Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Hyperthermia ; 33(3): 336-342, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27776457

RESUMO

PURPOSE: Heat shock induces DNA double-strand breaks (DSBs), but the precise mechanism of repairing heat-induced damage is unclear. Here, we investigated the DNA repair pathways involved in cell death induced by heat shock. MATERIALS AND METHODS: B02, a specific inhibitor of human RAD51 (homologous recombination; HR), and NU7026, a specific inhibitor of DNA-PK (non-homologous end-joining; NHEJ), were used for survival assays of human cancer cell lines with different p53-gene status. Mouse embryonic fibroblasts (MEFs) lacking Lig4 (NHEJ) and/or Rad54 (HR) were used for survival assays and a phosphorylated histone H2AX at Ser139 (γH2AX) assay. MEFs lacking Rad51d (HR) were used for survival assays. SPD8 cells were used to measure HR frequency after heat shock. RESULTS: Human cancer cells were more sensitive to heat shock in the presence of B02 despite their p53-gene status, and the effect of B02 on heat sensitivity was specific to the G2 phase. Rad54-deficient MEFs were sensitive to heat shock and showed prolonged γH2AX signals following heat shock. Rad51d-deficient MEFs were also sensitive to heat shock. Moreover, heat shock-stimulated cells had increased HR. CONCLUSIONS: The HR pathway plays an important role in the survival of mammalian cells against death induced by heat shock via the repair of heat-induced DNA DSBs.

3.
Nucleic Acids Res ; 39(1): 132-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20813759

RESUMO

DNA double-stranded breaks (DSBs) are among the most severe forms of DNA damage and responsible for chromosomal translocations that may lead to gene fusions. The RAD51 family plays an integral role in preserving genome stability by homology directed repair of DSBs. From a proteomics screen, we recently identified SFPQ/PSF as an interacting partner with the RAD51 paralogs, RAD51D, RAD51C and XRCC2. Initially discovered as a potential RNA splicing factor, SFPQ was later shown to have homologous recombination and non-homologous end joining related activities and also to bind and modulate the function of RAD51. Here, we demonstrate that SFPQ interacts directly with RAD51D and that deficiency of both proteins confers a severe loss of cell viability, indicating a synthetic lethal relationship. Surprisingly, deficiency of SFPQ alone also leads to sister chromatid cohesion defects and chromosome instability. In addition, SFPQ was demonstrated to mediate homology directed DNA repair and DNA damage response resulting from DNA crosslinking agents, alkylating agents and camptothecin. Taken together, these data indicate that SFPQ association with the RAD51 protein complex is essential for homologous recombination repair of DNA damage and maintaining genome integrity.


Assuntos
Cromátides/ultraestrutura , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas de Ligação a RNA/fisiologia , Alquilantes/toxicidade , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Instabilidade Cromossômica , Reagentes de Ligações Cruzadas/toxicidade , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Epistasia Genética , Deleção de Genes , Humanos , Camundongos , Mitomicina/toxicidade , Fator de Processamento Associado a PTB , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
4.
Oncotarget ; 11(22): 2026-2036, 2020 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-32547701

RESUMO

Ulcerative colitis has a significant impact on the quality of life for the patients, and can substantially increase the risk of colon cancer in patients suffering long-term. Conventional treatments provide only modest relief paired with a high risk of side effects, while complementary and alternative medicines can offer safe and effective options. Over the past decade, we have shown that both American ginseng and its hexane fraction (HAG) have anti-oxidant and anti-inflammatory properties that can suppress mouse colitis and prevent colitis-associated colon cancer. With the goal of isolating a single active compound, we further fractionated HAG, and found the most abundant molecule in this fraction was the polyacetylene, panaxynol (PA). After isolating and characterizing PA, we tested the efficacy of PA in the treatment and prevention of colitis in mice and studied the mechanism of action. We demonstrate here that PA effectively treats colitis in a Dextran Sulfate Sodium mouse model by targeting macrophages for DNA damage and apoptosis. This study provides additional mechanistic evidence that American ginseng can be used for conventional treatment of colitis and other diseases associated with macrophage dysfunction.

5.
Proteomics ; 9(16): 4071-86, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19658102

RESUMO

The RAD51 family of proteins is involved in homologous recombination (HR) DNA repair and maintaining chromosome integrity. To identify candidates that interact with HR proteins, the mouse RAD51C, RAD51D and XRCC2 proteins were purified using bacterial expression systems and each of them used to co-precipitate interacting partners from mouse embryonic fibroblast cellular extracts. Mass spectroscopic analysis was performed on protein bands obtained after 1-D SDS-PAGE of co-precipitation eluates from cell extracts of mitomycin C treated and untreated mouse embryonic fibroblasts. Profiling of the interacting proteins showed a clear bias toward nucleic acid binding and modification proteins. Interactions of four candidate proteins (SFPQ, NONO, MSH2 and mini chromosome maintenance protein 2) were confirmed by Western blot analysis of co-precipitation eluates and were also verified to form ex vivo complexes with RAD51D. Additional interacting proteins were associated with cell division, embryo development, protein and carbohydrate metabolism, cellular trafficking, protein synthesis, modification or folding, and cell structure or motility functions. Results from this study are an important step toward identifying interacting partners of the RAD51 paralogs and understanding the functional diversity of proteins that assist or regulate HR repair mechanisms.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteômica/métodos , Rad51 Recombinase/metabolismo , Animais , Western Blotting , Proteínas de Ligação a DNA/genética , Imunoprecipitação , Camundongos , Ligação Proteica , Rad51 Recombinase/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
6.
Dev Biol ; 319(2): 179-91, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18501887

RESUMO

Neural crest-derived structures that depend critically upon expression of the basic helix-loop-helix DNA binding protein Hand2 for normal development include craniofacial cartilage and bone, the outflow tract of the heart, cardiac cushion, and noradrenergic sympathetic ganglion neurons. Loss of Hand2 is embryonic lethal by E9.5, obviating a genetic analysis of its in-vivo function. We have overcome this difficulty by specific deletion of Hand2 in neural crest-derived cells by crossing our line of floxed Hand2 mice with Wnt1-Cre transgenic mice. Our analysis of Hand2 knock-out in neural crest-derived cells reveals effects on development in all neural crest-derived structures where Hand2 is expressed. In the autonomic nervous system, conditional disruption of Hand2 results in a significant and progressive loss of neurons as well as a significant loss of TH expression. Hand2 affects generation of the neural precursor pool of cells by affecting both the proliferative capacity of the progenitors as well as affecting expression of Phox2a and Gata3, DNA binding proteins important for the cell autonomous development of noradrenergic neurons. Our data suggest that Hand2 is a multifunctional DNA binding protein affecting differentiation and cell type-specific gene expression in neural crest-derived noradrenergic sympathetic ganglion neurons. Hand2 has a pivotal function in a non-linear cross-regulatory network of DNA binding proteins that affect cell autonomous control of differentiation and cell type-specific gene expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Deleção de Genes , Crista Neural/fisiologia , Neurônios/fisiologia , Células Ganglionares da Retina/fisiologia , Sistema Nervoso Simpático/embriologia , Animais , Regulação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Confocal , Sistema Nervoso Simpático/fisiologia , Transcrição Gênica
7.
BMC Mol Biol ; 10: 27, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19327148

RESUMO

BACKGROUND: The homologous recombination (HR) pathway is vital for maintaining genomic integrity through the restoration of double-stranded breaks and interstrand crosslinks. The RAD51 paralogs (RAD51B, RAD51C, RAD51D, XRCC2, XRCC3) are essential for this process in vertebrates, and the RAD51D paralog is unique in that it participates in both HR repair and telomere maintenance. RAD51D is also known to directly interact with the RAD51C and XRCC2 proteins. Rad51d splice variants have been reported in mouse and human tissues, supportive of a role for alternative splicing in HR regulation. The present study evaluated the interaction of the Rad51d splice isoform products with RAD51C and XRCC2 and their expression patterns. RESULTS: Yeast-2-hybrid analysis was used to determine that the Mus musculus Rad51d splice variant product RAD51DDelta7b (deleted for residues 219 through 223) was capable of interacting with both RAD51C and XRCC2 and that RAD51D+int3 interacted with XRCC2. In addition, the linker region (residues 54 through 77) of RAD51D was identified as a region that potentially mediates binding with XRCC2. Cellular localization, detected by EGFP fusion proteins, demonstrated that each of the splice variant products tested was distributed throughout the cell similar to the full-length protein. However, none of the splice variants were capable of restoring resistance of Rad51d-deficient cell lines to mitomycin C. RT-PCR expression analysis revealed that Rad51dDelta3 (deleted for exon 3) and Rad51dDelta5 (deleted for exon 5)transcripts display tissue specific expression patterns with Rad51dDelta3 being detected in each tissue except ovary and Rad51dDelta5 not detected in mammary gland and testis. These expression studies also led to the identification of two additional Rad51d ubiquitously expressed transcripts, one deleted for both exon 9 and 10 and one deleted for only exon 10. CONCLUSION: These results suggest Rad51d alternative splice variants potentially modulate mechanisms of HR by sequestering either RAD51C or XRCC2.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Proteínas de Ligação a DNA/metabolismo , Teste de Complementação Genética , Camundongos , Isoformas de Proteínas/química , Estrutura Terciária de Proteína , Rad51 Recombinase/metabolismo , Recombinação Genética/fisiologia , Deleção de Sequência
8.
Mol Carcinog ; 48(7): 586-91, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19347880

RESUMO

RAD51D, a paralog of the mammalian RAD51 gene, contributes towards maintaining genomic integrity by homologous recombination DNA repair and telomere maintenance. A RAD51D variant, E233G, was initially identified as a potential susceptibility allele in high-risk, site-specific, familial breast cancer. We describe in this report that the Rad51d (E233G) genetic variant confers increased cisplatin resistance and cell growth phenotypes in human breast carcinoma cell lines with a mutant p53 gene (BT20 and T47D) but not with a wild-type p53 gene (MCF-7). Treatment with a p53 specific inhibitor, pifithrin alpha, restored this resistant phenotype in the MCF-7 cell line. Additionally, Rad51d (E233G) conferred increased cisplatin resistance of an MCF7 cell line in which p53 expression was stably knocked down by shRNAp53, indicating that the effect of this variant is dependent upon p53 status. Further study of Rad51d (E233G) will provide mechanistic insight towards the role of RAD51D in cellular response to anticancer agents and as a potential target for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Cisplatino/farmacologia , Proteínas de Ligação a DNA/genética , Genes p53 , Sequência de Bases , Western Blotting , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Primers do DNA , Resistencia a Medicamentos Antineoplásicos , Humanos
9.
Methods Mol Biol ; 1999: 209-215, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31127578

RESUMO

A complementation assay was developed to determine whether alleles of DNA repair genes are necessary for repairing specific types of damage. The assay was established by measuring the resistance capacity of Rad51d-deficient mouse embryonic fibroblasts (MEFs) transfected with mammalian expression constructs. Here, we describe the methods used to assess colony survival following the treatment of transfected cells with genotoxic compounds. This approach provides a time-efficient and stringent strategy to screen genetic alleles for identifying regions or specific amino acid residues critical for function or regulation of DNA repair pathways.


Assuntos
Bioensaio/métodos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , Fibroblastos/efeitos dos fármacos , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Proteínas de Ligação a DNA/deficiência , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Higromicina B/toxicidade , Camundongos , Mitomicina/toxicidade , Transfecção
10.
Methods Mol Biol ; 1999: 3-29, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31127567

RESUMO

Double-strand DNA breaks (DSBs) are generated by ionizing radiation and as intermediates during the processing of DNA, such as repair of interstrand cross-links and collapsed replication forks. These potentially deleterious DSBs are repaired primarily by the homologous recombination (HR) and nonhomologous end joining (NHEJ) DNA repair pathways. HR utilizes a homologous template to accurately restore damaged DNA, whereas NHEJ utilizes microhomology to join breaks in close proximity. The pathway available for DSB repair is dependent upon the cell cycle stage; for example, HR primarily functions during the S/G2 stages while NHEJ can repair DSBs at any cell cycle stage. Posttranslational modifications (PTMs) promote activity of specific pathways and subpathways through enzyme activation and precisely timed protein recruitment and degradation. This chapter provides an overview of PTMs occurring during DSB repair. In addition, clinical phenotypes associated with HR-defective cancers, such as mutational signatures used to predict response to poly(ADP-ribose) polymerase inhibitors, are discussed. Understanding these processes will provide insight into mechanisms of genome maintenance and likely identify targets and new avenues for therapeutic interventions.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Reparo de DNA por Recombinação/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/genética , Humanos , Mutação , Neoplasias/genética , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Reparo de DNA por Recombinação/genética
11.
Environ Mol Mutagen ; 59(1): 38-48, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28945288

RESUMO

Thiopurines are part of a clinical regimen used for the treatment of autoimmune disorders and childhood acute lymphoblastic leukemia. However, despite these successes, there are also unintended consequences such as therapy-induced cancer in long-term survivors. Therefore, a better understanding of cellular responses to thiopurines will lead to improved and personalized treatment strategies. RAD51D is an important component of homologous recombination (HR), and our previous work established that mammalian cells defective for RAD51D are more sensitive to the thiopurine 6-thioguanine (6TG) and have dramatically increased numbers of multinucleated cells and chromosome instability. 6TG is capable of being incorporated into telomeres, and interestingly, RAD51D contributes to telomere maintenance, although the precise function of RAD51D at the telomeres remains unclear. We sought here to investigate: (1) the activity of RAD51D at telomeres, (2) the contribution of RAD51D to protect against 6TG-induced telomere damage, and (3) the fates of Rad51d-deficient cells following 6TG treatment. These results demonstrate that RAD51D is required for maintaining the telomeric 3' overhangs. As measured by γ-H2AX induction and foci formation, 6TG induced DNA damage in Rad51d-proficient and Rad51d-deficient cells. However, the extent of γ-H2AX telomere localization following 6TG treatment was higher in Rad51d-deficient cells than in Rad51d-proficient cells. Using live-cell imaging of 6TG-treated Rad51d-deficient cells, two predominant forms of mitotic catastrophe were found to contribute to the formation of multinucleated cells, failed division and restitution. Collectively, these findings provide a unique window into the role of the RAD51D HR protein during thiopurine induction of mitotic catastrophe. Environ. Mol. Mutagen. 59:38-48, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Proteínas de Ligação a DNA/deficiência , Recombinação Homóloga/efeitos dos fármacos , Mitose/efeitos dos fármacos , Tioguanina/farmacologia , Animais , Linhagem Celular , Instabilidade Cromossômica/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Camundongos , Telômero/efeitos dos fármacos
12.
Cancer Res ; 65(6): 2089-96, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781618

RESUMO

Homologous recombination is a double-strand break repair pathway required for resistance to DNA damage and maintaining genomic integrity. In mitotically dividing vertebrate cells, the primary proteins involved in homologous recombination repair are RAD51 and the five RAD51 paralogs, RAD51B, RAD51C, RAD51D, XRCC2, and XRCC3. In the absence of Rad51d, human and mouse cells fail to proliferate, and mice defective for Rad51d die before birth, likely as a result of genomic instability and p53 activation. Here, we report that a p53 deletion is sufficient to extend the life span of Rad51d-deficient embryos by up to 6 days and rescue the cell lethal phenotype. The Rad51d-/- Trp53-/- mouse embryo-derived fibroblasts were sensitive to DNA-damaging agents, particularly interstrand cross-links, and exhibited extensive chromosome instability including aneuploidy, chromosome fragments, deletions, and complex rearrangements. Additionally, loss of Rad51d resulted in increased centrosome fragmentation and reduced levels of radiation-induced RAD51-focus formation. Spontaneous frequencies of sister chromatid exchange were not affected by the absence of Rad51d, but sister chromatid exchange frequencies did fail to be induced upon challenge with the DNA cross-linking agent mitomycin C. These findings support a crucial role for mammalian RAD51D in normal development, recombination, and maintaining mammalian genome stability.


Assuntos
Instabilidade Cromossômica , Proteínas de Ligação a DNA/deficiência , Animais , Técnicas de Cultura de Células , Centrossomo , Cisplatino/farmacologia , Dano ao DNA , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/fisiologia , Metanossulfonato de Metila/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mitomicina/farmacologia , Troca de Cromátide Irmã , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
13.
DNA Repair (Amst) ; 42: 82-93, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27161866

RESUMO

The RAD51 family is integral for homologous recombination (HR) mediated DNA repair and maintaining chromosome integrity. RAD51D, the fourth member of the family, is a known ovarian cancer susceptibility gene and required for the repair of interstrand crosslink DNA damage and preserving chromosomal stability. In this report, we describe the RNF138 E3 ubiquitin ligase that interacts with and ubiquitinates the RAD51D HR protein. RNF138 is a member of an E3 ligase family that contains an amino-terminal RING finger domain and a putative carboxyl-terminal ubiquitin interaction motif. In mammalian cells, depletion of RNF138 increased the stability of the RAD51D protein, suggesting that RNF138 governs ubiquitin-proteasome-mediated degradation of RAD51D. However, RNF138 depletion conferred sensitivity to DNA damaging agents, reduced RAD51 focus formation, and increased chromosomal instability. Site-specific mutagenesis of the RNF138 RING finger domain demonstrated that it was necessary for RAD51D ubiquitination. Presence of RNF138 also enhanced the interaction between RAD51D and a known interacting RAD51 family member XRCC2 in a yeast three-hybrid assay. Therefore, RNF138 is a newly identified regulatory component of the HR mediated DNA repair pathway that has implications toward understanding how ubiquitination modifies the functions of the RAD51 paralog protein complex.


Assuntos
Cromossomos Humanos/genética , Reparo do DNA , Rad51 Recombinase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Células HeLa , Recombinação Homóloga , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Proteólise , Ubiquitinação
14.
J Med Chem ; 58(1): 433-42, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25454794

RESUMO

The cyclin groove is an important recognition site for substrates of the cell cycle cyclin dependent kinases and provides an opportunity for highly selective inhibition of kinase activity through a non-ATP competitive mechanism. The key peptide residues of the cyclin binding motif have been studied in order to precisely define the structure-activity relationship for CDK kinase inhibition. Through this information, new insights into the interactions of peptide CDK inhibitors with key subsites of the cyclin binding groove provide for the replacement of binding determinants with more druglike functionality through REPLACE, a strategy for the iterative conversion of peptidic blockers of protein-protein interactions into pharmaceutically relevant compounds. As a result, REPLACE is further exemplified in combining optimized peptidic sequences with effective N-terminal capping groups to generate more stable compounds possessing antitumor activity consistent with on-target inhibition of cell cycle CDKs. The compounds described here represent prototypes for a next generation of kinase therapeutics with high efficacy and kinome selectivity, thus avoiding problems observed with first generation CDK inhibitors.


Assuntos
Antineoplásicos/química , Quinases Ciclina-Dependentes/química , Ciclinas/química , Peptídeos/química , Inibidores de Proteínas Quinases/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Peptídeos/metabolismo , Peptídeos/farmacologia , Ligação Proteica , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
15.
Mol Cancer Res ; 9(2): 206-14, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21205838

RESUMO

Thiopurines are extensively used as immunosuppressants and in the treatment of childhood cancers, even though there is concern about therapy-induced leukemias and myelodysplastic syndromes resulting from thiopurine use. Following metabolic activation, thiopurines are incorporated into DNA and invoke mismatch repair (MMR). Recognition of 6-thioguanine (6-thioG) in DNA by key MMR proteins results in cell death rather than repair. There are suggestions that homologous recombination (HR) is involved downstream of MMR following thiopurine treatment, but the precise role of HR is poorly understood. In this study, we demonstrate that cells deficient in RAD51D (a RAD51 paralogue) are extremely sensitive to 6-thioG. This sensitivity is almost completely rescued by the deletion of Mlh1, which suggests that HR is involved in the repair of the 6-thioG-induced recombinogenic lesions generated by MMR. Furthermore, 6-thioG induces chromosome aberrations in the Rad51d-deficient cells. Interestingly, Rad51d-deficient cells show a striking increase in the frequency of triradial and quadriradial chromosomes in response to 6-thioG therapy. The presence of these chromatid exchange-type aberrations indicates that the deficiency in RAD51D-dependent HR results in profound chromosomal damage precipitated by the processing of 6-thioG by MMR. The radials are notable as an important source of chromosomal translocations, which are the most common class of mutations found in hematologic malignancies. This study thus suggests that HR insufficiency could be a potential risk factor for the development of secondary cancers that result from long-term use of thiopurines in patients.


Assuntos
Dano ao DNA , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Recombinação Genética/genética , Tioguanina/farmacologia , Animais , Linhagem Celular , Instabilidade Cromossômica/efeitos dos fármacos , DNA/metabolismo , Proteínas de Ligação a DNA/deficiência , Fase G2/efeitos dos fármacos , Células Gigantes/efeitos dos fármacos , Células Gigantes/patologia , Camundongos , Recombinação Genética/efeitos dos fármacos
16.
DNA Repair (Amst) ; 9(4): 458-67, 2010 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-20133210

RESUMO

S(N)1-type methylating agents generate O(6)-methyl guanine (O(6)-meG), which is a potently mutagenic, toxic, and recombinogenic DNA adduct. Recognition of O(6)-meG:T mismatches by mismatch repair (MMR) causes sister chromatid exchanges, which are representative of homologous recombination (HR) events. Although the MMR-dependent mutagenicity and toxicity caused by O(6)-meG has been studied, the mechanisms of recombination induced by O(6)-meG are poorly understood. To explore the HR and MMR genetic interactions in mammals, we used the Rad51d and Mlh1 mouse models. Ablation of Mlh1 did not appreciably influence the developmental phenotypes conferred by the absence of Rad51d. Mouse embryonic fibroblasts (MEFs) deficient in Rad51d can only proliferate in p53-deficient background. Therefore, Rad51d(-/-)Mlh1(-/-)Trp53(-/-) MEFs with a combined deficiency of HR and MMR were generated and comparisons between MLH1 and RAD51D status were made. To our knowledge, these MEFs are the first mammalian model system for combined HR and MMR defects. Rad51d-deficient MEFs were 5.3-fold sensitive to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) compared to the Rad51d-proficient MEFs. A pronounced G2/M arrest in Rad51d-deficient cells was accompanied by an accumulation of gamma-H2AX and apoptosis. Mlh1-deficient MEFs were resistant to MNNG and showed no G2/M arrest or apoptosis at the doses used. Importantly, loss of Mlh1 alleviated sensitivity of Rad51d-deficient cells to MNNG, in addition to reducing gamma-H2AX, G2/M arrest and apoptosis. Collectively, the data support the hypothesis that MMR-dependent sensitization of HR-deficient cells is specific for O(6)-meG and suggest that HR resolves DNA intermediates created by MMR recognition of O(6)-meG:T. This study provides insight into recombinogenic mechanisms of carcinogenesis and chemotherapy resulting from O(6)-meG adducts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/fisiologia , Guanina/análogos & derivados , Proteínas Nucleares/metabolismo , Rad51 Recombinase/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Fase G2 , Guanina/metabolismo , Metilnitronitrosoguanidina/toxicidade , Camundongos , Proteína 1 Homóloga a MutL , Proteínas Nucleares/genética , Rad51 Recombinase/genética
17.
Pharmacogenet Genomics ; 19(2): 153-60, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19033885

RESUMO

BACKGROUND AND OBJECTIVE: RAD51D, a paralog of the mammalian RAD51 gene, is an important component for DNA repair and telomere maintenance. A RAD51D variant, E233G, was initially identified as a potential susceptibility allele in high-risk, site-specific, familial breast cancer. We describe in this report, the effects of this amino acid change on RAD51D protein interaction and function. METHODS AND RESULTS: To examine the effect of the variant on cellular resistance to DNA damage, a complementation analysis by using Rad51d-deficient mouse embryonic fibroblasts was performed. Results indicated that the E233G variant actually increased the cellular resistance to the DNA-damaging agents, mitomycin C, cisplatin, methyl methane sulfonate, and ultraviolet light as well as to taxol. In addition, the E233G variant reduced the anaphase bridge index, a telomere dysfunction correlate, and conferred increased cellular proliferation, suggesting that the E to G substitution may affect telomere function. Yeast two-hybrid analyses demonstrated that interaction between RAD51C and RAD51D (E233G) was decreased by two fold, whereas normal levels of interaction between XRCC2 and the variant were maintained. Molecular modeling suggested that the glutamic acid-233 forms a salt bridge with lysine-23 in the N-terminal domain of RAD51D, and the glycine substitution may disrupt an interdomain interaction. CONCLUSION: Our findings suggest that the E233G variant affects RAD51D functions and protein interactions and increases cellular chemoresistance. This study is the first to analyze the functional effects of a clinically relevant RAD51D amino acid substitution. Further study of this variant will provide mechanistic insight into the role of RAD51D in cellular response to anticancer agents and as a molecular target for cancer therapy.


Assuntos
Proteínas de Ligação a DNA/genética , Variação Genética , Substituição de Aminoácidos/genética , Animais , Proliferação de Células , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Deleção de Genes , Camundongos , Modelos Moleculares , Conformação Proteica , Técnicas do Sistema de Duplo-Híbrido
18.
Biochem Pharmacol ; 76(6): 697-706, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18599024

RESUMO

The synthesis of thymidylate (TMP) occupies a convergence of two critical metabolic pathways: folate metabolism and pyrimidine biosynthesis. Thymidylate is formed from deoxyuridylate (dUMP) using N(5),N(10)-methylene tetrahydrofolate. The metabolic relationship between dUMP, TMP, and folate has been the subject of cancer research from prevention to chemotherapy. Thymidylate stress is induced by nutritional deficiency of folic acid, defects in folate metabolism, and by antifolate and fluoropyrimidine chemotherapeutics. Both classes of chemotherapeutics remain mainstay treatments against solid tumors. Because of the close relationship between dUMP and TMP, thymidylate stress is associated with increased incorporation of uracil into DNA. Genomic uracil is removed by uracil DNA glycosylases of base excision repair (BER). Unfortunately, BER is apparently problematic during thymidylate stress. Because BER requires a DNA resynthesis step, elevated dUTP causes reintroduction of genomic uracil. BER strand break intermediates are clastogenic if not repaired. Thus, BER during thymidylate stress appears to cause genome instability, yet might also contribute to the mechanism of action for antifolates and fluoropyrimidines. However, the precise roles of BER and its components during thymidylate stress remain unclear. In particular, links between BER and downstream events remain poorly defined, including damage signaling pathways and homologous recombination (HR). Evidence is growing that HR responds to persistent BER strand break intermediates and DNA damage signaling pathways mediate cross talk between BER and HR. Examination of crosstalk among BER, HR, and damage signaling may shed light on decades of investigation and provide insight for development of novel chemopreventive and chemotherapeutic approaches.


Assuntos
Antineoplásicos/metabolismo , DNA/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Uracila/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Deficiência de Ácido Fólico/tratamento farmacológico , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Humanos , Neoplasias/genética
19.
Results Probl Cell Differ ; 42: 343-67, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16903217

RESUMO

Meiosis is a unique form of cellular division by which a diploid cell produces genetically distinct haploid gametes. Initiation and regulation of mammalian meiosis differs between the sexes. In females, meiosis is initiated during embryo development and arrested shortly after birth during prophase I. In males, spermatogonial stem cells initiate meiosis at puberty and proceed through gametogenesis with no cell cycle arrest. Mouse genes required for early meiotic cell cycle events are being identified by comparative analysis with other eukaryotic systems, by virtue of gene knockout technology and by mouse mutagenesis screens for reproductive defects. This review focuses on mouse reproductive biology and describes the available mouse mutants with defects in the early meiotic cell cycle and prophase I regulatory events. These research tools will permit rapid advances in such medically relevant research areas as infertility, embryo lethality and developmental abnormalities.


Assuntos
Ciclo Celular , Regulação da Expressão Gênica , Células Germinativas/citologia , Animais , Feminino , Humanos , Masculino , Mamíferos , Meiose , Modelos Biológicos , Oócitos/citologia , Espermatozoides/citologia , Células-Tronco/citologia
20.
Chem Res Toxicol ; 19(12): 1580-94, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17173371

RESUMO

The chemical methylating agents methylmethane sulfonate (MMS) and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) have been used for decades as classical DNA damaging agents. These agents have been utilized to uncover and explore pathways of DNA repair, DNA damage response, and mutagenesis. MMS and MNNG modify DNA by adding methyl groups to a number of nucleophilic sites on the DNA bases, although MNNG produces a greater percentage of O-methyl adducts. There has been substantial progress elucidating direct reversal proteins that remove methyl groups and base excision repair (BER), which removes and replaces methylated bases. Direct reversal proteins and BER, thus, counteract the toxic, mutagenic, and clastogenic effects of methylating agents. Despite recent progress, the complexity of DNA damage responses to methylating agents is still being discovered. In particular, there is growing understanding of pathways such as homologous recombination, lesion bypass, and mismatch repair that react when the response of direct reversal proteins and BER is insufficient. Furthermore, the importance of proper balance within the steps in BER has been uncovered with the knowledge that DNA structural intermediates during BER are deleterious. A number of issues complicate the elucidation of the downstream responses when direct reversal is insufficient or BER is imbalanced. These include inter-species differences, cell-type-specific differences within mammals and between cancer cell lines, and the type of methyl damage or BER intermediate encountered. MMS also carries a misleading reputation of being a radiomimetic, that is, capable of directly producing strand breaks. This review focuses on the DNA methyl damage caused by MMS and MNNG for each site of potential methylation to summarize what is known about the repair of such damage and the downstream responses and consequences if the damage is not repaired.


Assuntos
Quebras de DNA , Metilação de DNA/efeitos dos fármacos , Reparo do DNA , Animais , Humanos , Metanossulfonato de Metila/farmacologia , Metilnitronitrosoguanidina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA