Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4770, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839769

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic , SMARCB1 Protein , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Humans , Animals , Cell Line, Tumor , Mice , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/genetics , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Chromatin Assembly and Disassembly/genetics , Mice, Nude , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays , Promoter Regions, Genetic/genetics , Cell Proliferation/genetics , Cell Proliferation/drug effects , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology
2.
J Med Chem ; 66(1): 695-715, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36572866

ABSTRACT

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.


Subject(s)
Histone Acetyltransferases , Histones , Humans , Mice , Animals , Histones/metabolism , Histone Acetyltransferases/metabolism , Acetylation , p300-CBP Transcription Factors , E1A-Associated p300 Protein
3.
Pharmaceuticals (Basel) ; 15(2)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35215239

ABSTRACT

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.

4.
Cancer Sci ; 111(3): 774-782, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31955490

ABSTRACT

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.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Neoplasms/genetics , Humans , Immunotherapy/methods , Molecular Targeted Therapy/methods , Precision Medicine/methods
5.
Biochem Biophys Res Commun ; 522(2): 342-347, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31761322

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/deficiency , Glutathione/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Transcription Factors/deficiency , Amino Acid Transport System y+/metabolism , Animals , Ascites/metabolism , Ascites/pathology , DNA-Binding Proteins/metabolism , Female , Glutathione/metabolism , Humans , Mice, Nude , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Gynecol Oncol ; 155(3): 489-498, 2019 12.
Article in English | MEDLINE | ID: mdl-31604667

ABSTRACT

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.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Deoxycytidine/analogs & derivatives , Nuclear Proteins/deficiency , Ovarian Neoplasms/drug therapy , Transcription Factors/deficiency , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , DNA-Binding Proteins , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Female , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Random Allocation , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
7.
Cancer Cell ; 35(2): 177-190.e8, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30686770

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutathione/metabolism , Nuclear Proteins/deficiency , Ovarian Neoplasms/drug therapy , Oxidative Stress/drug effects , Quinuclidines/pharmacology , Transcription Factors/deficiency , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , DNA-Binding Proteins , Female , Glutamate-Cysteine Ligase/metabolism , HCT116 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Oncotarget ; 9(5): 6228-6237, 2018 Jan 19.
Article in English | MEDLINE | ID: mdl-29464067

ABSTRACT

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.

9.
Cancer Discov ; 6(4): 430-45, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26603525

ABSTRACT

UNLABELLED: Loss-of-function mutations in the CBP/CREBBP gene, which encodes a histone acetyltransferase (HAT), are present in a variety of human tumors, including lung, bladder, gastric, and hematopoietic cancers. Consequently, development of a molecular targeting method capable of specifically killing CBP-deficient cancer cells would greatly improve cancer therapy. Functional screening of synthetic-lethal genes in CBP-deficient cancers identified the CBP paralog p300/EP300 Ablation of p300 in CBP-knockout and CBP-deficient cancer cells induced G1-S cell-cycle arrest, followed by apoptosis. Genome-wide gene expression analysis revealed that MYC is a major factor responsible for the synthetic lethality. Indeed, p300 ablation in CBP-deficient cells caused downregulation of MYC expression via reduction of histone acetylation in its promoter, and this lethality was rescued by exogenous MYC expression. The p300-HAT inhibitor C646 specifically suppressed the growth of CBP-deficient lung and hematopoietic cancer cells in vitro and in vivo; thus p300 is a promising therapeutic target for treatment of CBP-deficient cancers. SIGNIFICANCE: Targeting synthetic-lethal partners of genes mutated in cancer holds great promise for treating patients without activating driver gene alterations. Here, we propose a "synthetic lethal-based therapeutic strategy" for CBP-deficient cancers by inhibition of the p300 HAT activity. Patients with CBP-deficient cancers could benefit from therapy using p300-HAT inhibitors.


Subject(s)
Apoptosis/genetics , CREB-Binding Protein/deficiency , E1A-Associated p300 Protein/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Synthetic Lethal Mutations , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly , Disease Models, Animal , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Histone Acetyltransferases/antagonists & inhibitors , Humans , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , RNA Interference , Transcription, Genetic
10.
Nucleic Acids Res ; 43(16): 7931-44, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26206670

ABSTRACT

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.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Histone-Lysine N-Methyltransferase/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Methyltransferases/physiology , Protein Methyltransferases/physiology , Recombinational DNA Repair , Repressor Proteins/physiology , BRCA1 Protein/metabolism , Cells, Cultured , Chromatids , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , DNA Damage , DNA End-Joining Repair , G2 Phase , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/metabolism , Repressor Proteins/antagonists & inhibitors , Tumor Suppressor p53-Binding Protein 1
11.
Sci Rep ; 5: 11305, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26065573

ABSTRACT

Carbon-ion radiotherapy (CIRT) holds promise to treat inoperable locally-advanced non-small cell lung carcinoma (NSCLC), a disease poorly controlled by standard chemoradiotherapy using X-rays. Since CIRT is an extremely limited medical resource, selection of NSCLC patients likely to benefit from it is important; however, biological predictors of response to CIRT are ill-defined. The present study investigated the association between the mutational status of EGFR and KRAS, driver genes frequently mutated in NSCLC, and the relative biological effectiveness (RBE) of carbon-ion beams over X-rays. The assessment of 15 NSCLC lines of different EGFR/KRAS mutational status and that of isogenic NSCLC lines expressing wild-type or mutant EGFR revealed that EGFR-mutant NSCLC cells, but not KRAS-mutant cells, show low RBE. This was attributable to (i) the high X-ray sensitivity of EGFR-mutant cells, since EGFR mutation is associated with a defect in non-homologous end joining, a major pathway for DNA double-strand break (DSB) repair, and (ii) the strong cell-killing effect of carbon-ion beams due to poor repair of carbon-ion beam-induced DSBs regardless of EGFR mutation status. These data highlight the potential of EGFR mutation status as a predictor of response to CIRT, i.e., CIRT may show a high therapeutic index in EGFR mutation-negative NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Heavy Ion Radiotherapy , Humans , Lung Neoplasms/radiotherapy , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
12.
PLoS One ; 9(12): e115121, 2014.
Article in English | MEDLINE | ID: mdl-25531293

ABSTRACT

BACKGROUND AND PURPOSE: To understand the mechanisms involved in the strong killing effect of carbon-ion beam irradiation on cancer cells with TP53 tumor suppressor gene deficiencies. MATERIALS AND METHODS: DNA damage responses after carbon-ion beam or X-ray irradiation in isogenic HCT116 colorectal cancer cell lines with and without TP53 (p53+/+ and p53-/-, respectively) were analyzed as follows: cell survival by clonogenic assay, cell death modes by morphologic observation of DAPI-stained nuclei, DNA double-strand breaks (DSBs) by immunostaining of phosphorylated H2AX (γH2AX), and cell cycle by flow cytometry and immunostaining of Ser10-phosphorylated histone H3. RESULTS: The p53-/- cells were more resistant than the p53+/+ cells to X-ray irradiation, while the sensitivities of the p53+/+ and p53-/- cells to carbon-ion beam irradiation were comparable. X-ray and carbon-ion beam irradiations predominantly induced apoptosis of the p53+/+ cells but not the p53-/- cells. In the p53-/- cells, carbon-ion beam irradiation, but not X-ray irradiation, markedly induced mitotic catastrophe that was associated with premature mitotic entry with harboring long-retained DSBs at 24 h post-irradiation. CONCLUSIONS: Efficient induction of mitotic catastrophe in apoptosis-resistant p53-deficient cells implies a strong cancer cell-killing effect of carbon-ion beam irradiation that is independent of the p53 status, suggesting its biological advantage over X-ray treatment.


Subject(s)
Apoptosis/radiation effects , Radiation, Ionizing , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded/radiation effects , G2 Phase Cell Cycle Checkpoints/radiation effects , HCT116 Cells , Heavy Ion Radiotherapy , Histones/metabolism , Humans , M Phase Cell Cycle Checkpoints/radiation effects , Phosphorylation , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
14.
Cancer Res ; 74(9): 2465-75, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24788099

ABSTRACT

The SWI/SNF chromatin-remodeling family contains various protein complexes, which regulate gene expression during cellular development and influence DNA damage response in an ATP- and complex-dependent manner, of which details remain elusive. Recent human genome sequencing of various cancer cells revealed frequent mutations in SWI/SNF factors, especially ARID1A, a variant subunit in the BRG1-associated factor (BAF) complex of the SWI/SNF family. We combined live-cell analysis and gene-suppression experiments to show that suppression of either ARID1A or its paralog ARID1B led to reduced nonhomologous end joining activity of DNA double-strand breaks (DSB), decreased accumulation of KU70/KU80 proteins at DSB, and sensitivity to ionizing radiation, as well as to cisplatin and UV. Thus, in contrast to transcriptional regulation, both ARID1 proteins are required for cellular resistance to various types of DNA damage, including DSB. The suppression of other SWI/SNF factors, namely SNF5, BAF60a, BAF60c, BAF155, or BAF170, exhibits a similar phenotype. Of these factors, ARID1A, ARID1B, SNF5, and BAF60c are necessary for the immediate recruitment of the ATPase subunit of the SWI/SNF complex to DSB, arguing that both ARID1 proteins facilitate the damage response of the complex. Finally, we found interdependent protein stability among the SWI/SNF factors, suggesting their direct interaction within the complex and the reason why multiple factors are frequently lost in parallel in cancer cells. Taken together, we show that cancer cells lacking in the expression of certain SWI/SNF factors, including ARID1A, are deficient in DNA repair and potentially vulnerable to DNA damage.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Humans , Protein Stability , Protein Subunits/metabolism , Radiation Tolerance , Transcription Factors/metabolism
15.
Radiother Oncol ; 111(2): 222-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24746574

ABSTRACT

BACKGROUND AND PURPOSE: Chromatin remodeling through histone modifications, including acetylation, plays an important role in the appropriate response to DNA damage induced by ionizing radiation (IR). Here we investigated the radiosensitizing effect of C646, a selective small molecule inhibitor of p300 histone acetyltransferase, and explored the underlying mechanisms. MATERIALS AND METHODS: A549, H157 and H460 human non-small cell lung carcinoma (NSCLC) cells, and HFL-III human lung fibroblasts were assessed by clonogenic survival assay. Apoptosis and necrosis were assessed by annexin V staining. Senescence was assessed by Senescence-associated ß-galactosidase staining. Mitotic catastrophe was assessed by evaluating nuclear morphology with DAPI staining. Cell cycle profiles were analyzed by flow cytometry. Protein expression was analyzed by immunoblotting. RESULTS: C646 sensitized A549, H460 and H157 cells to IR with a dose enhancement ratio at 10% surviving fraction of 1.4, 1.2 and 1.2, respectively. C646 did not radiosensitize HFL-III cells. In A549 cells, but not in HFL-III cells, C646 (i) enhanced mitotic catastrophe but not apoptosis, necrosis, or senescence after IR; (ii) increased the hyperploid cell population after IR; and (iii) suppressed the phosphorylation of CHK1 after IR. CONCLUSIONS: C646 radiosensitizes NSCLC cells by enhancing mitotic catastrophe through the abrogation of G2 checkpoint maintenance.


Subject(s)
Benzoates/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Histone Acetyltransferases/antagonists & inhibitors , Lung Neoplasms/pathology , Mitosis/drug effects , Pyrazoles/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Checkpoint Kinase 1 , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Lung Neoplasms/drug therapy , Nitrobenzenes , Protein Kinases/metabolism , Pyrazolones , Radiation, Ionizing
16.
J Thorac Oncol ; 9(5): 622-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24722152

ABSTRACT

BACKGROUND: Oncogenic RET fusion, caused by an inversion in chromosome 10, was recently identified as a driver mutation for the development of lung adenocarcinoma (LADC). Nevertheless, the molecular mechanism(s) underlying the rearrangement of the RET locus during lung carcinogenesis are unknown. METHODS: Genomic segments containing breakpoint junctions for RET fusions were cloned and analyzed by genomic polymerase chain reaction and genome capture sequencing using a next-generation sequencer to identify the mechanisms involved in DNA strand breaks and illegitimate joining of DNA ends. Of the 18 cases studied, 16 were identified by screening 671 LADC cases and two were previously published. RESULTS: Almost all (17 of 18, 94%) of the breakpoints in RET were located within a 2.0-kb region spanning exon 11 to intron 11 and no breakpoint occurred within 4 bp of any other. This suggested that as in papillary thyroid carcinoma, DNA strand breaks formed at nonspecific sites within this region trigger RET fusion. Just over half of the RET fusions in LADC (10 of 18, 56%) were caused by simple reciprocal inversion, and two DNA-repair mechanisms, namely nonhomologous end joining and break-induced replication, were deduced to have contributed to the illegitimate joining of the DNA ends. CONCLUSIONS: Oncogenic RET fusion in LADC occurs through multiple pathways and involves the illegitimate repair of DNA strand breaks through mechanisms different from those identified in papillary thyroid carcinoma, where RET fusion also functions as a driver mutation.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis/genetics , Chromosome Breakpoints , Chromosome Inversion , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-ret/genetics , Adenocarcinoma/pathology , Cytoskeletal Proteins/genetics , DNA End-Joining Repair , DNA, Neoplasm/genetics , Exons , Genetic Variation , Humans , Introns , Kinesins/genetics , Lung Neoplasms/pathology , Molecular Sequence Data
17.
Clin Cancer Res ; 20(12): 3087-93, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24727320

ABSTRACT

PURPOSE: To identify druggable oncogenic fusions in invasive mucinous adenocarcinoma (IMA) of the lung, a malignant type of lung adenocarcinoma in which KRAS mutations frequently occur. EXPERIMENTAL DESIGN: From an IMA cohort of 90 cases, consisting of 56 cases (62%) with KRAS mutations and 34 cases without (38%), we conducted whole-transcriptome sequencing of 32 IMAs, including 27 cases without KRAS mutations. We used the sequencing data to identify gene fusions, and then performed functional analyses of the fusion gene products. RESULTS: We identified oncogenic fusions that occurred mutually exclusively with KRAS mutations: CD74-NRG1, SLC3A2-NRG1, EZR-ERBB4, TRIM24-BRAF, and KIAA1468-RET. NRG1 fusions were present in 17.6% (6/34) of KRAS-negative IMAs. The CD74-NRG1 fusion activated HER2:HER3 signaling, whereas the EZR-ERBB4 and TRIM24-BRAF fusions constitutively activated the ERBB4 and BRAF kinases, respectively. Signaling pathway activation and fusion-induced anchorage-independent growth/tumorigenicity of NIH3T3 cells expressing these fusions were suppressed by tyrosine kinase inhibitors approved for clinical use. CONCLUSIONS: Oncogenic fusions act as driver mutations in IMAs without KRAS mutations, and thus represent promising therapeutic targets for the treatment of such IMAs.


Subject(s)
Adenocarcinoma, Mucinous/drug therapy , Lung Neoplasms/drug therapy , Mutation/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Aged , Animals , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins p21(ras) , Signal Transduction/drug effects
18.
J Radiat Res ; 55(4): 613-28, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24522270

ABSTRACT

Chromatin-regulating proteins represent a large class of novel targets for cancer therapy. In the context of radiotherapy, acetylation and deacetylation of histones by histone acetyltransferases (HATs) and histone deacetylases (HDACs) play important roles in the repair of DNA double-strand breaks generated by ionizing irradiation, and are therefore attractive targets for radiosensitization. Small-molecule inhibitors of HATs (garcinol, anacardic acid and curcumin) and HDACs (vorinostat, sodium butyrate and valproic acid) have been shown to sensitize cancer cells to ionizing irradiation in preclinical models, and some of these molecules are being tested in clinical trials, either alone or in combination with radiotherapy. Meanwhile, recent large-scale genome analyses have identified frequent mutations in genes encoding chromatin-regulating proteins, especially in those encoding subunits of the SWI/SNF chromatin-remodeling complex, in various human cancers. These observations have driven researchers toward development of targeted therapies against cancers carrying these mutations. DOT1L inhibition in MLL-rearranged leukemia, EZH2 inhibition in EZH2-mutant or MLL-rearranged hematologic malignancies and SNF5-deficient tumors, BRD4 inhibition in various hematologic malignancies, and BRM inhibition in BRG1-deficient tumors have demonstrated promising anti-tumor effects in preclinical models, and these strategies are currently awaiting clinical application. Overall, the data collected so far suggest that targeting chromatin-regulating proteins is a promising strategy for tomorrow's cancer therapy, including radiotherapy and molecularly targeted chemotherapy.


Subject(s)
Chromatin/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Animals , Chromatin Assembly and Disassembly , Histone Acetyltransferases/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Molecular Targeted Therapy , Mutation , Neoplasms/genetics , Radiation-Sensitizing Agents/pharmacology , Radiotherapy
19.
Mol Cell ; 53(4): 617-30, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24560272

ABSTRACT

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.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Thiolester Hydrolases/chemistry , Carrier Proteins/metabolism , Cell Death , Cell Line, Tumor , DNA-Binding Proteins/chemistry , Gene Library , Gene Silencing , HeLa Cells , Histone Chaperones , Histones/chemistry , Homologous Recombination , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/chemistry , Mutation , Nuclear Proteins/metabolism , Plasmids/metabolism , RNA, Small Interfering/metabolism , Recombination, Genetic , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin/chemistry , Ubiquitin-Protein Ligases
20.
Jpn J Clin Oncol ; 43(9): 849-55, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23904343

ABSTRACT

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.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Germ-Line Mutation , Neoplasms/genetics , Transcription Factors/genetics , Adenocarcinoma/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Renal Cell/genetics , DNA Helicases/genetics , Female , Histones/genetics , Humans , Kidney Neoplasms/genetics , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Melanoma/genetics , Nuclear Proteins/genetics , Nucleosomes/genetics , Ovarian Neoplasms/genetics , SMARCB1 Protein , Skin Neoplasms/genetics , Stomach Neoplasms/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...