Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Clin Oncol ; 150(1): 27, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38263342

RESUMO

PURPOSE: Cervical cancer is a common gynecological malignancy, pathologically associated with persistent infection of high-risk types of human papillomavirus (HPV). Previous studies revealed that HPV-positive cervical cancer displays genomic instability; however, the underlying mechanism is not fully understood. METHODS: To investigate if DNA damage responses are aggravated in precancerous lesions of HPV-positive cervical epithelium, cervical tissues were biopsied and cryosectioned, and subjected to immunofluorescent staining. Cloned HA-tagged E6 and E7 genes of HPV16 subtype were transfected into HEK293T or C33A cells, and indirect immunofluorescent staining was applied to reveal the competency of double strand break (DSB) repair. To test the synthetic lethality of E7-indued HRD and PARP inhibitor (PARPi), we expressed E7 in C33A cells in the presence or absence of olaparib, and evaluated cell viability by colony formation. RESULTS: In precancerous lesions, endogenous DNA lesions were elevated along with the severity of CIN grade. Expressing high-risk viral factor (E7) in HPV-negative cervical cells did not impair checkpoint activation upon genotoxic insults, but affected the potential of DSB repair, leading to homologous recombination deficiency (HRD). Based on this HPV-induced genomic instability, the viability of E7-expressing cells was reduced upon exposure to PARPi in comparison with control cells. CONCLUSION: In aggregate, our findings demonstrate that HPV-E7 is a potential driver for genome instability and provides a new angle to understand its role in cancer development. The viral HRD could be employed to target HPV-positive cervical cancer via synthetic lethality.


Assuntos
Antineoplásicos , Infecções por Papillomavirus , Lesões Pré-Cancerosas , Neoplasias do Colo do Útero , Humanos , Feminino , Papillomavirus Humano , Inibidores de Poli(ADP-Ribose) Polimerases , Células HEK293 , Papillomaviridae , Instabilidade Genômica
2.
J Clin Invest ; 133(23)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37815873

RESUMO

Many cancers harbor homologous recombination defects (HRDs). A HRD is a therapeutic target that is being successfully utilized in treatment of breast/ovarian cancer via synthetic lethality. However, canonical HRD caused by BRCAness mutations do not prevail in liver cancer. Here we report a subtype of HRD caused by the perturbation of a proteasome variant (CDW19S) in hepatitis B virus-bearing (HBV-bearing) cells. This amalgamate protein complex contained the 19S proteasome decorated with CRL4WDR70 ubiquitin ligase, and assembled at broken chromatin in a PSMD4Rpn10- and ATM-MDC1-RNF8-dependent manner. CDW19S promoted DNA end processing via segregated modules that promote nuclease activities of MRE11 and EXO1. Contrarily, a proteasomal component, ADRM1Rpn13, inhibited resection and was removed by CRL4WDR70-catalyzed ubiquitination upon commitment of extensive resection. HBx interfered with ADRM1Rpn13 degradation, leading to the imposition of ADRM1Rpn13-dependent resection barrier and consequent viral HRD subtype distinguishable from that caused by BRCA1 defect. Finally, we demonstrated that viral HRD in HBV-associated hepatocellular carcinoma can be exploited to restrict tumor progression. Our work clarifies the underlying mechanism of a virus-induced HRD subtype.


Assuntos
Carcinoma Hepatocelular , Hepatite B , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Neoplasias Hepáticas/genética , Transativadores/genética , Transativadores/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição/genética , Hepatite B/genética , Recombinação Homóloga , Peptídeos e Proteínas de Sinalização Intracelular/genética
3.
Exp Cell Res ; 417(2): 113225, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35644414

RESUMO

Malignancies including ovarian cancer (OvCa) are genetically unstable. Genomic integrity is maintained by tumor suppressor p53 and DNA damage response network, which crosstalk to each other via not well characterized mechanisms. In this work, we characterize features of damage-related signals in cultured epithelial OvCa cells and tumor biopsies. We found that endogenous burden of DNA damage in OvCa tissues were ubiquitously accumulated in high-grade malignancies than lower grade of cancer that cannot be obviously explained by disturbed function of in DNA damage response (DDR). In contrast, CHK1 phosphorylation (CHK1-pS345) marking the checkpoint activation in nucleolar compartments are prevalent in high-grade OvCa, coincident to the elevated DNA damage in nucleoplasm. Generation of CHK1-pS345 requires the presence of p53 protein in addition to the well-known activities of ATM/ATR kinases. Apparently, mutant forms of p53 possess higher activity in triggering CHK1 phosphorylation than wild type, implying a potential role of p53 in maintaining rDNA integrity. Loss of p53 function would cause replication stress in nucleoli. Altogether, our study reveals endogenous nucleoli stress in OvCa that is coupled to perturbed function of p53 in DNA repair.


Assuntos
Neoplasias Ovarianas , Proteína Supressora de Tumor p53 , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Carcinoma Epitelial do Ovário/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/metabolismo , Dano ao DNA/genética , Feminino , Humanos , Neoplasias Ovarianas/genética , Fosforilação , Proteínas Quinases/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
4.
Nat Commun ; 13(1): 2861, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35606358

RESUMO

The atypical nuclease ENDOD1 functions with cGAS-STING in innate immunity. Here we identify a previously uncharacterized ENDOD1 function in DNA repair. ENDOD1 is enriched in the nucleus following H2O2 treatment and ENDOD1-/- cells show increased PARP chromatin-association. Loss of ENDOD1 function is synthetic lethal with homologous recombination defects, with affected cells accumulating DNA double strand breaks. Remarkably, we also uncover an additional synthetic lethality between ENDOD1 and p53. ENDOD1 depletion in TP53 mutated tumour cells, or p53 depletion in ENDOD1-/- cells, results in rapid single stranded DNA accumulation and cell death. Because TP53 is mutated in ~50% of tumours, ENDOD1 has potential as a wide-spectrum target for synthetic lethal treatments. To support this we demonstrate that systemic knockdown of mouse EndoD1 is well tolerated and whole-animal siRNA against human ENDOD1 restrains TP53 mutated tumour progression in xenograft models. These data identify ENDOD1 as a potential cancer-specific target for SL drug discovery.


Assuntos
Neoplasias , Mutações Sintéticas Letais , Animais , Reparo do DNA , Humanos , Peróxido de Hidrogênio , Camundongos , Neoplasias/patologia , Mutações Sintéticas Letais/genética , Proteína Supressora de Tumor p53/genética
5.
Carcinogenesis ; 42(4): 640-649, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33417668

RESUMO

Impairment of genome instability drives the development of cancer by disrupting anti-cancer barriers. Upon genotoxic insults, DNA damage responsive factors, notably ATM kinase, is crucial to protect genomic integrity while promoting cell death. Meanwhile, cytotoxic therapy-inducing DNA lesions is double-edged sword by causing cancer metastasis based on animal models and clinical observations. The underlying mechanisms for the procancer effect of cytotoxic therapies are poorly understood. Here, we report that cancer cells subjected to cytotoxic treatments elicit dramatic alteration of gene expression controlling the potential of epithelium-mesenchyme transition (EMT). Resultantly, EMT-dependent cell mobility is potently induced upon DNA damage. This stimulation of EMT is mainly Ataxia-Telangiectasia-mutated (ATM)-dependent, as the chemical inhibitor specifically inhibiting ATM kinase activity can suppress the EMT gene expression and thus cell mobility. At last, we show that cancer cells with ATM activation display increased metastatic potential in ovarian cancer tissues. Taken together, we reveal a novel role of ATM in promoting metastatic potential of cancer cells by favoring EMT gene expression.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Neoplasias/genética , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Instabilidade Genômica/genética , Humanos , Metástase Neoplásica , Neoplasias/patologia
6.
BMC Cancer ; 20(1): 1020, 2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087072

RESUMO

BACKGROUND: Loss of the genomic stability jeopardize genome stability and promote malignancies. A fraction of ovarian cancer (OvCa) arises from pathological mutations of DNA repair genes that result in highly mutagenic genomes. However, it remains elusive why the ovarian epithelial cells are particularly susceptible to the malfunction of genome surveillance system. METHODS: To explore the genotoxic responses in the unique context of microenvironment for ovarian epithelium that is periodically exposed to high-level steroid hormones, we examined estrogen-induced DNA damage by immunofluorescence in OvCa cell lines, animal and human samples. RESULTS: We found that OvCa cells are burdened with high levels of endogenous DNA damage that is not correlated with genomic replication. The elevation of damage burden is attributable to the excessive concentration of bioactive estrogen instead of its chemomimetic derivative (tamoxifen). Induction of DNA lesions by estrogen is dependent on the expression of hormone receptors, and occurs in G1 and non-G1 phases of cell cycle. Moreover, depletion of homologous recombination (HR) genes (BRCA1 and BRCA2) exacerbated the genotoxicity of estrogen, highlighting the role of HR to counteract hormone-induced genome instability. Finally, the estrogen-induced DNA damage was reproduced in the epithelial compartments of both ovarian and fallopian tubes. CONCLUSIONS: Taken together, our study disclose that estrogen-induced genotoxicity and HR deficiency perturb the genome stability of ovarian and fallopian epithelial cells, representing microenvironmental and genetic risk factors, respectively.


Assuntos
Dano ao DNA , Estrogênios/toxicidade , Tubas Uterinas/efeitos dos fármacos , Neoplasias Ovarianas/genética , Ovário/efeitos dos fármacos , Animais , Proteína BRCA1/genética , Proteína BRCA2/genética , Linhagem Celular Tumoral , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Tubas Uterinas/metabolismo , Feminino , Recombinação Homóloga , Humanos , Camundongos , Neoplasias Experimentais , Especificidade de Órgãos , Neoplasias Ovarianas/tratamento farmacológico , Ovário/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo
7.
Hepatology ; 69(6): 2546-2561, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30791110

RESUMO

Chronic infection of hepatitis B virus (HBV) is associated with an increased incidence of hepatocellular carcinoma (HCC). HBV encodes an oncoprotein, hepatitis B x protein (HBx), that is crucial for viral replication and interferes with multiple cellular activities including gene expression, histone modifications, and genomic stability. To date, it remains unclear how disruption of these activities contributes to hepatocarcinogenesis. Here, we report that HBV exhibits antiresection activity by disrupting DNA end resection, thus impairing the initial steps of homologous recombination (HR). This antiresection activity occurs in primary human hepatocytes undergoing a natural viral infection-replication cycle as well as in cells with integrated HBV genomes. Among the seven HBV-encoded proteins, we identified HBx as the sole viral factor that inhibits resection. By disrupting an evolutionarily conserved Cullin4A-damage-specific DNA binding protein 1-RING type of E3 ligase, CRL4WDR70 , through its H-box, we show that HBx inhibits H2B monoubiquitylation at lysine 120 at double-strand breaks, thus reducing the efficiency of long-range resection. We further show that directly impairing H2B monoubiquitylation elicited tumorigenesis upon engraftment of deficient cells in athymic mice, confirming that the impairment of CRL4WDR70 function by HBx is sufficient to promote carcinogenesis. Finally, we demonstrate that lack of H2B monoubiquitylation is manifest in human HBV-associated HCC when compared with HBV-free HCC, implying corresponding defects of epigenetic regulation and end resection. Conclusion: The antiresection activity of HBx induces an HR defect and genomic instability and contributes to tumorigenesis of host hepatocytes.


Assuntos
Carcinoma Hepatocelular/virologia , Instabilidade Genômica/genética , Hepatite B/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Transativadores/genética , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Epigênese Genética , Hepatite B/patologia , Vírus da Hepatite B/genética , Hepatócitos/citologia , Hepatócitos/fisiologia , Humanos , Litostatina/genética , Neoplasias Hepáticas/patologia , Camundongos , Sensibilidade e Especificidade , Proteínas Virais Reguladoras e Acessórias , Replicação Viral/genética
8.
BMC Cancer ; 18(1): 426, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29661159

RESUMO

BACKGROUND: Ovarian cancer is one of the most deadly gynecological malignancies and inclined to recurrence and drug resistance. Previous studies showed that the tumorigenesis of ovarian cancers and their major histotypes are associated with genomic instability caused by defined sets of pathogenic mutations. In contrast, the mechanism that influences the development of drug resistance and disease recurrence is not well elucidated. Solid tumors are prone to chromosomal instability (CIN) and micronuclei formation (MN). Although MN is traditionally regarded as the outcome of genomic instability, recent investigation on its origin and final consequences reveal that the abnormal DNA metabolism in MN is a driver force for some types of catastrophic genomic rearrangements, accelerating dramatic genetic variation of cancer cells. METHODS: We used Indirect Immunofluorescent staining to visualize micronuclei and activation of DNA repair factors in ovarian cancer cell lines and biopsies. RESULTS: We show that ovarian cancer cells are disposed to form micronuclei upon genotoxic insults. Double strand DNA breaks (DSBs)-triggered insurgence of micronuclei is associated with unrepaired chromosomes passing through mitosis. According to their morphology and DNA staining, micronuclei compartments are divided into early and late stages that can be further characterized by differential staining of γH2AX and 53BP1. We also show that MN compartments do not halt controlled DNA metabolism as sequestered nuclear repair factors are enriched at DNA breaks in MN compartments and efficiently process DNA ends to generate single-stranded DNA (ssDNA) structures. Interestingly, unknown factors are required for DNA end processing in MN in addition to the nuclear resection machinery. Finally, these hallmarks of micronuclei evolution depicted in cell culture were recapitulated in different stages of ovarian cancer biopsies. CONCLUSIONS: In aggregate, our findings demonstrate that ovarian cancer cells are inclined to form micronuclei that undergo robust DNA metabolism and generate ssDNA structures, potentially destabilizing genomic structures and triggering genetic variation.


Assuntos
Reparo do DNA por Junção de Extremidades/genética , DNA de Neoplasias/genética , Recidiva Local de Neoplasia/genética , Neoplasias Ovarianas/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/genética , Feminino , Instabilidade Genômica/genética , Humanos , Micronúcleos com Defeito Cromossômico , Mutação , Recidiva Local de Neoplasia/patologia , Neoplasias Ovarianas/patologia
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(5): 693-698, 2017 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-29130659

RESUMO

OBJECTIVE: To investigate the roles of enzyme DCAF proteinDNA damagebinding protein 1 (DDB1)/cullin4 (CRL4) complex family members CRL4WD40 repeat domain protein 70 (WDR70) in DNA repair process and its mutation in ovarian cancer. METHODS: Immunofluorescent assay was employed to measure H2AX (γH2AX) and phosphorylated replication protein A2 (RPA32) formed in siDDB1 or siWDR70 ovarian cancer cells after the treatments of chemical medicine and radioactive threapy. 5Brdu immunohistochemical staining was used to explore the function of WDR70 in DNA replication. The expressions of WDR70 and histone protein H2B monoubiquitination (uH2B) was measured by immunohistochemistry,the function of DNA repair,expression and mutations of CRL4 in ovarian cancer were detected by semiquantitative PCR and DNA sequencing. RESULTS: Immunofluorescent assay indicated that distinct subunits of CRL4 played different roles in checkpoint activation and H2Bmonoubiquitinationdepedendent homologous recombination,while the scaffold subunit DDB1 participated in both processes,WDR70 was only required for DNA end resection,chromatin loading of RPA32 and HR. The dose of WDR70 was not effect on DNA replication. Ovarian cancer had different expression of WDR70 and uH2B compared with normal tissue,transcripts of WDR70 was diminished or truncated in 50% of ovarian cancer,which corresponded to multiple mutations. CONCLUSION: CRL4 ubiquitin ligase plays multiple roles in DNA repair and is critical for genome stability. It may be an potential anticancer barrier against ovarian malignancies.


Assuntos
Reparo do DNA , Histonas , Neoplasias Ovarianas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Feminino , Humanos , Mutação , Neoplasias Ovarianas/genética , Ubiquitina-Proteína Ligases/genética
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 47(3): 316-20, 336, 2016 May.
Artigo em Chinês | MEDLINE | ID: mdl-27468472

RESUMO

OBJECTIVE: To assess the sensitisation effects of DDR inhibitors combined with conventional chemotherapeutics agents (cisplatin et al) in a drug-resistant ovarian cancer cell line(OVCAR-8). METHODS: Inhibitors of DDR regulators with cisplatin were applied to challenge OVCAR-8, and evaluated the DNA damage response (DDR) and cytotoxic effects of different combination of chemicals. Inhibition of proliferation to OVCAR-8 of different drugs was evaluated by MTT assay. The activation of phosphorylation of histone family 2A variant (yH2AX) and p53 binding protein 1 (53BP1) in OVCAR-8 were evaluated by immunofluorescence to observe their ability of recruitment and forming foci at DNA damage site. RESULTS: We observed that combined treatment of ataxia-telangiectasia mutated (ATM)/ATM and Rad 3-related (ATR) inhibitor and cisplatin can suppress the activation of damage repair mechanisms and weakened the proliferative activity of OVCAR-8 cells (P<0. 01) ; ATR pathway was suppressed and the signal of γH2AX weakened and cell survival rate significantly reduced when combination therapy of HU and Wortmannin (P < 0.05); poly ADP-ribose polymerase (PARP) inhibitor could not enhance chemosensitivity in OVCAR-8 cells when combined with cisplatin (P > 0.05). CONCLUSION: We substantiated that appropriate inhibitors of DNA damage response may have a potential to improve the anti-tumor effect of conventional chemotherapy drugs and prevent drug resistances.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Dano ao DNA , Neoplasias Ovarianas/patologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Linhagem Celular Tumoral/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosforilação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
11.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 47(4): 501-506, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-28591950

RESUMO

OBJECTIVES: To analyze the cellular function of the newly discovered DNA damage repair factor WDR70, and investigate the mutation in ovarian cancer to verify if function loss of the WDR70gene was associated with ovarian cancer. METHODS: The WDR70 gene was silenced by using siRNA technique or overexpressed its wild and mutation type by with lentivirus and plasmid in hunman cells. The subcellular localization and biochemical function of WDR70 was analyzes by indirect immunofluorescence and immunoblotting. The expression level of WDR70 and the mutations of its cDNA was checked with RT-PCR sequencing for 1 normal ovarian tissue and 16 ovarian cancer specimen. RESULTS: We found gene silencing of WDR70 or overexpression of WDR70 mutation type disrupts the phosphorylation level of homologous recombination functional protein RPA32 and the ability of recruitment at DNA damage site of recombinase RAD51, the loss of function of WDR70 also causes the elevation of the chromosome breakage in metaphase. Meanwhile, we also noticed that the existence of multiple mutations in genomic WDR70 in ovarian cancer specimen. CONCLUSIONS: Our results defined that in vitro system, WDR70 is a DNA damage repair gene, silencing of WDR70 or overexpression of WDR70 mutation type disrupts homologous recombination and chromosomal instability; the frequent mutations of WDR70 gene in genome of ovarian cancer specimens could also lead to DNA repair defeat and gene instability. Consequently WDR70 gene could represent an anti-cancer mechanism for ovarian cancer.


Assuntos
Dano ao DNA , Reparo do DNA , Neoplasias Ovarianas/genética , Feminino , Humanos , Mutação
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 47(6): 837-842, 2016 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-28598108

RESUMO

OBJECTIVES: To determine the function of human papillomavirus (HPV) E7 in DNA damage response of cervical intraepithelial neoplasia (CIN) 3 cells. METHODS: Samples of CIN 3 and child foreskin tissues were collected,with pathologically confirmed HPV positive and negative,respectively.Collagenase A was used for digesting tissues prior to primary culture.The HPV negative cells were infected with lentivirus E7 and pLV.Proteins(53BP1,NBS1,BRCA1 and RPA32) responsive to DNA double break damages were detected by indirect immunofluorescent staining after 0-8 h treatment with X-ray (2 or 5 Gy). RESULTS: After treatment with 2 or 5 Gy X-ray,53BP1,NBS1,BRCA1 and RPA32 foci in HPV+ cells increased compared with HPV- cells (P<0.05).Less 53BP1,RPA32,BRCA1 and NBS1 foci positive cells (foci>5) were found in E7 infected cells than in pLV infected cells(P<0.05).Both of them reached the peak at 6 h (2 Gy) or 4 h (5 Gy). CONCLUSIONS: We have successfully established a model to detect the function of HPV E7 in DNA damage response using primary culture of CIN fibroblasts.With the progression of CIN,HPV E7 can inhibit DNA double break repair.


Assuntos
Dano ao DNA , Proteínas E7 de Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Displasia do Colo do Útero/virologia , Criança , Feminino , Humanos , Proteínas Oncogênicas Virais , Papillomaviridae , Displasia do Colo do Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA