Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
EMBO J ; 42(20): e110844, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37661798

ABSTRACT

Homologous recombination (HR) is a prominent DNA repair pathway maintaining genome integrity. Mutations in many HR genes lead to cancer predisposition. Paradoxically, the implication of the pivotal HR factor RAD51 on cancer development remains puzzling. Particularly, no RAD51 mouse models are available to address the role of RAD51 in aging and carcinogenesis in vivo. We engineered a mouse model with an inducible dominant-negative form of RAD51 (SMRad51) that suppresses RAD51-mediated HR without stimulating alternative mutagenic repair pathways. We found that in vivo expression of SMRad51 led to replicative stress, systemic inflammation, progenitor exhaustion, premature aging and reduced lifespan, but did not trigger tumorigenesis. Expressing SMRAD51 in a breast cancer predisposition mouse model (PyMT) decreased the number and the size of tumors, revealing an anti-tumor activity of SMRAD51. We propose that these in vivo phenotypes result from chronic endogenous replication stress caused by HR decrease, which preferentially targets progenitors and tumor cells. Our work underlines the importance of RAD51 activity for progenitor cell homeostasis, preventing aging and more generally for the balance between cancer and aging.


Subject(s)
Neoplasms , Rad51 Recombinase , Animals , Mice , Aging/genetics , Carcinogenesis/genetics , Cell Transformation, Neoplastic , DNA Damage , DNA Repair , Homologous Recombination , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
3.
Nucleic Acids Res ; 50(17): 9909-9929, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36107774

ABSTRACT

DNA lesions in S phase threaten genome stability. The DNA damage tolerance (DDT) pathways overcome these obstacles and allow completion of DNA synthesis by the use of specialised translesion (TLS) DNA polymerases or through recombination-related processes. However, how these mechanisms coordinate with each other and with bulk replication remains elusive. To address these issues, we monitored the variation of replication intermediate architecture in response to ultraviolet irradiation using transmission electron microscopy. We show that the TLS polymerase η, able to accurately bypass the major UV lesion and mutated in the skin cancer-prone xeroderma pigmentosum variant (XPV) syndrome, acts at the replication fork to resolve uncoupling and prevent post-replicative gap accumulation. Repriming occurs as a compensatory mechanism when this on-the-fly mechanism cannot operate, and is therefore predominant in XPV cells. Interestingly, our data support a recombination-independent function of RAD51 at the replication fork to sustain repriming. Finally, we provide evidence for the post-replicative commitment of recombination in gap repair and for pioneering observations of in vivo recombination intermediates. Altogether, we propose a chronology of UV damage tolerance in human cells that highlights the key role of polη in shaping this response and ensuring the continuity of DNA synthesis.


Subject(s)
DNA Repair , Xeroderma Pigmentosum , DNA Damage , DNA Replication , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Ultraviolet Rays , Xeroderma Pigmentosum/genetics
4.
Front Immunol ; 13: 871766, 2022.
Article in English | MEDLINE | ID: mdl-35432321

ABSTRACT

Somatic hypermutation (SHM) of immunoglobulin (Ig) genes is a B cell specific process required for the generation of specific and high affinity antibodies during the maturation of the immune response against foreign antigens. This process depends on the activity of both activation-induced cytidine deaminase (AID) and several DNA repair factors. AID-dependent SHM creates the full spectrum of mutations in Ig variable (V) regions equally distributed at G/C and A/T bases. In most mammalian cells, deamination of deoxycytidine into uracil during S phase induces targeted G/C mutagenesis using either direct replication of uracils or TLS mediated bypass, however only the machinery of activated B lymphocytes can generate A/T mutagenesis around AID-created uracils. The molecular mechanism behind the latter remains incompletely understood to date. However, the lack of a cellular model that reproduces both G/C and A/T mutation spectra constitutes the major hurdle to elucidating it. The few available B cell lines used thus far to study Ig SHM indeed undergo mainly G/C mutations, that make them inappropriate or of limited use. In this report, we show that in the Ramos cell line that undergoes constitutive G/C-biased SHM in culture, the low rate of A/T mutations is due to an imbalance in the ubiquitination/deubiquitination reaction of PCNA, with the deubiquitination reaction being predominant. The inhibition of the deubiquitinase complex USP1-UAF1 or the expression of constitutive fusion of ubiquitin to PCNA provides the missing clue required for DNA polymerase η recruitment and thereafter the introduction of A/T base pair (bp) mutations during the process of IgV gene diversification. This study reports the establishment of the first modified human B cell line that recapitulates the mechanism of SHM of Ig genes in vitro.


Subject(s)
Immunoglobulin A , Somatic Hypermutation, Immunoglobulin , Animals , Cell Line , Humans , Immunoglobulin A/genetics , Mammals/metabolism , Mutation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin
5.
EMBO J ; 40(21): e104543, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34533226

ABSTRACT

The DNA polymerase zeta (Polζ) plays a critical role in bypassing DNA damage. REV3L, the catalytic subunit of Polζ, is also essential in mouse embryonic development and cell proliferation for reasons that remain incompletely understood. In this study, we reveal that REV3L protein interacts with heterochromatin components including repressive histone marks and localizes in pericentromeric regions through direct interaction with HP1 dimer. We demonstrate that Polζ/REV3L ensures progression of replication forks through difficult-to-replicate pericentromeric heterochromatin, thereby preventing spontaneous chromosome break formation. We also find that Rev3l-deficient cells are compromised in the repair of heterochromatin-associated double-stranded breaks, eliciting deletions in late-replicating regions. Lack of REV3L leads to further consequences that may be ascribed to heterochromatin replication and repair-associated functions of Polζ, with a disruption of the temporal replication program at specific loci. This is correlated with changes in epigenetic landscape and transcriptional control of developmentally regulated genes. These results reveal a new function of Polζ in preventing chromosome instability during replication of heterochromatic regions.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , DNA/genetics , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Heterochromatin/metabolism , Animals , Cell Line , Cell Line, Transformed , Cell Proliferation , Chromobox Protein Homolog 5/genetics , Chromobox Protein Homolog 5/metabolism , Chromosomal Instability , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Embryo, Mammalian , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , HeLa Cells , Heterochromatin/chemistry , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Signal Transduction
6.
Eur J Cancer ; 84: 290-303, 2017 10.
Article in English | MEDLINE | ID: mdl-28846956

ABSTRACT

Immune checkpoint inhibitors have demonstrated unprecedented clinical activity in a wide range of cancers. Significant therapeutic responses have recently been observed in patients presenting mismatch repair-deficient (MMRD) tumours. MMRD cancers exhibit a remarkably high rate of mutations, which can result in the formation of neoantigens, hypothesised to enhance the antitumour immune response. In addition to MMRD tumours, cancers mutated in the exonuclease domain of the catalytic subunit of the DNA polymerase epsilon (POLE) also exhibit an ultramutated genome and are thus likely to benefit from immunotherapy. In this review, we provide an overview of recent data on hypermutated tumours, including MMRD and POLE-mutated cancers, with a focus on their distinctive clinicopathological and molecular characteristics as well as their immune environment. We also discuss the emergence of immune therapy to treat these hypermutated cancers, and we comment on the recent Food and Drug Administration approval of an immune checkpoint inhibitor, the programmed cell death 1 antibody (pembrolizumab, Keytruda), for the treatment of patients with metastatic MMRD cancers regardless of the tumour type. This breakthrough represents a turning point in the management of these hypermutated tumours and paves the way for broader strategies in immunoprecision medicine.


Subject(s)
Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Immunotherapy/methods , Mutation , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine/methods , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , DNA Mismatch Repair , DNA Mutational Analysis , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Genetic Predisposition to Disease , Humans , Microsatellite Instability , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/pathology , Phenotype , Poly-ADP-Ribose Binding Proteins , Predictive Value of Tests , Tumor Escape , Tumor Microenvironment
7.
Nat Med ; 23(5): 568-578, 2017 May.
Article in English | MEDLINE | ID: mdl-28394329

ABSTRACT

Chromosomal instability (CIN), a feature of most adult neoplasms from their early stages onward, is a driver of tumorigenesis. However, several malignancy subtypes, including some triple-negative breast cancers, display a paucity of genomic aberrations, thus suggesting that tumor development may occur in the absence of CIN. Here we show that the differentiation status of normal human mammary epithelial cells dictates cell behavior after an oncogenic event and predetermines the genetic routes toward malignancy. Whereas oncogene induction in differentiated cells induces massive DNA damage, mammary stem cells are resistant, owing to a preemptive program driven by the transcription factor ZEB1 and the methionine sulfoxide reductase MSRB3. The prevention of oncogene-induced DNA damage precludes induction of the oncosuppressive p53-dependent DNA-damage response, thereby increasing stem cells' intrinsic susceptibility to malignant transformation. In accord with this model, a subclass of breast neoplasms exhibit unique pathological features, including high ZEB1 expression, a low frequency of TP53 mutations and low CIN.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Cell Differentiation/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , Methionine Sulfoxide Reductases/genetics , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin Immunoprecipitation , DNA Damage , Epithelial Cells/cytology , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoblotting , Mammary Glands, Human/cytology , Methionine Sulfoxide Reductases/metabolism , Mice, Inbred NOD , Middle Aged , Reactive Oxygen Species , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Stem Cells/cytology , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Young Adult , Zinc Finger E-box-Binding Homeobox 1/metabolism
8.
Oncotarget ; 8(23): 37104-37114, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28415748

ABSTRACT

Oncogenes trigger replicative stress that can lead to genetic instability, which participates in cancer progression. Thus, determining how cells cope with replicative stress can help our understanding of oncogenesis and lead to the identification of new antitumor treatment targets. We previously showed that constitutive overexpression of the oncogenic transcription factor Spi1/PU.1 leads to pre-leukemic cells that have a shortened S phase duration with an increased replication fork speed and increased mutability in the absence of DNA breaks. Here, we demonstrate that the S phase checkpoint protein CHK1 is maintained in a low phosphorylation state in Spi1/PU.1-overexpressing cells and provide evidence that this is not due to negative control of its primary kinase ATR. Notably, we found that the expression of the CHK1 phosphatase PP1α is increased in Spi1/PU.1-overexpressing cells. By exogenously modulating its activity, we demonstrate that PP1α is required to maintain CHK1 in a dephosphorylated state and, more importantly, that it is responsible for the accelerated replication fork progression in Spi1/PU.1-overexpressing cells. These results identify a novel pathway by which an oncogene influences replication in the absence of DNA damage.


Subject(s)
Checkpoint Kinase 1/metabolism , DNA Replication , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Cycle/genetics , Cells, Cultured , Checkpoint Kinase 1/genetics , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Mice, Transgenic , Phosphorylation , Protein Phosphatase 1/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , Trans-Activators/genetics
9.
Nat Commun ; 7: 13326, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27811911

ABSTRACT

Translesion polymerase eta (polη) was characterized for its ability to replicate ultraviolet-induced DNA lesions that stall replicative polymerases, a process promoted by Rad18-dependent PCNA mono-ubiquitination. Recent findings have shown that polη also acts at intrinsically difficult to replicate sequences. However, the molecular mechanisms that regulate its access to these loci remain elusive. Here, we uncover that polη travels with replication forks during unchallenged S phase and this requires its SUMOylation on K163. Abrogation of polη SUMOylation results in replication defects in response to mild replication stress, leading to chromosome fragments in mitosis and damage transmission to daughter cells. Rad18 plays a pivotal role, independently of its ubiquitin ligase activity, acting as a molecular bridge between polη and the PIAS1 SUMO ligase to promote polη SUMOylation. Our results provide the first evidence that SUMOylation represents a new way to target polη to replication forks, independent of the Rad18-mediated PCNA ubiquitination, thereby preventing under-replicated DNA.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Humans , Proliferating Cell Nuclear Antigen/genetics , Protein Inhibitors of Activated STAT/genetics , S Phase/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation/genetics , Ubiquitin-Protein Ligases/genetics
10.
DNA Repair (Amst) ; 29: 154-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25766642

ABSTRACT

Xeroderma pigmentosum variant (XP-V) is a rare genetic disease, characterized by sunlight sensitivity and predisposition to cutaneous malignancies. XP-V is caused by a deficiency in DNA polymerase eta (Polη) that plays a pivotal role in translesion synthesis by bypassing UV-induced pyrimidine dimers. Previously we identified a new Polη variant containing two missense mutations, one mutation within the bipartite NLS (T692A) and a second mutation on the stop codon (X714W) leading to a longer protein with an extra 8 amino acids (721 instead of 713 AA). First biochemical analysis revealed that this Polη missense variant was barely detectable by western blot. As this mutant is extremely unstable and is nearly undetectable, a definitive measure of its functional deficit in cells has not been explored. Here we report the molecular and cellular characterization of this missense variant. In cell free extracts, the extra 8 amino acids in the C-terminal of Polη(721) only slightly reduce the bypass efficiency through CPD lesions. In vivo, Polη(721) accumulates in replication factories and interacts with mUb-PCNA albeit at lower level than Polη(wt). XP-V cells overexpressing Polη(721) were only slightly UV-sensitive. Altogether, our data strongly suggest that Polη(721) is functional and that the patient displays a XP-V phenotype because the mutant protein is excessively unstable. We then investigated the molecular mechanisms involved in this excessive proteolysis. We showed that Polη(721) is degraded by the proteasome in an ubiquitin-dependent manner and that this proteolysis is independent of the E3 ligases, CRL4(cdt2) and Pirh2, reported to promote Polη degradation. We then demonstrated that the extra 8 amino acids of Polη(721) do not act as a degron but rather induce a conformational change of the Polη C-terminus exposing its bipartite NLS as well as a sequence close to its UBZ to the ubiquitin/proteasome system. Interestingly we showed that the clinically approved proteasome inhibitor, Bortezomib restores the levels of Polη(721) suggesting that this might be a therapeutic approach to preventing tumor development in certain XP-V patients harboring missense mutations.


Subject(s)
DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Mutation, Missense , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Codon, Terminator , Gene Expression , Humans , Nuclear Proteins/metabolism , Protein Stability , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/metabolism , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
11.
Nucleic Acids Res ; 43(4): 2116-25, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25662213

ABSTRACT

Switching between replicative and translesion synthesis (TLS) DNA polymerases are crucial events for the completion of genomic DNA synthesis when the replication machinery encounters lesions in the DNA template. In eukaryotes, the translesional DNA polymerase η (Polη) plays a central role for accurate bypass of cyclobutane pyrimidine dimers, the predominant DNA lesions induced by ultraviolet irradiation. Polη deficiency is responsible for a variant form of the Xeroderma pigmentosum (XPV) syndrome, characterized by a predisposition to skin cancer. Here, we show that the FF483-484 amino acids in the human Polη (designated F1 motif) are necessary for the interaction of this TLS polymerase with POLD2, the B subunit of the replicative DNA polymerase δ, both in vitro and in vivo. Mutating this motif impairs Polη function in the bypass of both an N-2-acetylaminofluorene adduct and a TT-CPD lesion in cellular extracts. By complementing XPV cells with different forms of Polη, we show that the F1 motif contributes to the progression of DNA synthesis and to the cell survival after UV irradiation. We propose that the integrity of the F1 motif of Polη, necessary for the Polη/POLD2 interaction, is required for the establishment of an efficient TLS complex.


Subject(s)
DNA Damage , DNA Polymerase III/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Cell Cycle , Cell Line , Cell Survival , Humans , Protein Interaction Domains and Motifs , Protein Subunits/metabolism , Ultraviolet Rays
12.
Mol Cell ; 57(1): 123-37, 2015 01 08.
Article in English | MEDLINE | ID: mdl-25533188

ABSTRACT

The SLX4 Fanconi anemia protein is a tumor suppressor that may act as a key regulator that engages the cell into specific genome maintenance pathways. Here, we show that the SLX4 complex is a SUMO E3 ligase that SUMOylates SLX4 itself and the XPF subunit of the DNA repair/recombination XPF-ERCC1 endonuclease. This SLX4-dependent activity is mediated by a remarkably specific interaction between SLX4 and the SUMO-charged E2 conjugating enzyme UBC9 and relies not only on newly identified SUMO-interacting motifs (SIMs) in SLX4 but also on its BTB domain. In contrast to its ubiquitin-binding UBZ4 motifs, SLX4 SIMs are dispensable for its DNA interstrand crosslink repair functions. Instead, while detrimental in response to global replication stress, the SUMO E3 ligase activity of the SLX4 complex is critical to prevent mitotic catastrophe following common fragile site expression.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Genome , Protein Subunits/metabolism , Recombinases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cell Line, Tumor , DNA Replication , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genomic Instability , Humans , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinases/genetics , Sequence Alignment , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics
13.
Hum Mutat ; 35(1): 117-28, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24130121

ABSTRACT

Xeroderma pigmentosum variant (XP-V) is a rare genetic disease, characterized by some sunlight sensitivity and predisposition to cutaneous malignancies. We described clinical and genetic features of the largest collection ever published of 23 XPV patients (ages between 21 and 86) from 20 unrelated families. Primary fibroblasts from patients showed normal nucleotide excision repair but UV-hypersensitivity in the presence of caffeine, a signature of the XP-V syndrome. 87% of patients developed skin tumors with a median age of 21 for the first occurrence. The median numbers of basal-cell carcinoma was 13 per patient, six for squamous-cell carcinoma, and five for melanoma. XP-V is due to defects in the translesion-synthesis DNA polymerase Polη coded by the POLH gene. DNA sequencing of POLH revealed 29 mutations, where 12 have not been previously identified, leading to truncated polymerases in 69% of patients. Four missense mutations are correlated with the protein stability by structural modeling of the Polη polymerase domain. There is a clear relationship between the types of missense mutations and clinical severity. For truncating mutations, which lead to an absence of or to inactive proteins, the life-cumulated UV exposure is probably the best predictor of cancer incidence, reinforcing the necessity to protect XP-Vs from sun exposure.


Subject(s)
DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Skin Neoplasms/genetics , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology , Adult , Aged , Aged, 80 and over , Caffeine , Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cells, Cultured , DNA Repair , Female , Fibroblasts/metabolism , Genetic Variation , Genotype , Humans , Male , Melanoma/epidemiology , Melanoma/genetics , Melanoma/pathology , Middle Aged , Models, Molecular , Mutation, Missense , Phenotype , Protein Stability , Retrospective Studies , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Ultraviolet Rays , Xeroderma Pigmentosum/complications , Young Adult
14.
Environ Mol Mutagen ; 53(9): 752-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23076824

ABSTRACT

During translesion synthesis (TLS), low-fidelity polymerases of the Y-family polymerases bypass DNA damages that block the progression of conventional processive DNA polymerases, thereby allowing the completion of DNA replication. Among the TLS polymerases, DNA polymerase eta (polη) performs nucleotide incorporation past ultraviolet (UV) photoproducts and is deficient in cancer-prone xeroderma pigmentosum variant (XPV) syndrome. Upon UV irradiation, the DNA sliding clamp PCNA is monoubiquitylated on its conserved Lys-164. This event is considered to facilitate the TLS process in vivo since polη preferentially interacts with monoubiquitylated PCNA through its ubiquitin-binding domain (UBZ) as well as its PCNA interacting peptide (PIP)-box. However, recent observations questioned this model. Therefore, in this study, we re-examined the relative contribution of the regulatory UBZ and PIP domains of polη in response to UVC. We show that simultaneous invalidation of both motifs confers sensitivity to UVC, sensitization by low concentrations of caffeine, prolonged inhibition of DNA synthesis and persistent S phase checkpoint activation, all characteristic features of XPV cells. While each domain is essential for efficient accumulation of polη in replication factories, mutational inactivation of UBZ or PIP motif only confers a slight sensitivity to UVC indicating that, although informative, polη focus analysis is not a reliable tool to assess the polη's ability to function in TLS in vivo. Taken together, these data indicate that PIP and UBZ motifs are not required for recruitment but for retention of polη at sites of stalled replication forks. We propose that this is a way to ensure that a sufficient amount of the protein is available for its bypass function.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , Cell Line, Transformed , DNA-Directed DNA Polymerase/chemistry , Flow Cytometry , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
15.
Mol Cell ; 43(4): 649-62, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855803

ABSTRACT

Posttranslational modification of PCNA by ubiquitin plays an important role in coordinating the processes of DNA damage tolerance during DNA replication. The monoubiquitination of PCNA was shown to facilitate the switch between the replicative DNA polymerase with the low-fidelity polymerase eta (η) to bypass UV-induced DNA lesions during replication. Here, we show that in response to oxidative stress, PCNA becomes transiently monoubiquitinated in an S phase- and USP1-independent manner. Moreover, Polη interacts with mUb-PCNA at sites of oxidative DNA damage via its PCNA-binding and ubiquitin-binding motifs. Strikingly, while functional base excision repair is not required for this modification of PCNA or Polη recruitment to chromatin, the presence of hMsh2-hMsh6 is indispensable. Our findings highlight an alternative pathway in response to oxidative DNA damage that may coordinate the removal of oxidatively induced clustered DNA lesions and could explain the high levels of oxidized DNA lesions in MSH2-deficient cells.


Subject(s)
DNA Damage , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase/physiology , MutS Homolog 2 Protein/physiology , Oxidative Stress , Proliferating Cell Nuclear Antigen/physiology , Arabidopsis Proteins , Cell Line , Chromatin/metabolism , DNA Polymerase beta/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Endopeptidases/metabolism , Humans , MutS Homolog 2 Protein/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Specific Proteases , Ubiquitination , X-ray Repair Cross Complementing Protein 1
16.
Hum Mol Genet ; 19(9): 1690-701, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20123862

ABSTRACT

DNA polymerase eta (poleta) performs translesion synthesis past ultraviolet (UV) photoproducts and is deficient in cancer-prone xeroderma pigmentosum variant (XP-V) syndrome. The slight sensitivity of XP-V cells to UV is dramatically enhanced by low concentrations of caffeine. So far, the biological explanation for this feature remains elusive. Using DNA combing, we showed that translesion synthesis defect leads to a strong reduction in the number of active replication forks and a high proportion of stalled forks in human cells, which contrasts with budding yeast. Moreover, extensive regions of single-strand DNA are formed during replication in irradiated XP-V cells, leading to an over-activation of ATR/Chk1 pathway after low UVC doses. Addition of a low concentration of caffeine post-irradiation, although inefficient to restore S-phase progression, significantly decreases Chk1 activation and abrogates DNA synthesis in XP-V cells. While inhibition of Chk1 activity by UCN-01 prevents UVC-induced S-phase delay in wild-type cells, it aggravates replication defect in XP-V cells by increasing fork stalling. Consequently, UCN-01 sensitizes XP-V cells to UVC as caffeine does. Our findings indicate that poleta acts at stalled forks to resume their progression, preventing the requirement for efficient replication checkpoint after low UVC doses. In the absence of poleta, Chk1 kinase becomes essential for replication resumption by alternative pathways, via fork stabilization.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA Replication/physiology , DNA/biosynthesis , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Xeroderma Pigmentosum/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Line , Checkpoint Kinase 1 , DNA Replication/genetics , DNA-Directed DNA Polymerase/metabolism , Flow Cytometry , Humans , RNA, Small Interfering/genetics , Transfection , Ultraviolet Rays/adverse effects
17.
EMBO J ; 27(21): 2883-95, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18923427

ABSTRACT

Human DNA polymerase iota (poliota) is a unique member of the Y-family of specialised polymerases that displays a 5'deoxyribose phosphate (dRP) lyase activity. Although poliota is well conserved in higher eukaryotes, its role in mammalian cells remains unclear. To investigate the biological importance of poliota in human cells, we generated fibroblasts stably downregulating poliota (MRC5-pol iota(KD)) and examined their response to several types of DNA-damaging agents. We show that cell lines downregulating poliota exhibit hypersensitivity to DNA damage induced by hydrogen peroxide (H(2)O(2)) or menadione but not to ethylmethane sulphonate (EMS), UVC or UVA. Interestingly, extracts from cells downregulating poliota show reduced base excision repair (BER) activity. In addition, poliota binds to chromatin after treatment of cells with H(2)O(2) and interacts with the BER factor XRCC1. Finally, green fluorescent protein-tagged poliota accumulates at the sites of oxidative DNA damage in living cells. This recruitment is partially mediated by its dRP lyase domain and ubiquitin-binding domains. These data reveal a novel role of human poliota in protecting cells from oxidative damage.


Subject(s)
Cytoprotection , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Oxidative Stress , Alkylating Agents/pharmacology , Cell Death/drug effects , Cell Death/radiation effects , Cell Extracts , Cell Line , Chromatin/enzymology , Cytoprotection/drug effects , Cytoprotection/radiation effects , DNA Damage , DNA Polymerase beta/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/deficiency , Down-Regulation/drug effects , Down-Regulation/radiation effects , Fibroblasts/drug effects , Fibroblasts/radiation effects , G1 Phase/drug effects , G1 Phase/radiation effects , Humans , Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Structure, Tertiary , Ultraviolet Rays , Uracil/metabolism , X-ray Repair Cross Complementing Protein 1 , DNA Polymerase iota
18.
Cancer Res ; 67(6): 2526-34, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363570

ABSTRACT

To study the relationships between different DNA repair pathways, we established a set of clones in which one specific DNA repair gene was silenced using long-term RNA interference in HeLa cell line. We focus here on genes involved in either nucleotide excision repair (XPA and XPC) or nonhomologous end joining (NHEJ; DNA-PKcs and XRCC4). As expected, XPA(KD) (knock down) and XPC(KD) cells were highly sensitive to UVC. DNA-PKcs(KD) and XRCC4(KD) cells presented an increased sensitivity to various inducers of double-strand breaks (DSBs) and a 70% to 80% reduction of in vitro NHEJ activity. Long-term silencing of XPC gene expression led to an increased sensitivity to etoposide, a topoisomerase II inhibitor that creates DSBs through the progression of DNA replication forks. XPC(KD) cells also showed intolerance toward acute gamma-ray irradiation. We showed that XPC(KD) cells exhibited an altered spectrum of NHEJ products with decreased levels of intramolecular joined products. Moreover, in both XPC(KD) and DNA-PKcs(KD) cells, XRCC4 and ligase IV proteins were mobilized on damaged nuclear structures at lower doses of DSB inducer. In XPC-proficient cells, XPC protein was released from nuclear structures after induction of DSBs. By contrast, silencing of XPA gene expression did not have any effect on sensitivity to DSB or NHEJ. Our results suggest that XPC deficiency, certainly in combination with other genetic defects, may contribute to impair DSB repair.


Subject(s)
DNA Damage , DNA Repair/physiology , DNA-Binding Proteins/genetics , Gene Silencing , DNA Ligase ATP , DNA Ligases/metabolism , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Activated Protein Kinase/biosynthesis , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Etoposide/pharmacology , Gamma Rays , HeLa Cells , Humans , RNA Interference
19.
Mol Cancer Res ; 3(9): 519-29, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16179499

ABSTRACT

We developed and characterized replicative small interfering RNA (siRNA) vectors for efficient, specific, and long-term gene silencing in human cells. We created stable XPA(KD) and XPC(KD) (knockdown) syngeneic cell lines to mimic human cancer-prone syndromes. We also silenced (HSA)KIN17. Several clones displaying undetectable protein levels of XPA, XPC, or (HSA)kin17 were grown for more than 300 days. This stability of gene silencing over several months of culture allows us to assess the specific involvement of these proteins in UVC sensitivity in syngeneic cells. Unlike XPA, (HSA)KIN17, and XPC gene silencing dramatically impeded HeLa cell growth for several weeks after transfection. As expected, XPA(KD) and XPC(KD) HeLa cells were highly UVC sensitive. They presented an impaired unscheduled DNA synthesis after UVC irradiation. Interestingly, XPC(KD) HeLa clones were more sensitive to UVC than their XPA(KD) or KIN17(KD) counterparts. Hygromycin B withdrawal led to the total disappearance of EBV vectors and the resumption of normal XPA or XPC protein levels. Whereas reverted XPA(KD) cells recovered a normal UVC sensitivity, XPC(KD) cells remained highly sensitive, suggestive of irreversible damage following long-term XPC silencing. Our results show that in HeLa cells, (HSA)kin17 participates indirectly in early events following UVC irradiation, and XPC deficiency strongly affects cell physiology and contributes to UVC sensitivity to a greater extent than does XPA. EBV-based siRNA vectors improve the interest of siRNA by permitting long-term gene silencing without the safety concerns inherent in viral-based siRNA vehicles.


Subject(s)
DNA Repair , Gene Silencing , Genetic Vectors/physiology , Herpesvirus 4, Human/genetics , RNA, Small Interfering/genetics , Ultraviolet Rays , Blotting, Southern , Blotting, Western , Cell Cycle/radiation effects , Cell Survival/radiation effects , DNA-Binding Proteins/genetics , Flow Cytometry , HeLa Cells/physiology , HeLa Cells/radiation effects , Humans , Nuclear Proteins/genetics , RNA-Binding Proteins , Tumor Stem Cell Assay , Xeroderma Pigmentosum Group A Protein
20.
Mol Cancer Res ; 1(7): 519-31, 2003 May.
Article in English | MEDLINE | ID: mdl-12754299

ABSTRACT

The Homo sapiens kin17 ((HSA)kin17) protein is a chromatin-associated protein conserved during evolution and overproduced in certain human tumor cell lines. For the first time, immunoelectron microscopy analysis of endogenous (HSA)kin17 protein revealed an ultrastructural co-localization of (HSA)kin17 and bromodeoxyuridine (BrdUrd) at sites of DNA replication after either short (15 min) or long (120 min) pulses of BrdUrd labeling. After hydroxyurea (HU) or L-mimosine (Mimo) block and withdrawal, we observed that (HSA)kin17 was recruited onto the chromatin during the re-entry and the progression in the S phase. These results are consistent with a major role of (HSA)kin17 protein in DNA replication factories. Other treatments hampering replication fork progression and/or inducing double-strand breaks also triggered an accumulation and a concentration of the chromatin-bound (HSA)kin17 protein into large intranuclear foci 24 h post-treatment. Moreover, HU- and Mimo-induced (HSA)kin17 foci were retained in the nucleus after detergent extraction, suggesting a strong association with nuclear structures. Gel filtration analyses of cellular extracts showed that endogenous (HSA)kin17 protein co-eluted with both replication proteins RPA32 and RPA70 in a fraction containing complexes of M(r) 600,000. Interestingly, HU-induced G(1)-S arrest triggered an increase in the molecular weight of complexes containing (HSA)kin17 protein. Hence, treatments interfering with either initiation and/or elongation of DNA replication also recruited chromatin-bound (HSA)kin17 protein. We hypothesize that in the presence of unrepaired DNA damage, (HSA)kin17 protein concentrated into high molecular weight complexes probably to create a bridge that contributes to the harmonization of DNA replication and repair.


Subject(s)
DNA Replication/physiology , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Animals , Antineoplastic Agents/toxicity , Bromodeoxyuridine , Carcinoma, Non-Small-Cell Lung , Cell Division , Colonic Neoplasms , DNA Replication/drug effects , DNA-Binding Proteins/analysis , Flow Cytometry , Humans , Hydroxyurea/toxicity , Lung Neoplasms , Microscopy, Immunoelectron , Mimosine/toxicity , Nuclear Proteins/analysis , RNA-Binding Proteins , Tumor Cells, Cultured , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...