Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2857, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565848

ABSTRACT

PARP2 is a DNA-dependent ADP-ribosyl transferase (ARTs) enzyme with Poly(ADP-ribosyl)ation activity that is triggered by DNA breaks. It plays a role in the Base Excision Repair pathway, where it has overlapping functions with PARP1. However, additional roles for PARP2 have emerged in the response of cells to replication stress. In this study, we demonstrate that PARP2 promotes replication stress-induced telomere fragility and prevents telomere loss following chronic induction of oxidative DNA lesions and BLM helicase depletion. Telomere fragility results from the activity of the break-induced replication pathway (BIR). During this process, PARP2 promotes DNA end resection, strand invasion and BIR-dependent mitotic DNA synthesis by orchestrating POLD3 recruitment and activity. Our study has identified a role for PARP2 in the response to replication stress. This finding may lead to the development of therapeutic approaches that target DNA-dependent ART enzymes, particularly in cancer cells with high levels of replication stress.


Subject(s)
DNA Repair , DNA , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , DNA/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , Telomere/genetics , Telomere/metabolism
2.
J Mol Biol ; 436(1): 168207, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37481154

ABSTRACT

Alternative DNA structures that differ from the canonical B-form of DNA can arise from repetitive sequences and play beneficial roles in many cellular processes such as gene regulation and chromatin organization. However, they also threaten genomic stability in several ways including mutagenesis and collisions with replication and/or transcription machinery, which lead to genomic instability that is associated with human disease. Thus, the careful regulation of non-B-DNA structure formation and resolution is crucial for the maintenance of genome integrity. Several protein factors have been demonstrated to associate with alternative DNA structures to facilitate their removal, one of which is the ADP-ribose transferase (ART) PARP1 (also called ADP-ribosyltransferase diphtheria toxin-like 1 or ARTD1), a multifaceted DNA repair enzyme that recognizes single- and double-stranded DNA breaks and synthesizes chains of poly (ADP-ribose) (PAR) to recruit DNA repair proteins. It is now well appreciated that PARP1 recognizes several nucleic acid structures beyond DNA lesions, including stalled replication forks, DNA hairpins and cruciforms, R-loops, and DNA G-quadruplexes (G4 DNA). In this review, we summarize the current evidence of a direct association of PARP1 with each of these aforementioned alternative DNA structures, as well as discuss the role of PARP1 in the prevention of non-B-DNA structure-induced genetic instability. We will focus on the mechanisms of the recognition and binding by PARP1 to each alternative structure and the structure-based stimulation of PARP1 catalytic activity upon binding. Finally, we will discuss some of the outstanding gaps in the literature and offer speculative insight for questions that remain to be experimentally addressed.


Subject(s)
DNA, Cruciform , Genomic Instability , Poly (ADP-Ribose) Polymerase-1 , Humans , DNA/chemistry , DNA Repair , Gene Expression Regulation , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Ribose/chemistry , Animals
3.
NAR Cancer ; 5(2): zcad019, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37180029

ABSTRACT

Centromeres play a crucial role in DNA segregation by mediating the cohesion and separation of sister chromatids during cell division. Centromere dysfunction, breakage or compromised centromeric integrity can generate aneuploidies and chromosomal instability, which are cellular features associated with cancer initiation and progression. Maintaining centromere integrity is thus essential for genome stability. However, the centromere itself is prone to DNA breaks, likely due to its intrinsically fragile nature. Centromeres are complex genomic loci that are composed of highly repetitive DNA sequences and secondary structures and require the recruitment and homeostasis of a centromere-associated protein network. The molecular mechanisms engaged to preserve centromere inherent structure and respond to centromeric damage are not fully understood and remain a subject of ongoing research. In this article, we provide a review of the currently known factors that contribute to centromeric dysfunction and the molecular mechanisms that mitigate the impact of centromere damage on genome stability. Finally, we discuss the potential therapeutic strategies that could arise from a deeper understanding of the mechanisms preserving centromere integrity.

4.
Nucleic Acids Res ; 51(5): 2215-2237, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36794853

ABSTRACT

PARP1 is a DNA-dependent ADP-Ribose transferase with ADP-ribosylation activity that is triggered by DNA breaks and non-B DNA structures to mediate their resolution. PARP1 was also recently identified as a component of the R-loop-associated protein-protein interaction network, suggesting a potential role for PARP1 in resolving this structure. R-loops are three-stranded nucleic acid structures that consist of a RNA-DNA hybrid and a displaced non-template DNA strand. R-loops are involved in crucial physiological processes but can also be a source of genome instability if persistently unresolved. In this study, we demonstrate that PARP1 binds R-loops in vitro and associates with R-loop formation sites in cells which activates its ADP-ribosylation activity. Conversely, PARP1 inhibition or genetic depletion causes an accumulation of unresolved R-loops which promotes genomic instability. Our study reveals that PARP1 is a novel sensor for R-loops and highlights that PARP1 is a suppressor of R-loop-associated genomic instability.


Subject(s)
Genomic Instability , Poly (ADP-Ribose) Polymerase-1 , R-Loop Structures , Humans , DNA/chemistry , DNA Repair , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA/chemistry
5.
Sci Transl Med ; 14(662): eabq3215, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103513

ABSTRACT

Arginine-rich dipeptide repeat proteins (R-DPRs), abnormal translational products of a GGGGCC hexanucleotide repeat expansion in C9ORF72, play a critical role in C9ORF72-related amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), the most common genetic form of the disorders (c9ALS/FTD). R-DPRs form liquid condensates in vitro, induce stress granule formation in cultured cells, aggregate, and sometimes coaggregate with TDP-43 in postmortem tissue from patients with c9ALS/FTD. However, how these processes are regulated is unclear. Here, we show that loss of poly(ADP-ribose) (PAR) suppresses neurodegeneration in c9ALS/FTD fly models and neurons differentiated from patient-derived induced pluripotent stem cells. Mechanistically, PAR induces R-DPR condensation and promotes R-DPR-induced stress granule formation and TDP-43 aggregation. Moreover, PAR associates with insoluble R-DPR and TDP-43 in postmortem tissue from patients. These findings identified PAR as a promoter of R-DPR toxicity and thus a potential target for treating c9ALS/FTD.


Subject(s)
Frontotemporal Dementia , Arginine , C9orf72 Protein/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dipeptides/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Poly Adenosine Diphosphate Ribose
6.
Res Sq ; 2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35441168

ABSTRACT

The repertoire of coronavirus disease 2019 (COVID-19)-mediated adverse health outcomes has continued to expand in infected patients, including the susceptibility to developing long-COVID; however, the molecular underpinnings at the cellular level are poorly defined. In this study, we report that SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection triggers host cell genome instability by modulating the expression of molecules of DNA repair and mutagenic translesion synthesis. Further, SARS-CoV-2 infection causes genetic alterations, such as increased mutagenesis, telomere dysregulation, and elevated microsatellite instability (MSI). The MSI phenotype was coupled to reduced MLH1, MSH6, and MSH2 in infected cells. Strikingly, pre-treatment of cells with the REV1-targeting translesion DNA synthesis inhibitor, JH-RE-06, suppresses SARS-CoV-2 proliferation and dramatically represses the SARS-CoV-2-dependent genome instability. Mechanistically, JH-RE-06 treatment induces autophagy, which we hypothesize limits SARS-CoV-2 proliferation and, therefore, the hijacking of host-cell genome instability pathways. These results have implications for understanding the pathobiological consequences of COVID-19.

7.
Cell Mol Life Sci ; 79(4): 215, 2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35348914

ABSTRACT

The ADP-ribose transferase (ART) family comprises 17 enzymes that catalyze mono- or poly-ADP-ribosylation, a post-translational modification of proteins. Present in all subcellular compartments, ARTs are implicated in a growing number of biological processes including DNA repair, replication, transcription regulation, intra- and extra-cellular signaling, viral infection and cell death. Five members of the family, PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 are mainly described for their crucial functions in the maintenance of genome stability. It is well established that the most describedrole of PARP1, 2 and 3 is the repair of DNA lesions while tankyrases 1 and 2 are crucial for maintaining the integrity of telomeres. Telomeres, nucleoprotein complexes located at the ends of eukaryotic chromosomes, utilize their unique structure and associated set of proteins to orchestrate the mechanisms necessary for their own protection and replication. While the functions of tankyrases 1 and 2 at telomeres are well known, several studies have also brought PARP1, 2 and 3 to the forefront of telomere protection. The singular quality of the telomeric environment has highlighted protein interactions and molecular pathways distinct from those described throughout the genome. The aim of this review is to provide an overview of the current knowledge on the multiple roles of PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 in the maintenance and preservation of telomere integrity.


Subject(s)
ADP Ribose Transferases , Telomere , ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , DNA Repair , Genomic Instability , Humans , Telomere/genetics , Telomere/metabolism
8.
Methods Mol Biol ; 2444: 141-159, 2022.
Article in English | MEDLINE | ID: mdl-35290636

ABSTRACT

Mammalian telomeres are guanine-rich sequences which cap the ends of linear chromosomes. While recognized as sites sensitive to oxidative stress, studies on the consequences of oxidative damage to telomeres have been primarily limited to experimental conditions which cause oxidative damage throughout the whole genome and cell. We developed a chemoptogenetic tool (FAP-mCER-TRF1) to specifically induce singlet oxygen at telomeres, resulting in the formation of the common oxidative lesion 8-oxo-guanine. Here, we describe this tool and detail how to generate cell lines which express FAP-mCER-TRF1 at telomeres and verify the formation of 8-oxo-guanine.


Subject(s)
DNA Damage , Telomere , Animals , Guanine/analogs & derivatives , Guanine/metabolism , Mammals/genetics , Oxidative Stress/genetics , Telomere/genetics , Telomere/metabolism
9.
Methods Mol Biol ; 2102: 237-249, 2020.
Article in English | MEDLINE | ID: mdl-31989559

ABSTRACT

A key component of sustained cellular proliferation is the preservation of telomere integrity. Telomeres are nucleoprotein structures that cap and protect linear chromosomes. Their linearity and repetitive sequence represent a challenge for the replication machinery and cause telomere shortening, fragility, and losses. Here we describe the common technique of quantitative fluorescent in situ hybridization that allows for the scoring of telomere aberrations and measurement of telomere length directly on metaphase chromosomes through the use of highly specific peptide nucleic acid probes.


Subject(s)
In Situ Hybridization, Fluorescence/methods , Telomere/genetics , Animals , Cells, Cultured , Humans , Metaphase , Microscopy, Fluorescence , Nucleic Acid Probes , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/genetics , Telomere Homeostasis , Workflow
10.
Proc Natl Acad Sci U S A ; 116(37): 18435-18444, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451640

ABSTRACT

Reactive oxygen species (ROS) play important roles in aging, inflammation, and cancer. Mitochondria are an important source of ROS; however, the spatiotemporal ROS events underlying oxidative cellular damage from dysfunctional mitochondria remain unresolved. To this end, we have developed and validated a chemoptogenetic approach that uses a mitochondrially targeted fluorogen-activating peptide (Mito-FAP) to deliver a photosensitizer MG-2I dye exclusively to this organelle. Light-mediated activation (660 nm) of the Mito-FAP-MG-2I complex led to a rapid loss of mitochondrial respiration, decreased electron transport chain complex activity, and mitochondrial fragmentation. Importantly, one round of singlet oxygen produced a persistent secondary wave of mitochondrial superoxide and hydrogen peroxide lasting for over 48 h after the initial insult. By following ROS intermediates, we were able to detect hydrogen peroxide in the nucleus through ratiometric analysis of the oxidation of nuclear cysteine residues. Despite mitochondrial DNA (mtDNA) damage and nuclear oxidative stress induced by dysfunctional mitochondria, there was a lack of gross nuclear DNA strand breaks and apoptosis. Targeted telomere analysis revealed fragile telomeres and telomere loss as well as 53BP1-positive telomere dysfunction-induced foci (TIFs), indicating that DNA double-strand breaks occurred exclusively in telomeres as a direct consequence of mitochondrial dysfunction. These telomere defects activated ataxia-telangiectasia mutated (ATM)-mediated DNA damage repair signaling. Furthermore, ATM inhibition exacerbated the Mito-FAP-induced mitochondrial dysfunction and sensitized cells to apoptotic cell death. This profound sensitivity of telomeres through hydrogen peroxide induced by dysregulated mitochondria reveals a crucial mechanism of telomere-mitochondria communication underlying the pathophysiological role of mitochondrial ROS in human diseases.


Subject(s)
Mitochondria/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Telomere/metabolism , Apoptosis/drug effects , Cell Cycle , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , DNA, Mitochondrial/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Membrane Potentials , Mitochondrial Diseases/metabolism , Oxidative Stress , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Signal Transduction , Superoxides/metabolism , Superoxides/toxicity , Tumor Suppressor p53-Binding Protein 1/metabolism
11.
Nat Struct Mol Biol ; 26(8): 695-703, 2019 08.
Article in English | MEDLINE | ID: mdl-31332353

ABSTRACT

UV-DDB, a key protein in human global nucleotide excision repair (NER), binds avidly to abasic sites and 8-oxo-guanine (8-oxoG), suggesting a noncanonical role in base excision repair (BER). We investigated whether UV-DDB can stimulate BER for these two common forms of DNA damage, 8-oxoG and abasic sites, which are repaired by 8-oxoguanine glycosylase (OGG1) and apurinic/apyrimidinic endonuclease (APE1), respectively. UV-DDB increased both OGG1 and APE1 strand cleavage and stimulated subsequent DNA polymerase ß-gap filling activity by 30-fold. Single-molecule real-time imaging revealed that UV-DDB forms transient complexes with OGG1 or APE1, facilitating their dissociation from DNA. Furthermore, UV-DDB moves to sites of 8-oxoG repair in cells, and UV-DDB depletion sensitizes cells to oxidative DNA damage. We propose that UV-DDB is a general sensor of DNA damage in both NER and BER pathways, facilitating damage recognition in the context of chromatin.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/physiology , Cell Line , DNA Damage , DNA Glycosylases/chemistry , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Mapping , Pyrimidine Dimers/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Single Molecule Imaging , Substrate Specificity , Xeroderma Pigmentosum/pathology
12.
Mol Cell ; 75(1): 117-130.e6, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31101499

ABSTRACT

Telomeres are essential for genome stability. Oxidative stress caused by excess reactive oxygen species (ROS) accelerates telomere shortening. Although telomeres are hypersensitive to ROS-mediated 8-oxoguanine (8-oxoG) formation, the biological effect of this common lesion at telomeres is poorly understood because ROS have pleiotropic effects. Here we developed a chemoptogenetic tool that selectively produces 8-oxoG only at telomeres. Acute telomeric 8-oxoG formation increased telomere fragility in cells lacking OGG1, the enzyme that removes 8-oxoG, but did not compromise cell survival. However, chronic telomeric 8-oxoG induction over time shortens telomeres and impairs cell growth. Accumulation of telomeric 8-oxoG in chronically exposed OGG1-deficient cells triggers replication stress, as evidenced by mitotic DNA synthesis at telomeres, and significantly increases telomere losses. These losses generate chromosome fusions, leading to chromatin bridges and micronucleus formation upon cell division. By confining base damage to the telomeres, we show that telomeric 8-oxoG accumulation directly drives telomere crisis.


Subject(s)
Chromosome Aberrations/radiation effects , DNA Glycosylases/genetics , DNA Repair/radiation effects , Genomic Instability/radiation effects , Guanine/analogs & derivatives , Telomere/radiation effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , DNA Damage , DNA Glycosylases/deficiency , DNA Replication/radiation effects , Gene Expression , Guanine/agonists , Guanine/biosynthesis , HeLa Cells , Humans , Light/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Optogenetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Oxidative Stress/radiation effects , Singlet Oxygen/agonists , Singlet Oxygen/metabolism , Telomere/metabolism , Telomere Homeostasis/radiation effects
13.
Methods Mol Biol ; 1999: 295-306, 2019.
Article in English | MEDLINE | ID: mdl-31127586

ABSTRACT

Telomere repeats at chromosomal ends are essential for genome stability and sustained cellular proliferation but are susceptible to DNA damage. Repair of damage at telomeres is influenced by numerous factors including telomeric binding proteins, sequence and structure. Ultraviolet (UV) light irradiation induces DNA photoproducts at telomeres that can interfere with telomere maintenance. Here we describe a highly sensitive method for quantifying the formation and removal of UV photoproducts in telomeres isolated from UV irradiated cultured human cells. Damage is detected by immunospot blotting of telomeres with highly specific antibodies against UV photoproducts. This method is adaptable for measuring other types of DNA damage at telomeres as well.


Subject(s)
Genomics/methods , Immunoblotting/methods , Pyrimidine Dimers/analysis , Telomere/radiation effects , Ultraviolet Rays/adverse effects , Antibodies/immunology , Cell Line , DNA/analysis , DNA/genetics , DNA/radiation effects , DNA Damage/radiation effects , DNA Repair , Genomic Instability , Humans , Pyrimidine Dimers/genetics , Pyrimidine Dimers/radiation effects , Telomere/genetics , Telomere/immunology , Telomere-Binding Proteins/immunology
14.
Mech Ageing Dev ; 177: 37-45, 2019 01.
Article in English | MEDLINE | ID: mdl-29604323

ABSTRACT

Telomeres are dynamic nucleoprotein-DNA structures that cap and protect linear chromosome ends. Because telomeres shorten progressively with each replication, they impose a functional limit on the number of times a cell can divide. Critically short telomeres trigger cellular senescence in normal cells, or genomic instability in pre-malignant cells, which contribute to numerous degenerative and aging-related diseases including cancer. Therefore, a detailed understanding of the mechanisms of telomere loss and preservation is important for human health. Numerous studies have shown that oxidative stress is associated with accelerated telomere shortening and dysfunction. Oxidative stress caused by inflammation, intrinsic cell factors or environmental exposures, contributes to the pathogenesis of many degenerative diseases and cancer. Here we review the studies demonstrating associations between oxidative stress and accelerated telomere attrition in human tissue, mice and cell culture, and discuss possible mechanisms and cellular pathways that protect telomeres from oxidative damage.


Subject(s)
Aging/metabolism , DNA Damage , Environmental Exposure/adverse effects , Oxidative Stress , Aging/pathology , Animals , Humans , Mice , Oxidation-Reduction
15.
Molecules ; 22(12)2017 Dec 02.
Article in English | MEDLINE | ID: mdl-29207465

ABSTRACT

Measurement of telomere length by fluorescent in situ hybridization is widely used for biomedical and epidemiological research, but there has been relatively little development of the technology in the 20 years since it was first reported. This report describes the use of dual gammaPNA (γPNA) probes that hybridize at alternating sites along a telomere and give rise to Förster resonance energy transfer (FRET) signals. Bright staining of telomeres is observed in nuclei, chromosome spreads and tissue samples. The use of FRET detection also allows for elimination of wash steps, normally required to remove unhybridized probes that would contribute to background signals. We found that these wash steps can diminish the signal intensity through the removal of bound, as well as unbound probes, so eliminating these steps not only accelerates the process but also enhances the quality of staining. Thus, γPNA FRET pairs allow for brighter and faster staining of telomeres in a wide range of research and clinical formats.


Subject(s)
DNA/metabolism , Fluorescence Resonance Energy Transfer/methods , In Situ Hybridization, Fluorescence/methods , Telomere/metabolism , Base Sequence , Cell Count , Cell Line , Fluorescent Dyes/chemistry , Humans , Molecular Structure , Nucleic Acid Hybridization , Optical Imaging/methods , Osteosarcoma , Peptide Nucleic Acids/metabolism
16.
J Vis Exp ; (125)2017 07 10.
Article in English | MEDLINE | ID: mdl-28715381

ABSTRACT

There are several different techniques for measuring telomere length, each with their own advantages and disadvantages. The traditional approach, Telomere Restriction Fragment (TRF) analysis, utilizes a DNA hybridization technique whereby genomic DNA samples are digested with restriction enzymes, leaving behind telomere DNA repeats and some sub-telomeric DNA. These are separated by agarose gel electrophoresis, transferred to a filter membrane and hybridized to oligonucleotide probes tagged with either chemiluminescence or radioactivity to visualize telomere restriction fragments. This approach, while requiring a larger quantity of DNA than other techniques such as PCR, can measure the telomere length distribution of a population of cells and allows measurement expressed in absolute kilobases. This manuscript demonstrates a modified DNA hybridization procedure for determining telomere length. Genomic DNA is first digested with restriction enzymes (that do not cut telomeres) and separated by agarose gel electrophoresis. The gel is then dried and the DNA is denatured and hybridized in situ to a radiolabeled oligonucleotide probe. This in situ hybridization avoids loss of telomere DNA and improves signal intensity. Following hybridization, the gels are imaged utilizing phosphor screens and the telomere length is quantified using a graphing program. This procedure was developed by the laboratories of Drs. Woodring Wright and Jerry Shay at the University of Texas Southwestern1,2. Here, we present a detailed description of this procedure, with some modifications.


Subject(s)
DNA Restriction Enzymes/genetics , DNA/genetics , In Situ Hybridization/methods , Telomere/metabolism , Humans
17.
Photochem Photobiol ; 93(1): 229-237, 2017 01.
Article in English | MEDLINE | ID: mdl-27861975

ABSTRACT

The fields of telomere biology and DNA repair have enjoyed a great deal of cross-fertilization and convergence in recent years. Telomeres function at chromosome ends to prevent them from being falsely recognized as chromosome breaks by the DNA damage response and repair machineries. Conversely, both canonical and nonconical functions of numerous DNA repair proteins have been found to be critical for preserving telomere structure and function. In 2009, Elizabeth Blackburn, Carol Greider and Jack Szostak were awarded the Nobel prize in Physiology or Medicine for the discovery of telomeres and telomerase. Four years later, pioneers in the field of DNA repair, Aziz Sancar, Tomas Lindahl and Paul Modrich were recognized for their seminal contributions by being awarded the Nobel Prize in Chemistry. This review is part of a special issue meant to celebrate this amazing achievement, and will focus in particular on the convergence of nucleotide excision repair and telomere biology, and will discuss the profound implications for human health.


Subject(s)
DNA Repair , Telomere , Chromosome Aberrations , DNA Damage , Deoxyribodipyrimidine Photo-Lyase/metabolism , Humans , Nobel Prize , Pyrimidine Dimers/metabolism , Telomerase/metabolism , Ultraviolet Rays
18.
Nat Struct Mol Biol ; 23(12): 1092-1100, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27820808

ABSTRACT

Changes in telomere length are associated with degenerative diseases and cancer. Oxidative stress and DNA damage have been linked to both positive and negative alterations in telomere length and integrity. Here we examined how the common oxidative lesion 8-oxo-7,8-dihydro-2'-deoxyguanine (8-oxoG) regulates telomere elongation by human telomerase. When 8-oxoG is present in the dNTP pool as 8-oxodGTP, telomerase utilization of the oxidized nucleotide during telomere extension is mutagenic and terminates further elongation. Depletion of MTH1, the enzyme that removes oxidized dNTPs, increases telomere dysfunction and cell death in telomerase-positive cancer cells with shortened telomeres. In contrast, a preexisting 8-oxoG within the telomeric DNA sequence promotes telomerase activity by destabilizing the G-quadruplex DNA structure. We show that the mechanism by which 8-oxoG arises in telomeres, either by insertion of oxidized nucleotides or by direct reaction with free radicals, dictates whether telomerase is inhibited or stimulated and thereby mediates the biological outcome.


Subject(s)
Deoxyguanine Nucleotides/metabolism , Oxidative Stress , Telomerase/metabolism , Telomere/metabolism , Base Sequence , Cell Death , Cell Line , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Damage , Deoxyguanine Nucleotides/chemistry , Enzyme Activation , G-Quadruplexes , Humans , Mutagens/chemistry , Mutagens/metabolism , Oxidation-Reduction , Telomere/chemistry , Telomere Shortening
19.
DNA Repair (Amst) ; 44: 159-168, 2016 08.
Article in English | MEDLINE | ID: mdl-27233113

ABSTRACT

Telomeres at chromosome ends are nucleoprotein structures consisting of tandem TTAGGG repeats and a complex of proteins termed shelterin. DNA damage and repair at telomeres is uniquely influenced by the ability of telomeric DNA to form alternate structures including loops and G-quadruplexes, coupled with the ability of shelterin proteins to interact with and regulate enzymes in every known DNA repair pathway. The role of shelterin proteins in preventing telomeric ends from being falsely recognized and processed as DNA double strand breaks is well established. Here we focus instead on recent developments in understanding the roles of shelterin proteins and telomeric DNA sequence and structure in processing genuine damage at telomeres induced by endogenous and exogenous DNA damage agents. We will highlight advances in double strand break repair, base excision repair and nucleotide excision repair at telomeres, and will discuss important questions remaining in the field.


Subject(s)
DNA Repair , DNA Replication , DNA/genetics , Protein Subunits/genetics , Telomere-Binding Proteins/genetics , Telomere/metabolism , Animals , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , G-Quadruplexes , Humans , Protein Subunits/metabolism , Shelterin Complex , Telomere/ultrastructure , Telomere Homeostasis , Telomere-Binding Proteins/metabolism
20.
Nat Commun ; 6: 8214, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26351258

ABSTRACT

Ultraviolet light induces cyclobutane pyrimidine dimers (CPD) and pyrimidine(6-4)pyrimidone photoproducts, which interfere with DNA replication and transcription. Nucleotide excision repair (NER) removes these photoproducts, but whether NER functions at telomeres is unresolved. Here we use immunospot blotting to examine the efficiency of photoproduct formation and removal at telomeres purified from UVC irradiated cells at various recovery times. Telomeres exhibit approximately twofold fewer photoproducts compared with the bulk genome in cells, and telomere-binding protein TRF1 significantly reduces photoproduct formation in telomeric fragments in vitro. CPD removal from telomeres occurs 1.5-fold faster than the bulk genome, and is completed by 48 h. 6-4PP removal is rapidly completed by 6 h in both telomeres and the overall genome. A requirement for XPA protein indicates the mechanism of telomeric photoproduct removal is NER. These data provide new evidence that telomeres are partially protected from ultraviolet irradiation and that NER preserves telomere integrity.


Subject(s)
DNA Damage/radiation effects , DNA Repair/physiology , Telomere/radiation effects , Cell Line , Cell Proliferation , Cell Survival , Fibroblasts/cytology , Fibroblasts/radiation effects , Humans , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...