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











Base de datos
Intervalo de año de publicación
1.
Lab Invest ; 104(2): 100297, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38008183

RESUMEN

The detection of microsatellite instability (MSI) and mismatch repair (MMR) deficiency has become mandatory for most tumors in recent years, owing to the development of immune checkpoint inhibitors as a highly effective therapy for MMR deficiency/MSI tumors. The timely and efficient detection of MSI is valuable, and new methods are increasingly being developed. To date, MMR assessment has been performed using immunohistochemistry of the 4 MMR proteins and/or microsatellite stability/MSI using PCR, mostly using the pentaplex panel. The implementation of next-generation sequencing (NGS) for MSI analysis would improve the effectiveness at a lower cost and in less time. This study describes the development of 8 new microsatellites combined with a classification algorithm, termed "Octaplex CaBio-MSID" (for Cancérologie Biologique MSI Detection tool), to assess MSI using NGS. A series of 303 colorectal cancer and 88 endometrial cancer samples were assessed via MSI testing using NGS using the Octaplex CaBio-MSID algorithm. The sensitivity and specificity of Octaplex CaBio-MSID were 98.4% and 98.4% for colorectal cancers, and 89.3% and 100% for endometrial cancers, respectively. This new NGS-based MSI detection method outperforms previously published methods (ie, Idylla [Biocartis], OncoMate MSI Dx [Promega], and Foundation One CDx [Roche Foundation Medicine]). Although highly efficient, Octaplex CaBio-MSID requires validation in a larger independent series of different tumor types.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Neoplasias Endometriales , Síndromes Neoplásicos Hereditarios , Femenino , Humanos , Inestabilidad de Microsatélites , Reparación de la Incompatibilidad de ADN/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos
2.
Cell Mol Gastroenterol Hepatol ; 16(5): 757-782, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37482243

RESUMEN

BACKGROUND & AIMS: Brain metastases (BMs) from colorectal cancer (CRC) are associated with significant morbidity and mortality, with chemoresistance and short overall survival. Migrating cancer stem cells with the ability to initiate BM have been described in breast and lung cancers. In this study, we describe the identification and characterization of cancer stem cells in BM from CRC. METHODS: Four brain metastasis stem cell lines from patients with colorectal cancer (BM-SC-CRC1 to BM-SC-CRC4) were obtained by mechanical dissociation of patient's tumors and selection of cancer stem cells by appropriate culture conditions. BM-SC-CRCs were characterized in vitro by clonogenic and limiting-dilution assays, as well as immunofluorescence and Western blot analyses. In ovo, a chicken chorioallantoic membrane (CAM) model and in vivo, xenograft experiments using BALB/c-nude mice were realized. Finally, a whole exome and RNA sequencing analyses were performed. RESULTS: BM-SC-CRC formed metaspheres and contained tumor-initiating cells with self-renewal properties. They expressed stem cell surface markers (CD44v6, CD44, and EpCAM) in serum-free medium and CRC markers (CK19, CK20 and CDX-2) in fetal bovine serum-enriched medium. The CAM model demonstrated their invasive and migratory capabilities. Moreover, mice intracranial xenotransplantation of BM-SC-CRCs adequately recapitulated the original patient BM phenotype. Finally, transcriptomic and genomic approaches showed a significant enrichment of invasiveness and specific stemness signatures and highlighted KMT2C as a potential candidate gene to potentially identify high-risk CRC patients. CONCLUSIONS: This original study represents the first step in CRC BM initiation and progression comprehension, and further investigation could open the way to new therapeutics avenues to improve patient prognosis.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Humanos , Ratones , Animales , Neoplasias Colorrectales/patología , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Xenoinjertos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología
3.
Cancer Med ; 8(18): 7556-7566, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31651105

RESUMEN

INTRODUCTION: ALK and ROS1 rearrangements are molecular targets of several tyrosine kinase inhibitors. RNA-sequencing approaches are regarded as the new standard for fusion gene detection, representing an alternative to standard immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) techniques. PATIENTS AND METHODS: We aimed to compare two recent amplicon-based RNA-sequencing techniques: FusionPlex® Alk Ret Ros1 v2 Kit (Archer® ) with FHS-003Z-12-Human Lung Cancer Panel (Qiagen® ) and assessed the accuracy of the data for therapy management. Thirty-seven formalin-fixed paraffin-embedded non-small cell carcinoma (NSCC) lesions initially explored by IHC and FISH were selected for RNA-sequencing analysis. RESULTS: Qiagen® and Archer® kits produced similar results and correctly identified 85.1% (23/27) and 81.5% (22/27) of IHC/FISH ALK- and ROS1-positive samples, respectively, and 100% (6/6) of the negative samples. With regard to the ambiguous IHC-positive/FISH-negative cases, RNA-sequencing confirmed 75% (3/4) of the FISH conclusion. Although not statistically significant, patients with common EML4-ALK variants presented shorter overall survival and progression-free survival compared with patients harboring rare variants. CONCLUSION: Our findings assessed the implementation of RNA-sequencing approaches to explore ALK and ROS1 rearrangements from formalin-fixed paraffin-embedded samples. We highlighted the similarities between Qiagen® and Archer® kits in terms of handling time, cost, and outcomes. We confirmed the feasibility of molecular testing in routine organization and its possible use not only as an alternative for standard IHC and FISH techniques, but as a supplementary technique helping to classify discrepant cases.


Asunto(s)
Análisis de Secuencia de ARN/métodos , Adulto , Anciano , Anciano de 80 o más Años , Quinasa de Linfoma Anaplásico/genética , Biomarcadores de Tumor , Biopsia , Carcinoma de Pulmón de Células no Pequeñas/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Análisis de Secuencia de ARN/normas , Secuenciación del Exoma
4.
Int J Mol Sci ; 19(10)2018 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-30282933

RESUMEN

"Glioma Stem Cells" (GSCs) are known to play a role in glioblastoma (GBM) recurrence. Homologous recombination (HR) defects and cell cycle checkpoint abnormalities can contribute concurrently to the radioresistance of GSCs. DNA repair protein RAD51 homolog 1 (RAD51) is a crucial protein for HR and its inhibition has been shown to sensitize GSCs to irradiation. The aim of this study was to examine the consequences of ionizing radiation (IR) for cell cycle progression in GSCs. In addition, we intended to assess the potential effect of RAD51 inhibition on cell cycle progression. Five radiosensitive GSC lines and five GSC lines that were previously characterized as radioresistant were exposed to 4Gy IR, and cell cycle analysis was done by fluorescence-activated cell sorting (FACS) at 24, 48, 72, and 96 h with or without RAD51 inhibitor. Following 4Gy IR, all GSC lines presented a significant increase in G2 phase at 24 h, which was maintained over 72 h. In the presence of RAD51 inhibitor, radioresistant GSCs showed delayed G2 arrest post-irradiation for up to 48 h. This study demonstrates that all GSCs can promote G2 arrest in response to radiation-induced DNA damage. However, following RAD51 inhibition, the cell cycle checkpoint response differed. This study contributes to the characterization of the radioresistance mechanisms of GSCs, thereby supporting the rationale of targeting RAD51-dependent repair pathways in view of radiosensitizing GSCs.


Asunto(s)
Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Recombinasa Rad51/genética , Radiación Ionizante , Línea Celular Tumoral , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Recombinasa Rad51/metabolismo , Tolerancia a Radiación/genética
5.
Cancer Med ; 6(11): 2625-2634, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28960893

RESUMEN

Glioblastoma stem cells (GSCs) are believed to be involved in the mechanisms of tumor resistance, therapeutic failures, and recurrences after conventional glioblastoma therapy. Therefore, elimination of GSCs might be a prerequisite for the development of successful therapeutic strategies. ALK, ROS1, and MET are targeted by Crizotinib, a tyrosine kinase inhibitor which has been approved for treatment of ALK-rearranged non-small-cell lung cancer. In this study we investigated ALK, ROS1, and MET status in nine glioblastoma stem cell lines and tumors from which they arise. Fluorescent in situ hybridization (FISH), Sanger's direct sequencing, and immunohistochemistry were used to screen genomic rearrangements (or amplifications), genomic mutations, and protein expression, respectively. The immunohistochemical and FISH studies revealed no significant dysregulation of ROS1 in GSCs and associated tumors. Neither amplification nor polysomy of ALK was observed in GSC, but weak overexpression was detected by IHC in three of nine GSCs. Similarly, no MET amplification was found by FISH but three GSCs presented significant immunohistochemical staining. No ALK or MET mutation was found by Sanger's direct sequencing. In this study, we show no molecular rearrangement of ALK, ROS1, and MET that would lead us not to propose, as a valid strategy, the use of crizotinib to eradicate GSCs. However, MET was overexpressed in all GSCs with mesenchymal subtype and three GSCs presented an overexpression of ALK. Therefore, our study corroborates the idea that MET and ALK may assume a role in the tumorigenicity of GSC.


Asunto(s)
Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas c-met , Proteínas Proto-Oncogénicas , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras , Anciano , Quinasa de Linfoma Anaplásico , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/metabolismo , Crizotinib , Análisis Mutacional de ADN , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Células Madre Neoplásicas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Pirazoles/metabolismo , Piridinas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo
6.
Int J Mol Sci ; 17(12)2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27916952

RESUMEN

Approximately 30% of patients with wild type RAS metastatic colorectal cancer are non-responders to anti-epidermal growth factor receptor monoclonal antibodies (anti-EGFR mAbs), possibly due to undetected tumoral subclones harboring RAS mutations. The aim of this study was to analyze the distribution of RAS mutations in different areas of the primary tumor, metastatic lymph nodes and distant metastasis. A retrospective cohort of 18 patients with a colorectal cancer (CRC) was included in the study. Multiregion analysis was performed in 60 spatially separated tumor areas according to the pathological tumor node metastasis (pTNM) staging and KRAS, NRAS and BRAF mutations were tested using pyrosequencing. In primary tumors, intra-tumoral heterogeneity for RAS mutation was found in 33% of cases. Inter-tumoral heterogeneity for RAS mutation between primary tumors and metastatic lymph nodes or distant metastasis was found in 36% of cases. Moreover, 28% of tumors had multiple RAS mutated subclones in the same tumor. A high proportion of CRCs presented intra- and/or inter-tumoral heterogeneity, which has relevant clinical implications for anti-EGFR mAbs prescription. These results suggest the need for multiple RAS testing in different parts of the same tumor and/or more sensitive techniques.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , GTP Fosfohidrolasas/genética , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Cetuximab/administración & dosificación , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Femenino , Heterogeneidad Genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia
7.
BMC Cancer ; 16: 604, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27495836

RESUMEN

BACKGROUND: Radioresistant glioblastoma stem cells (GSCs) contribute to tumor recurrence and identification of the molecular targets involved in radioresistance mechanisms is likely to enhance therapeutic efficacy. This study analyzed the DNA damage response following ionizing radiation (IR) in 10 GSC lines derived from patients. METHODS: DNA damage was quantified by Comet assay and DNA repair effectors were assessed by Low Density Array. The effect of RAD51 inhibitor, RI-1, was evaluated by comet and annexin V assays. RESULTS: While all GSC lines displayed efficient DNA repair machinery following ionizing radiation, our results demonstrated heterogeneous responses within two distinct groups showing different intrinsic radioresistance, up to 4Gy for group 1 and up to 8Gy for group 2. Radioresistant cell group 2 (comprising 5 out of 10 GSCs) showed significantly higher RAD51 expression after IR. In these cells, inhibition of RAD51 prevented DNA repair up to 180 min after IR and induced apoptosis. In addition, RAD51 protein expression in glioblastoma seems to be associated with poor progression-free survival. CONCLUSION: These results underscore the importance of RAD51 in radioresistance of GSCs. RAD51 inhibition could be a therapeutic strategy helping to treat a significant number of glioblastoma, in combination with radiotherapy.


Asunto(s)
Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Recombinasa Rad51/metabolismo , Tolerancia a Radiación/fisiología , Western Blotting , Línea Celular Tumoral , Ensayo Cometa , Daño del ADN/efectos de la radiación , Citometría de Flujo , Humanos , Inmunohistoquímica , Células Madre Neoplásicas/efectos de la radiación , Análisis de Matrices Tisulares
8.
Exp Mol Pathol ; 99(2): 207-11, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26163758

RESUMEN

In advanced colorectal carcinoma (CRC) patients, extended RAS mutations testing (KRAS exons 2 to 4 and NRAS exons 2 to 4) is a prerequisite for patient stratification to anti-EGFr therapy. Accurately distinguishing mutant patients from potential responders has a clinically critical impact, and thus effective and low cost methods are needed for identification of the mutation status. We have developed quantitative pyrosequencing assays for sensitive and rapid detection of mutant RAS alleles in formalin-fixed, paraffin-embedded tissues. Exons 2 to 4 of KRAS and NRAS genes were PCR amplified and analyzed by pyrosequencing. For validation, PCR products were sequenced by conventional Sanger sequencing. Analytical sensitivity of these assays was determined by calculating the limit of detection. The results showed that low levels of mutant RAS alleles (2-13%) can be detected with pyrosequencing assays.


Asunto(s)
Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/métodos , Receptores ErbB/genética , Pruebas Genéticas/métodos , Mutación/genética , Reacción en Cadena de la Polimerasa/métodos , Proteínas ras/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Evaluación de Resultado en la Atención de Salud , Adhesión en Parafina
9.
Mol Cancer Ther ; 13(9): 2159-69, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053823

RESUMEN

Recent data suggest that inhibition of the Hedgehog pathway could be a therapeutic target for glioblastoma. Alkaloid cyclopamine inhibits Hedgehog signaling, depleting stem-like cancer cells derived from glioblastoma. However, this compound is toxic for somatic stem cells, preventing its use for clinical applications. In this study, we tested a derivatization product of cyclopamine in the form of cyclopamine glucuronide prodrug (CGP-2). This compound was used in vitro and in vivo toward glioblastoma-initiating cells (GIC). Results obtained in vitro indicate that CGP-2 is active only in the presence of ß-glucuronidase, an enzyme detected in high levels in necrotic areas of glioblastomas. CGP-2 decreased proliferation and inhibited the self-renewal of all GIC lines tested. Hedgehog pathway blockade by 10 µmol/L of CGP-2 induced a 99% inhibition of clonogenicity on GICs, similar to cyclopamine treatment. Combination of CGP-2 with radiation decreased clonogenic survival in all GIC lines compared with CGP-2 alone. In a subcutaneous glioblastoma xenograft model, a two-week CGP-2 treatment prevented tumor growth with 75% inhibition at 8 weeks, and this inhibition was still significant after 14 weeks. Unlike cyclopamine, CGP-2 had no detectable toxic effects in intestinal crypts. Our study suggests that inhibition of the Hedgehog pathway with CGP-2 is more effective than conventional temozolomide adjuvant, with much lower concentrations, and seems to be an effective therapeutic strategy for targeting GICs.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Glucurónidos/química , Células Madre Neoplásicas/citología , Profármacos/química , Alcaloides de Veratrum/química , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Concentración 50 Inhibidora , Metilcelulosa/química , Ratones , Ratones Desnudos , Trasplante de Neoplasias
10.
PLoS One ; 8(11): e80460, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260394

RESUMEN

Anti-RhD prophylaxis of haemolytic disease of the fetus and newborn (HDFN) is highly effective, but as the suppressive mechanism remains uncertain, a mouse model would be of interest. Here we have generated transgenic mice expressing human RhAG and RhD erythrocyte membrane proteins in the presence and, for human RhAG, in the absence, of mouse Rhag. Human RhAG associates with mouse Rh but not mouse Rhag on red blood cells. In Rhag knockout mice transgenic for human RHAG, the mouse Rh protein is "rescued" (re-expressed), and co-immunoprecipitates with human RhAG, indicating the presence of hetero-complexes which associate mouse and human proteins. RhD antigen was expressed from a human RHD gene on a BAC or from RHD cDNA under control of ß-globin regulatory elements. RhD was never observed alone, strongly indicative that its expression absolutely depends on the presence of transgenic human RhAG. This first expression of RhD in mice is an important step in the creation of a mouse model of RhD allo-immunisation and HDFN, in conjunction with the Rh-Rhag knockout mice we have developed previously.


Asunto(s)
Proteínas Sanguíneas/genética , Expresión Génica , Glicoproteínas de Membrana/genética , Sistema del Grupo Sanguíneo Rh-Hr/genética , Compuestos de Amonio/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Sistema del Grupo Sanguíneo Rh-Hr/química , Sistema del Grupo Sanguíneo Rh-Hr/metabolismo , Transcripción Genética , Globinas beta/metabolismo
11.
Cancer Chemother Pharmacol ; 72(2): 397-403, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23765179

RESUMEN

It is now well established that metastatic colorectal cancer patients without KRAS mutation (codon 12) benefit from treatment with an epidermal growth factor receptor monoclonal antibody (anti-EGFR mAb). Recently, EFGR and KRAS mutations have been shown to exist in patients who developed resistance to anti-EGFR mAb. We analyzed KRAS, BRAF V600E and EGFR S492R mutations in 37 post-anti-EGFR mAb tumor samples from 23 patients treated with chemotherapy plus anti-EGFR mAb. No EGFR S492R mutation was detected. A KRAS mutation was found after anti-EGFR mAb in only one tumor. Our results suggest that acquired EGFR S492R and KRAS mutations do not constitute the main mechanism of resistance to anti-EGFR mAb in combination with chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Receptores ErbB/genética , Receptores ErbB/inmunología , Mutación/genética , Mutación/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Anciano , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/patología , Terapia Combinada , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas p21(ras) , Resultado del Tratamiento
12.
Int J Mol Sci ; 13(6): 6983-6994, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22837675

RESUMEN

Glioblastoma (GBM) is the most malignant type of primary brain tumor with a very poor prognosis. The actual standard protocol of treatment for GBM patients consists of radiotherapy and concomitant temozolomide (TMZ). However, the therapeutic efficacy of this treatment is limited due to tumor recurrence and TMZ resistance. Recently isolated, glioma stem-like cells (GSCs) are thought to represent the population of tumorigenic cells responsible for GBM resistance and recurrence following surgery and chemotherapy. In addition, MGMT (O6-methylguanine-methyltransferase) methylation is considered as one of the principal mechanisms contributing to TMZ sensitivity of GBM. In this study we have isolated GSCs from 10 adult GBM patients and investigated the relationship between MGMT methylation status and Temozolomide (TMZ) sensitivity of these lines grown either in stem-like or differentiation promoting conditions. Sensitivity to TMZ was significantly associated with MGMT methylation status in cells committed to differentiation but not in stem-like cells. In addition, patients harboring highly methylated MGMT promoters had a longer overall survival. These results reveal the importance of the differentiation process when considering the predictive value of MGMT status in GSCs for clinical response to TMZ.


Asunto(s)
Neoplasias Encefálicas/genética , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioma/genética , Células Madre Neoplásicas/citología , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/genética , Anciano , Algoritmos , Diferenciación Celular , Línea Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/química , Femenino , Humanos , Concentración 50 Inhibidora , Masculino , Microscopía Confocal , Persona de Mediana Edad , Análisis de Secuencia de ADN , Temozolomida
13.
Int J Cancer ; 128(4): 826-38, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473906

RESUMEN

Glioblastoma (GBM), the highest-grade form of gliomas, is the most frequent and the most aggressive. Recently, a subpopulation of cells with stem cells characteristics, commonly named "tumor-initiating stem cells" (TISCs) or "cancer stem cells" (CSCs) were identified in GBM. These cells were shown to be highly resistant to chemotherapeutic drugs and to ionizing radiations. Consequently, the knowledge of the signals that regulate the functions and survival of TISCs is crucial. In our work, we describe a neurosphere-initiating cell (NS-IC) assay to quantify TISC/CSCs from patients with GBM and show that these cells are tumorigenic in vivo. We demonstrate that the intracellular signal transducer and activator of transcription STAT3 is constitutively activated by phosphorylation preferentially on serine 727 in these cells. Moreover, we demonstrate that the selective inhibition of STAT3 by the chemical compound Stattic or by siRNA STAT3 abrogates TISC/CSC proliferation and NS-IC suggesting that self-renewal of GBM "stem-like" cells depends on the presence of STAT3 for their maintenance. Finally, we show that inhibition of STAT3 by Stattic sensitizes TISC/CSCs to the inhibitory action of Temozolomide with a strong synergistic effect of both drugs. Overall, these results suggest that strategies focused on STAT3 inhibition are efficient at the level of "stem-like" cells and could be of interest for therapeutic purposes in patients with malignant GBM.


Asunto(s)
Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Óxidos S-Cíclicos/farmacología , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/antagonistas & inhibidores , Temozolomida , Células Tumorales Cultivadas
14.
Carcinogenesis ; 31(12): 2058-65, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926829

RESUMEN

Poly(ADP-ribose) metabolism, mediated mainly by poly(ADP-ribose) polymerase (PARP) 1 and poly(ADP-ribose) glycohydrolase (PARG), regulates various cellular processes in response to genotoxic stress. PARP1 has been shown to be important in multiple cellular processes, including DNA repair, chromosomal stability, chromatin function, apoptosis and transcriptional regulation. However, whether PARP1's polymer synthesizing activity or polymer homeostasis is responsible for these functions remains largely unknown. Given a concerted action of multiple PARPs and unique PARG in the homeostasis of poly(ADP-ribosyl)ation, PARG is hypothesized to function in these processes. The lethal phenotype of the PARG null mutation in mouse embryos, however, hampers further investigation on biological function of PARG. Here, we show that mouse embryonic fibroblasts carrying a hypomorphic mutation of PARG, i.e. lacking the nuclear 110 kD isoform (PARG(110)(-/-)), have defects in the repair of DNA damage caused by various genotoxic agents. PARG(110)(-/-) cells exhibit genomic instability, characterized by a high frequency of sister chromatid exchange, micronuclei formation and chromosomal aberrations. Moreover, mutant cells contain supernumerary centrosomes, another hallmark of genomic instability, which correlates with an accumulation of S-phase cells after replication poison. Intriguingly, PARG(110)(-/-) cells accumulate more Rad51 foci in response to hydroxyurea, indicative of a defective repair of replication fork damage. Finally, PARG(110)(-/-) mice are susceptible to diethylnitrosamine-induced hepatocellular carcinoma. These data demonstrate that the homeostasis of poly(ADP-ribosyl)ation is important for an efficient DNA repair of damaged replication forks and for stabilizing the genome, thereby preventing carcinogenesis.


Asunto(s)
Núcleo Celular/enzimología , Reparación del ADN , Inestabilidad Genómica , Glicósido Hidrolasas/fisiología , Isoenzimas/fisiología , Neoplasias/etiología , Animales , Células Cultivadas , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Replicación del ADN , Neoplasias Hepáticas Experimentales/inducido químicamente , Ratones , Micronúcleos con Defecto Cromosómico , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/fisiología , Recombinasa Rad51/fisiología , Intercambio de Cromátides Hermanas
15.
J Cereb Blood Flow Metab ; 26(5): 684-95, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16177811

RESUMEN

Poly(ADP-ribose) (PAR) is a polymer synthesized by poly(ADP-ribose) polymerases (PARPs) and metabolized into free adenosine diphosphate (ADP)-ribose units by poly(ADP-ribose) glycohydrolase (PARG). Perturbations in PAR synthesis have been shown to play a key role in brain disorders including postischemic brain damage. A single parg gene but two PARG isoforms (110 and 60 kDa) have been detected in mouse cells. Complete suppression of parg gene causes early embryonic lethality, whereas mice selectively lacking the 110 kDa PARG isoform (PARG(110)(-/-)) develop normally. We used PARG(110)(-/-) mice to evaluate the importance of PAR catabolism to postischemic brain damage. Poly(ADP-ribose) contents were higher in the brain tissue of PARG(110)(-/-) than PARG(110)(+/+) mice, both under basal conditions and after PARP activation. Distal middle cerebral artery occlusion caused higher increase of brain PAR levels and larger infarct volumes in PARG(110)(-/-) mice than in wild-type counterparts. Of note, the brain of PARG(110)(-/-) mice showed reduced heat-shock protein (HSP)-70 and increased cyclooxygenase-2 expression under both control and ischemic conditions. No differences were detected in brain expression/activation of procaspase-3, PARP-1, Akt, HSP-25 and interleukin-1beta. Our findings show that PAR accumulation worsens ischemic brain injury, and highlight the therapeutic potential of strategies capable of maintaining PAR homeostasis.


Asunto(s)
Isquemia Encefálica/patología , Glicósido Hidrolasas/metabolismo , Isoenzimas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Isquemia Encefálica/metabolismo , Activación Enzimática , Glicósido Hidrolasas/genética , Homeostasis , Técnicas In Vitro , Infarto de la Arteria Cerebral Media , Isoenzimas/genética , Ratones , Ratones Noqueados , NAD/metabolismo , Fármacos Neuroprotectores/metabolismo , Neurotoxinas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo
16.
FASEB J ; 19(6): 558-66, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15791006

RESUMEN

Poly (ADP-ribosyl)ation, an early post-translational modification in response to DNA damage, is catalyzed by poly (ADP-ribose) polymerase (PARP-1) and catabolized by poly(ADP-ribose) glycohydrolase (PARG). The aim of this study was to investigate the role of PARG on the modulation of the inflammatory response caused by splanchnic ischemia and reperfusion. SAO shock in rats and wild-type (WT) mice was associated with a significant neutrophil infiltration in the ileum and production of tumor necrosis factor-alpha (TNF-alpha). Reperfused ileum tissue sections from SAO-shocked WT mice and rats showed positive staining for P-selectin and ICAM-1 localized mainly in the vascular endothelial cells. Genetic disruption of the PARG gene in mice or pharmacological inhibition of PARG by PARG inhibitors significantly improved the histological status of the reperfused tissues associated with reduced expression of P-selectin and ICAM-1, neutrophil infiltration into the reperfused intestine, and TNF-alpha production. These results suggest that PARG activity modulates the inflammatory response in ischemia/reperfusion and participates in end (target) organ damage under these conditions.


Asunto(s)
Glicósido Hidrolasas/fisiología , Intestinos/irrigación sanguínea , Daño por Reperfusión/enzimología , Circulación Esplácnica , Animales , Arterias , Arteria Celíaca , Constricción , Inhibidores Enzimáticos/farmacología , Fluorenos/farmacología , Glicósido Hidrolasas/antagonistas & inhibidores , Glicósido Hidrolasas/deficiencia , Íleon/irrigación sanguínea , Íleon/química , Íleon/patología , Inmunohistoquímica , Inflamación , Molécula 1 de Adhesión Intercelular/análisis , Intestinos/química , Intestinos/patología , Masculino , Arteria Mesentérica Superior , Ratones , Ratones Noqueados , Neutrófilos/patología , Selectina-P/análisis , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Choque , Factor de Necrosis Tumoral alfa/análisis
17.
J Am Soc Nephrol ; 16(3): 712-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15677308

RESUMEN

The role of poly(ADP-ribose) (PAR) glycohydrolase (PARG) in the pathophysiology of renal ischemia/reperfusion (I/R) injury is not known. Poly(ADP-ribosyl)ation is rapidly stimulated in cells after DNA damage caused by the generation of reactive oxygen and nitrogen species during I/R. Continuous or excessive activation of poly(ADP-ribose) polymerase-1 produces extended chains of ADP-ribose on nuclear proteins and results in a substantial depletion of intracellular NAD(+) and subsequently, ATP, leading to cellular dysfunction and, ultimately, cell death. The key enzyme involved in polymer turnover is PARG, which possesses mainly exoglycosidase activity but can remove olig(ADP-ribose) fragments via endoglycosidic cleavage. Thus, the aim of this study was to investigate whether the absence of PARG(110) reduced the renal dysfunction, injury, and inflammation caused by I/R of the mouse kidney. Here, the renal dysfunction and injury caused by I/R (bilateral renal artery occlusion [30 min] followed by reperfusion [24 h]) in mice lacking PARG(110), the major nuclear isoform of PARG, was investigated. The following markers of renal dysfunction and injury were measured: Plasma urea, creatinine, aspartate aminotransferase, and histology. The following markers of inflammation were also measured: Myeloperoxidase activity, malondialdehyde levels, and plasma nitrite/nitrate. The degree of renal injury and dysfunction caused by I/R was significantly reduced in PARG(110)-deficient mice when compared with their wild-type littermates, and there were no differences in any of the biochemical parameters measured between sham-operated PARG(110)(-/-) mice and sham-operated wild-type littermates. Thus, it is proposed that endogenous PARG(110) plays a pivotal role in the pathophysiology of I/R injury of the kidney.


Asunto(s)
Glicósido Hidrolasas/genética , Isoenzimas/genética , Riñón/fisiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Animales , Aspartato Aminotransferasas/sangre , Creatinina/sangre , Glicósido Hidrolasas/metabolismo , Isoenzimas/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Nefritis/metabolismo , Nefritis/patología , Nefritis/fisiopatología , Nitratos/metabolismo , Nitritos/metabolismo , Peroxidasa/metabolismo , Daño por Reperfusión/patología , Urea/sangre
18.
J Clin Invest ; 114(8): 1072-81, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15489954

RESUMEN

Poly(ADP-ribosyl)ation is rapidly formed in cells following DNA damage and is regulated by poly(ADP-ribose) polymerase-1 (PARP-1). PARP-1 is known to be involved in various cellular processes, such as DNA repair, genomic stability, transcription, and cell death. During apoptosis, PARP-1 is cleaved by caspases to generate 89-kDa and 24-kDa fragments, a hallmark of apoptosis. This cleavage is thought to be a regulatory event for cellular death. In order to understand the biological significance of PARP-1 cleavage, we generated a PARP-1 knockin (PARP-1(KI/KI)) mouse model, in which the caspase cleavage site of PARP-1, DEVD(214), was mutated to render the protein resistant to caspases during apoptosis. While PARP-1(KI/KI) mice developed normally, they were highly resistant to endotoxic shock and to intestinal and renal ischemia-reperfusions, which were associated with reduced inflammatory responses in the target tissues and cells due to the compromised production of specific inflammatory mediators. Despite normal binding of NF-kappaB to DNA, NF-kappaB-mediated transcription activity was impaired in the presence of caspase-resistant PARP-1. This study provides a novel insight into the function of PARP-1 in inflammation and ischemia-related pathophysiologies.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Inflamación/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Daño por Reperfusión/metabolismo , Animales , Inhibidores de Caspasas , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Íleon/citología , Íleon/metabolismo , Íleon/patología , Interleucina-1/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Estrés Oxidativo , Mutación Puntual , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Recombinación Genética , Daño por Reperfusión/patología , Choque Séptico/metabolismo , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
19.
Mol Cell Biol ; 24(16): 7163-78, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15282315

RESUMEN

Poly(ADP-ribosylation) is rapidly stimulated in cells following DNA damage. This posttranslational modification is regulated by the synthesizing enzyme poly(ADP-ribose) polymerase 1 (PARP-1) and the degrading enzyme poly(ADP-ribose) glycohydrolase (PARG). Although the role of PARP-1 in response to DNA damage has been studied extensively, the function of PARG and the impact of poly(ADP-ribose) homeostasis in various cellular processes are largely unknown. Here we show that by gene targeting in embryonic stem cells and mice, we specifically deleted the 110-kDa PARG protein (PARG(110)) normally found in the nucleus and that depletion of PARG(110) severely compromised the automodification of PARP-1 in vivo. PARG(110)-deficient mice were viable and fertile, but these mice were hypersensitive to alkylating agents and ionizing radiation. In addition, these mice were susceptible to streptozotocin-induced diabetes and endotoxic shock. These data indicate that PARG(110) plays an important role in DNA damage responses and in pathological processes.


Asunto(s)
Daño del ADN , Glicósido Hidrolasas/metabolismo , Isoformas de Proteínas/metabolismo , Choque Séptico , Alquilantes/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN/efectos de los fármacos , ADN/efectos de la radiación , Diabetes Mellitus Experimental , Susceptibilidad a Enfermedades , Femenino , Rayos gamma , Marcación de Gen , Glicósido Hidrolasas/genética , Humanos , Lipopolisacáridos/administración & dosificación , Masculino , Metilnitrosourea/farmacología , Ratones , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células Madre/citología , Células Madre/fisiología , Tasa de Supervivencia
20.
Oncogene ; 23(21): 3872-82, 2004 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15021907

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP-1) is an abundant DNA end-sensing and binding molecule. Inactivation of PARP-1 by chemicals and genetic mutations slows cell proliferation, increases sister chromatid exchange (SCE), micronuclei formation and chromosome instability, and shortens telomeres. Given its affinity to DNA breaks and temporal occupation on DNA strand break sites, PARP-1 is proposed to prevent inappropriate DNA recombination. We investigated the potential role of PARP-1 in repair of DNA double-strand breaks (DSBs) and stalled replication forks. PARP-1-/- embryonic stem cells and embryonic fibroblast cells exhibited normal repair of DNA DSBs by either homologous recombination (HR) or nonhomologous end-joining (NHEJ) pathways. Inactivation of PARP-1 did not interfere with gene-targeting efficiency in ES cells. However, PARP-1-/- cells were hypersensitive to the replication damage agent hydroxyurea (HU)-induced cell death and exhibited enhanced SCE formation. Ablation of PARP-1 delayed reactivation of stalled replication forks imposed by HU and re-entry into the G2-M phase after HU release. These data indicate that PARP-1 is dispensable in HR induced by DSBs, but is involved in the repair and reactivation of stalled replication forks.


Asunto(s)
Daño del ADN , Replicación del ADN , Poli(ADP-Ribosa) Polimerasas/fisiología , Animales , Proteínas de Unión al ADN/análisis , Hidroxiurea/farmacología , Ratones , Recombinasa Rad51 , Recombinación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA