Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
G3 (Bethesda) ; 14(5)2024 05 07.
Article in English | MEDLINE | ID: mdl-38478595

ABSTRACT

DDX11/Chl1R is a conserved DNA helicase with roles in genome maintenance, DNA replication, and chromatid cohesion. Loss of DDX11 in humans leads to the rare cohesinopathy Warsaw breakage syndrome. DDX11 has also been implicated in human cancer where it has been proposed to have an oncogenic role and possibly to constitute a therapeutic target. Given the multiple roles of DDX11 in genome stability and its potential as an anticancer target, we set out to define a complete genetic interaction profile of DDX11 loss in human cell lines. Screening the human genome with clustered regularly interspaced short palindromic repeats (CRISPR) guide RNA drop out screens in DDX11-wildtype (WT) or DDX11-deficient cells revealed a strong enrichment of genes with functions related to sister chromatid cohesion. We confirm synthetic lethal relationships between DDX11 and the tumor suppressor cohesin subunit STAG2, which is frequently mutated in several cancer types and the kinase HASPIN. This screen highlights the importance of cohesion in cells lacking DDX11 and suggests DDX11 may be a therapeutic target for tumors with mutations in STAG2.


Subject(s)
Cell Cycle Proteins , Chromatids , DEAD-box RNA Helicases , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatids/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cohesins , Epistasis, Genetic , DNA Helicases/genetics , Cell Line
2.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33782138

ABSTRACT

Genetic screens can identify synthetic lethal (SL) interactions and uncover potential anticancer therapeutic targets. However, most SL screens have utilized knockout or knockdown approaches that do not accurately mimic chemical inhibition of a target protein. Here, we test whether missense mutations can be utilized as a model for a type of protein inhibition that creates a dominant gain-of-function cytotoxicity. We expressed missense mutations in the FEN1 endonuclease and the replication-associated helicase, CHL1, that inhibited enzymatic activity but retained substrate binding, and found that these mutations elicited a dominant SL phenotype consistent with the generation of cytotoxic protein-DNA or protein-protein intermediates. Genetic screens with nuclease-defective hFEN1 and helicase-deficient yCHL1 captured dominant SL interactions, in which ectopic expression of the mutant form, in the presence of the wild-type form, caused SL in specific mutant backgrounds. Expression of nuclease-defective hFEN1 in yeast elicited DNA binding-dependent dominant SL with homologous recombination mutants. In contrast, dominant SL interactions with helicase-deficient yCHL1 were observed in spindle-associated, Ctf18-alternative replication factor C (Ctf18-RFC) clamp loader complex, and cohesin mutant backgrounds. These results highlight the different mechanisms underlying SL interactions that occur in the presence of an inhibited form of the target protein and point to the utility of modeling trapping mutations in pursuit of more clinically relevant SL interactions.


Subject(s)
DNA/metabolism , Flap Endonucleases/metabolism , Mutation, Missense , Synthetic Lethal Mutations , Antineoplastic Agents/toxicity , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , Drug Development/methods , Flap Endonucleases/genetics , Genetic Techniques , Humans , Protein Binding , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
3.
Genetics ; 215(3): 609-621, 2020 07.
Article in English | MEDLINE | ID: mdl-32414869

ABSTRACT

New anticancer therapeutics require extensive in vivo characterization to identify endogenous and exogenous factors affecting efficacy, to measure toxicity and mutagenicity, and to determine genotypes that result in therapeutic sensitivity or resistance. We used Caenorhabditis elegans as a platform with which to characterize properties of the anticancer therapeutic CX-5461. To understand the processes that respond to CX-5461-induced damage, we generated pharmacogenetic profiles for a panel of C. elegans DNA replication and repair mutants with common DNA-damaging agents for comparison with the profile of CX-5461. We found that multiple repair pathways, including homology-directed repair, microhomology-mediated end joining, nucleotide excision repair, and translesion synthesis, were needed for CX-5461 tolerance. To determine the frequency and spectrum of CX-5461-induced mutations, we used a genetic balancer to capture CX-5461-induced mutations. We found that CX-5461 is mutagenic, resulting in both large copy number variations and a high frequency of single-nucleotide variations (SNVs), which are consistent with the pharmacogenetic profile for CX-5461. Whole-genome sequencing of CX-5461-exposed animals found that CX-5461-induced SNVs exhibited a distinct mutational signature. We also phenocopied the CX-5461 photoreactivity observed in clinical trials and demonstrated that CX-5461 generates reactive oxygen species when exposed to UVA radiation. Together, the data from C. elegans demonstrate that CX-5461 is a multimodal DNA-damaging anticancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Benzothiazoles/toxicity , Caenorhabditis elegans/genetics , Carcinogenicity Tests/methods , Genome-Wide Association Study/methods , Mutagens/toxicity , Naphthyridines/toxicity , Pharmacogenomic Variants , Animals , Caenorhabditis elegans/drug effects , DNA Repair , Drug Resistance, Neoplasm , Genome, Helminth , Mutation , Polymorphism, Single Nucleotide
4.
Genetics ; 214(3): 735-747, 2020 03.
Article in English | MEDLINE | ID: mdl-31937519

ABSTRACT

Cross-species complementation can be used to generate humanized yeast, which is a valuable resource with which to model and study human biology. Humanized yeast can be used as an in vivo platform to screen for chemical inhibition of human protein drug targets. To this end, we report the systematic complementation of nonessential yeast genes implicated in chromosome instability (CIN) with their human homologs. We identified 20 human-yeast complementation pairs that are replaceable in 44 assays that test rescue of chemical sensitivity and/or CIN defects. We selected a human-yeast pair (hFEN1/yRAD27), which is frequently overexpressed in cancer and is an anticancer therapeutic target, to perform in vivo inhibitor assays using a humanized yeast cell-based platform. In agreement with published in vitro assays, we demonstrate that HU-based PTPD is a species-specific hFEN1 inhibitor. In contrast, another reported hFEN1 inhibitor, the arylstibonic acid derivative NSC-13755, was determined to have off-target effects resulting in a synthetic lethal phenotype with yRAD27-deficient strains. Our study expands the list of human-yeast complementation pairs to nonessential genes by defining novel cell-based assays that can be utilized as a broad resource to study human drug targets.


Subject(s)
Flap Endonucleases/genetics , Fungal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Chromosomal Instability/drug effects , Chromosomal Instability/genetics , Drug Development/methods , Flap Endonucleases/antagonists & inhibitors , Fungal Proteins/antagonists & inhibitors , Genetic Complementation Test , Humans , Mutation/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors
5.
Genome Res ; 28(2): 266-274, 2018 02.
Article in English | MEDLINE | ID: mdl-29273626

ABSTRACT

Advances in long-read single molecule sequencing have opened new possibilities for 'benchtop' whole-genome sequencing. The Oxford Nanopore Technologies MinION is a portable device that uses nanopore technology that can directly sequence DNA molecules. MinION single molecule long sequence reads are well suited for de novo assembly of complex genomes as they facilitate the construction of highly contiguous physical genome maps obviating the need for labor-intensive physical genome mapping. Long sequence reads can also be used to delineate complex chromosomal rearrangements, such as those that occur in tumor cells, that can confound analysis using short reads. Here, we assessed MinION long-read-derived sequences for feasibility concerning: (1) the de novo assembly of a large complex genome, and (2) the elucidation of complex rearrangements. The genomes of two Caenorhabditis elegans strains, a wild-type strain and a strain containing two complex rearrangements, were sequenced with MinION. Up to 42-fold coverage was obtained from a single flow cell, and the best pooled data assembly produced a highly contiguous wild-type C. elegans genome containing 48 contigs (N50 contig length = 3.99 Mb) covering >99% of the 100,286,401-base reference genome. Further, the MinION-derived genome assembly expanded the C. elegans reference genome by >2 Mb due to a more accurate determination of repetitive sequence elements and assembled the complete genomes of two co-extracted bacteria. MinION long-read sequence data also facilitated the elucidation of complex rearrangements in a mutagenized strain. The sequence accuracy of the MinION long-read contigs (∼98%) was improved using Illumina-derived sequence data to polish the final genome assembly to 99.8% nucleotide accuracy when compared to the reference assembly.


Subject(s)
Caenorhabditis elegans/genetics , Genome/genetics , Molecular Sequence Annotation , Animals , Chromosome Mapping , Gene Rearrangement/genetics , High-Throughput Nucleotide Sequencing , Repetitive Sequences, Nucleic Acid/genetics
6.
Nat Rev Genet ; 18(10): 613-623, 2017 10.
Article in English | MEDLINE | ID: mdl-28649135

ABSTRACT

A synthetic lethal interaction occurs between two genes when the perturbation of either gene alone is viable but the perturbation of both genes simultaneously results in the loss of viability. Key to exploiting synthetic lethality in cancer treatment are the identification and the mechanistic characterization of robust synthetic lethal genetic interactions. Advances in next-generation sequencing technologies are enabling the identification of hundreds of tumour-specific mutations and alterations in gene expression that could be targeted by a synthetic lethality approach. The translation of synthetic lethality to therapy will be assisted by the synthesis of genetic interaction data from model organisms, tumour genomes and human cell lines.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Animals , Epistasis, Genetic , Humans , Models, Animal
7.
Nat Commun ; 8: 14432, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28211448

ABSTRACT

G-quadruplex DNAs form four-stranded helical structures and are proposed to play key roles in different cellular processes. Targeting G-quadruplex DNAs for cancer treatment is a very promising prospect. Here, we show that CX-5461 is a G-quadruplex stabilizer, with specific toxicity against BRCA deficiencies in cancer cells and polyclonal patient-derived xenograft models, including tumours resistant to PARP inhibition. Exposure to CX-5461, and its related drug CX-3543, blocks replication forks and induces ssDNA gaps or breaks. The BRCA and NHEJ pathways are required for the repair of CX-5461 and CX-3543-induced DNA damage and failure to do so leads to lethality. These data strengthen the concept of G4 targeting as a therapeutic approach, specifically for targeting HR and NHEJ deficient cancers and other tumours deficient for DNA damage repair. CX-5461 is now in advanced phase I clinical trial for patients with BRCA1/2 deficient tumours (Canadian trial, NCT02719977, opened May 2016).


Subject(s)
BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , G-Quadruplexes , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Neoplasms/drug therapy , Animals , Base Sequence , Benzoxazines/pharmacology , Caenorhabditis elegans/drug effects , Cell Line, Tumor , Chromosomal Instability/genetics , DNA Damage , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Ribosomal/genetics , Female , G-Quadruplexes/drug effects , Genome, Human , Genotype , Homologous Recombination/drug effects , Humans , Mice , Quinolones/pharmacology , Saccharomyces cerevisiae/metabolism , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
8.
Genetics ; 197(2): 611-23, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24653001

ABSTRACT

Many tumors contain mutations that confer defects in the DNA-damage response and genome stability. DNA-damaging agents are powerful therapeutic tools that can differentially kill cells with an impaired DNA-damage response. The response to DNA damage is complex and composed of a network of coordinated pathways, often with a degree of redundancy. Tumor-specific somatic mutations in DNA-damage response genes could be exploited by inhibiting the function of a second gene product to increase the sensitivity of tumor cells to a sublethal concentration of a DNA-damaging therapeutic agent, resulting in a class of conditional synthetic lethality we call synthetic cytotoxicity. We used the Saccharomyces cerevisiae nonessential gene-deletion collection to screen for synthetic cytotoxic interactions with camptothecin, a topoisomerase I inhibitor, and a null mutation in TEL1, the S. cerevisiae ortholog of the mammalian tumor-suppressor gene, ATM. We found and validated 14 synthetic cytotoxic interactions that define at least five epistasis groups. One class of synthetic cytotoxic interaction was due to telomere defects. We also found that at least one synthetic cytotoxic interaction was conserved in Caenorhabditis elegans. We have demonstrated that synthetic cytotoxicity could be a useful strategy for expanding the sensitivity of certain tumors to DNA-damaging therapeutics.


Subject(s)
Camptothecin/chemistry , DNA Damage , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Epistasis, Genetic , Oligonucleotide Array Sequence Analysis , Saccharomyces cerevisiae/drug effects , Telomere/genetics , Topoisomerase I Inhibitors/chemistry
9.
Mol Cancer Ther ; 13(3): 724-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24356817

ABSTRACT

Recent data have identified STAG2, a core subunit of the multifunctional cohesin complex, as a highly recurrently mutated gene in several types of cancer. We sought to identify a therapeutic strategy to selectively target cancer cells harboring inactivating mutations of STAG2 using two independent pairs of isogenic glioblastoma cell lines containing either an endogenous mutant STAG2 allele or a wild-type STAG2 allele restored by homologous recombination. We find that mutations in STAG2 are associated with significantly increased sensitivity to inhibitors of the DNA repair enzyme PARP. STAG2-mutated, PARP-inhibited cells accumulated in G2 phase and had a higher percentage of micronuclei, fragmented nuclei, and chromatin bridges compared with wild-type STAG2 cells. We also observed more 53BP1 foci in STAG2-mutated glioblastoma cells, suggesting that these cells have defects in DNA repair. Furthermore, cells with mutations in STAG2 were more sensitive than cells with wild-type STAG2 when PARP inhibitors were used in combination with DNA-damaging agents. These data suggest that PARP is a potential target for tumors harboring inactivating mutations in STAG2, and strongly recommend that STAG2 status be determined and correlated with therapeutic response to PARP inhibitors, both prospectively and retrospectively, in clinical trials.


Subject(s)
Antigens, Nuclear/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Glioblastoma/genetics , Poly(ADP-ribose) Polymerases/genetics , Cell Line, Tumor , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/pathology , Humans , Mutation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Cohesins
10.
PLoS Genet ; 9(7): e1003582, 2013.
Article in English | MEDLINE | ID: mdl-23874209

ABSTRACT

The generation and resolution of joint molecule recombination intermediates is required to ensure bipolar chromosome segregation during meiosis. During wild type meiosis in Caenorhabditis elegans, SPO-11-generated double stranded breaks are resolved to generate a single crossover per bivalent and the remaining recombination intermediates are resolved as noncrossovers. We discovered that early recombination intermediates are limited by the C. elegans BLM ortholog, HIM-6, and in the absence of HIM-6 by the structure specific endonuclease MUS-81. In the absence of both MUS-81 and HIM-6, recombination intermediates persist, leading to chromosome breakage at diakinesis and inviable embryos. MUS-81 has an additional role in resolving late recombination intermediates in C. elegans. mus-81 mutants exhibited reduced crossover recombination frequencies suggesting that MUS-81 is required to generate a subset of meiotic crossovers. Similarly, the Mus81-related endonuclease XPF-1 is also required for a subset of meiotic crossovers. Although C. elegans gen-1 mutants have no detectable meiotic defect either alone or in combination with him-6, mus-81 or xpf-1 mutations, mus-81;xpf-1 double mutants are synthetic lethal. While mus-81;xpf-1 double mutants are proficient for the processing of early recombination intermediates, they exhibit defects in the post-pachytene chromosome reorganization and the asymmetric disassembly of the synaptonemal complex, presumably triggered by crossovers or crossover precursors. Consistent with a defect in resolving late recombination intermediates, mus-81; xpf-1 diakinetic bivalents are aberrant with fine DNA bridges visible between two distinct DAPI staining bodies. We were able to suppress the aberrant bivalent phenotype by microinjection of activated human GEN1 protein, which can cleave Holliday junctions, suggesting that the DNA bridges in mus-81; xpf-1 diakinetic oocytes are unresolved Holliday junctions. We propose that the MUS-81 and XPF-1 endonucleases act redundantly to process late recombination intermediates to form crossovers during C. elegans meiosis.


Subject(s)
Caenorhabditis elegans Proteins/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Meiosis/genetics , Recombination, Genetic , Animals , Caenorhabditis elegans/genetics , Chromosome Segregation/genetics , Crossing Over, Genetic , DNA, Cruciform/genetics , Endodeoxyribonucleases/genetics , Humans , Mutation
11.
Trends Genet ; 29(5): 290-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23333522

ABSTRACT

Cohesins are mutated in a significant number of tumors of various types making them attractive targets for chemotherapeutic intervention. However, cohesins have a spectrum of cellular roles including sister chromatid cohesion, transcription, replication, and repair. Which of these roles are central to cancer biology and which roles can be exploited for therapeutic intervention? Genetic interaction networks in yeast have identified synthetic lethal interactions between mutations in cohesin and replication fork mediators. These interactions are conserved in worms and in human cells suggesting that inhibition of replication fork stability mediators such as poly (ADP-ribose) polymerase (PARP) could result in the specific killing of tumors with cohesin mutations. These findings also highlight the utility of genetic interaction networks in model organisms for the identification of clinically relevant interactions. Here, we review this type of approach, emphasizing the power of synthetic lethal interactions to reveal new avenues for developing cancer therapeutics.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Replication/genetics , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Humans , Poly (ADP-Ribose) Polymerase-1 , Cohesins
12.
Nat Genet ; 44(12): 1310-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104009

ABSTRACT

Endometrial cancer is the sixth most commonly diagnosed cancer in women worldwide, causing ~74,000 deaths annually. Serous endometrial cancers are a clinically aggressive subtype with a poorly defined genetic etiology. We used whole-exome sequencing to comprehensively search for somatic mutations within ~22,000 protein-encoding genes in 13 primary serous endometrial tumors. We subsequently resequenced 18 genes, which were mutated in more than 1 tumor and/or were components of an enriched functional grouping, from 40 additional serous tumors. We identified high frequencies of somatic mutations in CHD4 (17%), EP300 (8%), ARID1A (6%), TSPYL2 (6%), FBXW7 (29%), SPOP (8%), MAP3K4 (6%) and ABCC9 (6%). Overall, 36.5% of serous tumors had a mutated chromatin-remodeling gene, and 35% had a mutated ubiquitin ligase complex gene, implicating frequent mutational disruption of these processes in the molecular pathogenesis of one of the deadliest forms of endometrial cancer.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Endometrioid/genetics , Chromatin Assembly and Disassembly/genetics , Endometrial Neoplasms/genetics , Exome/genetics , Ubiquitin-Protein Ligase Complexes/genetics , ATP-Binding Cassette Transporters/genetics , Adult , Autoantigens/genetics , Base Sequence , Cell Cycle Proteins/genetics , DNA-Binding Proteins , E1A-Associated p300 Protein/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Gene Frequency , Humans , MAP Kinase Kinase Kinase 4/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Repressor Proteins/genetics , Sequence Analysis, DNA , Sulfonylurea Receptors , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
13.
PLoS Genet ; 8(3): e1002574, 2012.
Article in English | MEDLINE | ID: mdl-22412391

ABSTRACT

Synthetic lethality has been proposed as a way to leverage the genetic differences found in tumor cells to affect their selective killing. Cohesins, which tether sister chromatids together until anaphase onset, are mutated in a variety of tumor types. The elucidation of synthetic lethal interactions with cohesin mutants therefore identifies potential therapeutic targets. We used a cross-species approach to identify robust negative genetic interactions with cohesin mutants. Utilizing essential and non-essential mutant synthetic genetic arrays in Saccharomyces cerevisiae, we screened genome-wide for genetic interactions with hypomorphic mutations in cohesin genes. A somatic cell proliferation assay in Caenorhabditis elegans demonstrated that the majority of interactions were conserved. Analysis of the interactions found that cohesin mutants require the function of genes that mediate replication fork progression. Conservation of these interactions between replication fork mediators and cohesin in both yeast and C. elegans prompted us to test whether other replication fork mediators not found in the yeast were required for viability in cohesin mutants. PARP1 has roles in the DNA damage response but also in the restart of stalled replication forks. We found that a hypomorphic allele of the C. elegans SMC1 orthologue, him-1(e879), genetically interacted with mutations in the orthologues of PAR metabolism genes resulting in a reduced brood size and somatic cell defects. We then demonstrated that this interaction is conserved in human cells by showing that PARP inhibitors reduce the viability of cultured human cells depleted for cohesin components. This work demonstrates that large-scale genetic interaction screening in yeast can identify clinically relevant genetic interactions and suggests that PARP inhibitors, which are currently undergoing clinical trials as a treatment of homologous recombination-deficient cancers, may be effective in treating cancers that harbor cohesin mutations.


Subject(s)
Caenorhabditis elegans , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Poly(ADP-ribose) Polymerases , Saccharomyces cerevisiae , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/genetics , Cell Proliferation , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA Damage/genetics , Epistasis, Genetic , Genes, Lethal , HCT116 Cells , Homologous Recombination/genetics , Humans , Mutation , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Cohesins
14.
DNA Repair (Amst) ; 10(11): 1174-82, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21968058

ABSTRACT

A family of helicases that are important in maintaining genome stability is the iron-sulfur group. Members of this family include DOG-1/FANCJ, RTEL1, XPD and Chl1p/DDX11. In Caenorhabitis elegans, the predicted gene M03C11.2 has orthology to the CHL1 (Chromosome loss 1) gene in Saccharomyces cerevisiae and DDX11 (DEAD/H box polypeptide 11) in humans. In this paper, we show that the chl-1 gene in C. elegans is required for normal development and fertility. Mutants have lineage-independent cell proliferation defects that result in a Stu (sterile uncoordinated) phenotype, characterized by gonadal abnormalities and a reduced number of D motor neurons and seam cells. A chromosome stability defect is present in the germ cells, where an abnormal number of DAPI-staining chromosomes appear in diakinesis. CHL-1 function is required for the integrity of poly-guanine/poly-cytosine DNA in the absence of DOG-1/FANCJ: the loss of CHL-1 alone does not result in the deletion of G-tracts, but it does increase the number of deletions observed in the dog-1; chl-1 double mutant, indicating a role for CHL-1 during replication and repair. In addition, we observed that cohesin defects increased the number of deletions in the absence of DOG-1/FANCJ. Our results demonstrate a role for CHL-1 in cell proliferation and maintaining normal chromosome numbers, and implicate CHL-1 in chromosome stability and repair of unresolved secondary structures during replication.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Chromosomal Instability , DNA Helicases/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , Gene Order , Homozygote , Humans , Male , Mutation , Phylogeny , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Cohesins
15.
BMC Genomics ; 11: 131, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20178641

ABSTRACT

BACKGROUND: The original sequencing and annotation of the Caenorhabditis elegans genome along with recent advances in sequencing technology provide an exceptional opportunity for the genomic analysis of wild-type and mutant strains. Using the Illumina Genome Analyzer, we sequenced the entire genome of Rec-1, a strain that alters the distribution of meiotic crossovers without changing the overall frequency. Rec-1 was derived from ethylmethane sulfonate (EMS)-treated strains, one of which had a high level of transposable element mobility. Sequencing of this strain provides an opportunity to examine the consequences on the genome of altering the distribution of meiotic recombination events. RESULTS: Using Illumina sequencing and MAQ software, 83% of the base pair sequence reads were aligned to the reference genome available at Wormbase, providing a 21-fold coverage of the genome. Using the software programs MAQ and Slider, we observed 1124 base pair differences between Rec-1 and the reference genome in Wormbase (WS190), and 441 between the mutagenized Rec-1 (BC313) and the wild-type N2 strain (VC2010). The most frequent base-substitution was G:C to A:T, 141 for the entire genome most of which were on chromosomes I or X, 55 and 31 respectively. With this data removed, no obvious pattern in the distribution of the base differences along the chromosomes was apparent. No major chromosomal rearrangements were observed, but additional insertions of transposable elements were detected. There are 11 extra copies of Tc1, and 8 of Tc2 in the Rec-1 genome, most likely the remains of past high-hopper activity in a progenitor strain. CONCLUSION: Our analysis of high-throughput sequencing was able to detect regions of direct repeat sequences, deletions, insertions of transposable elements, and base pair differences. A subset of sequence alterations affecting coding regions were confirmed by an independent approach using oligo array comparative genome hybridization. The major phenotype of the Rec-1 strain is an alteration in the preferred position of the meiotic recombination event with no other significant phenotypic consequences. In this study, we observed no evidence of a mutator effect at the nucleotide level attributable to the Rec-1 mutation.


Subject(s)
Caenorhabditis elegans/genetics , Genome, Helminth , Recombination, Genetic , Animals , Base Sequence , Comparative Genomic Hybridization , DNA Transposable Elements , DNA, Helminth/genetics , Meiosis , Molecular Sequence Data , Mutagenesis, Insertional , Repetitive Sequences, Nucleic Acid , Sequence Analysis, DNA , Software
16.
Mol Biol Cell ; 20(24): 5306-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19846659

ABSTRACT

Somatic mutations causing chromosome instability (CIN) in tumors can be exploited for selective killing of cancer cells by knockdown of second-site genes causing synthetic lethality. We tested and statistically validated synthetic lethal (SL) interactions between mutations in six Saccharomyces cerevisiae CIN genes orthologous to genes mutated in colon tumors and five additional CIN genes. To identify which SL interactions are conserved in higher organisms and represent potential chemotherapeutic targets, we developed an assay system in Caenorhabditis elegans to test genetic interactions causing synthetic proliferation defects in somatic cells. We made use of postembryonic RNA interference and the vulval cell lineage of C. elegans as a readout for somatic cell proliferation defects. We identified SL interactions between members of the cohesin complex and CTF4, RAD27, and components of the alternative RFC(CTF18) complex. The genetic interactions tested are highly conserved between S. cerevisiae and C. elegans and suggest that the alternative RFC components DCC1, CTF8, and CTF18 are ideal therapeutic targets because of their mild phenotype when knocked down singly in C. elegans. Furthermore, the C. elegans assay system will contribute to our knowledge of genetic interactions in a multicellular animal and is a powerful approach to identify new cancer therapeutic targets.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Multiprotein Complexes/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division , Cell Lineage , Cell Proliferation , Chromosomal Instability/genetics , Chromosomal Proteins, Non-Histone/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Genes, Neoplasm/genetics , Mutation/genetics , Saccharomyces cerevisiae/genetics , Cohesins
17.
Mol Genet Genomics ; 282(1): 37-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19330515

ABSTRACT

Genomic rearrangements are widely used in Caenorhabditis elegans research but many remain incompletely characterized at the physical level. We have used oligo-array comparative genomic analysis to assess the physical structure of 20 deficiencies and a single duplication of chromosome V. We find that while deletions internal to the chromosome appear simple in structure, terminal deletions are complex, containing duplications in addition to the deletion. Additionally, we confirm that transposon-induced deficiencies contain breakpoints that initiate at Tc1 elements. Finally, 13 of these deficiencies are known to suppress recombination far beyond the extent of the deletion. These deficiencies fall into two classes: strong and weak suppressors of adjacent recombination. Analysis of the deleted regions in these deficiencies reveals no common physical sites to explain the observed differences in recombination suppression. However, we find a strong correlation between the size of the rearranged chromosome and the severity of recombination suppression. Rearranged chromosomes that have a minor effect on recombination fall within 2% of normal chromosome size. Our observations highlight the use of array-based approaches for the analysis of rearranged genomes, revealing previously unidentified deficiency characteristics and addressing biologically relevant questions.


Subject(s)
Caenorhabditis elegans/genetics , Genes, Helminth , Animals , Chromosome Breakage , Chromosome Deletion , Chromosome Mapping , Comparative Genomic Hybridization , Crossing Over, Genetic , Gene Rearrangement , Genome, Helminth , Models, Genetic , Mutation , Recombination, Genetic , Translocation, Genetic
18.
Cell ; 135(2): 261-71, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18957201

ABSTRACT

Homologous recombination (HR) is an important conserved process for DNA repair and ensures maintenance of genome integrity. Inappropriate HR causes gross chromosomal rearrangements and tumorigenesis in mammals. In yeast, the Srs2 helicase eliminates inappropriate recombination events, but the functional equivalent of Srs2 in higher eukaryotes has been elusive. Here, we identify C. elegans RTEL-1 as a functional analog of Srs2 and describe its vertebrate counterpart, RTEL1, which is required for genome stability and tumor avoidance. We find that rtel-1 mutant worms and RTEL1-depleted human cells share characteristic phenotypes with yeast srs2 mutants: lethality upon deletion of the sgs1/BLM homolog, hyperrecombination, and DNA damage sensitivity. In vitro, purified human RTEL1 antagonizes HR by promoting the disassembly of D loop recombination intermediates in a reaction dependent upon ATP hydrolysis. We propose that loss of HR control after deregulation of RTEL1 may be a critical event that drives genome instability and cancer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , DNA Helicases/metabolism , Genomic Instability , Recombination, Genetic , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DNA/metabolism , DNA Helicases/genetics , DNA Repair , Humans , Mutation , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism
19.
DNA Repair (Amst) ; 7(11): 1846-54, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18708164

ABSTRACT

The Caenorhabditis elegans ortholog of the Fanconi anemia pathway component J (FANCJ) is DOG-1, which is essential for genome stability. Previous studies have shown that disruption of the dog-1 gene generates small deletions of poly-C/poly-G tracts detectable by PCR and results in a mutator phenotype. In this paper, we describe the isolation and characterization of lethal mutations resulting from the loss of dog-1 function. The mutant strains were analyzed using a combination of techniques including genetic mapping, SNP mapping, and oaCGH (oligo array Comparative Genome Hybridization). Using the eT1 balancer system to recover lethal mutants, we isolated, in addition to small deletions, large chromosomal rearrangements, including duplications, translocations and deficiencies. The forward mutation frequency was 10-fold higher than the spontaneous frequency for eT1, and equivalent to that observed for low doses of standard mutagens. From a screen for suppressors of mdf-1/MAD1 lethality, we previously had isolated such-4(h2168), shown here to be a large tandem duplication. Thus, the range of mutational events caused by lack of DOG-1/FANCJ is much broader than previously described.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA Helicases/metabolism , DNA Mutational Analysis , Animals , Caenorhabditis elegans , Chromosome Mapping , Chromosomes/ultrastructure , DNA Damage , Gene Deletion , Genetic Markers , Models, Biological , Models, Genetic , Mutagens , Mutation , Nucleic Acid Hybridization , Polymorphism, Single Nucleotide
20.
Mol Cell Biol ; 28(5): 1470-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18086896

ABSTRACT

Fanconi anemia (FA) is a cancer susceptibility syndrome characterized by defective DNA interstrand cross-link (ICL) repair. Here, we show that DOG-1 is the Caenorhabditis elegans homologue of FANCJ, a helicase mutated in FA-J patients. DOG-1 performs a conserved role in ICL repair, as dog-1 mutants are hypersensitive to ICL-inducing agents, but not to UVC irradiation or X rays. Genetic analysis indicated that dog-1 is epistatic with fcd-2 (C. elegans FANCD2) but is nonepistatic with brc-1 (C. elegans BRCA1), thus establishing the existence of two distinct pathways of ICL repair in worms. Furthermore, DOG-1 is dispensable for FCD-2 and RAD-51 focus formation, suggesting that DOG-1 operates downstream of FCD-2 and RAD-51 in ICL repair. DOG-1 was previously implicated in poly(G)/poly(C) (G/C) tract maintenance during DNA replication. G/C tracts remain stable in the absence of ATL-1, CLK-2 (FA pathway activators), FCD-2, BRC-2, and MLH-1 (associated FA components), implying that DOG-1 is the sole FA component required for G/C tract maintenance in a wild-type background. However, FCD-2 is required to promote deletion-free repair at G/C tracts in dog-1 mutants, consistent with a role for FA factors at the replication fork. The functional conservation between DOG-1 and FANCJ suggests a possible role for FANCJ in G/C tract maintenance in human cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , DNA Helicases/physiology , DNA Repair/physiology , DNA-Binding Proteins/physiology , Fanconi Anemia Complementation Group Proteins/physiology , RNA Helicases/physiology , Amino Acid Sequence , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cross-Linking Reagents/pharmacology , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Immunohistochemistry , Molecular Sequence Data , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL