Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Am J Hum Genet ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38906146

ABSTRACT

Genome-wide association studies (GWASs) have identified numerous lung cancer risk-associated loci. However, decoding molecular mechanisms of these associations is challenging since most of these genetic variants are non-protein-coding with unknown function. Here, we implemented massively parallel reporter assays (MPRAs) to simultaneously measure the allelic transcriptional activity of risk-associated variants. We tested 2,245 variants at 42 loci from 3 recent GWASs in East Asian and European populations in the context of two major lung cancer histological types and exposure to benzo(a)pyrene. This MPRA approach identified one or more variants (median 11 variants) with significant effects on transcriptional activity at 88% of GWAS loci. Multimodal integration of lung-specific epigenomic data demonstrated that 63% of the loci harbored multiple potentially functional variants in linkage disequilibrium. While 22% of the significant variants showed allelic effects in both A549 (adenocarcinoma) and H520 (squamous cell carcinoma) cell lines, a subset of the functional variants displayed a significant cell-type interaction. Transcription factor analyses nominated potential regulators of the functional variants, including those with cell-type-specific expression and those predicted to bind multiple potentially functional variants across the GWAS loci. Linking functional variants to target genes based on four complementary approaches identified candidate susceptibility genes, including those affecting lung cancer cell growth. CRISPR interference of the top functional variant at 20q13.33 validated variant-to-gene connections, including RTEL1, SOX18, and ARFRP1. Our data provide a comprehensive functional analysis of lung cancer GWAS loci and help elucidate the molecular basis of heterogeneity and polygenicity underlying lung cancer susceptibility.

2.
Am J Hum Genet ; 109(12): 2210-2229, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36423637

ABSTRACT

The most recent genome-wide association study (GWAS) of cutaneous melanoma identified 54 risk-associated loci, but functional variants and their target genes for most have not been established. Here, we performed massively parallel reporter assays (MPRAs) by using malignant melanoma and normal melanocyte cells and further integrated multi-layer annotation to systematically prioritize functional variants and susceptibility genes from these GWAS loci. Of 1,992 risk-associated variants tested in MPRAs, we identified 285 from 42 loci (78% of the known loci) displaying significant allelic transcriptional activities in either cell type (FDR < 1%). We further characterized MPRA-significant variants by motif prediction, epigenomic annotation, and statistical/functional fine-mapping to create integrative variant scores, which prioritized one to six plausible candidate variants per locus for the 42 loci and nominated a single variant for 43% of these loci. Overlaying the MPRA-significant variants with genome-wide significant expression or methylation quantitative trait loci (eQTLs or meQTLs, respectively) from melanocytes or melanomas identified candidate susceptibility genes for 60% of variants (172 of 285 variants). CRISPRi of top-scoring variants validated their cis-regulatory effect on the eQTL target genes, MAFF (22q13.1) and GPRC5A (12p13.1). Finally, we identified 36 melanoma-specific and 45 melanocyte-specific MPRA-significant variants, a subset of which are linked to cell-type-specific target genes. Analyses of transcription factor availability in MPRA datasets and variant-transcription-factor interaction in eQTL datasets highlighted the roles of transcription factors in cell-type-specific variant functionality. In conclusion, MPRAs along with variant scoring effectively prioritized plausible candidates for most melanoma GWAS loci and highlighted cellular contexts where the susceptibility variants are functional.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/genetics , Skin Neoplasms/genetics , Genome-Wide Association Study , Biological Assay , Transcription Factors , Receptors, G-Protein-Coupled , Melanoma, Cutaneous Malignant
3.
Cell ; 134(2): 279-90, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18662543

ABSTRACT

In quiescent cells, mitochondria are the primary source of reactive oxygen species (ROS), which are generated by leakiness of the electron transport chain (ETC). High levels of ROS can trigger cell death, whereas lower levels drive diverse and important cellular functions. We show here by employing a newly developed mitochondrial matrix-targeted superoxide indicator, that individual mitochondria undergo spontaneous bursts of superoxide generation, termed "superoxide flashes." Superoxide flashes occur randomly in space and time, exhibit all-or-none properties, and provide a vital source of superoxide production across many different cell types. Individual flashes are triggered by transient openings of the mitochondrial permeability transition pore stimulating superoxide production by the ETC. Furthermore, we observe a flurry of superoxide flash activity during reoxygenation of cardiomyocytes after hypoxia, which is inhibited by the cardioprotective compound adenosine. We propose that superoxide flashes could serve as a valuable biomarker for a wide variety of oxidative stress-related diseases.


Subject(s)
Mitochondria/metabolism , Superoxides/metabolism , Adenoviridae/genetics , Animals , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Humans , Luminescent Proteins/metabolism , Muscle Cells/metabolism , Myocytes, Cardiac/metabolism , Neurons/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
4.
Nucleic Acids Res ; 44(10): 4871-80, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27131364

ABSTRACT

The Fanconi anemia protein SLX4 assembles a genome and telomere maintenance toolkit, consisting of the nucleases SLX1, MUS81 and XPF. Although it is known that SLX4 acts as a scaffold for building this complex, the molecular basis underlying this function of SLX4 remains unclear. Here, we report that functioning of SLX4 is dependent on its dimerization via an oligomerization motif called the BTB domain. We solved the crystal structure of the SLX4BTB dimer, identifying key contacts (F681 and F708) that mediate dimerization. Disruption of BTB dimerization abrogates nuclear foci formation and telomeric localization of not only SLX4 but also of its associated nucleases. Furthermore, dimerization-deficient SLX4 mutants cause defective cellular response to DNA interstrand crosslinking agent and telomere maintenance, underscoring the contribution of BTB domain-mediated dimerization of SLX4 in genome and telomere maintenance.


Subject(s)
Endonucleases/metabolism , Recombinases/chemistry , Cell Line , Hydrophobic and Hydrophilic Interactions , Mitomycin/toxicity , Protein Domains , Protein Multimerization , Recombinases/metabolism , Telomere/enzymology , Telomere/ultrastructure
5.
Nucleic Acids Res ; 43(12): 5912-23, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25990736

ABSTRACT

SLX4 assembles a toolkit of endonucleases SLX1, MUS81 and XPF, which is recruited to telomeres via direct interaction of SLX4 with TRF2. Telomeres present an inherent obstacle for DNA replication and repair due to their high propensity to form branched DNA intermediates. Here we provide novel insight into the mechanism and regulation of the SLX4 complex in telomere preservation. SLX4 associates with telomeres throughout the cell cycle, peaking in late S phase and under genotoxic stress. Disruption of SLX4's interaction with TRF2 or SLX1 and SLX1's nuclease activity independently causes telomere fragility, suggesting a requirement of the SLX4 complex for nucleolytic resolution of branched intermediates during telomere replication. Indeed, the SLX1-SLX4 complex processes a variety of telomeric joint molecules in vitro. The nucleolytic activity of SLX1-SLX4 is negatively regulated by telomeric DNA-binding proteins TRF1 and TRF2 and is suppressed by the RecQ helicase BLM in vitro. In vivo, in the presence of functional BLM, telomeric circle formation and telomere sister chromatid exchange, both arising out of nucleolytic processing of telomeric homologous recombination intermediates, are suppressed. We propose that the SLX4-toolkit is a telomere accessory complex that, in conjunction with other telomere maintenance proteins, ensures unhindered, but regulated telomere maintenance.


Subject(s)
Recombinases/metabolism , Telomere/metabolism , Cell Cycle , DNA/metabolism , Endodeoxyribonucleases , Endonucleases/metabolism , HeLa Cells , Homologous Recombination , Humans , RecQ Helicases/metabolism , Sister Chromatid Exchange , Telomere-Binding Proteins/metabolism
6.
J Biol Chem ; 290(9): 5502-11, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25572391

ABSTRACT

Uracil in the genome can result from misincorporation of dUTP instead of dTTP during DNA synthesis, and is primarily removed by uracil DNA glycosylase (UNG) during base excision repair. Telomeres contain long arrays of TTAGGG repeats and may be susceptible to uracil misincorporation. Using model telomeric DNA substrates, we showed that the position and number of uracil substitutions of thymine in telomeric DNA decreased recognition by the telomere single-strand binding protein, POT1. In primary mouse hematopoietic cells, uracil was detectable at telomeres, and UNG deficiency further increased uracil loads and led to abnormal telomere lengthening. In UNG-deficient cells, the frequencies of sister chromatid exchange and fragility in telomeres also significantly increased in the absence of telomerase. Thus, accumulation of uracil and/or UNG deficiency interferes with telomere maintenance, thereby underscoring the necessity of UNG-initiated base excision repair for the preservation of telomere integrity.


Subject(s)
Bone Marrow Cells/metabolism , DNA Repair , Telomere/metabolism , Uracil/metabolism , Animals , Base Sequence , Cells, Cultured , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , In Situ Hybridization, Fluorescence , Mice, Knockout , Protein Binding , Shelterin Complex , Telomere/genetics , Telomere Homeostasis/genetics , Telomere-Binding Proteins , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Thymine/metabolism , Uracil-DNA Glycosidase/deficiency , Uracil-DNA Glycosidase/genetics
7.
EMBO J ; 29(18): 3140-55, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20711169

ABSTRACT

BLM, the helicase defective in Bloom syndrome, is part of a multiprotein complex that protects genome stability. Here, we show that Rif1 is a novel component of the BLM complex and works with BLM to promote recovery of stalled replication forks. First, Rif1 physically interacts with the BLM complex through a conserved C-terminal domain, and the stability of Rif1 depends on the presence of the BLM complex. Second, Rif1 and BLM are recruited with similar kinetics to stalled replication forks, and the Rif1 recruitment is delayed in BLM-deficient cells. Third, genetic analyses in vertebrate DT40 cells suggest that BLM and Rif1 work in a common pathway to resist replication stress and promote recovery of stalled forks. Importantly, vertebrate Rif1 contains a DNA-binding domain that resembles the αCTD domain of bacterial RNA polymerase α; and this domain preferentially binds fork and Holliday junction (HJ) DNA in vitro and is required for Rif1 to resist replication stress in vivo. Our data suggest that Rif1 provides a new DNA-binding interface for the BLM complex to restart stalled replication forks.


Subject(s)
Carrier Proteins/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Telomere-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Chickens , DNA/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA, Small Interfering/pharmacology , RecQ Helicases/antagonists & inhibitors , RecQ Helicases/genetics , Sequence Homology, Amino Acid , Telomere-Binding Proteins/antagonists & inhibitors , Telomere-Binding Proteins/genetics
8.
bioRxiv ; 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37808664

ABSTRACT

Genome-wide association studies (GWAS) identified over fifty loci associated with lung cancer risk. However, the genetic mechanisms and target genes underlying these loci are largely unknown, as most risk-associated-variants might regulate gene expression in a context-specific manner. Here, we generated a barcode-shared transcriptome and chromatin accessibility map of 117,911 human lung cells from age/sex-matched ever- and never-smokers to profile context-specific gene regulation. Accessible chromatin peak detection identified cell-type-specific candidate cis-regulatory elements (cCREs) from each lung cell type. Colocalization of lung cancer candidate causal variants (CCVs) with these cCREs prioritized the variants for 68% of the GWAS loci, a subset of which was also supported by transcription factor abundance and footprinting. cCRE colocalization and single-cell based trait relevance score nominated epithelial and immune cells as the main cell groups contributing to lung cancer susceptibility. Notably, cCREs of rare proliferating epithelial cell types, such as AT2-proliferating (0.13%) and basal cells (1.8%), overlapped with CCVs, including those in TERT. A multi-level cCRE-gene linking system identified candidate susceptibility genes from 57% of lung cancer loci, including those not detected in tissue- or cell-line-based approaches. cCRE-gene linkage uncovered that adjacent genes expressed in different cell types are correlated with distinct subsets of coinherited CCVs, including JAML and MPZL3 at the 11q23.3 locus. Our data revealed the cell types and contexts where the lung cancer susceptibility genes are functional.

9.
J Clin Invest ; 130(11): 6034-6040, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32780721

ABSTRACT

Air pollution involving particulate matter smaller than 2.5 µm in size (PM2.5) is the world's leading environmental risk factor contributing to mortality through cardiometabolic pathways. In this study, we modeled early life exposure using chow-fed C57BL/6J male mice that were exposed to real-world inhaled, concentrated PM2.5 (~10 times ambient levels/~60-120 µg/m3) or filtered air over a 14-week period. We investigated the effects of PM2.5 on phenotype, the transcriptome, and chromatin accessibility and compared these with the effects of a prototypical high-fat diet (HFD) as well as cessation of exposure on phenotype reversibility. Exposure to PM2.5 impaired glucose and insulin tolerance and reduced energy expenditure and 18FDG-PET uptake in brown adipose tissue. Multiple differentially expressed gene clusters in pathways involving metabolism and circadian rhythm were noted in insulin-responsive tissues. Although the magnitude of transcriptional change detected with PM2.5 exposure was lower than that observed with a HFD, the degree of alteration in chromatin accessibility after PM2.5 exposure was significant. The novel chromatin remodeler SMARCA5 (SWI/SNF complex) was regulated in response to PM2.5 exposure, the cessation of which was associated with a reversal of insulin resistance and restoration of chromatin accessibility and nucleosome positioning near transcription start sites, as well as a reversal of exposure-induced changes in the transcriptome, including SMARCA5. These changes indicate pliable epigenetic control mechanisms following cessation of exposure.


Subject(s)
Adipose Tissue, Brown , Air Pollutants/toxicity , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Environmental Exposure/adverse effects , Insulin Resistance , Adenosine Triphosphatases/metabolism , Adipose Tissue, Brown/diagnostic imaging , Adipose Tissue, Brown/metabolism , Animals , Chromatin Assembly and Disassembly/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Fluorodeoxyglucose F18/pharmacology , Mice , Positron-Emission Tomography , Transcriptome/drug effects
10.
Nat Commun ; 11(1): 2718, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483191

ABSTRACT

Genome-wide association studies (GWAS) have identified ~20 melanoma susceptibility loci, most of which are not functionally characterized. Here we report an approach integrating massively-parallel reporter assays (MPRA) with cell-type-specific epigenome and expression quantitative trait loci (eQTL) to identify susceptibility genes/variants from multiple GWAS loci. From 832 high-LD variants, we identify 39 candidate functional variants from 14 loci displaying allelic transcriptional activity, a subset of which corroborates four colocalizing melanocyte cis-eQTL genes. Among these, we further characterize the locus encompassing the HIV-1 restriction gene, MX2 (Chr21q22.3), and validate a functional intronic variant, rs398206. rs398206 mediates the binding of the transcription factor, YY1, to increase MX2 levels, consistent with the cis-eQTL of MX2 in primary human melanocytes. Melanocyte-specific expression of human MX2 in a zebrafish model demonstrates accelerated melanoma formation in a BRAFV600E background. Our integrative approach streamlines GWAS follow-up studies and highlights a pleiotropic function of MX2 in melanoma susceptibility.


Subject(s)
Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Melanoma/genetics , Mutation , Myxovirus Resistance Proteins/genetics , Polymorphism, Single Nucleotide , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation , Genes, Reporter/genetics , HEK293 Cells , Humans , Melanocytes/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Quantitative Trait Loci/genetics , Zebrafish/genetics , Zebrafish/metabolism
11.
Gene ; 391(1-2): 26-38, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17250975

ABSTRACT

Mutations in a human RecQ helicase homologue, RECQL4, have been identified in patients with Type II Rothmund-Thomson syndrome (RTS) with osteosarcoma predisposition, RAPADILINO syndrome, and Baller-Gerold syndrome. A role in DNA replication initiation has been demonstrated and mapped to the amino terminus upstream of the helicase domain; however, no nuclear localization signal (NLS) has been identified by sequence analysis. Here, we show both endogenous and green fluorescent protein (GFP)-tagged RECQL4 are nuclear and cytoplasmic in transformed cell lines. Using GFP-tagged constructs we identified a major nuclear localization domain within amino acids (aa) 363-492 (exons 5-8) sufficient for nuclear localization of GFP and necessary for nuclear localization of RECQL4 as GFP-RECQL4 deleted for aa 363-492 is entirely cytoplasmic. Additional mapping within this domain revealed that a conserved block of 22 basic amino acids (aa 365-386; exons 5-6) is sufficient for nuclear localization of GFP, but not required for nuclear import of RECQL4. Conversely, even though the region encoded by exon 7-8 is not sufficient for nuclear import of GFP, GFP-RECQL4 deleted for exon 7 (aa 420-463), a mutation found in all reported patients with RAPADILINO syndrome, is cytoplasmic. Nuclear localization of the exon 7 deletion construct is increased in cells treated with leptomycin B suggesting that exon 7 encodes a domain required for nuclear retention of RECQL4. This retention activity is partially conveyed by a conserved VLPLY motif (aa 450-454) in exon 7 of the human sequence. In summary, unlike other RecQ proteins with carboxyl terminal NLS, RECQL4 nuclear localization and retention activities are amino terminal. This location would provide nuclear transport of putative truncated proteins encoded by RTS mutant alleles consistent with the proposed essential replication function in the amino terminus of RECQL4.


Subject(s)
Cell Nucleus/metabolism , RecQ Helicases/genetics , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Conserved Sequence/genetics , Cytoplasm/metabolism , Fatty Acids, Unsaturated/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Nuclear Localization Signals/genetics , RecQ Helicases/chemistry , RecQ Helicases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
12.
Nat Genet ; 46(5): 482-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24686846

ABSTRACT

Although CDKN2A is the most frequent high-risk melanoma susceptibility gene, the underlying genetic factors for most melanoma-prone families remain unknown. Using whole-exome sequencing, we identified a rare variant that arose as a founder mutation in the telomere shelterin gene POT1 (chromosome 7, g.124493086C>T; p.Ser270Asn) in five unrelated melanoma-prone families from Romagna, Italy. Carriers of this variant had increased telomere lengths and numbers of fragile telomeres, suggesting that this variant perturbs telomere maintenance. Two additional rare POT1 variants were identified in all cases sequenced in two separate Italian families, one variant per family, yielding a frequency for POT1 variants comparable to that for CDKN2A mutations in this population. These variants were not found in public databases or in 2,038 genotyped Italian controls. We also identified two rare recurrent POT1 variants in US and French familial melanoma cases. Our findings suggest that POT1 is a major susceptibility gene for familial melanoma in several populations.


Subject(s)
Genetic Predisposition to Disease/genetics , Melanoma/genetics , Models, Molecular , Mutation, Missense/genetics , Neoplasms, Connective Tissue/genetics , Telomere Homeostasis/genetics , Telomere-Binding Proteins/genetics , Amino Acid Sequence , Base Sequence , Computational Biology , Exome/genetics , France , Humans , In Situ Hybridization, Fluorescence , Italy , Molecular Sequence Data , Pedigree , Sequence Alignment , Sequence Analysis, DNA , Shelterin Complex , Skin Neoplasms , Telomere-Binding Proteins/chemistry , United States , Melanoma, Cutaneous Malignant
13.
Aging Cell ; 12(4): 635-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23590194

ABSTRACT

Reactive oxygen species (ROS) are proposed to play a major role in telomere length alterations during aging. The mechanisms by which ROS disrupt telomeres remain unclear. In Saccharomyces cerevisiae, telomere DNA consists of TG(1-3) repeats, which are maintained primarily by telomerase. Telomere length maintenance can be modulated by the expression level of telomerase subunits and telomerase activity. Additionally, telomerase-mediated telomere repeat addition is negatively modulated by the levels of telomere-bound Rap1-Rif1-Rif2 protein complex. Using a yeast strain defective in the major peroxiredoxin Tsa1 that is involved in ROS neutralization, we have investigated the effect of defective ROS detoxification on telomere DNA, telomerase, telomere-binding proteins, and telomere length. Surprisingly, the tsa1 mutant does not show significant increase in steady-state levels of oxidative DNA lesions at telomeres. The tsa1 mutant displays abnormal telomere lengthening, and reduction in oxidative exposure alleviates this phenotype. The telomere lengthening in the tsa1 cells was abolished by disruption of Est2, subtelomeric DNA, Rap1 C-terminus, or Rif2, but not by Rif1 deletion. Although telomerase expression and activity are not altered, telomere-bound Est2 is increased, while telomere-bound Rap1 is reduced in the tsa1 mutant. We propose that defective ROS scavenging can interfere with pathways that are critical in controlling telomere length homeostasis.


Subject(s)
Gene Deletion , Gene Expression Regulation, Enzymologic , Peroxidases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/enzymology , Telomere Homeostasis , Telomere/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , Enzyme Activation , Gene Expression Regulation, Fungal , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Shelterin Complex , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Cell Rep ; 4(5): 861-9, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24012755

ABSTRACT

SLX4 interacts with several endonucleases to resolve structural barriers in DNA metabolism. SLX4 also interacts with telomeric protein TRF2 in human cells. The molecular mechanism of these interactions at telomeres remains unknown. Here, we report the crystal structure of the TRF2-binding motif of SLX4 (SLX4TBM) in complex with the TRFH domain of TRF2 (TRF2TRFH) and map the interactions of SLX4 with endonucleases SLX1, XPF, and MUS81. TRF2 recognizes a unique HxLxP motif on SLX4 via the peptide-binding site in its TRFH domain. Telomeric localization of SLX4 and associated nucleases depend on the SLX4-endonuclease and SLX4-TRF2 interactions and the protein levels of SLX4 and TRF2. SLX4 assembles an endonuclease toolkit that negatively regulates telomere length via SLX1-catalyzed nucleolytic resolution of telomere DNA structures. We propose that the SLX4-TRF2 complex serves as a double-layer scaffold bridging multiple endonucleases with telomeres for recombination-based telomere maintenance.


Subject(s)
DNA Repair , Endonucleases/metabolism , Recombinases/metabolism , Telomere/metabolism , Endonucleases/genetics , Humans , Recombinases/genetics , Telomere/genetics
15.
Mol Cell Biol ; 30(6): 1382-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20065033

ABSTRACT

Cellular and biochemical studies support a role for all five human RecQ helicases in DNA replication; however, their specific functions during this process are unclear. Here we investigate the in vivo association of the five human RecQ helicases with three well-characterized human replication origins. We show that only RECQ1 (also called RECQL or RECQL1) and RECQ4 (also called RECQL4) associate with replication origins in a cell cycle-regulated fashion in unperturbed cells. RECQ4 is recruited to origins at late G(1), after ORC and MCM complex assembly, while RECQ1 and additional RECQ4 are loaded at origins at the onset of S phase, when licensed origins begin firing. Both proteins are lost from origins after DNA replication initiation, indicating either disassembly or tracking with the newly formed replisome. Nascent-origin DNA synthesis and the frequency of origin firing are reduced after RECQ1 depletion and, to a greater extent, after RECQ4 depletion. Depletion of RECQ1, though not that of RECQ4, also suppresses replication fork rates in otherwise unperturbed cells. These results indicate that RECQ1 and RECQ4 are integral components of the human replication complex and play distinct roles in DNA replication initiation and replication fork progression in vivo.


Subject(s)
DNA Replication , RecQ Helicases/metabolism , Cell Line , Cell Proliferation , Chromatin/metabolism , DNA/biosynthesis , DNA Replication Timing , Down-Regulation , G1 Phase , Humans , Lamin Type B/genetics , Lamin Type B/metabolism , Models, Biological , Protein Binding , RNA, Small Interfering/metabolism , Replication Origin/genetics , S Phase , beta-Globins/genetics , beta-Globins/metabolism
16.
Mol Cell Biol ; 29(8): 2042-52, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19223468

ABSTRACT

Human DNA ligase I (hLigI) participates in DNA replication and excision repair via an interaction with proliferating cell nuclear antigen (PCNA), a DNA sliding clamp. In addition, hLigI interacts with and is inhibited by replication factor C (RFC), the clamp loader complex that loads PCNA onto DNA. Here we show that a mutant version of hLigI, which mimics the hyperphosphorylated M-phase form of hLigI, does not interact with and is not inhibited by RFC, demonstrating that inhibition of ligation is dependent upon the interaction between hLigI and RFC. To examine the biological relevance of hLigI phosphorylation, we isolated derivatives of the hLigI-deficient cell line 46BR.1G1 that stably express mutant versions of hLigI in which four serine residues phosphorylated in vivo were replaced with either alanine or aspartic acid. The cell lines expressing the phosphorylation site mutants of hLigI exhibited a dramatic reduction in proliferation and DNA synthesis and were also hypersensitive to DNA damage. The dominant-negative effects of the hLigI phosphomutants on replication and repair are due to the activation of cellular senescence, presumably because of DNA damage arising from replication abnormalities. Thus, appropriate phosphorylation of hLigI is critical for its participation in DNA replication and repair.


Subject(s)
DNA Ligases/metabolism , DNA Repair , DNA Replication , Replication Protein C/metabolism , Cell Line , Cell Proliferation , Cellular Senescence , DNA Ligase ATP , DNA Ligases/antagonists & inhibitors , DNA Ligases/genetics , Humans , Mutant Proteins , Phosphorylation , Replication Protein C/physiology
17.
Proc Natl Acad Sci U S A ; 103(11): 4068-73, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537486

ABSTRACT

BLM encodes a member of the highly conserved RecQ DNA helicase family, which is essential for the maintenance of genome stability. Homozygous inactivation of BLM gives rise to the cancer predisposition disorder Bloom's syndrome. A common feature of many RecQ helicase mutants is a hyperrecombination phenotype. In Bloom's syndrome, this phenotype manifests as an elevated frequency of sister chromatid exchanges and interhomologue recombination. We have shown previously that BLM, together with its evolutionarily conserved binding partner topoisomerase IIIalpha (hTOPO IIIalpha), can process recombination intermediates that contain double Holliday junctions into noncrossover products by a mechanism termed dissolution. Here we show that a recently identified third component of the human BLM/hTOPO IIIalpha complex, BLAP75/RMI1, promotes dissolution catalyzed by hTOPO IIIalpha. This activity of BLAP75/RMI1 is specific for dissolution catalyzed by hTOPO IIIalpha because it has no effect in reactions containing either Escherichia coli Top1 or Top3, both of which can also catalyze dissolution in a BLM-dependent manner. We present evidence that BLAP75/RMI1 acts by recruiting hTOPO IIIalpha to double Holliday junctions. Implications of the conserved ability of type IA topoisomerases to catalyze dissolution and how the evolution of factors such as BLAP75/RMI1 might confer specificity on the execution of this process are discussed.


Subject(s)
Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , DNA Helicases/metabolism , Recombination, Genetic , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA-Binding Proteins , Escherichia coli/genetics , Humans , In Vitro Techniques , Multiprotein Complexes , Nuclear Proteins , Phenotype , Protein Binding , RecQ Helicases , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sister Chromatid Exchange
18.
Cell Cycle ; 4(1): 80-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15611632

ABSTRACT

Fanconi anemia (FA) proteins function in a DNA damage response pathway that appears to be part of the network including breast cancer susceptibility gene products, BRCA1 and BRCA2. In response to DNA damage or replication signals, a nuclear FA core complex of at least 6 FA proteins (FANCA, FANCC, FANCE, FANCF, FANCG and FANCL) is activated and leads to monoubiquitination of the downstream FA protein, FANCD2. One puzzling question for this pathway is the role of BRCA2. A previous study has proposed that BRCA2 could be identical to two FA proteins: FANCD1, which functions either downstream or in a parallel pathway; and FANCB, which functions upstream of the FANCD2 monoubiquitination. Now, a new study shows that the real FANCB protein is not BRCA2, but a previously uncharacterized component of the FA core complex, FAAP95, suggesting that BRCA2 does not act upstream of the FA pathway. Interestingly, the newly discovered FANCB gene is X-linked and subject to X-inactivation. The presence of a single active copy of FANCB and its essentiality for a functional FA-BRCA pathway make it a potentially vulnerable component of the cellular machinery that maintains genomic integrity.


Subject(s)
BRCA1 Protein/physiology , BRCA2 Protein/physiology , DNA Damage , Fanconi Anemia Complementation Group Proteins/physiology , Fanconi Anemia/physiopathology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Repair/genetics , DNA Repair/physiology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression Regulation , Genes, BRCA1 , Genes, BRCA2 , Genes, X-Linked , Humans , Receptors, Peptide/genetics , Receptors, Peptide/physiology , Signal Transduction
19.
EMBO J ; 24(7): 1465-76, 2005 Apr 06.
Article in English | MEDLINE | ID: mdl-15775963

ABSTRACT

Bloom's syndrome (BS) is a rare human genetic disorder characterized by dwarfism, immunodeficiency, genomic instability and cancer predisposition. We have previously purified three complexes containing BLM, the helicase mutated in this disease. Here we demonstrate that BLAP75, a novel protein containing a putative OB-fold nucleic acid binding domain, is an integral component of BLM complexes, and is essential for their stability in vivo. Consistent with a role in BLM-mediated processes, BLAP75 colocalizes with BLM in subnuclear foci in response to DNA damage, and its depletion impairs the recruitment of BLM to these foci. Depletion of BLAP75 by siRNA also results in deficient phosphorylation of BLM during mitosis, as well as defective cell proliferation. Moreover, cells depleted of BLAP75 display an increased level of sister-chromatid exchange, similar to cells depleted of BLM by siRNA. Thus, BLAP75 is an essential component of the BLM-associated cellular machinery that maintains genome integrity.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Helicases/metabolism , Multiprotein Complexes/metabolism , Amino Acid Sequence , Base Sequence , Cell Extracts , Cell Fractionation , DNA-Binding Proteins , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoprecipitation , Molecular Sequence Data , Multiprotein Complexes/genetics , Nuclear Proteins , Oligonucleotides , Phosphorylation , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RecQ Helicases , Sequence Analysis, DNA
20.
Hum Mol Genet ; 13(20): 2421-30, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15317757

ABSTRACT

The Rothmund-Thomson syndrome (growth retardation, skin and bone defects, predisposition to cancer) and the RAPADILINO syndrome are caused by mutations in the RECQL4 gene. The 133 kDa RECQL4 is a putative DNA helicase, a member of the family that includes the BLM and WRN helicases. The latter are mutated, respectively, in the Bloom and Werner syndromes, whose manifestations include predisposition to cancer. Using antibodies to human RECQL4, we found that the bulk of RECQL4 was present in a cytoplasmic extract of HeLa cells, in contrast to the largely nuclear BLM and WRN helicases. However, in untransformed WI-38 fibroblasts, RECQL4 was found to be largely nuclear, and was present at significantly lower total levels than in transformed HeLa cells. RECQL4 from HeLa cells was isolated as a stable complex with UBR1 and UBR2. These 200 kDa proteins are ubiquitin ligases of the N-end rule pathway, whose substrates include proteins with destabilizing N-terminal residues. The functions of this proteolytic pathway include the regulation of peptide import, chromosome stability, meiosis, apoptosis and cardiovascular development. Although the known role of UBR1 and UBR2 is to mediate polyubiquitylation (and subsequent degradation) of their substrates, the UBR1/2-bound RECQL4 was not ubiquitylated in vivo, and was a long-lived protein in HeLa cells. The isolated RECQL4-UBR1/2 complex had a DNA-stimulated ATPase activity, but was inactive in DNA-based assays for helicases and translocases, the assays in which the BLM helicase was active. We discuss ramifications of these results, possible functions of RECQL4, and the involvement of the N-end rule pathway.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/genetics , Cell Nucleus/chemistry , Cytoplasm/chemistry , DNA Helicases/analysis , DNA Helicases/genetics , HeLa Cells , Humans , Immunoprecipitation , Mutation/genetics , RecQ Helicases , Rothmund-Thomson Syndrome/genetics , Syndrome , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL