Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Cancer Res ; 81(18): 4861-4873, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34333454

RESUMEN

Isocitrate dehydrogenase-mutant low-grade gliomas (IDHmut-LGG) grow slowly but frequently undergo malignant transformation, which eventually leads to premature death. Chemotherapy and radiotherapy treatments prolong survival, but can also induce genetic (or epigenetic) alterations involved in transformation. Here, we developed a mathematical model of tumor progression based on serial tumor volume data and treatment history of 276 IDHmut-LGGs classified by chromosome 1p/19q codeletion (IDHmut/1p19qcodel and IDHmut/1p19qnoncodel) and performed genome-wide mutational analyses, including targeted sequencing and longitudinal whole-exome sequencing data. These analyses showed that tumor mutational burden correlated positively with malignant transformation rate, and chemotherapy and radiotherapy significantly suppressed tumor growth but increased malignant transformation rate per cell by 1.8 to 2.8 times compared with before treatment. This model revealed that prompt adjuvant chemoradiotherapy prolonged malignant transformation-free survival in small IDHmut-LGGs (≤ 50 cm3). Furthermore, optimal treatment differed according to genetic alterations for large IDHmut-LGGs (> 50 cm3); adjuvant therapies delayed malignant transformation in IDHmut/1p19qnoncodel but often accelerated it in IDHmut/1p19qcodel. Notably, PI3K mutation was not associated with malignant transformation but increased net postoperative proliferation rate and decreased malignant transformation-free survival, prompting the need for adjuvant therapy in IDHmut/1p19qcodel. Overall, this model uncovered therapeutic strategies that could prevent malignant transformation and, consequently, improve overall survival in patients with IDHmut-LGGs. SIGNIFICANCE: A mathematical model successfully estimates malignant transformation-free survival and reveals a link between genetic alterations and progression, identifying precision medicine approaches for optimal treatment of IDH-mutant low-grade gliomas.


Asunto(s)
Transformación Celular Neoplásica/genética , Análisis Mutacional de ADN/métodos , Glioma/genética , Glioma/patología , Isocitrato Deshidrogenasa/genética , Modelos Teóricos , Mutación , Adulto , Biomarcadores de Tumor , Variaciones en el Número de Copia de ADN , Manejo de la Enfermedad , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Glioma/mortalidad , Glioma/terapia , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Pronóstico , Resultado del Tratamiento , Carga Tumoral
3.
Nat Cell Biol ; 21(7): 879-888, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31263265

RESUMEN

Most human tumours are heterogeneous, composed of cellular clones with different properties present at variable frequencies. Highly heterogeneous tumours have poor clinical outcomes, yet the underlying mechanism remains poorly understood. Here, we show that minor subclones of breast cancer cells expressing IL11 and FIGF (VEGFD) cooperate to promote metastatic progression and generate polyclonal metastases composed of driver and neutral subclones. Expression profiling of the epithelial and stromal compartments of monoclonal and polyclonal primary and metastatic lesions revealed that this cooperation is indirect, mediated through the local and systemic microenvironments. We identified neutrophils as a leukocyte population stimulated by the IL11-expressing minor subclone and showed that the depletion of neutrophils prevents metastatic outgrowth. Single-cell RNA-seq of CD45+ cell populations from primary tumours, blood and lungs demonstrated that IL11 acts on bone-marrow-derived mesenchymal stromal cells, which induce pro-tumorigenic and pro-metastatic neutrophils. Our results indicate key roles for non-cell-autonomous drivers and minor subclones in metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/patología , Neutrófilos/metabolismo , Microambiente Tumoral , Animales , Carcinogénesis/metabolismo , Progresión de la Enfermedad , Humanos , Pulmón/patología , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/citología
4.
PLoS One ; 14(4): e0215409, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31026288

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) exhibits a variety of phenotypes with regard to disease progression and treatment response. This variability complicates clinical decision-making despite the improvement of survival due to the recent introduction of FOLFIRINOX (FFX) and nab-paclitaxel. Questions remain as to the timing and sequence of therapies and the role of radiotherapy for unresectable PDAC. Here we developed a computational analysis platform to investigate the dynamics of growth, metastasis and treatment response to FFX, gemcitabine (GEM), and GEM+nab-paclitaxel. Our approach was informed using data of 1,089 patients treated at the Massachusetts General Hospital and validated using an independent cohort from Osaka Medical College. Our framework establishes a logistic growth pattern of PDAC and defines the Local Advancement Index (LAI), which determines the eventual primary tumor size and predicts the number of metastases. We found that a smaller LAI leads to a larger metastatic burden. Furthermore, our analyses ascertain that i) radiotherapy after induction chemotherapy improves survival in cases receiving induction FFX or with larger LAI, ii) neoadjuvant chemotherapy improves survival in cases with resectable PDAC, and iii) temporary cessations of chemotherapies do not impact overall survival, which supports the feasibility of treatment holidays for patients with FFX-associated adverse effects. Our findings inform clinical decision-making for PDAC patients and allow for the rational design of clinical strategies using FFX, GEM, GEM+nab-paclitaxel, neoadjuvant chemotherapy, and radiation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/terapia , Desoxicitidina/análogos & derivados , Modelos Biológicos , Neoplasias Pancreáticas/terapia , Anciano , Albúminas/uso terapéutico , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Quimioradioterapia/métodos , Toma de Decisiones Clínicas , Simulación por Computador , Desoxicitidina/uso terapéutico , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Esquema de Medicación , Estudios de Factibilidad , Femenino , Fluorouracilo/uso terapéutico , Humanos , Irinotecán/uso terapéutico , Estimación de Kaplan-Meier , Leucovorina/uso terapéutico , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Oxaliplatino/uso terapéutico , Paclitaxel/uso terapéutico , Páncreas/patología , Páncreas/cirugía , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Inducción de Remisión/métodos , Carga Tumoral , Gemcitabina
5.
JCO Clin Cancer Inform ; 3: 1-11, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30901235

RESUMEN

Despite recent progress in diagnostic and multimodal treatment approaches, most cancer deaths are still caused by metastatic spread and the subsequent growth of tumor cells in sites distant from the primary organ. So far, few quantitative studies are available that allow for the estimation of metastatic parameters and the evaluation of alternative treatment strategies. Most computational studies have focused on situations in which the tumor cell population expands exponentially over time; however, tumors may eventually be subject to resource and space limitations so that their growth patterns deviate from exponential growth to adhere to density-dependent growth models. In this study, we developed a stochastic evolutionary model of cancer progression that considers alterations in metastasis-related genes and intercellular growth competition leading to density effects described by logistic growth. Using this stochastic model, we derived analytical approximations for the time between the initiation of tumorigenesis and diagnosis, the expected number of metastatic sites, the total number of metastatic cells, the size of the primary tumor, and survival. Furthermore, we investigated the effects of drug administration and surgical resection on these quantities and predicted outcomes for different treatment regimens. Parameter values used in the analysis were estimated from data obtained from a pancreatic cancer rapid autopsy program. Our theoretical approach allows for flexible modeling of metastatic progression dynamics.


Asunto(s)
Evolución Clonal , Modelos Biológicos , Neoplasias Pancreáticas/patología , Algoritmos , Evolución Clonal/genética , Progresión de la Enfermedad , Humanos , Modelos Logísticos , Metástasis de la Neoplasia , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/mortalidad , Pronóstico , Procesos Estocásticos
7.
Cancer Cell ; 34(6): 939-953.e9, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30472020

RESUMEN

Members of the KDM5 histone H3 lysine 4 demethylase family are associated with therapeutic resistance, including endocrine resistance in breast cancer, but the underlying mechanism is poorly defined. Here we show that genetic deletion of KDM5A/B or inhibition of KDM5 activity increases sensitivity to anti-estrogens by modulating estrogen receptor (ER) signaling and by decreasing cellular transcriptomic heterogeneity. Higher KDM5B expression levels are associated with higher transcriptomic heterogeneity and poor prognosis in ER+ breast tumors. Single-cell RNA sequencing, cellular barcoding, and mathematical modeling demonstrate that endocrine resistance is due to selection for pre-existing genetically distinct cells, while KDM5 inhibitor resistance is acquired. Our findings highlight the importance of cellular phenotypic heterogeneity in therapeutic resistance and identify KDM5A/B as key regulators of this process.


Asunto(s)
Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteína 2 de Unión a Retinoblastoma/genética , Transcriptoma/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Fulvestrant/farmacología , Heterogeneidad Genética , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células MCF-7 , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteína 2 de Unión a Retinoblastoma/metabolismo , Transcriptoma/efectos de los fármacos , Secuenciación del Exoma/métodos
8.
Cancer Res ; 77(12): 3325-3335, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381541

RESUMEN

Phenotypic diversity in pancreatic ductal adenocarcinoma (PDAC) results in a variety of treatment responses. Rapid autopsy studies have revealed a subgroup of PDAC patients with a lower propensity to develop metastatic disease, challenging the common perception that all patients die of widely metastatic disease, but questions remain about root causes of this difference and the potential impact on treatment strategies. In this study, we addressed these questions through the development of a mathematical model of PDAC progression that incorporates the major alteration status of specific genes with predictive utility. The model successfully reproduced clinical outcomes regarding metastatic patterns and the genetic alteration status of patients from two independent cohorts from the United States and Japan. Using this model, we defined a candidate predictive signature in patients with low metastatic propensity. If a primary tumor contained a small fraction of cells with KRAS and additional alterations to CDKN2A, TP53, or SMAD4 genes, the patient was likely to exhibit low metastatic propensity. By using this predictive signature, we computationally simulated a set of clinical trials to model whether this subgroup would benefit from locally intensive therapies such as surgery or radiation therapy. The largest overall survival benefit resulted from complete resection, followed by adjuvant chemoradiation therapy and salvage therapies for isolated recurrence. While requiring prospective validation in a clinical trial, our results suggest a new tool to help personalize care in PDAC patients in seeking the most effective therapeutic modality for each individual. Cancer Res; 77(12); 3325-35. ©2017 AACR.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Modelos Teóricos , Neoplasias Pancreáticas/patología , Medicina de Precisión/métodos , Transcriptoma , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/terapia , Humanos , Estimación de Kaplan-Meier , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas
9.
Sci Rep ; 6: 34214, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27694914

RESUMEN

Although the capacity of the liver to recover its size after resection has enabled extensive liver resection, post-hepatectomy liver failure remains one of the most lethal complications of liver resection. Therefore, it is clinically important to discover reliable predictive factors after resection. In this study, we established a novel mathematical framework which described post-hepatectomy liver regeneration in each patient by incorporating quantitative clinical data. Using the model fitting to the liver volumes in series of computed tomography of 123 patients, we estimated liver regeneration rates. From the estimation, we found patients were divided into two groups: i) patients restored the liver to its original size (Group 1, n = 99); and ii) patients experienced a significant reduction in size (Group 2, n = 24). From discriminant analysis in 103 patients with full clinical variables, the prognosis of patients in terms of liver recovery was successfully predicted in 85-90% of patients. We further validated the accuracy of our model prediction using a validation cohort (prediction = 84-87%, n = 39). Our interdisciplinary approach provides qualitative and quantitative insights into the dynamics of liver regeneration. A key strength is to provide better prediction in patients who had been judged as acceptable for resection by current pragmatic criteria.


Asunto(s)
Regeneración Hepática , Modelos Estadísticos , Periodo Posoperatorio , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
Sci Rep ; 5: 15886, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26515895

RESUMEN

Metastasis is a leading cause of cancer-related deaths. Carcinoma generally initiates at a specific organ as a primary tumor, but eventually metastasizes and forms tumor sites in other organs. In this report, we developed a mathematical model of cancer progression with alterations in metastasis-related genes. In cases in which tumor cells acquire metastatic ability through two steps of genetic alterations, we derive formulas for the probability, the expected number, and the distribution of the number of metastases. Moreover, we investigate practical pancreatic cancer disease progression in cases in which both one and two steps of genetic alterations are responsible for metastatic formation. Importantly, we derive a mathematical formula for the survival outcome validated using clinical data as well as direct simulations. Our model provides theoretical insights into how invisible metastases distribute upon diagnosis with respect to growth rates, (epi)genetic alteration rates, metastatic rate, and detection size. Prediction of survival outcome using the formula is of clinical importance in terms of determining therapeutic strategies.


Asunto(s)
Modelos Teóricos , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Progresión de la Enfermedad , Epigénesis Genética , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidad , Análisis de Supervivencia
11.
Cell Rep ; 11(1): 71-84, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25843711

RESUMEN

Upon systemic bacterial infection, hematopoietic stem and progenitor cells (HSPCs) migrate to the periphery in order to supply a sufficient number of immune cells. Although pathogen-associated molecular patterns reportedly mediate HSPC activation, how HSPCs detect pathogen invasion in vivo remains elusive. Bacteria use the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) for a variety of activities. Here, we report that c-di-GMP comprehensively regulated both HSPCs and their niche cells through an innate immune sensor, STING, thereby inducing entry into the cell cycle and mobilization of HSPCs while decreasing the number and repopulation capacity of long-term hematopoietic stem cells. Furthermore, we show that type I interferon acted as a downstream target of c-di-GMP to inhibit HSPC expansion in the spleen, while transforming growth factor-ß was required for c-di-GMP-dependent splenic HSPC expansion. Our results define machinery underlying the dynamic regulation of HSPCs and their niches during bacterial infection through c-di-GMP/STING signaling.


Asunto(s)
GMP Cíclico/análogos & derivados , Células Madre Hematopoyéticas/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Proteínas de la Membrana/genética , Animales , Bacterias/metabolismo , Bacterias/patogenicidad , GMP Cíclico/administración & dosificación , GMP Cíclico/inmunología , GMP Cíclico/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/microbiología , Factor 3 Regulador del Interferón/inmunología , Proteínas de la Membrana/inmunología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Nicho de Células Madre/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
12.
PLoS One ; 9(8): e105724, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25158060

RESUMEN

Platinum drugs and PARP inhibitors ("PARPis") are considered to be effective in BRCA-associated cancers with impaired DNA repair. These agents cause stalled and collapsed replication forks and create double-strand breaks effectively in the absence of repair mechanisms, resulting in arrest of the cell cycle and induction of cell death. However, recent studies have shown failure of these chemotherapeutic agents due to emerging drug resistance. In this study, we developed a stochastic model of BRCA-associated cancer progression in which there are four cancer populations: those with (i) functional BRCA, (ii) dysfunctional BRCA, (iii) functional BRCA and a growth advantage, and (iv) dysfunctional BRCA and a growth advantage. These four cancer populations expand from one cancer cell with normal repair function until the total cell number reaches a detectable amount. We derived formulas for the probability and expected numbers of each population at the time of detection. Furthermore, we extended the model to consider the tumor dynamics during treatment. Results from the model were validated and showed good agreement with clinical and experimental evidence in BRCA-associated cancers. Based on the model, we investigated conditions in which drug resistance during the treatment course originated from either a pre-existing drug-resistant population or a de novo population, due to secondary mutations. Finally, we found that platinum drugs and PARPis were effective if (i) BRCA inactivation is present, (ii) the cancer was diagnosed early, and (iii) tumor growth is rapid. Our results indicate that different types of cancers have a preferential way of acquiring resistance to platinum drugs and PARPis according to their growth and mutational characteristics.


Asunto(s)
Antineoplásicos/farmacología , Carboplatino/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Progresión de la Enfermedad , Evolución Molecular , Genes BRCA1 , Genes BRCA2 , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/patología
13.
PLoS One ; 8(4): e60043, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573231

RESUMEN

Morphological analysis of mitotic chromosomes is used to detect mutagenic chemical compounds and to estimate the dose of ionizing radiation to be administered. It has long been believed that chromosomal breaks are always associated with double-strand breaks (DSBs). We here provide compelling evidence against this canonical theory. We employed a genetic approach using two cell lines, chicken DT40 and human Nalm-6. We measured the number of chromosomal breaks induced by three replication-blocking agents (aphidicolin, 5-fluorouracil, and hydroxyurea) in DSB-repair-proficient wild-type cells and cells deficient in both homologous recombination and nonhomologous end-joining (the two major DSB-repair pathways). Exposure of cells to the three replication-blocking agents for at least two cell cycles resulted in comparable numbers of chromosomal breaks for RAD54(-/-/)KU70(-/-) DT40 clones and wild-type cells. Likewise, the numbers of chromosomal breaks induced in RAD54(-/-/)LIG4(-/-) Nalm-6 clones and wild-type cells were also comparable. These data indicate that the replication-blocking agents can cause chromosomal breaks unassociated with DSBs. In contrast with DSB-repair-deficient cells, chicken DT40 cells deficient in PIF1 or ATRIP, which molecules contribute to the completion of DNA replication, displayed higher numbers of mitotic chromosomal breaks induced by aphidicolin than did wild-type cells, suggesting that single-strand gaps left unreplicated may result in mitotic chromosomal breaks.


Asunto(s)
Rotura Cromosómica , Replicación del ADN/efectos de los fármacos , Mitosis , Animales , Antígenos Nucleares/genética , Afidicolina/farmacología , Apoptosis , Línea Celular Tumoral , Pollos , Roturas del ADN de Doble Cadena , ADN Helicasas/fisiología , Reparación del ADN , Enzimas Reparadoras del ADN/fisiología , Proteínas de Unión al ADN/genética , Fluorouracilo/farmacología , Técnicas de Inactivación de Genes , Humanos , Hidroxiurea/farmacología , Autoantígeno Ku
14.
PLoS Genet ; 7(7): e1002148, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21779174

RESUMEN

RAD51 recombinase polymerizes at the site of double-strand breaks (DSBs) where it performs DSB repair. The loss of RAD51 causes extensive chromosomal breaks, leading to apoptosis. The polymerization of RAD51 is regulated by a number of RAD51 mediators, such as BRCA1, BRCA2, RAD52, SFR1, SWS1, and the five RAD51 paralogs, including XRCC3. We here show that brca2-null mutant cells were able to proliferate, indicating that RAD51 can perform DSB repair in the absence of BRCA2. We disrupted the BRCA1, RAD52, SFR1, SWS1, and XRCC3 genes in the brca2-null cells. All the resulting double-mutant cells displayed a phenotype that was very similar to that of the brca2-null cells. We suggest that BRCA2 might thus serve as a platform to recruit various RAD51 mediators at the appropriate position at the DNA-damage site.


Asunto(s)
Proteína BRCA2/genética , Epistasis Genética , Recombinación Homóloga , Recombinasa Rad51/genética , Animales , Camptotecina/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Pollos , Aberraciones Cromosómicas/efectos de los fármacos , Aberraciones Cromosómicas/efectos de la radiación , Cisplatino/farmacología , Células Clonales , Daño del ADN , Epistasis Genética/efectos de los fármacos , Epistasis Genética/efectos de la radiación , Rayos gamma , Conversión Génica/efectos de los fármacos , Conversión Génica/efectos de la radiación , Eliminación de Gen , Sitios Genéticos/genética , Genoma/genética , Recombinación Homóloga/efectos de los fármacos , Recombinación Homóloga/efectos de la radiación , Modelos Biológicos , Fenotipo , Ftalazinas/farmacología , Piperazinas/farmacología , Recombinasa Rad51/deficiencia
15.
Environ Mol Mutagen ; 52(7): 547-61, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21538559

RESUMEN

Included among the quantitative high throughput screens (qHTS) conducted in support of the US Tox21 program are those being evaluated for the detection of genotoxic compounds. One such screen is based on the induction of increased cytotoxicity in seven isogenic chicken DT40 cell lines deficient in DNA repair pathways compared to the parental DNA repair-proficient cell line. To characterize the utility of this approach for detecting genotoxic compounds and identifying the type(s) of DNA damage induced, we evaluated nine of 42 compounds identified as positive for differential cytotoxicity in qHTS (actinomycin D, adriamycin, alachlor, benzotrichloride, diglycidyl resorcinol ether, lovastatin, melphalan, trans-1,4-dichloro-2-butene, tris(2,3-epoxypropyl)isocyanurate) and one non-cytotoxic genotoxic compound (2-aminothiamine) for (1) clastogenicity in mutant and wild-type cells; (2) the comparative induction of γH2AX positive foci by melphalan; (3) the extent to which a 72-hr exposure duration increased assay sensitivity or specificity; (4) the use of 10 additional DT40 DNA repair-deficient cell lines to better analyze the type(s) of DNA damage induced; and (5) the involvement of reactive oxygen species in the induction of DNA damage. All compounds but lovastatin and 2-aminothiamine were more clastogenic in at least one DNA repair-deficient cell line than the wild-type cells. The differential responses across the various DNA repair-deficient cell lines provided information on the type(s) of DNA damage induced. The results demonstrate the utility of this DT40 screen for detecting genotoxic compounds, for characterizing the nature of the DNA damage, and potentially for analyzing mechanisms of mutagenesis.


Asunto(s)
Daño del ADN/efectos de los fármacos , Trastornos por Deficiencias en la Reparación del ADN/genética , Mutágenos/toxicidad , Acetamidas/toxicidad , Animales , Línea Celular , Pollos , Aberraciones Cromosómicas , Daño del ADN/genética , Dactinomicina/toxicidad , Doxorrubicina/toxicidad , Lovastatina/toxicidad , Melfalán/toxicidad , Pruebas de Mutagenicidad , Resorcinoles/toxicidad , Tolueno/análogos & derivados , Tolueno/toxicidad
16.
Proc Natl Acad Sci U S A ; 108(16): 6492-6, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21464321

RESUMEN

Interstrand cross-links (ICLs) block replication and transcription and thus are highly cytotoxic. In higher eukaryotes, ICLs processing involves the Fanconi anemia (FA) pathway and homologous recombination. Stalled replication forks activate the eight-subunit FA core complex, which ubiquitylates FANCD2-FANCI. Once it is posttranslationally modified, this heterodimer recruits downstream members of the ICL repairosome, including the FAN1 nuclease. However, ICL processing has been shown to also involve MUS81-EME1 and XPF-ERCC1, nucleases known to interact with SLX4, a docking protein that also can bind another nuclease, SLX1. To investigate the role of SLX4 more closely, we disrupted the SLX4 gene in avian DT40 cells. SLX4 deficiency caused cell death associated with extensive chromosomal aberrations, including a significant fraction of isochromatid-type breaks, with sister chromatids broken at the same site. SLX4 thus appears to play an essential role in cell proliferation, probably by promoting the resolution of interchromatid homologous recombination intermediates. Because ubiquitylation plays a key role in the FA pathway, and because the N-terminal region of SLX4 contains a ubiquitin-binding zinc finger (UBZ) domain, we asked whether this domain is required for ICL processing. We found that SLX4(-/-) cells expressing UBZ-deficient SLX4 were selectively sensitive to ICL-inducing agents, and that the UBZ domain was required for interaction of SLX4 with ubiquitylated FANCD2 and for its recruitment to DNA-damage foci generated by ICL-inducing agents. Our findings thus suggest that ubiquitylated FANCD2 recruits SLX4 to DNA damage sites, where it mediates the resolution of recombination intermediates generated during the processing of ICLs.


Asunto(s)
Reparación del ADN/fisiología , Endonucleasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Recombinasas/metabolismo , Recombinación Genética/fisiología , Ubiquitinación/fisiología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Pollos , Cromátides/genética , Cromátides/metabolismo , Aberraciones Cromosómicas/efectos de los fármacos , Reactivos de Enlaces Cruzados/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Reparación del ADN/efectos de los fármacos , Endonucleasas/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Recombinasas/genética , Recombinación Genética/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Dedos de Zinc
17.
DNA Repair (Amst) ; 9(12): 1292-8, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21030320

RESUMEN

Chemicals used industrially and commercially are required by law to be assessed for their genotoxic potential. However, all currently used assays have major limitations and despite intense effort, there is no universal agreement on which tests should be employed, or how to interpret results. We have developed a new assay system using the chicken DT40 B cell line that offers a number of significant advantages over current methodologies. Our assay could provide enhanced sensitivity using genetically defined and phenotypically characterized mutants defective in DNA repair pathways. Furthermore, analysis of the mutants, using DNA repair proficient wild-type cells as a negative control, minimizes false negative outcomes. Assessing the different responses of a panel of mutants representative of all repair pathways, mechanistic detail of genotoxicity can be determined. This unique feature, as well as reducing the false positive rate, strengthens positive identifications and is useful when extrapolating results to the human context. Our panel of mutants is likely to be useful in screening large compound libraries for an emerging class of chemotherapeutic drugs, which includes inhibitors of DNA repair enzymes such as PARP and DNA polymerases.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , Replicación del ADN/fisiología , Pruebas de Mutagenicidad/métodos , Mutágenos/metabolismo , Mutación/genética , Animales , Línea Celular , Pollos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...