Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Aging Cell ; 22(9): e13893, 2023 09.
Article in English | MEDLINE | ID: mdl-37547972

ABSTRACT

Cellular senescence constitutes a generally irreversible proliferation barrier, accompanied by macromolecular damage and metabolic rewiring. Several senescence types have been identified based on the initiating stimulus, such as replicative (RS), stress-induced (SIS) and oncogene-induced senescence (OIS). These senescence subtypes are heterogeneous and often develop subset-specific phenotypes. Reduced protein synthesis is considered a senescence hallmark, but whether this trait pertains to various senescence subtypes and if distinct molecular mechanisms are involved remain largely unknown. Here, we analyze large published or experimentally produced RNA-seq and Ribo-seq datasets to determine whether major translation-regulating entities such as ribosome stalling, the presence of uORFs/dORFs and IRES elements may differentially contribute to translation deficiency in senescence subsets. We show that translation-regulating mechanisms may not be directly relevant to RS, however uORFs are significantly enriched in SIS. Interestingly, ribosome stalling, uORF/dORF patterns and IRES elements comprise predominant mechanisms upon OIS, strongly correlating with Notch pathway activation. Our study provides for the first time evidence that major translation dysregulation mechanisms/patterns occur during cellular senescence, but at different rates depending on the stimulus type. The degree at which those mechanisms accumulate directly correlates with translation deficiency levels. Our thorough analysis contributes to elucidating crucial and so far unknown differences in the translation machinery between senescence subsets.


Subject(s)
Cellular Senescence , Ribosomes , Cellular Senescence/genetics , Ribosomes/genetics , Ribosomes/metabolism , Protein Biosynthesis
2.
Nucleic Acids Res ; 51(16): 8575-8586, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37470822

ABSTRACT

In addition to being essential for gene expression, transcription is crucial for the maintenance of genome integrity. Here, we undertook a systematic approach, to monitor the assembly kinetics of the pre-initiating RNA Polymerase (Pol) II at promoters at steady state and different stages during recovery from UV irradiation-stress, when pre-initiation and initiation steps have been suggested to be transiently shut down. Taking advantage of the reversible dissociation of pre-initiating Pol II after high salt treatment, we found that de novo recruitment of the available Pol II molecules at active promoters not only persists upon UV at all times tested but occurs significantly faster in the early phase of recovery (2 h) than in unexposed human fibroblasts at the majority of active genes. Our method unveiled groups of genes with significantly different pre-initiation complex (PIC) assembly dynamics after UV that present distinct rates of UV-related mutational signatures in melanoma tumours, providing functional relevance to the importance of keeping transcription initiation active during UV recovery. Our findings uncover novel mechanistic insights further detailing the multilayered transcriptional response to genotoxic stress and link PIC assembly dynamics after exposure to genotoxins with cancer mutational landscapes.


Subject(s)
RNA Polymerase II , Transcription Initiation, Genetic , Humans , DNA Damage , Mutagenesis , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription, Genetic , Ultraviolet Rays , Fibroblasts/metabolism , DNA Repair
3.
Comput Struct Biotechnol J ; 21: 614-629, 2023.
Article in English | MEDLINE | ID: mdl-36659919

ABSTRACT

The timing and location of writing and erasing of histone modifications determine gene expression programs and are tightly controlled processes. One such modification is the monoubiquitination of histone H2B (H2Bub), whose precise level during transcription elongation is dynamically regulated by the synergistic action of RNF20/40 ubiquitin-ligase and the de-ubiquitinase (DUB) of the ATXN7L3-containing DUB modules. Here, we characterize the dynamics of H2Bub in transcription and explore its role in perspective with the recently updated model of UV damage-induced transcription reorganization. Employing integrative analysis of genome-wide high-throughput approaches, transcription inhibitors and ATXN7L3-DUB knockdown cells, we find that H2Bub levels and patterns depend on intron-exon architecture both in steady state and upon UV. Importantly, our analysis reveals a widespread redistribution of this histone mark, rather than a uniform loss as previously suggested, which closely mirrors the post-UV dynamics of elongating RNA Polymerase II (RNAPII) at transcribed loci. The observed effects are due to a direct inter-dependence on RNAPII local concentration and speed, and we show that deficient ATXN7L3-mediated DUB activity leads to increased elongation rates in both non-irradiated and irradiated conditions. Our data and the implementation of a high-resolution computational framework reveal that the H2Bub pattern follows that of RNAPII, both in the ATXNL3 knockdown and in response to UV guaranteeing faithful elongation speed, especially in the context of the transcription-driven DNA damage response.

4.
Genome Med ; 14(1): 78, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35879783

ABSTRACT

BACKGROUND: Synovial fibroblasts (SFs) are specialized cells of the synovium that provide nutrients and lubricants for the proper function of diarthrodial joints. Recent evidence appreciates the contribution of SF heterogeneity in arthritic pathologies. However, the normal SF profiles and the molecular networks that govern the transition from homeostatic to arthritic SF heterogeneity remain poorly defined. METHODS: We applied a combined analysis of single-cell (sc) transcriptomes and epigenomes (scRNA-seq and scATAC-seq) to SFs derived from naïve and hTNFtg mice (mice that overexpress human TNF, a murine model for rheumatoid arthritis), by employing the Seurat and ArchR packages. To identify the cellular differentiation lineages, we conducted velocity and trajectory analysis by combining state-of-the-art algorithms including scVelo, Slingshot, and PAGA. We integrated the transcriptomic and epigenomic data to infer gene regulatory networks using ArchR and custom-implemented algorithms. We performed a canonical correlation analysis-based integration of murine data with publicly available datasets from SFs of rheumatoid arthritis patients and sought to identify conserved gene regulatory networks by utilizing the SCENIC algorithm in the human arthritic scRNA-seq atlas. RESULTS: By comparing SFs from healthy and hTNFtg mice, we revealed seven homeostatic and two disease-specific subsets of SFs. In healthy synovium, SFs function towards chondro- and osteogenesis, tissue repair, and immune surveillance. The development of arthritis leads to shrinkage of homeostatic SFs and favors the emergence of SF profiles marked by Dkk3 and Lrrc15 expression, functioning towards enhanced inflammatory responses and matrix catabolic processes. Lineage inference analysis indicated that specific Thy1+ SFs at the root of trajectories lead to the intermediate Thy1+/Dkk3+/Lrrc15+ SF states and culminate in a destructive and inflammatory Thy1- SF identity. We further uncovered epigenetically primed gene programs driving the expansion of these arthritic SFs, regulated by NFkB and new candidates, such as Runx1. Cross-species analysis of human/mouse arthritic SF data determined conserved regulatory and transcriptional networks. CONCLUSIONS: We revealed a dynamic SF landscape from health to arthritis providing a functional genomic blueprint to understand the joint pathophysiology and highlight the fibroblast-oriented therapeutic targets for combating chronic inflammatory and destructive arthritic disease.


Subject(s)
Arthritis, Rheumatoid , Single-Cell Analysis , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Humans , Inflammation/metabolism , Membrane Proteins/metabolism , Mice , Synovial Membrane/metabolism , Synovial Membrane/pathology
7.
Cells ; 10(4)2021 04 10.
Article in English | MEDLINE | ID: mdl-33920220

ABSTRACT

Cockayne syndrome (CS) is a DNA repair syndrome characterized by a broad spectrum of clinical manifestations such as neurodegeneration, premature aging, developmental impairment, photosensitivity and other symptoms. Mutations in Cockayne syndrome protein B (CSB) are present in the vast majority of CS patients and in other DNA repair-related pathologies. In the literature, the role of CSB in different DNA repair pathways has been highlighted, however, new CSB functions have been identified in DNA transcription, mitochondrial biology, telomere maintenance and p53 regulation. Herein, we present an overview of identified structural elements and processes that impact on CSB activity and its post-translational modifications, known to balance the different roles of the protein not only during normal conditions but most importantly in stress situations. Moreover, since CSB has been found to be overexpressed in a number of different tumors, its role in cancer is presented and possible therapeutic targeting is discussed.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Animals , Cockayne Syndrome/metabolism , DNA Damage , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Repair , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Gene Expression Regulation, Neoplastic , Humans , Models, Molecular , Mutation , Neoplasms/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Conformation , Protein Processing, Post-Translational
8.
Nucleic Acids Res ; 49(11): e64, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33693861

ABSTRACT

Specific capture of chromatin fractions with distinct and well-defined features has emerged as both challenging and a key strategy towards a comprehensive understanding of genome biology. In this context, we developed aniFOUND (accelerated native isolation of factors on unscheduled nascent DNA), an antibody-free method, which can label, capture, map and characterise nascent chromatin fragments that are synthesized in response to specific cues outside S-phase. We used the 'unscheduled' DNA synthesis (UDS) that takes place during the repair of UV-induced DNA lesions and coupled the captured chromatin to high-throughput analytical technologies. By mass-spectrometry we identified several factors with no previously known role in UVC-DNA damage response (DDR) as well as known DDR proteins. We experimentally validated the repair-dependent recruitment of the chromatin remodeller RSF1 and the cohesin-loader NIPBL at sites of UVC-induced photolesions. Developing aniFOUND-seq, a protocol for mapping UDS activity with high resolution, allowed us to monitor the landscape of UVC repair-synthesis events genome wide. We further resolved repair efficacy of the rather unexplored repeated genome, in particular rDNA and telomeres. In summary, aniFOUND delineates the proteome composition and genomic landscape of chromatin loci with specific features by integrating state-of-the-art 'omics' technologies to promote a comprehensive view of their function.


Subject(s)
Chromatin/metabolism , DNA Repair , Genomics/methods , Proteomics/methods , Cell Cycle Proteins/metabolism , Cell Line , DNA/biosynthesis , DNA/radiation effects , High-Throughput Nucleotide Sequencing , Humans , Mass Spectrometry , Nuclear Proteins/metabolism , Proteome , Repetitive Sequences, Nucleic Acid , Trans-Activators/metabolism , Ultraviolet Rays
9.
J Immunol ; 206(3): 607-620, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33443087

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by exuberant deposition of extracellular matrix components, leading to the deterioration of lung architecture and respiratory functions. Profibrotic mechanisms are controlled by multiple regulatory molecules, including MAPKs, in turn regulated by multiple phosphorylation cascades. MAP3K8 is an MAPK kinase kinase suggested to pleiotropically regulate multiple pathogenic pathways in the context of inflammation and cancer; however, a possible role in the pathogenesis of IPF has not been investigated. In this report, MAP3K8 mRNA levels were found decreased in the lungs of IPF patients and of mice upon bleomycin-induced pulmonary fibrosis. Ubiquitous genetic deletion of Map3k8 in mice exacerbated the modeled disease, whereas bone marrow transfer experiments indicated that although MAP3K8 regulatory functions are active in both hematopoietic and nonhematopoietic cells, Map3k8 in hematopoietic cells has a more dominant role. Macrophage-specific deletion of Map3k8 was further found to be sufficient for disease exacerbation thus confirming a major role for macrophages in pulmonary fibrotic responses and suggesting a main role for Map3k8 in the homeostasis of their effector functions in the lung. Map3k8 deficiency was further shown to be associated with decreased Cox-2 expression, followed by a decrease in PGE2 production in the lung; accordingly, exogenous administration of PGE2 reduced inflammation and reversed the exacerbated fibrotic profile of Map3k8 -/- mice. Therefore, MAP3K8 has a central role in the regulation of inflammatory responses and Cox-2-mediated PGE2 production in the lung, and the attenuation of its expression is integral to pulmonary fibrosis development.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Inflammation/metabolism , Lung/pathology , MAP Kinase Kinase Kinases/genetics , Proto-Oncogene Proteins/genetics , Pulmonary Fibrosis/metabolism , Animals , Bone Marrow Transplantation , Cells, Cultured , Fibrosis , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Cell Mol Life Sci ; 78(7): 3443-3465, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33247761

ABSTRACT

During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.


Subject(s)
Calcium Signaling , Central Nervous System/pathology , Homeodomain Proteins/metabolism , Neuroblastoma/pathology , Neuronal Outgrowth , Neurons/pathology , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Central Nervous System/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Neuroblastoma/metabolism , Neurons/metabolism , Phosphorylation , Signal Transduction , Tumor Suppressor Proteins/genetics
11.
Nat Commun ; 11(1): 916, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060325

ABSTRACT

Inhibition of transcription caused by DNA damage-impaired RNA polymerase II (Pol II) elongation conceals a local increase in de novo transcription, slowly progressing from Transcription Start Sites (TSSs) to gene ends. Although associated with accelerated repair of Pol II-encountered lesions and limited mutagenesis, it is still unclear how this mechanism is maintained during genotoxic stress-recovery. Here we uncover a widespread gain in chromatin accessibility and preservation of the active H3K27ac mark after UV-irradiation. The concomitant increase in Pol II escape from promoter-proximal pause (PPP) sites of most active genes, PROMPTs and enhancer RNAs favors unrestrained initiation, as evidenced by the synthesis of nascent RNAs including start RNAs. Accordingly, drug-inhibition of PPP-release replenishes levels of pre-initiating Pol II at TSSs after UV. Our data show that such continuous engagement of Pol II molecules ensures maximal transcription-driven repair throughout expressed genes and regulatory loci. Importantly, revealing this unanticipated regulatory layer of UV-response provides physiological relevant traction to the emerging concept that Pol II initiation rate is determined by pause-release dynamics.


Subject(s)
DNA Repair , Transcription Initiation Site , Transcription, Genetic , Chromatin/genetics , Chromatin/metabolism , DNA Damage , Gene Expression Regulation , Humans , Promoter Regions, Genetic , RNA/genetics , RNA/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Regulatory Sequences, Nucleic Acid
12.
Nat Commun ; 8(1): 2076, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233992

ABSTRACT

Complex molecular responses preserve gene expression accuracy and genome integrity in the face of environmental perturbations. Here we report that, in response to UV irradiation, RNA polymerase II (RNAPII) molecules are dynamically and synchronously released from promoter-proximal regions into elongation to promote uniform and accelerated surveillance of the whole transcribed genome. The maximised influx of de novo released RNAPII correlates with increased damage-sensing, as confirmed by RNAPII progressive accumulation at dipyrimidine sites and by the average slow-down of elongation rates in gene bodies. In turn, this transcription elongation 'safe' mode guarantees efficient DNA repair regardless of damage location, gene size and transcription level. Accordingly, we detect low and homogenous rates of mutational signatures associated with UV exposure or cigarette smoke across all active genes. Our study reveals a novel advantage for transcription regulation at the promoter-proximal level and provides unanticipated insights into how active transcription shapes the mutagenic landscape of cancer genomes.


Subject(s)
DNA Damage/genetics , Mutation Rate , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , Transcription Elongation, Genetic/radiation effects , Cell Line , DNA Damage/radiation effects , DNA Repair/radiation effects , Humans , RNA Polymerase II/radiation effects , Ultraviolet Rays/adverse effects
13.
Adv Exp Med Biol ; 1007: 17-39, 2017.
Article in English | MEDLINE | ID: mdl-28840550

ABSTRACT

DNA damage poses a constant threat to genome integrity taking a variety of shapes and arising by normal cellular metabolism or environmental insults. Human syndromes, characterized by increased cancer pre-disposition or early onset of age-related pathology and developmental abnormalities, often result from defective DNA damage responses and compromised genome integrity. Over the last decades intensive research worldwide has made important contributions to our understanding of the molecular mechanisms underlying genomic instability and has substantiated the importance of DNA repair in cancer prevention in the general population. In this chapter, we discuss Nucleotide Excision Repair pathway, the causative role of its components in disease-related pathology and recent technological achievements that decipher mutational landscapes and may facilitate pathological classification and personalized therapy.


Subject(s)
DNA Damage , DNA Repair , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Genomic Instability , Humans
14.
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
15.
Cold Spring Harb Perspect Biol ; 5(8): a012625, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23906714

ABSTRACT

Transcriptional arrest caused by DNA damage is detrimental for cells and organisms as it impinges on gene expression and thereby on cell growth and survival. To alleviate transcriptional arrest, cells trigger a transcription-dependent genome surveillance pathway, termed transcription-coupled nucleotide excision repair (TC-NER) that ensures rapid removal of such transcription-impeding DNA lesions and prevents persistent stalling of transcription. Defective TC-NER is causatively linked to Cockayne syndrome, a rare severe genetic disorder with multisystem abnormalities that results in patients' death in early adulthood. Here we review recent data on how damage-arrested transcription is actively coupled to TC-NER in mammals and discuss new emerging models concerning the role of TC-NER-specific factors in this process.


Subject(s)
Chromatin/physiology , Cockayne Syndrome/genetics , DNA Damage/genetics , DNA Repair/physiology , Mammals/physiology , Models, Genetic , Transcription, Genetic/physiology , Animals , Chromatin/genetics , DNA Repair/genetics , Mammals/genetics , Transcription, Genetic/genetics
16.
Nat Genet ; 44(5): 598-602, 2012 May.
Article in English | MEDLINE | ID: mdl-22466611

ABSTRACT

Transcription-coupled nucleotide-excision repair (TC-NER) is a subpathway of NER that efficiently removes the highly toxic RNA polymerase II blocking lesions in DNA. Defective TC-NER gives rise to the human disorders Cockayne syndrome and UV-sensitive syndrome (UV(S)S). NER initiating factors are known to be regulated by ubiquitination. Using a SILAC-based proteomic approach, we identified UVSSA (formerly known as KIAA1530) as part of a UV-induced ubiquitinated protein complex. Knockdown of UVSSA resulted in TC-NER deficiency. UVSSA was found to be the causative gene for UV(S)S, an unresolved NER deficiency disorder. The UVSSA protein interacts with elongating RNA polymerase II, localizes specifically to UV-induced lesions, resides in chromatin-associated TC-NER complexes and is implicated in stabilizing the TC-NER master organizing protein ERCC6 (also known as CSB) by delivering the deubiquitinating enzyme USP7 to TC-NER complexes. Together, these findings indicate that UVSSA-USP7­mediated stabilization of ERCC6 represents a critical regulatory mechanism of TC-NER in restoring gene expression.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cockayne Syndrome/genetics , DNA Helicases/chemistry , DNA Repair Enzymes/chemistry , DNA Repair/genetics , Transcription, Genetic , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Carrier Proteins/antagonists & inhibitors , Cells, Cultured , Chromatin/genetics , DNA Damage/genetics , DNA Damage/radiation effects , DNA Helicases/genetics , DNA Repair/radiation effects , DNA Repair Enzymes/genetics , Humans , Immunoprecipitation , Mutation/genetics , Poly-ADP-Ribose Binding Proteins , Proteomics , RNA Polymerase II/metabolism , RNA, Small Interfering/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ultraviolet Rays
17.
DNA Repair (Amst) ; 10(7): 743-50, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21622031

ABSTRACT

A network of DNA damage surveillance systems is triggered by sensing of DNA lesions and the initiation of a signal transduction cascade that activates genome-protection pathways including nucleotide excision repair (NER). NER operates through coordinated assembly of repair factors into pre- and post-incision complexes. Recent work identifies RPA as a key regulator of the transition from dual incision to repair-synthesis in UV-irradiated non-cycling cells, thereby averting the generation of unprocessed repair intermediates. These intermediates could lead to recombinogenic events and trigger a persistent ATR-dependent checkpoint signaling. It is now evident that DNA damage signaling is not limited to NER proficient cells. ATR-dependent checkpoint activation also occurs in UV-exposed non-cycling repair deficient cells coinciding with the formation of endonuclease APE1-mediated DNA strand breaks. In addition, the encounter of elongating RNA polymerase II (RNAPIIo) with DNA damage lesions and its persistent stalling provides a strong DNA damage signaling leading to cell cycle arrest, apoptosis and increased mutagenesis. The mechanism underlying the strong and strand specific induction of UV-induced mutations in NER deficient cells has been recently resolved by the finding that gene transcription itself increases UV-induced mutagenesis in a strand specific manner via increased deamination of cytosines. The cell removes the RNAPIIo-blocking DNA lesions by transcription-coupled repair (TC-NER) without displacement of the DNA damage stalled RNAPIIo. Deficiency in TC-NER associates with mutations in the CSA and CSB genes giving rise to the rare human disorder Cockayne syndrome (CS). CSB functions as a repair coupling factor to attract NER proteins, chromatin remodelers and the CSA-E3-ubiquitin ligase complex to the stalled RNAPIIo; CSA is dispensable for attraction of NER proteins, yet in cooperation with CSB is required to recruit XAB2, the nucleosomal binding protein HMGN1 and TFIIS. The molecular mechanisms by which these proteins bring about efficient TC-NER and trigger signaling after transcription arrest remain elusive; particularly the role of chromatin remodeling in TC-NER needs to be clarified in the context of anticipated structural changes that allow repair and transcription restart.


Subject(s)
DNA Damage , DNA Repair , DNA/genetics , Genomic Instability , Transcription, Genetic , Chromatin Assembly and Disassembly , DNA/metabolism , DNA/radiation effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , DNA Replication , Genome, Human , Humans , Mutagenesis , Mutation , Phosphorylation , Signal Transduction , Ultraviolet Rays
18.
J Cell Biol ; 192(3): 401-15, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21282463

ABSTRACT

Single-stranded DNA gaps that might arise by futile repair processes can lead to mutagenic events and challenge genome integrity. Nucleotide excision repair (NER) is an evolutionarily conserved repair mechanism, essential for removal of helix-distorting DNA lesions. In the currently prevailing model, NER operates through coordinated assembly of repair factors into pre- and post-incision complexes; however, its regulation in vivo is poorly understood. Notably, the transition from dual incision to repair synthesis should be rigidly synchronized as it might lead to accumulation of unprocessed repair intermediates. We monitored NER regulatory events in vivo using sequential UV irradiations. Under conditions that allow incision yet prevent completion of repair synthesis or ligation, preincision factors can reassociate with new damage sites. In contrast, replication protein A remains at the incomplete NER sites and regulates a feedback loop from completion of DNA repair synthesis to subsequent damage recognition, independently of ATR signaling. Our data reveal an important function for replication protein A in averting further generation of DNA strand breaks that could lead to mutagenic and recombinogenic events.


Subject(s)
DNA Repair , Replication Protein A/physiology , Cells, Cultured , DNA Replication , DNA, Single-Stranded/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Genome, Human , Humans , Replication Protein A/genetics , Replication Protein A/metabolism , Ultraviolet Rays/adverse effects
19.
Mol Cell Biol ; 30(20): 4828-39, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20713449

ABSTRACT

Nucleotide excision repair (NER) operates through coordinated assembly of repair factors into pre- and postincision complexes. The postincision step of NER includes gap-filling DNA synthesis and ligation. However, the exact composition of this NER-associated DNA synthesis complex in vivo and the dynamic interactions of the factors involved are not well understood. Using immunofluorescence, chromatin immunoprecipitation, and live-cell protein dynamic studies, we show that replication factor C (RFC) is implicated in postincision NER in mammalian cells. Small interfering RNA-mediated knockdown of RFC impairs upstream removal of UV lesions and abrogates the downstream recruitment of DNA polymerase delta. Unexpectedly, RFC appears dispensable for PCNA recruitment yet is required for the subsequent recruitment of DNA polymerases to PCNA, indicating that RFC is essential to stably load the polymerase clamp to start DNA repair synthesis at 3' termini. The kinetic studies are consistent with a model in which RFC exchanges dynamically at sites of repair. However, its persistent localization at stalled NER complexes suggests that RFC remains targeted to the repair complex even after loading of PCNA. We speculate that RFC associates with the downstream 5' phosphate after loading; such interaction would prevent possible signaling events initiated by the RFC-like Rad17 and may assist in unloading of PCNA.


Subject(s)
DNA Polymerase III/metabolism , DNA Repair/physiology , Replication Protein C/metabolism , Binding Sites , Cell Line , Cytarabine/pharmacology , DNA Damage , DNA Replication , Fluorescence Recovery After Photobleaching , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydroxyurea/pharmacology , Kinetics , Models, Biological , Nucleic Acid Synthesis Inhibitors/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Replication Protein C/antagonists & inhibitors , Replication Protein C/genetics , Ultraviolet Rays
20.
Mol Cell ; 38(5): 637-48, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20541997

ABSTRACT

Transcription-coupled nucleotide excision repair (TC-NER) allows RNA polymerase II (RNAPII)-blocking lesions to be rapidly removed from the transcribed strand of active genes. Defective TCR in humans is associated with Cockayne syndrome (CS), typically caused by defects in either CSA or CSB. Here, we show that CSB contains a ubiquitin-binding domain (UBD). Cells expressing UBD-less CSB (CSB(del)) have phenotypes similar to those of cells lacking CSB, but these can be suppressed by appending a heterologous UBD, so ubiquitin binding is essential for CSB function. Surprisingly, CSB(del) remains capable of assembling nucleotide excision repair factors and repair synthesis proteins around damage-stalled RNAPII, but such repair complexes fail to excise the lesion. Together, our results indicate an essential role for protein ubiquitylation and CSB's UBD in triggering damage incision during TC-NER and allow us to integrate the function of CSA and CSB in a model for the process.


Subject(s)
DNA Helicases , DNA Repair Enzymes , DNA Repair , Ubiquitin/metabolism , Amino Acid Sequence , Cell Line/radiation effects , Cell Nucleus/metabolism , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Humans , Molecular Sequence Data , Mutation , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Tetrahydrofolate Dehydrogenase/genetics , Ubiquitin/genetics , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...