Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Front Aging Neurosci ; 15: 1290681, 2023.
Article in English | MEDLINE | ID: mdl-38161589

ABSTRACT

Ataxia with oculomotor apraxia type 1 (AOA1) is a progressive neurodegenerative disorder characterized by a gradual loss of coordination of hand movements, speech, and eye movements. AOA1 is caused by an inactivation mutation in the APTX gene. APTX resolves abortive DNA ligation intermediates. APTX deficiency may lead to the accumulation of 5'-AMP termini, especially in the mitochondrial genome. The consequences of APTX deficiency includes impaired mitochondrial function, increased DNA single-strand breaks, elevated reactive oxygen species production, and altered mitochondrial morphology. All of these processes can cause misplacement of nuclear and mitochondrial DNA, which can activate innate immune sensors to elicit an inflammatory response. This study explores the impact of APTX knockout in microglial cells, the immune cells of the brain. RNA-seq analysis revealed significant differences in the transcriptomes of wild-type and APTX knockout cells, especially in response to viral infections and innate immune pathways. Specifically, genes and proteins involved in the cGAS-STING and RIG-I/MAVS pathways were downregulated in APTX knockout cells, which suggests an impaired immune response to cytosolic DNA and RNA. The clinical relevance of these findings was supported by analyzing publicly available RNA-seq data from AOA1 patient cell lines. Comparisons between APTX-deficient patient cells and healthy control cells also revealed altered immune responses and dysregulated DNA- and RNA-sensing pathways in the patient cells. Overall, this study highlights the critical role of APTX in regulating innate immunity, particularly in DNA- and RNA-sensing pathways. Our findings contribute to a better understanding of the underlying molecular mechanisms of AOA1 pathology and highlights potential therapeutic targets for this disease.

2.
Front Cell Dev Biol ; 10: 984245, 2022.
Article in English | MEDLINE | ID: mdl-36158192

ABSTRACT

Mitochondria are the primary sites for cellular energy production and are required for many essential cellular processes. Mitochondrial DNA (mtDNA) is a 16.6 kb circular DNA molecule that encodes only 13 gene products of the approximately 90 different proteins of the respiratory chain complexes and an estimated 1,200 mitochondrial proteins. MtDNA is, however, crucial for organismal development, normal function, and survival. MtDNA maintenance requires mitochondrially targeted nuclear DNA repair enzymes, a mtDNA replisome that is unique to mitochondria, and systems that control mitochondrial morphology and quality control. Here, we provide an overview of the current literature on mtDNA repair and transcription machineries and discuss how dynamic functional interactions between the components of these systems regulate mtDNA maintenance and transcription. A profound understanding of the molecular mechanisms that control mtDNA maintenance and transcription is important as loss of mtDNA integrity is implicated in normal process of aging, inflammation, and the etiology and pathogenesis of a number of diseases.

3.
Cells ; 10(12)2021 12 15.
Article in English | MEDLINE | ID: mdl-34944052

ABSTRACT

Aging is the consequence of a lifelong accumulation of stochastic damage to tissues and cellular components. Advancing age closely associates with elevated markers of innate immunity and low-grade chronic inflammation, probably reflecting steady increasing incidents of cellular and tissue damage over the life course. The DNA sensing cGAS-STING signaling pathway is activated by misplaced cytosolic self-DNA, which then initiates the innate immune responses. Here, we hypothesize that the stochastic release of various forms of DNA from the nucleus and mitochondria, e.g., because of DNA damage, altered nucleus integrity, and mitochondrial damage, can result in chronic activation of inflammatory responses that characterize the aging process. This cytosolic self-DNA-innate immunity axis may perturb tissue homeostasis and function that characterizes human aging and age-associated pathology. Proper techniques and experimental models are available to investigate this axis to develop therapeutic interventions.


Subject(s)
Aging/pathology , Cytosol/metabolism , DNA/metabolism , Inflammation/pathology , Autophagy , Cellular Senescence , Humans
4.
Aging (Albany NY) ; 13(21): 23876-23894, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34751671

ABSTRACT

Tauopathies are a group of progressive neurodegenerative disorders characterized by the presence of insoluble intracellular tau filaments in the brain. Evidence suggests that there is a tight connection between mitochondrial dysfunction and tauopathies, including Alzheimer's disease. However, whether mitochondrial dysfunction occurs prior to the detection of tau aggregates in tauopathies remains elusive. Here, we utilized transgenic nematodes expressing the full length of wild type tau in neuronal cells and monitored mitochondrial morphology alterations over time. Although tau-expressing nematodes did not accumulate detectable levels of tau aggregates during larval stages, they displayed increased mitochondrial damage and locomotion defects compared to the control worms. Chelating calcium restored mitochondrial activity and improved motility in the tau-expressing larvae suggesting a link between mitochondrial damage, calcium homeostasis and neuronal impairment in these animals. Our findings suggest that defective mitochondrial function is an early pathogenic event of tauopathies, taking place before tau aggregation and undermining neuronal homeostasis and organismal fitness. Understanding the molecular mechanisms causing mitochondrial dysfunction early in tauopathy will be of significant clinical and therapeutic value and merits further investigation.


Subject(s)
Homeostasis/physiology , Mitochondria , Tauopathies , tau Proteins/metabolism , Alzheimer Disease , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Disease Models, Animal , Humans , Larva/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Tauopathies/metabolism , Tauopathies/physiopathology
5.
Cell Rep ; 36(10): 109668, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34496255

ABSTRACT

Aging, genomic stress, and mitochondrial dysfunction are risk factors for neurodegenerative pathologies, such as Parkinson disease (PD). Although genomic instability is associated with aging and mitochondrial impairment, the underlying mechanisms are poorly understood. Here, we show that base excision repair generates genomic stress, promoting age-related neurodegeneration in a Caenorhabditis elegans PD model. A physiological level of NTH-1 DNA glycosylase mediates mitochondrial and nuclear genomic instability, which promote degeneration of dopaminergic neurons in older nematodes. Conversely, NTH-1 deficiency protects against α-synuclein-induced neurotoxicity, maintaining neuronal function with age. This apparent paradox is caused by modulation of mitochondrial transcription in NTH-1-deficient cells, and this modulation activates LMD-3, JNK-1, and SKN-1 and induces mitohormesis. The dependance of neuroprotection on mitochondrial transcription highlights the integration of BER and transcription regulation during physiological aging. Finally, whole-exome sequencing of genomic DNA from patients with idiopathic PD suggests that base excision repair might modulate susceptibility to PD in humans.


Subject(s)
Aging , DNA Repair/physiology , DNA, Single-Stranded/metabolism , Parkinson Disease/pathology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , DNA Damage/drug effects , DNA Repair/genetics , Dopaminergic Neurons/metabolism , Endonucleases/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Parkinson Disease/genetics
6.
Nucleic Acids Res ; 48(12): 6530-6546, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32432680

ABSTRACT

OGG1 initiated base excision repair (BER) is the major pathway for repair of oxidative DNA base damage 8-oxoguanine (8-oxoG). Here, we report that RECQL4 DNA helicase, deficient in the cancer-prone and premature aging Rothmund-Thomson syndrome, physically and functionally interacts with OGG1. RECQL4 promotes catalytic activity of OGG1 and RECQL4 deficiency results in defective 8-oxoG repair and increased genomic 8-oxoG. Furthermore, we show that acute oxidative stress leads to increased RECQL4 acetylation and its interaction with OGG1. The NAD+-dependent protein SIRT1 deacetylates RECQL4 in vitro and in cells thereby controlling the interaction between OGG1 and RECQL4 after DNA repair and maintaining RECQL4 in a low acetylated state. Collectively, we find that RECQL4 is involved in 8-oxoG repair through interaction with OGG1, and that SIRT1 indirectly modulates BER of 8-oxoG by controlling RECQL4-OGG1 interaction.


Subject(s)
DNA Glycosylases/metabolism , DNA Repair , RecQ Helicases/metabolism , Sirtuin 1/metabolism , Acetylation , Cell Line, Tumor , Guanosine/analogs & derivatives , Guanosine/genetics , HEK293 Cells , Humans , Oxidative Stress , Protein Binding
7.
Acta Neuropathol Commun ; 8(1): 25, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32131898

ABSTRACT

Insoluble intracellular aggregation of tau proteins into filaments and neurodegeneration are histopathological hallmarks of Alzheimer disease (AD) and other tauopathies. Recently, prefibrillar, soluble, oligomeric tau intermediates have emerged as relevant pathological tau species; however, the molecular mechanisms of neuronal responses to tau oligomers are not fully understood. Here, we show that hippocampal neurons in six-month-old transgenic mouse model of tauopathy, THY-Tau22, are enriched with oligomeric tau, contain elongated mitochondria, and display cellular stress, but no overt cytotoxicity compared to the control mice. The levels of several key mitochondrial proteins were markedly different between the THY-Tau22 and control mice hippocampi including the mitochondrial SIRT3, PINK1, ANT1 and the fission protein DRP1. DNA base excision repair (BER) is the primary defense system against oxidative DNA damage and it was elevated in six-month-old transgenic mice. DNA polymerase ß, the key BER DNA polymerase, was enriched in the cytoplasm of hippocampal neurons in six-month-old transgenic mice and localized with and within mitochondria. Polß also co-localized with mitochondria in human AD brains in neurons containing oligomeric tau. Most of these altered mitochondrial and DNA repair events were specific to the transgenic mice at 6 months of age and were not different from control mice at 12 months of age when tau pathology reaches its maximum and oligomeric forms of tau are no longer detectable. In summary, our data suggests that we have identified key cellular stress responses at early stages of tau pathology to preserve neuronal integrity and to promote survival. To our knowledge, this work provides the first description of multiple stress responses involving mitochondrial homeostasis and BER early during the progression of tau pathology, and represents an important advance in the etiopathogenesis of tauopathies.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Neurons/metabolism , Oxidative Stress , tau Proteins/metabolism , Adenine Nucleotide Translocator 1/metabolism , Aged , Animals , DNA Damage , DNA Polymerase beta/metabolism , DNA Repair , Disease Models, Animal , Dynamins/metabolism , Frontal Lobe/cytology , Frontal Lobe/metabolism , Hippocampus/cytology , Homeostasis , Humans , Male , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Middle Aged , Mitochondria/ultrastructure , Neurofibrillary Tangles , Neurons/ultrastructure , Protein Kinases/metabolism , Sirtuin 3/metabolism , tau Proteins/genetics
8.
Nucleic Acids Res ; 47(22): 11709-11728, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31647095

ABSTRACT

The A-type lamins (lamin A/C), encoded by the LMNA gene, are important structural components of the nuclear lamina. LMNA mutations lead to degenerative disorders known as laminopathies, including the premature aging disease Hutchinson-Gilford progeria syndrome. In addition, altered lamin A/C expression is found in various cancers. Reports indicate that lamin A/C plays a role in DNA double strand break repair, but a role in DNA base excision repair (BER) has not been described. We provide evidence for reduced BER efficiency in lamin A/C-depleted cells (Lmna null MEFs and lamin A/C-knockdown U2OS). The mechanism involves impairment of the APE1 and POLß BER activities, partly effectuated by associated reduction in poly-ADP-ribose chain formation. Also, Lmna null MEFs displayed reduced expression of several core BER enzymes (PARP1, LIG3 and POLß). Absence of Lmna led to accumulation of 8-oxoguanine (8-oxoG) lesions, and to an increased frequency of substitution mutations induced by chronic oxidative stress including GC>TA transversions (a fingerprint of 8-oxoG:A mismatches). Collectively, our results provide novel insights into the functional interplay between the nuclear lamina and cellular defenses against oxidative DNA damage, with implications for cancer and aging.


Subject(s)
DNA Repair/genetics , Lamin Type A/physiology , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Cells, Cultured , DNA Damage/physiology , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Microarray Analysis , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Oxidative Stress/genetics , Progeria/genetics
9.
Ageing Res Rev ; 54: 100940, 2019 09.
Article in English | MEDLINE | ID: mdl-31415807

ABSTRACT

Genetic and pharmacological intervention studies have identified evolutionarily conserved and functionally interconnected networks of cellular energy homeostasis, nutrient-sensing, and genome damage response signaling pathways, as prominent regulators of longevity and health span in various species. Mitochondria are the primary sites of ATP production and are key players in several other important cellular processes. Mitochondrial dysfunction diminishes tissue and organ functional performance and is a commonly considered feature of the aging process. Here we review the evidence that through reciprocal and multilevel functional interactions, mitochondria are implicated in the lifespan modulation function of these pathways, which altogether constitute a highly dynamic and complex system that controls the aging process. An important characteristic of these pathways is their extensive crosstalk and apparent malleability to modification by non-invasive pharmacological, dietary, and lifestyle interventions, with promising effects on lifespan and health span in animal models and potentially also in humans.


Subject(s)
Longevity , Mitochondria/metabolism , Signal Transduction , Aging , Animals , DNA Repair , Energy Metabolism , Humans , Mitochondria/physiology
10.
Nucleic Acids Res ; 47(8): 4086-4110, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30986824

ABSTRACT

Ataxia with oculomotor apraxia type 1 (AOA1) is an early onset progressive spinocerebellar ataxia caused by mutation in aprataxin (APTX). APTX removes 5'-AMP groups from DNA, a product of abortive ligation during DNA repair and replication. APTX deficiency has been suggested to compromise mitochondrial function; however, a detailed characterization of mitochondrial homeostasis in APTX-deficient cells is not available. Here, we show that cells lacking APTX undergo mitochondrial stress and display significant changes in the expression of the mitochondrial inner membrane fusion protein optic atrophy type 1, and components of the oxidative phosphorylation complexes. At the cellular level, APTX deficiency impairs mitochondrial morphology and network formation, and autophagic removal of damaged mitochondria by mitophagy. Thus, our results show that aberrant mitochondrial function is a key component of AOA1 pathology. This work corroborates the emerging evidence that impaired mitochondrial function is a characteristic of an increasing number of genetically diverse neurodegenerative disorders.


Subject(s)
DNA-Binding Proteins/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitophagy/genetics , Nuclear Proteins/genetics , Spinocerebellar Ataxias/congenital , Cell Line, Transformed , Cell Line, Tumor , DNA-Binding Proteins/deficiency , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , GTP Phosphohydrolases/deficiency , Gene Expression Profiling , Gene Expression Regulation , Homeostasis/genetics , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Mitochondria/metabolism , Mitochondria/ultrastructure , Nuclear Proteins/deficiency , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteoblasts/pathology , Oxidative Phosphorylation , Signal Transduction , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
11.
Nat Neurosci ; 22(3): 401-412, 2019 03.
Article in English | MEDLINE | ID: mdl-30742114

ABSTRACT

Accumulation of damaged mitochondria is a hallmark of aging and age-related neurodegeneration, including Alzheimer's disease (AD). The molecular mechanisms of impaired mitochondrial homeostasis in AD are being investigated. Here we provide evidence that mitophagy is impaired in the hippocampus of AD patients, in induced pluripotent stem cell-derived human AD neurons, and in animal AD models. In both amyloid-ß (Aß) and tau Caenorhabditis elegans models of AD, mitophagy stimulation (through NAD+ supplementation, urolithin A, and actinonin) reverses memory impairment through PINK-1 (PTEN-induced kinase-1)-, PDR-1 (Parkinson's disease-related-1; parkin)-, or DCT-1 (DAF-16/FOXO-controlled germline-tumor affecting-1)-dependent pathways. Mitophagy diminishes insoluble Aß1-42 and Aß1-40 and prevents cognitive impairment in an APP/PS1 mouse model through microglial phagocytosis of extracellular Aß plaques and suppression of neuroinflammation. Mitophagy enhancement abolishes AD-related tau hyperphosphorylation in human neuronal cells and reverses memory impairment in transgenic tau nematodes and mice. Our findings suggest that impaired removal of defective mitochondria is a pivotal event in AD pathogenesis and that mitophagy represents a potential therapeutic intervention.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Mitophagy , Neurons/metabolism , Neurons/pathology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Disease Models, Animal , Female , Induced Pluripotent Stem Cells , Male , Memory , Mice , Neural Stem Cells
12.
Mech Ageing Dev ; 161(Pt A): 83-94, 2017 01.
Article in English | MEDLINE | ID: mdl-27105872

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. Autosomal dominant, familial AD (fAD) is very rare and caused by mutations in amyloid precursor protein (APP), presenilin-1 (PSEN-1), and presenilin-2 (PSEN-2) genes. The pathogenesis of sporadic AD (sAD) is more complex and variants of several genes are associated with an increased lifetime risk of AD. Nuclear and mitochondrial DNA integrity is pivotal during neuronal development, maintenance and function. DNA damage and alterations in cellular DNA repair capacity have been implicated in the aging process and in age-associated neurodegenerative diseases, including AD. These findings are supported by research using animal models of AD and in DNA repair deficient animal models. In recent years, novel mechanisms linking DNA damage to neuronal dysfunction have been identified and have led to the development of noninvasive treatment strategies. Further investigations into the molecular mechanisms connecting DNA damage to AD pathology may help to develop novel treatment strategies for this debilitating disease. Here we provide an overview of the role of genome instability and DNA repair deficiency in AD pathology and discuss research strategies that include genome instability as a component.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Genomic Instability , Mutation , Neurons/metabolism , Presenilin-1/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , DNA Damage , DNA Repair , Humans , Neurons/pathology , Presenilin-1/genetics
13.
Sci Rep ; 5: 12876, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26256098

ABSTRACT

Aborted DNA ligation events in eukaryotic cells can generate 5'-adenylated (5'-AMP) DNA termini that can be removed from DNA by aprataxin (APTX). Mutations in APTX cause an inherited human disease syndrome characterized by early-onset progressive ataxia with ocular motor apraxia (AOA1). APTX is found in the nuclei and mitochondria of eukaryotic cells. Depletion of APTX causes mitochondrial dysfunction and renders the mitochondrial genome, but not the nuclear genome susceptible to damage. The biochemical processes that link APTX deficiency to mitochondrial dysfunction have not been well elucidated. Here, we monitored the repair of 5'-AMP DNA damage in nuclear and mitochondrial extracts from human APTX(+/+) and APTX(-/-) cells. The efficiency of repair of 5'-AMP DNA was much lower in mitochondrial than in nuclear protein extracts, and resulted in persistent DNA repair intermediates in APTX deficient cells. Moreover, the removal of 5'-AMP from DNA was significantly slower in the mitochondrial extracts from human cell lines and mouse tissues compared with their corresponding nuclear extracts. These results suggest that, contrary to nuclear DNA repair, mitochondrial DNA repair is not able to compensate for APTX deficiency resulting in the accumulation of mitochondrial DNA damage.


Subject(s)
Adenosine Monophosphate/metabolism , DNA Repair , DNA, Mitochondrial/metabolism , Depsipeptides/genetics , Mitochondria/genetics , Animals , Brain/metabolism , Cell Line , DNA Damage , DNA, Mitochondrial/genetics , Databases, Factual , Depsipeptides/deficiency , Humans , Liver/metabolism , Mice , Mitochondria/metabolism
14.
DNA Repair (Amst) ; 32: 172-179, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26002197

ABSTRACT

Chemical modification and spontaneous loss of nucleotide bases from DNA are estimated to occur at the rate of thousands per human cell per day. DNA base excision repair (BER) is a critical mechanism for repairing such lesions in nuclear and mitochondrial DNA. Defective expression or function of proteins required for BER or proteins that regulate BER have been consistently associated with neurological dysfunction and disease in humans. Recent studies suggest that DNA lesions in the nuclear and mitochondrial compartments and the cellular response to those lesions have a profound effect on cellular energy homeostasis, mitochondrial function and cellular bioenergetics, with especially strong influence on neurological function. Further studies in this area could lead to novel approaches to prevent and treat human neurodegenerative disease.


Subject(s)
Brain/metabolism , DNA Repair , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/genetics , Brain/pathology , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Mitochondrial/chemistry , Gene Expression Regulation , Homeostasis , Humans , Mitochondria/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins
15.
DNA Repair (Amst) ; 16: 44-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24674627

ABSTRACT

Base excision repair (BER) is the most prominent DNA repair pathway in human mitochondria. BER also results in a temporary generation of AP-sites, single-strand breaks and nucleotide gaps. Thus, incomplete BER can result in the generation of DNA repair intermediates that can disrupt mitochondrial DNA replication and transcription and generate mutations. We carried out BER analysis in highly purified mitochondrial extracts from human cell lines U2OS and HeLa, and mouse brain using a circular DNA substrate containing a lesion at a specific position. We found that DNA ligation is significantly slower than the preceding mitochondrial BER steps. Overexpression of DNA ligase III in mitochondria improved the rate of overall BER, increased cell survival after menadione induced oxidative stress and reduced autophagy following the inhibition of the mitochondrial electron transport chain complex I by rotenone. Our results suggest that the amount of DNA ligase III in mitochondria may be critical for cell survival following prolonged oxidative stress, and demonstrate a functional link between mitochondrial DNA damage and repair, cell survival upon oxidative stress, and removal of dysfunctional mitochondria by autophagy.


Subject(s)
Brain/metabolism , DNA Ligases/metabolism , DNA Repair , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Oxidative Stress/drug effects , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Ligase ATP , DNA Ligases/genetics , Electron Transport Complex I/metabolism , HeLa Cells , Humans , Mice , Mitochondria/genetics , Poly-ADP-Ribose Binding Proteins , Rotenone/pharmacology , Vitamin K 3/pharmacology , Xenopus Proteins
16.
Int J Mol Sci ; 13(12): 17210-29, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23247283

ABSTRACT

X-ray Repair Cross Complementing protein 1 (XRCC1) acts as a scaffolding protein in the converging base excision repair (BER) and single strand break repair (SSBR) pathways. XRCC1 also interacts with itself and rapidly accumulates at sites of DNA damage. XRCC1 can thus mediate the assembly of large multiprotein DNA repair complexes as well as facilitate the recruitment of DNA repair proteins to sites of DNA damage. Moreover, XRCC1 is present in constitutive DNA repair complexes, some of which associate with the replication machinery. Because of the critical role of XRCC1 in DNA repair, its common variants Arg194Trp, Arg280His and Arg399Gln have been extensively studied. However, the prevalence of these variants varies strongly in different populations, and their functional influence on DNA repair and disease remains elusive. Here we present the current knowledge about the role of XRCC1 and its variants in BER and human disease/cancer.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , DNA-Binding Proteins/genetics , Humans , Neoplasm Proteins/genetics , X-ray Repair Cross Complementing Protein 1
17.
Aging Cell ; 11(3): 456-66, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22296597

ABSTRACT

RECQL4 is associated with Rothmund-Thomson Syndrome (RTS), a rare autosomal recessive disorder characterized by premature aging, genomic instability, and cancer predisposition. RECQL4 is a member of the RecQ helicase family, and has many similarities to WRN protein, which is also implicated in premature aging. There is no information about whether any of the RecQ helicases play roles in mitochondrial biogenesis, which is strongly implicated in the aging process. Here, we used microscopy to visualize RECQL4 in mitochondria. Fractionation of human and mouse cells also showed that RECQL4 was present in mitochondria. Q-PCR amplification of mitochondrial DNA demonstrated that mtDNA damage accumulated in RECQL4-deficient cells. Microarray analysis suggested that mitochondrial bioenergetic pathways might be affected in RTS. Measurements of mitochondrial bioenergetics showed a reduction in the mitochondrial reserve capacity after lentiviral knockdown of RECQL4 in two different primary cell lines. Additionally, biochemical assays with RECQL4, mitochondrial transcription factor A, and mitochondrial DNA polymerase γ showed that the polymerase inhibited RECQL4's helicase activity. RECQL4 is the first 3'-5' RecQ helicase to be found in both human and mouse mitochondria, and the loss of RECQL4 alters mitochondrial integrity.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondria/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Age Factors , Aged, 80 and over , Animals , Cell Fractionation/methods , Cell Line, Tumor , DNA Damage , Genomic Instability , HeLa Cells , Humans , Mice
18.
DNA Repair (Amst) ; 11(4): 357-66, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22281126

ABSTRACT

XRCC1 functions as a non-enzymatic, scaffold protein in single strand break repair (SSBR) and base excision repair (BER). Here, we examine different regions of XRCC1 for their contribution to the scaffolding functions of the protein. We found that the central BRCT1 domain is essential for recruitment of XRCC1 to sites of DNA damage and DNA replication. Also, we found that ectopic expression of the region from residue 166-436 partially rescued the methyl methanesulfonate (MMS) hypersensitivity of XRCC1-deficient EM9 cells, suggesting a key role for this region in mediating DNA repair. The three most common amino acid variants of XRCC1, Arg194Trp, Arg280His and Arg399Gln, are located within the region comprising the NLS and BRCT1 domains, and these variants may be associated with increased incidence of specific types of cancer. While we could not detect differences in the intra-nuclear localization or the ability to support recruitment of POLß or PNKP to micro-irradiated sites for these variants relative to the conservative protein, we did observe lower foci intensity after micro-irradiation and a reduced stability of the foci with the Arg280His and Arg399Gln variants, respectively. Furthermore, when challenged with MMS or hydrogen peroxide, we detected small but consistent differences in the repair profiles of cells expressing these two variants in comparison to the conservative protein.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Polymorphism, Single Nucleotide , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Damage , DNA Polymerase beta/metabolism , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA Repair Enzymes/metabolism , DNA Replication/drug effects , DNA Replication/genetics , DNA Replication/radiation effects , DNA-Binding Proteins/genetics , Humans , Methyl Methanesulfonate/pharmacology , Nuclear Localization Signals , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , Protein Transport/radiation effects , X-ray Repair Cross Complementing Protein 1
19.
Mutat Res ; 736(1-2): 33-8, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-21736882

ABSTRACT

The base excision repair (BER) pathway corrects many different DNA base lesions and is important for genomic stability. The mechanism of BER cannot easily be investigated in intact cells and therefore in vitro methods that reflect the in vivo processes are in high demand. Reconstitution of BER using purified proteins essentially mirror properties of the proteins used, and does not necessarily reflect the mechanism as it occurs in the cell. Nuclear extracts from cultured cells have the capacity to carry out complete BER and can give important information on the mechanism. Furthermore, candidate proteins in extracts can be inhibited or depleted in a controlled way, making defined extracts an important source for mechanistic studies. The major drawback is that there is no standardized method of preparing nuclear extract for BER studies, and it does not appear to be a topic given much attention. Here we have examined BER activity of nuclear cell extracts from HeLa cells, using as substrate a circular DNA molecule with either uracil or an AP-site in a defined position. We show that BER activity of nuclear extracts from the same batch of cells varies inversely with the volume of nuclear extraction buffer relative to nuclei volume, in spite of identical protein concentrations in the BER assay mixture. Surprisingly, the uracil-DNA glycosylase activity (mainly UNG2), but not amount of UNG2, also correlated negatively with the volume of extraction buffer. These studies demonstrate that the method for preparation of nuclear extract is an important factor to consider for in vitro BER analysis and conditions used in comparative studies must be carefully worked out.


Subject(s)
Buffers , Cell Nucleus/chemistry , DNA Repair , Cell Nucleus/metabolism , Cells, Cultured , HeLa Cells , Humans
20.
DNA Repair (Amst) ; 11(1): 82-91, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22153281

ABSTRACT

Single-strand DNA binding proteins protect DNA from nucleolytic damage, prevent formation of secondary structures and prevent premature reannealing of DNA in DNA metabolic transactions. In eukaryotes, the nuclear single-strand DNA binding protein RPA is essential for chromosomal DNA replication and transcription and directly participates in several DNA repair processes by binding to and modulating the activity of repair factors. Much less is known about the involvement of the only mitochondrial single-strand binding protein mtSSB in the context of DNA repair. Here we demonstrate that mtSSB impedes excision of uracil and oxidative demethylation of 3meC in single-stranded DNA by UNG1 and ABH1, respectively, whereas excision by NEIL1 was partially inhibited. mtSSB also effectively inhibited nicking of single-stranded DNA by APE1 and ABH1 and partially inhibited the lyase activity of NEIL1. Finally we identified a putative surface motif in mtSSB that may recruit UNG1 to DNA-bound mtSSB. We suggest that the massive amount of mtSSB in mitochondria effectively prevents processing of uracil and other types of damaged bases to avoid introduction of nicks in single-stranded mtDNA formed during replication. Local enrichment of UNG1 at DNA-bound mtSSB may furthermore facilitate rapid access to- and processing of the damage once the dsDNA conformation is restored. This could be of potential biological importance, since mitochondria have no or limited capacity for homologous recombination to process nicks at the replication fork.


Subject(s)
DNA Glycosylases/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , DNA/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nucleic Acid Conformation , Uracil/metabolism , Amino Acid Sequence , Base Sequence , Carboxylic Ester Hydrolases/metabolism , Cell Extracts , DNA/metabolism , DNA Damage , DNA Glycosylases/chemistry , DNA Methylation , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/chemistry , HeLa Cells , Humans , Membrane Proteins/metabolism , Mitochondrial Proteins/chemistry , Molecular Sequence Data , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL