Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Cell ; 61(1): 161-9, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26626482

ABSTRACT

G quadruplexes (G4s) can present potent blocks to DNA replication. Accurate and timely replication of G4s in vertebrates requires multiple specialized DNA helicases and polymerases to prevent genetic and epigenetic instability. Here we report that PrimPol, a recently described primase-polymerase (PrimPol), plays a crucial role in the bypass of leading strand G4 structures. While PrimPol is unable to directly replicate G4s, it can bind and reprime downstream of these structures. Disruption of either the catalytic activity or zinc-finger of PrimPol results in extreme G4-dependent epigenetic instability at the BU-1 locus in avian DT40 cells, indicative of extensive uncoupling of the replicative helicase and polymerase. Together, these observations implicate PrimPol in promoting restart of DNA synthesis downstream of, but closely coupled to, G4 replication impediments.


Subject(s)
Avian Proteins/metabolism , DNA Primase/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA/biosynthesis , G-Quadruplexes , Multifunctional Enzymes/metabolism , Animals , Avian Proteins/genetics , Base Sequence , Cell Line , Chickens , Chromatin Assembly and Disassembly , DNA/chemistry , DNA Primase/genetics , DNA-Directed DNA Polymerase/genetics , Epigenesis, Genetic , Genomic Instability , Histones/metabolism , Molecular Sequence Data , Multifunctional Enzymes/genetics , Transfection
2.
Mol Cell ; 52(4): 566-73, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267451

ABSTRACT

DNA damage can stall the DNA replication machinery, leading to genomic instability. Thus, numerous mechanisms exist to complete genome duplication in the absence of a pristine DNA template, but identification of the enzymes involved remains incomplete. Here, we establish that Primase-Polymerase (PrimPol; CCDC111), an archaeal-eukaryotic primase (AEP) in eukaryotic cells, is involved in chromosomal DNA replication. PrimPol is required for replication fork progression on ultraviolet (UV) light-damaged DNA templates, possibly mediated by its ability to catalyze translesion synthesis (TLS) of these lesions. This PrimPol UV lesion bypass pathway is not epistatic with the Pol η-dependent pathway and, as a consequence, protects xeroderma pigmentosum variant (XP-V) patient cells from UV-induced cytotoxicity. In addition, we establish that PrimPol is also required for efficient replication fork progression during an unperturbed S phase. These and other findings indicate that PrimPol is an important player in replication fork progression in eukaryotic cells.


Subject(s)
Chromosomes, Human/genetics , DNA Adducts/genetics , DNA Primase/physiology , DNA Replication , DNA-Directed DNA Polymerase/physiology , Multifunctional Enzymes/physiology , Amino Acid Sequence , Animals , Cell Proliferation , Cell Survival , Chickens , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Damage , DNA Primase/chemistry , DNA, Single-Stranded/chemistry , DNA-Directed DNA Polymerase/chemistry , G2 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Multifunctional Enzymes/chemistry , Ultraviolet Rays , Xenopus
3.
Nucleic Acids Res ; 47(8): 4026-4038, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30715459

ABSTRACT

Eukaryotic Primase-Polymerase (PrimPol) is an enzyme that maintains efficient DNA duplication by repriming replication restart downstream of replicase stalling lesions and structures. To elucidate the cellular requirements for PrimPol in human cells, we generated PrimPol-deleted cell lines and show that it plays key roles in maintaining active replication in both the nucleus and mitochondrion, even in the absence of exogenous damage. Human cells lacking PrimPol exhibit delayed recovery after UV-C damage and increased mutation frequency, micronuclei and sister chromatin exchanges but are not sensitive to genotoxins. PrimPol is also required during mitochondrial replication, with PrimPol-deficient cells having increased mtDNA copy number but displaying a significant decrease in replication. Deletion of PrimPol in XPV cells, lacking functional polymerase Eta, causes an increase in DNA damage sensitivity and pronounced fork stalling after UV-C treatment. We show that, unlike canonical TLS polymerases, PrimPol is important for allowing active replication to proceed, even in the absence of exogenous damage, thus preventing the accumulation of excessive fork stalling and genetic mutations. Together, these findings highlight the importance of PrimPol for maintaining efficient DNA replication in unperturbed cells and its complementary roles, with Pol Eta, in damage tolerance in human cells.


Subject(s)
Cell Nucleus/radiation effects , DNA Primase/genetics , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/genetics , DNA/genetics , Mitochondria/radiation effects , Multifunctional Enzymes/genetics , 4-Nitroquinoline-1-oxide/pharmacology , Bleomycin/pharmacology , Cell Line, Transformed , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cisplatin/pharmacology , DNA/drug effects , DNA/metabolism , DNA Primase/deficiency , DNA Replication/drug effects , DNA-Directed DNA Polymerase/deficiency , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Deletion , Humans , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Mitochondria/drug effects , Mitochondria/genetics , Multifunctional Enzymes/deficiency , Mutagens/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/radiation effects , Quinolones/pharmacology , Sister Chromatid Exchange/drug effects , Sister Chromatid Exchange/radiation effects , Ultraviolet Rays/adverse effects
4.
Proc Natl Acad Sci U S A ; 113(30): E4276-85, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402764

ABSTRACT

The genetic information in mammalian mitochondrial DNA is densely packed; there are no introns and only one sizeable noncoding, or control, region containing key cis-elements for its replication and expression. Many molecules of mitochondrial DNA bear a third strand of DNA, known as "7S DNA," which forms a displacement (D-) loop in the control region. Here we show that many other molecules contain RNA as a third strand. The RNA of these R-loops maps to the control region of the mitochondrial DNA and is complementary to 7S DNA. Ribonuclease H1 is essential for mitochondrial DNA replication; it degrades RNA hybridized to DNA, so the R-loop is a potential substrate. In cells with a pathological variant of ribonuclease H1 associated with mitochondrial disease, R-loops are of low abundance, and there is mitochondrial DNA aggregation. These findings implicate ribonuclease H1 and RNA in the physical segregation of mitochondrial DNA, perturbation of which represents a previously unidentified disease mechanism.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mutation , Ribonuclease H/genetics , Animals , Cell Line, Tumor , Cells, Cultured , DNA Replication , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Nucleic Acid Conformation , Ribonuclease H/metabolism
5.
Nucleic Acids Res ; 44(7): 3317-29, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26984527

ABSTRACT

Translesion synthesis (TLS) employs specialized DNA polymerases to bypass replication fork stalling lesions. PrimPol was recently identified as a TLS primase and polymerase involved in DNA damage tolerance. Here, we identify a novel PrimPol binding partner, PolDIP2, and describe how it regulates PrimPol's enzymatic activities. PolDIP2 stimulates the polymerase activity of PrimPol, enhancing both its capacity to bind DNA and the processivity of the catalytic domain. In addition, PolDIP2 stimulates both the efficiency and error-free bypass of 8-oxo-7,8-dihydrodeoxyguanosine (8-oxoG) lesions by PrimPol. We show that PolDIP2 binds to PrimPol's catalytic domain and identify potential binding sites. Finally, we demonstrate that depletion of PolDIP2 in human cells causes a decrease in replication fork rates, similar to that observed in PrimPol(-/-)cells. However, depletion of PolDIP2 in PrimPol(-/-)cells does not produce a further decrease in replication fork rates. Together, these findings establish that PolDIP2 can regulate the TLS polymerase and primer extension activities of PrimPol, further enhancing our understanding of the roles of PolDIP2 and PrimPol in eukaryotic DNA damage tolerance.


Subject(s)
DNA Damage , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/metabolism , Nuclear Proteins/metabolism , Cells, Cultured , DNA/metabolism , DNA Primase/antagonists & inhibitors , DNA Replication , DNA-Binding Proteins/metabolism , Guanine/analogs & derivatives , Humans , Multifunctional Enzymes/antagonists & inhibitors , Proliferating Cell Nuclear Antigen/metabolism
6.
Biochem Soc Trans ; 45(2): 513-529, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28408491

ABSTRACT

PrimPol, (primase-polymerase), the most recently identified eukaryotic polymerase, has roles in both nuclear and mitochondrial DNA maintenance. PrimPol is capable of acting as a DNA polymerase, with the ability to extend primers and also bypass a variety of oxidative and photolesions. In addition, PrimPol also functions as a primase, catalysing the preferential formation of DNA primers in a zinc finger-dependent manner. Although PrimPol's catalytic activities have been uncovered in vitro, we still know little about how and why it is targeted to the mitochondrion and what its key roles are in the maintenance of this multicopy DNA molecule. Unlike nuclear DNA, the mammalian mitochondrial genome is circular and the organelle has many unique proteins essential for its maintenance, presenting a differing environment within which PrimPol must function. Here, we discuss what is currently known about the mechanisms of DNA replication in the mitochondrion, the proteins that carry out these processes and how PrimPol is likely to be involved in assisting this vital cellular process.


Subject(s)
DNA Primase/metabolism , DNA Replication , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/metabolism , Animals , Cell Nucleus/genetics , DNA Primase/genetics , DNA-Directed DNA Polymerase/genetics , Humans , Mitochondria/genetics , Multifunctional Enzymes/genetics , Mutation
7.
Nucleic Acids Res ; 43(2): 1056-68, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25550423

ABSTRACT

PrimPol is a recently identified polymerase involved in eukaryotic DNA damage tolerance, employed in both re-priming and translesion synthesis mechanisms to bypass nuclear and mitochondrial DNA lesions. In this report, we investigate how the enzymatic activities of human PrimPol are regulated. We show that, unlike other TLS polymerases, PrimPol is not stimulated by PCNA and does not interact with it in vivo. We identify that PrimPol interacts with both of the major single-strand binding proteins, RPA and mtSSB in vivo. Using NMR spectroscopy, we characterize the domains responsible for the PrimPol-RPA interaction, revealing that PrimPol binds directly to the N-terminal domain of RPA70. In contrast to the established role of SSBs in stimulating replicative polymerases, we find that SSBs significantly limit the primase and polymerase activities of PrimPol. To identify the requirement for this regulation, we employed two forward mutation assays to characterize PrimPol's replication fidelity. We find that PrimPol is a mutagenic polymerase, with a unique error specificity that is highly biased towards insertion-deletion errors. Given the error-prone disposition of PrimPol, we propose a mechanism whereby SSBs greatly restrict the contribution of this enzyme to DNA replication at stalled forks, thus reducing the mutagenic potential of PrimPol during genome replication.


Subject(s)
DNA Primase/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Mitochondrial Proteins/metabolism , Multifunctional Enzymes/metabolism , Replication Protein A/metabolism , DNA Primers/biosynthesis , DNA Replication , Humans , Mutagenesis , Proliferating Cell Nuclear Antigen/metabolism , Protein Interaction Domains and Motifs , Replication Protein A/chemistry
8.
Nucleic Acids Res ; 42(19): 12102-11, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25262353

ABSTRACT

PrimPol is a primase-polymerase found in humans, and other eukaryotes, involved in bypassing lesions encountered during DNA replication. PrimPol employs both translesion synthesis and repriming mechanisms to facilitate lesion bypass by the replisome. PrimPol has been reported to be a potential susceptibility gene associated with the development of myopia. Mutation of tyrosine 89 to aspartic acid (PrimPolY89D) has been identified in a number of cases of high myopia, implicating it in the aetiology of this disorder. Here, we examined whether this mutation resulted in any changes in the molecular and cellular activities associated with human PrimPol. We show that PrimPolY89D has a striking decrease in primase and polymerase activities. The hydrophobic ring of tyrosine is important for retaining wild-type extension activity. We also demonstrate that the decreased activity of PrimPolY89D is associated with reduced affinities for DNA and nucleotides, resulting in diminished catalytic efficiency. Although the structure and stability of PrimPolY89D is altered, its fidelity remains unchanged. This mutation also reduces cell viability after DNA damage and significantly slows replication fork rates in vivo. Together, these findings establish that the major DNA replication defect associated with this PrimPol mutant is likely to contribute to the onset of high myopia.


Subject(s)
DNA Primase/genetics , DNA Replication , DNA-Directed DNA Polymerase/genetics , Multifunctional Enzymes/genetics , Myopia/genetics , Point Mutation , DNA/metabolism , DNA Primase/chemistry , DNA Primase/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Humans , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/metabolism
9.
Nucleic Acids Res ; 42(9): 5830-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24682820

ABSTRACT

PrimPol is a primase-polymerase involved in nuclear and mitochondrial DNA replication in eukaryotic cells. Although PrimPol is predicted to possess an archaeo-eukaryotic primase and a UL52-like zinc finger domain, the role of these domains has not been established. Here, we report that the proposed zinc finger domain of human PrimPol binds zinc ions and is essential for maintaining primase activity. Although apparently dispensable for its polymerase activity, the zinc finger also regulates the processivity and fidelity of PrimPol's extension activities. When the zinc finger is disrupted, PrimPol becomes more promutagenic, has an altered translesion synthesis spectrum and is capable of faithfully bypassing cyclobutane pyrimidine dimer photolesions. PrimPol's polymerase domain binds to both single- and double-stranded DNA, whilst the zinc finger domain binds only to single-stranded DNA. We additionally report that although PrimPol's primase activity is required to restore wild-type replication fork rates in irradiated PrimPol-/- cells, polymerase activity is sufficient to maintain regular replisome progression in unperturbed cells. Together, these findings provide the first analysis of the molecular architecture of PrimPol, describing the activities associated with, and interplay between, its functional domains and defining the requirement for its primase and polymerase activities during nuclear DNA replication.


Subject(s)
DNA Primase/chemistry , DNA-Directed DNA Polymerase/chemistry , Multifunctional Enzymes/chemistry , Animals , Catalytic Domain , Cell Line , DNA Primase/physiology , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/physiology , Electrophoretic Mobility Shift Assay , Humans , Manganese/chemistry , Multifunctional Enzymes/physiology , Protein Binding , Xenopus Proteins/chemistry , Zinc/chemistry
10.
Nucleic Acids Res ; 37(7): 2327-35, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19244310

ABSTRACT

Expression of a proof-reading deficient form of mitochondrial DNA (mtDNA) polymerase gamma, POLG, causes early death accompanied by features of premature ageing in mouse. However, the mechanism of cellular senescence remains unresolved. In addition to high levels of point mutations of mtDNA, the POLG mutator mouse harbours linear mtDNAs. Using one- and two-dimensional agarose gel electrophoresis, we show that the linear mtDNAs derive from replication intermediates and are indicative of replication pausing and chromosomal breakage at the accompanying fragile sites. Replication fork arrest is not random but occurs at specific sites close to two cis-elements known as O(H) and O(L). Pausing at these sites may be enhanced in the case of exonuclease-deficient POLG owing to delayed resumption of DNA replication, or replisome instability. In either case, the mtDNA replication cycle is perturbed and this might explain the progeroid features of the POLG mutator mouse.


Subject(s)
Chromosome Breakage , Chromosome Fragile Sites , DNA Replication , DNA, Mitochondrial/biosynthesis , DNA-Directed DNA Polymerase/genetics , Animals , DNA Polymerase gamma , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/metabolism , Female , Liver/metabolism , Mice , Mice, Mutant Strains , Mitochondria/enzymology , Progeria/genetics , Sequence Analysis, DNA , Single-Strand Specific DNA and RNA Endonucleases
11.
Methods Mol Biol ; 2277: 299-329, 2021.
Article in English | MEDLINE | ID: mdl-34080159

ABSTRACT

In light of accumulating evidence suggestive of cell type-specific vulnerabilities as a result of normal aging processes that adversely affect the brain, as well as age-related neurodegenerative disorders such as Parkinson's disease (PD), the current chapter highlights how we study mitochondrial DNA (mtDNA) changes at a single-cell level. In particular, we comment on increasing questioning of the narrow neurocentric view of such pathologies, where microglia and astrocytes have traditionally been considered bystanders rather than players in related pathological processes. Here we review the contribution made by single-cell mtDNA alterations towards neuronal vulnerability seen in neurodegenerative disorders, focusing on PD as a prominent example. In addition, we give an overview of methodologies that support such experimental investigations. In considering the significant advances that have been made in recent times for developing mitochondria-specific therapies, investigations to account for cell type-specific mitochondrial patterns and how these are altered by disease hold promise for delivering more effective disease-modifying therapeutics.


Subject(s)
Brain/pathology , DNA, Mitochondrial/analysis , DNA, Mitochondrial/genetics , Neurodegenerative Diseases/pathology , Single-Cell Analysis/methods , Aging/genetics , Humans , Neurodegenerative Diseases/genetics , Parkinson Disease/genetics , Polymerase Chain Reaction/methods
12.
Sci Adv ; 7(49): eabh1004, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34860556

ABSTRACT

Replication stress and DNA damage stall replication forks and impede genome synthesis. During S phase, damage tolerance pathways allow lesion bypass to ensure efficient genome duplication. One such pathway is repriming, mediated by Primase-Polymerase (PrimPol) in human cells. However, the mechanisms by which PrimPol is regulated are poorly understood. Here, we demonstrate that PrimPol is phosphorylated by Polo-like kinase 1 (PLK1) at a conserved residue between PrimPol's RPA binding motifs. This phosphorylation is differentially modified throughout the cell cycle, which prevents aberrant recruitment of PrimPol to chromatin. Phosphorylation can also be delayed and reversed in response to replication stress. The absence of PLK1-dependent regulation of PrimPol induces phenotypes including chromosome breaks, micronuclei, and decreased survival after treatment with camptothecin, olaparib, and UV-C. Together, these findings establish that deregulated repriming leads to genomic instability, highlighting the importance of regulating this damage tolerance pathway following fork stalling and throughout the cell cycle.

13.
Nat Commun ; 11(1): 5863, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203852

ABSTRACT

Stalled replication forks can be restarted and repaired by RAD51-mediated homologous recombination (HR), but HR can also perform post-replicative repair after bypass of the obstacle. Bulky DNA adducts are important replication-blocking lesions, but it is unknown whether they activate HR at stalled forks or behind ongoing forks. Using mainly BPDE-DNA adducts as model lesions, we show that HR induced by bulky adducts in mammalian cells predominantly occurs at post-replicative gaps formed by the DNA/RNA primase PrimPol. RAD51 recruitment under these conditions does not result from fork stalling, but rather occurs at gaps formed by PrimPol re-priming and resection by MRE11 and EXO1. In contrast, RAD51 loading at double-strand breaks does not require PrimPol. At bulky adducts, PrimPol promotes sister chromatid exchange and genetic recombination. Our data support that HR at bulky adducts in mammalian cells involves post-replicative gap repair and define a role for PrimPol in HR-mediated DNA damage tolerance.


Subject(s)
DNA Adducts/genetics , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination/physiology , Multifunctional Enzymes/metabolism , 4-Nitroquinoline-1-oxide/toxicity , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , Benz(a)Anthracenes/administration & dosage , Benz(a)Anthracenes/toxicity , Cell Line , DNA Adducts/metabolism , DNA Primase/genetics , DNA, Single-Stranded , DNA-Directed DNA Polymerase/genetics , Humans , Multifunctional Enzymes/genetics , Quinolones/toxicity , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Single Molecule Imaging , Sister Chromatid Exchange
14.
Am J Prev Med ; 35(4): 380-5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687567

ABSTRACT

BACKGROUND: If public health research does not progress beyond descriptive research to testing the effectiveness of interventions that can provide causal data, its contribution to evidence-based practice will not be realized. This paper examined the number and percentage of public health research publications over three time periods (1987-1988, 1997-1998, 2005-2006) for three preventive health issues: tobacco use, alcohol use, and inadequate physical activity. METHODS: A computer-based literature search was conducted, using the Medline and PsycINFO databases. A random sample of 1000 abstracts for each preventive health issue was examined per time period. The abstracts were first categorized as public health or not, and then as data-based or non-data-based public health research. Data-based publications were classified according to research type as measurement-, descriptive/epidemiologic-, or intervention-oriented. Data analysis occurred in 2007. RESULTS: The number of data-based public health research publications examined for each topic ranged between 180 and 346 in 1987-1988, 199 and 322 in 1997-1998, and 302 and 364 in 2005-2006. Despite a significant increase over time in the percentage of intervention publications for tobacco (from 10% in 1987-1988 to 18% in 2005-2006, p=0.008), the majority of publications in all three time periods were descriptive/epidemiologic for all topics (62%-87% in 1987-1988, 64%-85% in 1997-1998, 78%-79% in 2005-2006). There were fewer measurement (3%-7% in 1987-1988, 2%-6% in 1997-1998, 4%-10% in 2005-2006) or intervention publications (9%-31% in 1987-1988, 10%-30% in 1997-1998, 12%-18% in 2005-2006). CONCLUSIONS: Descriptive research does not provide optimal evidence for how to reduce preventable illness. Reasons for the lack of measurement and intervention public health research are explored.


Subject(s)
Bibliometrics , Public Health , Publishing/statistics & numerical data , Research , Alcohol Drinking/prevention & control , Chi-Square Distribution , Evidence-Based Medicine , Exercise/physiology , Humans , Tobacco Use Disorder/prevention & control
15.
Front Cell Neurosci ; 12: 220, 2018.
Article in English | MEDLINE | ID: mdl-30123109

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and is distinguished from other dementias by observation of extracellular Amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles, comprised of fibrils of Aß and tau protein, respectively. At early stages, AD is characterized by minimal neurodegeneration, oxidative stress, nucleolar stress, and altered protein synthesis machinery. It is generally believed that Aß oligomers are the neurotoxic species and their levels in the AD brain correlate with the severity of dementia suggesting that they play a critical role in the pathogenesis of the disease. Here, we show that the incubation of differentiated human neuroblastoma cells (SHSY5Y) with freshly prepared Aß42 oligomers initially resulted in oxidative stress and subtle nucleolar stress in the absence of DNA damage or cell death. The presence of exogenous Aß oligomers resulted in altered nuclear tau levels as well as phosphorylation state, leading to altered distribution of nucleolar tau associated with nucleolar stress. These markers of cellular dysfunction worsen over time alongside a reduction in ribosomal RNA synthesis and processing, a decrease in global level of newly synthesized RNA and reduced protein synthesis. The interplay between Aß and tau in AD remains intriguing and Aß toxicity has been linked to tau phosphorylation and changes in localization. These findings provide evidence for the involvement of Aß42 effects on nucleolar tau and protein synthesis machinery dysfunction in cultured cells. Protein synthesis dysfunction is observed in mild cognitive impairment and early AD in the absence of significant neuronal death.

16.
Acta Neuropathol Commun ; 6(1): 70, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30064522

ABSTRACT

Tau is known for its pathological role in neurodegenerative diseases, including Alzheimer's disease (AD) and other tauopathies. Tau is found in many subcellular compartments such as the cytosol and the nucleus. Although its normal role in microtubule binding is well established, its nuclear role is still unclear. Here, we reveal that tau localises to the nucleolus in undifferentiated and differentiated neuroblastoma cells (SHSY5Y), where it associates with TIP5, a key player in heterochromatin stability and ribosomal DNA (rDNA) transcriptional repression. Immunogold labelling on human brain sample confirms the physiological relevance of this finding by showing tau within the nucleolus colocalises with TIP5. Depletion of tau results in an increase in rDNA transcription with an associated decrease in heterochromatin and DNA methylation, suggesting that under normal conditions tau is involved in silencing of the rDNA. Cellular stress induced by glutamate causes nucleolar stress associated with the redistribution of nucleolar non-phosphorylated tau, in a similar manner to fibrillarin, and nuclear upsurge of phosphorylated tau (Thr231) which doesn't colocalise with fibrillarin or nucleolar tau. This suggests that stress may impact on different nuclear tau species. In addition to involvement in rDNA transcription, nucleolar non-phosphorylated tau also undergoes stress-induced redistribution similar to many nucleolar proteins.


Subject(s)
Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glutamic Acid/pharmacology , tau Proteins/metabolism , Brain/metabolism , Brain/ultrastructure , Cell Differentiation/physiology , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/ultrastructure , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Gene Expression Regulation, Neoplastic/genetics , Heterochromatin/physiology , Histones/metabolism , Humans , Immunoprecipitation , Microscopy, Confocal , Microscopy, Electron , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Transport/drug effects , RNA, Messenger , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Transfection , tau Proteins/genetics , tau Proteins/ultrastructure
17.
Nat Cell Biol ; 20(2): 162-174, 2018 02.
Article in English | MEDLINE | ID: mdl-29335528

ABSTRACT

Mitochondria are subcellular organelles that are critical for meeting the bioenergetic and biosynthetic needs of the cell. Mitochondrial function relies on genes and RNA species encoded both in the nucleus and mitochondria, and on their coordinated translation, import and respiratory complex assembly. Here, we characterize EXD2 (exonuclease 3'-5' domain-containing 2), a nuclear-encoded gene, and show that it is targeted to the mitochondria and prevents the aberrant association of messenger RNAs with the mitochondrial ribosome. Loss of EXD2 results in defective mitochondrial translation, impaired respiration, reduced ATP production, increased reactive oxygen species and widespread metabolic abnormalities. Depletion of the Drosophila melanogaster EXD2 orthologue (CG6744) causes developmental delays and premature female germline stem cell attrition, reduced fecundity and a dramatic extension of lifespan that is reversed with an antioxidant diet. Our results define a conserved role for EXD2 in mitochondrial translation that influences development and ageing.


Subject(s)
Drosophila Proteins/physiology , Exonucleases/genetics , Longevity/genetics , Mitochondrial Proteins/physiology , Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Exonucleases/physiology , Germ Cells/metabolism , Homeostasis , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Stem Cells/metabolism
18.
Nat Commun ; 8: 15222, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28534480

ABSTRACT

DNA damage and secondary structures can stall the replication machinery. Cells possess numerous tolerance mechanisms to complete genome duplication in the presence of such impediments. In addition to translesion synthesis (TLS) polymerases, most eukaryotic cells contain a multifunctional replicative enzyme called primase-polymerase (PrimPol) that is capable of directly bypassing DNA damage by TLS, as well as repriming replication downstream of impediments. Here, we report that PrimPol is recruited to reprime through its interaction with RPA. Using biophysical and crystallographic approaches, we identify that PrimPol possesses two RPA-binding motifs and ascertained the key residues required for these interactions. We demonstrate that one of these motifs is critical for PrimPol's recruitment to stalled replication forks in vivo. In addition, biochemical analysis reveals that RPA serves to stimulate the primase activity of PrimPol. Together, these findings provide significant molecular insights into PrimPol's mode of recruitment to stalled forks to facilitate repriming and restart.


Subject(s)
DNA Primase/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/metabolism , Replication Protein A/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Chickens , Chromatin/metabolism , Crystallography, X-Ray , DNA Primase/chemistry , DNA-Directed DNA Polymerase/chemistry , HEK293 Cells , Humans , Models, Biological , Multifunctional Enzymes/chemistry , Protein Binding , Protein Domains , Replication Protein A/chemistry , Xenopus
19.
Cell Cycle ; 15(7): 908-18, 2016.
Article in English | MEDLINE | ID: mdl-26694751

ABSTRACT

PrimPol is a recently identified member of the archaeo-eukaryote primase (AEP) family of primase-polymerases. It has been shown that this mitochondrial and nuclear localized enzyme plays roles in the maintenance of both unperturbed replication fork progression and in the bypass of lesions after DNA damage. Here, we utilized an avian (DT40) knockout cell line to further study the consequences of loss of PrimPol (PrimPol(-/-)) on the downstream maintenance of cells after UV damage. We report that PrimPol(-/-) cells are more sensitive to UV-C irradiation in colony survival assays than Pol η-deficient cells. Although this increased UV sensitivity is not evident in cell viability assays, we show that this discrepancy is due to an enhanced checkpoint arrest after UV-C damage in the absence of PrimPol. PrimPol(-/-) arrested cells become stalled in G2, where they are protected from UV-induced cell death. Despite lacking an enzyme required for the bypass and maintenance of replication fork progression in the presence of UV damage, we show that PrimPol(-/-) cells actually have an advantage in the presence of a Chk1 inhibitor due to their slow progression through S-phase.


Subject(s)
DNA Damage , DNA Primase/physiology , DNA-Directed DNA Polymerase/physiology , G2 Phase Cell Cycle Checkpoints , Ultraviolet Rays , Animals , Cell Death , Cell Line , Cell Proliferation , Cell Survival/radiation effects , Checkpoint Kinase 1 , Chickens , DNA Primase/genetics , DNA-Directed DNA Polymerase/genetics , G2 Phase/radiation effects , Gene Knockout Techniques , Mitosis/radiation effects , p38 Mitogen-Activated Protein Kinases/physiology
20.
Cell Cycle ; 15(15): 1997-2008, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27230014

ABSTRACT

PrimPol is a DNA damage tolerance enzyme possessing both translesion synthesis (TLS) and primase activities. To uncover its potential role in TLS-mediated IgVλ hypermutation and define its interplay with other TLS polymerases, PrimPol(-/-) and PrimPol(-/-)/Polη(-/-)/Polζ (-/-) gene knockouts were generated in avian cells. Loss of PrimPol had no significant impact on the rate of hypermutation or the mutation spectrum of IgVλ. However, PrimPol(-/-) cells were sensitive to methylmethane sulfonate, suggesting that it may bypass abasic sites at the IgVλ segment by repriming DNA synthesis downstream of these sites. PrimPol(-/-) cells were also sensitive to cisplatin and hydroxyurea, indicating that it assists in maintaining / restarting replication at a variety of lesions. To accurately measure the relative contribution of the TLS and primase activities, we examined DNA damage sensitivity in PrimPol(-/-) cells complemented with polymerase or primase-deficient PrimPol. Polymerase-defective, but not primase-deficient, PrimPol suppresses the hypersensitivity of PrimPol(-/-) cells. This indicates that its primase, rather than TLS activity, is pivotal for DNA damage tolerance. Loss of TLS polymerases, Polη and Polζ has an additive effect on the sensitivity of PrimPol(-/-) cells. Moreover, we found that PrimPol and Polη-Polζ redundantly prevented cell death and facilitated unperturbed cell cycle progression. PrimPol(-/-) cells also exhibited increased sensitivity to a wide variety of chain-terminating nucleoside analogs (CTNAs). PrimPol could perform close-coupled repriming downstream of CTNAs and oxidative damage in vitro. Together, these results indicate that PrimPol's repriming activity plays a central role in reinitiating replication downstream from CTNAs and other specific DNA lesions.


Subject(s)
DNA Damage , DNA Primase/metabolism , DNA Replication , Nucleosides/metabolism , Animals , Biocatalysis/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Chickens , Cisplatin/pharmacology , DNA Replication/drug effects , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin lambda-Chains/genetics , Methyl Methanesulfonate/pharmacology , Somatic Hypermutation, Immunoglobulin/drug effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL