Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 10: 1071786, 2022.
Article in English | MEDLINE | ID: mdl-36605718

ABSTRACT

Switch/Sucrose non-fermenting (SWI/SNF) chromatin remodelers hydrolyze ATP to push and slide nucleosomes along the DNA thus modulating access to various genomic loci. These complexes are the most frequently mutated epigenetic regulators in human cancers. SWI/SNF complexes are well known for their function in transcription regulation, but more recent work has uncovered a role for these complexes in the repair of DNA double strand breaks (DSBs). As radiotherapy and most chemotherapeutic agents kill cancer cells by inducing double strand breaks, by identifying a role for these complexes in double strand break repair we are also identifying a DNA repair vulnerability that can be exploited therapeutically in the treatment of SWI/SNF-mutated cancers. In this review we summarize work describing the function of various SWI/SNF subunits in the repair of double strand breaks with a focus on homologous recombination repair and discuss the implication for the treatment of cancers with SWI/SNF mutations.

2.
Cell Cycle ; 19(22): 3096-3114, 2020 11.
Article in English | MEDLINE | ID: mdl-33044911

ABSTRACT

DNA double strand breaks (DSBs) are among the most toxic DNA lesions and can be repaired accurately through homologous recombination (HR). HR requires processing of the DNA ends by nucleases (DNA end resection) in order to generate the required single-stranded DNA (ssDNA) regions. The SWI/SNF chromatin remodelers are 10-15 subunit complexes that contain one ATPase (BRG1 or BRM). Multiple subunits of these complexes have recently been identified as a novel family of tumor suppressors. These complexes are capable of remodeling chromatin by pushing nucleosomes along the DNA. More recent studies have identified these chromatin remodelers as important factors in DNA repair. Using the DR-U2OS reporter system, we show that the down regulation of BRG1 significantly reduces HR efficiency, while BRM has a minor effect. Inactivation of BRG1 impairs DSB repair and results in a defect in DNA end resection, as measured by the amount of BrdU-containing ssDNA generated after DNA damage. Inactivation of BRG1 also impairs the activation of the ATR kinase, reduces the levels of chromatin-bound RPA, and reduces the number of RPA and RAD51 foci after DNA damage. This defect in DNA end resection is explained by the defective recruitment of GFP-CtIP to laser-induced DSBs in the absence of BRG1. Importantly, we show that BRG1 reduces nucleosome density at DSBs. Finally, inactivation of BRG1 renders cells sensitive to anti-cancer drugs that induce DSBs. This study identifies BRG1 as an important factor for HR, which suggests that BRG1-mutated cancers have a DNA repair vulnerability that can be exploited therapeutically.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA Helicases/metabolism , Endodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Recombinational DNA Repair/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Damage , DNA Helicases/genetics , Down-Regulation/genetics , Endodeoxyribonucleases/genetics , Humans , Nuclear Proteins/genetics , Replication Protein A/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transfection
3.
Cell Cycle ; 19(18): 2260-2269, 2020 09.
Article in English | MEDLINE | ID: mdl-32787501

ABSTRACT

The E2F1 transcription factor and RB tumor suppressor are best known for their roles in regulating the expression of genes important for cell cycle progression but, they also have transcription-independent functions that facilitate DNA repair at sites of damage. Depending on the type of DNA damage, E2F1 can recruit either the GCN5 or p300/CBP histone acetyltransferases to deposit different histone acetylation marks in flanking chromatin. At DNA double-strand breaks, E2F1 also recruits RB and the BRG1 ATPase to remodel chromatin and promote loading of the MRE11-RAD50-NBS1 complex. Knock-in mouse models demonstrate important roles for E2F1 post-translational modifications in regulating DNA repair and physiological responses to DNA damage. This review highlights how E2F1 moonlights in DNA repair, thus revealing E2F1 as a versatile protein that recruits many of the same chromatin-modifying enzymes to sites of DNA damage to promote repair that it recruits to gene promoters to regulate transcription.


Subject(s)
DNA Damage , DNA Repair , E2F1 Transcription Factor/metabolism , Retinoblastoma Protein/metabolism , Acetylation , Animals , Chromatin Assembly and Disassembly , E2F1 Transcription Factor/genetics , Histones/metabolism , Humans , Retinoblastoma Protein/genetics , p300-CBP Transcription Factors/metabolism
4.
Nat Commun ; 10(1): 4951, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666529

ABSTRACT

E2F1 and retinoblastoma (RB) tumor-suppressor protein not only regulate the periodic expression of genes important for cell proliferation, but also localize to DNA double-strand breaks (DSBs) to promote repair. E2F1 is acetylated in response to DNA damage but the role this plays in DNA repair is unknown. Here we demonstrate that E2F1 acetylation creates a binding motif for the bromodomains of the p300/KAT3B and CBP/KAT3A acetyltransferases and that this interaction is required for the recruitment of p300 and CBP to DSBs and the induction of histone acetylation at sites of damage. A knock-in mutation that blocks E2F1 acetylation abolishes the recruitment of p300 and CBP to DSBs and also the accumulation of other chromatin modifying activities and repair factors, including Tip60, BRG1 and NBS1, and renders mice hypersensitive to ionizing radiation (IR). These findings reveal an important role for E2F1 acetylation in orchestrating the remodeling of chromatin structure at DSBs to facilitate repair.


Subject(s)
CREB-Binding Protein/metabolism , DNA Breaks, Double-Stranded , E1A-Associated p300 Protein/metabolism , E2F1 Transcription Factor/metabolism , Histones/metabolism , Acetylation , Animals , Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , DNA Repair/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Gene Knock-In Techniques , Histone Acetyltransferases , Lysine Acetyltransferase 5/metabolism , Mice , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Radiation, Ionizing , Trans-Activators/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
5.
Int J Mol Sci ; 18(8)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28812991

ABSTRACT

The retinoblastoma (RB) tumor suppressor is known as a master regulator of the cell cycle. RB is mutated or functionally inactivated in the majority of human cancers. This transcriptional regulator exerts its function in cell cycle control through its interaction with the E2F family of transcription factors and with chromatin remodelers and modifiers that contribute to the repression of genes important for cell cycle progression. Over the years, studies have shown that RB participates in multiple processes in addition to cell cycle control. Indeed, RB is known to interact with over 200 different proteins and likely exists in multiple complexes. RB, in some cases, acts through its interaction with E2F1, other members of the pocket protein family (p107 and p130), and/or chromatin remodelers and modifiers. RB is a tumor suppressor with important chromatin regulatory functions that affect genomic stability. These functions include the role of RB in DNA repair, telomere maintenance, chromosome condensation and cohesion, and silencing of repetitive regions. In this review we will discuss recent advances in RB biology related to RB, partner proteins, and their non-transcriptional functions fighting back against genomic instability.


Subject(s)
DNA Repair , Gene Silencing , Genomic Instability , Retinoblastoma Protein/metabolism , Telomere Homeostasis , Animals , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism
6.
Genes Dev ; 30(22): 2500-2512, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27940962

ABSTRACT

The retinoblastoma (RB) tumor suppressor is recognized as a master regulator that controls entry into the S phase of the cell cycle. Its loss leads to uncontrolled cell proliferation and is a hallmark of cancer. RB works by binding to members of the E2F family of transcription factors and recruiting chromatin modifiers to the promoters of E2F target genes. Here we show that RB also localizes to DNA double-strand breaks (DSBs) dependent on E2F1 and ATM kinase activity and promotes DSB repair through homologous recombination (HR), and its loss results in genome instability. RB is necessary for the recruitment of the BRG1 ATPase to DSBs, which stimulates DNA end resection and HR. A knock-in mutation of the ATM phosphorylation site on E2F1 (S29A) prevents the interaction between E2F1 and TopBP1 and recruitment of RB, E2F1, and BRG1 to DSBs. This knock-in mutation also impairs DNA repair, increases genomic instability, and renders mice hypersensitive to IR. Importantly, depletion of RB in osteosarcoma and breast cancer cell lines results in sensitivity to DNA-damaging drugs, which is further exacerbated by poly-ADP ribose polymerase (PARP) inhibitors. We uncovered a novel, nontranscriptional function for RB in HR, which could contribute to genome instability associated with RB loss.


Subject(s)
DNA Breaks, Double-Stranded , DNA Helicases/metabolism , Homologous Recombination/genetics , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Helicases/genetics , DNA Repair/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gamma Rays , Gene Knock-In Techniques , Genomic Instability/genetics , Humans , Male , Mice , Mutagens/pharmacology , Mutation , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Transport/genetics , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Whole-Body Irradiation/mortality
7.
Mech Ageing Dev ; 134(5-6): 234-42, 2013.
Article in English | MEDLINE | ID: mdl-23562425

ABSTRACT

Cockayne syndrome (CS) is a rare genetic disorder characterized by a variety of growth and developmental defects, photosensitivity, cachectic dwarfism, hearing loss, skeletal abnormalities, progressive neurological degeneration, and premature aging. CS arises due to mutations in the CSA and CSB genes. Both gene products are required for the transcription-coupled (TC) branch of the nucleotide excision repair (NER) pathway, however, the severe phenotype of CS patients is hard to reconcile with a sole defect in TC-NER. Studies using cells from patients and mouse models have shown that the CSB protein is involved in a variety of cellular pathways and plays a major role in the cellular response to stress. CSB has been shown to regulate processes such as the transcriptional recovery after DNA damage, the p53 transcriptional response, the response to hypoxia, the response to insulin-like growth factor-1 (IGF-1), transactivation of nuclear receptors, transcription of housekeeping genes and the transcription of rDNA. Some of these processes are also affected in combined XP/CS patients. These new advances in the function(s) of CSB shed light onto the etiology of the clinical features observed in CS patients and could potentially open therapeutic avenues for these patients in the future. Moreover, the study of CS could further our knowledge of the aging process.


Subject(s)
Cockayne Syndrome/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Transcription, Genetic , Animals , Cell Hypoxia/drug effects , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA, Ribosomal/biosynthesis , DNA, Ribosomal/genetics , DNA-Binding Proteins , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Poly-ADP-Ribose Binding Proteins , Proteins/genetics , Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
DNA Repair (Amst) ; 12(4): 293-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23419237

ABSTRACT

In the present study we show that a number of cancer cell lines from different tissues display dramatically increased expression of the Cockayne Syndrome group B (CSB) protein, a DNA repair factor, that has recently been shown to be involved in cell robustness. Furthermore, we demonstrated that ablation of this protein by antisense technology causes devastating effects on tumor cells through a drastic reduction of cell proliferation and massive induction of apoptosis, while non-transformed cells remain unaffected. Finally, suppression of CSB in cancer cells makes these cells hypersensitive to a variety of commonly used cancer chemotherapeutic agents. Based on these results, we conclude that cancer cells overexpress CSB protein in order to enhance their anti-apoptotic capacity. The fact that CSB suppression specifically affects only cancerous cells, without harming healthy cells, suggests that CSB may be a very attractive target for the development of new anticancer therapies.


Subject(s)
Apoptosis/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Gene Expression , HeLa Cells , Humans , MCF-7 Cells , Poly-ADP-Ribose Binding Proteins , RNA, Small Interfering
9.
Proc Natl Acad Sci U S A ; 110(3): E212-20, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23267107

ABSTRACT

Specific mutations in the XPD subunit of transcription factor IIH result in combined xeroderma pigmentosum (XP)/Cockayne syndrome (CS), a severe DNA repair disorder characterized at the cellular level by a transcriptional arrest following UV irradiation. This transcriptional arrest has always been thought to be the result of faulty transcription-coupled repair. In the present study, we showed that, following UV irradiation, XP-D/CS cells displayed a gross transcriptional dysregulation compared with "pure" XP-D cells or WT cells. Furthermore, global RNA-sequencing analysis showed that XP-D/CS cells repressed the majority of genes after UV, whereas pure XP-D cells did not. By using housekeeping genes as a model, we demonstrated that XP-D/CS cells were unable to reassemble these gene promoters and thus to restart transcription after UV irradiation. Furthermore, we found that the repression of these promoters in XP-D/CS cells was not a simple consequence of deficient repair but rather an active heterochromatinization process mediated by the histone deacetylase Sirt1. Indeed, RNA-sequencing analysis showed that inhibition of and/or silencing of Sirt1 changed the chromatin environment at these promoters and restored the transcription of a large portion of the repressed genes in XP-D/CS cells after UV irradiation. Our work demonstrates that a significant part of the transcriptional arrest displayed by XP-D/CS cells arises as a result of an active repression process and not simply as a result of a DNA repair deficiency. This dysregulation of Sirt1 function that results in transcriptional repression may be the cause of various severe clinical features in patients with XP-D/CS that cannot be explained by a DNA repair defect.


Subject(s)
Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , RNA/biosynthesis , Sirtuin 1/metabolism , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Cells, Cultured , Cockayne Syndrome/complications , DNA Repair , Heterochromatin/genetics , Heterochromatin/metabolism , Heterochromatin/radiation effects , Humans , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription, Genetic/radiation effects , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/complications , Xeroderma Pigmentosum Group D Protein/chemistry
10.
Int J Mol Sci ; 13(10): 13554-68, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23202967

ABSTRACT

Many of the biochemical details of nucleotide excision repair (NER) have been established using purified proteins and DNA substrates. In cells however, DNA is tightly packaged around histones and other chromatin-associated proteins, which can be an obstacle to efficient repair. Several cooperating mechanisms enhance the efficiency of NER by altering chromatin structure. Interestingly, many of the players involved in modifying chromatin at sites of DNA damage were originally identified as regulators of transcription. These include ATP-dependent chromatin remodelers, histone modifying enzymes and several transcription factors. The p53 and E2F1 transcription factors are well known for their abilities to regulate gene expression in response to DNA damage. This review will highlight the underappreciated, transcription-independent functions of p53 and E2F1 in modifying chromatin structure in response to DNA damage to promote global NER.


Subject(s)
DNA Repair , E2F1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA/metabolism , DNA Damage , Histones/metabolism , Humans
11.
Cell Cycle ; 10(21): 3719-30, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22032989

ABSTRACT

Mutations in Cockayne syndrome (CS) A and B genes (CSA and CSB) result in a rare genetic disease that affects the development and homeostasis of a wide range of tissues and organs. We previously correlated the degenerative phenotype of patients to the enhanced apoptotic response, exhibited by CS cells, which is associated with the exceptional induction of p53 protein, upon a variety of stress stimuli. Here we showed that the elevated and persistent levels of p53 displayed by CS cells are due to the insufficient ubiquitination of the p53 protein. We further demonstrated that CSA and CSB proteins associate in a unique complex with p53 and Mdm2; this interaction greatly stimulates the ubiquitination of p53 in an Mdm2-dependent manner. Tandem affinity purification and immunoprecipitations combined with mass spectrometry studies indicate that CSA and CSB associate within a Cullin Ring Ubiquitin Ligase complex responsible, under certain circumstances, for p53 ubiquitination. This study identifies CSA and CSB as the key elements of a regulatory mechanism that equilibrate beneficial and detrimental effects of p53 activity upon cellular stress. The deregulation of p53, in absence of either of the CS proteins, can potentially explain the early onset degeneration of tissues and organs observed in CS patients.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Proto-Oncogene Proteins c-mdm2/physiology , Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology , Cockayne Syndrome/pathology , DNA Helicases/metabolism , DNA Helicases/physiology , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/physiology , Feedback, Physiological , Gene Expression Regulation , Humans , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/metabolism , Stress, Physiological , Transcription Factors/metabolism , Transcription Factors/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitination
12.
Mol Cell ; 38(1): 54-66, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20385089

ABSTRACT

Upon gene activation, we found that RNA polymerase II transcription machinery assembles sequentially with the nucleotide excision repair (NER) factors at the promoter. This recruitment occurs in absence of exogenous genotoxic attack, is sensitive to transcription inhibitors, and depends on the XPC protein. The presence of these repair proteins at the promoter of activated genes is necessary in order to achieve optimal DNA demethylation and histone posttranslational modifications (H3K4/H3K9 methylation, H3K9/14 acetylation) and thus efficient RNA synthesis. Deficiencies in some NER factors impede the recruitment of others and affect nuclear receptor transactivation. Our data suggest that there is a functional difference between the presence of the NER factors at the promoters (which requires XPC) and the NER factors at the distal regions of the gene (which requires CSB). While the latter may be a repair function, the former is a function with respect to transcription unveiled in the current study.


Subject(s)
Chromatin/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Mutagens/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Chromatin Assembly and Disassembly , DNA Damage , DNA Methylation , DNA-Binding Proteins/genetics , HeLa Cells , Histones/metabolism , Humans , Models, Molecular , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
13.
Eur J Hum Genet ; 16(3): 320-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18183039

ABSTRACT

Cockayne syndrome is an autosomal recessive neurodegenerative disorder characterized by a specific defect in the repair of UV-induced DNA lesions. Most cases of Cockayne syndrome are caused by mutations in the CSB gene but the pathophysiological mechanisms are poorly understood. We report the clinical and molecular data of two severely affected Cockayne patients with undetectable CSB protein and mRNA. Both patients showed severe growth failure, microcephaly, mental retardation, congenital cataracts, retinal pigmentary degeneration, photosensitivity and died at the ages of 6 and 8 years. UV irradiation assays demonstrated that both patients had the classical DNA repair defect. Genomic DNA sequencing of the CSB gene showed a homozygous deletion involving non-coding exon 1 and upstream regulatory sequences, but none of the coding exons. Functional complementation using a wild-type CSB expression plasmid fully corrected the DNA repair defect in transfected fibroblasts. Horibata et al recently proposed that all type of CSB mutations result in a defect in UV damage repair that is responsible for the photosensitivity observed in the syndrome, but that only truncated CSB polypeptides generated by nonsense mutations have some additional inhibitory functions in transcription or in oxidative damage repair, which are necessary to lead to the other features of the phenotype. Our patients do not fit the proposed paradigm and new hypotheses are required to account for the pathophysiology of Cockayne syndrome, at the crossroads between DNA repair and transcription.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Sequence Deletion , Brain/pathology , Cockayne Syndrome/physiopathology , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Microsatellite Repeats/genetics , Poly-ADP-Ribose Binding Proteins , Polymerase Chain Reaction , RNA, Messenger/genetics
14.
Biochemistry ; 44(10): 3972-81, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15751973

ABSTRACT

DNA adducts are mutagenic and clastogenic. Because of their harmful nature, lesions are recognized by many proteins involved in DNA repair. However, mounting evidence suggests that lesions also are recognized by proteins with no obvious role in repair processes. One such protein is topoisomerase II, an essential enzyme that removes knots and tangles from the DNA. Because topoisomerase II generates a protein-linked double-stranded DNA break during its catalytic cycle, it has the potential to fragment the genome. Previous studies indicate that abasic sites and other lesions that distort the double helix stimulate topoisomerase II-mediated DNA cleavage. Therefore, to further explore interactions between DNA lesions and the enzyme, the effects of exocyclic adducts on DNA cleavage mediated by human topoisomerase IIalpha were determined. When located within the four-base overhang of a topoisomerase II cleavage site (at the +2 or +3 position 3' relative to the scissile bond), 3,N(4)-ethenodeoxycytidine, 3,N(4)-etheno-2'-ribocytidine, 1,N(2)-ethenodeoxyguanosine, pyrimido[1,2-a]purin-10(3H)-one deoxyribose (M(1)dG), and 1,N(2)-propanodeoxyguanosine increased DNA scission approximately 5-17-fold. Enhanced cleavage did not result from an increased affinity of topoisomerase IIalpha for adducted DNA or a decreased rate of religation. Therefore, it is concluded that these exocyclic lesions act by accelerating the forward rate of enzyme-mediated DNA scission. Finally, treatment of cultured human cells with 2-chloroacetaldehyde, a reactive metabolite of vinyl chloride that generates etheno adducts, increased cellular levels of DNA cleavage by topoisomerase IIalpha. This finding suggests that type II topoisomerases interact with exocyclic DNA lesions in physiological systems.


Subject(s)
Acetaldehyde/analogs & derivatives , Antigens, Neoplasm/chemistry , DNA Adducts/toxicity , DNA Damage , DNA Topoisomerases, Type II/chemistry , DNA-Binding Proteins/chemistry , Deoxyguanosine/analogs & derivatives , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Alkylating Agents/metabolism , Alkylating Agents/toxicity , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , DNA Adducts/metabolism , DNA Damage/drug effects , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyguanosine/metabolism , Deoxyguanosine/toxicity , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein , Proto-Oncogenes/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
15.
Biochemistry ; 43(42): 13416-23, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15491148

ABSTRACT

The DNA ligation reaction of topoisomerase II is essential for genomic integrity. However, it has been impossible to examine many fundamental aspects of this reaction because ligation assays historically required the enzyme to cleave a DNA substrate before sealing the nucleic acid break. Recently, a cleavage-independent DNA ligation assay was developed for human topoisomerase IIalpha [Bromberg, K. D., Hendricks, C., Burgin, A. B., and Osheroff, N. (2002) J. Biol. Chem. 277, 31201-31206]. This assay overcomes the requirement for DNA cleavage by monitoring the ability of the enzyme to ligate a nicked oligonucleotide in which the 5'-terminal phosphate at the nick has been activated by covalent attachment to the tyrosine mimic, p-nitrophenol. The cleavage-independent ligation assay was used to more fully characterize the DNA ligation activity of human topoisomerase IIalpha. Results suggest that the active site tyrosine contributes little to the catalysis of DNA ligation beyond its primary role as an activating/leaving group. Although arginine 804 (the residue immediately N-terminal to the active site tyrosine) has been proposed to help anchor the 5'-DNA terminus during cleavage, conversion of this residue to alanine had only a modest effect on DNA ligation. Thus, it appears that arginine 804 does not play an essential role in DNA strand joining. In contrast, disruption of base pairing at the 5'-DNA terminus abrogated DNA ligation in the absence of a covalent enzyme-DNA bond. Therefore, it is proposed that base pairing represents a secondary mechanism for aligning the 5'-DNA termini for ligation. Finally, the human enzyme appears to ligate the two scissile bonds of a cleavage site in a nonconcerted fashion.


Subject(s)
DNA Repair , DNA Topoisomerases, Type II/metabolism , Antigens, Neoplasm , Base Pairing , Catalysis , DNA Damage , DNA, Superhelical/metabolism , DNA-Binding Proteins , Humans , Hydrolysis , Oligonucleotides/metabolism , Plasmids , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...