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










Publication year range
1.
Nat Commun ; 14(1): 4445, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488098

ABSTRACT

RAD51C is an enigmatic predisposition gene for breast, ovarian, and prostate cancer. Currently, missing structural and related functional understanding limits patient mutation interpretation to homology-directed repair (HDR) function analysis. Here we report the RAD51C-XRCC3 (CX3) X-ray co-crystal structure with bound ATP analog and define separable RAD51C replication stability roles informed by its three-dimensional structure, assembly, and unappreciated polymerization motif. Mapping of cancer patient mutations as a functional guide confirms ATP-binding matching RAD51 recombinase, yet highlights distinct CX3 interfaces. Analyses of CRISPR/Cas9-edited human cells with RAD51C mutations combined with single-molecule, single-cell and biophysics measurements uncover discrete CX3 regions for DNA replication fork protection, restart and reversal, accomplished by separable functions in DNA binding and implied 5' RAD51 filament capping. Collective findings establish CX3 as a cancer-relevant replication stress response complex, show how HDR-proficient variants could contribute to tumor development, and identify regions to aid functional testing and classification of cancer mutations.


Subject(s)
Prostatic Neoplasms , Male , Humans , Rad51 Recombinase , Mutation , DNA Replication , Adenosine Triphosphate , DNA-Binding Proteins
2.
Oncotarget ; 7(38): 61874-61889, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27542204

ABSTRACT

NUCKS1 is a 27 kD vertebrate-specific protein, with a role in the DNA damage response. Here, we show that after 4 Gy total-body X-irradiation, Trp53+/- Nucks1+/- mice more rapidly developed tumors, particularly thymic lymphoma (TL), than Trp53+/- mice. TLs in both cohorts showed loss of heterozygosity (LOH) of the Trp53+ allele in essentially all cases. In contrast, LOH of the Nucks1+ allele was rare. Nucks1 expression correlated well with Nucks1 gene dosage in normal thymi, but was increased in the majority of TLs from Trp53+/- Nucks1+/- mice, suggesting that elevated Nucks1 message may be associated with progression towards malignancy in vivo. Trp53+/- Nucks1+/- mice frequently succumbed to CD4- CD8- TLs harboring translocations involving Igh but not Tcra/d, indicating TLs in Trp53+/- Nucks1+/- mice mostly originated prior to the double positive stage and at earlier lineage than TLs in Trp53+/- mice. Monoclonal rearrangements at Tcrb were more prevalent in TLs from Trp53+/- Nucks1+/- mice, as was infiltration of primary TL cells to distant organs (liver, kidney and spleen). We propose that, in the context of Trp53 deficiency, wild type levels of Nucks1 are required to suppress radiation-induced TL, likely through the role of the NUCKS1 protein in the DNA damage response.


Subject(s)
Lymphoma/genetics , Neoplasms, Radiation-Induced/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , Alleles , Animals , Antibodies, Monoclonal/chemistry , Comparative Genomic Hybridization , DNA Damage , Female , Gene Dosage , Genotype , Haploinsufficiency , Immunophenotyping , Kidney/metabolism , Liver/metabolism , Loss of Heterozygosity , Lymphoma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Radiation-Induced/metabolism , Spleen/metabolism , Up-Regulation
3.
Nucleic Acids Res ; 43(20): 9817-34, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26323318

ABSTRACT

NUCKS1 (nuclear casein kinase and cyclin-dependent kinase substrate 1) is a 27 kD chromosomal, vertebrate-specific protein, for which limited functional data exist. Here, we demonstrate that NUCKS1 shares extensive sequence homology with RAD51AP1 (RAD51 associated protein 1), suggesting that these two proteins are paralogs. Similar to the phenotypic effects of RAD51AP1 knockdown, we find that depletion of NUCKS1 in human cells impairs DNA repair by homologous recombination (HR) and chromosome stability. Depletion of NUCKS1 also results in greatly increased cellular sensitivity to mitomycin C (MMC), and in increased levels of spontaneous and MMC-induced chromatid breaks. NUCKS1 is critical to maintaining wild type HR capacity, and, as observed for a number of proteins involved in the HR pathway, functional loss of NUCKS1 leads to a slow down in DNA replication fork progression with a concomitant increase in the utilization of new replication origins. Interestingly, recombinant NUCKS1 shares the same DNA binding preference as RAD51AP1, but binds to DNA with reduced affinity when compared to RAD51AP1. Our results show that NUCKS1 is a chromatin-associated protein with a role in the DNA damage response and in HR, a DNA repair pathway critical for tumor suppression.


Subject(s)
Genomic Instability , Nuclear Proteins/physiology , Phosphoproteins/physiology , Recombinational DNA Repair , Cell Line , Chromatin/metabolism , Chromosome Aberrations , DNA/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , HeLa Cells/physiology , Humans , Mitomycin/pharmacology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/radiation effects , RNA-Binding Proteins , Rad51 Recombinase/metabolism , S Phase/radiation effects , Sequence Homology, Amino Acid , X-Rays
4.
DNA Repair (Amst) ; 24: 87-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25288561

ABSTRACT

RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Cell Line/drug effects , Cell Line/radiation effects , Chickens , Cisplatin/pharmacology , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , Genetic Complementation Test , Humans , Hydroxyurea/pharmacology , Molecular Sequence Data , RNA-Binding Proteins , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation, Ionizing , Vertebrates/genetics
5.
J Biol Chem ; 287(15): 12343-7, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22375013

ABSTRACT

Homologous recombination catalyzed by the RAD51 recombinase is essential for maintaining genome integrity upon the induction of DNA double strand breaks and other DNA lesions. By enhancing the recombinase activity of RAD51, RAD51AP1 (RAD51-associated protein 1) serves a key role in homologous recombination-mediated chromosome damage repair. We show here that RAD51AP1 harbors two distinct DNA binding domains that are both needed for maximal protein activity under physiological conditions. We have finely mapped the two DNA binding domains in RAD51AP1 and generated mutant variants that are impaired in either or both of the DNA binding domains. Examination of these mutants reveals that both domains are indispensable for RAD51AP1 function in cells. These and other results illuminate the mechanistic basis of RAD51AP1 action in homologous DNA repair.


Subject(s)
DNA Repair , DNA-Binding Proteins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Conserved Sequence , DNA/chemistry , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Mapping , Phenotype , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Deletion
6.
J Biol Chem ; 286(43): 37328-34, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21903585

ABSTRACT

Homologous recombination (HR) reactions mediated by the RAD51 recombinase are essential for DNA and replication fork repair, genome stability, and tumor suppression. RAD51-associated protein 1 (RAD51AP1) is an important HR factor that associates with and stimulates the recombinase activity of RAD51. We have recently shown that RAD51AP1 also partners with the meiotic recombinase DMC1, displaying isoform-specific interactions with DMC1. Here, we have characterized the DMC1 interaction site in RAD51AP1 by a series of truncations and point mutations to uncover a highly conserved WVPP motif critical for DMC1 interaction but dispensable for RAD51 association. This RAD51AP1 motif is reminiscent of the FVPP motif in the tumor suppressor protein BRCA2 that mediates DMC1 interaction. These results further implicate RAD51AP1 in meiotic HR via RAD51 and DMC1.


Subject(s)
Cell Cycle Proteins/chemistry , DNA-Binding Proteins/chemistry , Amino Acid Motifs , BRCA1 Protein/chemistry , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Protein Binding , RNA-Binding Proteins , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 108(9): 3560-5, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21307306

ABSTRACT

Homologous recombination is needed for meiotic chromosome segregation, genome maintenance, and tumor suppression. RAD51AP1 (RAD51 associated protein 1) has been shown to interact with and enhance the recombinase activity of RAD51. Accordingly, genetic ablation of RAD51AP1 leads to enhanced sensitivity to and also chromosome aberrations upon DNA damage, demonstrating a role for RAD51AP1 in mitotic homologous recombination. Here we show physical association of RAD51AP1 with the meiosis-specific recombinase DMC1 and a stimulatory effect of RAD51AP1 on the DMC1-mediated D-loop reaction. Mechanistic studies have revealed that RAD51AP1 enhances the ability of the DMC1 presynaptic filament to capture the duplex-DNA partner and to assemble the synaptic complex, in which the recombining DNA strands are homologously aligned. We also provide evidence that functional cooperation is dependent on complex formation between DMC1 and RAD51AP1 and that distinct epitopes in RAD51AP1 mediate interactions with RAD51 and DMC1. Finally, we show that RAD51AP1 is expressed in mouse testes, and that RAD51AP1 foci colocalize with a subset of DMC1 foci in spermatocytes. These results suggest that RAD51AP1 also serves an important role in meiotic homologous recombination.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Meiosis , Nuclear Proteins/metabolism , Recombinases/metabolism , Animals , Chromatin/metabolism , Chromosome Pairing , DNA-Binding Proteins/isolation & purification , Humans , Male , Mice , Mutant Proteins/metabolism , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Phosphate-Binding Proteins , Protein Binding , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Protein Transport , RNA-Binding Proteins , Rad51 Recombinase/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism
8.
Nat Struct Mol Biol ; 17(10): 1255-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20871616

ABSTRACT

Homologous recombination mediated by RAD51 recombinase helps eliminate chromosomal lesions, such as DNA double-strand breaks induced by radiation or arising from injured DNA replication forks. The tumor suppressors BRCA2 and PALB2 act together to deliver RAD51 to chromosomal lesions to initiate repair. Here we document a new function of PALB2: to enhance RAD51's ability to form the D loop. We show that PALB2 binds DNA and physically interacts with RAD51. Notably, although PALB2 alone stimulates D-loop formation, it has a cooperative effect with RAD51AP1, an enhancer of RAD51. This stimulation stems from the ability of PALB2 to function with RAD51 and RAD51AP1 to assemble the synaptic complex. Our results demonstrate the multifaceted role of PALB2 in chromosome damage repair. Because PALB2 mutations can cause cancer or Fanconi anemia, our findings shed light on the mechanism of tumor suppression in humans.


Subject(s)
BRCA2 Protein/physiology , Breast Neoplasms/metabolism , DNA Repair/physiology , DNA, Neoplasm/metabolism , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Rad51 Recombinase/physiology , Recombination, Genetic/physiology , Tumor Suppressor Proteins/physiology , Apoptosis Regulatory Proteins , BRCA2 Protein/chemistry , DNA-Binding Proteins/chemistry , Fanconi Anemia Complementation Group N Protein , Female , Humans , Multiprotein Complexes , Neoplasm Proteins/chemistry , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Mapping , RNA-Binding Proteins , Rad51 Recombinase/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
9.
Radiat Res ; 173(1): 27-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20041757

ABSTRACT

To test the contribution of homologous recombinational repair (HRR) in repairing DNA damage sites induced by high-energy iron ions, we used (1) HRR-deficient rodent cells carrying a deletion in the RAD51D gene and (2) syngeneic human cells impaired for HRR by RAD51D or RAD51 knockdown using RNA interference. We found that in response to exposure to iron ions, HRR contributed to cell survival in rodent cells and that HRR deficiency abrogated RAD51 focus formation. Complementation of the HRR defect by human RAD51D rescues both enhanced cytotoxicity and RAD51 focus formation. For human cells irradiated with iron ions, cell survival was decreased, and in p53 mutant cells, the levels of mutagenesis were increased when HRR was impaired. Human cells synchronized in S phase exhibited a more pronounced resistance to iron ions compared with cells in G(1) phase, and this increase in radioresistance was diminished by RAD51 knockdown. These results indicate a role for RAD51-mediated DNA repair (i.e. HRR) in removing a fraction of clustered lesions induced by charged-particle radiation. Our results are the first to directly show the requirement for an intact HRR pathway in human cells in ensuring DNA repair and cell survival after exposure to high-energy high-LET radiation.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , Iron/toxicity , Recombination, Genetic , Animals , Base Sequence , CHO Cells , Cell Survival/genetics , Cell Survival/radiation effects , Cricetinae , Cricetulus , G2 Phase/genetics , G2 Phase/radiation effects , Gene Knockdown Techniques , Humans , Mutagenesis/radiation effects , Mutation/radiation effects , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , S Phase/genetics , S Phase/radiation effects , Thymidine Kinase/genetics , Tumor Suppressor Protein p53/metabolism
10.
Nucleic Acids Res ; 38(4): 1061-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19942681

ABSTRACT

RAD51, a key protein in the homologous recombinational DNA repair (HRR) pathway, is the major strand-transferase required for mitotic recombination. An important early step in HRR is the formation of single-stranded DNA (ss-DNA) coated by RPA (a ss-DNA-binding protein). Displacement of RPA by RAD51 is highly regulated and facilitated by a number of different proteins known as the 'recombination mediators'. To assist these recombination mediators, a second group of proteins also is required and we are defining these proteins here as 'recombination co-mediators'. Defects in either recombination mediators or co-mediators, including BRCA1 and BRCA2, lead to impaired HRR that can genetically be complemented for (i.e. suppressed) by overexpression of RAD51. Defects in HRR have long been known to contribute to genomic instability leading to tumor development. Since genomic instability also slows cell growth, precancerous cells presumably require genomic re-stabilization to gain a growth advantage. RAD51 is overexpressed in many tumors, and therefore, we hypothesize that the complementing ability of elevated levels of RAD51 in tumors with initial HRR defects limits genomic instability during carcinogenic progression. Of particular interest, this model may also help explain the high frequency of TP53 mutations in human cancers, since wild-type p53 represses RAD51 expression.


Subject(s)
DNA Repair , Genomic Instability , Neoplasms/genetics , Rad51 Recombinase/metabolism , Recombination, Genetic , Animals , Genes, p53 , Humans , Models, Genetic , Mutation , Neoplasms/metabolism
11.
Mol Cell ; 28(3): 482-90, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17996711

ABSTRACT

Homologous recombination (HR) repairs chromosome damage and is indispensable for tumor suppression in humans. RAD51 mediates the DNA strand-pairing step in HR. RAD51 associated protein 1 (RAD51AP1) is a RAD51-interacting protein whose function has remained elusive. Knockdown of RAD51AP1 in human cells by RNA interference engenders sensitivity to different types of genotoxic stress, and RAD51AP1 is epistatic to the HR protein XRCC3. Moreover, RAD51AP1-depleted cells are impaired for the recombinational repair of a DNA double-strand break and exhibit chromatid breaks both spontaneously and upon DNA-damaging treatment. Purified RAD51AP1 binds both dsDNA and a D loop structure and, only when able to interact with RAD51, greatly stimulates the RAD51-mediated D loop reaction. Biochemical and cytological results show that RAD51AP1 functions at a step subsequent to the assembly of the RAD51-ssDNA nucleoprotein filament. Our findings provide evidence that RAD51AP1 helps maintain genomic integrity via RAD51 recombinase enhancement.


Subject(s)
DNA-Binding Proteins/physiology , Genomic Instability , Rad51 Recombinase/metabolism , Recombination, Genetic , Chromatids/metabolism , DNA Breaks , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Mutation , Nucleic Acid Conformation , RNA-Binding Proteins
12.
Nucleic Acids Res ; 34(18): 5081-92, 2006.
Article in English | MEDLINE | ID: mdl-16990250

ABSTRACT

Many interacting proteins regulate and/or assist the activities of RAD51, a recombinase which plays a critical role in both DNA repair and meiotic recombination. Yeast two-hybrid screening of a human testis cDNA library revealed a new protein, RAD51AP2 (RAD51 Associated Protein 2), that interacts strongly with RAD51. A full-length cDNA clone predicts a novel vertebrate-specific protein of 1159 residues, and the RAD51AP2 transcript was observed only in meiotic tissue (i.e. adult testis and fetal ovary), suggesting a meiotic-specific function for RAD51AP2. In HEK293 cells the interaction of RAD51 with an ectopically-expressed recombinant large fragment of RAD51AP2 requires the C-terminal 57 residues of RAD51AP2. This RAD51-binding region shows 81% homology to the C-terminus of RAD51AP1/PIR51, an otherwise totally unrelated RAD51-binding partner that is ubiquitously expressed. Analyses using truncations and point mutations in both RAD51AP1 and RAD51AP2 demonstrate that these proteins use the same structural motif for RAD51 binding. RAD54 shares some homology with this RAD51-binding motif, but this homologous region plays only an accessory role to the adjacent main RAD51-interacting region, which has been narrowed here to 40 amino acids. A novel protein, RAD51AP2, has been discovered that interacts with RAD51 through a C-terminal motif also present in RAD51AP1.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Rad51 Recombinase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , DNA Damage , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Expression , Humans , Meiosis , Mitosis , Molecular Sequence Data , RNA-Binding Proteins , Two-Hybrid System Techniques
13.
Nucleic Acids Res ; 34(9): 2833-43, 2006.
Article in English | MEDLINE | ID: mdl-16717288

ABSTRACT

In vertebrates, homologous recombinational repair (HRR) requires RAD51 and five RAD51 paralogs (XRCC2, XRCC3, RAD51B, RAD51C and RAD51D) that all contain conserved Walker A and B ATPase motifs. In human RAD51D we examined the requirement for these motifs in interactions with XRCC2 and RAD51C, and for survival of cells in response to DNA interstrand crosslinks (ICLs). Ectopic expression of wild-type human RAD51D or mutants having a non-functional A or B motif was used to test for complementation of a rad51d knockout hamster CHO cell line. Although A-motif mutants complement very efficiently, B-motif mutants do not. Consistent with these results, experiments using the yeast two- and three-hybrid systems show that the interactions between RAD51D and its XRCC2 and RAD51C partners also require a functional RAD51D B motif, but not motif A. Similarly, hamster Xrcc2 is unable to bind to the non-complementing human RAD51D B-motif mutants in co-immunoprecipitation assays. We conclude that a functional Walker B motif, but not A motif, is necessary for RAD51D's interactions with other paralogs and for efficient HRR. We present a model in which ATPase sites are formed in a bipartite manner between RAD51D and other RAD51 paralogs.


Subject(s)
Adenosine Triphosphatases/chemistry , DNA-Binding Proteins/chemistry , Recombination, Genetic , Amino Acid Motifs , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Complementation Test , Humans , Immunoprecipitation , Molecular Sequence Data , Mutation , Rad51 Recombinase/metabolism , Two-Hybrid System Techniques
14.
J Biol Chem ; 279(40): 42313-20, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15292210

ABSTRACT

The highly conserved Rad51 protein plays an essential role in repairing DNA damage through homologous recombination. In vertebrates, five Rad51 paralogs (Rad51B, Rad51C, Rad51D, XRCC2, and XRCC3) are expressed in mitotically growing cells and are thought to play mediating roles in homologous recombination, although their precise functions remain unclear. Among the five paralogs, Rad51C was found to be a central component present in two complexes, Rad51C-XRCC3 and Rad51B-Rad51C-Rad51D-XRCC2. We have shown previously that the human Rad51C protein exhibits three biochemical activities, including DNA binding, ATPase, and DNA duplex separation. Here we report the use of RNA interference to deplete expression of Rad51C protein in human HT1080 and HeLa cells. In HT1080 cells, depletion of Rad51C by small interfering RNA caused a significant reduction of frequency in homologous recombination. The level of XRCC3 protein was also sharply reduced in Rad51C-depleted HeLa cells, suggesting that XRCC3 is dependent for its stability upon heterodimerization with Rad51C. In addition, Rad51C-depleted HeLa cells showed hypersensitivity to the DNA-cross-linking agent mitomycin C and moderately increased sensitivity to ionizing radiation. Importantly, the radiosensitivity of Rad51C-deficient HeLa cells was evident in S and G(2)/M phases of the cell cycle but not in G(1) phase. Together, these results provide direct cellular evidence for the function of human Rad51C in homologous recombinational repair.


Subject(s)
DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Interphase/radiation effects , Radiation Tolerance , Recombination, Genetic , Cell Line, Tumor , DNA-Binding Proteins/physiology , Epithelial Cells , G2 Phase/radiation effects , Humans , Protein Binding , RNA, Small Interfering/pharmacology , S Phase/radiation effects
15.
Mutat Res ; 509(1-2): 49-78, 2002 Nov 30.
Article in English | MEDLINE | ID: mdl-12427531

ABSTRACT

We review the genes and proteins related to the homologous recombinational repair (HRR) pathway that are implicated in cancer through either genetic disorders that predispose to cancer through chromosome instability or the occurrence of somatic mutations that contribute to carcinogenesis. Ataxia telangiectasia (AT), Nijmegen breakage syndrome (NBS), and an ataxia-like disorder (ATLD), are chromosome instability disorders that are defective in the ataxia telangiectasia mutated (ATM), NBS, and Mre11 genes, respectively. These genes are critical in maintaining cellular resistance to ionizing radiation (IR), which kills largely by the production of double-strand breaks (DSBs). Bloom syndrome involves a defect in the BLM helicase, which seems to play a role in restarting DNA replication forks that are blocked at lesions, thereby promoting chromosome stability. The Werner syndrome gene (WRN) helicase, another member of the RecQ family like BLM, has very recently been found to help mediate homologous recombination. Fanconi anemia (FA) is a genetically complex chromosomal instability disorder involving seven or more genes, one of which is BRCA2. FA may be at least partially caused by the aberrant production of reactive oxidative species. The breast cancer-associated BRCA1 and BRCA2 proteins are strongly implicated in HRR; BRCA2 associates with Rad51 and appears to regulate its activity. We discuss in detail the phenotypes of the various mutant cell lines and the signaling pathways mediated by the ATM kinase. ATM's phosphorylation targets can be grouped into oxidative stress-mediated transcriptional changes, cell cycle checkpoints, and recombinational repair. We present the DNA damage response pathways by using the DSB as the prototype lesion, whose incorrect repair can initiate and augment karyotypic abnormalities.


Subject(s)
DNA Repair , Genetic Diseases, Inborn/complications , Recombination, Genetic , Ataxia Telangiectasia/genetics , Bloom Syndrome/genetics , Chromosome Breakage , Fanconi Anemia/genetics , Genetic Diseases, Inborn/genetics , Humans , Neoplasms/genetics , Werner Syndrome
16.
Nucleic Acids Res ; 30(4): 1009-15, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11842113

ABSTRACT

Genetic studies in rodent and chicken mutant cell lines have suggested that Rad51 paralogs (XRCC2, XRCC3, Rad51B/Rad51L1, Rad51C/Rad51L2 and Rad51D/Rad51L3) play important roles in homologous recombinational repair of DNA double-strand breaks and in maintaining chromosome stability. Previous studies using yeast two- and three-hybrid systems have shown interactions among these proteins, but it is not clear whether these interactions occur simultaneously or sequentially in vivo. By utilizing immunoprecipitation with extracts of human cells expressing epitope-tagged Rad51 paralogs, we demonstrate that XRCC2 and Rad51D, while stably interacting with each other, co-precipitate with Rad51C but not with XRCC3. In contrast, Rad51C is pulled down with XRCC3, whereas XRCC2 and Rad51D are not. In addition, Rad51B could be pulled down with Rad51C and Rad51D, but not with XRCC3. These results suggest that Rad51C is involved in two distinct in vivo complexes: Rad51B-Rad51C-Rad51D-XRCC2 and Rad51C-XRCC3. In addition, we demonstrate that Rad51 co-precipitates with XRCC3 but not with XRCC2 or Rad51D, suggesting that Rad51 can be present in an XRCC3-Rad51C-Rad51 complex. These complexes may act as functional units and serve accessory roles for Rad51 in the presynapsis stage of homologous recombinational repair.


Subject(s)
DNA-Binding Proteins/metabolism , Animals , Avian Proteins , Cell Line , Cricetinae , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gamma Rays , HeLa Cells , Humans , Macromolecular Substances , Microscopy, Fluorescence , Models, Biological , Precipitin Tests , Rad51 Recombinase , Recombinant Fusion Proteins/metabolism , Transfection
17.
Nucleic Acids Res ; 30(4): 1001-8, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11842112

ABSTRACT

Homologous recombinational repair of DNA double-strand breaks and crosslinks in human cells is likely to require Rad51 and the five Rad51 paralogs (XRCC2, XRCC3, Rad51B/Rad51L1, Rad51C/Rad51L2 and Rad51D/Rad51L3), as has been shown in chicken and rodent cells. Previously, we reported on the interactions among these proteins using baculovirus and two- and three-hybrid yeast systems. To test for interactions involving XRCC3 and Rad51C, stable human cell lines have been isolated that express (His)6-tagged versions of XRCC3 or Rad51C. Ni2+-binding experiments demonstrate that XRCC3 and Rad51C interact in human cells. In addition, we find that Rad51C, but not XRCC3, interacts directly or indirectly with Rad51B, Rad51D and XRCC2. These results argue that there are at least two complexes of Rad51 paralogs in human cells (Rad51C-XRCC3 and Rad51B-Rad51C-Rad51D-XRCC2), both containing Rad51C. Moreover, Rad51 is not found in these complexes. X-ray treatment did not alter either the level of any Rad51 paralog or the observed interactions between paralogs. However, the endogenous level of Rad51C is moderately elevated in the XRCC3-overexpressing cell line, suggesting that dimerization between these proteins might help stabilize Rad51C.


Subject(s)
DNA-Binding Proteins/metabolism , Avian Proteins , Cell Death , Cell Line , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Humans , Kinetics , Lymphocytes/metabolism , Lymphocytes/radiation effects , Macromolecular Substances , Models, Biological , Precipitin Tests , Rad51 Recombinase , Recombinant Fusion Proteins/metabolism , Transfection , X-Rays
18.
J Biol Chem ; 277(10): 8406-11, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11744692

ABSTRACT

RAD51B and RAD51C are two of five known paralogs of the human RAD51 protein that are thought to function in both homologous recombination and DNA double-strand break repair. This work describes the in vitro and in vivo identification of the RAD51B/RAD51C heterocomplex. The RAD51B/RAD51C heterocomplex was isolated and purified by immunoaffinity chromatography from insect cells co-expressing the recombinant proteins. Moreover, co-immunoprecipitation of the RAD51B and RAD51C proteins from HeLa, MCF10A, and MCF7 cells strongly suggests the existence of an endogenous RAD51B/RAD51C heterocomplex. We extended these observations to examine the interaction between the RAD51B/RAD51C complex and the other RAD51 paralogs. Immunoprecipitation using protein-specific antibodies showed that RAD51C is central to a single large protein complex and/or several smaller complexes with RAD51B, RAD51D, XRCC2, and XRCC3. However, our experiments showed no evidence for the inclusion of RAD51 within these complexes. Further analysis is required to elucidate the function of the RAD51B/RAD51C heterocomplex and its association with the other RAD51 paralogs in the processes of homologous recombination and DNA double-strand break repair.


Subject(s)
DNA-Binding Proteins/chemistry , Animals , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Chromatography , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Insecta , Precipitin Tests , Protein Binding , Rad51 Recombinase , Recombinant Proteins/metabolism , Recombination, Genetic , Tumor Cells, Cultured
19.
Phys Occup Ther Pediatr ; 12(4): 117-118, 1993.
Article in English | MEDLINE | ID: mdl-28368732

ABSTRACT

No abstract available for this article.

SELECTION OF CITATIONS
SEARCH DETAIL
...