Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(10): e0258269, 2021.
Article in English | MEDLINE | ID: mdl-34614038

ABSTRACT

Ionizing radiation (IR) is widely used in cancer therapy and accidental or environmental exposure is a major concern. However, little is known about the genome-wide effects IR exerts on germ cells and the relative contribution of DNA repair pathways for mending IR-induced lesions. Here, using C. elegans as a model system and using primary sequencing data from our recent high-level overview of the mutagenic consequences of 11 genotoxic agents, we investigate in detail the genome-wide mutagenic consequences of exposing wild-type and 43 DNA repair and damage response defective C. elegans strains to a Caesium (Cs-137) source, emitting γ-rays. Cs-137 radiation induced single nucleotide variants (SNVs) at a rate of ~1 base substitution per 3 Gy, affecting all nucleotides equally. In nucleotide excision repair mutants, this frequency increased 2-fold concurrently with increased dinucleotide substitutions. As observed for DNA damage induced by bulky DNA adducts, small deletions were increased in translesion polymerase mutants, while base changes decreased. Structural variants (SVs) were augmented with dose, but did not arise with significantly higher frequency in any DNA repair mutants tested. Moreover, 6% of all mutations occurred in clusters, but clustering was not significantly altered in any DNA repair mutant background. Our data is relevant for better understanding how DNA repair pathways modulate IR-induced lesions.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Genome, Helminth , Radiation, Ionizing , Animals , Caenorhabditis elegans/drug effects , Cisplatin/pharmacology , DNA Repair/drug effects , Humans , Mutation/genetics , Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Ultraviolet Rays
2.
PLoS One ; 16(4): e0250291, 2021.
Article in English | MEDLINE | ID: mdl-33905417

ABSTRACT

Maintaining genome integrity is particularly important in germ cells to ensure faithful transmission of genetic information across generations. Here we systematically describe germ cell mutagenesis in wild-type and 61 DNA repair mutants cultivated over multiple generations. ~44% of the DNA repair mutants analysed showed a >2-fold increased mutagenesis with a broad spectrum of mutational outcomes. Nucleotide excision repair deficiency led to higher base substitution rates, whereas polh-1(Polη) and rev-3(Polζ) translesion synthesis polymerase mutants resulted in 50-400 bp deletions. Signatures associated with defective homologous recombination fall into two classes: 1) brc-1/BRCA1 and rad-51/RAD51 paralog mutants showed increased mutations across all mutation classes, 2) mus-81/MUS81 and slx-1/SLX1 nuclease, and him-6/BLM, helq-1/HELQ or rtel-1/RTEL1 helicase mutants primarily accumulated structural variants. Repetitive and G-quadruplex sequence-containing loci were more frequently mutated in specific DNA repair backgrounds. Tandem duplications embedded in inverted repeats were observed in helq-1 helicase mutants, and a unique pattern of 'translocations' involving homeologous sequences occurred in rip-1 recombination mutants. atm-1/ATM checkpoint mutants harboured structural variants specifically enriched in subtelomeric regions. Interestingly, locally clustered mutagenesis was only observed for combined brc-1 and cep-1/p53 deficiency. Our study provides a global view of how different DNA repair pathways contribute to prevent germ cell mutagenesis.


Subject(s)
Caenorhabditis elegans/genetics , DNA Repair , DNA, Helminth/genetics , Gene Expression Regulation , Genome, Helminth , Germ Cells/metabolism , Mutation , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Proliferation , Chromosome Mapping , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , DNA, Helminth/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Germ Cells/cytology , Isoenzymes/genetics , Isoenzymes/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
3.
DNA Repair (Amst) ; 95: 102957, 2020 11.
Article in English | MEDLINE | ID: mdl-32980770

ABSTRACT

Genome integrity is constantly challenged by exogenous and endogenous insults, and mutations are associated with inherited disease and cancer. Here we summarize recent studies that utilized C. elegans whole genome next generation sequencing to experimentally determine mutational signatures associated with mutagen exposure, DNA repair deficiency or a combination of both and discuss the implications of these results for the understanding of cancer genome evolution. The experimental analysis of wild-type and DNA repair deficient nematodes propagated under unchallenged conditions over many generations revealed increased mutagenesis in approximately half of all DNA repair deficient strains, its rate, except for DNA mismatch repair, only being moderately increased. The exposure of wild-type and DNA repair defective strains to selected genotoxins, including UV-B and ionizing radiation, alkylating compounds, aristolochic acid, aflatoxin-B1, and cisplatin enabled the systematic analysis of the relative contributions of redundant repair modalities that mend DNA damage. Combining genotoxin exposure with DNA repair deficiency can manifest as altered mutation rates and/or as a change in mutational profiles, and reveals how different DNA alterations induced by one genotoxin are repaired by separate DNA repair pathways, often in a highly redundant way. Cancer genomes provide a snapshot of all mutational events that happened prior to cancer detection and sequencing, necessitating computational models to deduce mutational signatures using mathematical best fit approaches. While computationally deducing signatures from cancer genomes has been tremendously successful in associating some signatures to known mutagenic causes, many inferred signatures lack a clear link to a known mutagenic process. Moreover, analytical signatures might not reflect any distinct mutagenic processes. Nonetheless, combined effects of mutagen exposure and DNA damage-repair deficiency are also present in cancer genomes, but cannot be as easily detected owing to the unknown histories of genotoxic exposures and because biallelic in contrast to monoallelic DNA repair deficiency is rare. The impact of damage-repair interactions also manifests through selective pressure for DNA repair gene inactivation during cancer evolution. Using these considerations, we discuss a theoretical framework that explains why minute mutagenic changes, possibly too small to manifest as change in a signature, can have major effects in oncogenesis. Overall, the experimental analysis of mutational processes underscores that the interpretation of mutational signatures requires considering both the primary DNA lesion and repair status and imply that mutational signatures derived from cancer genomes may be more variable than currently anticipated.


Subject(s)
Caenorhabditis elegans/genetics , Genomics , Mutation , Neoplasms/genetics , Animals , Humans
4.
Nat Commun ; 11(1): 2169, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358516

ABSTRACT

Cells possess an armamentarium of DNA repair pathways to counter DNA damage and prevent mutation. Here we use C. elegans whole genome sequencing to systematically quantify the contributions of these factors to mutational signatures. We analyse 2,717 genomes from wild-type and 53 DNA repair defective backgrounds, exposed to 11 genotoxins, including UV-B and ionizing radiation, alkylating compounds, aristolochic acid, aflatoxin B1, and cisplatin. Combined genotoxic exposure and DNA repair deficiency alters mutation rates or signatures in 41% of experiments, revealing how different DNA alterations induced by the same genotoxin are mended by separate repair pathways. Error-prone translesion synthesis causes the majority of genotoxin-induced base substitutions, but averts larger deletions. Nucleotide excision repair prevents up to 99% of point mutations, almost uniformly across the mutation spectrum. Our data show that mutational signatures are joint products of DNA damage and repair and suggest that multiple factors underlie signatures observed in cancer genomes.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , Animals , Caenorhabditis elegans/genetics , DNA Damage/genetics , DNA Repair/genetics , Genomics/methods , Humans , Mutation/genetics , Point Mutation/genetics
5.
Genome Res ; 28(5): 666-675, 2018 05.
Article in English | MEDLINE | ID: mdl-29636374

ABSTRACT

Throughout their lifetime, cells are subject to extrinsic and intrinsic mutational processes leaving behind characteristic signatures in the genome. DNA mismatch repair (MMR) deficiency leads to hypermutation and is found in different cancer types. Although it is possible to associate mutational signatures extracted from human cancers with possible mutational processes, the exact causation is often unknown. Here, we use C. elegans genome sequencing of pms-2 and mlh-1 knockouts to reveal the mutational patterns linked to C. elegans MMR deficiency and their dependency on endogenous replication errors and errors caused by deletion of the polymerase ε subunit pole-4 Signature extraction from 215 human colorectal and 289 gastric adenocarcinomas revealed three MMR-associated signatures, one of which closely resembles the C. elegans MMR spectrum and strongly discriminates microsatellite stable and unstable tumors (AUC = 98%). A characteristic difference between human and C. elegans MMR deficiency is the lack of elevated levels of NCG > NTG mutations in C. elegans, likely caused by the absence of cytosine (CpG) methylation in worms. The other two human MMR signatures may reflect the interaction between MMR deficiency and other mutagenic processes, but their exact cause remains unknown. In summary, combining information from genetically defined models and cancer samples allows for better aligning mutational signatures to causal mutagenic processes.


Subject(s)
Adenocarcinoma/genetics , Caenorhabditis elegans/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair , Mutation , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Animals , Base Sequence , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Mutational Analysis/methods , DNA Polymerase II/deficiency , DNA Polymerase II/genetics , Humans , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/genetics , Stomach Neoplasms/metabolism
6.
Nat Commun ; 9(1): 728, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463814

ABSTRACT

Faithful chromosome segregation and genome maintenance requires the removal of all DNA bridges that physically link chromosomes before cells divide. Using C. elegans embryos we show that the LEM-3/Ankle1 nuclease defines a previously undescribed genome integrity mechanism by processing DNA bridges right before cells divide. LEM-3 acts at the midbody, the structure where abscission occurs at the end of cytokinesis. LEM-3 localization depends on factors needed for midbody assembly, and LEM-3 accumulation is increased and prolonged when chromatin bridges are trapped at the cleavage plane. LEM-3 locally processes chromatin bridges that arise from incomplete DNA replication, unresolved recombination intermediates, or the perturbance of chromosome structure. Proper LEM-3 midbody localization and function is regulated by AIR-2/Aurora B kinase. Strikingly, LEM-3 acts cooperatively with the BRC-1/BRCA1 homologous recombination factor to promote genome integrity. These findings provide a molecular basis for the suspected role of the LEM-3 orthologue Ankle1 in human breast cancer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Chromatin/metabolism , Endodeoxyribonucleases/metabolism , Mitosis , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Chromatin/genetics , Cytokinesis , DNA/genetics , DNA/metabolism , DNA Replication , Endodeoxyribonucleases/genetics
7.
G3 (Bethesda) ; 7(12): 3875-3885, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29046436

ABSTRACT

Relatively little is known about the cross-talk between the spindle assembly checkpoint and the DNA damage response, especially in multicellular organisms. We performed a Caenorhabditis elegans forward genetic screen to uncover new genes involved in the repair of DNA damage induced by ionizing radiation. We isolated a mutation, gt2000, which confers hypersensitivity to ionizing radiation and showed that gt2000 introduces a premature stop in bub-3 BUB-3 is a key component of the spindle assembly checkpoint. We provide evidence that BUB-3 acts during development and in the germline; irradiated bub-3(gt2000) larvae are developmentally retarded and form abnormal vulvae. Moreover, bub-3(gt2000) embryos sired from irradiated worms show increased levels of lethality. Both bub-3 and san-1 (the C. elegans homolog of MAD3) deletion alleles confer hypersensitivity to ionizing radiation, consistent with the notion that the spindle assembly checkpoint pathway is required for the DNA damage response. bub-3(gt2000) is moderately sensitive to the cross-linking drug cisplatin but not to ultraviolet light or methyl methanesulfonate. This is consistent with a role in dealing with DNA double-strand breaks and not with base damage. Double mutant analysis revealed that bub-3 does not act within any of the three major pathways involved in the repair of double-strand breaks. Finally, the cdc-20 gain-of-function mutant cdc-20/fzy-1(av15), which is refractory to the cell cycle delay conferred by the spindle checkpoint, showed phenotypes similar to bub-3 and san-1 mutants. We speculate that BUB-3 is involved in the DNA damage response through regulation of cell cycle timing.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Cdc20 Proteins/genetics , Cell Cycle Proteins/genetics , M Phase Cell Cycle Checkpoints/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/radiation effects , Cell Cycle Proteins/physiology , Cell Cycle Proteins/radiation effects , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Damage/radiation effects , Gene Expression Regulation/radiation effects , Genomic Instability/genetics , Genomic Instability/radiation effects , M Phase Cell Cycle Checkpoints/radiation effects , Mutation , Radiation, Ionizing
8.
PLoS Genet ; 12(3): e1005872, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27010650

ABSTRACT

Meiotic recombination is essential for the repair of programmed double strand breaks (DSBs) to generate crossovers (COs) during meiosis. The efficient processing of meiotic recombination intermediates not only needs various resolvases but also requires proper meiotic chromosome structure. The Smc5/6 complex belongs to the structural maintenance of chromosome (SMC) family and is closely related to cohesin and condensin. Although the Smc5/6 complex has been implicated in the processing of recombination intermediates during meiosis, it is not known how Smc5/6 controls meiotic DSB repair. Here, using Caenorhabditis elegans we show that the SMC-5/6 complex acts synergistically with HIM-6, an ortholog of the human Bloom syndrome helicase (BLM) during meiotic recombination. The concerted action of the SMC-5/6 complex and HIM-6 is important for processing recombination intermediates, CO regulation and bivalent maturation. Careful examination of meiotic chromosomal morphology reveals an accumulation of inter-chromosomal bridges in smc-5; him-6 double mutants, leading to compromised chromosome segregation during meiotic cell divisions. Interestingly, we found that the lethality of smc-5; him-6 can be rescued by loss of the conserved BRCA1 ortholog BRC-1. Furthermore, the combined deletion of smc-5 and him-6 leads to an irregular distribution of condensin and to chromosome decondensation defects reminiscent of condensin depletion. Lethality conferred by condensin depletion can also be rescued by BRC-1 depletion. Our results suggest that SMC-5/6 and HIM-6 can synergistically regulate recombination intermediate metabolism and suppress ectopic recombination by controlling chromosome architecture during meiosis.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Meiosis/genetics , Recombination, Genetic , Animals , Caenorhabditis elegans/genetics , Chromosomes/genetics , Humans , Multiprotein Complexes/genetics
9.
Nat Genet ; 47(3): 257-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25642631

ABSTRACT

DNA replication-associated mutations are repaired by two components: polymerase proofreading and mismatch repair. The mutation consequences of disruption to both repair components in humans are not well studied. We sequenced cancer genomes from children with inherited biallelic mismatch repair deficiency (bMMRD). High-grade bMMRD brain tumors exhibited massive numbers of substitution mutations (>250/Mb), which was greater than all childhood and most cancers (>7,000 analyzed). All ultra-hypermutated bMMRD cancers acquired early somatic driver mutations in DNA polymerase ɛ or δ. The ensuing mutation signatures and numbers are unique and diagnostic of childhood germ-line bMMRD (P < 10(-13)). Sequential tumor biopsy analysis revealed that bMMRD/polymerase-mutant cancers rapidly amass an excess of simultaneous mutations (∼600 mutations/cell division), reaching but not exceeding ∼20,000 exonic mutations in <6 months. This implies a threshold compatible with cancer-cell survival. We suggest a new mechanism of cancer progression in which mutations develop in a rapid burst after ablation of replication repair.


Subject(s)
Base Pair Mismatch , Brain Neoplasms/genetics , DNA Mismatch Repair , DNA Replication/genetics , DNA Repair , DNA-Directed DNA Polymerase/genetics , Exons , Germ-Line Mutation , Humans , Microsatellite Instability
10.
Exp Cell Res ; 329(1): 35-41, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25131498

ABSTRACT

Genetic information is under constant attack from endogenous and exogenous sources, and the use of model organisms has provided important frameworks to understand how genome stability is maintained and how various DNA lesions are repaired. The advance of high throughput next generation sequencing (NGS) provides new inroads for investigating mechanisms needed for genome maintenance. These emerging studies, which aim to link genetic toxicology and mechanistic analyses of DNA repair processes in vivo, rely on defining mutational signatures caused by faulty replication, endogenous DNA damaging metabolites, or exogenously applied genotoxins; the analysis of their nature, their frequency and distribution. In contrast to classical studies, where DNA repair deficiency is assessed by reduced cellular survival, the localization of DNA repair factors and their interdependence as well as limited analysis of single locus reporter assays, NGS based approaches reveal the direct, quantal imprint of mutagenesis genome-wide, at the DNA sequence level. As we will show, such investigations require the analysis of DNA derived from single genotoxin treated cells, or DNA from cell populations regularly passaged through single cell bottlenecks when naturally occurring mutation accumulation is investigated. We will argue that the life cycle of the nematode Caenorhabditis elegans, its genetic malleability combined with whole genome sequencing provides an exciting model system to conduct such analysis.


Subject(s)
Caenorhabditis elegans Proteins/genetics , DNA Damage/genetics , DNA Repair/genetics , Genome , High-Throughput Nucleotide Sequencing , Animals , Caenorhabditis elegans/genetics
11.
Genome Res ; 24(10): 1624-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25030888

ABSTRACT

Mutation is associated with developmental and hereditary disorders, aging, and cancer. While we understand some mutational processes operative in human disease, most remain mysterious. We used Caenorhabditis elegans whole-genome sequencing to model mutational signatures, analyzing 183 worm populations across 17 DNA repair-deficient backgrounds propagated for 20 generations or exposed to carcinogens. The baseline mutation rate in C. elegans was approximately one per genome per generation, not overtly altered across several DNA repair deficiencies over 20 generations. Telomere erosion led to complex chromosomal rearrangements initiated by breakage-fusion-bridge cycles and completed by simultaneously acquired, localized clusters of breakpoints. Aflatoxin B1 induced substitutions of guanines in a GpC context, as observed in aflatoxin-induced liver cancers. Mutational burden increased with impaired nucleotide excision repair. Cisplatin and mechlorethamine, DNA crosslinking agents, caused dose- and genotype-dependent signatures among indels, substitutions, and rearrangements. Strikingly, both agents induced clustered rearrangements resembling "chromoanasynthesis," a replication-based mutational signature seen in constitutional genomic disorders, suggesting that interstrand crosslinks may play a pathogenic role in such events. Cisplatin mutagenicity was most pronounced in xpf-1 mutants, suggesting that this gene critically protects cells against platinum chemotherapy. Thus, experimental model systems combined with genome sequencing can recapture and mechanistically explain mutational signatures associated with human disease.


Subject(s)
Caenorhabditis elegans/genetics , Carcinogens/pharmacology , DNA Repair , Mutation , Sequence Analysis, DNA/methods , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/genetics , DNA Helicases/genetics , Genome , Models, Animal
12.
PLoS Genet ; 9(7): e1003591, 2013.
Article in English | MEDLINE | ID: mdl-23901331

ABSTRACT

Holliday junctions (HJs) are cruciform DNA structures that are created during recombination events. It is a matter of considerable importance to determine the resolvase(s) that promote resolution of these structures. We previously reported that C. elegans GEN-1 is a symmetrically cleaving HJ resolving enzyme required for recombinational repair, but we could not find an overt role in meiotic recombination. Here we identify C. elegans proteins involved in resolving meiotic HJs. We found no evidence for a redundant meiotic function of GEN-1. In contrast, we discovered two redundant HJ resolution pathways likely coordinated by the SLX-4 scaffold protein and also involving the HIM-6/BLM helicase. SLX-4 associates with the SLX-1, MUS-81 and XPF-1 nucleases and has been implicated in meiotic recombination in C. elegans. We found that C. elegans [mus-81; xpf-1], [slx-1; xpf-1], [mus-81; him-6] and [slx-1; him-6] double mutants showed a similar reduction in survival rates as slx-4. Analysis of meiotic diakinesis chromosomes revealed a distinct phenotype in these double mutants. Instead of wild-type bivalent chromosomes, pairs of "univalents" linked by chromatin bridges occur. These linkages depend on the conserved meiosis-specific transesterase SPO-11 and can be restored by ionizing radiation, suggesting that they represent unresolved meiotic HJs. This suggests the existence of two major resolvase activities, one provided by XPF-1 and HIM-6, the other by SLX-1 and MUS-81. In all double mutants crossover (CO) recombination is reduced but not abolished, indicative of further redundancy in meiotic HJ resolution. Real time imaging revealed extensive chromatin bridges during the first meiotic division that appear to be eventually resolved in meiosis II, suggesting back-up resolution activities acting at or after anaphase I. We also show that in HJ resolution mutants, the restructuring of chromosome arms distal and proximal to the CO still occurs, suggesting that CO initiation but not resolution is likely to be required for this process.


Subject(s)
Caenorhabditis elegans Proteins/genetics , DNA Helicases/genetics , DNA, Cruciform/genetics , DNA-Binding Proteins/genetics , Deoxyribonucleases/genetics , Endonucleases/genetics , Meiosis/genetics , Animals , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromosome Segregation/genetics , Crossing Over, Genetic , DNA Breaks, Double-Stranded , Humans , Meiotic Prophase I/genetics , Mice , Mutation , Recombination, Genetic
13.
EMBO J ; 28(22): 3549-63, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19779462

ABSTRACT

The telomerase reverse transcriptase adds de novo DNA repeats to chromosome termini. Here we define Caenorhabditis elegans MRT-1 as a novel factor required for telomerase-mediated telomere replication and the DNA-damage response. MRT-1 is composed of an N-terminal domain homologous to the second OB-fold of POT1 telomere-binding proteins and a C-terminal SNM1 family nuclease domain, which confer single-strand DNA-binding and processive 3'-to-5' exonuclease activity, respectively. Furthermore, telomerase activity in vivo depends on a functional MRT-1 OB-fold. We show that MRT-1 acts in the same telomere replication pathway as telomerase and the 9-1-1 DNA-damage response complex. MRT-1 is dispensable for DNA double-strand break repair, but functions with the 9-1-1 complex to promote DNA interstrand cross-link (ICL) repair. Our data reveal MRT-1 as a dual-domain protein required for telomerase function and ICL repair, which raises the possibility that telomeres and ICL lesions may share a common feature that plays a critical role in de novo telomere repeat addition.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , DNA Repair/genetics , Deoxyribonucleases/physiology , Telomerase/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Damage/physiology , DNA, Single-Stranded/metabolism , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Enzyme Activation/genetics , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid , Telomere/metabolism
14.
Genetics ; 180(2): 741-54, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18780750

ABSTRACT

Critically shortened telomeres can be subjected to DNA repair events that generate end-to-end chromosome fusions. The resulting dicentric chromosomes can enter breakage-fusion-bridge cycles, thereby impeding elucidation of the structures of the initial fusion events and a mechanistic understanding of their genesis. Current models for the molecular basis of fusion of critically shortened, uncapped telomeres rely on PCR assays that typically capture fusion breakpoints created by direct ligation of chromosome ends. Here we use independent approaches that rely on distinctive features of Caenorhabditis elegans to study the frequency of direct end-to-end chromosome fusion in telomerase mutants: (1) holocentric chromosomes that allow for genetic isolation of stable end-to-end fusions and (2) unique subtelomeric sequences that allow for thorough PCR analysis of samples of genomic DNA harboring multiple end-to-end fusions. Surprisingly, only a minority of end-to-end fusion events resulted from direct end joining with no additional genome rearrangements. We also demonstrate that deficiency for the C. elegans Ku DNA repair heterodimer does not affect telomere length or cause synthetic effects in the absence of telomerase.


Subject(s)
Caenorhabditis elegans/genetics , Telomere/metabolism , Animals , Caenorhabditis elegans/metabolism , Chromosomes/metabolism , DNA Ligases/metabolism , DNA Repair , Models, Genetic , Mutation , Telomerase/genetics , Telomerase/metabolism
15.
Am J Dent ; 20(1): 59-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17380810

ABSTRACT

PURPOSE: To design an artificial mouth in order to evaluate if a new diagnostic tool (Clinpro Cario Diagnosis) can be used for early detection of secondary caries at resin composite margins in vitro. METHODS: 32 intact human third molars received standardized Class-V resin composite restorations (Tetric Ceram bonded with Syntac SC). After storage for 4 weeks at 37 degrees C, teeth were subjected to 5,000 or 10,000 thermocycles (+/- 5 degrees C and +/- 55 degrees C) and polysiloxane impressions were taken. Streptococcus mutans 10449 (SM) was used in a nutrition medium to initiate a secondary caries process. Daily, the teeth were incubated for 2 x 2.5 hours in SM containing nutrition medium followed by 2 x 9.5 hours incubation in artificial saliva. Teeth were investigated after total incubation periods of 4, 6, and 8 weeks. After the different incubation protocols, the restoration margins were evaluated for infection and secondary caries processes in using Clinpro Cario Diagnosis which measures site-specifically the lactic acid production of SM in response to a sucrose challenge. The color signal was read 5 minutes after removal of the diagnostic impression. After thermocycling and biological load cycling, precision polysiloxane impressions were taken and replicas were investigated under a light microscope for gap widths at enamel and dentin margins. Demineralization was evaluated by fluorescence microscopy in using a special FITC filter. The demineralization depths at the cavity margin were calculated with Xpert for Windows using a pixel distance of 5 microm. RESULTS: After the different thermocycling protocols, no differences in gap widths and demineralization depths were found (P > 0.05). After SM incubation, gap widths and demineralization depths were significantly dependent on SM incubation time and previous number of thermocycles (P < 0.05). Lactic acid formations of SM were detectable by Clinpro Cario Diagnosis at dentin cavosurface margins formed after 6 weeks of incubation with SM (P < 0.05).


Subject(s)
Composite Resins/chemistry , Dental Caries Susceptibility , Dental Restoration, Permanent , Dental Caries/microbiology , Dental Caries/pathology , Dental Enamel/ultrastructure , Dental Impression Materials/chemistry , Dental Restoration, Permanent/classification , Dentin/ultrastructure , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Lactic Acid/analysis , Recurrence , Resin Cements/chemistry , Risk Assessment , Saliva, Artificial/chemistry , Siloxanes/chemistry , Streptococcus mutans/isolation & purification , Streptococcus mutans/metabolism , Surface Properties , Temperature , Time Factors , Tooth Demineralization/pathology
16.
Curr Biol ; 16(7): R249-51, 2006 Apr 04.
Article in English | MEDLINE | ID: mdl-16581501

ABSTRACT

Faithful recombination and chromosome segregation in meiosis require regulated steps of homolog recognition and association which are monitored by meiotic checkpoints. A recent study in the nematode Caenorhabditis elegans has identified a checkpoint mechanism that monitors chromosome pairing during meiosis.


Subject(s)
Caenorhabditis elegans/genetics , Chromosome Pairing/physiology , Meiosis/genetics , Animals , Apoptosis/physiology , Caenorhabditis elegans/cytology , DNA Damage , Female , Germ Cells/cytology , Germ Cells/physiology , Male , Mutation , Pachytene Stage/physiology , Recombination, Genetic
17.
PLoS Genet ; 2(2): e18, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16477310

ABSTRACT

Mutants of trt-1, the Caenorhabditis elegans telomerase reverse transcriptase, reproduce normally for several generations but eventually become sterile as a consequence of telomere erosion and end-to-end chromosome fusions. Telomere erosion and uncapping do not cause an increase in apoptosis in the germlines of trt-1 mutants. Instead, late-generation trt-1 mutants display chromosome segregation defects that are likely to be the direct cause of sterility. trt-1 functions in the same telomere replication pathway as mrt-2, a component of the Rad9/Rad1/Hus1 (9-1-1) proliferating cell nuclear antigen-like sliding clamp. Thus, the 9-1-1 complex may be required for telomerase to act at chromosome ends in C. elegans. Although telomere erosion limits replicative life span in human somatic cells, neither trt-1 nor telomere shortening affects postmitotic aging in C. elegans. These findings illustrate effects of telomere dysfunction in C. elegans mutants lacking the catalytic subunit of telomerase, trt-1.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Telomerase/genetics , Telomere/ultrastructure , Animals , Apoptosis , Caenorhabditis elegans , Caenorhabditis elegans Proteins/physiology , Catalysis , Catalytic Domain , DNA Replication , Genes, Helminth , Germ-Line Mutation , Mitosis
18.
J Biol Chem ; 279(1): 86-94, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14551211

ABSTRACT

The Saccharomyces cerevisiae Ku heterodimer comprising Yku70p and Yku80p is involved in telomere maintenance and DNA repair by the pathway of non-homologous end joining. It is also a key regulator of transcriptional silencing of genes placed in close proximity to telomeres. Here, we describe the identification of separation-of-function mutants of Yku80p that exhibit defects in silencing but not DNA repair and show that these mutations map to an evolutionarily conserved domain within Yku80p. Furthermore, we reveal that Yku80p interacts with the silent information regulator protein Sir4p and that this interaction is mediated by the N-terminal 200 amino acid residues of Sir4p. Notably, this interaction also requires the region of Yku80p that contains the sites of the silencing defective mutations. Finally, we show that these mutations impair the Yku80p-Sir4p interaction and recruitment of Sir3p to telomeric regions in vivo. Taken together with other data, these findings indicate that the Yku80p-Sir4p interaction plays a vital role in the assembly of telomeric heterochromatin.


Subject(s)
Antigens, Nuclear/genetics , DNA Helicases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Silencing , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Telomere/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Genotype , Humans , Ku Autoantigen , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics
19.
J Neuroimmunol ; 131(1-2): 21-30, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12458033

ABSTRACT

Using Cyclosporin A (CsA) as an unconditioned stimulus has previously demonstrated that behaviorally conditioned inhibition of splenocyte proliferation and cytokine production is mediated via the splenic nerve. Therefore, we currently examined the adrenergic modulation of conditioned suppression of splenocyte function. Chemical sympathectomy via 6-OHDA completely blocked the conditioned suppression of splenocyte proliferation to mitogens and cytokine (IL-2, IFN-gamma) production. Furthermore, administration of beta-adrenoceptor antagonist propranolol abrogated the conditioned effect on splenocyte proliferation. Supporting the position that conditioning is beta-adrenergic-dependent, addition of beta-adrenoceptor agonist, but not alpha-adrenoceptor agonists, to splenocytes in vitro mimicked the conditioned suppression of splenocyte functions, with these effects blocked by propranolol. Therefore, these data indicate that behavioral conditioning of splenocyte function in the rat is regulated by the sympathetic nervous system, predominantly via beta-adrenergic mechanisms.


Subject(s)
Neuroimmunomodulation , Norepinephrine/physiology , Receptors, Adrenergic, beta/physiology , Spleen/immunology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Behavior, Animal , Cell Division , Cells, Cultured , Immunosuppression Therapy , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Male , Oxidopamine/pharmacology , Propranolol/pharmacology , RNA, Messenger/biosynthesis , Rats , Spleen/cytology , Sympathectomy, Chemical , Sympatholytics/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...