Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.226
Filtrar
1.
Oncotarget ; 15: 699-713, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39352803

RESUMO

Monoclonal antibody therapies for cancer have demonstrated extraordinary clinical success in recent years. However, these strategies are thus far mostly limited to specific cell surface antigens, even though many disease targets are found intracellularly. Here we report studies on the humanization of a full-length, nucleic acid binding, monoclonal lupus-derived autoantibody, 3E10, which exhibits a novel mechanism of cell penetration and tumor specific targeting. Comparing humanized variants of 3E10, we demonstrate that cell uptake depends on the nucleoside transporter ENT2, and that faster cell uptake and superior in vivo tumor targeting are associated with higher affinity nucleic acid binding. We show that one human variant retains the ability of the parental 3E10 to bind RAD51, serving as a synthetically lethal inhibitor of homology-directed repair in vitro. These results provide the basis for the rational design of a novel antibody platform for therapeutic tumor targeting with high specificity following systemic administration.


Assuntos
Rad51 Recombinase , Humanos , Animais , Rad51 Recombinase/antagonistas & inibidores , Rad51 Recombinase/metabolismo , Rad51 Recombinase/imunologia , Camundongos , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Peptídeos Penetradores de Células/farmacologia , Peptídeos Penetradores de Células/química
2.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(5): 1323-1333, 2024 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-39479812

RESUMO

OBJECTIVE: To investigate the mechanism of DNA damage and repair in MLL -rearranged acute myeloid leukemia( MLL-r AML)cells by the combination of Chidamide and the BRD4 inhibitor(+)-JQ-1. METHODS: MLL-r AML cell lines Molm-13, MV4-11 and non- MLL-r AML cell line Kasumi were divided into control group(contr), Chidamide group(chida), (+)-JQ-1 group and Combination group(combi), respectively. Cell viability of Molm-13 was measured by CCK-8 to determine optimal the concentrations of Chidamide and(+)-JQ-1. The cell cycle was detected by flow cytometry, and apoptosis-related factors Bcl-2, Bax and caspase-3 were detected by Western blot. DNA damage marker γH2AX was detected by immunofluorescence. The protein expressions of DNA damage factor γH2AX, DNA damage checkpoint kinases p-ATR, p-CHK1, p-ATM, p-CHK2 and DNA damage repair factors Rad51 and 53BP1 were detected by Western blot. The expression of DNA damage repair factors Rad51 and 53BP1 mRNA was detected by qRT-PCR. RESULTS: Under the treatment of Chidamide (300 nmol/L) and (+)-JQ-1 (400 nmol/L), the proportion of G1 phase cells in MLL-r AML cell lines Molm-13 and MV4-11 was increased in combination group compared with control group. In non- MLL-r AML cell line Kasumi, compared with control group, the proportion of G1 phase cells in combination group was increased (P < 0.05). In Molm-13 and MV4-11 cell lines, compared with control group, the expression level of DNA damage marker γH2AX in combination group was increased (P < 0.05). The expression levels of DNA damage checkpoint and damage repair factors p-ATR, p-CHK1, p-ATM, p-CHK2, Rad51, 53BP1 were decreased (P < 0.05). In Kasumi cell line, compared with control group, there was no significant change in the expression of some of the above factors in combination group (P >0.05), but the expression trend of some factors was opposite. In MLL-r AML cell lines Molm-13 and MV4-11, compared with control group, the expression levels of Bax and caspase-3 protein were increased in combination group, while the expression levels of Bcl-2 protein were decreased (P < 0.05). In non- MLL-r AML cell line Kasumi, there was no significant change in apoptotic factor protein expression in combination group compared with control group (P >0.05). CONCLUSION: Chidamide combined with (+)-JQ-1 can inhibit the proliferation of MLL-r AML cells, inhibit the initiation of protective self-repair of these leukemia cells by inhibiting the DNA damage response pathway, and ultimately increase the apoptosis of these cells, but non- MLL-r AML cells have no similar results.


Assuntos
Apoptose , Dano ao DNA , Leucemia Mieloide Aguda , Proteína de Leucina Linfoide-Mieloide , Humanos , Linhagem Celular Tumoral , Proteína de Leucina Linfoide-Mieloide/genética , Apoptose/efeitos dos fármacos , Rad51 Recombinase/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fatores de Transcrição/metabolismo , Rearranjo Gênico , Ciclo Celular , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA , Histona-Lisina N-Metiltransferase
3.
Nature ; 634(8033): 492-500, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39261728

RESUMO

DNA double-strand break (DSB) repair by homologous recombination is initiated by DNA end resection, a process involving the controlled degradation of the 5'-terminated strands at DSB sites1,2. The breast cancer suppressor BRCA1-BARD1 not only promotes resection and homologous recombination, but it also protects DNA upon replication stress1,3-9. BRCA1-BARD1 counteracts the anti-resection and pro-non-homologous end-joining factor 53BP1, but whether it functions in resection directly has been unclear10-16. Using purified recombinant proteins, we show here that BRCA1-BARD1 directly promotes long-range DNA end resection pathways catalysed by the EXO1 or DNA2 nucleases. In the DNA2-dependent pathway, BRCA1-BARD1 stimulates DNA unwinding by the Werner or Bloom helicase. Together with MRE11-RAD50-NBS1 and phosphorylated CtIP, BRCA1-BARD1 forms the BRCA1-C complex17,18, which stimulates resection synergistically to an even greater extent. A mutation in phosphorylated CtIP (S327A), which disrupts its binding to the BRCT repeats of BRCA1 and hence the integrity of the BRCA1-C complex19-21, inhibits resection, showing that BRCA1-C is a functionally integrated ensemble. Whereas BRCA1-BARD1 stimulates resection in DSB repair, it paradoxically also protects replication forks from unscheduled degradation upon stress, which involves a homologous recombination-independent function of the recombinase RAD51 (refs. 4-6,8). We show that in the presence of RAD51, BRCA1-BARD1 instead inhibits DNA degradation. On the basis of our data, the presence and local concentration of RAD51 might determine the balance between the pronuclease and the DNA protection functions of BRCA1-BARD1 in various physiological contexts.


Assuntos
Proteína BRCA1 , Quebras de DNA de Cadeia Dupla , DNA , Reparo de DNA por Recombinação , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Humanos , Proteína BRCA1/química , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , DNA/química , DNA/genética , DNA/metabolismo , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/química , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/metabolismo , Fosforilação , Ligação Proteica , Rad51 Recombinase/metabolismo , RecQ Helicases , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Helicase da Síndrome de Werner , Proteína Homóloga a MRE11/metabolismo , Proteínas de Ciclo Celular/metabolismo
4.
Nature ; 634(8033): 482-491, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39261729

RESUMO

The licensing step of DNA double-strand break repair by homologous recombination entails resection of DNA ends to generate a single-stranded DNA template for assembly of the repair machinery consisting of the RAD51 recombinase and ancillary factors1. DNA end resection is mechanistically intricate and reliant on the tumour suppressor complex BRCA1-BARD1 (ref. 2). Specifically, three distinct nuclease entities-the 5'-3' exonuclease EXO1 and heterodimeric complexes of the DNA endonuclease DNA2, with either the BLM or WRN helicase-act in synergy to execute the end resection process3. A major question concerns whether BRCA1-BARD1 directly regulates end resection. Here, using highly purified protein factors, we provide evidence that BRCA1-BARD1 physically interacts with EXO1, BLM and WRN. Importantly, with reconstituted biochemical systems and a single-molecule analytical tool, we show that BRCA1-BARD1 upregulates the activity of all three resection pathways. We also demonstrate that BRCA1 and BARD1 harbour stand-alone modules that contribute to the overall functionality of BRCA1-BARD1. Moreover, analysis of a BARD1 mutant impaired in DNA binding shows the importance of this BARD1 attribute in end resection, both in vitro and in cells. Thus, BRCA1-BARD1 enhances the efficiency of all three long-range DNA end resection pathways during homologous recombination in human cells.


Assuntos
Proteína BRCA1 , Quebras de DNA de Cadeia Dupla , Exodesoxirribonucleases , Recombinação Homóloga , RecQ Helicases , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Humanos , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , DNA/metabolismo , DNA/genética , DNA Helicases , Reparo do DNA , Enzimas Reparadoras do DNA , DNA de Cadeia Simples/metabolismo , Exodesoxirribonucleases/metabolismo , Ligação Proteica , Rad51 Recombinase/metabolismo , Reparo de DNA por Recombinação , RecQ Helicases/metabolismo , RecQ Helicases/genética , Imagem Individual de Molécula , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima , Helicase da Síndrome de Werner/metabolismo , Helicase da Síndrome de Werner/genética
5.
Nucleic Acids Res ; 52(19): 11738-11752, 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-39268578

RESUMO

Homologous recombination (HR) factors are crucial for DSB repair and processing stalled replication forks. RAD51 paralogs, including RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3, have emerged as essential tumour suppressors, forming two subcomplexes, BCDX2 and CX3. Mutations in these genes are associated with cancer susceptibility and Fanconi anaemia, yet their biochemical activities remain unclear. This study reveals a linear arrangement of BCDX2 subunits compared to the RAD51 ring. BCDX2 shows a strong affinity towards single-stranded DNA (ssDNA) via unique binding mechanism compared to RAD51, and a contribution of DX2 subunits in binding branched DNA substrates. We demonstrate that BCDX2 facilitates RAD51 loading on ssDNA by suppressing the cooperative requirement of RAD51 binding to DNA and stabilizing the filament. Notably, BCDX2 also promotes RAD51 loading on short ssDNA and reversed replication fork substrates. Moreover, while mutants defective in ssDNA binding retain the ability to bind branched DNA substrates, they still facilitate RAD51 loading onto reversed replication forks. Our study provides mechanistic insights into how the BCDX2 complex stimulates the formation of BRCA2-independent RAD51 filaments on short stretches of ssDNA present at ssDNA gaps or stalled replication forks, highlighting its role in genome maintenance and DNA repair.


Assuntos
Replicação do DNA , DNA de Cadeia Simples , Proteínas de Ligação a DNA , Rad51 Recombinase , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Humanos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Replicação do DNA/genética , Ligação Proteica , Mutação
6.
Nat Commun ; 15(1): 7797, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39242676

RESUMO

Ribosomal DNA (rDNA) encodes the ribosomal RNA genes and represents an intrinsically unstable genomic region. However, the underlying mechanisms and implications for genome integrity remain elusive. Here, we use Bloom syndrome (BS), a rare genetic disease characterized by DNA repair defects and hyper-unstable rDNA, as a model to investigate the mechanisms leading to rDNA instability. We find that in Bloom helicase (BLM) proficient cells, the homologous recombination (HR) pathway in rDNA resembles that in nuclear chromatin; it is initiated by resection, replication protein A (RPA) loading and BRCA2-dependent RAD51 filament formation. However, BLM deficiency compromises RPA-loading and BRCA1/2 recruitment to rDNA, but not RAD51 accumulation. RAD51 accumulates at rDNA despite depletion of long-range resection nucleases and rDNA damage results in micronuclei when BLM is absent. In summary, our findings indicate that rDNA is permissive to RAD51 accumulation in the absence of BLM, leading to micronucleation and potentially global genomic instability.


Assuntos
DNA Ribossômico , Instabilidade Genômica , Rad51 Recombinase , RecQ Helicases , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Humanos , RecQ Helicases/metabolismo , RecQ Helicases/genética , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , Recombinação Homóloga , Síndrome de Bloom/genética , Síndrome de Bloom/metabolismo , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Reparo do DNA
7.
Nat Commun ; 15(1): 8292, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39333100

RESUMO

BRCA2 is essential for DNA repair by homologous recombination in mitosis and meiosis. It interacts with recombinases RAD51 and DMC1 to facilitate the formation of nucleoprotein filaments on resected DNA ends that catalyse recombination-mediated repair. BRCA2's BRC repeats bind and disrupt RAD51 and DMC1 filaments, whereas its PhePP motifs bind recombinases and stabilise their nucleoprotein filaments. However, the mechanism of filament stabilisation has hitherto remained unknown. Here, we report the crystal structure of a BRCA2-DMC1 complex, revealing how core interaction sites of PhePP motifs bind to recombinases. The interaction mode is conserved for RAD51 and DMC1, which selectively bind to BRCA2's two distinct PhePP motifs via subtly divergent binding pockets. PhePP motif sequences surrounding their core interaction sites protect nucleoprotein filaments from BRC-mediated disruption. Hence, we report the structural basis of how BRCA2's PhePP motifs stabilise RAD51 and DMC1 nucleoprotein filaments for their essential roles in mitotic and meiotic recombination.


Assuntos
Proteína BRCA2 , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Ligação Proteica , Rad51 Recombinase , Rad51 Recombinase/metabolismo , Rad51 Recombinase/química , Proteína BRCA2/metabolismo , Proteína BRCA2/química , Proteína BRCA2/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Humanos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Nucleoproteínas/metabolismo , Nucleoproteínas/química , Nucleoproteínas/genética , Cristalografia por Raios X , Meiose , Sítios de Ligação , Motivos de Aminoácidos , Modelos Moleculares , Mitose
8.
Cell Physiol Biochem ; 58(5): 459-476, 2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-39248186

RESUMO

BACKGROUND/AIMS: One of the treatments for breast cancer is surgical resection of the tumour and prevention of recurrence with postoperative radiotherapy. Unfortunately, radiotherapy is not always effective enough due to the low sensitivity of cancer cells to ionising radiation. This study aimed to evaluate the radiosensitising properties of resveratrol, piceatannol and polydatin on breast cancer cells, which differ in the presence of hormonal receptors on their surface. METHODS: The experimental part was carried out on triple-negative breast cancer cells (HCC38) and hormone-dependent cells (MCF7). The study assessed the level of cell death, changes in the expression of genes (BAX, BCL-2) and proteins related to the apoptosis process (CASPASE 3, 8 and P53), changes in the expression of antioxidant enzymes (CATALASE, SOD, GPx1/2) and NRF-2. Additionally, the expression level of RAD51 protein and histone H2AX, which are involved in DNA repair processes, was assessed. Statistical significance was evaluated by a two-way analysis of variance (ANOVA) followed by Tukey's post hoc test (p < 0.05). RESULTS: Ionising radiation in combination with resveratrol or piceatannol activates the apoptosis process by internal and external pathways. Greater sensitivity of MCF7 cells compared to HCC38 cells to ionising radiation in combination with resveratrol is associated with a weaker antioxidant response of cells and reduced intensity of DNA damage repair. DNA repair induced by ionising radiation occurs more effectively in HCC38 cells than in MCF7 cells. CONCLUSION: Resveratrol has the highest radiosensitising potential among the tested stilbene for cells of both lines. The effectiveness of ionizing radiation in combination with resveratrol (to a lesser extent with piceatannol) is more significant in MCF7 than in HCC38 cells.


Assuntos
Apoptose , Radiação Ionizante , Radiossensibilizantes , Resveratrol , Estilbenos , Humanos , Estilbenos/farmacologia , Resveratrol/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Feminino , Radiossensibilizantes/farmacologia , Linhagem Celular Tumoral , Células MCF-7 , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Neoplasias da Mama/tratamento farmacológico , Histonas/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Rad51 Recombinase/metabolismo , Caspase 3/metabolismo , Glucosídeos
9.
Mol Biol Cell ; 35(11): ar136, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39292916

RESUMO

In yeasts and higher eukaryotes, chromatin motions may be tuned to genomic functions, with transcriptional activation and the DNA damage response both leading to profound changes in chromatin dynamics. The RAD51 recombinase is a key mediator of chromatin mobility following DNA damage. As functions of RAD51 beyond DNA repair are being discovered, we asked whether RAD51 modulates chromatin dynamics in the absence of DNA damage and found that inhibition or depletion of RAD51 alters chromatin motions in undamaged cells. Inhibition of RAD51 increased nucleosome clustering. Predictions from polymer models are that chromatin clusters reduce chain mobility and, indeed, we measured reduced motion of individual chromatin loci in cells treated with a RAD51 inhibitor. This effect was conserved in mammalian cells, yeasts, and plant cells. In contrast, RAD51 depletion or inhibition increased global chromatin motions at the microscale. The results uncover a role for RAD51 in regulating local and global chromatin dynamics independently from DNA damage and highlight the importance of considering different physical scales when studying chromatin dynamics.


Assuntos
Cromatina , Dano ao DNA , Rad51 Recombinase , Rad51 Recombinase/metabolismo , Cromatina/metabolismo , Humanos , Reparo do DNA , Nucleossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Animais , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
10.
Asian Pac J Cancer Prev ; 25(8): 2645-2654, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39205561

RESUMO

BACKGROUND: Radiotherapy (RT) is a crucial treatment for head and neck cancer however, it causes adverse reactions to the normal tissue and organs adjacent to target tumor. The present study was carried out to investigate possible association of single nucleotide polymorphism in DNA repair genes with toxicity effects of radiotherapy on normal tissue. METHODS: Three hundred and fifty head and neck cancer patients receiving radiotherapy treatment were enrolled in this study. The adverse after effects of radiotherapy on the normal tissue in the form of skin reactions were recorded. Single nucleotide polymorphisms of APE1 (rs1130409), hOGG1 (rs1052133) and Rad51 (rs1801320, rs1801321) genes were studied by polymerase chain reaction-Restriction fragment length polymorphism (PCR-RFLP) and direct DNA sequencing methods and their association with development of severe radio-toxicity effects was evaluated logistic regression analysis. RESULTS: The 172G/T polymorphism of Rad51 was 2.85 times higher and significantly associated with skin reactions (OR=2.85, 95% CI: 1.50-5.41; p=0.001) and severe oral mucositis (OR=4.96, 95% CI: 2.40-10.25; p<0.0001). These results suggested that the polymorphic nature of Rad51 is responsible for risk of radiotherapy adverse effects in HNC patients. The variant 326Cys and heterozygous 326Ser/Cys genotype of hOGG1 was significantly associated with high tumor grade (OR=3.16 95% CI: 1.66-5.99; p=0.0004, and OR=3.97 95% CI: 2.15-7.34; p=<0.0001 respectively). The homozygous variant 172TT genotype of Rad51 showed positive association with poor response of both tumor and nodes towards radiotherapy treatment (p=0.007 and p=0.022). CONCLUSIONS: Interpretation of our results revealed significant association of rs1801321 SNP of Rad51 with development of adverse toxicity reactions in normal tissue of head and neck cancer patients treated with radiotherapy.


Assuntos
DNA Glicosilases , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Neoplasias de Cabeça e Pescoço , Polimorfismo de Nucleotídeo Único , Rad51 Recombinase , Humanos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Masculino , Neoplasias de Cabeça e Pescoço/radioterapia , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Feminino , Rad51 Recombinase/genética , Pessoa de Meia-Idade , DNA Glicosilases/genética , Seguimentos , Prognóstico , Lesões por Radiação/genética , Lesões por Radiação/etiologia , Idoso , Adulto , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/patologia , Genótipo , Reparo do DNA/genética , Biomarcadores Tumorais/genética , Radioterapia/efeitos adversos
11.
Neoplasia ; 57: 101037, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39142065

RESUMO

Ovarian cancer (OC) is the deadliest malignancy of the female reproductive system. The standard first-line therapy for OC involves cytoreductive surgical debulking followed by chemotherapy based on platinum and paclitaxel. Despite these treatments, there remains a high rate of tumor recurrence and resistance to platinum. Recent studies have highlighted the potential anti-tumor properties of metformin (met), a traditional diabetes drug. In our study, we investigated the impact of met on the anticancer activities of cisplatin (cDDP) both in vitro and in vivo. Our findings revealed that combining met with cisplatin significantly reduced apoptosis in OC cells, decreased DNA damage, and induced resistance to cDDP. Furthermore, our mechanistic study indicated that the resistance induced by met is primarily driven by the inhibition of the ATM/CHK2 pathway and the upregulation of the Rad51 protein. Using an ATM inhibitor, KU55933, effectively reversed the cisplatin resistance phenotype. In conclusion, our results suggest that met can antagonize the effects of cDDP in specific types of OC cells, leading to a reduction in the chemotherapeutic efficacy of cDDP.


Assuntos
Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Quinase do Ponto de Checagem 2 , Cisplatino , Metformina , Neoplasias Ovarianas , Rad51 Recombinase , Ensaios Antitumorais Modelo de Xenoenxerto , Feminino , Cisplatino/farmacologia , Humanos , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Metformina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Animais , Camundongos , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2/metabolismo , Quinase do Ponto de Checagem 2/genética , Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Transdução de Sinais/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
12.
Nucleic Acids Res ; 52(18): 10999-11013, 2024 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-39180395

RESUMO

Eukaryotic chromosomal replication occurs in a segmented, temporal manner wherein open euchromatin and compact heterochromatin replicate during early and late S-phase respectively. Using single molecule DNA fiber analyses coupled with cell synchronization, we find that newly synthesized strands remain stable at perturbed forks in early S-phase. Unexpectedly, stalled forks are susceptible to nucleolytic digestion during late replication resulting in defective fork restart. This inherent vulnerability to nascent strand degradation is dependent on fork reversal enzymes and resection nucleases MRE11, DNA2 and EXO1. Inducing chromatin compaction elicits digestion of nascent DNA in response to fork stalling due to reduced association of RAD51 with nascent DNA. Furthermore, RAD51 occupancy at stalled forks in late S-phase is diminished indicating that densely packed chromatin limits RAD51 accessibility to mediate replication fork protection. Genetic analyses reveal that susceptibility of late replicating forks to nascent DNA digestion is dependent on EXO1 via DNA mismatch repair (MMR) and that the BRCA2-mediated replication fork protection blocks MMR from degrading nascent DNA. Overall, our findings illustrate differential regulation of fork protection between early and late replication and demonstrate nascent strand degradation as a critical determinant of heterochromatin instability in response to replication stress.


Assuntos
Reparo de Erro de Pareamento de DNA , Replicação do DNA , Exodesoxirribonucleases , Rad51 Recombinase , Fase S , Fase S/genética , Humanos , Exodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , DNA/metabolismo , DNA/genética , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Proteína Homóloga a MRE11/metabolismo , Proteína Homóloga a MRE11/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Heterocromatina/metabolismo , Heterocromatina/genética , Cromatina/metabolismo
13.
Nat Commun ; 15(1): 7197, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39169038

RESUMO

Templated DNA repair that occurs during homologous recombination and replication stress relies on RAD51. RAD51 activity is positively regulated by BRCA2 and the RAD51 paralogs. The Shu complex is a RAD51 paralog-containing complex consisting of SWSAP1, SWS1, and SPIDR. We demonstrate that SWSAP1-SWS1 binds RAD51, maintains RAD51 filament stability, and enables strand exchange. Using single-molecule confocal fluorescence microscopy combined with optical tweezers, we show that SWSAP1-SWS1 decorates RAD51 filaments proficient for homologous recombination. We also find SWSAP1-SWS1 enhances RPA diffusion on ssDNA. Importantly, we show human sgSWSAP1 and sgSWS1 knockout cells are sensitive to pharmacological inhibition of PARP and APE1. Lastly, we identify cancer variants in SWSAP1 that alter Shu complex formation. Together, we show that SWSAP1-SWS1 stimulates RAD51-dependent high-fidelity repair and may be an important new cancer therapeutic target.


Assuntos
DNA de Cadeia Simples , Rad51 Recombinase , Proteína de Replicação A , Humanos , Reparo do DNA , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Recombinação Homóloga , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Ligação Proteica , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , Imagem Individual de Molécula , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
14.
EMBO J ; 43(19): 4356-4383, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39174851

RESUMO

Meiosis is the developmental program that generates gametes. To produce healthy gametes, meiotic recombination creates reciprocal exchanges between each pair of homologous chromosomes that facilitate faithful chromosome segregation. Using fission yeast and biochemical, genetic, and cytological approaches, we have studied the role of CDK (cyclin-dependent kinase) in the control of Swi5-Sfr1, a Rad51-recombinase auxiliary factor involved in homolog invasion during recombination. We show that Sfr1 is a CDK target, and its phosphorylation downregulates Swi5-Sfr1 function in the meiotic prophase. Expression of a phospho-mimetic sfr1-7D mutant inhibits Rad51 binding, its robust chromosome loading, and subsequently decreases interhomolog recombination. On the other hand, the non-phosphorylatable sfr1-7A mutant alters Rad51 dynamics at late prophase, and exacerbates chromatin segregation defects and Rad51 retention observed in dbl2 deletion mutants when combined with them. We propose Sfr1 phospho-inhibition as a novel cell-cycle-dependent mechanism, which ensures timely resolution of recombination intermediates and successful chromosome distribution into the gametes. Furthermore, the N-terminal disordered part of Sfr1, an evolutionarily conserved feature, serves as a regulatory platform coordinating this phospho-regulation, protein localization and stability, with several CDK sites and regulatory sequences being conserved.


Assuntos
Meiose , Rad51 Recombinase , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Fosforilação , Quinases Ciclina-Dependentes/metabolismo , Quinases Ciclina-Dependentes/genética , Regulação para Baixo , Segregação de Cromossomos
15.
Proc Natl Acad Sci U S A ; 121(34): e2402262121, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39145931

RESUMO

Homologous recombination (HR) is essential for the maintenance of genome stability. During HR, Replication Protein A (RPA) rapidly coats the 3'-tailed single-strand DNA (ssDNA) generated by end resection. Then, the ssDNA-bound RPA must be timely replaced by Rad51 recombinase to form Rad51 nucleoprotein filaments that drive homology search and HR repair. How cells regulate Rad51 assembly dynamics and coordinate RPA and Rad51 actions to ensure proper HR remains poorly understood. Here, we identified that Rtt105, a Ty1 transposon regulator, acts to stimulate Rad51 assembly and orchestrate RPA and Rad51 actions during HR. We found that Rtt105 interacts with Rad51 in vitro and in vivo and restrains the adenosine 5' triphosphate (ATP) hydrolysis activity of Rad51. We showed that Rtt105 directly stimulates dynamic Rad51-ssDNA assembly, strand exchange, and D-loop formation in vitro. Notably, we found that Rtt105 physically regulates the binding of Rad51 and RPA to ssDNA via different motifs and that both regulations are necessary and epistatic in promoting Rad51 nucleation, strand exchange, and HR repair. Consequently, disrupting either of the interactions impaired HR and conferred DNA damage sensitivity, underscoring the importance of Rtt105 in orchestrating the actions of Rad51 and RPA. Our work reveals additional layers of mechanisms regulating Rad51 filament dynamics and the coordination of HR.


Assuntos
DNA de Cadeia Simples , Rad51 Recombinase , Reparo de DNA por Recombinação , Proteína de Replicação A , Proteínas de Saccharomyces cerevisiae , Rad51 Recombinase/metabolismo , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Ligação Proteica
16.
J Surg Res ; 302: 250-258, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39111128

RESUMO

INTRODUCTION: RAD51 is a pivotal DNA repair gene managing double-stranded DNA break recognition and repair. RAD51 high expression was associated with adverse outcomes in other cancer types. This study aims to investigate the tumor microenvironment and immune landscape in the RAD51 high-expressed Hepatocellular Carcinoma (HCCs). METHODS: A total of 467 patients from two large independent cohorts with clinical and transcriptomic data were obtained. The cohort was dichotomized based on the median RAD51 gene expression. xCell and Gene Set Enrichment Analysis (GSEA) were used. RESULTS: RAD51 high-expressed HCCs were associated with worse recurrence-free, progression-free, disease-specific, and overall survival (all P < 0.05). While RAD51 high-expressed HCCs were associated with intratumoral heterogeneity, homologous recombination deficiency, and fraction altered scores, mutation or neoantigens were not increased in this group. xCell analysis demonstrated inconsistent immune cell infiltration between two cohorts. Cytolytic activity as well as GSEA with immune-related gene sets also demonstrated inconsistent results between two cohorts as well. On the other hand, RAD51 expression was significantly increased in higher-grade tumors, larger tumors, and higher clinical stages. RAD51 high-expressed HCCs were found to have elevated proliferation score. Furthermore, GSEA exhibited significant enrichment of all the cell proliferation-related gene sets in the Hallmark collection, including E2F targets, G2M checkpoint, Mitotic spindle, MYC targets, and MTORC1 signaling consistently in both cohorts (all false discovery rate < 0.25). CONCLUSIONS: RAD51 high-expressed HCCs were associated with worse survival and with increased cell proliferation and were not necessarily associated with immune infiltration or inflammation.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Rad51 Recombinase , Microambiente Tumoral , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Microambiente Tumoral/imunologia
17.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167481, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39186963

RESUMO

Radiotherapy stands as an effective method in the clinical treatment of hepatocellular carcinoma (HCC) patients. However, both primary and acquired radioresistance limit its clinical application in HCC. Therefore, investigating the mechanism of radioresistance may provide other options for treating HCC. Based on single-cell RNA sequencing (scRNA-seq) and HCC transcriptome datasets, 227 feature genes with prognostic value were selected to establish the tSNE score. The tSNE score emerged as an independent prognostic factor for HCC and correlated with cell proliferation and radioresistance-related biological functions. UBAP2 was identified as the most relevant gene with the tSNE score, consistently elevated in human HCC samples, and positively associated with patient prognosis. Functionally, UBAP2 knockdown impeded HCC development and reduced radiation resistance in vitro and in vivo. The ectopic expression of SLC27A5 reversed the effects of UBAP2. Mechanically, we uncovered that UBAP2, through the ubiquitin-proteasome system, decreased the homologous recombination-related gene RAD51, not the non-homologous end-joining (NHEJ)-related gene CTIP, by degrading the antioncogene SLC27A5, thereby generating radioresistance in HCC. The findings recapitulated that UBAP2 promoted HCC progression and radioresistance via SLC27A5 stability mediated by the ubiquitin-proteasome pathway. It was also suggested that targeting the UBAP2/SLC27A5 axis could be a valuable radiosensitization strategy in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Tolerância a Radiação , Ubiquitinação , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/radioterapia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/metabolismo , Tolerância a Radiação/genética , Camundongos , Animais , Recombinação Homóloga , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Prognóstico , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Proliferação de Células/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Camundongos Nus , Masculino , Proteínas de Transporte
18.
Nat Commun ; 15(1): 7015, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39147779

RESUMO

During meiosis, nucleoprotein filaments of the strand exchange proteins RAD51 and DMC1 are crucial for repairing SPO11-generated DNA double-strand breaks (DSBs) by homologous recombination (HR). A balanced activity of positive and negative RAD51/DMC1 regulators ensures proper recombination. Fidgetin-like 1 (FIGNL1) was previously shown to negatively regulate RAD51 in human cells. However, FIGNL1's role during meiotic recombination in mammals remains unknown. Here, we decipher the meiotic functions of FIGNL1 and FIGNL1 Interacting Regulator of Recombination and Mitosis (FIRRM) using male germline-specific conditional knock-out (cKO) mouse models. Both FIGNL1 and FIRRM are required for completing meiotic prophase in mouse spermatocytes. Despite efficient recruitment of DMC1 on ssDNA at meiotic DSB hotspots, the formation of late recombination intermediates is defective in Firrm cKO and Fignl1 cKO spermatocytes. Moreover, the FIGNL1-FIRRM complex limits RAD51 and DMC1 accumulation on intact chromatin, independently from the formation of SPO11-catalyzed DSBs. Purified human FIGNL1ΔN alters the RAD51/DMC1 nucleoprotein filament structure and inhibits strand invasion in vitro. Thus, this complex might regulate RAD51 and DMC1 association at sites of meiotic DSBs to promote proficient strand invasion and processing of recombination intermediates.


Assuntos
Proteínas de Ciclo Celular , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA , Meiose , Camundongos Knockout , Rad51 Recombinase , Espermatócitos , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Animais , Masculino , Meiose/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Humanos , Camundongos , Espermatócitos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Recombinação Homóloga , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Dano ao DNA , Endodesoxirribonucleases/metabolismo , Endodesoxirribonucleases/genética , Cromatina/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/genética
19.
Nat Commun ; 15(1): 7081, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39152168

RESUMO

DSS1, essential for BRCA2-RAD51 dependent homologous recombination (HR), associates with the helical domain (HD) and OB fold 1 (OB1) of the BRCA2 DSS1/DNA-binding domain (DBD) which is frequently targeted by cancer-associated pathogenic variants. Herein, we reveal robust ss/dsDNA binding abilities in HD-OB1 subdomains and find that DSS1 shuts down HD-OB1's DNA binding to enable ssDNA targeting of the BRCA2-RAD51 complex. We show that C-terminal helix mutations of DSS1, including the cancer-associated R57Q mutation, disrupt this DSS1 regulation and permit dsDNA binding of HD-OB1/BRCA2-DBD. Importantly, these DSS1 mutations impair BRCA2/RAD51 ssDNA loading and focus formation and cause decreased HR efficiency, destabilization of stalled forks and R-loop accumulation, and hypersensitize cells to DNA-damaging agents. We propose that DSS1 restrains the intrinsic dsDNA binding of BRCA2-DBD to ensure BRCA2/RAD51 targeting to ssDNA, thereby promoting optimal execution of HR, and potentially replication fork protection and R-loop suppression.


Assuntos
Proteína BRCA2 , Replicação do DNA , DNA de Cadeia Simples , DNA , Recombinação Homóloga , Mutação , Rad51 Recombinase , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/química , Humanos , DNA/metabolismo , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Homeostase , Ligação Proteica , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Domínios Proteicos , Linhagem Celular Tumoral , Dano ao DNA , Complexo de Endopeptidases do Proteassoma
20.
Nucleic Acids Res ; 52(17): 10490-10503, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39189448

RESUMO

Telomeric repeat containing RNA (TERRA) is a noncoding RNA that is transcribed from telomeres. Previous study showed that TERRA trans anneals by invading into the telomeric duplex to form an R-loop in mammalian cells. Here, we elucidate the molecular mechanism underlying TERRA recruitment and invasion into telomeres in the context of shelterin proteins, RAD51 and RNase H using single molecule (sm) assays. We demonstrate that TERRA trans annealing into telomeric DNA exhibits dynamic movement that is stabilized by TRF2. TERRA annealing to the telomeric duplex results in the formation of a stable triplex structure which differs from a conventional R-loop. We identified that the presence of a sub-telomeric DNA and a telomeric overhang in the form of a G-quadruplex significantly enhances TERRA annealing to telomeric duplex. We also demonstrate that RAD51-TERRA complex invades telomere duplex more efficiently than TERRA alone. Additionally, TRF2 increases TERRA affinity to telomeric duplex and protects it from RNase H digestion. In contrast, TRF1 represses TERRA annealing to telomeric duplex and fails to provide protection against RNase H digestion. Our findings provide an in-depth molecular mechanism underpinning TERRA recruitment and annealing to the telomere.


Assuntos
Rad51 Recombinase , Ribonuclease H , Telômero , Proteína 1 de Ligação a Repetições Teloméricas , Proteína 2 de Ligação a Repetições Teloméricas , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Ribonuclease H/metabolismo , Rad51 Recombinase/metabolismo , Humanos , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/genética , Quadruplex G , DNA/metabolismo , DNA/química , DNA/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteínas de Ligação a Telômeros/genética , Estruturas R-Loop , RNA não Traduzido/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/química , Complexo Shelterina/metabolismo , Imagem Individual de Molécula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...