Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 149(4): 795-806, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22579284

ABSTRACT

T loops and telomeric G-quadruplex (G4) DNA structures pose a potential threat to genome stability and must be dismantled to permit efficient telomere replication. Here we implicate the helicase RTEL1 in the removal of telomeric DNA secondary structures, which is essential for preventing telomere fragility and loss. In the absence of RTEL1, T loops are inappropriately resolved by the SLX4 nuclease complex, resulting in loss of the telomere as a circle. Depleting SLX4 or blocking DNA replication abolished telomere circles (TCs) and rescued telomere loss in RTEL1(-/-) cells but failed to suppress telomere fragility. Conversely, stabilization of telomeric G4-DNA or loss of BLM dramatically enhanced telomere fragility in RTEL1-deficient cells but had no impact on TC formation or telomere loss. We propose that RTEL1 performs two distinct functions at telomeres: it disassembles T loops and also counteracts telomeric G4-DNA structures, which together ensure the dynamics and stability of the telomere.


Subject(s)
DNA Helicases/metabolism , G-Quadruplexes , Telomere/metabolism , Animals , DNA Replication , Fibroblasts/metabolism , Mice , Nucleic Acid Conformation , Recombinases/metabolism
2.
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
3.
Genes Dev ; 29(18): 1969-79, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26385965

ABSTRACT

The Caenorhabditis elegans gene rec-1 was the first genetic locus identified in metazoa to affect the distribution of meiotic crossovers along the chromosome. We report that rec-1 encodes a distant paralog of HIM-5, which was discovered by whole-genome sequencing and confirmed by multiple genome-edited alleles. REC-1 is phosphorylated by cyclin-dependent kinase (CDK) in vitro, and mutation of the CDK consensus sites in REC-1 compromises meiotic crossover distribution in vivo. Unexpectedly, rec-1; him-5 double mutants are synthetic-lethal due to a defect in meiotic double-strand break formation. Thus, we uncovered an unexpected robustness to meiotic DSB formation and crossover positioning that is executed by HIM-5 and REC-1 and regulated by phosphorylation.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/genetics , Crossing Over, Genetic/genetics , DNA Breaks, Double-Stranded , Animals , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Meiosis/genetics
4.
Molecules ; 27(6)2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35335240

ABSTRACT

Helminth infections continue to be a neglected global threat in tropical regions, and there have been growing cases of anthelmintic resistance reported towards the existing anthelmintic drugs. Thus, the search for a novel anthelmintic agent has been increasing, especially those derived from plants. Leucaena leucocephala (LL) is a leguminous plant that is known to have several pharmacological activities, including anthelmintic activity. It is widely known to contain a toxic compound called mimosine, which we believed could be a potential lead candidate that could exert a potent anthelmintic effect. Hence, this study aimed to validate the presence of mimosine in LL extract and to investigate the anthelmintic effect of LL extract and mimosine on head thrashing, egg-laying, and pharyngeal pumping activities using the animal model Caenorhabditis elegans (C. elegans). Mimosine content in LL extract was confirmed through an HPLC analysis of spiking LL extract with different mimosine concentrations, whereby an increasing trend in peak heights was observed at a retention time of 0.9 min. LL extract and mimosine caused a significant dose-dependent increase in the percentage of worm mortality, which produced LC50s of 73 mg/mL and 6.39 mg/mL, respectively. Exposure of C. elegans to different concentrations of LL extract and mimosine significantly decreased the head thrashing, egg-laying, and mean pump amplitude of pharyngeal pumping activity. We speculated that these behavioral changes are due to the inhibitory effect of LL extract and mimosine on an L-type calcium channel called EGL-19. Our findings provide evidential support for the potential of LL extract and its active compound, mimosine, as novel anthelmintic candidates. However, the underlying mechanism of the anthelmintic action has yet to be elucidated.


Subject(s)
Anthelmintics , Fabaceae , Animals , Anthelmintics/pharmacology , Caenorhabditis elegans , Mimosine/pharmacology , Plant Extracts/pharmacology
5.
Nature ; 518(7538): 258-62, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25642963

ABSTRACT

Large-scale genomic studies have shown that half of epithelial ovarian cancers (EOCs) have alterations in genes regulating homologous recombination (HR) repair. Loss of HR accounts for the genomic instability of EOCs and for their cellular hyper-dependence on alternative poly-ADP ribose polymerase (PARP)-mediated DNA repair mechanisms. Previous studies have implicated the DNA polymerase θ (Polθ also known as POLQ, encoded by POLQ) in a pathway required for the repair of DNA double-strand breaks, referred to as the error-prone microhomology-mediated end-joining (MMEJ) pathway. Whether Polθ interacts with canonical DNA repair pathways to prevent genomic instability remains unknown. Here we report an inverse correlation between HR activity and Polθ expression in EOCs. Knockdown of Polθ in HR-proficient cells upregulates HR activity and RAD51 nucleofilament assembly, while knockdown of Polθ in HR-deficient EOCs enhances cell death. Consistent with these results, genetic inactivation of an HR gene (Fancd2) and Polq in mice results in embryonic lethality. Moreover, Polθ contains RAD51 binding motifs and it blocks RAD51-mediated recombination. Our results reveal a synthetic lethal relationship between the HR pathway and Polθ-mediated repair in EOCs, and identify Polθ as a novel druggable target for cancer therapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Amino Acid Motifs , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle , Cell Death , Cell Line, Tumor , DNA End-Joining Repair/genetics , DNA Replication , DNA-Directed DNA Polymerase/deficiency , Embryo Loss , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Genomic Instability , Homologous Recombination/genetics , Humans , Mice , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Protein Binding , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Recombinational DNA Repair/genetics , DNA Polymerase theta
6.
Mol Cell ; 45(1): 75-86, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22153967

ABSTRACT

Inappropriate homologous recombination (HR) causes genomic instability and cancer. In yeast, the UvrD family helicase Srs2 is recruited to sites of DNA replication by SUMO-modified PCNA, where it acts to restrict HR by disassembling toxic RAD51 nucleofilaments. How human cells control recombination at replication forks is unknown. Here, we report that the protein PARI, containing a UvrD-like helicase domain, is a PCNA-interacting partner required for preservation of genome stability in human and DT40 chicken cells. Using cell-based and biochemical assays, we show that PARI restricts unscheduled recombination by interfering with the formation of RAD51-DNA HR structures. Finally, we show that PARI knockdown suppresses the genomic instability of Fanconi Anemia/BRCA pathway-deficient cells. Thus, we propose that PARI is a long sought-after factor that suppresses inappropriate recombination events at mammalian replication forks.


Subject(s)
Carrier Proteins/physiology , Homologous Recombination/physiology , Proliferating Cell Nuclear Antigen/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chickens/genetics , DNA Repair , DNA-Binding Proteins , Gene Expression Regulation , Genomic Instability , HEK293 Cells , HeLa Cells , Humans , Rad51 Recombinase/metabolism
7.
Mol Cell ; 37(2): 259-72, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20122407

ABSTRACT

Homologous recombination (HR) is essential for repair of meiotic DNA double-strand breaks (DSBs). Although the mechanisms of RAD-51-DNA filament assembly and strand exchange are well characterized, the subsequent steps of HR are less well defined. Here, we describe a synthetic lethal interaction between the C. elegans helicase helq-1 and RAD-51 paralog rfs-1, which results in a block to meiotic DSB repair after strand invasion. Whereas RAD-51-ssDNA filaments assemble at meiotic DSBs with normal kinetics in helq-1, rfs-1 double mutants, persistence of RAD-51 foci and genetic interactions with rtel-1 suggest a failure to disassemble RAD-51 from strand invasion intermediates. Indeed, purified HELQ-1 and RFS-1 independently bind to and promote the disassembly of RAD-51 from double-stranded, but not single-stranded, DNA filaments via distinct mechanisms in vitro. These results indicate that two compensating activities are required to promote postsynaptic RAD-51 filament disassembly, which are collectively essential for completion of meiotic DSB repair.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/enzymology , DNA Breaks, Double-Stranded , DNA Repair/physiology , DNA-Binding Proteins/physiology , Meiosis , Rad51 Recombinase/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Helicases/physiology , DNA Repair/genetics , DNA, Helminth/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation , Recombination, Genetic
8.
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
9.
Biomed Pharmacother ; 143: 112066, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560550

ABSTRACT

Gynura procumbens (GP) is a herbal medicinal plant of South-East Asian origin, popularly recognised as 'Sambung nyawa'. The plant has been used traditionally to treat various diseases including hypertension. The anti-hypertensive activity of this plant has also been scientifically proven both in vivo and in vitro yet the investigation on its mechanisms of actions remains limited. Our previous study has demonstrated the vasodilatory action of both aqueous and methanol GP extracts possibly via activation of the cholinergic pathway and that kaempferol 3-O-rutinoside is the active ingredient responsible in mediating this effect. Hence, in this study we further confirm the involvement of the cholinergic pathway by using several pharmacological interventions, focusing on the downstream mechanism of this pathway. Our results showed that in the presence of endothelium, GP extracts induced vasodilation via activation of the muscarinic M3 receptors. However, in the absence of endothelium, GP mediated vasodilation possibly via stimulation of other muscarinic receptors and/or involvement of nicotinic receptors, a speculation that needs further investigations. GP-induced relaxation was markedly inhibited by nitric oxide (NO) blocker, L-NAME, suggesting that GP elicited ACh endothelium-dependent relaxation by producing NO in rat aortic rings. In conclusion, these data demonstrate that the vasodilatory effect of GP extracts appears to be mediated via cholinergic pathway.


Subject(s)
Aorta, Thoracic/drug effects , Asteraceae , Muscarinic Agonists/pharmacology , Plant Extracts/pharmacology , Receptor, Muscarinic M3/agonists , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/metabolism , Asteraceae/chemistry , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Kaempferols/isolation & purification , Kaempferols/pharmacology , Male , Methanol/chemistry , Muscarinic Agonists/isolation & purification , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Plant Extracts/isolation & purification , Plant Leaves , Rats, Sprague-Dawley , Receptor, Muscarinic M3/metabolism , Signal Transduction , Solvents/chemistry , Vasodilator Agents/isolation & purification , Water/chemistry
10.
Heliyon ; 5(9): e02476, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31687571

ABSTRACT

In this study, the physicochemical, antioxidant, antibacterial properties, and the toxicity of propolis particles produced by stingless bee Heterotrigona itama found in Brunei Darussalam were investigated. Propolis particles of different sizes were extracted from raw propolis using various volume fractions of ethanol in water. Spectroscopic analyses were utilized to characterize the chemical structures, functional groups, as well as absorbance and fluorescence properties. The total antioxidant capacity of propolis particles, which was assessed using DPPH (2,2-diphenyl-1-picrylhydrazyl) assay, was found to increase with volume fraction of ethanol. The maximum antioxidant capacity was as high as 317.65 mg ascorbic acid equivalent per gram of propolis particles. All of the propolis particles showed antibacterial activity against Gram-positive (Bacillus subtilis and Staphylococcus aureus) and Gram-negative bacteria (Escherichia coli and Pseudomonas aeruginosa). The diameters of the inhibition zone were either significantly higher or equivalent to those of two standard antibiotics (rifampicin and streptomycin), suggesting strong antibacterial activity. The toxicity studies of propolis particles against Caenorhabditis elegans revealed that they are non-toxic after 24 h exposure. Overall findings suggest that H. itama propolis particles are not only an important source of natural antioxidants that could be beneficial for human health, but they have potentials as antimicrobial against bacteria.

11.
Mol Cell Biol ; 25(8): 3127-39, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15798199

ABSTRACT

The BRCA2 tumor suppressor is implicated in DNA double-strand break (DSB) repair by homologous recombination (HR), where it regulates the RAD51 recombinase. We describe a BRCA2-related protein of Caenorhabditis elegans (CeBRC-2) that interacts directly with RAD-51 via a single BRC motif and that binds preferentially to single-stranded DNA through an oligonucleotide-oligosaccharide binding fold. Cebrc-2 mutants fail to repair meiotic or radiation-induced DSBs by HR due to inefficient RAD-51 nuclear localization and a failure to target RAD-51 to sites of DSBs. Genetic and cytological comparisons of Cebrc-2 and rad-51 mutants revealed fundamental phenotypic differences that suggest a role for Cebrc-2 in promoting the use of an alternative repair pathway in the absence of rad-51 and independent of nonhomologous end joining (NHEJ). Unlike rad-51 mutants, Cebrc-2 mutants also accumulate RPA-1 at DSBs, and abnormal chromosome aggregates that arise during the meiotic prophase can be rescued by blocking the NHEJ pathway. CeBRC-2 also forms foci in response to DNA damage and can do so independently of rad-51. Thus, CeBRC-2 not only regulates RAD-51 during HR but can also function independently of rad-51 in DSB repair processes.


Subject(s)
Caenorhabditis elegans/physiology , DNA Damage , DNA Repair/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins , DNA/radiation effects , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Embryo, Nonmammalian , Gene Deletion , Genes, Lethal , Germ Cells/chemistry , Germ Cells/metabolism , Germ Cells/radiation effects , Molecular Sequence Data , Rad51 Recombinase , Replication Protein A
12.
J Mol Biol ; 361(2): 231-42, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16843491

ABSTRACT

The BRCA2 tumour suppressor regulates the RAD-51 recombinase during double-strand break (DSB) repair by homologous recombination (HR) but how BRCA2 executes its functions is not well understood. We previously described a functional homologue of BRCA2 in Caenorhabditis elegans (CeBRC-2) that binds preferentially to single-stranded DNA via an OB-fold domain and associates directly with RAD-51 via a single BRC domain. Consistent with a direct role in HR, Cebrc-2 mutants are defective for repair of meiotic and radiation-induced DSBs due to an inability to regulate RAD-51. Here, we explore the function of CeBRC-2 in HR processes using purified proteins. We show that CeBRC-2 stimulates RAD-51-mediated D-loop formation and reduces the rate of ATP hydrolysis catalysed by RAD-51. These functions of CeBRC-2 are dependent upon direct association with RAD-51 via its BRC motif and on its DNA-binding activity, as point mutations in the BRC domain that abolish RAD-51 binding or the BRC domain of CeBRC-2 alone, lacking the DNA-binding domain, fail to stimulate RAD-51-mediated D-loop formation and do not reduce the rate of ATP hydrolysis by RAD-51. Phenotypic comparison of Cebrc-2 and rad-51 mutants also revealed a role for CeBRC-2 in an error-prone DSB repair pathway independent of rad-51 and non-homologous end joining, raising the possibility that CeBRC-2 may have replaced the role of vertebrate Rad52 in DNA single-strand annealing (SSA), which is missing from C. elegans. Indeed, we show here that CeBRC-2 mediates SSA of RPA-oligonucleotide complexes similar to Rad52. These results reveal RAD-51-dependent and -independent functions of CeBRC-2 that provide an explanation for the difference in DNA repair defects observed in Cebrc-2 and rad-51 mutants, and define mechanistic roles for CeBRC-2 in HR and in the SSA pathway for DSB repair.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA Repair/physiology , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Recombination, Genetic , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , DNA Damage , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , Hydrolysis , Molecular Sequence Data , Rad51 Recombinase/genetics
13.
Biol Open ; 5(5): 537-49, 2016 May 15.
Article in English | MEDLINE | ID: mdl-27010030

ABSTRACT

Studies in Caenorhabditis elegans have revealed osmoregulatory systems engaged when worms experience hypertonic conditions, but less is known about measures employed when faced with hypotonic stress. Inactivation of fmo-4, which encodes flavin-containing monooxygenase-4, results in dramatic hypoosmotic hypersensitivity; worms are unable to prevent overwhelming water influx and swell rapidly, finally rupturing due to high internal hydrostatic pressure. fmo-4 is expressed prominently in hypodermis, duct and pore cells but is excluded from the excretory cell. Thus, FMO-4 plays a crucial osmoregulatory role by promoting clearance of excess water that enters during hypotonicity, perhaps by synthesizing an osmolyte that acts to establish an osmotic gradient from excretory cell to duct and pore cells. C. elegans FMO-4 contains a C-terminal extension conserved in all nematode FMO-4s. The coincidently numbered human FMO4 also contains an extended C-terminus with features similar to those of FMO-4. Although these shared sequence characteristics suggest potential orthology, human FMO4 was unable to rescue the fmo-4 osmoregulatory defect. Intriguingly, however, mammalian FMO4 is expressed predominantly in the kidney - an appropriate site if it too is, or once was, involved in osmoregulation.

14.
Gene ; 346: 83-96, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15716098

ABSTRACT

The flavin-containing monooxygenase (FMO) gene family is conserved and ancient with representatives present in almost all phyla so far examined. The genes encode FAD-, NADP- and O(2)-dependent enzymes that catalyse oxygenation of soft-nucleophilic heteroatom centres in a range of substrates. Although usually classified as xenobiotic-metabolising enzymes, examples of FMOs exist that have evolved to metabolise specific endogenous substrates as part of a discrete physiological process. The genome of Caenorhabditis elegans contains five predicted genes encoding putative homologs of mammalian FMOs, K08C7.2, K08C7.5, Y39A1A.19, F53F4.5 and H24K24.5, which we have named fmo and numbered fmo-1 to fmo-5, respectively. As a first step towards determining their functional role(s), we have experimentally characterised these C. elegans fmo genes including analysing reporter gene expression patterns and RNAi phenotypes. Two major gene expression patterns were observed, either intestinal or hypodermal, but no gross RNAi phenotypes were found possibly due to functional redundancy. The internal structures of fmo-2, fmo-3 and fmo-4 have been compared with orthologs identified in the related nematode C. briggsae. For each orthologous pair, a global comparison of the paired upstream intergenic regions was performed and a number of conserved noncoding sequences, which may represent potential cis-regulatory elements, identified. Phylogenetic analysis reveals that several of the fmo homologs are the result of gene duplication along the lineage leading to the nematodes.


Subject(s)
Caenorhabditis elegans/genetics , Gene Expression , Genome , Oxygenases/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Phylogeny , RNA Interference , Species Specificity
15.
Cell Rep ; 6(5): 827-35, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24561255

ABSTRACT

Human RTEL1 is an essential, multifunctional helicase that maintains telomeres, regulates homologous recombination, and helps prevent bone marrow failure. Here, we show that RTEL1 also blocks trinucleotide repeat expansions, the causal mutation for 17 neurological diseases. Increased expansion frequencies of (CTG⋅CAG) repeats occurred in human cells following knockdown of RTEL1, but not the alternative helicase Fbh1, and purified RTEL1 efficiently unwound triplet repeat hairpins in vitro. The expansion-blocking activity of RTEL1 also required Rad18 and HLTF, homologs of yeast Rad18 and Rad5. These findings are reminiscent of budding yeast Srs2, which inhibits expansions, unwinds hairpins, and prevents triplet-repeat-induced chromosome fragility. Accordingly, we found expansions and fragility were suppressed in yeast srs2 mutants expressing RTEL1, but not Fbh1. We propose that RTEL1 serves as a human analog of Srs2 to inhibit (CTG⋅CAG) repeat expansions and fragility, likely by unwinding problematic hairpins.


Subject(s)
Chromosome Fragility , DNA Helicases/genetics , Trinucleotide Repeat Expansion , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Mutation , Polymorphism, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Science ; 342(6155): 239-42, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24115439

ABSTRACT

Regulator of telomere length 1 (RTEL1) is an essential DNA helicase that disassembles telomere loops (T loops) and suppresses telomere fragility to maintain the integrity of chromosome ends. We established that RTEL1 also associates with the replisome through binding to proliferating cell nuclear antigen (PCNA). Mouse cells disrupted for the RTEL1-PCNA interaction (PIP mutant) exhibited accelerated senescence, replication fork instability, reduced replication fork extension rates, and increased origin usage. Although T-loop disassembly at telomeres was unaffected in the mutant cells, telomere replication was compromised, leading to fragile sites at telomeres. RTEL1-PIP mutant mice were viable, but loss of the RTEL1-PCNA interaction accelerated the onset of tumorigenesis in p53-deficient mice. We propose that RTEL1 plays a critical role in both telomere and genome-wide replication, which is crucial for genetic stability and tumor avoidance.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA Helicases/metabolism , DNA Replication , Genome/genetics , Proliferating Cell Nuclear Antigen/metabolism , Telomere/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , DNA Helicases/genetics , Mice , Mice, Mutant Strains , Tumor Suppressor Protein p53/genetics
19.
Proc Natl Acad Sci U S A ; 104(20): 8299-304, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17483448

ABSTRACT

Mutations in BRCA2 predispose individuals to breast cancer, a consequence of the role of BRCA2 in DNA repair. Human BRCA2 interacts with the recombinase RAD51 via eight BRC repeats. Controversy has existed, however, about whether the BRC interactions are primarily with RAD51 monomers or with the RAD51-DNA helical polymer, and whether there is a single interaction or multiple ones. We show here that the single BRC motif in the Caenorhabditis elegans BRCA2 homolog, CeBRC-2, contains two different RAD-51-binding regions. One of these regions binds only weakly to RAD-51-DNA filaments but strongly to RAD-51 alone and corresponds to the part of human BRC4 crystallized with RAD51. Injection of a peptide corresponding to this region into worms inhibits the normal formation of RAD-51 foci in response to ionizing radiation (IR). Conversely, peptides corresponding to the second region bind strongly to RAD-51-DNA filaments but do not bind to RAD-51 alone. Three-dimensional reconstructions from electron micrographs show that this peptide binds to the RAD-51 N-terminal domain, which has been shown to have a regulatory function. Injection of this peptide into worms before IR leads to a dramatic increase and persistence of IR-induced RAD-51 foci. This peptide also inhibits the RAD-51 ATPase activity, required for filament depolymerization. These results support a model where an interaction with RAD-51 alone is likely involved in filament nucleation, whereas a second independent interaction is involved in stabilization of RAD-51 filaments by BRCA2. The multiple interactions between BRCA2-like molecules and RAD51 provide insights into why mutations in BRCA2 lead to cancer.


Subject(s)
BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , Caenorhabditis elegans/metabolism , DNA, Helminth/metabolism , Rad51 Recombinase/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , DNA, Helminth/ultrastructure , Genes, Dominant , Humans , Hydrolysis , Molecular Sequence Data , Protein Structure, Tertiary , Rad51 Recombinase/ultrastructure , Structure-Activity Relationship , Time Factors
20.
EMBO J ; 25(10): 2178-88, 2006 May 17.
Article in English | MEDLINE | ID: mdl-16628214

ABSTRACT

The BRCA1 tumour suppressor and its heterodimeric partner BARD1 constitute an E3-ubiquitin (Ub) ligase and function in DNA repair by unknown mechanisms. We show here that the Caenorhabditis elegans BRCA1/BARD1 (CeBCD) complex possesses an E3-Ub ligase responsible for ubiquitylation at DNA damage sites following ionizing radiation (IR). The DNA damage checkpoint promotes the association of the CeBCD complex with E2-Ub conjugating enzyme, Ubc5(LET-70), leading to the formation of an active E3-Ub ligase on chromatin following IR. Correspondingly, defects in Ubc5(let-70) or the DNA damage checkpoint genes atl-1 or mre-11 abolish CeBCD-dependent ubiquitylation in vivo. Extending these findings to human cells reveals a requirement for UbcH5c, the MRN complex, gamma-H2AX and a co-dependence for ATM and ATR kinases for BRCA1-dependent ubiquitylation at DNA damage sites. Furthermore, we demonstrate that the DNA damage checkpoint promotes the association between BRCA1 and UbcH5c to form an active E3-Ub ligase on chromatin after IR. These data reveal that BRCA1-dependent ubiquitylation is activated at sites of DNA repair by the checkpoint as part of a conserved DNA damage response.


Subject(s)
BRCA1 Protein/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA Damage , Signal Transduction/physiology , Ubiquitin/metabolism , Animals , BRCA1 Protein/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cell Line , Chromatin/metabolism , DNA Repair , Enzyme Activation , Humans , Multiprotein Complexes , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL