Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 53(4): 617-30, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24560272

RESUMEN

DNA double-strand breaks (DSBs) are deleterious lesions that lead to genetic mutations and cell death. Protein ubiquitination mediated by the E3 ubiquitin ligase RNF8 within the regions surrounding DSBs recruits DNA DSB response (DDR) factors and induces chromatin remodeling, which supports cell survival after DNA damage. Nevertheless, the impact of RNF8-mediated ubiquitination on DNA repair remains to be elucidated. Here, we report that depletion of the deubiquitinating enzyme OTUB2 enhances RNF8-mediated ubiquitination in an early phase of the DDR and promotes faster DSB repair but suppresses homologous recombination. The rapid ubiquitination results in accelerated accumulation of 53BP1 and RAP80 at DSBs, which in turn protects DSB ends from resection in OTUB2-depleted cells. Mechanistically, OTUB2 suppresses RNF8-mediated L3MBTL1 ubiquitination and Lys 63-linked ubiquitin chain formation in a deubiquitinating activity-dependent manner. Thus, OTUB2 fine-tunes the speed of DSB-induced ubiquitination so that the appropriate DNA repair pathway is chosen.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Tioléster Hidrolasas/química , Proteínas Portadoras/metabolismo , Muerte Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/química , Biblioteca de Genes , Silenciador del Gen , Células HeLa , Chaperonas de Histonas , Histonas/química , Recombinación Homóloga , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/química , Mutación , Proteínas Nucleares/metabolismo , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Recombinación Genética , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/química , Ubiquitina-Proteína Ligasas
2.
Cancer Sci ; 111(3): 774-782, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31955490

RESUMEN

The SWI/SNF chromatin remodeling complex is composed of approximately 15 subunits, and approximately 20% of all cancers carry mutations in the genes encoding these subunits. Most of the genetic alterations in these genes are loss-of-function mutations. The identification of vulnerability based on synthetic lethality in cancers with SWI/SNF chromatin remodeling complex deficiency contributes to precision medicine. The SWI/SNF chromatin remodeling complex is involved in transcription, DNA repair, DNA replication, and chromosomal segregation. Cancers with deficiency in the SWI/SNF chromatin remodeling complex show increased vulnerability derived from the loss of these functions. Synthetic lethal targets have been identified based on vulnerabilities in the functions of the SWI/SNF chromatin remodeling complex. In this review article, we propose a precision medicine strategy using chemotherapeutic methods, such as molecular targeted therapy and immunotherapy, based on harnessing synthetic lethality in cancers with deficiency in the SWI/SNF chromatin remodeling complex.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , Neoplasias/genética , Humanos , Inmunoterapia/métodos , Terapia Molecular Dirigida/métodos , Medicina de Precisión/métodos
3.
Biochem Biophys Res Commun ; 522(2): 342-347, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31761322

RESUMEN

ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, increases the intracellular levels of glutathione (GSH) by upregulating solute carrier family 7 member 11 (SLC7A11). Diffuse-type gastric cancer is an aggressive tumor that is frequently associated with ARID1A deficiency. Here, we investigated the efficacy of GSH inhibition for the treatment of diffuse-type gastric cancer with ARID1A deficiency using ARID1A-proficient or -deficient patient-derived cells (PDCs). ARID1A-deficient PDCs were selectively sensitive to the GSH inhibitor APR-246, the GCLC inhibitor buthionine sulfoximine, and the SLC7A11 inhibitor erastin. Expression of SLC7A11, which is required for incorporation of cystine, and the basal level of GSH were lower in ARID1A-deficient than in ARID1A-proficient PDCs. Treatment with APR-246 decreased intracellular GSH levels, leading to the excessive production of reactive oxygen species (ROS), and these phenotypes are suppressed by supply of cystine and GSH compensators. Taken together, vulnerability of ARID1A-deficient gastric cancer cells to GSH inhibition is caused by decreased GSH synthesis due to diminished SLC7A11 expression. The present results suggest that GSH inhibition is a promising strategy for the treatment of diffuse-type gastric cancers with ARID1A deficiency.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Glutatión/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Factores de Transcripción/deficiencia , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Ascitis/metabolismo , Ascitis/patología , Proteínas de Unión al ADN/metabolismo , Femenino , Glutatión/metabolismo , Humanos , Ratones Desnudos , Neoplasias Gástricas/patología , Factores de Transcripción/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Gynecol Oncol ; 155(3): 489-498, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31604667

RESUMEN

OBJECTIVE: Ovarian clear cell carcinoma (OCCC) is often resistant to conventional, standard chemotherapy using cytotoxic drugs. OCCC harbors a unique genomic feature of frequent (approximately 50%) ARID1A deficiency. The present study was performed to investigate standard chemotherapeutic options suitable for ARID1A-deficient OCCC patients. METHODS: Drugs with selective toxicity to ARID1A-deficient OCCC cells were identified among six cytotoxic drugs used in standard chemotherapy for OCCC by employing multiple ARID1A-knockout cell lines and an OCCC cell line panel. Anti-tumor effects of drug treatment were assessed using a xenograft model. To obtain proof of concept in patients, seven OCCC patients who received single-agent therapy with gemcitabine were identified in a retrospective cohort of 149 OCCC patients. Patient samples and cases were analyzed for association between therapeutic response and ARID1A deficiency. RESULTS: ARID1A-knockout and ARID1A-deficient OCCC cells had selective sensitivity to gemcitabine. IC50 values for gemcitabine of ARID1A-deficient cells were significantly lower than those of ARID1A-proficient cells (p = 0.0001). Growth of OCCC xenografts with ARID1A deficiency was inhibited by administration of gemcitabine, and gemcitabine treatment effectively induced apoptosis in ARID1A-deficient OCCC cells. Three ARID1A-deficient OCCC patients had significantly longer progression-free survival after gemcitabine treatment than four ARID1A-proficient OCCC patients (p = 0.02). An ARID1A-deficient case that was resistant to multiple cytotoxic drugs, including paclitaxel plus carboplatin in the adjuvant and etoposide plus irinotecan in the first-line treatment, exhibited a dramatic response to gemcitabine in the second-line treatment. CONCLUSION: ARID1A-deficient OCCC patients could benefit from gemcitabine treatment in clinical settings.


Asunto(s)
Adenocarcinoma de Células Claras/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Proteínas Nucleares/deficiencia , Neoplasias Ováricas/tratamiento farmacológico , Factores de Transcripción/deficiencia , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Adulto , Anciano , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proteínas de Unión al ADN , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Femenino , Técnicas de Inactivación de Genes , Células HCT116 , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Distribución Aleatoria , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
5.
Mol Cell ; 40(6): 976-87, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21172662

RESUMEN

DNA double-strand breaks (DSBs) are repaired via nonhomologous end-joining (NHEJ) or homologous recombination (HR), but cellular repair processes remain elusive. We show here that the ATP-dependent chromatin-remodeling factors, ACF1 and SNF2H, accumulate rapidly at DSBs and are required for DSB repair in human cells. If the expression of ACF1 or SNF2H is suppressed, cells become extremely sensitive to X-rays and chemical treatments producing DSBs, and DSBs remain unrepaired. ACF1 interacts directly with KU70 and is required for the accumulation of KU proteins at DSBs. The KU70/80 complex becomes physically more associated with the chromatin-remodeling factors of the CHRAC complex, which includes ACF1, SNF2H, CHRAC15, and CHRAC17, after treatments producing DSBs. Furthermore, the frequency of NHEJ as well as HR induced by DSBs in chromosomal DNA is significantly decreased in cells depleted of either of these factors. Thus, ACF1 and its complexes play important roles in DSBs repair.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Factores de Transcripción/metabolismo , Antígenos Nucleares/metabolismo , Células Cultivadas , ADN Polimerasa III/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Autoantígeno Ku , Nucleoproteínas/metabolismo , Rayos Ultravioleta
6.
Nucleic Acids Res ; 43(16): 7931-44, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26206670

RESUMEN

Recent studies have shown that homologous recombination (HR) requires chromatin repression as well as relaxation at DNA double strand breaks (DSBs). HP1 and SUV39H1/2 are repressive factors essential for HR. Here, we identify SETDB1 as an additional compacting factor promoting HR. Depletion of HP1, SUV39, SETDB1 or BRCA1 confer identical phenotypes. The repressive factors, like BRCA1, are dispensable for the initiation of resection but promote the extension step causing diminished RPA or RAD51 foci and HR in irradiated G2 cells. Depletion of the compacting factors does not inhibit BRCA1 recruitment but at 8 h post IR, BRCA1 foci are smaller and aberrantly positioned compared to control cells. BRCA1 promotes 53BP1 repositioning to the periphery of enlarged foci and formation of a devoid core with BRCA1 becoming enlarged and localized internally to 53BP1. Depletion of the compacting factors precludes these changes at irradiation-induced foci. Thus, the repressive factors are required for BRCA1 function in promoting the repositioning of 53BP1 during HR. Additionally, depletion of these repressive factors in undamaged cells causes diminished sister chromatid association at centromeric sequences. We propose a model for how these findings may be functionally linked.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metiltransferasas/fisiología , Proteína Metiltransferasas/fisiología , Reparación del ADN por Recombinación , Proteínas Represoras/fisiología , Proteína BRCA1/metabolismo , Células Cultivadas , Cromátides , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Daño del ADN , Reparación del ADN por Unión de Extremidades , Fase G2 , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Metiltransferasas/antagonistas & inhibidores , Proteína Metiltransferasas/antagonistas & inhibidores , Proteína Metiltransferasas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteína 1 de Unión al Supresor Tumoral P53
7.
Nat Commun ; 15(1): 4770, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839769

RESUMEN

SMARCB1, a subunit of the SWI/SNF chromatin remodeling complex, is the causative gene of rhabdoid tumors and epithelioid sarcomas. Here, we identify a paralog pair of CBP and p300 as a synthetic lethal target in SMARCB1-deficient cancers by using a dual siRNA screening method based on the "simultaneous inhibition of a paralog pair" concept. Treatment with CBP/p300 dual inhibitors suppresses growth of cell lines and tumor xenografts derived from SMARCB1-deficient cells but not from SMARCB1-proficient cells. SMARCB1-containing SWI/SNF complexes localize with H3K27me3 and its methyltransferase EZH2 at the promotor region of the KREMEN2 locus, resulting in transcriptional downregulation of KREMEN2. By contrast, SMARCB1 deficiency leads to localization of H3K27ac, and recruitment of its acetyltransferases CBP and p300, at the KREMEN2 locus, resulting in transcriptional upregulation of KREMEN2, which cooperates with the SMARCA1 chromatin remodeling complex. Simultaneous inhibition of CBP/p300 leads to transcriptional downregulation of KREMEN2, followed by apoptosis induction via monomerization of KREMEN1 due to a failure to interact with KREMEN2, which suppresses anti-apoptotic signaling pathways. Taken together, our findings indicate that simultaneous inhibitors of CBP/p300 could be promising therapeutic agents for SMARCB1-deficient cancers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteína SMARCB1 , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Humanos , Animales , Línea Celular Tumoral , Ratones , Factores de Transcripción p300-CBP/metabolismo , Factores de Transcripción p300-CBP/genética , Proteína p300 Asociada a E1A/metabolismo , Proteína p300 Asociada a E1A/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Ensamble y Desensamble de Cromatina/genética , Ratones Desnudos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Regiones Promotoras Genéticas/genética , Proliferación Celular/genética , Proliferación Celular/efectos de los fármacos , Tumor Rabdoide/genética , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patología
8.
Carcinogenesis ; 34(11): 2486-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23825154

RESUMEN

Inhibitors of poly(ADP-ribose) polymerase (PARP) are promising anticancer drugs, particularly for the treatment of tumors deficient in the DNA damage response (DDR). However, it is challenging to design effective therapeutic strategies for use of these compounds against cancers without DDR deficiencies. In this context, combination therapies in which PARP inhibitors are used alongside DDR inhibitors have elicited a great deal of interest. Curcumin, a component of turmeric (Curcuma longa), has been tested in clinical studies for its chemosensitizing potential; however, the mechanisms of chemosensitization by curcumin have not been fully elucidated. This study demonstrates that curcumin suppresses three major DDR pathways: non-homologous end joining (NHEJ), homologous recombination (HR) and the DNA damage checkpoint. Curcumin suppresses the histone acetylation at DNA double-strand break (DSB) sites by inhibiting histone acetyltransferase activity, thereby reducing recruitment of the key NHEJ factor KU70/KU80 to DSB sites. Curcumin also suppresses HR by reducing expression of the BRCA1 gene, which regulates HR, by impairing histone acetylation at the BRCA1 promoter. Curcumin also inhibits ataxia telangiectasia and Rad3-related protein (ATR) kinase (IC50 in vitro = 493 nM), resulting in impaired activation of ATR-CHK1 signaling, which is necessary for HR and the DNA damage checkpoint pathway. Thus, curcumin suppresses three DDR pathways by inhibiting histone acetyltransferases and ATR. Concordantly, curcumin sensitizes cancer cells to PARP inhibitors by enhancing apoptosis and mitotic catastrophe via inhibition of both the DNA damage checkpoint and DSB repair. Our results indicate that curcumin is a promising sensitizer for PARP inhibitor-based therapy.


Asunto(s)
Curcumina/farmacología , Daño del ADN/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Transducción de Señal/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/metabolismo , Western Blotting , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Radioisótopos de Cobalto , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Rayos gamma , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Recombinación Homóloga/efectos de la radiación , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Regiones Promotoras Genéticas , Proteínas Quinasas/metabolismo , Sialoglicoproteínas/antagonistas & inhibidores , Sialoglicoproteínas/metabolismo , Transducción de Señal/efectos de la radiación , Células Tumorales Cultivadas , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Factores de Transcripción p300-CBP/metabolismo
9.
Jpn J Clin Oncol ; 43(9): 849-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23904343

RESUMEN

Chromosomal deoxyribonucleic acid and histone proteins form a highly condensed structure known as chromatin. Chromatin remodeling proteins regulate deoxyribonucleic acid transcription, synthesis and repair by changing nucleosomal composition in an adenosine triphosphate-dependent manner and mediate access of deoxyribonucleic acid-binding proteins to deoxyribonucleic acid double strands. Recently, large-scale genome sequencing studies identified somatic mutations in genes encoding chromatin remodeling proteins in a variety of human solid cancers. Notably, inactivating mutations in genes encoding the catalytic and regulatory subunits of the switch/sucrose non-fermenting chromatin remodeling complex have been detected in several solid cancers: sucrose non-fermenting/switch/sucrose non-fermenting-related, matrix-associated, actin-dependent regulator of chromatin, subfamily b, member 1/Brahma-related gene 1-associated factor 47/integrase interactor 1 mutations in rhabdoid tumors; AT-rich interactive domain-containing protein 1 A/Brahma-related gene 1-associated factor 250a mutations in ovarian clear cell carcinoma, hepatocellular carcinoma and gastric adenocarcinoma; polybromo 1/Brahma-related gene 1-associated factor 180 mutations in renal clear cell carcinoma; Brahma-related gene 1/switch/sucrose non-fermenting-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 mutations in non-small-cell lung carcinoma and AT-rich interactive domain-containing protein 2/Brahma-related gene 1-associated factor 200 mutations in hepatocellular carcinoma and malignant melanoma. This suggests that the switch/sucrose non-fermenting complex has a tumor-suppressive function, and that switch/sucrose non-fermenting gene deficiencies may affect the properties of cancer cells, which could be of value for the development of novel therapeutic strategies.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Mutación de Línea Germinal , Neoplasias/genética , Factores de Transcripción/genética , Adenocarcinoma/genética , Carcinoma Hepatocelular/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Renales/genética , ADN Helicasas/genética , Femenino , Histonas/genética , Humanos , Neoplasias Renales/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Melanoma/genética , Proteínas Nucleares/genética , Nucleosomas/genética , Neoplasias Ováricas/genética , Proteína SMARCB1 , Neoplasias Cutáneas/genética , Neoplasias Gástricas/genética , Relación Estructura-Actividad
10.
J Med Chem ; 66(1): 695-715, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36572866

RESUMEN

Histone acetylation is a post-translational modification of histones that is catalyzed by histone acetyltransferases (HATs) and plays an essential role in cellular processes. The HAT domain of EP300/CBP has recently emerged as a potential drug target for cancer therapy. Here, we describe the identification of the novel, highly potent, and selective EP300/CBP HAT inhibitor DS-9300. Our optimization efforts using a structure-based drug design approach based on the cocrystal structures of the EP300 HAT domain in complex with compounds 2 and 3 led to the identification of compounds possessing low-nanomolar EP300 HAT inhibitory potency and the ability to inhibit cellular acetylation of histone H3K27. Optimization of the pharmacokinetic properties in this series resulted in compounds with excellent oral systemic exposure, and once-daily oral administration of 16 (DS-9300) demonstrated potent antitumor effects in a castrated VCaP xenograft mouse model without significant body weight loss.


Asunto(s)
Histona Acetiltransferasas , Histonas , Humanos , Ratones , Animales , Histonas/metabolismo , Histona Acetiltransferasas/metabolismo , Acetilación , Factores de Transcripción p300-CBP , Proteína p300 Asociada a E1A
11.
J Biol Chem ; 286(42): 36368-77, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21885877

RESUMEN

Polynucleotide kinase and aprataxin-like forkhead-associated protein (PALF, also called aprataxin- and PNK-like factor (APLF)) has been shown to have nuclease activity and to use its forkhead-associated domain to bind to x-ray repair complementing defective repair in Chinese hamster cells 4 (XRCC4). Because XRCC4 is a key component of the ligase IV complex that is central to the nonhomologous DNA end joining (NHEJ) pathway, this raises the possibility that PALF might play a role in NHEJ. For this reason, we further studied the nucleolytic properties of PALF, and we searched for any modulation of PALF by NHEJ components. We verified that PALF has 3' exonuclease activity. However, PALF also possesses single-stranded DNA endonuclease activity. This single-stranded DNA endonuclease activity can act at all single-stranded sites except those within four nucleotides 3' of a double-stranded DNA junction, suggesting that PALF minimally requires approximately four nucleotides of single-strandedness. Ku, DNA-dependent protein kinase catalytic subunit, and XRCC4-DNA ligase IV do not modulate PALF nuclease activity on single-stranded DNA or overhangs of duplex substrates. PALF does not open DNA hairpins. However, in a reconstituted end joining assay that includes Ku, XRCC4-DNA ligase IV, and PALF, PALF is able to resect 3' overhanging nucleotides and permit XRCC4-DNA ligase IV to complete the joining process in a manner that is as efficient as Artemis. Reduction of PALF in vivo reduces the joining of incompatible DNA ends. Hence, PALF can function in concert with other NHEJ proteins.


Asunto(s)
ADN Helicasas/metabolismo , Reparación del ADN/fisiología , ADN de Cadena Simple/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Proteínas de Unión al ADN/metabolismo , Exonucleasas/metabolismo , Animales , Línea Celular , Cricetinae , Cricetulus , ADN Helicasas/química , ADN Helicasas/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Exonucleasas/química , Exonucleasas/genética , Humanos , Autoantígeno Ku , Proteínas de Unión a Poli-ADP-Ribosa
12.
Pharmaceuticals (Basel) ; 15(2)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35215239

RESUMEN

In the field of drug repurposing, the use of statins for treating dyslipidemia is considered promising in ovarian cancer treatment based on epidemiological studies and basic research findings. Biomarkers should be established to identify patients who will respond to statin treatment to achieve clinical application. In the present study, we demonstrated that statins have a multifaceted mode of action in ovarian cancer and involve pathways other than protein prenylation. To identify biomarkers that predict the response to statins, we subjected ovarian cancer cells to microarray analysis and calculated Pearson's correlation coefficients between gene expression and cell survival after statin treatment. The results showed that VDAC1 and LDLRAP1 were positively and negatively correlated with the response to statins, respectively. Histoculture drug response assays revealed that statins were effective in clinical samples. We also confirmed the synergistic effects of statins with paclitaxel and panobinostat and determined that statins are hematologically safe to administer to statin-treated mice. Future clinical trials based on the expression of the biomarkers identified in this study for repurposing statins for ovarian cancer treatment are warranted.

13.
Genes Chromosomes Cancer ; 49(4): 342-52, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20073072

RESUMEN

A total of 176 genes homozygously deleted in human lung cancer were identified by DNA array-based whole genome scanning of 52 lung cancer cell lines and subsequent genomic PCR in 74 cell lines, including the 52 cell lines scanned. One or more exons of these genes were homozygously deleted in one (1%) to 20 (27%) cell lines. These genes included known tumor suppressor genes, e.g., CDKN2A/p16, RB1, and SMAD4, and candidate tumor suppressor genes whose hemizygous or homozygous deletions were reported in several types of human cancers, such as FHIT, KEAP1, and LRP1B/LRP-DIP. CDKN2A/p16 and p14ARF located in 9p21 were most frequently deleted (20/74, 27%). The PTPRD gene was most frequently deleted (8/74, 11%) among genes mapping to regions other than 9p21. Somatic mutations, including a nonsense mutation, of the PTPRD gene were detected in 8/74 (11%) of cell lines and 4/95 (4%) of surgical specimens of lung cancer. Reduced PTPRD expression was observed in the majority (>80%) of cell lines and surgical specimens of lung cancer. Therefore, PTPRD is a candidate tumor suppressor gene in lung cancer. Microarray-based expression profiling of 19 lung cancer cell lines also indicated that some of the 176 genes, such as KANK and ADAMTS1, are preferentially inactivated by epigenetic alterations. Genetic/epigenetic as well as functional studies of these 176 genes will increase our understanding of molecular mechanisms behind lung carcinogenesis.


Asunto(s)
Eliminación de Gen , Genes Supresores de Tumor , Neoplasias Pulmonares/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Línea Celular Tumoral , Hibridación Genómica Comparativa , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Homocigoto , Humanos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados
14.
DNA Repair (Amst) ; 7(6): 879-89, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18396468

RESUMEN

The protein Rad52 is a key player in various types of homologous recombination and is essential to maintenance of genomic integrity. Although evidence indicates that Rad52 is modified by SUMO, the physiological relevance of this sumoylation remains unclear. Here, we identify the conditions under which Rad52 sumoylation is induced, and clarify the role of this modification in homologous recombination. Oligomerization of Rad52 was a prerequisite for sumoylation, and the modification occurred in the cell proceeding S phase being exposed to the DNA-damaging agent methyl methanesulfonate (MMS). Following exposure to MMS, sumoylated Rad52 accumulated in rad51 cells, but not in the recombination-related gene mutants, rad54, rad55, rad59, sgs1, or srs2. The accumulation of sumoylated Rad52 was suppressed in rad51 cells expressing Rad51-K191R, an ATPase-defective protein presumed to be recruited to ssDNA. Although the sumoylation defective mutant rad52-3KR (K10R/K11R/K220R) showed no defect in mating-type switching, which did not lead to Rad52 sumoylation in wild-type cells, the mutant did demonstrate a partial defect in MMS-induced interchromosomal homologous recombination.


Asunto(s)
Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Secuencia de Bases , ADN/efectos de los fármacos , Cartilla de ADN , Metilmetanosulfonato/farmacología , Mutagénesis Sitio-Dirigida , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteínas de Saccharomyces cerevisiae/genética
15.
Nucleic Acids Res ; 35(2): 353-62, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17170004

RESUMEN

The inaccurate repair of DNA double-strand breaks (DSBs) can result in genomic instability, and additionally cell death or the development of cancer. Elg1, which forms an alternative RFC-like complex with RFC2-5, is required for the maintenance of genome stability in Saccharomyces cerevisiae, and its function has been linked to DNA replication or damage checkpoint response. Here, we show that Elg1 is involved in homologous recombination (HR)-mediated DSB repair. Mutants of elg1 were partially defective in HR induced by methylmethanesufonate (MMS) and phleomycin. Deletion of ELG1 resulted in less efficient repair of phleomycin-induced DSBs in G2/M phase-arrested cells. During HR between MAT and HML loci, Elg1 associated with both the MAT locus near the HO endonuclease-induced DSB site, and the HML homologous donor locus. The association of Elg1 with the MAT locus was not dependent on Rad52. However, Elg1 association with the HML locus depended on Rad52. Importantly, we found that two of the later steps in HR-mediated repair of an HO endonuclease-induced DSB, primer extension after strand invasion and ligation, were less efficient in elg1 mutants. Our results suggest that Elg1 is involved in DSB repair by HR.


Asunto(s)
Proteínas Portadoras/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas Portadoras/genética , Ciclo Celular , Genes del Tipo Sexual de los Hongos , Mutación , Fleomicinas/toxicidad , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Recombinación Genética , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae
16.
Nucleic Acids Res ; 35(15): 4989-5000, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17636314

RESUMEN

The efficient repair of double-strand breaks (DSBs) is crucial in maintaining genomic integrity. Sister chromatid cohesion is important for not only faithful chromosome segregation but also for proper DSB repair. During DSB repair, the Smc1-Smc3 cohesin complex is loaded onto chromatin around the DSB to support recombination-mediated DSB repair. In this study, we investigated whether Ctf18, a factor implicated in the establishment of sister chromatid cohesion, is involved in DSB repair in budding yeast. Ctf18 was recruited to HO-endonuclease induced DSB sites in an Mre11-dependent manner and to damaged chromatin in G2/M phase-arrested cells. The ctf18 mutant cells showed high sensitivity to DSB-inducible genotoxic agents and defects in DSB repair, as well as defects in damage-induced recombination between sister chromatids and between homologous chromosomes. These results suggest that Ctf18 is involved in damage-induced homologous recombination.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona , Replicación del ADN , Endodesoxirribonucleasas/fisiología , Exodesoxirribonucleasas/fisiología , Eliminación de Gen , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Nucleic Acids Res ; 35(9): 3109-17, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17452364

RESUMEN

The actin-related proteins (Arps) comprise a conserved protein family. Arp4p is found in large multisubunits of the INO80 and SWR1 chromatin remodeling complexes and in the NuA4 histone acetyltransferase complex. Here we show that arp4 (arp4S23A/D159A) temperature-sensitive cells are defective in G2/M phase function. arp4 mutants are sensitive to the microtubule depolymerizing agent benomyl and arrest at G2/M phase at restrictive temperature. Arp4p is associated with centromeric and telomeric regions throughout cell cycle. Ino80p, Esa1p and Swr1p, components of the INO80, NuA4 and SWR1 complexes, respectively, also associate with centromeres. The association of many kinetochore components including Cse4p, a component of the centromere nucleosome, Mtw1p and Ctf3p is partially impaired in arp4 cells, suggesting that the G2/M arrest of arp4 mutant cells is due to a defect in formation of the chromosomal segregation apparatus.


Asunto(s)
Actinas/fisiología , Ciclo Celular , Cinetocoros/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Actinas/análisis , Actinas/genética , Sitios de Unión , Ciclo Celular/efectos de los fármacos , División Celular , Fase G2 , Genómica , Mutación , Nocodazol/farmacología , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/análisis , Proteínas de Saccharomyces cerevisiae/genética
18.
Cancer Cell ; 35(2): 177-190.e8, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30686770

RESUMEN

ARID1A encodes an SWI/SNF chromatin-remodeling factor and is frequently mutated in various cancers. This study demonstrates that ARID1A-deficient cancer cells are specifically vulnerable to inhibition of the antioxidant glutathione (GSH) and the glutamate-cysteine ligase synthetase catalytic subunit (GCLC), a rate-limiting enzyme for GSH synthesis. Inhibition of GCLC markedly decreased GSH in ARID1A-deficient cancer cells, leading to apoptotic cell death triggered by excessive amounts of reactive oxygen species. The vulnerability of ARID1A-deficient cancer cells results from low basal levels of GSH due to impaired expression of SLC7A11. The SLC7A11-encoded cystine transporter supplies cells with cysteine, a key source of GSH, and its expression is enhanced by ARID1A-mediated chromatin remodeling. Thus, ARID1A-deficient cancers are susceptible to synthetic lethal targeting of GCLC.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutatión/metabolismo , Proteínas Nucleares/deficiencia , Neoplasias Ováricas/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Quinuclidinas/farmacología , Factores de Transcripción/deficiencia , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Proteínas de Unión al ADN , Femenino , Glutamato-Cisteína Ligasa/metabolismo , Células HCT116 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Factores de Transcripción/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nucleic Acids Res ; 34(11): 3389-98, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16840526

RESUMEN

Dpb11 is required for the loading of DNA polymerases alpha and epsilon on to DNA in chromosomal DNA replication and interacts with the DNA damage checkpoint protein Ddc1 in Saccharomyces cerevisiae. The interaction between the homologs of Dpb11 and Ddc1 in human cells and fission yeast is thought to reflect their involvement in the checkpoint response. Here we show that dpb11-1 cells, carrying a mutated Dpb11 that cannot interact with Ddc1, are defective in the repair of methyl methanesulfonate (MMS)-induced DNA damage but not in the DNA damage checkpoint at the permissive temperature. Epistatic analyses suggested that Dpb11 is involved in the Rad51/Rad52-dependent recombination pathway. Ddc1 as well as Dpb11 were required for homologous recombination induced by MMS. Moreover, we found the in vivo association of Dpb11 and Ddc1 with not only the HO-induced double-strand break (DSB) site at MAT locus but also the donor sequence HML during homologous recombination between MAT and HML. Rad51 was required for their association with the HML donor locus, but not with DSB site at the MAT locus. In addition, the association of Dpb11 with the MAT and HML locus after induction of HO-induced DSB was dependent on Ddc1. These results indicate that, besides the involvement in the replication and checkpoint, Dpb11 functions with Ddc1 in the recombination repair process itself.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Reparación del ADN , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/genética , Daño del ADN , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Metilmetanosulfonato/toxicidad , Mutación , Fosfoproteínas/fisiología , Recombinasa Rad51/fisiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
20.
Oncotarget ; 9(5): 6228-6237, 2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29464067

RESUMEN

There has been little improvement in the prognosis for adolescent and young adult (AYA) tumor patients. Hence, there is an urgent need to understand the etiology of tumor development and identify actionable gene aberrations to improve prevention and therapy. Here, 76 sporadic tumors (48 breast, 22 ovarian, and six uterine) from 76 AYA females (age range, 25-39 years) were subjected to whole exome and RNA sequencing to determine their mutational signatures and actionable gene profiles. Two individuals with breast cancer (4.2% of cases) and one with ovarian cancer (5.3% of cases) carried germline BRCA2 mutations. The two cases with breast tumors also each carried an additional deleterious germline mutation: one in TP53 and the other in CHEK2. Mutational signature analysis of the 76 tumors indicated that spontaneous deamination of 5-methylcytosine and activity of the APOBEC cytidine deaminase protein family are major causes of mutagenesis. In addition, 18 breast or ovarian tumors (18/70, 26%), including the three cases with germline BRCA2 mutations, exhibited a predominant "BRCAness" mutational signature, an indicator of functional BRCA1/BRCA2 deficiency. Actionable aberrations and high tumor mutation burdens were detected in 24 breast (50%), 17 ovarian (77%), and five uterine (83%) tumor cases. Thus, mutational processes and aberrant genes in AYA tumors are largely shared with those identified in non-AYA tumors. The efficacy of molecular targeting and immune checkpoint inhibitory therapies should be explored for both AYA and non-AYA patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA