Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Science ; 381(6664): eadi3448, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37590370

ABSTRACT

CDC45-MCM2-7-GINS (CMG) helicase assembly is the central event in eukaryotic replication initiation. In yeast, a multi-subunit "pre-loading complex" (pre-LC) accompanies GINS to chromatin-bound MCM2-7, leading to CMG formation. Here, we report that DONSON, a metazoan protein mutated in microcephalic primordial dwarfism, is required for CMG assembly in vertebrates. Using AlphaFold to screen for protein-protein interactions followed by experimental validation, we show that DONSON scaffolds a vertebrate pre-LC containing GINS, TOPBP1, and DNA pol ε. Our evidence suggests that DONSON docks the pre-LC onto MCM2-7, delivering GINS to its binding site in CMG. A patient-derived DONSON mutation compromises CMG assembly and recapitulates microcephalic dwarfism in mice. These results unify our understanding of eukaryotic replication initiation, implicate defective CMG assembly in microcephalic dwarfism, and illustrate how in silico protein-protein interaction screening accelerates mechanistic discovery.


Subject(s)
Cell Cycle Proteins , DNA Replication , DNA-Binding Proteins , Minichromosome Maintenance Proteins , Nuclear Proteins , Animals , Humans , Mice , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins , Protein Interaction Mapping/methods , Computer Simulation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Dwarfism/genetics , Microcephaly/genetics , Xenopus laevis
2.
Diagn Microbiol Infect Dis ; 104(1): 115732, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35728458

ABSTRACT

Throughout the COVID-19 pandemic nasopharyngeal or nose and/or throat swabs (NTS) have been the primary approach for collecting patient samples for the subsequent detection of viral RNA. However, this procedure, if undertaken correctly, can be unpleasant and therefore deters individuals from providing high quality samples. To overcome these limitations other modes of sample collection have been explored. In a cohort of frontline health care workers we have compared saliva and gargle samples to gold-standard NTS. 93% of individuals preferred providing saliva or gargle samples, with little sex-dependent variation. Viral titers collected in samples were analyzed using standard methods and showed that gargle and saliva were similarly comparable for identifying COVID-19 positive individuals compared to NTS (92% sensitivity; 98% specificity). We suggest that gargle and saliva collection are viable alternatives to NTS swabs and may encourage testing to provide better disease diagnosis and population surveillance.


Subject(s)
COVID-19 , COVID-19/diagnosis , COVID-19 Testing , Humans , Mouthwashes , Nasopharynx , Pandemics , RNA, Viral/genetics , SARS-CoV-2 , Saliva , Specimen Handling/methods
4.
J Exp Med ; 219(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35262626

ABSTRACT

Aberrant induction of type I IFN is a hallmark of the inherited encephalopathy Aicardi-Goutières syndrome (AGS), but the mechanisms triggering disease in the human central nervous system (CNS) remain elusive. Here, we generated human models of AGS using genetically modified and patient-derived pluripotent stem cells harboring TREX1 or RNASEH2B loss-of-function alleles. Genome-wide transcriptomic analysis reveals that spontaneous proinflammatory activation in AGS astrocytes initiates signaling cascades impacting multiple CNS cell subsets analyzed at the single-cell level. We identify accumulating DNA damage, with elevated R-loop and micronuclei formation, as a driver of STING- and NLRP3-related inflammatory responses leading to the secretion of neurotoxic mediators. Importantly, pharmacological inhibition of proapoptotic or inflammatory cascades in AGS astrocytes prevents neurotoxicity without apparent impact on their increased type I IFN responses. Together, our work identifies DNA damage as a major driver of neurotoxic inflammation in AGS astrocytes, suggests a role for AGS gene products in R-loop homeostasis, and identifies common denominators of disease that can be targeted to prevent astrocyte-mediated neurotoxicity in AGS.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Astrocytes/metabolism , Autoimmune Diseases of the Nervous System/genetics , DNA Damage , Humans , Inflammation/genetics , Inflammation/metabolism , Nervous System Malformations/genetics
5.
Nature ; 602(7898): 623-631, 2022 02.
Article in English | MEDLINE | ID: mdl-35140396

ABSTRACT

The mutational landscape is shaped by many processes. Genic regions are vulnerable to mutation but are preferentially protected by transcription-coupled repair1. In microorganisms, transcription has been demonstrated to be mutagenic2,3; however, the impact of transcription-associated mutagenesis remains to be established in higher eukaryotes4. Here we show that ID4-a cancer insertion-deletion (indel) mutation signature of unknown aetiology5 characterized by short (2 to 5 base pair) deletions -is due to a transcription-associated mutagenesis process. We demonstrate that defective ribonucleotide excision repair in mammals is associated with the ID4 signature, with mutations occurring at a TNT sequence motif, implicating topoisomerase 1 (TOP1) activity at sites of genome-embedded ribonucleotides as a mechanistic basis. Such TOP1-mediated deletions occur somatically in cancer, and the ID-TOP1 signature is also found in physiological settings, contributing to genic de novo indel mutations in the germline. Thus, although topoisomerases protect against genome instability by relieving topological stress6, their activity may also be an important source of mutations in the human genome.


Subject(s)
DNA Topoisomerases, Type I , Germ Cells , Mutagenesis , Neoplasms , Animals , DNA Repair/genetics , DNA Topoisomerases, Type I/metabolism , Germ Cells/metabolism , Humans , Mutagenesis/genetics , Mutation , Neoplasms/genetics , Ribonucleotides/genetics
6.
Mol Microbiol ; 116(3): 909-925, 2021 09.
Article in English | MEDLINE | ID: mdl-34181784

ABSTRACT

The Escherichia coli dnaE gene encodes the α-catalytic subunit (pol IIIα) of DNA polymerase III, the cell's main replicase. Like all high-fidelity DNA polymerases, pol III possesses stringent base and sugar discrimination. The latter is mediated by a so-called "steric gate" residue in the active site of the polymerase that physically clashes with the 2'-OH of an incoming ribonucleotide. Our structural modeling data suggest that H760 is the steric gate residue in E.coli pol IIIα. To understand how H760 and the adjacent S759 residue help maintain genome stability, we generated DNA fragments in which the codons for H760 or S759 were systematically changed to the other nineteen naturally occurring amino acids and attempted to clone them into a plasmid expressing pol III core (α-θ-ε subunits). Of the possible 38 mutants, only nine were successfully sub-cloned: three with substitutions at H760 and 6 with substitutions at S759. Three of the plasmid-encoded alleles, S759C, S759N, and S759T, exhibited mild to moderate mutator activity and were moved onto the chromosome for further characterization. These studies revealed altered phenotypes regarding deoxyribonucleotide base selectivity and ribonucleotide discrimination. We believe that these are the first dnaE mutants with such phenotypes to be reported in the literature.


Subject(s)
Catalytic Domain , DNA Polymerase III/chemistry , DNA Polymerase III/genetics , DNA/chemistry , DNA/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Alleles , Amino Acid Substitution , DNA Mismatch Repair , DNA Polymerase III/metabolism , DNA Replication , Deoxyribonucleotides/chemistry , Escherichia coli/enzymology , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genomic Instability , Models, Molecular , Mutation , Phenotype , Ribonucleotides/chemistry
7.
PLoS Biol ; 18(12): e3001030, 2020 12.
Article in English | MEDLINE | ID: mdl-33320856

ABSTRACT

With the ongoing COVID-19 (Coronavirus Disease 2019) pandemic, caused by the novel coronavirus SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), there is a need for sensitive, specific, and affordable diagnostic tests to identify infected individuals, not all of whom are symptomatic. The most sensitive test involves the detection of viral RNA using RT-qPCR (quantitative reverse transcription PCR), with many commercial kits now available for this purpose. However, these are expensive, and supply of such kits in sufficient numbers cannot always be guaranteed. We therefore developed a multiplex assay using well-established SARS-CoV-2 targets alongside a human cellular control (RPP30) and a viral spike-in control (Phocine Herpes Virus 1 [PhHV-1]), which monitor sample quality and nucleic acid extraction efficiency, respectively. Here, we establish that this test performs as well as widely used commercial assays, but at substantially reduced cost. Furthermore, we demonstrate >1,000-fold variability in material routinely collected by combined nose and throat swabbing and establish a statistically significant correlation between the detected level of human and SARS-CoV-2 nucleic acids. The inclusion of the human control probe in our assay therefore provides a quantitative measure of sample quality that could help reduce false-negative rates. We demonstrate the feasibility of establishing a robust RT-qPCR assay at approximately 10% of the cost of equivalent commercial assays, which could benefit low-resource environments and make high-volume testing affordable.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , RNA, Viral/analysis , SARS-CoV-2/isolation & purification , COVID-19 Testing/economics , Humans , Multiplex Polymerase Chain Reaction/economics , Reverse Transcriptase Polymerase Chain Reaction/economics , SARS-CoV-2/genetics
8.
Genes Dev ; 34(21-22): 1520-1533, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33060134

ABSTRACT

DNA replication is fundamental for cell proliferation in all organisms. Nonetheless, components of the replisome have been implicated in human disease, and here we report PRIM1 encoding the catalytic subunit of DNA primase as a novel disease gene. Using a variant classification agnostic approach, biallelic mutations in PRIM1 were identified in five individuals. PRIM1 protein levels were markedly reduced in patient cells, accompanied by replication fork asymmetry, increased interorigin distances, replication stress, and prolonged S-phase duration. Consequently, cell proliferation was markedly impaired, explaining the patients' extreme growth failure. Notably, phenotypic features distinct from those previously reported with DNA polymerase genes were evident, highlighting differing developmental requirements for this core replisome component that warrant future investigation.


Subject(s)
DNA Primase/genetics , Dwarfism/genetics , Fetal Growth Retardation/genetics , DNA Primase/chemistry , DNA Primase/deficiency , Dwarfism/diagnostic imaging , Dwarfism/pathology , Female , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology , Genetic Variation , Humans , Infant , Male , Pedigree , Syndrome
9.
Hum Mutat ; 40(8): 1063-1070, 2019 08.
Article in English | MEDLINE | ID: mdl-31045292

ABSTRACT

Microcephalic primordial dwarfism (MPD) is a group of rare single-gene disorders characterized by the extreme reduction in brain and body size from early development onwards. Proteins encoded by MPD-associated genes play important roles in fundamental cellular processes, notably genome replication and repair. Here we report the identification of four MPD individuals with biallelic variants in DNA2, which encodes an adenosine triphosphate (ATP)-dependent helicase/nuclease involved in DNA replication and repair. We demonstrate that the two intronic variants (c.1764-38_1764-37ins(53) and c.74+4A>C) found in these individuals substantially impair DNA2 transcript splicing. Additionally, we identify a missense variant (c.1963A>G), affecting a residue of the ATP-dependent helicase domain that is highly conserved between humans and yeast, with the resulting substitution (p.Thr655Ala) predicted to directly impact ATP/ADP (adenosine diphosphate) binding by DNA2. Our findings support the pathogenicity of these variants as biallelic hypomorphic mutations, establishing DNA2 as an MPD disease gene.


Subject(s)
DNA Helicases/genetics , Dwarfism/genetics , Genetic Variation , Microcephaly/genetics , Adolescent , Alleles , DNA Helicases/chemistry , Female , Genetic Predisposition to Disease , Humans , Introns , Male , Middle Aged , Models, Molecular , Mutagenesis, Insertional , Mutation, Missense , Polymorphism, Single Nucleotide
10.
Nat Genet ; 51(1): 96-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30478443

ABSTRACT

DNA methylation and Polycomb are key factors in the establishment of vertebrate cellular identity and fate. Here we report de novo missense mutations in DNMT3A, which encodes the DNA methyltransferase DNMT3A. These mutations cause microcephalic dwarfism, a hypocellular disorder of extreme global growth failure. Substitutions in the PWWP domain abrogate binding to the histone modifications H3K36me2 and H3K36me3, and alter DNA methylation in patient cells. Polycomb-associated DNA methylation valleys, hypomethylated domains encompassing developmental genes, become methylated with concomitant depletion of H3K27me3 and H3K4me3 bivalent marks. Such de novo DNA methylation occurs during differentiation of Dnmt3aW326R pluripotent cells in vitro, and is also evident in Dnmt3aW326R/+ dwarf mice. We therefore propose that the interaction of the DNMT3A PWWP domain with H3K36me2 and H3K36me3 normally limits DNA methylation of Polycomb-marked regions. Our findings implicate the interplay between DNA methylation and Polycomb at key developmental regulators as a determinant of organism size in mammals.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Dwarfism/genetics , Gain of Function Mutation/genetics , Microcephaly/genetics , Polycomb-Group Proteins/genetics , Animals , Cell Line, Tumor , Cells, Cultured , DNA Methyltransferase 3A , DNA Modification Methylases/genetics , Female , HeLa Cells , Histones/genetics , Humans , Male , Mice , Mice, Transgenic/genetics , Protein Binding/genetics , Regulatory Sequences, Nucleic Acid/genetics
11.
Gastroenterology ; 156(1): 145-159.e19, 2019 01.
Article in English | MEDLINE | ID: mdl-30273559

ABSTRACT

BACKGROUND & AIMS: RNase H2 is a holoenzyme, composed of 3 subunits (ribonuclease H2 subunits A, B, and C), that cleaves RNA:DNA hybrids and removes mis-incorporated ribonucleotides from genomic DNA through ribonucleotide excision repair. Ribonucleotide incorporation by eukaryotic DNA polymerases occurs during every round of genome duplication and produces the most frequent type of naturally occurring DNA lesion. We investigated whether intestinal epithelial proliferation requires RNase H2 function and whether RNase H2 activity is disrupted during intestinal carcinogenesis. METHODS: We generated mice with epithelial-specific deletion of ribonuclease H2 subunit B (H2bΔIEC) and mice that also had deletion of tumor-suppressor protein p53 (H2b/p53ΔIEC); we compared phenotypes with those of littermate H2bfl/fl or H2b/p53fl/fl (control) mice at young and old ages. Intestinal tissues were collected and analyzed by histology. We isolated epithelial cells, generated intestinal organoids, and performed RNA sequence analyses. Mutation signatures of spontaneous tumors from H2b/p53ΔIEC mice were characterized by exome sequencing. We collected colorectal tumor specimens from 467 patients, measured levels of ribonuclease H2 subunit B, and associated these with patient survival times and transcriptome data. RESULTS: The H2bΔIEC mice had DNA damage to intestinal epithelial cells and proliferative exhaustion of the intestinal stem cell compartment compared with controls and H2b/p53ΔIEC mice. However, H2b/p53ΔIEC mice spontaneously developed small intestine and colon carcinomas. DNA from these tumors contained T>G base substitutions at GTG trinucleotides. Analyses of transcriptomes of human colorectal tumors associated lower levels of RNase H2 with shorter survival times. CONCLUSIONS: In analyses of mice with disruption of the ribonuclease H2 subunit B gene and colorectal tumors from patients, we provide evidence that RNase H2 functions as a colorectal tumor suppressor. H2b/p53ΔIEC mice can be used to study the roles of RNase H2 in tissue-specific carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/enzymology , Genomic Instability , Intestinal Neoplasms/prevention & control , Intestine, Small/enzymology , Ribonuclease H/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/chemically induced , Colitis/enzymology , Colitis/genetics , Colitis/pathology , DNA Damage , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/pathology , Female , Genetic Predisposition to Disease , Humans , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Mice, Knockout , Phenotype , Ribonuclease H/deficiency , Ribonuclease H/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
Cancer Res ; 78(20): 5917-5926, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30154151

ABSTRACT

Because of imperfect discrimination against ribonucleoside triphosphates by the replicative DNA polymerases, large numbers of ribonucleotides are incorporated into the eukaryotic nuclear genome during S-phase. Ribonucleotides, by far the most common DNA lesion in replicating cells, destabilize the DNA, and an evolutionarily conserved DNA repair machinery, ribonucleotide excision repair (RER), ensures ribonucleotide removal. Whereas complete lack of RER is embryonically lethal, partial loss-of-function mutations in the genes encoding subunits of RNase H2, the enzyme essential for initiation of RER, cause the SLE-related type I interferonopathy Aicardi-Goutières syndrome. Here, we demonstrate that selective inactivation of RER in mouse epidermis results in spontaneous DNA damage and epidermal hyperproliferation associated with loss of hair follicle stem cells and hair follicle function. The animals developed keratinocyte intraepithelial neoplasia and invasive squamous cell carcinoma with complete penetrance, despite potent type I interferon production and skin inflammation. These results suggest that compromises to RER-mediated genome maintenance might represent an important tumor-promoting principle in human cancer.Significance: Selective inactivation of ribonucleotide excision repair by loss of RNase H2 in the murine epidermis results in spontaneous DNA damage, type I interferon response, skin inflammation, and development of squamous cell carcinoma. Cancer Res; 78(20); 5917-26. ©2018 AACR.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/prevention & control , DNA Repair , Ribonucleotides/genetics , Skin Neoplasms/genetics , Skin Neoplasms/prevention & control , Animals , Autoimmune Diseases of the Nervous System/metabolism , Cell Proliferation , DNA Damage , DNA Replication , Epidermis/metabolism , Humans , Inflammation , Interferons/metabolism , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mutation , Nervous System Malformations/metabolism , Ribonuclease H/metabolism , S Phase , Stem Cells/metabolism , Transcriptome
13.
Nature ; 559(7713): 285-289, 2018 07.
Article in English | MEDLINE | ID: mdl-29973717

ABSTRACT

The observation that BRCA1- and BRCA2-deficient cells are sensitive to inhibitors of poly(ADP-ribose) polymerase (PARP) has spurred the development of cancer therapies that use these inhibitors to target deficiencies in homologous recombination1. The cytotoxicity of PARP inhibitors depends on PARP trapping, the formation of non-covalent protein-DNA adducts composed of inhibited PARP1 bound to DNA lesions of unclear origins1-4. To address the nature of such lesions and the cellular consequences of PARP trapping, we undertook three CRISPR (clustered regularly interspersed palindromic repeats) screens to identify genes and pathways that mediate cellular resistance to olaparib, a clinically approved PARP inhibitor1. Here we present a high-confidence set of 73 genes, which when mutated cause increased sensitivity to PARP inhibitors. In addition to an expected enrichment for genes related to homologous recombination, we discovered that mutations in all three genes encoding ribonuclease H2 sensitized cells to PARP inhibition. We establish that the underlying cause of the PARP-inhibitor hypersensitivity of cells deficient in ribonuclease H2 is impaired ribonucleotide excision repair5. Embedded ribonucleotides, which are abundant in the genome of cells deficient in ribonucleotide excision repair, are substrates for cleavage by topoisomerase 1, resulting in PARP-trapping lesions that impede DNA replication and endanger genome integrity. We conclude that genomic ribonucleotides are a hitherto unappreciated source of PARP-trapping DNA lesions, and that the frequent deletion of RNASEH2B in metastatic prostate cancer and chronic lymphocytic leukaemia could provide an opportunity to exploit these findings therapeutically.


Subject(s)
CRISPR-Cas Systems , DNA Damage , Gene Editing , Neoplasms/genetics , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Ribonucleotides/genetics , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Cell Line , DNA Damage/drug effects , DNA Repair/genetics , DNA Replication , DNA Topoisomerases, Type I/metabolism , Female , Genes, BRCA1 , Genome/genetics , HeLa Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Ribonuclease H/deficiency , Ribonuclease H/genetics , Ribonuclease H/metabolism , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays
14.
Nucleic Acids Res ; 45(22): 12808-12815, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29106596

ABSTRACT

All DNA polymerases misincorporate ribonucleotides despite their preference for deoxyribonucleotides, and analysis of cultured cells indicates that mammalian mitochondrial DNA (mtDNA) tolerates such replication errors. However, it is not clear to what extent misincorporation occurs in tissues, or whether this plays a role in human disease. Here, we show that mtDNA of solid tissues contains many more embedded ribonucleotides than that of cultured cells, consistent with the high ratio of ribonucleotide to deoxynucleotide triphosphates in tissues, and that riboadenosines account for three-quarters of them. The pattern of embedded ribonucleotides changes in a mouse model of Mpv17 deficiency, which displays a marked increase in rGMPs in mtDNA. However, while the mitochondrial dGTP is low in the Mpv17-/- liver, the brain shows no change in the overall dGTP pool, leading us to suggest that Mpv17 determines the local concentration or quality of dGTP. Embedded rGMPs are expected to distort the mtDNA and impede its replication, and elevated rGMP incorporation is associated with early-onset mtDNA depletion in liver and late-onset multiple deletions in brain of Mpv17-/- mice. These findings suggest aberrant ribonucleotide incorporation is a primary mtDNA abnormality that can result in pathology.


Subject(s)
DNA, Mitochondrial/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Ribonucleotides/genetics , Sequence Deletion , Animals , Base Sequence , Brain/metabolism , Cell Line , Cell Line, Tumor , Disease Models, Animal , Humans , Liver/metabolism , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/deficiency
15.
Acta Neuropathol ; 134(6): 905-922, 2017 12.
Article in English | MEDLINE | ID: mdl-29030706

ABSTRACT

In search of novel germline alterations predisposing to tumors, in particular to gliomas, we studied a family with two brothers affected by anaplastic gliomas, and their father and paternal great-uncle diagnosed with prostate carcinoma. In this family, whole-exome sequencing yielded rare, simultaneously heterozygous variants in the Aicardi-Goutières syndrome (AGS) genes ADAR and RNASEH2B co-segregating with the tumor phenotype. AGS is a genetically induced inflammatory disease particularly of the brain, which has not been associated with a consistently increased cancer risk to date. By targeted sequencing, we identified novel ADAR and RNASEH2B variants, and a 3- to 17-fold frequency increase of the AGS mutations ADAR,c.577C>G;p.(P193A) and RNASEH2B,c.529G>A;p.(A177T) in the germline of familial glioma patients as well as in test and validation cohorts of glioblastomas and prostate carcinomas versus ethnicity-matched controls, whereby rare RNASEH2B variants were significantly more frequent in familial glioma patients. Tumors with ADAR or RNASEH2B variants recapitulated features of AGS, such as calcification and increased type I interferon expression. Patients carrying ADAR or RNASEH2B variants showed upregulation of interferon-stimulated gene (ISG) transcripts in peripheral blood as seen in AGS. An increased ISG expression was also induced by ADAR and RNASEH2B variants in tumor cells and was blocked by the JAK inhibitor Ruxolitinib. Our data implicate rare variants in the AGS genes ADAR and RNASEH2B and a type I interferon signature in glioma and prostate carcinoma risk and tumorigenesis, consistent with a genetic basis underlying inflammation-driven malignant transformation in glioma and prostate carcinoma development.


Subject(s)
Adenosine Deaminase/genetics , Genetic Predisposition to Disease , Interferon Type I/metabolism , Neoplasms/genetics , Neoplasms/metabolism , RNA-Binding Proteins/genetics , Ribonuclease H/genetics , Adenosine Deaminase/metabolism , Adult , Animals , Cells, Cultured , Cohort Studies , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Fibroblasts/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Male , Mice, Knockout , Molecular Dynamics Simulation , Neoplasms/drug therapy , Neoplasms/pathology , Phenotype , Polymorphism, Single Nucleotide , Protein Stability , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/genetics
16.
Nature ; 548(7668): 461-465, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28738408

ABSTRACT

DNA is strictly compartmentalized within the nucleus to prevent autoimmunity; despite this, cyclic GMP-AMP synthase (cGAS), a cytosolic sensor of double-stranded DNA, is activated in autoinflammatory disorders and by DNA damage. Precisely how cellular DNA gains access to the cytoplasm remains to be determined. Here, we report that cGAS localizes to micronuclei arising from genome instability in a mouse model of monogenic autoinflammation, after exogenous DNA damage and spontaneously in human cancer cells. Such micronuclei occur after mis-segregation of DNA during cell division and consist of chromatin surrounded by its own nuclear membrane. Breakdown of the micronuclear envelope, a process associated with chromothripsis, leads to rapid accumulation of cGAS, providing a mechanism by which self-DNA becomes exposed to the cytosol. cGAS is activated by chromatin, and consistent with a mitotic origin, micronuclei formation and the proinflammatory response following DNA damage are cell-cycle dependent. By combining live-cell laser microdissection with single cell transcriptomics, we establish that interferon-stimulated gene expression is induced in micronucleated cells. We therefore conclude that micronuclei represent an important source of immunostimulatory DNA. As micronuclei formed from lagging chromosomes also activate this pathway, recognition of micronuclei by cGAS may act as a cell-intrinsic immune surveillance mechanism that detects a range of neoplasia-inducing processes.


Subject(s)
Genomic Instability/immunology , Immunity, Innate/genetics , Micronuclei, Chromosome-Defective , Nucleotidyltransferases/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Chromatin/metabolism , Chromothripsis , Cytoplasm/enzymology , Cytoplasm/genetics , DNA/metabolism , DNA Damage , Female , Genomic Instability/genetics , Humans , Inflammation/enzymology , Inflammation/genetics , Lasers , Male , Mice , Microdissection , Mitosis , Nuclear Envelope/metabolism , Nucleotidyltransferases/genetics , Single-Cell Analysis , Transcriptome
17.
Nat Genet ; 48(1): 36-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595769

ABSTRACT

DNA lesions encountered by replicative polymerases threaten genome stability and cell cycle progression. Here we report the identification of mutations in TRAIP, encoding an E3 RING ubiquitin ligase, in patients with microcephalic primordial dwarfism. We establish that TRAIP relocalizes to sites of DNA damage, where it is required for optimal phosphorylation of H2AX and RPA2 during S-phase in response to ultraviolet (UV) irradiation, as well as fork progression through UV-induced DNA lesions. TRAIP is necessary for efficient cell cycle progression and mutations in TRAIP therefore limit cellular proliferation, providing a potential mechanism for microcephaly and dwarfism phenotypes. Human genetics thus identifies TRAIP as a component of the DNA damage response to replication-blocking DNA lesions.


Subject(s)
DNA Damage , Dwarfism/genetics , Mutation , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cell Proliferation/genetics , Child, Preschool , DNA Damage/radiation effects , Facies , Histones/genetics , Histones/metabolism , Humans , Microcephaly/genetics , Molecular Sequence Data , Phosphorylation , Replication Protein A/metabolism , S Phase/radiation effects , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ultraviolet Rays
18.
Nat Protoc ; 10(9): 1433-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26313479

ABSTRACT

Ribonucleotides are the most common noncanonical nucleotides incorporated into the genome of replicating cells. They are efficiently removed by ribonucleotide excision repair initiated by RNase H2 cleavage. In the absence of RNase H2, such embedded ribonucleotides can be used to track DNA polymerase activity in vivo. To determine their precise location in Saccharomyces cerevisiae, we developed embedded ribonucleotide sequencing (emRiboSeq), which uses recombinant RNase H2 to selectively create ligatable 3'-hydroxyl groups, in contrast to alternative methods that use alkaline hydrolysis. EmRiboSeq allows reproducible, strand-specific and potentially quantitative detection of embedded ribonucleotides at single-nucleotide resolution. For the genome-wide mapping of other noncanonical bases, RNase H2 can be replaced with specific nicking endonucleases in this protocol; we term this method endonuclease sequencing (EndoSeq). With the protocol taking <5 d to complete, these methods allow the in vivo study of DNA replication and repair, including the identification of replication origins and termination regions.


Subject(s)
Genomics/methods , Ribonuclease H/metabolism , Sequence Analysis/methods , Chromosome Mapping , Saccharomyces cerevisiae
19.
Nature ; 518(7540): 502-506, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25624100

ABSTRACT

The origin of mutations is central to understanding evolution and of key relevance to health. Variation occurs non-randomly across the genome, and mechanisms for this remain to be defined. Here we report that the 5' ends of Okazaki fragments have significantly increased levels of nucleotide substitution, indicating a replicative origin for such mutations. Using a novel method, emRiboSeq, we map the genome-wide contribution of polymerases, and show that despite Okazaki fragment processing, DNA synthesized by error-prone polymerase-α (Pol-α) is retained in vivo, comprising approximately 1.5% of the mature genome. We propose that DNA-binding proteins that rapidly re-associate post-replication act as partial barriers to Pol-δ-mediated displacement of Pol-α-synthesized DNA, resulting in incorporation of such Pol-α tracts and increased mutation rates at specific sites. We observe a mutational cost to chromatin and regulatory protein binding, resulting in mutation hotspots at regulatory elements, with signatures of this process detectable in both yeast and humans.


Subject(s)
DNA Replication/genetics , DNA/biosynthesis , DNA/genetics , Genome, Human/genetics , Mutation/genetics , Binding Sites , Chromatin/chemistry , Chromatin/metabolism , Conserved Sequence/genetics , DNA Polymerase I/metabolism , DNA Polymerase III/metabolism , DNA-Binding Proteins/metabolism , Evolution, Molecular , Humans , Models, Biological , Mutagenesis/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism
20.
J Clin Invest ; 125(1): 413-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25500883

ABSTRACT

Genome integrity is continuously challenged by the DNA damage that arises during normal cell metabolism. Biallelic mutations in the genes encoding the genome surveillance enzyme ribonuclease H2 (RNase H2) cause Aicardi-Goutières syndrome (AGS), a pediatric disorder that shares features with the autoimmune disease systemic lupus erythematosus (SLE). Here we determined that heterozygous parents of AGS patients exhibit an intermediate autoimmune phenotype and demonstrated a genetic association between rare RNASEH2 sequence variants and SLE. Evaluation of patient cells revealed that SLE- and AGS-associated mutations impair RNase H2 function and result in accumulation of ribonucleotides in genomic DNA. The ensuing chronic low level of DNA damage triggered a DNA damage response characterized by constitutive p53 phosphorylation and senescence. Patient fibroblasts exhibited constitutive upregulation of IFN-stimulated genes and an enhanced type I IFN response to the immunostimulatory nucleic acid polyinosinic:polycytidylic acid and UV light irradiation, linking RNase H2 deficiency to potentiation of innate immune signaling. Moreover, UV-induced cyclobutane pyrimidine dimer formation was markedly enhanced in ribonucleotide-containing DNA, providing a mechanism for photosensitivity in RNase H2-associated SLE. Collectively, our findings implicate RNase H2 in the pathogenesis of SLE and suggest a role of DNA damage-associated pathways in the initiation of autoimmunity.


Subject(s)
Autoimmunity/genetics , DNA Repair , Lupus Erythematosus, Systemic/genetics , Pyrimidine Dimers/metabolism , Cell Proliferation , Cells, Cultured , DNA Mutational Analysis , Gene Expression , Heterozygote , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Pyrimidine Dimers/genetics , Ribonuclease H/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...