. However, treatment resistance inevitably develops leading to mortality. In a subset of patients, systemic therapy appears to control the majority of tumors leaving 5 or less to progress, a phenomenon described as oligoprogression. Reasoning that the majority of lesions remain responsive to ongoing systemic chemotherapy, we hypothesized that local treatment of the progressing lesions would confer a benefit. The present study describes the cases of 5 patients whose metastatic disease was largely controlled by chemotherapy. The oligoprogressive lesions (≤5) were treated with stereotactic body radiotherapy (SBRT), justifying continued use of an effective systemic regimen. A total of 5 patients with metastatic disease on chemotherapy, with ≤5 progressing lesions amenable to SBRT, were treated with ablative intent. Primary tumor site and histology were as follows: 2 with metastatic colon adenocarcinoma, 2 with metastatic rectal adenocarcinoma and 1 with metastatic pancreatic adenocarcinoma. Imaging was performed prior to SBRT and every 3 months after SBRT. In total, 4 out of the 5 patients achieved disease control for >7 months with SBRT, without changing chemotherapy regimen. The median time to chemotherapy change was 9 months, with a median follow-up time of 9 months. The patient who failed to respond developed progressive disease outside of the SBRT field at 3 months. In conclusion, the addition of SBRT to chemotherapy is an option for the overall systemic control of oligoprogressive disease.

14.
Radiat Oncol ; 10: 199, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26391010

RESUMEN

BACKGROUND: In patients at high risk for regional node metastasis from squamous cell carcinoma (SCC) of the skin of the face, ear, or scalp, radiotherapy to the regional nodes is an alternative to parotid or neck surgery. Data on the efficacy of elective nodal radiotherapy in this setting are scarce such that there is no publication specifically addressing the subject. The purpose of our study is to fill this void in the skin cancer literature. METHODS: This is a single-institution study of outcomes following elective nodal radiotherapy in 71 consecutively treated adults with SCC of the face, ears, or scalp. Primary site stage distribution per the American Joint Committee on Cancer, 7(th) Edition, was as follows: T1, 15 %; T2, 34 %; T3, 1 %; and T4, 50 %. Other disease characteristics included the following: clinical perineural invasion, 13 %; pathological perineural invasion, 78 %; recurrent disease, 32 %; and positive or close margin, 67 %. The median radiation dose to the first- and second-echelon nodal area was 50 Gy. Acute and late toxicity were graded per the Common Terminology Criteria for Adverse Events, version 4.0. Regional control was assessed using the Kaplan-Meier product limit method. RESULTS: Median followup was 4.5 years for all patients. The actuarial regional control rate at 5 years was 96 %. There were no (0 %) grade 3 or higher complications from elective nodal irradiation. CONCLUSIONS: Elective nodal irradiation in patients with high-risk SCC of the face, ears and scalp is safe and effective.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeza y Cuello/radioterapia , Metástasis Linfática/prevención & control , Neoplasias Cutáneas/radioterapia , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Neoplasias del Oído/patología , Neoplasias del Oído/radioterapia , Cara/patología , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Ganglios Linfáticos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Cuero Cabelludo/patología , Neoplasias Cutáneas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello
15.
Ann N Y Acad Sci ; 1310: 89-97, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24528169

RESUMEN

Chromosome translocations are caused by inappropriate religation of two DNA double-strand breaks (DSBs) in heterologous chromosomes. These DSBs can be generated by endogenous or exogenous sources. Endogenous sources of DSBs leading to translocations include inappropriate recombination activating gene (RAG) or activation-induced deaminase (AID) activity during immune receptor maturation. Endogenous DSBs can also occur at noncanonical DNA structures or at collapsed replication forks. Exogenous sources of DSBs leading to translocations include ionizing radiation (IR) and cancer chemotherapy. Spatial proximity of the heterologous chromosomes is also important for translocations. While three distinct pathways for DNA DSB repair exist, mounting evidence supports alternative nonhomologous end joining (aNHEJ) as the predominant pathway through which the majority of translocations occur. Initiated by poly (ADP-ribose) polymerase 1 (PARP1), aNHEJ is utilized less frequently in DNA DSB repair than other forms of DSB repair. We recently found that PARP1 is essential for chromosomal translocations to occur and that small molecule PARP1 inhibitors, already in clinical use, can inhibit translocations generated by IR or topoisomerase II inhibition. These data confirm the central role of PARP1 in aNHEJ-mediated chromosomal translocations and raise the possibility of using clinically available PARP1 inhibitors in patients who are at high risk for secondary oncogenic chromosomal translocations.


Asunto(s)
Transformación Celular Neoplásica/genética , Poli(ADP-Ribosa) Polimerasas/fisiología , Translocación Genética/genética , Animales , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/fisiología , Reparación del ADN/fisiología , Desoxirribonucleasas/genética , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Receptores Inmunológicos/genética
16.
Cell Div ; 9: 1, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024738

RESUMEN

Chk1 both arrests replication forks and enhances repair of DNA damage by phosphorylation of downstream effectors. Metnase (also termed SETMAR) is a SET histone methylase and transposase nuclease protein that promotes both DNA double strand break (DSB) repair and re-start of stalled replication forks. We previously found that Chk1 phosphorylation of Metnase on S495 enhanced its DNA DSB repair activity but decreased its ability to re-start stalled replication forks. Here we show that phosphorylated Metnase feeds back to increase the half-life of Chk1. Chk1 half-life is regulated by DDB1 targeting it to Cul4A for ubiquitination and destruction. Metnase decreases Chk1 interaction with DDB1, and decreases Chk1 ubiquitination. These data define a novel pathway for Chk1 regulation, whereby a target of Chk1, Metnase, feeds back to amplify Chk1 stability, and therefore enhance replication fork arrest.

17.
Prog Mol Biol Transl Sci ; 110: 207-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22749147

RESUMEN

DNA damage occurs continuously as a result of various factors-intracellular metabolism, replication, and exposure to genotoxic agents, such as ionizing radiation and chemotherapy. If left unrepaired, this damage could result in changes or mutations within the cell genomic material. There are a number of different pathways that the cell can utilize to repair these DNA breaks. However, it is of utmost interest to know how the DNA damage is signaled to the various DNA pathways. As DNA damage occurs within the chromatin, we postulate that modifications of histones are important for signaling the position of DNA damage, recruiting the DNA repair proteins to the site of damage, and creating an open structure such that the repair proteins can access the site of damage. We discuss the modifications that occur on the histones and the manner in which they relate to the type of damage that has occurred as well as the DNA repair pathways that are activated.


Asunto(s)
Reparación del ADN/genética , Código de Histonas/genética , Animales , Reparación del ADN por Unión de Extremidades/genética , Recombinación Homóloga/genética , Humanos , Modelos Biológicos , Procesamiento Proteico-Postraduccional/genética
18.
Cancer Genet Cytogenet ; 200(2): 184-90, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20620605

RESUMEN

Chromosomal translocations are common in leukemia, but little is known about their mechanism. Metnase (also termed SETMAR) is a fusion of a histone methylase and transposase protein that arose specifically in primates. Transposases were thought to be extinct in primates because they would mediate deleterious DNA movement. In primates, Metnase interacts with DNA Ligase IV (Lig IV) and promotes nonhomologous end-joining (NHEJ) DNA repair. We show here that the primate-specific protein Metnase can also enhance NHEJ in murine cells and can also interact with murine Lig IV, indicating that it integrated into the preexisting NHEJ pathway after its development in primates. Significantly, expressing Metnase in murine cells significantly reduces chromosomal translocations. We propose that the fusion of the histone methylase SET domain and the transposase domain in the anthropoid lineage to form primate Metnase promotes accurate intrachromosomal NHEJ and thereby suppresses interchromosomal translocations. Metnase may have been selected for because it has a function opposing transposases and may thus play a key role in suppressing translocations that underlie oncogenicity.


Asunto(s)
Proteínas de Unión al ADN/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteínas Recombinantes de Fusión/fisiología , Translocación Genética , Transposasas/fisiología , Animales , ADN Ligasa (ATP) , ADN Ligasas/fisiología , Reparación del ADN , Ratones , Células 3T3 NIH
19.
Cell Cycle ; 8(18): 3019-24, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19713748

RESUMEN

The p21 (CDKN1A, Waf1 or Cip1) protein is widely known as an inhibitor of cyclin-dependent kinase (CDK), which plays a critical role in regulation of the G(1)-S transition during the cell cycle progression. The inhibition of G(1)-S transition by p21 is mainly mediated in the nucleus. However, the cytoplasmic p21 has been shown to play a pro-proliferation and anti-apoptosis role. Thus, the regulation of p21's intracellular distribution has a significant implication for cell fate determination. BCCIP is a BRCA2 and CDKN1A Interacting Protein. Previous reports showed that BCCIP enhances the p21 suppression activity towards CDK2, and BCCIP downregulation reduces p21 expression by abrogating p53 transcription activity. In this report, we demonstrate that the BCCIP-p21 interaction is enhanced in response to DNA damage using Fluorescent Resonance Energy Transfer (FRET) technique. We found that the downregulation of BCCIP reduces nuclear p21 and increases cytoplasmic p21. This p21 redistribution is not caused by the reduced expression of endogenous p21 resulting from BCCIP downregulation, because exogenously expressed p21 also preferably distributes in the cytoplasm. The BCCIP regulation of p21 distribution is not related to the status of Thr-145 phosphorylation that is known to cause cytoplasmic distribution. These data suggest that regulation of p21 intracellular distribution as a new mechanism for BCCIP to modulate p21 functions.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas de Unión al Calcio/fisiología , Proteínas de Ciclo Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Daño del ADN , Transferencia Resonante de Energía de Fluorescencia , Humanos , Proteínas Nucleares/genética , Fosforilación , ARN Interferente Pequeño/farmacología
20.
PLoS One ; 4(4): e5323, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19390626

RESUMEN

DNA replication produces tangled, or catenated, chromatids, that must be decatenated prior to mitosis or catastrophic genomic damage will occur. Topoisomerase IIalpha (Topo IIalpha) is the primary decatenating enzyme. Cells monitor catenation status and activate decatenation checkpoints when decatenation is incomplete, which occurs when Topo IIalpha is inhibited by chemotherapy agents such as the anthracyclines and epididophyllotoxins. We recently demonstrated that the DNA repair component Metnase (also called SETMAR) enhances Topo IIalpha-mediated decatenation, and hypothesized that Metnase could mediate resistance to Topo IIalpha inhibitors. Here we show that Metnase interacts with Topo IIalpha in breast cancer cells, and that reducing Metnase expression significantly increases metaphase decatenation checkpoint arrest. Repression of Metnase sensitizes breast cancer cells to Topo IIalpha inhibitors, and directly blocks the inhibitory effect of the anthracycline adriamycin on Topo IIalpha-mediated decatenation in vitro. Thus, Metnase may mediate resistance to Topo IIalpha inhibitors, and could be a biomarker for clinical sensitivity to anthracyclines. Metnase could also become an important target for combination chemotherapy with current Topo IIalpha inhibitors, specifically in anthracycline-resistant breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Proteínas de Unión al ADN/antagonistas & inhibidores , Resistencia a Antineoplásicos , N-Metiltransferasa de Histona-Lisina/metabolismo , Inhibidores de Topoisomerasa II , Antraciclinas/farmacología , Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA