Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 145(47): 25686-25694, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-37931025

ABSTRACT

Hexagonal boron nitride (hBN) is a highly selective catalyst for the oxidative dehydrogenation of propane (ODHP) to propylene. Using a variety of ex situ characterization techniques, the activity of the catalyst has been attributed to the formation of an amorphous boron oxyhydroxide surface layer. The ODHP reaction mechanism proceeds via a combination of surface mediated and gas phase propagated radical reactions with the relative importance of both depending on the surface-to-void-volume ratio. Here we demonstrate the unique capability of operando X-ray Raman spectroscopy (XRS) to investigate the oxyfunctionalization of the catalyst under reaction conditions (1 mm outer diameter reactor, 500 to 550 °C, P = 30 kPa C3H8, 15 kPa O2, 56 kPa He). We probe the effect of a water cofeed on the surface of the activated catalyst and find that water removes boron oxyhydroxide from the surface, resulting in a lower reaction rate when the surface reaction dominates and an enhanced reaction rate when the gas phase contribution dominates. Computational description of the surface transformations at an atomic-level combined with high precision XRS spectra simulations with the OCEAN code rationalize the experimental observations. This work establishes XRS as a powerful technique for the investigation of light element-containing catalysts under working conditions.

2.
Carcinogenesis ; 43(2): 160-169, 2022 03 24.
Article in English | MEDLINE | ID: mdl-34919656

ABSTRACT

The prevalent cancer predisposition Lynch syndrome (LS, OMIM #120435) is caused by an inherited heterozygous defect in any of the four core DNA mismatch repair (MMR) genes MSH2, MSH6, MLH1 or PMS2. MMR repairs errors by the replicative DNA polymerases in all proliferating tissues. Its deficiency, following somatic loss of the wild-type copy, results in a spontaneous mutator phenotype that underlies the rapid development of, predominantly, colorectal cancer (CRC) in LS. Here, we have addressed the hypothesis that aberrant responses of intestinal stem cells to diet-derived mutagens may be causally involved in the restricted cancer tropism of LS. To test this we have generated a panel of isogenic mouse embryonic stem (mES) cells with heterozygous or homozygous disruption of multiple MMR genes and investigated their responses to the common dietary mutagen and carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Our data reveal that PhIP can inactivate the wild-type allele of heterozygous mES cells via the induction of either loss of heterozygosity (LOH) or intragenic mutations. Moreover, while protective DNA damage signaling (DDS) is compromised, PhIP induces more mutations in Msh2, Mlh1, Msh6 or Pms2-deficient mES cells than in wild-type cells. Combined with their spontaneous mutator phenotypes, this results in a compound hypermutator phenotype. Together, these results indicate that dietary mutagens may promote CRC development in LS at multiple levels, providing a rationale for dietary modifications in the management of LS.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Animals , Brain Neoplasms , Colorectal Neoplasms , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Damage , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Diet/adverse effects , Germ-Line Mutation , Mice , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Mutagens/toxicity , Neoplastic Syndromes, Hereditary
3.
DNA Repair (Amst) ; 93: 102923, 2020 09.
Article in English | MEDLINE | ID: mdl-33087264

ABSTRACT

Canonical DNA mismatch repair (MMR) excises base-base mismatches to increase the fidelity of DNA replication. Thus, loss of MMR leads to increased spontaneous mutagenesis. MMR genes also are involved in the suppression of mutagenic, and the induction of protective, responses to various types of DNA damage. In this review we describe these non-canonical roles of MMR at different lesion types. Loss of non-canonical MMR gene functions may have important ramifications for the prevention, development and treatment of colorectal cancer associated with inherited MMR gene defects in Lynch syndrome. This graphical review pays tribute to Samuel H. Wilson. Sam not only made seminal contributions to understanding base excision repair, particularly with respect to structure-function relationships in DNA polymerase ß but also, as Editor of DNA Repair, has maintained a high standard of the journal.


Subject(s)
Colorectal Neoplasms/genetics , DNA Damage , DNA Mismatch Repair , Colorectal Neoplasms, Hereditary Nonpolyposis , DNA/metabolism , DNA Replication , Humans , Mutagenesis
4.
Cell Res ; 29(10): 783-784, 2019 10.
Article in English | MEDLINE | ID: mdl-31434995
5.
Ultramicroscopy ; 188: 52-58, 2018 05.
Article in English | MEDLINE | ID: mdl-29554486

ABSTRACT

The key to understanding the performance of Li-O2 batteries is to study the chemical and structural properties of their discharge product(s) at the nanometer scale. Using TEM for this purpose poses challenges due to the sensitivity of samples to air and electron beams. This paper describes our use of in situ EELS to evaluate experimental procedures to reduce electron-beam degradation and presents methods to deal with air sensitivity. Our results show that Li2O2 decomposition is dependent on the total dose and is approximately 4-5 times more pronounced at 80 than at 200 kV. We also demonstrate the benefits of using low-dose-rate STEM. We show further that a "graphene cell", which encapsulates the sample within graphene sheets, can protect the sample against air and e-beam damage.

6.
Org Biomol Chem ; 15(28): 5944-5948, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28675229

ABSTRACT

Utilizing rhodium catalysis, aryl nucleophiles generated via carbon-carbon single bond activation successfully undergo oxidative coupling with Michael acceptors. The reaction scope encompasses a broad range of nucleophiles generated from quinolinyl ketones as well as a series of electron deficient terminal alkenes, illustrating the broad potential of intersecting carbon-carbon bond activation with synthetically useful coupling methodologies. The demonstrated oxidative coupling produces a range of cinnamyl derivatives, several of which are challenging to prepare via conventional routes.

7.
Oncotarget ; 6(23): 19342-3, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26305846
8.
Nucleic Acids Res ; 43(17): 8325-39, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26187992

ABSTRACT

REV1 is a eukaryotic member of the Y-family of DNA polymerases involved in translesion DNA synthesis and genome mutagenesis. Recently, REV1 is also found to function in homologous recombination. However, it remains unclear how REV1 is recruited to the sites where homologous recombination is processed. Here, we report that loss of mammalian REV1 results in a specific defect in replication-associated gene conversion. We found that REV1 is targeted to laser-induced DNA damage stripes in a manner dependent on its ubiquitin-binding motifs, on RAD18, and on monoubiquitinated FANCD2 (FANCD2-mUb) that associates with REV1. Expression of a FANCD2-Ub chimeric protein in RAD18-depleted cells enhances REV1 assembly at laser-damaged sites, suggesting that FANCD2-mUb functions downstream of RAD18 to recruit REV1 to DNA breaks. Consistent with this suggestion we found that REV1 and FANCD2 are epistatic with respect to sensitivity to the double-strand break-inducer camptothecin. REV1 enrichment at DNA damage stripes also partially depends on BRCA1 and BRCA2, components of the FANCD2/BRCA supercomplex. Intriguingly, analogous to FANCD2-mUb and BRCA1/BRCA2, REV1 plays an unexpected role in protecting nascent replication tracts from degradation by stabilizing RAD51 filaments. Collectively these data suggest that REV1 plays multiple roles at stalled replication forks in response to replication stress.


Subject(s)
DNA Damage , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/physiology , Nuclear Proteins/physiology , Nucleotidyltransferases/physiology , Camptothecin/toxicity , Cell Line , DNA/metabolism , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Conversion , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Protein Interaction Domains and Motifs , Stress, Physiological/genetics , Topoisomerase I Inhibitors/toxicity , Ubiquitin-Protein Ligases
9.
10.
Nat Commun ; 6: 7199, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26068067

ABSTRACT

Möbius syndrome (MBS) is a neurological disorder that is characterized by paralysis of the facial nerves and variable other congenital anomalies. The aetiology of this syndrome has been enigmatic since the initial descriptions by von Graefe in 1880 and by Möbius in 1888, and it has been debated for decades whether MBS has a genetic or a non-genetic aetiology. Here, we report de novo mutations affecting two genes, PLXND1 and REV3L in MBS patients. PLXND1 and REV3L represent totally unrelated pathways involved in hindbrain development: neural migration and DNA translesion synthesis, essential for the replication of endogenously damaged DNA, respectively. Interestingly, analysis of Plxnd1 and Rev3l mutant mice shows that disruption of these separate pathways converge at the facial branchiomotor nucleus, affecting either motoneuron migration or proliferation. The finding that PLXND1 and REV3L mutations are responsible for a proportion of MBS patients suggests that de novo mutations in other genes might account for other MBS patients.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Mobius Syndrome/genetics , Mutation , Animals , DNA Damage , Exome , Heterozygote , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins , Mice , Mice, Mutant Strains
11.
J Cell Biol ; 209(1): 33-46, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25869665

ABSTRACT

In addition to correcting mispaired nucleotides, DNA mismatch repair (MMR) proteins have been implicated in mutagenic, cell cycle, and apoptotic responses to agents that induce structurally aberrant nucleotide lesions. Here, we investigated the mechanistic basis for these responses by exposing cell lines with single or combined genetic defects in nucleotide excision repair (NER), postreplicative translesion synthesis (TLS), and MMR to low-dose ultraviolet light during S phase. Our data reveal that the MMR heterodimer Msh2/Msh6 mediates the excision of incorrect nucleotides that are incorporated by TLS opposite helix-distorting, noninstructive DNA photolesions. The resulting single-stranded DNA patches induce canonical Rpa-Atr-Chk1-mediated checkpoints and, in the next cell cycle, collapse to double-stranded DNA breaks that trigger apoptosis. In conclusion, a novel MMR-related DNA excision repair pathway controls TLS a posteriori, while initiating cellular responses to environmentally relevant densities of genotoxic lesions. These results may provide a rationale for the colorectal cancer tropism in Lynch syndrome, which is caused by inherited MMR gene defects.


Subject(s)
DNA Damage , DNA Mismatch Repair , Animals , Apoptosis , Cell Line , DNA-Binding Proteins/physiology , Embryonic Stem Cells/physiology , Epistasis, Genetic , Humans , Mice, 129 Strain , MutS Homolog 2 Protein/physiology , Mutagenesis
12.
DNA Repair (Amst) ; 29: 56-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25655219

ABSTRACT

Most spontaneous and DNA damage-induced nucleotide substitutions in eukaryotes depend on translesion synthesis polymerases Rev1 and Pol ζ, the latter consisting of the catalytic subunit Rev3 and the accessory protein Rev7. Here we review the regulation, and the biochemical and cellular functions, of Rev1/Pol ζ-dependent translesion synthesis. These are correlated with phenotypes of mouse models with defects in Rev1, Rev3 or Rev7. The data indicate that Rev1/Pol ζ-mediated translesion synthesis is important for adaptive immunity while playing paradoxical roles in oncogenesis. On the other hand, by enabling the replication of endogenously damaged templates, Rev1/Pol ζ -dependent translesion synthesis protects stem cells, thereby preventing features of ageing. In conclusion, Rev1/Pol ζ-dependent translesion synthesis at DNA helix-distorting nucleotide lesions orchestrates pleiotropic responses that determine organismal fitness and disease.


Subject(s)
DNA Repair/physiology , DNA Replication/physiology , DNA-Directed DNA Polymerase/metabolism , Genomic Instability , Mutagenesis , Animals , DNA/metabolism , Disease/genetics , Mad2 Proteins/metabolism , Mice , Nucleotidyltransferases/metabolism
13.
Nucleic Acids Res ; 43(1): 282-94, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25505145

ABSTRACT

Translesion synthesis (TLS) provides a highly conserved mechanism that enables DNA synthesis on a damaged template. TLS is performed by specialized DNA polymerases of which polymerase (Pol) κ is important for the cellular response to DNA damage induced by benzo[a]pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), ultraviolet (UV) light and the alkylating agent methyl methanesulfonate (MMS). As TLS polymerases are intrinsically error-prone, tight regulation of their activity is required. One level of control is provided by ubiquitination of the homotrimeric DNA clamp PCNA at lysine residue 164 (PCNA-Ub). We here show that Polκ can function independently of PCNA modification and that Polη can function as a backup during TLS of MMS-induced lesions. Compared to cell lines deficient for PCNA modification (Pcna(K164R)) or Polκ, double mutant cell lines display hypersensitivity to MMS but not to BPDE or UV-C. Double mutant cells also displayed delayed post-replicative TLS, accumulate higher levels of replication stress and delayed S-phase progression. Furthermore, we show that Polη and Polκ are redundant in the DNA damage bypass of MMS-induced DNA damage. Taken together, we provide evidence for PCNA-Ub-independent activation of Polκ and establish Polη as an important backup polymerase in the absence of Polκ in response to MMS-induced DNA damage.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/physiology , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Survival , Cells, Cultured , Checkpoint Kinase 1 , DNA Replication , DNA-Directed DNA Polymerase/genetics , Methyl Methanesulfonate/toxicity , Mice, Knockout , Mutation , Proliferating Cell Nuclear Antigen/genetics , Protein Kinases/metabolism , S Phase
14.
Nucleic Acids Res ; 42(17): 11071-82, 2014.
Article in English | MEDLINE | ID: mdl-25170086

ABSTRACT

Short-wave ultraviolet light induces both mildly helix-distorting cyclobutane pyrimidine dimers (CPDs) and severely distorting (6-4) pyrimidine pyrimidone photoproducts ((6-4)PPs). The only DNA polymerase (Pol) that is known to replicate efficiently across CPDs is Polη, a member of the Y family of translesion synthesis (TLS) DNA polymerases. Phenotypes of Polη deficiency are transient, suggesting redundancy with other DNA damage tolerance pathways. Here we performed a comprehensive analysis of the temporal requirements of Y-family Pols ι and κ as backups for Polη in (i) bypassing genomic CPD and (6-4)PP lesions in vivo, (ii) suppressing DNA damage signaling, (iii) maintaining cell cycle progression and (iv) promoting cell survival, by using mouse embryonic fibroblast lines with single and combined disruptions in these Pols. The contribution of Polι is restricted to TLS at a subset of the photolesions. Polκ plays a dominant role in rescuing stalled replication forks in Polη-deficient mouse embryonic fibroblasts, both at CPDs and (6-4)PPs. This dampens DNA damage signaling and cell cycle arrest, and results in increased survival. The role of relatively error-prone Pols ι and κ as backups for Polη contributes to the understanding of the mutator phenotype of xeroderma pigmentosum variant, a syndrome caused by Polη defects.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/physiology , Ultraviolet Rays/adverse effects , Animals , Cell Cycle , Cell Line , DNA Breaks, Double-Stranded , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , Genome , Mice , Pyrimidine Dimers/metabolism , DNA Polymerase iota
15.
Nucleic Acids Res ; 42(7): 4406-13, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464993

ABSTRACT

Rev3, the catalytic subunit of DNA polymerase ζ, is essential for translesion synthesis of cytotoxic DNA photolesions, whereas the Rev1 protein plays a noncatalytic role in translesion synthesis. Here, we reveal that mammalian Rev3(-/-) and Rev1(-/-) cell lines additionally display a nucleotide excision repair (NER) defect, specifically during S phase. This defect is correlated with the normal recruitment but protracted persistence at DNA damage sites of factors involved in an early stage of NER, while repair synthesis is affected. Remarkably, the NER defect becomes apparent only at 2 h post-irradiation indicating that Rev3 affects repair synthesis only indirectly, rather than performing an enzymatic role in NER. We provide evidence that the NER defect is caused by scarceness of Replication protein A (Rpa) available to NER, resulting from its sequestration at stalled replication forks. Also the induction of replicative stress using hydroxyurea precludes the accumulation of Rpa at photolesion sites, both in Rev3(-/-) and in wild-type cells. These data support a model in which the limited Rpa pool coordinates replicative stress and NER, resulting in increased cytotoxicity of ultraviolet light when replicative stress exceeds a threshold.


Subject(s)
DNA Repair , DNA Replication , Replication Protein A/metabolism , Animals , Cell Line , Cell Proliferation , DNA-Directed DNA Polymerase/genetics , Mice , Transcription, Genetic , Ultraviolet Rays/adverse effects
16.
Mutat Res ; 751-752: 8-14, 2013.
Article in English | MEDLINE | ID: mdl-24144844

ABSTRACT

Ultraviolet radiation is a highly mutagenic agent that damages the DNA by the formation of mutagenic photoproducts at dipyrimidine sites and by oxidative DNA damages via reactive oxygen species (ROS). ROS can also give rise to mutations via oxidation of dNTPs in the nucleotide pool, e.g. 8-oxo-dGTP and 2-OH-dATP and subsequent incorporation during DNA replication. Here we show that expression of human MutT homolog 1 (hMTH1) which sanitizes the nucleotide pool by dephosphorylating oxidized dNTPs, protects against mutagenesis induced by long wave UVA light and by UVB light but not by short wave UVC light. Mutational spectra analyses of UVA-induced mutations at the endogenous Thymidine kinase gene in human lymphoblastoid cells revealed that hMTH1 mainly protects cells from transitions at GC and AT base pairs.


Subject(s)
DNA Repair Enzymes/genetics , Mutation/radiation effects , Phosphoric Monoester Hydrolases/genetics , Ultraviolet Rays , Base Pairing/radiation effects , Cell Line , DNA Repair Enzymes/metabolism , Gene Knockdown Techniques , Humans , Mutagenesis/radiation effects , Mutation Rate , Nucleotides/metabolism , Phosphoric Monoester Hydrolases/metabolism , Thymidine Kinase/genetics
17.
Ultramicroscopy ; 134: 94-101, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23822882

ABSTRACT

The Stobbs factor problem, a major difference of absolute contrast between experimental and simulated high resolution transmission electron microscopy images has been reported frequently. In this respect, some modifications to the multislice simulation techniques were proposed to improve the correspondence to the experiment. The influence of different suggestions on the simulated contrast is investigated by numerical simulations. The agreement between experiment and simulations is then checked by comparison with high-resolution micrographs of crystalline gold. The experimental data is therefore compared to simulated intensities computed for the thickness and orientation of the specimen measured by refinements of diffraction patterns. The agreement of both intensity and contrast is investigated and the remaining contrast discrepancy is determined. The results show a good agreement for a small objective aperture, while for a larger aperture a difference of contrast by a factor of 1.2 can still be observed. Without any aperture, the deviation between experiment and simulations is largest.


Subject(s)
Electron Microscope Tomography/methods , Microscopy, Electron, Transmission/methods , Computer Simulation , Crystallization/methods
18.
DNA Repair (Amst) ; 11(6): 550-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22521143

ABSTRACT

Replicative polymerases (Pols) arrest at damaged DNA nucleotides, which induces ubiquitination of the DNA sliding clamp PCNA (PCNA-Ub) and DNA damage signaling. PCNA-Ub is associated with the recruitment or activation of translesion synthesis (TLS) DNA polymerases of the Y family that can bypass the lesions, thereby rescuing replication and preventing replication fork collapse and consequent formation of double-strand DNA breaks. Here, we have used gene-targeted mouse embryonic fibroblasts to perform a comprehensive study of the in vivo roles of PCNA-Ub and of the Y family TLS Pols η, ι, κ, Rev1 and the B family TLS Polζ in TLS and in the suppression of DNA damage signaling and genome instability after exposure to UV light. Our data indicate that TLS Pols ι and κ and the N-terminal BRCT domain of Rev1, that previously was implicated in the regulation of TLS, play minor roles in TLS of DNA photoproducts. PCNA-Ub is critical for an early TLS pathway that replicates both strongly helix-distorting (6-4) pyrimidine-pyrimidone ((6-4)PP) and mildly distorting cyclobutane pyrimidine dimer (CPD) photoproducts. The role of Polη is mainly restricted to early TLS of CPD photoproducts, whereas Rev1 and, in particular, Polζ are essential for the bypass of (6-4)PP photoproducts, both early and late after exposure. Thus, structurally distinct photoproducts at the mammalian genome are bypassed by different TLS Pols in temporally different, PCNA-Ub-dependent and independent fashions.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Repair , Genome/radiation effects , Signal Transduction/genetics , Ultraviolet Rays , Animals , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line , Cell Proliferation/radiation effects , DNA Replication/genetics , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genome/genetics , Histones/metabolism , Immunoblotting , Mammals/genetics , Mice , Mutation , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
19.
Toxicol Sci ; 127(1): 130-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22331492

ABSTRACT

DNA lesions, induced by genotoxic compounds, block the processive replication fork but can be bypassed by specialized translesion synthesis (TLS) DNA polymerases (Pols). TLS safeguards the completion of replication, albeit at the expense of nucleotide substitution mutations. We studied the in vivo role of individual TLS Pols in cellular responses to benzo[a]pyrene diolepoxide (BPDE), a polycyclic aromatic hydrocarbon, and 4-hydroxynonenal (4-HNE), a product of lipid peroxidation. To this aim, we used mouse embryonic fibroblasts with targeted disruptions in the TLS-associated Pols η, ι, κ, and Rev1 as well as in Rev3, the catalytic subunit of TLS Polζ. After exposure, cellular survival, replication fork progression, DNA damage responses (DDR), and the induction of micronuclei were investigated. The results demonstrate that Rev1, Rev3, and, to a lesser extent, Polη are involved in TLS and the prevention of DDR and of DNA breaks, in response to both agents. Conversely, Polκ and the N-terminal BRCT domain of Rev1 are specifically involved in TLS of BPDE-induced DNA damage. We furthermore describe a novel role of Polι in TLS of 4-HNE-induced DNA damage in vivo. We hypothesize that different sets of TLS polymerases act on structurally different genotoxic DNA lesions in vivo, thereby suppressing genomic instability associated with cancer. Our experimental approach may provide a significant contribution in delineating the molecular bases of the genotoxicity in vivo of different classes of DNA-damaging agents.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Aldehydes/toxicity , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Mutagens/toxicity , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Cytokinesis , DNA Adducts/drug effects , DNA-Directed DNA Polymerase/genetics , Fibroblasts/drug effects , Food Contamination , Mice , Mice, Knockout , Micronuclei, Chromosome-Defective/chemically induced , Micronucleus Tests/methods
20.
DNA Repair (Amst) ; 10(10): 1051-9, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21889916

ABSTRACT

The generation of high affinity antibodies in B cells critically depends on translesion synthesis (TLS) polymerases that introduce mutations into immunoglobulin genes during somatic hypermutation (SHM). The majority of mutations at A/T base pairs during SHM require ubiquitination of PCNA at lysine 164 (PCNA-Ub), which activates TLS polymerases. By comparing the mutation spectra in B cells of WT, TLS polymerase η (Polη)-deficient, PCNA(K164R)-mutant, and PCNA(K164R);Polη double-mutant mice, we now find that most PCNA-Ub-independent A/T mutagenesis during SHM is mediated by Polη. In addition, upon exposure to various DNA damaging agents, PCNA(K164R) mutant cells display strongly impaired recruitment of TLS polymerases, reduced daughter strand maturation and hypersensitivity. Interestingly, compared to the single mutants, PCNA(K164R);Polη double-mutant cells are dramatically delayed in S phase progression and far more prone to cell death following UV exposure. Taken together, these data support the existence of PCNA ubiquitination-dependent and -independent activation pathways of Polη during SHM and DNA damage tolerance.


Subject(s)
B-Lymphocytes/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Somatic Hypermutation, Immunoglobulin/genetics , Ubiquitination , Animals , B-Lymphocytes/cytology , Enzyme Activation , Lysine/genetics , Mice , Mice, Inbred C57BL , Mutagenesis , Mutation , Proliferating Cell Nuclear Antigen/genetics , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...