Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
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
2.
Hum Mol Genet ; 31(18): 3083-3094, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35512351

ABSTRACT

BACKGROUND: TASP1 encodes an endopeptidase activating histone methyltransferases of the KMT2 family. Homozygous loss-of-function variants in TASP1 have recently been associated with Suleiman-El-Hattab syndrome. We report six individuals with Suleiman-El-Hattab syndrome and provide functional characterization of this novel histone modification disorder in a multi-omics approach. METHODS: Chromosomal microarray/exome sequencing in all individuals. Western blotting from fibroblasts in two individuals. RNA sequencing and proteomics from fibroblasts in one individual. Methylome analysis from blood in two individuals. Knock-out of tasp1 orthologue in zebrafish and phenotyping. RESULTS: All individuals had biallelic TASP1 loss-of-function variants and a phenotype including developmental delay, multiple congenital anomalies (including cardiovascular and posterior fossa malformations), a distinct facial appearance and happy demeanor. Western blot revealed absence of TASP1. RNA sequencing/proteomics showed HOX gene downregulation (HOXA4, HOXA7, HOXA1 and HOXB2) and dysregulation of transcription factor TFIIA. A distinct methylation profile intermediate between control and Kabuki syndrome (KMT2D) profiles could be produced. Zebrafish tasp1 knock-out revealed smaller head size and abnormal cranial cartilage formation in tasp1 crispants. CONCLUSION: This work further delineates Suleiman-El-Hattab syndrome, a recognizable neurodevelopmental syndrome. Possible downstream mechanisms of TASP1 deficiency include perturbed HOX gene expression and dysregulated TFIIA complex. Methylation pattern suggests that Suleiman-El-Hattab syndrome can be categorized into the group of histone modification disorders including Wiedemann-Steiner and Kabuki syndrome.


Subject(s)
Histone Code , Zebrafish , Abnormalities, Multiple , Animals , Endopeptidases/genetics , Face/abnormalities , Hematologic Diseases , Histone Methyltransferases/genetics , Phenotype , Transcription Factor TFIIA/genetics , Vestibular Diseases , Zebrafish/genetics
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.
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
5.
Am J Med Genet A ; 173(9): 2511-2516, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28688171

ABSTRACT

We describe the characterization of Xeroderma Pigmentosum variant (XPV) in a young Caucasian patient with phototype I, who exhibited a high sensitivity to sunburn and multiple cutaneous tumors at the age of 15 years. Two novel mutations in the POLH gene, which encodes the translesion DNA polymerase η, with loss of function due to two independent exon skippings, are reported to be associated as a compound heterozygous state in the patient. Western blot analysis performed on proteins from dermal fibroblasts derived from the patient and analysis of the mutation spectrum on immunoglobulin genes produced during the somatic hypermutation process in his memory B cells, show the total absence of translesion polymerase η activity in the patient. The total lack of Polη activity, necessary to bypass in an error-free manner UVR-induced pyrimidine dimers following sun exposure, explains the early unusual clinical appearance of this patient.


Subject(s)
DNA-Directed DNA Polymerase/genetics , Skin Neoplasms/genetics , Sunburn/genetics , Xeroderma Pigmentosum/genetics , Adolescent , DNA Damage/genetics , DNA Repair/genetics , Fibroblasts/metabolism , Humans , Male , Mutation , Skin Neoplasms/physiopathology , Sunburn/physiopathology , Sunlight , Xeroderma Pigmentosum/physiopathology
6.
Nucleic Acids Res ; 41(13): 6501-13, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23677613

ABSTRACT

During replication, DNA damage can challenge replication fork progression and cell viability. Homologous Recombination (HR) and Translesion Synthesis (TLS) pathways appear as major players involved in the resumption and completion of DNA replication. How both pathways are coordinated in human cells to maintain genome stability is unclear. Numerous helicases are involved in HR regulation. Among them, the helicase FBH1 accumulates at sites of DNA damage and potentially constrains HR via its anti-recombinase activity. However, little is known about its regulation in vivo. Here, we report a mechanism that controls the degradation of FBH1 after DNA damage. Firstly, we found that the sliding clamp Proliferating Cell Nuclear Antigen (PCNA) is critical for FBH1 recruitment to replication factories or DNA damage sites. We then showed the anti-recombinase activity of FBH1 is partially dependent on its interaction with PCNA. Intriguingly, after its re-localization, FBH1 is targeted for degradation by the Cullin-ring ligase 4-Cdt2 (CRL4(Cdt2))-PCNA pathway via a PCNA-interacting peptide (PIP) degron. Importantly, expression of non-degradable FBH1 mutant impairs the recruitment of the TLS polymerase eta to chromatin in UV-irradiated cells. Thus, we propose that after DNA damage, FBH1 might be required to restrict HR and then degraded by the Cdt2-proteasome pathway to facilitate TLS pathway.


Subject(s)
DNA Damage , DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Chromatin/enzymology , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination , Humans , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ultraviolet Rays
7.
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
8.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35896284

ABSTRACT

BACKGROUND: Clinical studies have highlighted the efficacy of anti-programmed death 1 (αPD-1) monoclonal antibodies in patients with DNA mismatch repair-deficient (MMRD) tumors. However, the responsiveness of MMRD cancers to αPD-1 therapy is highly heterogeneous, and the origins of this variability remain not fully understood. METHODS: 4T1 and CT26 mouse tumor cell lines were inactivated for the MMRD gene Msh2, leading to a massive accumulation of mutations after serial passages of cells. Insertions/deletion events and mutation load were evaluated by whole exome sequencing. Mice bearing highly mutated MMRD tumor or parental tumors were treated with αPD-1 and tumor volume was monitored. Immune cell type abundance was dynamically assessed in the tumor microenvironment and the blood by flow cytometry. Neutrophils were depleted in mice using αLY6G antibody, and regulatory T (Treg) cell population was reduced with αCD25 or anti-cytotoxic T-lymphocytes-associated protein 4 (αCTLA-4) antibodies. Patients with MMRD tumors treated with immune checkpoint blockade-based therapy were retrospectively identified and neutrophil-to-lymphocyte ratio (NLR) was evaluated and examined for correlation with clinical benefit. RESULTS: By recapitulating mismatch repair deficiency in different mouse tumor models, we revealed that elevated circulating tumor-induced neutrophils (TIN) in hypermutated MMRD tumors hampered response to αPD-1 monotherapy. Importantly, depletion of TIN using αLy-6G antibody reduced Treg cells and restored αPD-1 response. Conversely, targeting Treg cells by αCD25 or αCTLA-4 antibodies limited peripheral TIN accumulation and elicited response in αPD-1-resistant MMRD tumors, highlighting a crosstalk between TIN and Treg cells. Thus, αPD-1+αCTLA-4 combination overcomes TIN-induced resistance to αPD-1 in mice bearing MMRD tumors. Finally, in a cohort of human (high microsatellite instability)/MMRD tumors we revealed that early on-treatment change in the NLR ratio may predict resistance to αPD-1 therapy. CONCLUSIONS: TIN countered αPD-1 efficacy in MMRD tumors. Since αCTLA-4 could restrict TIN accumulation, αPD-1+αCTLA-4 combination overcomes αPD-1 resistance in hosts with hypermutated MMRD tumors displaying abnormal neutrophil accumulation.


Subject(s)
Neutrophils , Animals , Humans , Mice , Brain Neoplasms , Colorectal Neoplasms , Microsatellite Instability , Neoplastic Syndromes, Hereditary , Retrospective Studies , Tumor Microenvironment
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
SELECTION OF CITATIONS
SEARCH DETAIL