Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Adicionar filtros








Intervalo de ano
1.
Artigo | IMSEAR | ID: sea-220361

RESUMO

DNA repair enzymes are proteins that detect and repair physical damage to DNA induced by radiation, ultraviolet light, or reactive oxygen species. The repair of DNA damage prevents the loss of genetic information, the creation of double-strand breaks, and the formation of DNA crosslinks. The time-dependent reduction of functional properties is known as aging. Mitochondrial malfunction and the buildup of genetic damage are two common factors of aging. In fact, the poor maintenance of nuclear and mitochondrial DNA is likely a major factor in aging. When the DNA repair machinery isn't operating fine, DNA lesions and mutations can occur, which can lead to cancer development. In fact, the poor maintenance of nuclear and mitochondrial DNA is likely a major factor in aging. When the DNA repair enzymes isn't operating fine, DNA lesions and mutations can occur, which can lead to cancer development. The large number of alterations per cell, which can reach 105, has been identified as a driving mechanism in oncogenesis. These findings show that abnormalities in the DNA repair pathway contribute to the senescence as well as cancer. Nucleotide excision repair (NER), base excision repair (BER), double-strand break repair, mismatch repair (MMR), are all major DNA repair processes in mammalian cells. BER excises mostly oxidative and alkylation DNA damage, NER removes bulky, helix-distorting lesions from DNA (e.g., ultraviolet (UV) photodimers), MMR corrects replication errors

2.
Chinese Journal of Radiation Oncology ; (6): 614-618, 2021.
Artigo em Chinês | WPRIM | ID: wpr-910437

RESUMO

Objective:To clarify the role of classic Wnt signaling pathway in the radioresistance of esophageal cancer cells (ECC), and investigate the underlying mechanism, aiming to identify critical molecular targets for clinically enhancing the radiosensitivity of esophageal cancer.Methods:The radiosensitivity of four types of ECCs (EC9706, ECA109, KYSE70 and KYSE150) were assessed by colony formation assay. Western blot and RT-PCR were used to detect the activation of classical Wnt signaling pathway after irradiation. Classic Wnt signaling pathway activator (AZD2858) and inhibitor (XAV-939) were utilized to comprehensively evaluate the effect of classic Wnt signaling pathway on the radiosensitivity of ECCs. Cellular immunofluorescence staining was performed to detect the production and repair of DNA double-strand breaks (DSB), as well as the foci formation of DSB repair proteins after irradiation.Results:The results of colony formation assay showed that the radiosensitivity of four types of ECCs from high to low was EC9706, ECA109, KYSE70 and KYSE150. In KYSE150, a radioresistant cell type, the level of nuclear β-catenin and the transcription of c-Myc gene were significantly increased after irradiation (both P<0.05). However, in EC9706, a radiosensitive cell type, the level of nuclear β-catenin and c-Myc gene transcription were not affected by irradiation (both P>0.05). Moreover, EC9706 cells showed enhanced radioresistance in the presence of AZD2858( P<0.05), whereas XAV-939 treatment decreased the radioresistance in KYSE150 cells ( P<0.05). AZD2858 accelerated the DSB repair in EC9706 cells ( P<0.05), whereas XAV-939 delayed the DSB repair in KYSE150 cells ( P<0.05). Furthermore, the results of immunofluorescence staining showed that XAV-939 reduced the DSB repair capacity by inhibiting homologous recombination repair-related proteins (BRCA1 and RAD51) rather than non-homologous end junction repair-related proteins (Ku80 and XRCC4). Conclusions:The classic Wnt signaling pathway participates in the regulation of radiosensitivity in ECCs by regulating the homologous recombination repair of DSB after irradiation. Inhibition of the classic Wnt signaling pathway can counteract the radioresistance of ECCs and enhance the killing effect of irradiation on ECCs.

3.
Journal of Zhejiang University. Science. B ; (12): 63-72, 2021.
Artigo em Inglês | WPRIM | ID: wpr-880709

RESUMO

DNA is the hereditary material in humans and almost all other organisms. It is essential for maintaining accurate transmission of genetic information. In the life cycle, DNA replication, cell division, or genome damage, including that caused by endogenous and exogenous agents, may cause DNA aberrations. Of all forms of DNA damage, DNA double-strand breaks (DSBs) are the most serious. If the repair function is defective, DNA damage may cause gene mutation, genome instability, and cell chromosome loss, which in turn can even lead to tumorigenesis. DNA damage can be repaired through multiple mechanisms. Homologous recombination (HR) and non-homologous end joining (NHEJ) are the two main repair mechanisms for DNA DSBs. Increasing amounts of evidence reveal that protein modifications play an essential role in DNA damage repair. Protein deubiquitination is a vital post-translational modification which removes ubiquitin molecules or polyubiquitinated chains from substrates in order to reverse the ubiquitination reaction. This review discusses the role of deubiquitinating enzymes (DUBs) in repairing DNA DSBs. Exploring the molecular mechanisms of DUB regulation in DSB repair will provide new insights to combat human diseases and develop novel therapeutic approaches.

4.
Acta Pharmaceutica Sinica ; (12): 2535-2548, 2020.
Artigo em Chinês | WPRIM | ID: wpr-837517

RESUMO

Applying poly(ADP-ribose) polymerase inhibitors (PARPi) to the treatment of cancers with homologous recombination deficiency (HRDness) has been a great advance in the field of molecular therapeutics. However, in the clinic patients lacking the specific mutations or developing reverse mutations in the process of PARPi treatment may not benefit from PARPi monotherapy. Therefore, targeting homologous recombination (HR) repair with molecularly targeted agents is becoming an attractive research focus and is raising the concept of "chemical HRDness". HR repair is an evolutionarily conserved and extensively regulated process that employs sister chromatids as the template to repair DNA double-strand breaks with high fidelity. In addition to directly targeting HR components, modulation of regulatory pathways controlling HR repair is effective in achieving the "HRDness" phenotype; this includes modulation of the cell cycle checkpoint regulatory pathway, the phosphatidylinositol 3-kinase (PI3K) signaling pathway, the chromatin remodeling pathway, etc. Targeting HR repair can not only result in "synthetic lethality" when combined with PARPi, but also sensitizes cancers to traditional radio/chemotherapy and novel immunotherapy. In this review we describe the HR repair pathway and its regulatory pathways, summarize the preclinical and clinical outcomes of targeting HR repair, discuss the remaining problems in this field and provide a prospective on its application in tumor therapy.

5.
International Journal of Biomedical Engineering ; (6): 154-160, 2019.
Artigo em Chinês | WPRIM | ID: wpr-751605

RESUMO

The small ubiquitin-like modified protein (SUMO) is a protein structurally similar to ubiquitin which is involved in post-translational modification of proteins. SUMOylation refers to the process that SUMO molecule covalently binding to the specific lysine site of target proteins through maturation, activation, binding and ligation by ubiquitin-like specific protease 1 (Ulp1), E1 activating enzyme, E2 binding enzyme, and E3 ligase. SUMOylation alters the activity of target proteins, which is involved in the regulation of various cellular functions such as transcriptional regulation, regulation of embryonic development, cellular stress, maintenance of chromatin structure and genomic stability. In recent years, it has been found that SUMOylation modification is also widely involved in DNA damage repair, especially DNA double-strand breaks (DSBs), which are the most serious types of DNA damage. SUMOylation is involved in almost all processes of DSBs repair, so its role in DNA damage repair has become a research hotspot. In this paper, the research progress of the regulation of SUMOylation in DSBs repair was reviewed.

6.
Chinese Journal of Radiation Oncology ; (6): 1103-1108, 2017.
Artigo em Chinês | WPRIM | ID: wpr-613007

RESUMO

The stability of cell genetic material is influenced by a variety of factors, both internal and external, which can cause various types of DNA damage, such as DNA alkylation, oxidation, mismatching, loop structure, atypical DNA structure, single-strand break, and double-strand break.These DNA damages disrupt cellular homeostasis and dynamic equilibrium, which cause gene mutations, chromosomal abnormalities, and even degradation, aging, and death at different biological levels.By searching and identifying DNA damage sites, the cell activates a series of biochemical pathways, coordinates the progress of DNA replication and transcription, and then repairs the DNA damage.In this way, the cell maintains its independence and stability.While radiotherapy plays a role in eliminating tumors by DNA damages, it also initiates DNA damage responses.Among the responses, base excision repair, nucleotide excision repair, mismatch repair, double-strand break repair, and post-translesion synthesis repair play a key role in repairing the damages.The dysfunction of these repair pathways will cause differences in tumor radiation sensitivity.This paper summarizes recent research results in DNA damage repair, and focuses on the types of DNA damage and their repair mechanisms, so as to promote the understanding of the great significance of this field and to provide a theoretical basis for exploring the application of DNA damage repair pathways in tumor therapy.

7.
Chinese Pharmacological Bulletin ; (12): 323-326,327, 2016.
Artigo em Chinês | WPRIM | ID: wpr-603510

RESUMO

Aim To explore the relationship between Mre11/Rad50/Nbs1 ( MRN ) complex focus formation and DNA double-strand breaks( DSBs) caused by cinob-ufagin in human hepatocellular carcinoma HepG2 cells. Methods The Na+,K+-ATPaseα1 subunit expression level in liver cancer tissues was detected by immunohis-tochemistry. After HepG2 cells were treated with 5μmol·L-1 cinobufagin for 6, 12 and 24 h, the drug-in-duced DSBs were assessed by single cell gel electro-phroesis ( SCGE ) , the gene transcription and protein levels of Mrel1, Nbs1, Rad50 and p53 were evaluated by Real time-PCR and Western blot. The cell cycle in parallel was analyzed by flow cytometry. Results The Na+, K+-ATPase α1 subunit expression level in liver cancer tissues was significantly increased compared with the tissue adjacent to carcinoma ( P <0. 05 ) . The 5μmol · L-1 cinobufagin could induce the DSBs in a time-dependent manner (P <0. 05), and it could up-regulate the gene expression levels of Mre11, Nbs1, Rad50 and p53 in HepG2 cells ( P<0. 05 ) . The pro-portions of HepG2 cells in S phase were ( 21. 32 ± 4. 21) % in the control group, and (33. 25 ± 5. 72) %, (56. 72 ± 6. 29) % and (67. 32 ± 9. 42) % in HepG2 cells treated with 5 μmol · L-1 cinobufagin for 6, 12 and 24 h, respectively. The proportions of cells in S phase in cinobufagin groups were significantly increased compared with the control group ( P<0. 05 ) . Conclu-sion Cinobufagin could induce the cell cycle arrest in liver cancer HepG2 cells by activation of Mre11/Rad50/Nbs1 Complex.

8.
Braz. j. med. biol. res ; 48(6): 502-508, 06/2015. graf
Artigo em Inglês | LILACS | ID: lil-748225

RESUMO

Hormesis is an adaptive response to a variety of oxidative stresses that renders cells resistant to harmful doses of stressing agents. Caffeic acid (CaA) is an important antioxidant that has protective effects against DNA damage caused by reactive oxygen species (ROS). However, whether CaA-induced protection is a hormetic effect remains unknown, as is the molecular mechanism that is involved. We found that a low concentration (10 μM) of CaA increased human liver L-02 cell viability, attenuated hydrogen peroxide (H2O2)-mediated decreases in cell viability, and decreased the extent of H2O2-induced DNA double-strand breaks (DSBs). In L-02 cells exposed to H2O2, CaA treatment reduced ROS levels, which might have played a protective role. CaA also activated the extracellular signal-regulated kinase (ERK) signal pathway in a time-dependent manner. Inhibition of ERK by its inhibitor U0126 or by its specific small interfering RNA (siRNA) blocked the CaA-induced improvement in cell viability and the protective effects against H2O2-mediated DNA damage. This study adds to the understanding of the antioxidant effects of CaA by identifying a novel molecular mechanism of enhanced cell viability and protection against DNA damage.


Assuntos
Humanos , Antioxidantes/farmacologia , Ácidos Cafeicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Espécies Reativas de Oxigênio/análise , Análise de Variância , Western Blotting , Células Cultivadas , Linhagem Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fígado , Estresse Oxidativo/efeitos dos fármacos , Reprodutibilidade dos Testes , Fatores de Tempo
9.
J Biosci ; 2015 Mar; 40 (1): 91-111
Artigo em Inglês | IMSEAR | ID: sea-162021

RESUMO

Whether nucleic acids that circulate in blood have any patho-physiological functions in the host have not been explored. We report here that far from being inert molecules, circulating nucleic acids have significant biological activities of their own that are deleterious to healthy cells of the body. Fragmented DNA and chromatin (DNAfs and Cfs) isolated from blood of cancer patients and healthy volunteers are readily taken up by a variety of cells in culture to be localized in their nuclei within a few minutes. The intra-nuclear DNAfs and Cfs associate themselves with host cell chromosomes to evoke a cellular DNAdamage- repair-response (DDR) followed by their incorporation into the host cell genomes. Whole genome sequencing detected the presence of tens of thousands of human sequence reads in the recipient mouse cells. Genomic incorporation of DNAfs and Cfs leads to dsDNA breaks and activation of apoptotic pathways in the treated cells. When injected intravenously into Balb/C mice, DNAfs and Cfs undergo genomic integration into cells of their vital organs resulting in activation of DDR and apoptotic proteins in the recipient cells. Cfs have significantly greater activity than DNAfs with respect to all parameters examined, while both DNAfs and Cfs isolated from cancer patients are more active than those from normal volunteers. All the above pathological actions of DNAfs and Cfs described above can be abrogated by concurrent treatment with DNase I and/or anti-histone antibody complexed nanoparticles both in vitro and in vivo. Taken together, our results suggest that circulating DNAfs and Cfs are physiological, continuously arising, endogenous DNA damaging agents with implications for ageing and a multitude of human pathologies including initiation of cancer.

10.
Chinese Journal of Pathophysiology ; (12): 924-928, 2015.
Artigo em Chinês | WPRIM | ID: wpr-464252

RESUMO

AIM:To discover the effect of MCPH1 on the DNA damage induced by ionizing radiation in esoph-ageal cancer cells.METHODS:ECA109 cancer cells were radiated at dose of 8 Gy.The nuclear foci of relevant factors were detected 1 h after irradiation in the ECA109 cells after silence of MDC1 gene.A cell line was established that was sta-ble low expression of MCPH1.The nuclear foci induced by ionizing radiation after silence of MCPH1 were determined.RE-SULTS:The MCPH1 gene silenced ECA109 cell line was successfully constructed.A strong relationship between MDC1, MCPH1 andγ-H2AX was observed 1 h after 8 Gy irradiation.Silence of MDC1 did not affect the nuclear foci formation ofγ-H2AX and MCPH1.The nuclear foci of MDC1 but notγ-H2AX significantly reduced after silencing of MCPH1.CON-CLUSION:MCPH1 is located in the downstream of H2AX and upstream formation of MDC1, and regulates the nuclear fo-ci formation of MDC1 during DNA damage response.

11.
Biosalud ; 13(2): 95-110, jul.-dic. 2014. ilus
Artigo em Espanhol | LILACS | ID: lil-760908

RESUMO

Las células cuentan con mecanismos complejos que vigilan la integridad del ADN, activando mecanismos de reparación cuando hay deficiencias o errores durante la replicación. Una consecuencia potencial de los daños son las alteraciones permanentes en la estructura del ADN que pueden generar mutaciones, transformación carcinogénica y muerte celular. Estos son atribuidos a diferentes agentes endógenos como los radicales libres de oxígeno (RLO) provenientes de la respiración, los cuales son considerados el centro de la carcinogénesis y el envejecimiento por daño genómico; agentes exógenos como la luz ultravioleta que inducen dímeros de pirimidina y la radiación ionizante que produce una gran variedad de daños sobre las bases, muchos de ellos por efecto indirecto. También se encuentran las genotoxinas presentes en los alimentos, humo de tabaco y agentes quimioterapéuticos, con grandes cualidades para alterar la estructura de la molécula ADN e interferir con su expresión. De esta manera, cerca de 10(5) lesiones espontáneas por día son inducidas en nuestros genes, en donde los mecanismos de reparación detectan daños y perturbaciones durante el crecimiento y división celular. Esto es posible gracias a las funciones específicas de reconocimiento, corrección o eliminación de daños que asegura la integridad del genoma. En este artículo se presentan los principales mecanismos de reparación del ADN, su relación y activación de acuerdo al tipo de daño.


Cells have complex mechanisms that monitor DNA integrity that activate repair mechanisms when there are deficiencies or errors during replication. A potential result of the damage is a permanent alteration in DNA structure that can generate mutations, carcinogenic transformation and cell death. These are attributed to different endogenous agents such as oxygen free radicals (OFR) from respiration, which are considered the center of carcinogenesis and aging process due to genomic damage; exogenous agents, such as ultraviolet light, induce pyrimidine dimers and ionizing radiation that produce a variety of damage on the bases, many by indirect effect. Genotoxins present in food, tobacco and chemotherapeutic agents are also found with high potential in altering the DNA molecule structure and interfering with its expression. Thus, around 10(5) spontaneous lesions are induced per day in our genes, where the repair mechanisms can detect damages and disturbances during cell growth and division. This is possible thanks to the specific recognition, correction or elimination of damage functions, ensuring the integrity of the genome. In this article the main mechanisms of DNA repair, as well as their relationship and activation according to the type of damage, are presented.

12.
Chinese Journal of Radiological Medicine and Protection ; (12): 248-251, 2013.
Artigo em Chinês | WPRIM | ID: wpr-434857

RESUMO

Objective To establish an experimental model for the study of α-particle-induced bystander effect of DNA damage and investigate the characteristics of bystander DNA double-strand break (DSB).Methods The red fluorescence fusion protein of HsBrkl-RFP was used to mark the cytoplasm of one cell line to distinguish the irradiated target cells (HFS-RFP) and the non-irradiated bystander cells (HFS) in the co-culture cellular model.After α-particle irradiation,cellular DSB and its repair kinetics were analyzed by the immunofluorescence staining of γH2AX and laser confocal microscope observation.Results A bystander studying model was established by co-culturing human HFS-RFP cells with its partner HSF cells.After 0.1 Gy or 0.2 Gy α-particle irradiation,the similar kinetics of γH2AX foci production and abatement were observed in both irradiated HFS-RFP cells and non-irradiated bystander HFS cells,in which the highest level of γH2AX foci was detected at 1 h post-irradiation.The second peak of γH2AX foci formation appeared at 8 h post-irradiation,which possibly indicates the occurrence of secondary DSB.However,the production of secondary DSB in the bystander cells was weaker than that in the irradiated cells.Conclusions The cell co-culture model can be used for bystander effect investigation.Bystander DSB can be effectively induce by irradiation and the secondary breakage of DNA DSB in the bystander cells may relative to the consequential biochemical processing of clustered DNA damage.

13.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 147-153, 2011.
Artigo em Chinês | WPRIM | ID: wpr-298650

RESUMO

DNA damage response (DDR) in different cell cycle starus of human peripheral blood lymphocytes (PBLs) and the role of H2AX in DDR were investigated.The PBLs were stimulated into cell cycle with phytohemagglutinin (PHA).The apoptotic ratio and the phosphorylation H2AX (S139)were flow cytometrically measured in resting and proliferating PBLs after treatment with camptothecin (CPT) or X-ray.The expressions of γH2AX,Bcl-2,caspase-3 and caspase-9 were detected by Western blotting.DDR in 293T cells was detected after H2AX was silenced by RNAi method.Our results showed that DNA double strand breaks (DSBs) were both induced in quiescent and proliferating PBLs after CPT or X-ray treatment.The phosphorylation of H2AX and apoptosis were more sensitive in proliferating PBLs compared with quiescent lymphocytes (P<0.05).The expression levels of anti-apoptotic proteins Bcl-2 were reduced and cleaved caspase-3 and caspase-9 were increased.No significant changes were observed in CPT-induced apoptosis in 293T cells between H2AX knocking down group and controls.It was concluded that proliferating PBLs were more vulnerable to DNA damage compared to non-stimulated lymphocytes and had higher apoptosis rates.γH2AX may only serve as a marker of DNA damage but exert no effect on apoptosis regulation.

14.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 359-364, 2011.
Artigo em Chinês | WPRIM | ID: wpr-298611

RESUMO

The present study investigated the enhanced radiosensitivity of U-251 cells induced by sodium butyrate (NaB) and its possible mechanisms.Increased radiosensitivity of U251 cells was examined by clonogenic cell survival assays.The expression of Ku70 mRNA and protein was detected by using RT-PCR and Western blotting respectively.γ-H2AX foci were measured at different time points after ionizing irradiation alone or combined with NaB treatment.The results showed that cell survival rate was significantly reduced,both D0 and Dq values were decreased (D0:1.43 Gy vs.1.76 Gy; Dq:1.22 Gy vs.2.05 Gy) after the combined treatment as compared with irradiation alone,and sensitivity enhancing ratio (SER) reached 1.23.The average number ofγ-H2AX foci per cell receiving the combined treatment was significantly increased at different time points,and the expression levels of Ku70mRNA and protein were suppressed by NaB in a dose-dependent manner.It was concluded that enhanced radiosensitivity induced by NaB involves an inhibited expression of Ku70 and an increase in γ-H2AX foci,which suggests decreased ability in DSB repair.

15.
Genet. mol. biol ; 33(2): 368-373, 2010. ilus
Artigo em Inglês | LILACS | ID: lil-548813

RESUMO

DNA double-strand breaks (DSBs) are a serious threat to the cell, for if not or miss-repaired, they can lead to chromosomal aberration, mutation and cancer. DSBs in human cells are repaired via non-homologous DNA end joining (NHEJ) and homologous recombination repair pathways. In the former process, the structure of DNA termini plays an important role, as does the genetic constitution of the cells, through being different in normal and pathological cells. In order to investigate the dependence of NHEJ on DSB structure in normal and cancer cells, we used linearized plasmids with various, complementary or non-complementary, single-stranded or blunt DNA termini, as well as whole-cell extract isolated from normal human lymphocytes, chronic myeloid leukemia K562 cells and lung cancer A549 cells. We observed a pronounced variability in the efficacy of NHEJ reaction depending on the type of ends. Plasmids with complementary and blunt termini were more efficiently repaired than the substrate with 3' protruding single-strand ends. The hierarchy of the effectiveness of NHEJ was on average, from the most effective to the least, A549/ normal lymphocytes/ K562. Our results suggest that the genetic constitution of the cells together with the substrate terminal structure may contribute to the efficacy of the NHEJ reaction. This should be taken into account on considering its applicability in cancer chemo-or radiotherapy by pharmacologically modulating NHEJ cellular responses.

16.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 265-270, 2010.
Artigo em Chinês | WPRIM | ID: wpr-349838

RESUMO

Hepatitis B virus (HBV)-induced hepatocellular carcinoma (HCC) is one of the most frequently occurring cancers. Hepadnaviral DNA integrations are considered to be essential agents which can promote the process of the hepatocarcinogenesis. More and more researches were designed to find the relationship of the two. In this study, we investigated whether HBV DNA integration occurred at sites of DNA double-strand breaks (DSBs), one of the most detrimental DNA damage. An 18-bp I-SceI homing endonuclease recognition site was introduced into the DNA of HepG2 cell line by stable DNA transfection, then cells were incubated in patients' serum with high HBV DNA copies and at the same time, DSBs were induced by transient expression of I-SceI after transfection of an I-SceI expression vector. By using nest PCR, the viral DNA was detected at the sites of the break. It appeared that integration occurred between part of HBV x gene and the I-Scel induced breaks. The results suggested that DSBs, as the DNA damages, may serve as potential targets for bepadnaviral DNA insertion and the integrants would lead to widespread host genome changes necessarily. It provided a new site to investigate the integration.

17.
Journal of the Korean Association of Oral and Maxillofacial Surgeons ; : 1-6, 2009.
Artigo em Coreano | WPRIM | ID: wpr-139668

RESUMO

DNA double-strand breaks (DSBs) occur commonly in the all living and in cycling cells. They constitute one of the most severe form of DNA damage, because they affect both strand of DNA. DSBs result in cell death or a genetic alterations including deletion, loss of heterozygosity, translocation, and chromosome loss. DSBs arise from endogenous sources like metabolic products and reactive oxygen, and also exogenous factors like ionizing radiation. Defective DNA DSBs can lead to toxicity and large scale sequence rearrangement that can cause cancer and promote premature aging. There are two major pathways for their repair: homologous recombination(HR) and non-homologous end-joining(NHEJ). The HR pathway is a known "error-free" repair mechanism, in which a homologous sister chromatid serves as a template. NHEJ, on the other hand, is a "error-prone" pathway, in which the two termini of the broken DNA molecule are used to form compatible ends that are directly ligated. This review aims to provide a fundamental understanding of how HR and NHEJ pathways operate, cause genome instability, and what kind of genes during the pathways are associated with head and neck cancer.


Assuntos
Humanos , Senilidade Prematura , Morte Celular , Cromátides , DNA , Dano ao DNA , Instabilidade Genômica , Mãos , Cabeça , Neoplasias de Cabeça e Pescoço , Perda de Heterozigosidade , Oxigênio , Radiação Ionizante , Irmãos
18.
Journal of the Korean Association of Oral and Maxillofacial Surgeons ; : 1-6, 2009.
Artigo em Coreano | WPRIM | ID: wpr-139665

RESUMO

DNA double-strand breaks (DSBs) occur commonly in the all living and in cycling cells. They constitute one of the most severe form of DNA damage, because they affect both strand of DNA. DSBs result in cell death or a genetic alterations including deletion, loss of heterozygosity, translocation, and chromosome loss. DSBs arise from endogenous sources like metabolic products and reactive oxygen, and also exogenous factors like ionizing radiation. Defective DNA DSBs can lead to toxicity and large scale sequence rearrangement that can cause cancer and promote premature aging. There are two major pathways for their repair: homologous recombination(HR) and non-homologous end-joining(NHEJ). The HR pathway is a known "error-free" repair mechanism, in which a homologous sister chromatid serves as a template. NHEJ, on the other hand, is a "error-prone" pathway, in which the two termini of the broken DNA molecule are used to form compatible ends that are directly ligated. This review aims to provide a fundamental understanding of how HR and NHEJ pathways operate, cause genome instability, and what kind of genes during the pathways are associated with head and neck cancer.


Assuntos
Humanos , Senilidade Prematura , Morte Celular , Cromátides , DNA , Dano ao DNA , Instabilidade Genômica , Mãos , Cabeça , Neoplasias de Cabeça e Pescoço , Perda de Heterozigosidade , Oxigênio , Radiação Ionizante , Irmãos
19.
Chinese Journal of Radiological Medicine and Protection ; (12): 5-8, 2009.
Artigo em Chinês | WPRIM | ID: wpr-396127

RESUMO

Objective To investigate ff the γ-H12AX foci is a precise index for the DSB formation and repair in mature neurons of brain in vivo after clinically relevant doses irradiation. Methods For the DSB formation experiment, the mature neurons in the neocortex of brain tissue of C57BL/6 mice were analyzed at 10 rain after whole-body irradiation with 0.1, 0.5 and 1.0 Gy. For the DSB repair kinetics experiment, the mature neurons in the neocortex of brain tissue of repair-proficient (C57BL/6 mice) and repair-deficient mouse strains (BALB/c, A-T and SCID mice) were analyzed at 0.5, 2.5, 5, 24 and 48 h after whole-body irradiation with 2 Gy. The mature neurons in the neocortex of brain tissue of sham-irradiated mice of each strain served as controls. γ-H2AX immunohistochemistry and γ-H2AX and NeuN double immunofluorescence analysis was used to measure DSBs formation and repair in the mature neurons in the neocortex of brain tissue of the different mouse strains. Results For the DSB formation experiment, γ-H2AX foci levels with a clear linear dose correlation and very low backgrounds in the nuclei in the neocortex of brain tissue were observed. Scoring the loss of γ-H12AX foci allowed us to verify the different, genetically determined DSB repair deficiencies, including the minor impairment of BALB/c mice. Repair-proficient C57BL/6 mice exhibited the fastest decrease in foei number with time, and displayed low levels of residual damage at 24 h and 48 h post-irradiation. In contrast, SCID mice showed highly increased γ-H2AX foci levels at all repair times (0.5 h to 48 h) while A-T mice exhibited a lesser defect which was most significant at later repair times (≥ 5 h). Radiosensitive BALB/c mice exhibited slighdy elevated foei numbers compared with C57BI./6 mice at 5 h and 24 h but not at 48 h post-irradiation. Conclusion Quantifying the γ-H2AX loci in normal tissue represents a sensitive tool for the detection of induction and repair of radiation-induced DSBs at clinically relevant doses in vivo.

20.
Progress in Biochemistry and Biophysics ; (12)2006.
Artigo em Chinês | WPRIM | ID: wpr-592952

RESUMO

Epigenetic changes are important etiological factors of human tumor. The integrity of the genome is frequently challenged by the damage of DNA. However, the highly condensed structure of chromatin imposes significant obstacles on the repair processes. Eukaryotes have developed intricate mechanisms to overcome this repressive barrier imposed by chromatin. Covalent histone modifications and ATP-dependent chromatin remodeling play important roles in the process of DNA repair. Recent advances of the epigenetic regulations in the repair process were summarized. New findings in the cellular responses to DNA double strand breaks and how histone modifications and chromatin remodeling contributes to DNA double strand break repair were introduced. Future challenges in this field are also discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA