Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607954

ABSTRACT

BRCA1 germline mutations are associated with an increased risk of breast and ovarian cancer. Recent findings of others suggest that BRCA1 mutation carriers also bear an increased risk of esophageal and gastric cancer. Here, we employ a Brca1/Trp53 mouse model to show that unresolved replication stress (RS) in BRCA1 heterozygous cells drives esophageal tumorigenesis in a model of the human equivalent. This model employs 4-nitroquinoline-1-oxide (4NQO) as an RS-inducing agent. Upon drinking 4NQO-containing water, Brca1 heterozygous mice formed squamous cell carcinomas of the distal esophagus and forestomach at a much higher frequency and speed (∼90 to 120 d) than did wild-type (WT) mice, which remained largely tumor free. Their esophageal tissue, but not that of WT control mice, revealed evidence of overt RS as reflected by intracellular CHK1 phosphorylation and 53BP1 staining. These Brca1 mutant tumors also revealed higher genome mutation rates than those of control animals; the mutational signature SBS4, which is associated with tobacco-induced tumorigenesis; and a loss of Brca1 heterozygosity (LOH). This uniquely accelerated Brca1 tumor model is also relevant to human esophageal squamous cell carcinoma, an often lethal tumor.


Subject(s)
BRCA1 Protein/genetics , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Loss of Heterozygosity/genetics , Tumor Suppressor Protein p53/genetics , 4-Nitroquinoline-1-oxide/toxicity , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Checkpoint Kinase 1/metabolism , Disease Models, Animal , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/chemically induced , Esophageal Squamous Cell Carcinoma/pathology , Female , Germ-Line Mutation/genetics , Heterozygote , Humans , Loss of Heterozygosity/drug effects , Male , Mice , Mice, Knockout , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Nat Commun ; 12(1): 3542, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112789

ABSTRACT

R-loop structures act as modulators of physiological processes such as transcription termination, gene regulation, and DNA repair. However, they can cause transcription-replication conflicts and give rise to genomic instability, particularly at telomeres, which are prone to forming DNA secondary structures. Here, we demonstrate that BRCA1 binds TERRA RNA, directly and physically via its N-terminal nuclear localization sequence, as well as telomere-specific shelterin proteins in an R-loop-, and a cell cycle-dependent manner. R-loop-driven BRCA1 binding to CpG-rich TERRA promoters represses TERRA transcription, prevents TERRA R-loop-associated damage, and promotes its repair, likely in association with SETX and XRN2. BRCA1 depletion upregulates TERRA expression, leading to overly abundant TERRA R-loops, telomeric replication stress, and signs of telomeric aberrancy. Moreover, BRCA1 mutations within the TERRA-binding region lead to an excess of TERRA-associated R-loops and telomeric abnormalities. Thus, normal BRCA1/TERRA binding suppresses telomere-centered genome instability.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage/genetics , R-Loop Structures , RNA, Long Noncoding/metabolism , Telomere/metabolism , BRCA1 Protein/genetics , Cell Cycle/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Liquid , CpG Islands , DNA Helicases/metabolism , Exoribonucleases/metabolism , Humans , In Situ Hybridization, Fluorescence , Mass Spectrometry , Multifunctional Enzymes/metabolism , Mutation , Promoter Regions, Genetic , Protein Binding , R-Loop Structures/genetics , RNA Helicases/metabolism , RNA, Long Noncoding/genetics , RNA, Small Interfering , Telomere/genetics
3.
Nature ; 591(7851): 665-670, 2021 03.
Article in English | MEDLINE | ID: mdl-33536619

ABSTRACT

Strong connections exist between R-loops (three-stranded structures harbouring an RNA:DNA hybrid and a displaced single-strand DNA), genome instability and human disease1-5. Indeed, R-loops are favoured in relevant genomic regions as regulators of certain physiological processes through which homeostasis is typically maintained. For example, transcription termination pause sites regulated by R-loops can induce the synthesis of antisense transcripts that enable the formation of local, RNA interference (RNAi)-driven heterochromation6. Pause sites are also protected against endogenous single-stranded DNA breaks by BRCA17. Hypotheses about how DNA repair is enacted at pause sites include a role for RNA, which is emerging as a normal, albeit unexplained, regulator of genome integrity8. Here we report that a species of single-stranded, DNA-damage-associated small RNA (sdRNA) is generated by a BRCA1-RNAi protein complex. sdRNAs promote DNA repair driven by the PALB2-RAD52 complex at transcriptional termination pause sites that form R-loops and are rich in single-stranded DNA breaks. sdRNA repair operates in both quiescent (G0) and proliferating cells. Thus, sdRNA repair can occur in intact tissue and/or stem cells, and may contribute to tumour suppression mediated by BRCA1.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Fanconi Anemia Complementation Group N Protein/metabolism , RNA Interference , Rad52 DNA Repair and Recombination Protein/metabolism , Argonaute Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , Eukaryotic Initiation Factors/metabolism , HeLa Cells , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Resting Phase, Cell Cycle , Ribonuclease III/metabolism
4.
Cancer Cell ; 37(2): 139-140, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32049041

ABSTRACT

PARP inhibition (PARPi) kills tumor cells defective in homologous recombination-based repair (HR-) but not their HR+ competent counterparts. In this issue of Cancer Cell, it is shown that, when EZH2 is functionally silenced, HR+, CARM1-high, high-grade serous ovarian cancer cells become PARPi sensitive, undergo mitotic catastrophe, and die.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Enhancer of Zeste Homolog 2 Protein , Female , Homologous Recombination , Humans , Protein-Arginine N-Methyltransferases
5.
Proc Natl Acad Sci U S A ; 117(4): 2084-2091, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31932421

ABSTRACT

BRCA1 promotes error-free, homologous recombination-mediated repair (HRR) of DNA double-stranded breaks (DSBs). When excessive and uncontrolled, BRCA1 HRR activity promotes illegitimate recombination and genome disorder. We and others have observed that the BRCA1-associated protein RAP80 recruits BRCA1 to postdamage nuclear foci, and these chromatin structures then restrict the amplitude of BRCA1-driven HRR. What remains unclear is how this process is regulated. Here we report that both BRCA1 poly-ADP ribosylation (PARsylation) and the presence of BRCA1-bound RAP80 are critical for the normal interaction of BRCA1 with some of its partners (e.g., CtIP and BACH1) that are also known components of the aforementioned focal structures. Surprisingly, the simultaneous loss of RAP80 and failure therein of BRCA1 PARsylation results in the dysregulated accumulation in these foci of BRCA1 complexes. This in turn is associated with the intracellular development of a state of hyper-recombination and gross chromosomal disorder. Thus, physiological RAP80-BRCA1 complex formation and BRCA1 PARsylation contribute to the kinetics by which BRCA1 HRR-sustaining complexes normally concentrate in nuclear foci. These events likely contribute to aneuploidy suppression.


Subject(s)
BRCA1 Protein/metabolism , DNA-Binding Proteins/metabolism , Histone Chaperones/metabolism , Recombinational DNA Repair , BRCA1 Protein/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chromosomes/genetics , Chromosomes/metabolism , DNA Damage , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Histone Chaperones/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly ADP Ribosylation , Protein Binding
6.
Cell ; 178(1): 135-151.e19, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31251913

ABSTRACT

Loss of BRCA1 p220 function often results in basal-like breast cancer (BLBC), but the underlying disease mechanism is largely opaque. In mammary epithelial cells (MECs), BRCA1 interacts with multiple proteins, including NUMB and HES1, to form complexes that participate in interstrand crosslink (ICL) DNA repair and MEC differentiation control. Unrepaired ICL damage results in aberrant transdifferentiation to a mesenchymal state of cultured, human basal-like MECs and to a basal/mesenchymal state in primary mouse luminal MECs. Loss of BRCA1, NUMB, or HES1 or chemically induced ICL damage in primary murine luminal MECs results in persistent DNA damage that triggers luminal to basal/mesenchymal transdifferentiation. In vivo single-cell analysis revealed a time-dependent evolution from normal luminal MECs to luminal progenitor-like tumor cells with basal/mesenchymal transdifferentiation during murine BRCA1 BLBC development. Growing DNA damage accompanied this malignant transformation.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Cell Transdifferentiation/genetics , DNA Damage/genetics , DNA Repair/genetics , Mammary Glands, Animal/pathology , Animals , BRCA1 Protein/metabolism , Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Transformation, Neoplastic , Disease Models, Animal , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , MCF-7 Cells , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Transcription Factor HES-1/metabolism , Transfection
7.
Proc Natl Acad Sci U S A ; 115(41): E9600-E9609, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30254159

ABSTRACT

BRCA1 is an established breast and ovarian tumor suppressor gene that encodes multiple protein products whose individual contributions to human cancer suppression are poorly understood. BRCA1-IRIS (also known as "IRIS"), an alternatively spliced BRCA1 product and a chromatin-bound replication and transcription regulator, is overexpressed in various primary human cancers, including breast cancer, lung cancer, acute myeloid leukemia, and certain other carcinomas. Its naturally occurring overexpression can promote the metastasis of patient-derived xenograft (PDX) cells and other human cancer cells in mouse models. The IRIS-driven metastatic mechanism results from IRIS-dependent suppression of phosphatase and tensin homolog (PTEN) transcription, which in turn perturbs the PI3K/AKT/GSK-3ß pathway leading to prolyl hydroxylase-independent HIF-1α stabilization and activation in a normoxic environment. Thus, despite the tumor-suppressing genetic origin of IRIS, its properties more closely resemble those of an oncoprotein that, when spontaneously overexpressed, can, paradoxically, drive human tumor progression.


Subject(s)
Alternative Splicing , BRCA1 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , BRCA1 Protein/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Neoplasms/genetics , Neoplasms/pathology , PTEN Phosphohydrolase/genetics
8.
Clin Cancer Res ; 23(5): 1263-1273, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27573169

ABSTRACT

Purpose: Ovarian cancer is the leading cause of death from gynecologic malignancy in the United States, with high rates of recurrence and eventual resistance to cytotoxic chemotherapy. Model systems that allow for accurate and reproducible target discovery and validation are needed to support further drug development in this disease.Experimental Design: Clinically annotated patient-derived xenograft (PDX) models were generated from tumor cells isolated from the ascites or pleural fluid of patients undergoing clinical procedures. Models were characterized by IHC and by molecular analyses. Each PDX was luciferized to allow for reproducible in vivo assessment of intraperitoneal tumor burden by bioluminescence imaging (BLI). Plasma assays for CA125 and human LINE-1 were developed as secondary tests of in vivo disease burden.Results: Fourteen clinically annotated and molecularly characterized luciferized ovarian PDX models were generated. Luciferized PDX models retain fidelity to both the nonluciferized PDX and the original patient tumor, as demonstrated by IHC, array CGH, and targeted and whole-exome sequencing analyses. Models demonstrated diversity in specific genetic alterations and activation of PI3K signaling pathway members. Response of luciferized PDX models to standard-of-care therapy could be reproducibly monitored by BLI or plasma markers.Conclusions: We describe the establishment of a collection of 14 clinically annotated and molecularly characterized luciferized ovarian PDX models in which orthotopic tumor burden in the intraperitoneal space can be followed by standard and reproducible methods. This collection is well suited as a platform for proof-of-concept efficacy and biomarker studies and for validation of novel therapeutic strategies in ovarian cancer. Clin Cancer Res; 23(5); 1263-73. ©2016 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Xenograft Model Antitumor Assays/methods , Animals , Ascites/pathology , CA-125 Antigen/blood , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Disease Models, Animal , Humans , Long Interspersed Nucleotide Elements/genetics , Membrane Proteins/blood , Mice , Neoplasms, Glandular and Epithelial/blood , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Signal Transduction/drug effects
9.
Proc Natl Acad Sci U S A ; 113(48): E7701-E7709, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849576

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron dysfunction disease that leads to paralysis and death. There is currently no established molecular pathogenesis pathway. Multiple proteins involved in RNA processing are linked to ALS, including FUS and TDP43, and we propose a disease mechanism in which loss of function of at least one of these proteins leads to an accumulation of transcription-associated DNA damage contributing to motor neuron cell death and progressive neurological symptoms. In support of this hypothesis, we find that FUS or TDP43 depletion leads to increased sensitivity to a transcription-arresting agent due to increased DNA damage. Thus, these proteins normally contribute to the prevention or repair of transcription-associated DNA damage. In addition, both FUS and TDP43 colocalize with active RNA polymerase II at sites of DNA damage along with the DNA damage repair protein, BRCA1, and FUS and TDP43 participate in the prevention or repair of R loop-associated DNA damage, a manifestation of aberrant transcription and/or RNA processing. Gaining a better understanding of the role(s) that FUS and TDP43 play in transcription-associated DNA damage could shed light on the mechanisms underlying ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , RNA-Binding Protein FUS/physiology , Cell Line , Humans , Motor Neurons/metabolism , Protein Transport , Transcription, Genetic
10.
Mol Cell ; 63(2): 277-292, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27373334

ABSTRACT

An abnormal differentiation state is common in BRCA1-deficient mammary epithelial cells, but the underlying mechanism is unclear. Here, we report a convergence between DNA repair and normal, cultured human mammary epithelial (HME) cell differentiation. Surprisingly, depleting BRCA1 or FANCD2 (Fanconi anemia [FA] proteins) or BRG1, a mSWI/SNF subunit, caused HME cells to undergo spontaneous epithelial-to-mesenchymal transition (EMT) and aberrant differentiation. This also occurred when wild-type HMEs were exposed to chemicals that generate DNA interstrand crosslinks (repaired by FA proteins), but not in response to double-strand breaks. Suppressed expression of ΔNP63 also occurred in each of these settings, an effect that links DNA damage to the aberrant differentiation outcome. Taken together with somatic breast cancer genome data, these results point to a breakdown in a BRCA/FA-mSWI/SNF-ΔNP63-mediated DNA repair and differentiation maintenance process in mammary epithelial cells that may contribute to sporadic breast cancer development.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/prevention & control , Cell Differentiation , DNA Damage , DNA Helicases/metabolism , DNA Repair , Epithelial Cells/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Mammary Glands, Human/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetaldehyde/pharmacology , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cisplatin/pharmacology , DNA Helicases/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Formaldehyde/pharmacology , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mutation , Nuclear Proteins/genetics , Phenotype , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Cancer Res ; 75(7): 1177-80, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25788695

ABSTRACT

This symposium was held in Trento, Italy, on June 19-21, 2014, and was focused on advances in biology, physiology, and pathology of neoplasms affected by hormones, especially breast and prostate cancers. The stem cell function, the genetic and epigenetic interactions with hormones, the mechanisms of estrogen receptor transcription, biochemical markers and therapeutic targets in breast cancer, promotion of breast cancer carcinogenesis by progesterone, the basis for prostate cancer progression and the relevance of DNA repair processes, androgen receptor programming during prostate carcinogenesis, the metabolic stress role in tumor survival, and the diagnostic use of imaging in prostate cancer were discussed.


Subject(s)
Breast Neoplasms/metabolism , Hormones/physiology , Prostatic Neoplasms/metabolism , Animals , Female , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Translational Research, Biomedical
12.
Mol Cell ; 57(4): 636-647, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25699710

ABSTRACT

The mechanisms contributing to transcription-associated genomic instability are both complex and incompletely understood. Although R-loops are normal transcriptional intermediates, they are also associated with genomic instability. Here, we show that BRCA1 is recruited to R-loops that form normally over a subset of transcription termination regions. There it mediates the recruitment of a specific, physiological binding partner, senataxin (SETX). Disruption of this complex led to R-loop-driven DNA damage at those loci as reflected by adjacent γ-H2AX accumulation and ssDNA breaks within the untranscribed strand of relevant R-loop structures. Genome-wide analysis revealed widespread BRCA1 binding enrichment at R-loop-rich termination regions (TRs) of actively transcribed genes. Strikingly, within some of these genes in BRCA1 null breast tumors, there are specific insertion/deletion mutations located close to R-loop-mediated BRCA1 binding sites within TRs. Thus, BRCA1/SETX complexes support a DNA repair mechanism that addresses R-loop-based DNA damage at transcriptional pause sites.


Subject(s)
BRCA1 Protein/physiology , DNA Repair , Models, Genetic , RNA Helicases/physiology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Damage , DNA Helicases , HeLa Cells , Humans , Multifunctional Enzymes , RNA Helicases/genetics , RNA Helicases/metabolism , Transcription Termination, Genetic , Transcription, Genetic
13.
J Virol ; 89(5): 2857-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25540383

ABSTRACT

UNLABELLED: Many of the small DNA tumor viruses encode transforming proteins that function by targeting critical cellular pathways involved in cell proliferation and survival. In this study, we have examined whether some of the functions of the polyomavirus small T antigens (ST) are shared by the E6 and E7 oncoproteins of two oncogenic papillomaviruses. Using three different assays, we have found that E7 can provide some simian virus 40 (SV40) or murine polyomavirus (PyV) ST functions. Both human papillomavirus 16 (HPV16) and bovine papillomavirus (BPV1) E7 proteins are capable of partially substituting for SV40 ST in a transformation assay that also includes SV40 large T antigen, the catalytic subunit of cellular telomerase, and oncogenic Ras. Like SV40 ST, HPV16 E7 has the ability to override a quiescence block induced by mitogen deprivation. Like PyV ST, it also has the ability to inhibit myoblast differentiation. At least two of these activities are dependent upon the interaction of HPV16 E7 with retinoblastoma protein family members. For small T antigens, interaction with PP2A is needed for each of these functions. Even though there is no strong evidence that E6 or E7 share the ability of small T to interact with PP2A, E7 provides these functions related to cellular transformation. IMPORTANCE: DNA tumor viruses have provided major insights into how cancers develop. Some viruses, like the human papillomaviruses, can cause cancer directly. Both the papillomaviruses and the polyomaviruses have served as tools for understanding pathways that are often perturbed in cancer. Here, we have compared the functions of transforming proteins from several DNA tumor viruses, including two papillomaviruses and two polyomaviruses. We tested the papillomavirus E6 and E7 oncoproteins in three functional assays and found that E7 can provide some or all of the functions of the SV40 small T antigen, another well-characterized oncoprotein, in two of these assays. In a third assay, papillomavirus E7 has the same effect as the murine polyomavirus small T protein. In summary, we report several new functions for the papillomavirus E7 proteins, which will contribute new insights into the roles of viruses in cancer and the cellular pathways they perturb in carcinogenesis.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Viral , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Genetic Complementation Test , Human papillomavirus 16/genetics , Human papillomavirus 16/physiology , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Simian virus 40/genetics , Simian virus 40/physiology
14.
Proc Natl Acad Sci U S A ; 112(1): 232-7, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535366

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is the most common and aggressive form of epithelial ovarian cancer, for which few targeted therapies exist. To search for new therapeutic target proteins, we performed an in vivo shRNA screen using an established human HGSOC cell line growing either subcutaneously or intraperitoneally in immunocompromised mice. We identified genes previously implicated in ovarian cancer such as AURKA1, ERBB3, CDK2, and mTOR, as well as several novel candidates including BRD4, VRK1, and GALK2. We confirmed, using both genetic and pharmacologic approaches, that the activity of BRD4, an epigenetic transcription modulator, is necessary for proliferation/survival of both an established human ovarian cancer cell line (OVCAR8) and a subset of primary serous ovarian cancer cell strains (DFs). Among the DFs tested, the strains sensitive to BRD4 inhibition revealed elevated expression of either MYCN or c-MYC, with MYCN expression correlating closely with JQ1 sensitivity. Accordingly, primary human xenografts derived from high-MYCN or c-MYC strains exhibited sensitivity to BRD4 inhibition. These data suggest that BRD4 inhibition represents a new therapeutic approach for MYC-overexpressing HGSOCs.


Subject(s)
Genetic Testing , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/therapy , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Transcription Factors/metabolism , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Female , Genetic Association Studies , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
15.
Nat Commun ; 5: 5496, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25400221

ABSTRACT

BRCA1-a breast and ovarian cancer suppressor gene-promotes genome integrity. To study the functionality of BRCA1 in the heterozygous state, we established a collection of primary human BRCA1(+/+) and BRCA1(mut/+) mammary epithelial cells and fibroblasts. Here we report that all BRCA1(mut/+) cells exhibited multiple normal BRCA1 functions, including the support of homologous recombination- type double-strand break repair (HR-DSBR), checkpoint functions, centrosome number control, spindle pole formation, Slug expression and satellite RNA suppression. In contrast, the same cells were defective in stalled replication fork repair and/or suppression of fork collapse, that is, replication stress. These defects were rescued by reconstituting BRCA1(mut/+) cells with wt BRCA1. In addition, we observed 'conditional' haploinsufficiency for HR-DSBR in BRCA1(mut/+) cells in the face of replication stress. Given the importance of replication stress in epithelial cancer development and of an HR defect in breast cancer pathogenesis, both defects are candidate contributors to tumorigenesis in BRCA1-deficient mammary tissue.


Subject(s)
DNA Replication/physiology , Genes, BRCA1/physiology , Haploinsufficiency/physiology , Animals , Breast/cytology , Cells, Cultured , Centrosome/physiology , DNA Replication/genetics , Female , Haploinsufficiency/genetics , Heterozygote , Humans , Mice , RNA, Satellite/genetics , RNA, Satellite/physiology , Rad51 Recombinase/genetics , Rad51 Recombinase/physiology , Recombinational DNA Repair/genetics , Recombinational DNA Repair/physiology , Spindle Poles/genetics , Spindle Poles/physiology
16.
Cancer Discov ; 4(12): 1430-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25252691

ABSTRACT

UNLABELLED: BRCA1 promotes homologous recombination-mediated DNA repair (HRR). However, HRR must be tightly regulated to prevent illegitimate recombination. We previously found that BRCA1 HRR function is regulated by the RAP80 complex, but the mechanism was unclear. We have now observed that PARP1 interacts with and poly-ADP-ribosylates (aka PARsylates) BRCA1. PARsylation is directed at the BRCA1 DNA binding domain and downmodulates its function. Moreover, RAP80 contains a poly-ADP-ribose-interacting domain that binds PARsylated BRCA1 and helps to maintain the stability of PARP1-BRCA1-RAP80 complexes. BRCA1 PARsylation is a key step in BRCA1 HRR control. When BRCA1 PARsylation is defective, it gives rise to excessive HRR and manifestations of genome instability. BRCA1 PARsylation and/or RAP80 expression is defective in a subset of sporadic breast cancer cell lines and patient-derived tumor xenograft models. These observations are consistent with the possibility that such defects, when chronic, contribute to tumor development in BRCA1+/+ individuals. SIGNIFICANCE: We propose a model that describes how BRCA1 functions to both support and restrict HRR. BRCA1 PARsylation is a key event in this process, failure of which triggers hyper-recombination and chromosome instability. Thus, hyperfunctioning BRCA1 can elicit genomic abnormalities similar to those observed in the absence of certain BRCA1 functions.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Homologous Recombination , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , BRCA1 Protein/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosomes, Human , DNA Damage , DNA-Binding Proteins , Female , Gene Expression , Genomic Instability , HeLa Cells , Histone Chaperones , Humans , Models, Biological , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments , Poly (ADP-Ribose) Polymerase-1 , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Subunits/metabolism , Signal Transduction
17.
Genes Dev ; 28(17): 1957-75, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25184681

ABSTRACT

BRCA1 is a breast and ovarian tumor suppressor. Given its numerous incompletely understood functions and the possibility that more exist, we performed complementary systematic screens in search of new BRCA1 protein-interacting partners. New BRCA1 functions and/or a better understanding of existing ones were sought. Among the new interacting proteins identified, genetic interactions were detected between BRCA1 and four of the interactors: TONSL, SETX, TCEANC, and TCEA2. Genetic interactions were also detected between BRCA1 and certain interactors of TONSL, including both members of the FACT complex. From these results, a new BRCA1 function in the response to transcription-associated DNA damage was detected. Specifically, new roles for BRCA1 in the restart of transcription after UV damage and in preventing or repairing damage caused by stabilized R loops were identified. These roles are likely carried out together with some of the newly identified interactors. This new function may be important in BRCA1 tumor suppression, since the expression of several interactors, including some of the above-noted transcription proteins, is repeatedly aberrant in both breast and ovarian cancers.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage/genetics , DNA Repair/genetics , Transcription, Genetic/genetics , BRCA1 Protein/genetics , Cell Line, Tumor , HeLa Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding , Protein Interaction Mapping , Ultraviolet Rays
18.
Virology ; 468-470: 311-321, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25222147

ABSTRACT

An important step in the malignant progression of HPV-associated lesions is the dysregulation of expression of the viral E6 and E7 oncogenes. This is often achieved through the loss of expression of E2, which represses the HPV LCR promoter and E6/E7 expression. Our previous studies confirmed a role for Brd4 in mediating the E2 transcriptional repression function, and identified JARID1C/SMCX and EP400 as contributors to E2-mediated repression. Here we show that TIP60, a component of the TIP60/TRRAP histone acetyltransferase complex, also contributes to the E2 repression function, and we extend our studies on SMCX. Di- and tri-methyl marks on histone H3K4 are reduced in the presence of E2 and SMCX, suggesting a mechanism by which SMCX contributes to E2-mediated repression of the HPV LCR. Together, these findings lead us to hypothesize that E2 recruits histone-modifying cellular proteins to the HPV LCR, resulting in transcriptional repression of E6 and E7.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Oncogene Proteins, Viral/metabolism , Oxidoreductases, N-Demethylating/metabolism , Cell Cycle Proteins , Cell Line , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Histone Acetyltransferases/genetics , Histone Demethylases , Humans , Lysine Acetyltransferase 5 , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Oxidoreductases, N-Demethylating/genetics , Plasmids , Promoter Regions, Genetic , Protein Binding , Protein Subunits , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Mol Cell Biol ; 34(20): 3828-42, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25092866

ABSTRACT

Sporadic basal-like cancers (BLCs) are a common subtype of breast cancer that share multiple biological properties with BRCA1-mutated breast tumors. Despite being BRCA1(+/+), sporadic BLCs are widely viewed as phenocopies of BRCA1-mutated breast cancers, because they are hypothesized to manifest a BRCA1 functional defect or breakdown of a pathway(s) in which BRCA1 plays a major role. The role of BRCA1 in the repair of double-strand DNA breaks by homologous recombination (HR) is its best understood function and the function most often implicated in BRCA1 breast cancer suppression. Therefore, it is suspected that sporadic BLCs exhibit a defect in HR. To test this hypothesis, multiple DNA damage repair assays focused on several types of repair were performed on a group of cell lines classified as sporadic BLCs and on controls. The sporadic BLC cell lines failed to exhibit an overt HR defect. Rather, they exhibited defects in the repair of stalled replication forks, another BRCA1 function. These results provide insight into why clinical trials of poly(ADP-ribose) polymerase (PARP) inhibitors, which require an HR defect for efficacy, have been unsuccessful in sporadic BLCs, unlike cisplatin, which elicits DNA damage that requires stalled fork repair and has shown efficacy in sporadic BLCs.


Subject(s)
BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Breast Neoplasms , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Replication , Female , Humans , Inhibitory Concentration 50 , Methyl Methanesulfonate/pharmacology , Mutagens/pharmacology , Neoplasms, Basal Cell , Recombinational DNA Repair
20.
Mol Cell ; 54(6): 932-945, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24857551

ABSTRACT

Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.


Subject(s)
G1 Phase/genetics , G2 Phase/genetics , Oncogene Protein p21(ras)/genetics , Protein Phosphatase 2/genetics , Resting Phase, Cell Cycle/physiology , Cell Line , Cyclin E/biosynthesis , Humans , MCF-7 Cells , Mitosis/genetics , Protein Subunits/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...