Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell ; 37(5): 705-719.e6, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32243838

ABSTRACT

While KRAS mutations are common in non-small cell lung cancer (NSCLC), effective treatments are lacking. Here, we report that half of KRAS-mutant NSCLCs aberrantly express the homeobox protein HOXC10, largely due to unappreciated defects in PRC2, which confers sensitivity to combined BET/MEK inhibitors in xenograft and PDX models. Efficacy of the combination is dependent on suppression of HOXC10 by BET inhibitors. We further show that HOXC10 regulates the expression of pre-replication complex (pre-RC) proteins in sensitive tumors. Accordingly, BET/MEK inhibitors suppress pre-RC proteins in cycling cells, triggering stalled replication, DNA damage, and death. These studies reveal a promising therapeutic strategy for KRAS-mutant NSCLCs, identify a predictive biomarker of response, and define a subset of NSCLCs with a targetable epigenetic vulnerability.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Acrylonitrile/analogs & derivatives , Acrylonitrile/pharmacology , Aniline Compounds/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proteins/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Genes Dev ; 29(5): 489-94, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25737278

ABSTRACT

Hereditary cancers derive from gene defects that often compromise DNA repair. Thus, BRCA-associated cancers are sensitive to DNA-damaging agents such as cisplatin. The efficacy of cisplatin is limited, however, by the development of resistance. One cisplatin resistance mechanism is restoration of homologous recombination (HR), which can result from BRCA reversion mutations. However, in BRCA2 mutant cancers, cisplatin resistance can occur independently of restored HR by a mechanism that remains unknown. Here we performed a genome-wide shRNA screen and found that loss of the nucleosome remodeling factor CHD4 confers cisplatin resistance. Restoration of cisplatin resistance is independent of HR but correlates with restored cell cycle progression, reduced chromosomal aberrations, and enhanced DNA damage tolerance. Suggesting clinical relevance, cisplatin-resistant clones lacking genetic reversion of BRCA2 show de novo loss of CHD4 expression in vitro. Moreover, BRCA2 mutant ovarian cancers with reduced CHD4 expression significantly correlate with shorter progression-free survival and shorter overall survival. Collectively, our findings indicate that CHD4 modulates therapeutic response in BRCA2 mutant cancer cells.


Subject(s)
Autoantigens/genetics , Drug Resistance, Neoplasm/genetics , Genes, BRCA2/physiology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Ovarian Neoplasms/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Mutation/genetics , Ovarian Neoplasms/drug therapy
3.
Cancer Res ; 74(3): 932-44, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24351291

ABSTRACT

Nucleotide excision repair (NER) is critical for the repair of DNA lesions induced by UV radiation, but its contribution in replicating cells is less clear. Here, we show that dual incision by NER endonucleases, including XPF and XPG, promotes the S-phase accumulation of the BRCA1 and Fanconi anemia-associated DNA helicase FANCJ to sites of UV-induced damage. FANCJ promotes replication protein A phosphorylation and the arrest of DNA synthesis following UV irradiation. Interaction defective mutants of FANCJ reveal that BRCA1 binding is not required for FANCJ localization, whereas interaction with the mismatch repair (MMR) protein MLH1 is essential. Correspondingly, we find that FANCJ, its direct interaction with MLH1, and the MMR protein MSH2 function in a common pathway in response to UV irradiation. FANCJ-deficient cells are not sensitive to killing by UV irradiation, yet we find that DNA mutations are significantly enhanced. Thus, we considered that FANCJ deficiency could be associated with skin cancer. Along these lines, in melanoma we found several somatic mutations in FANCJ, some of which were previously identified in hereditary breast cancer and Fanconi anemia. Given that, mutations in XPF can also lead to Fanconi anemia, we propose collaborations between Fanconi anemia, NER, and MMR are necessary to initiate checkpoint activation in replicating human cells to limit genomic instability.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , DNA Mismatch Repair , Fanconi Anemia Complementation Group Proteins/metabolism , Genomic Instability/radiation effects , Ultraviolet Rays/adverse effects , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair , DNA Replication/radiation effects , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Models, Biological , Mutation/radiation effects , Phosphorylation , Protein Transport , Replication Protein A/metabolism , S Phase/genetics , S Phase/radiation effects
4.
PLoS Genet ; 8(7): e1002786, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22792074

ABSTRACT

BRCA1 promotes DNA repair through interactions with multiple proteins, including CtIP and FANCJ (also known as BRIP1/BACH1). While CtIP facilitates DNA end resection when de-acetylated, the function of FANCJ in repair processing is less well defined. Here, we report that FANCJ is also acetylated. Preventing FANCJ acetylation at lysine 1249 does not interfere with the ability of cells to survive DNA interstrand crosslinks (ICLs). However, resistance is achieved with reduced reliance on recombination. Mechanistically, FANCJ acetylation facilitates DNA end processing required for repair and checkpoint signaling. This conclusion was based on the finding that FANCJ and its acetylation were required for robust RPA foci formation, RPA phosphorylation, and Rad51 foci formation in response to camptothecin (CPT). Furthermore, both preventing and mimicking FANCJ acetylation at lysine 1249 disrupts FANCJ function in checkpoint maintenance. Thus, we propose that the dynamic regulation of FANCJ acetylation is critical for robust DNA damage response, recombination-based processing, and ultimately checkpoint maintenance.


Subject(s)
Acetylation , Basic-Leucine Zipper Transcription Factors , DNA Damage/genetics , DNA , Fanconi Anemia Complementation Group Proteins , Lysine/metabolism , BRCA1 Protein/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , DNA/genetics , DNA/metabolism , DNA Repair , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Homologous Recombination , Humans , Mutation
5.
Future Oncol ; 7(2): 253-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21345144

ABSTRACT

It is clear that FANCJ, also known as BACH1 or BRIP1, is an essential tumor suppressor gene based on the identification of clinically relevant mutations not only in breast cancer, but also the childhood cancer syndrome, Fanconi anemia. This conclusion is further supported by the direct and functional interaction between FANCJ and the hereditary breast cancer-associated gene product BRCA1. In the absence of the FANCJ DNA helicase or its interaction with BRCA1, cells have defects in several aspects of the DNA damage response. In particular, the BRCA1-FANCJ interaction is essential for promoting error-free repair, checkpoint control and for limiting DNA damage tolerance. As the number of FANCJ clinical mutations and affected patients accumulate, it will be critical to understand whether the associated tumors resemble BRCA-associated tumors. If so, FANCJ patients could also benefit from new therapies that selectively sensitize DNA repair-defective tumors and spare healthy cells. In this article, we summarize the breast cancer-associated FANCJ mutations and discuss functional outcomes for DNA repair and tumor suppression.


Subject(s)
BRCA1 Protein/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Mutation , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , BRCA1 Protein/genetics , Basic-Leucine Zipper Transcription Factors/genetics , DNA Repair , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans
6.
Cancer Prev Res (Phila) ; 3(11): 1409-16, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20978114

ABSTRACT

Defects in MLH1, as with other mismatch repair (MMR) proteins, are the primary cause of hereditary nonpolyposis colon cancer (HNPCC). Mutations in MMR genes often disrupt mismatch repair and MMR signaling functions. However, some HNPCC-associated mutations have unknown pathogenicity. Here, we uncover an MLH1 clinical mutation with a leucine (L)-to-histidine (H) amino acid change at position 607 that ablates MLH1 binding to FANCJ. Given that a DNA helicase is not essential for mammalian MMR in vitro, we considered that loss of MLH1 binding to FANCJ could alter MMR signaling. Consistent with this hypothesis, FANCJ-deficient cells exhibit delayed MMR signaling and apoptotic responses that generate resistance to agents that induce O(6)-methylguanine lesions. Our data indicate that the delay in MMR signaling provides time for the methylguanine methyltransferase (MGMT) enzyme to reverse DNA methylation. In essence, FANCJ deficiency alters the competition between two pathways: MGMT-prosurvival versus MMR-prodeath. This outcome could explain the HNPCC familial cancers that present as microsatellite stable and with intact MMR, such as MLH(L607H). Importantly, the link between FANCJ and HNPCC provides insight toward directed therapies because loss of the FANCJ/MLH1 interaction also uniquely sensitizes cells to DNA cross-linking agents.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Mutation , Nuclear Proteins/genetics , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Cell Line, Tumor , DNA Mismatch Repair/physiology , Drug Resistance, Neoplasm , Humans , Immunoprecipitation , MutL Protein Homolog 1 , Nuclear Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...