CHOSUN

The effect of DNA repair system on the oxidative stress-mediated apoptosis.

Metadata Downloads
Author(s)
윤차경
Issued Date
2006
Abstract
본 연구는8-oxoguanine DNA glycosylase(hOGG1)와 human 8-oxo-dGTPase(MutT) homologue1(hMTH1)의 발현 억제가 산화성 스트레스에 의한 세포 사멸사에 미치는 영향에 관한 것이다.
hOGG1은 세포 내 돌연변이체인 8-oxoG를 제거하는 효소이다. Small interfering RNA (siRNA)를 사용하여 hOGG1의 효소 기능을 억제한 섬유아세포( fibroblast GM00637)에서는 H2O2관련 세포사멸이 증가한다. 이러한 세포 사멸은 p53과 관련된 세포 사멸사 경로( apoptotic pathway)를 통한 것이다. 또한 hOGG1의 효소기능이 억제된 세포는 유전적 불안정성도 증가한다. 세포내hOGG1의 활성이 저하되더라도 p53 기능이 억제된 경우에는 같은 농도의 H2O2를 처리하였어도 세포사멸(apoptosis)이 나타나지 않았다. hOGG1의 활성이 억제된 세포에서는 H2O2를 처리 하였을 때 p21, Noxa, caspase-3/7의 활성이 증가하였지만 p53 기능을 siRNA로 억제 하였을 때는 이러한 단백질의 활성이 증가하지 않으면서 세포 생존율이 증가하였다. p53유전자가 발현되지 않는 세포인 H1299 에서 hOGG1의 효소 활성을 siRNA로 억제하고 p53을 과발현 시켰을 때, H2O2에 대한 세포 생존율은 p53이 없을 때보다 감소하였다. 섬유아세포 GM00637에서 hOGG1 효소 활성을 증가시키면, hOGG1이 정상적으로 발현되고 있을 때와 비교하여, 같은 농도의 H2O2를 처리하였을 경우 세포 생존율이 증가하고 p53활성이 감소하였다. 이러한 결과는 hOGG1이 H2O2가 유발하는 산화적 스트레스에 대항하여 유전적인 안정성을 증가시킴으로 p53이 매개하는 세포사멸로부터 세포를 보호하는 역할을 함을 시사한다.
hMTH1활성이 억제된 세포는 정상적인 세포에 비해서 H2O2관련 세포사멸(apoptosis)이 증가하게 되는데, 이러한 세포사멸은 p53 단백질이 매개하는 신호전달이 활성화된 결과이다. hMTH1활성이 억제된 세포에서는 H2O2 에 의한Noxa 단백질의 발현과 H2AX 단백질의 인산화(γ-H2AX )가 증가한다. Noxa와 p53이 기능을 하지 못하게 되면, hMTH1이 억제된 세포는 같은 농도의 H2O2에서도 정상 세포에 비해 세포사멸이 감소한다. hMTH1 활성이 억제된 세포에서 H2O2관련 세포사멸의 증가는, p53단백질의 인산화로 인한 Noxa 단백질의 과발현에 의한 것으로 추측된다. 또한 hMTH1-siRNA로 hMTH1 단백질의 발현을 억제시킨 세포에서 H2O2에 의한 유전인자의 불안정성이 증가되는 것을 확인하였다. hMTH의 활성이 억제된 세포를120일간 키웠을 때, 외부 자극 없이도 세포 내에서 발생하는 reactive oxygen species (ROS)가 증가하고 이와 동시에 p53이 매개하는 세포사멸(apoptosis)이 진행됨을 관찰하였다. 이러한 현상은 오랜 기간 동안 세포내 DNA손상이 복구 되지 못하고 축적된 결과로 추측된다. 이러한 연구 결과를 종합해 볼 때 hMTH1은 H2O2와 같은 산화성 스트레스에 의한 DNA 손상을 제거함으로써 유전인자의 안정성을 유지하고 세포사멸로부터 세포를 보호하는 중요한 역할을 하고 있는 것으로 사료된다.|Ⅰ. hOGG1-deficient fibroblasts undergo p53-dependent oxidative stress-induced apoptosis
Human 8-oxoguanine DNA glycosylase (hOGG1) is the main defense enzyme against the mutagenic effects of cellular 8-oxoG. In this study, we investigated the biological role of hOGG1 in response to H2O2-derived oxidative stress in human fibroblast GM00637 cells. A large proportion of hOGG1 silencing cells, which were generated by stably introducing small interfering RNA (siRNA), led to a significant increase in apoptotic cell death through the activation of p53-mediated apoptotic pathway, and a range of genomic instability of human fibroblasts in response to H2O2 exposure. The hOGG1-depleted cells lacking p53 did not undergo apoptosis upon treatment with similar H2O2 concentrations. The p53-siRNA transfection markedly inhibited the activation of p21, Noxa and caspase-3/7, which were significantly induced by H2O2 treatment, resulting in the increased cell viability in hOGG1-deficient cells. The overexpression of p53 in p53-deficient H1299 cells lacking hOGG1 by siRNA transfection significantly caused in the decrease of their cell viability in a H2O2 dose dependent manner. The overexpression of hOGG1 in GM00637 resulted a significant increase in the cell viability and a decrease in p53 phosphorylation upon exposure to H2O2. These results indicate that hOGG1 plays an important role in protecting cells against p53-mediated apoptosis, and maintaining the genomic stability in response to H2O2-induced oxidative stress.
Ⅱ. hMTH1 knockdown by small interfering RNA increases oxidative stress-induced cell death and chromosomal instability in human fibroblast GM00637 cells
Human MTH1 (hMTH1) exhibits oxidized purine nucleotide triphosphatase activity, which repairs oxidized forms of dGTP such as 8-oxo-2 -deoxyguanosine 5 -triphosphate (8-oxo-dGTP) and 2-hydroxy-2 -deoxyadenosine 5 -triphosphate (2-OH-dATP). In this study, we investigated the biological role of hMTH1 in response to H2O2-derived oxidative stress using hMTH1-depleted GM00637 cells, which were generated by stably introducing hMTH1-siRNA. The hMTH1 deficient cells caused a significant increase in apoptotic cell death through the activation of p53-mediated apoptotic pathway in response to H2O2-oxidative stress. The level of H2O2-induced Noxa expression and H2AX phosphorylation ( -H2AX) was significantly higher in hMTH1 deficient cells than in hMTH1 proficient cells. The cells lacking Noxa and p53 did not undergo cell death in response to the similar H2O2 concentration in hMTH1-depleted cells. Apoptosis of the hMTH1-depleted cells associated with Noxa expression is dependent on p53 phosphorylation. Furthermore, the ablation of hMTH1 protein by hMTH1-siRNA occurred over a range of genomic instability of human fibroblasts in response to H2O2-oxidative stress. After the long term culture of the hMTH1-depleted cells, increased intracellular ROS generation and apoptosis. These results suggest that hMTH1 plays an important role in protecting cells against oxidative stress and maintaining the genomic stability caused by H2O2-induced DNA damage.
Alternative Title
DNA 수복효소가 산화성 스트레스에 의한 세포 사멸사에 미치는 영향
Alternative Author(s)
Youn, Cha-Kyung
Affiliation
조선대학교 대학원
Department
일반대학원 생물신소재학과
Advisor
유호진
Awarded Date
2007-02
Table Of Contents
ABSTRACT = 1
Ⅰ. hOGG1-deficient fibroblasts undergo p53-dependent oxidative stress-induced apoptosis = 1
Ⅱ. hMTH1 knockdown by small interfering RNA increases oxidative stress-induced cell death and chromosomal instability in human fibroblast GM00637cells = 2
INTRODUCTION = 4
Ⅰ. hOGG1-deficient fibroblasts undergo p53-dependent oxidative stress-induced apoptosis = 11
Ⅱ. hMTH1 knockdown by small interfering RNA increases oxidative stress-induced cell death and chromosomal instability in human fibroblast GM00637cells = 13
MATERIALS AND METHODS = 16
1. Maintenance of Cell Lines = 16
2. Plasmid Constructs of hOGG1 and transfection to GM00637 cells = 16
3. hOGG1-siRNA design, synthesis and transfection = 17
4. Western blot analysis = 17
5. Semiquantative Reverse Transcriptase-Polymerase Chain Reaction = 19
6. 8-oxoG Glycosylase Activity Assay( Endonuclease Nicking Assay) = 19
7. Cytotoxicity Assay by trypan blue = 20
8. Cytotoxicity Assay by MTT = 20
9. Flow cytometry by PI staining = 21
10. Caspase-3/7 activity assay = 21
11. p53-siRNA transfection and Cytotoxicity Assay by MTT = 21
12. Transfection with p53 plasmid and Cytotoxicity Assay by MTT = 22
13. Bacterial artificial chromosome (BAC)-array comparative genomic hybridization (array-CGH) = 22
14. Statistical analysis = 23
15. Maintenance of Cell Lines = 24
16. Western blot analysis = 24
17. Semiquantative Reverse Transcriptase-Polymerase Chain Reaction = 25
18. hMTH1-siRNA design, synthesis and transfection = 25
19. Cytotoxicity Assay by trypan blue = 26
20. Cytotoxicity Assay by MTT = 27
21. Flow cytometry by PI staining = 27
22. Noxa-siRNA transfection and Cytotoxicity Assay by MTT = 27
23. p53-siRNA transfection and Cytotoxicity Assay by MTT = 28
24. Immunolocalization of phosphorylated-H2AX (H2AX) = 28
25. Measurement of intracellular ROS by FACS = 29
26. Bacterial artificial chromosome (BAC)-array comparative genomic hybridization (array-CGH) = 29
27. Statistical analysis = 30
RESULTS = 31
Ⅰ. hOGG1-deficient fibroblasts undergo p53-dependent oxidative stress-induced apoptosis = 31
1. Suppression of hOGG1 gene expression enhances the cytotoxic effects of H2O2 in human fibroblast GM00637 = 31
2. hOGG1-knockdown GM00637 cells lead to the significant increases of caspase-3 and caspase-7 activities in response to H2O2-oxidative stress = 35
3. Requirement of p53 activation for H2O2-induced cell death in hOGG1 deficient cells = 38
4. hOGG1 overexpression plays an important role to protect GM00637 against H2O2-induced apoptosis = 44
5. Involvement of ATM and DNA-PK in the H2O2-induced p53 phosphorylation of hOGG1-deficient GM00637 cells = 78
6. Array CGH characterization of the hOGG1 deficient fibroblasts following H2O2 treatment = 50
Ⅱ. hMTH1 knockdown by small interfering RNA increases oxidative stress-induced cell death and chromosomal instability in human fibroblast GM00637 cells = 53
7. The inhibition of hMTH1 expression by siRNA leads to an increase of H2O2-induced cytotoxicity in GM00637 = 53
8. hMTH1 knockdown augments the expression of Noxa, but it does not affect p53 phosphorylation and caspase-3/7 activation in response to H2O2-oxidative stress = 57
9. Noxa and p53 knockdown in hMTH1 deficient cells restores the decreased cell viability in response to H2O2-oxidative stress = 60
10. Requirement of p53 for Noxa expression and H2O2-induced cell death in hMTH1 deficient cells = 63
11. DNA damage accumulation induced intra cellular ROS and apoptosis = 66
12. hMTH1 knockdown increases H2O2-induced histone H2AX phosphorylation in GM00637 cells = 72
13. Increased chromosomal instability in hMTH1-siRNA transfected GM00637 = 74
DISCUSSION = 77
Ⅰ. hOGG1-deficient fibroblasts undergo p53-dependent oxidative stress-induced apoptosis = 77
Ⅱ. hMTH1 knockdown by small interfering RNA increases oxidative stress-induced cell death and chromosomal instability in human fibroblast GM00637 cells = 84
REFERENCES = 91
Degree
Doctor
Publisher
조선대학교 대학원
Citation
윤차경. (2006). The effect of DNA repair system on the oxidative stress-mediated apoptosis.
Type
Dissertation
URI
https://oak.chosun.ac.kr/handle/2020.oak/6519
http://chosun.dcollection.net/common/orgView/200000233890
Appears in Collections:
General Graduate School > 4. Theses(Ph.D)
Authorize & License
  • AuthorizeOpen
  • Embargo2008-09-10
Files in This Item:

Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.