Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 20(12): 766-784, 2019 12.
Article in English | MEDLINE | ID: mdl-31558824

ABSTRACT

The spatiotemporal control of RNA polymerase II (Pol II)-mediated gene transcription is tightly and intricately regulated. In addition, preservation of the integrity of the DNA template is required so as to ensure unperturbed transcription, particularly since DNA is continually challenged by different types of damaging agents that can form transcription-blocking DNA lesions (TBLs), which impede transcription elongation and cause transcription stress. To overcome the highly cytotoxic effects of TBLs, an intricate cellular response has evolved, in which the transcription-coupled nucleotide excision repair (TC-NER) pathway has a central role in removing TBLs specifically from the transcribed strand. Damage detection by stalling of the transcribing Pol II is highly efficient, but a stalled Pol II complex may create an even bigger problem by interfering with repair of the lesions, and overall with transcription and replication. In this Review, we discuss the effects of different types of DNA damage on Pol II, important concepts of transcription stress, the manner in which TBLs are removed by TC-NER and how different tissues respond to TBLs. We also discuss the role of TBLs in ageing and the complex genotype-phenotype correlations of TC-NER hereditary disorders.


Subject(s)
DNA Damage , DNA Repair , DNA Replication , Genetic Diseases, Inborn , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , RNA Polymerase II/genetics
2.
Mol Cell ; 82(7): 1343-1358.e8, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35271816

ABSTRACT

Nucleotide excision repair (NER) counteracts the onset of cancer and aging by removing helix-distorting DNA lesions via a "cut-and-patch"-type reaction. The regulatory mechanisms that drive NER through its successive damage recognition, verification, incision, and gap restoration reaction steps remain elusive. Here, we show that the RAD5-related translocase HLTF facilitates repair through active eviction of incised damaged DNA together with associated repair proteins. Our data show a dual-incision-dependent recruitment of HLTF to the NER incision complex, which is mediated by HLTF's HIRAN domain that binds 3'-OH single-stranded DNA ends. HLTF's translocase motor subsequently promotes the dissociation of the stably damage-bound incision complex together with the incised oligonucleotide, allowing for an efficient PCNA loading and initiation of repair synthesis. Our findings uncover HLTF as an important NER factor that actively evicts DNA damage, thereby providing additional quality control by coordinating the transition between the excision and DNA synthesis steps to safeguard genome integrity.


Subject(s)
DNA Repair , DNA-Binding Proteins , DNA/genetics , DNA/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/genetics
3.
Nature ; 613(7942): 187-194, 2023 01.
Article in English | MEDLINE | ID: mdl-36544021

ABSTRACT

R-loops are RNA-DNA-hybrid-containing nucleic acids with important cellular roles. Deregulation of R-loop dynamics can lead to DNA damage and genome instability1, which has been linked to the action of endonucleases such as XPG2-4. However, the mechanisms and cellular consequences of such processing have remained unclear. Here we identify a new population of RNA-DNA hybrids in the cytoplasm that are R-loop-processing products. When nuclear R-loops were perturbed by depleting the RNA-DNA helicase senataxin (SETX) or the breast cancer gene BRCA1 (refs. 5-7), we observed XPG- and XPF-dependent cytoplasmic hybrid formation. We identify their source as a subset of stable, overlapping nuclear hybrids with a specific nucleotide signature. Cytoplasmic hybrids bind to the pattern recognition receptors cGAS and TLR3 (ref. 8), activating IRF3 and inducing apoptosis. Excised hybrids and an R-loop-induced innate immune response were also observed in SETX-mutated cells from patients with ataxia oculomotor apraxia type 2 (ref. 9) and in BRCA1-mutated cancer cells10. These findings establish RNA-DNA hybrids as immunogenic species that aberrantly accumulate in the cytoplasm after R-loop processing, linking R-loop accumulation to cell death through the innate immune response. Aberrant R-loop processing and subsequent innate immune activation may contribute to many diseases, such as neurodegeneration and cancer.


Subject(s)
Cytoplasm , DNA , Innate Immunity Recognition , Nucleic Acid Heteroduplexes , R-Loop Structures , RNA , Humans , Apoptosis , Cytoplasm/immunology , Cytoplasm/metabolism , DNA/chemistry , DNA/immunology , DNA Helicases/genetics , DNA Helicases/metabolism , Genes, BRCA1 , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , Neoplasms , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/immunology , R-Loop Structures/immunology , RNA/chemistry , RNA/immunology , RNA Helicases/genetics , RNA Helicases/metabolism , Spinocerebellar Ataxias/genetics
4.
Cell ; 153(5): 988-99, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706737

ABSTRACT

Lymphocyte activation is initiated by a global increase in messenger RNA synthesis. However, the mechanisms driving transcriptome amplification during the immune response are unknown. By monitoring single-stranded DNA genome wide, we show that the genome of naive cells is poised for rapid activation. In G0, ∼90% of promoters from genes to be expressed in cycling lymphocytes are polymerase loaded but unmelted and support only basal transcription. Furthermore, the transition from abortive to productive elongation is kinetically limiting, causing polymerases to accumulate nearer to transcription start sites. Resting lymphocytes also limit the expression of the transcription factor IIH complex, including XPB and XPD helicases involved in promoter melting and open complex extension. To date, two rate-limiting steps have been shown to control global gene expression in eukaryotes: preinitiation complex assembly and polymerase pausing. Our studies identify promoter melting as a third key regulatory step and propose that this mechanism ensures a prompt lymphocyte response to invading pathogens.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , Lymphocyte Activation , Lymphocytes/metabolism , Promoter Regions, Genetic , Animals , B-Lymphocytes/immunology , Cell Line, Tumor , DNA, Single-Stranded/metabolism , Enhancer Elements, Genetic , Genome-Wide Association Study , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Transcription Factor TFIIH/metabolism , Transcription, Genetic
5.
Cell ; 150(6): 1182-95, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980979

ABSTRACT

Ubiquitin-dependent signaling during the DNA damage response (DDR) to double-strand breaks (DSBs) is initiated by two E3 ligases, RNF8 and RNF168, targeting histone H2A and H2AX. RNF8 is the first ligase recruited to the damage site, and RNF168 follows RNF8-dependent ubiquitination. This suggests that RNF8 initiates H2A/H2AX ubiquitination with K63-linked ubiquitin chains and RNF168 extends them. Here, we show that RNF8 is inactive toward nucleosomal H2A, whereas RNF168 catalyzes the monoubiquitination of the histones specifically on K13-15. Structure-based mutagenesis of RNF8 and RNF168 RING domains shows that a charged residue determines whether nucleosomal proteins are recognized. We find that K63 ubiquitin chains are conjugated to RNF168-dependent H2A/H2AX monoubiquitination at K13-15 and not on K118-119. Using a mutant of RNF168 unable to target histones but still catalyzing ubiquitin chains at DSBs, we show that ubiquitin chains per se are insufficient for signaling, but RNF168 target ubiquitination is required for DDR.


Subject(s)
Histones/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Histones/chemistry , Humans , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Structure, Tertiary , Scattering, Small Angle , Ubiquitin-Protein Ligases/chemistry , X-Ray Diffraction
6.
Nat Rev Mol Cell Biol ; 15(7): 465-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24954209

ABSTRACT

Nucleotide excision repair (NER) eliminates various structurally unrelated DNA lesions by a multiwise 'cut and patch'-type reaction. The global genome NER (GG-NER) subpathway prevents mutagenesis by probing the genome for helix-distorting lesions, whereas transcription-coupled NER (TC-NER) removes transcription-blocking lesions to permit unperturbed gene expression, thereby preventing cell death. Consequently, defects in GG-NER result in cancer predisposition, whereas defects in TC-NER cause a variety of diseases ranging from ultraviolet radiation-sensitive syndrome to severe premature ageing conditions such as Cockayne syndrome. Recent studies have uncovered new aspects of DNA-damage detection by NER, how NER is regulated by extensive post-translational modifications, and the dynamic chromatin interactions that control its efficiency. Based on these findings, a mechanistic model is proposed that explains the complex genotype-phenotype correlations of transcription-coupled repair disorders.


Subject(s)
Aging/genetics , DNA Repair/physiology , Neoplasms/genetics , DNA Damage , DNA Repair/genetics , Humans , Models, Biological , Ubiquitin/physiology
8.
Nucleic Acids Res ; 51(18): e93, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37522336

ABSTRACT

Transcription-coupled nucleotide excision repair (TC-NER) is an important DNA repair mechanism that protects against the negative effects of transcription-blocking DNA lesions. Hereditary TC-NER deficiencies cause pleiotropic and often severe neurodegenerative and progeroid symptoms. While multiple assays have been developed to determine TC-NER activity for clinical and research purposes, monitoring TC-NER is hampered by the low frequency of repair events occurring in transcribed DNA. 'Recovery of RNA Synthesis' is widely used as indirect TC-NER assay based on the notion that lesion-blocked transcription only resumes after successful TC-NER. Here, we show that measuring novel synthesis of a protein after its compound-induced degradation prior to DNA damage induction is an equally effective but more versatile manner to indirectly monitor DNA repair activity in transcribed genes. This 'Recovery of Protein Synthesis' (RPS) assay can be adapted to various degradable proteins and readouts, including imaging and immunoblotting. Moreover, RPS allows real-time monitoring of TC-NER activity in various living cells types and even in differentiated tissues of living organisms. To illustrate its utility, we show that DNA repair in transcribed genes declines in aging muscle tissue of C. elegans. Therefore, the RPS assay constitutes an important novel clinical and research tool to investigate transcription-coupled DNA repair.


Subject(s)
Caenorhabditis elegans , DNA Repair , Protein Biosynthesis , Transcription, Genetic , Animals , Caenorhabditis elegans/physiology , DNA/metabolism , DNA Damage , Aging/metabolism , Muscles/metabolism
9.
Nucleic Acids Res ; 51(17): 9055-9074, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37470997

ABSTRACT

The SWI/SNF family of ATP-dependent chromatin remodeling complexes is implicated in multiple DNA damage response mechanisms and frequently mutated in cancer. The BAF, PBAF and ncBAF complexes are three major types of SWI/SNF complexes that are functionally distinguished by their exclusive subunits. Accumulating evidence suggests that double-strand breaks (DSBs) in transcriptionally active DNA are preferentially repaired by a dedicated homologous recombination pathway. We show that different BAF, PBAF and ncBAF subunits promote homologous recombination and are rapidly recruited to DSBs in a transcription-dependent manner. The PBAF and ncBAF complexes promote RNA polymerase II eviction near DNA damage to rapidly initiate transcriptional silencing, while the BAF complex helps to maintain this transcriptional silencing. Furthermore, ARID1A-containing BAF complexes promote RNaseH1 and RAD52 recruitment to facilitate R-loop resolution and DNA repair. Our results highlight how multiple SWI/SNF complexes perform different functions to enable DNA repair in the context of actively transcribed genes.


Subject(s)
Chromosomal Proteins, Non-Histone , R-Loop Structures , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA , DNA Repair/genetics , Homologous Recombination/genetics , Humans
10.
N Engl J Med ; 385(16): 1485-1495, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34644472

ABSTRACT

BACKGROUND: Cell-culture-derived influenza vaccines may enable a closer antigenic match to circulating strains of influenza virus by avoiding egg-adapted mutations. METHODS: We evaluated the efficacy of a cell-culture-derived quadrivalent inactivated influenza vaccine (IIV4c) using a Madin-Darby canine kidney cell line in children and adolescents 2 to less than 18 years of age. During three influenza seasons, participants from eight countries were enrolled in an observer-blinded, randomized clinical trial comparing IIV4c with a noninfluenza vaccine (meningococcal ACWY). All the participants received a dose of a trial vaccine. Children 2 to less than 9 years of age without previous influenza vaccination who were assigned to the IIV4c group received a second dose on day 29; their counterparts who were assigned to the comparator group received placebo. Participants were followed for at least 180 days for efficacy and safety. The presence of influenza virus in nasopharyngeal swabs from participants with influenza-like illness was confirmed by reverse-transcriptase-polymerase-chain-reaction assay and viral culture. A Cox proportional-hazards model was used to evaluate the efficacy of IIV4c as measured by the first occurrence of laboratory-confirmed type A or B influenza (primary end point). RESULTS: Between 2017 and 2019, a total of 4514 participants were randomly assigned to receive IIV4c or the meningococcal ACWY vaccine. Laboratory-confirmed influenza occurred in 175 of 2257 participants (7.8%) in the IIV4c group and in 364 of 2252 participants (16.2%) in the comparator group, and the efficacy of IIV4c was 54.6% (95% confidence interval [CI], 45.7 to 62.1). Efficacy was 80.7% (95% CI, 69.2 to 87.9) against influenza A/H1N1, 42.1% (95% CI, 20.3 to 57.9) against influenza A/H3N2, and 47.6% (95% CI, 31.4 to 60.0) against influenza B. IIV4c showed consistent vaccine efficacy in subgroups according to age, sex, race, and previous influenza vaccination. The incidences of adverse events were similar in the IIV4c group and the comparator group. CONCLUSIONS: IIV4c provided protection against influenza in healthy children and adolescents across seasons, regardless of previous influenza vaccination. (Funded by Seqirus; EudraCT number, 2016-002883-15; ClinicalTrials.gov number, NCT03165617.).


Subject(s)
Immunogenicity, Vaccine , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Adolescent , Antibodies, Viral/blood , Child , Child, Preschool , Female , Humans , Influenza Vaccines/adverse effects , Male , Meningococcal Vaccines/immunology , Orthomyxoviridae/isolation & purification , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction , Single-Blind Method , Vaccines, Inactivated/immunology
12.
Hum Mol Genet ; 30(18): 1711-1720, 2021 08 28.
Article in English | MEDLINE | ID: mdl-33909043

ABSTRACT

Trichothiodystrophy (TTD) is a rare hereditary neurodevelopmental disorder defined by sulfur-deficient brittle hair and nails and scaly skin, but with otherwise remarkably variable clinical features. The photosensitive TTD (PS-TTD) forms exhibits in addition to progressive neuropathy and other features of segmental accelerated aging and is associated with impaired genome maintenance and transcription. New factors involved in various steps of gene expression have been identified for the different non-photosensitive forms of TTD (NPS-TTD), which do not appear to show features of premature aging. Here, we identify alanyl-tRNA synthetase 1 and methionyl-tRNA synthetase 1 variants as new gene defects that cause NPS-TTD. These variants result in the instability of the respective gene products alanyl- and methionyl-tRNA synthetase. These findings extend our previous observations that TTD mutations affect the stability of the corresponding proteins and emphasize this phenomenon as a common feature of TTD. Functional studies in skin fibroblasts from affected individuals demonstrate that these new variants also impact on the rate of tRNA charging, which is the first step in protein translation. The extension of reduced abundance of TTD factors to translation as well as transcription redefines TTD as a syndrome in which proteins involved in gene expression are unstable.


Subject(s)
Alanine-tRNA Ligase/genetics , Methionine-tRNA Ligase/genetics , Trichothiodystrophy Syndromes/genetics , Alanine-tRNA Ligase/metabolism , Child , Enzyme Stability/genetics , Female , Humans , Methionine-tRNA Ligase/metabolism , Trichothiodystrophy Syndromes/enzymology , Trichothiodystrophy Syndromes/pathology , Whole Genome Sequencing
13.
Mol Cell ; 59(6): 885-6, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26384662

ABSTRACT

In this issue, Li et al. (2015) uncover roles for the XPB and XPD helicases and for XPA during damage verification in nucleotide excision repair, supporting a novel tripartite damage checking mechanism that combines extreme versatility with narrow specificity.


Subject(s)
DNA Adducts/genetics , DNA-Binding Proteins/physiology , Transcription Factor TFIIH/physiology , Xeroderma Pigmentosum Group A Protein/physiology , Animals , Humans
14.
Cell Mol Life Sci ; 79(3): 166, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35230528

ABSTRACT

The XPG/ERCC5 endonuclease was originally identified as the causative gene for Xeroderma Pigmentosum complementation group G. Ever since its discovery, in depth biochemical, structural and cell biological studies have provided detailed mechanistic insight into its function in excising DNA damage in nucleotide excision repair, together with the ERCC1-XPF endonuclease. In recent years, it has become evident that XPG has additional important roles in genome maintenance that are independent of its function in NER, as XPG has been implicated in protecting replication forks by promoting homologous recombination as well as in resolving R-loops. Here, we provide an overview of the multitasking of XPG in genome maintenance, by describing in detail how its activity in NER is regulated and the evidence that points to important functions outside of NER. Furthermore, we present the various disease phenotypes associated with inherited XPG deficiency and discuss current ideas on how XPG deficiency leads to these different types of disease.


Subject(s)
DNA-Binding Proteins/genetics , Endonucleases/genetics , Genome/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , DNA Repair/genetics , DNA Replication/genetics , Humans , Xeroderma Pigmentosum/genetics
15.
Neurogenetics ; 23(4): 271-274, 2022 10.
Article in English | MEDLINE | ID: mdl-35920923

ABSTRACT

Cockayne syndrome is a rare inherited DNA repair multisystemic disorder. Here, we aim to raise awareness of the phenotypic resemblances between Cockayne syndrome and the neurodevelopmental disorder caused by pathogenic variants in MORC2, a gene also involved in DNA repair. Using exome sequencing, we identified a de novo pathogenic variant in MORC2 in our patient. Our patient's phenotype was characterized by multiple features evocative of Cockayne syndrome. Based on our patient's phenotype, in addition to the phenotypic description of patients with pathogenic variants in MORC2 reported in the literature, we suggest that pathogenic variants in this gene are associated with a Cockayne-like phenotype.


Subject(s)
Cockayne Syndrome , Neurodevelopmental Disorders , Humans , Cockayne Syndrome/genetics , Phenotype , Neurodevelopmental Disorders/genetics , Exome Sequencing , Transcription Factors/genetics
16.
Am J Hum Genet ; 105(2): 434-440, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31374204

ABSTRACT

Brittle and "tiger-tail" hair is the diagnostic hallmark of trichothiodystrophy (TTD), a rare recessive disease associated with a wide spectrum of clinical features including ichthyosis, intellectual disability, decreased fertility, and short stature. As a result of premature abrogation of terminal differentiation, the hair is brittle and fragile and contains reduced cysteine content. Hypersensitivity to UV light is found in about half of individuals with TTD; all of these individuals harbor bi-allelic mutations in components of the basal transcription factor TFIIH, and these mutations lead to impaired nucleotide excision repair and basal transcription. Different genes have been found to be associated with non-photosensitive TTD (NPS-TTD); these include MPLKIP (also called TTDN1), GTF2E2 (also called TFIIEß), and RNF113A. However, a relatively large group of these individuals with NPS-TTD have remained genetically uncharacterized. Here we present the identification of an NPS-TTD-associated gene, threonyl-tRNA synthetase (TARS), found by next-generation sequencing of a group of uncharacterized individuals with NPS-TTD. One individual has compound heterozygous TARS variants, c.826A>G (p.Lys276Glu) and c.1912C>T (p.Arg638∗), whereas a second individual is homozygous for the TARS variant: c.680T>C (p.Leu227Pro). We showed that these variants have a profound effect on TARS protein stability and enzymatic function. Our results expand the spectrum of genes involved in TTD to include genes implicated in amino acid charging of tRNA, which is required for the last step in gene expression, namely protein translation. We previously proposed that some of the TTD-specific features derive from subtle transcription defects as a consequence of unstable transcription factors. We now extend the definition of TTD from a transcription syndrome to a "gene-expression" syndrome.


Subject(s)
Hair Diseases/pathology , Mutation , Threonine-tRNA Ligase/genetics , Trichothiodystrophy Syndromes/pathology , Alleles , Amino Acid Sequence , Case-Control Studies , Hair Diseases/genetics , Humans , Phenotype , Sequence Homology , Transcription Factor TFIIH/genetics , Trichothiodystrophy Syndromes/genetics
17.
Am J Hum Genet ; 104(3): 520-529, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30824121

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes responsible for charging tRNA molecules with cognate amino acids. Consistent with the essential function and ubiquitous expression of ARSs, mutations in 32 of the 37 ARS-encoding loci cause severe, early-onset recessive phenotypes. Previous genetic and functional data suggest a loss-of-function mechanism; however, our understanding of the allelic and locus heterogeneity of ARS-related disease is incomplete. Cysteinyl-tRNA synthetase (CARS) encodes the enzyme that charges tRNACys with cysteine in the cytoplasm. To date, CARS variants have not been implicated in any human disease phenotype. Here, we report on four subjects from three families with complex syndromes that include microcephaly, developmental delay, and brittle hair and nails. Each affected person carries bi-allelic CARS variants: one individual is compound heterozygous for c.1138C>T (p.Gln380∗) and c.1022G>A (p.Arg341His), two related individuals are compound heterozygous for c.1076C>T (p.Ser359Leu) and c.1199T>A (p.Leu400Gln), and one individual is homozygous for c.2061dup (p.Ser688Glnfs∗2). Measurement of protein abundance, yeast complementation assays, and assessments of tRNA charging indicate that each CARS variant causes a loss-of-function effect. Compared to subjects with previously reported ARS-related diseases, individuals with bi-allelic CARS variants are unique in presenting with a brittle-hair-and-nail phenotype, which most likely reflects the high cysteine content in human keratins. In sum, our efforts implicate CARS variants in human inherited disease, expand the locus and clinical heterogeneity of ARS-related clinical phenotypes, and further support impaired tRNA charging as the primary mechanism of recessive ARS-related disease.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/etiology , Developmental Disabilities/etiology , Hair Diseases/etiology , Microcephaly/etiology , Mutation , Nail Diseases/etiology , Adult , Amino Acid Sequence , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/pathology , Developmental Disabilities/enzymology , Developmental Disabilities/pathology , Female , Genes, Recessive , Genetic Predisposition to Disease , Hair Diseases/enzymology , Hair Diseases/pathology , Humans , Male , Microcephaly/enzymology , Microcephaly/pathology , Nail Diseases/enzymology , Nail Diseases/pathology , Pedigree , Phenotype , Prognosis , Sequence Homology , Young Adult
18.
Nucleic Acids Res ; 48(1): 231-248, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31722399

ABSTRACT

Cockayne Syndrome (CS) is a severe neurodegenerative and premature aging autosomal-recessive disease, caused by inherited defects in the CSA and CSB genes, leading to defects in transcription-coupled nucleotide excision repair (TC-NER) and consequently hypersensitivity to ultraviolet (UV) irradiation. TC-NER is initiated by lesion-stalled RNA polymerase II, which stabilizes the interaction with the SNF2/SWI2 ATPase CSB to facilitate recruitment of the CSA E3 Cullin ubiquitin ligase complex. However, the precise biochemical connections between CSA and CSB are unknown. The small ubiquitin-like modifier SUMO is important in the DNA damage response. We found that CSB, among an extensive set of other target proteins, is the most dynamically SUMOylated substrate in response to UV irradiation. Inhibiting SUMOylation reduced the accumulation of CSB at local sites of UV irradiation and reduced recovery of RNA synthesis. Interestingly, CSA is required for the efficient clearance of SUMOylated CSB. However, subsequent proteomic analysis of CSA-dependent ubiquitinated substrates revealed that CSA does not ubiquitinate CSB in a UV-dependent manner. Surprisingly, we found that CSA is required for the ubiquitination of the largest subunit of RNA polymerase II, RPB1. Combined, our results indicate that the CSA, CSB, RNA polymerase II triad is coordinated by ubiquitin and SUMO in response to UV irradiation. Furthermore, our work provides a resource of SUMO targets regulated in response to UV or ionizing radiation.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA Repair , Poly-ADP-Ribose Binding Proteins/genetics , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Ubiquitin/genetics , Cell Line, Transformed , Cell Line, Tumor , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Expression Regulation , Gene Regulatory Networks , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Transcription Factors/metabolism , Ubiquitin/metabolism , Ubiquitination , Ultraviolet Rays
19.
Nature ; 523(7558): 53-8, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26106861

ABSTRACT

In response to DNA damage, tissue homoeostasis is ensured by protein networks promoting DNA repair, cell cycle arrest or apoptosis. DNA damage response signalling pathways coordinate these processes, partly by propagating gene-expression-modulating signals. DNA damage influences not only the abundance of messenger RNAs, but also their coding information through alternative splicing. Here we show that transcription-blocking DNA lesions promote chromatin displacement of late-stage spliceosomes and initiate a positive feedback loop centred on the signalling kinase ATM. We propose that initial spliceosome displacement and subsequent R-loop formation is triggered by pausing of RNA polymerase at DNA lesions. In turn, R-loops activate ATM, which signals to impede spliceosome organization further and augment ultraviolet-irradiation-triggered alternative splicing at the genome-wide level. Our findings define R-loop-dependent ATM activation by transcription-blocking lesions as an important event in the DNA damage response of non-replicating cells, and highlight a key role for spliceosome displacement in this process.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage/physiology , Signal Transduction , Spliceosomes/metabolism , Alternative Splicing/physiology , Cell Line , Chromatin/metabolism , DNA-Directed RNA Polymerases/metabolism , Enzyme Activation , Humans , Ultraviolet Rays
20.
Mol Cell ; 51(4): 469-79, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973375

ABSTRACT

Chromatin remodeling is tightly linked to all DNA-transacting activities. To study chromatin remodeling during DNA repair, we established quantitative fluorescence imaging methods to measure the exchange of histones in chromatin in living cells. We show that particularly H2A and H2B are evicted and replaced at an accelerated pace at sites of UV-induced DNA damage. This accelerated exchange of H2A/H2B is facilitated by SPT16, one of the two subunits of the histone chaperone FACT (facilitates chromatin transcription) but largely independent of its partner SSRP1. Interestingly, SPT16 is targeted to sites of UV light-induced DNA damage-arrested transcription and is required for efficient restart of RNA synthesis upon damage removal. Together, our data uncover an important role for chromatin dynamics at the crossroads of transcription and the UV-induced DNA damage response.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Histones/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , Ultraviolet Rays , Blotting, Western , Cell Cycle Proteins , Chromatin Immunoprecipitation , Cross-Linking Reagents/pharmacology , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , HeLa Cells , High Mobility Group Proteins/genetics , Histones/genetics , Humans , Nucleosomes/genetics , RNA/genetics , RNA/metabolism , Transcription Factors , Transcriptional Elongation Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL