博碩士論文 103881005 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:8 、訪客IP:18.116.42.208
姓名 林衡道(Heng-Dao Lin)  查詢紙本館藏   畢業系所 生命科學系
論文名稱 探討DNA損傷反應與慢性暴露4-胺基聯苯產生之肝臟毒性
(DNA Damage Response and Chronic Liver Toxicity Resulting from Exposure to 4-Aminobiphenyl)
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 ( 永不開放)
摘要(中) 4-胺基聯苯 (4-ABP)是一種人類膀胱致癌物,常見於偶氮染料及香菸、環境污染物產生的煙霧中。我們先前的研究發現4-ABP可在HepG2細胞誘導DNA損傷,顯示4-ABP可對人類肝細胞造成遺傳毒性。此外,流行病學調查也間接指出4-ABP可能對人類肝臟產生影響,但目前對於4-ABP是否對於人類肝臟產生致癌作用仍具有爭議。我們的目標是研究4-ABP短期高劑量暴露下產生的DNA損傷反應之分子機制以及長期低劑量暴露後是否可誘導肝癌產生。在第一部分,我們以L-02,HepG2和Hep3B等作為測試模型探討ROS及miRNA是否可參與4-ABP在人類肝細胞誘導的DNA損傷反應。實驗結果顯示高劑量4-ABP可誘導肝細胞產生ROS,並通過調節miR-513a-5p和miR-630的表現量進而抑制DNA同源重組修復活性。在第二部分中,我們利用人類肝細胞株和斑馬魚建立了肝細胞癌變模型,以模擬長期暴露於低劑量的4-ABP(1 nM-10 µM)的肝癌發展。結果表明,低劑量4-ABP長期暴露可促進肝細胞增殖和遷移。此外, HBx, Src(p53-)轉基因斑馬魚暴露3個月4-ABP後及野生型斑馬魚中暴露6個月4-ABP都發現肝細胞癌形成。我們同時也發現細胞及斑馬魚模型中Ras-ERK途徑活化與4-ABP誘導的肝細胞癌之間的相關性。這些結果表明4-ABP可對肝臟產生遺傳毒性並促進肝細胞癌進展。據我們所知,這是首份關於慢性4-ABP暴露後促進人類細胞和斑馬魚產生肝癌之研究。
摘要(英) 4-Aminobiphenyl (4-ABP) is a recognized human urinary bladder carcinogen that was commonly found in the manufacture of azo dyes, the compositions of cigarette smoke or polluted air. In our previous study, we found 4-ABP can induce DNA damage in HepG2 cells. Many epidemiologic studies have investigated the potential effects of 4-ABP on liver, but the oncogenic effect of 4-ABP on human liver remains controversial. Our purpose is to investigate the molecular mechanism of DNA damage response after acute 4-ABP treatment and whether 4-ABP induced liver carcinogenesis after chronic exposure. In the first part of our studies, we used human liver cells like L-02, HepG2 and Hep3B as a test model to investigate whether the ROS and miRNA affect DNA damage response during exposure to various concentrations of 4-ABP (75-300 µM) for 24 h. Our results showed that 4-ABP induces ROS production which inhibiting DNA homologous recombination (HR) repair activity via regulated miR-513a-5p and miR-630 expression. In the second part, we have established a liver cell carcinogenesis model in human liver cell lines and zebrafish to mimic liver cancer development associated with long-term exposure to low doses of 4-ABP (1 nM-10 µM). The results demonstrated that chronic 4-ABP exposure promoted cellular proliferation and migration in vitro. In addition, 4-ABP induces hepatocellular carcinoma (HCC) formation in HBx, Src (p53-) transgenic zebrafish at four months of age and in wild-type zebrafish at seven months of age. We also observed a correlation between the Ras-ERK pathway and 4-ABP-induced HCC in vitro and in vivo. These results indicated that 4-ABP has effects on genotoxicity in liver and HCC progression. To our knowledge, this is the first report on the promotion of liver carcinogenesis in human cells and zebrafish following 4-ABP exposure.
關鍵字(中) ★ 4-胺基聯苯
★ 微小RNA
★ DNA修補
★ 氧化壓力
★ 肝細胞癌
★ 斑馬魚
關鍵字(英) ★ 4-Aminobiphenyl
★ microRNA
★ DNA repair
★ oxidative stress
★ HCC
★ zebrafish
論文目次 Table of Contents
Page
Declaration I
Publications II
(A). Referred papers: II
(B). Abstracts presented in meetings: III
中文摘要 IV
Abstract V
Acknowledgments VI
Table of Contents VII
List of Figures X
List of Tables XIV
Abbreviations XV
Chapter 1: General Introduction 1
1-1. 4-Aminobiphenyl (4-ABP) 1
Overview of 4-ABP 1
Bioactivation and genotoxicity of 4-ABP 2
Genetic toxicity assessment of aromatic amine using liver cells 4
The association between miRNA and DNA repair during exposure to 4-ABP 5
The potential link between 4-ABP and HCC 5
1-2. The interplay between ROS and miRNA in DNA damage response 7
Reactive oxygen species (ROS) 7
Micro RNA (miRNA) 8
ROS and miRNA 9
1-3. DNA damage response (DDR) 9
Aromatic amines and DNA damage 9
DNA damage response 10
DNA double-strand breaks repair 10
The role of p53 in DNA damage response 12
The role of MCM8-9 complex in DNA repair 13
1-4. Human liver cancer 14
Overview of liver cancer 14
The risk factor of HCC 14
Oxidative stress and inflammation contribute to HCC 15
1-5. Zebrafish liver cancer model 16
Overview of Zebrafish in liver disease model 16
Zebrafish cancer model 17
Xenotransplantation of zebrafish 18
1-6. Specific aim of the thesis 18
Chapter 2: Material and methods 21
2-1. Cell culture and transfection 21
2-2. Cell viability assay 21
2-3. Comet assay 21
2-4. RNA extraction, cDNA synthesis and qPCR 22
2-5. miRNA extraction 23
2-6. miRNA qPCR 23
2-7. Luciferase assay 23
2-8. ChIP assay 24
2-9. DNA HR activity assay 24
2-10. Cell cycle analysis 25
2-11. ROS assay 25
2-12. Protein extraction and Western blot 25
2-13. Colony formation assay 26
2-14. Soft agar formation 26
2-15. BrdU assay 27
2-16. Transwell assay 27
2-17. Zebrafish Maintenance and Transgenic Zebrafish Lines 27
2-18. Embryos Collection 28
2-19. Xenotransplantation of Hepatoma in Zebrafish 29
2-20. Body length, Weight Measurement and Liver Collection 29
2-21. Hematoxylin and eosin (H&E) staining 30
2-22. Immunohistochemistry Staining 31
2-23. Isolation of Zebrafish liver and RNA extraction 32
2-24. Hepatotoxicity test in zebrafish embryo 32
2-25. Embryotoxicity test 33
2-26. Statistical analysis 33
Chapter 3: Result 35
3-1. The epigenetic regulation of HR repair during exposure to 4-ABP 35
4-ABP-induced DNA double-strand breaks in liver cell lines 35
4-ABP regulated miRNA expression 35
4-ABP inhibit DNA HR repair via miR-513a-5p and miR-630 37
ROS plays a critical role in 4-ABP-induced DNA damage response 38
3-2. Chronic exposure to 4-ABP induce or promote hepatocarcinogenesis in vitro and in vivo 40
Effect of 4-ABP exposure on liver cells 40
Zebrafish xenotransplation model for 4-ABP chronic treated cells 42
H&E staining and IHC analysis of 4-ABP chronic treated zebrafish 43
Chapter 4: Discussion 47
4-1. Whether L-02, HepG2 and Hep3B cells can be used as a model to assessed the genotoxicity mechanism of 4-ABP? 47
4-2. DNA damage response and miR-513a-5p with 4-ABP treatment 48
4-3. DNA damage response and miR-630 with 4-ABP treatment 49
4-4. Oxidative stress and DNA damage response with 4-ABP treatment 52
4-5. The concentration of -4-ABP in the acute and chronic study 52
4-6. In vitro model of 4-ABP chronic exposure and xenotransplantation of the zebrafish embryo 53
4-7. Cytochrome P450 family in zebrafish 55
4-8. Chronic exposure to 4-ABP induce or promote hepatocarcinogenesis in the zebrafish model 56
Summary 57
Reference 58
Figures 77



List of Figures
Figure 1. Effects of 4-ABP on human liver cell viability. 77
Figure 2. Effect of 4-ABP on DNA damage in human liver cells. 78
Figure 3. Effect of 4-ABP on DNA DSBs in liver cells. 79
Figure 4. 4-ABP induces DSBs in liver cells. 80
Figure 5. Dysregulation of miR-513a-5p or miR-630 in liver cells after 4-ABP treatment. 81
Figure 6. p53 was predicted to bind to the promoter of miR-513a-5p. 82
Figure 7. CREB was predicted to bind to the 5’-UTR of miR-630 host gene ARIH1 and attenuated by NAC pretreatment during 4-ABP exposure. 83
Figure 8. miR-513a-5p mediates p53 expression in liver cells with 4-ABP treatment. 85
Figure 9. miR-513a-5p inhibitor or TP53 overexpression partially rescues DNA damage during exposure to 4-ABP. 87
Figure 10. Validation of genes involved in the p53 signaling pathway by qPCR. 88
Figure11. Validation of genes involved in the DNA double-strand break repair pathway. 89
Figure 12. miR-630 mediates MCM8 expression in liver cells with 4-ABP treatment. 90
Figure 13. RAD18 and MCM8 overexpression or miR-630 inhibitor partially inhibit 4-ABP-induced DNA DSBs. 91
Figure 14. miR-513a-5p inhibitor or TP53 overexpression partially rescues homologous recombination activity during 4-ABP exposure. 92
Figure 15. RAD18 and MCM8 overexpression or miR-630 inhibitor partially rescue DNA HR activity during 4-ABP exposure. 93
Figure 16. Effects of 4-ABP, miR-630 mimic or miR-630 inhibitor on the expression of MCM9. 94
Figure 17. 4-ABP induces S phase arrest in HepG2 and L-02 cells. 95
Figure 18. Transfection of pLenti-p53 or miR-513a-5p inhibitor rescue HepG2 or L-02 cells survival. 96
Figure 19. Transfection of pLenti-RAD18 or miR-630 inhibitor rescue HepG2 or Hep3B cells survival. 97
Figure 20. The ROS scavenger NAC suppressed ROS generation in liver cells during 4-ABP exposure. 98
Figure 21. NAC suppressed 4-ABP-induced DNA damage in liver cells. 99
Figure 22. NAC suppressed 4-ABP-induced DNA DSBs in liver cells. 100
Figure 23. NAC partially rescued DNA HR activity in liver cells during 4-ABP exposure. 101
Figure 24. NAC partially attenuates 4-ABP-induced miR-513a-5p and miR-630 expression in liver cells. 102
Figure 25. NAC partially rescued p53, RAD18 or MCM8 expression level in liver cells during 4-ABP exposure. 103
Figure 26. The effects of NAC on the expression of p53 or CREB during exposure to 4-ABP. 104
Figure 27. The effect of 4-ABP repeated treatment on human liver cells. 105
Figure 28. Progression of carcinogenesis induced by 4-ABP. 106
Figure 29. 4-ABP exposure for 20 cycles induced cell proliferation and migration. 108
Figure 30. GO (gene ontology) analysis of the differentially expressed genes in HepG2-ABP20 cells. 109
Figure 31. The KEGG enrichment analysis of differentially expressed genes in HepG2-ABP20 cells. 110
Figure 32. Overview of significant pathways in network marker in HepG2-ABP20 cells. 111
Figure 33. 4-ABP induced H-Ras expression level with 4-ABP repeated treatment. 112
Figure 34. 4-ABP exposure for 20 cycles upregulated RAS-MEK-ERK pathway protein expression level. 113
Figure 35. Low dose of 4-ABP induced ROS level in human liver cells. 114
Figure 36. NAC partially attenuates 4-ABP-induced induced carcinogenesis, cell proliferation and migration. 115
Figure 37. NAC partially attenuates 4-ABP-induced RAS protein expression level. 117
Figure 38. NAC partially attenuates 4-ABP-induced Ras-MEK-ERK pathway protein expression level. 118
Figure 39. 4-ABP repeated treatment increased human liver cell lines proliferation in the zebrafish embryo. 119
Figure 40. Embryonic toxicity assay for 4-ABP treatment of wild-type zebrafish or Tg(fabp10a: HBx, src, p53-/-). 121
Figure 41. LC50 test for 4-ABP treatment of wild-type zebrafish or Tg(fabp10a: HBx, src, p53-/-). 122
Figure 42. Hepatoxicity assay for 4-ABP treatment of WT zebrafish or Tg(fabp10a: HBx, src, p53-/-). 123
Figure 43. WT zebrafish or Tg(fabp10a: HBx, src, p53-/-) as model organism for studies of chronic exposure to 4-ABP. 124
Figure 44. 4-ABP induced hepatocarcinogenesis in WT or Tg(fabp10a: HBx, src, p53-/-) at 7 months of age. 125
Figure 45. Expression analysis of cell proliferation relating genes in liver of WT or Tg(fabp10a: HBx, src, p53-/-) at 4 and 7 months of age during exposure to 4-ABP. 126
Figure 46. The immunoreactive score (IRS) of immunohistochemistry for the proliferating cell nuclear antigen (PCNA) was greater in 4-ABP treatment group than in control group at 4 and 7 months of age. 127
Figure 47. Expression of inflammatory genes s in the liver of WT or Tg(fabp10a: HBx, src, p53-/-) at 4 and 7 months of age during exposure to 4-ABP. 128
Figure 48. The immunoreactive score (IRS) of immunohistochemistry for the p-ERK was greater in 4-ABP treatment group than in control group at 4 and 7 months of age. 129


List of Tables
Table 1. List of primer sequences for qPCR (F: Forward, R: Reverse) 130
Table 2. List of primer sequences for qPCR in zebrafish study 132
Table 3. List of gene-specific PCR primer sequences for cloning 133
Table 4. List of primer sequences for ChIP-qPCR 134

參考文獻 Reference
1. Vineis P, Pirastu R. Aromatic amines and cancer. Cancer Causes Control. 1997;8(3):346–55.
2. Wang L-F. Mutagenicity and aromatic amine content of fumes from heated cooking oils produced in Taiwan. Food Chem Toxicol. 1999;37(2–3):125–34.
3. Makena PS, Chung K-T. Evidence that 4‐aminobiphenyl, benzidine, and benzidine congeners produce genotoxicity through reactive oxygen species. Environ Mol Mutagen. 2007;48(5):404–13.
4. Torino JL, Burger MS, Reznikoff CA, Swaminathan S. Role of TP53 in repair of N-(deoxyguanosin-8-yl)-4-aminobiphenyl adducts in human transitional cell carcinoma of the urinary bladder. Carcinogenesis. 2001;22(1):147–54.
5. Murata M, Kawanishi S. Mechanisms of oxidative DNA damage induced by carcinogenic arylamines. Front Biosci J Virt Lib. 2011;16:1132–43.
6. David BA. The health consequences of smoking—50 years of progress. A Report of the Surgeon General. 2014.
7. Wang L-Y, Chen C-J, Zhang Y-J, Tsai W-Y, Lee P-H, Feitelson MA, et al. 4-Aminobiphenyl DNA damage in liver tissue of hepatocellular carcinoma patients and controls. Am J Epidemiol. 1998;147(3):315–23.
8. Penning TM. Metabolic activation of chemical carcinogens. Chemical Carcinogenesis. 2011; p. 135–158.
9. Moonen HJ, Briedé JJ, van Maanen JM, Kleinjans JC, de Kok TM. Generation of free radicals and induction of DNA adducts by activation of heterocyclic aromatic amines via different metabolic pathways in vitro. Mol Carcinog. 2002;35(4):196–203.
10. Walker K, Ginsberg G, Hattis D, Johns DO, Guyton KZ, Sonawane B. Genetic polymorphism in N-acetyltransferase (NAT): population distribution of NAT1 and NAT2 activity. J Toxicol Environ Health Part B. 2009;12(5–6):440–72.
11. Turesky RJ, Le Marchand L. Metabolism and biomarkers of heterocyclic aromatic amines in molecular epidemiology studies: lessons learned from aromatic amines. Chem Res Toxicol. 2011;24(8):1169–214.
12. Schut HA, Snyderwine EG. DNA adducts of heterocyclic amine food mutagens: implications for mutagenesis and carcinogenesis. Carcinogenesis. 1999;20(3):353–68.
13. Baird WM, Hooven LA, Mahadevan B. Carcinogenic polycyclic aromatic hydrocarbon‐DNA adducts and mechanism of action. Environ Mol Mutagen. 2005;45(2‐3):106–14.
14. Gupta PK. Fundamentals of toxicology: essential concepts and applications. Academic Press. 2016.
15. Cimino MC. Comparative overview of current international strategies and guidelines for genetic toxicology testing for regulatory purposes. Environ Mol Mutagen. 2006;47(5):362–90.
16. Jena GB, Kaul CL, Ramarao P. Genotoxicity testing, a regulatory requirement for drug discovery and development: impact of ICH guidelines. Indian J Pharmacol. 2002;34(2):86–99.
17. Heinonen T, Louekari K, Tähti H. Need for harmonized strategies and improved assessment of carcinogenic and genotoxic potencies of chemical substances. J Transl Toxicol. 2014;1(1):76–87.
18. Nesslany F. The current limitations of in vitro genotoxicity testing and their relevance to the in vivo situation. Food Chem Toxicol. 2017;106:609–15.
19. Corvi R, Madia F. In vitro genotoxicity testing–Can the performance be enhanced? Food Chem Toxicol. 2017;106:600–8.
20. Ehrlich V, Darroudi F, Uhl M, Steinkellner H, Gann M, Majer BJ, et al. Genotoxic effects of ochratoxin A in human-derived hepatoma (HepG2) cells. Food Chem Toxicol. 2002;40(8):1085–90.
21. Knasmüller S, Mersch-Sundermann V, Kevekordes S, Darroudi F, Huber WW, Hoelzl C, et al. Use of human-derived liver cell lines for the detection of environmental and dietary genotoxicants; current state of knowledge. Toxicology. 2004;198(1–3):315–28.
22. Marnett LJ. Oxyradicals and DNA damage. Carcinogenesis. 2000;21(3):361–70.
23. Burger MS, Torino JL, Swaminathan S. DNA damage in human transitional cell carcinoma cells after exposure to the proximate metabolite of the bladder carcinogen 4‐aminobiphenyl. Environ Mol Mutagen. 2001;38(1):1–11.
24. Chen T. The role of MicroRNA in chemical carcinogenesis. J Environ Sci Health Part C. 2010;28(2):89–124.
25. Wang SC, Chung J-G, Chen C-H, Chen S-C. 2-and 4-Aminobiphenyls induce oxidative DNA damage in human hepatoma (HepG2) cells via different mechanisms. Mutat Res Mol Mech Mutagen. 2006;593(1–2):9–21.
26. Lin C-H, Wu J-C, Chiou B-H, Chen C-H, Ma N, Chang CY, et al. MicroRNA regulation of DNA repair gene expression in 4-aminobiphenyl-treated HepG2 cells. Toxicology. 2014;322:69–77.
27. Bartek J, Lukas J, Bartkova J. DNA damage response as an anti-cancer barrier: damage threshold and the concept of’conditional haploinsufficiency’. Cell Cycle. 2007;6(19):2344–7.
28. Roos WP, Thomas AD, Kaina B. DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer. 2016;16(1):20.
29. Patil P, Patil K, Hande S, Pawar V, Mujumdar A, Mhatre A. Conceptual study of Dooshivisha (Cumulative toxicity) wrt Ancient classics and Modern science. J Ayurveda Integr Med. 2016;1(3):144–50.
30. Ambs S, Neumann H-G. Acute and chronic toxicity of aromatic amines studied in the isolated perfused rat liver. Toxicol Appl Pharmacol. 1996;139(1):186–94.
31. Larcher T, Perrichon P, Vignet C, Ledevin M, Le Menach K, Lyphout L, et al. Chronic dietary exposure of zebrafish to PAH mixtures results in carcinogenic but not genotoxic effects. Environ Sci Pollut Res. 2014;21(24):13833–49.
32. Wang S, Sugamori KS, Tung A, McPherson JP, Grant DM. N-hydroxylation of 4-aminobiphenyl by CYP2E1 produces oxidative stress in a mouse model of chemically induced liver cancer. Toxicol Sci. 2015;144(2):393–405.
33. Yu MC, Skipper PL, Taghizadeh K, Tannenbaum SR, Chan KK, Henderson BE, et al. Acetylator phenotype, aminobiphenyl-hemoglobin adduct levels, and bladder cancer risk in white, black, and Asian men in Los Angeles, California. JNCI J Natl Cancer Inst. 1994;86(9):712–6.
34. Ross RK, Jones PA, Yu MC. Bladder cancer epidemiology and pathogenesis. In seminars in oncology. 1996; p. 536.
35. Mimi CY, Skipper PL, Tannenbaum SR, Chan KK, Ross RK. Arylamine exposures and bladder cancer risk. Mutat Res Mol Mech Mutagen. 2002;506:21–8.
36. Chen S-Y, Wang L-Y, Lunn RM, Tsai W-Y, Lee P-H, Lee C-S, et al. Polycyclic aromatic hydrocarbon‐DNA adducts in liver tissues of hepatocellular carcinoma patients and controls. Int J Cancer. 2002;99(1):14–21.
37. Lee Y-CA, Cohet C, Yang Y-C, Stayner L, Hashibe M, Straif K. Meta-analysis of epidemiologic studies on cigarette smoking and liver cancer. Int J Epidemiol. 2009;38(6):1497–511.
38. D’Autréaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007;8(10):813–24.
39. Amer J, Ghoti H, Rachmilewitz E, Koren A, Levin C, Fibach E. Red blood cells, platelets and polymorphonuclear neutrophils of patients with sickle cell disease exhibit oxidative stress that can be ameliorated by antioxidants. Br J Haematol. 2006;132(1):108–13.
40. Bardaweel SK, Gul M, Alzweiri M, Ishaqat A, ALSalamat HA, Bashatwah RM. Reactive oxygen species: the dual role in physiological and pathological conditions of the human body. Eurasian J Med. 2018;50(3):193.
41. Hall EJ, Rossi HH, Kellerer AM, Goodman L, Marino S. Radiobiological studies with monoenergetic neutrons. Radiat Res. 1973;54(3):431–43.
42. Salehi F, Behboudi H, Kavoosi G, Ardestani SK. Oxidative DNA damage induced by ROS-modulating agents with the ability to target DNA: A comparison of the biological characteristics of citrus pectin and apple pectin. Sci Rep. 2018;8(1):1–16.
43. Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, Alexeyev MF. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 2009;37(8):2539–48.
44. Bravard A, Vacher M, Gouget B, Coutant A, de Boisferon FH, Marsin S, et al. Redox regulation of human OGG1 activity in response to cellular oxidative stress. Mol Cell Biol. 2006;26(20):7430–6.
45. Lee BWL, Ghode P, Ong DST. Redox regulation of cell state and fate. Redox Biol. 2019;25:101056.
46. Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med. 2012;4(3):143–59.
47. Wan G, Mathur R, Hu X, Zhang X, Lu X. miRNA response to DNA damage. Trends Biochem Sci. 2011;36(9):478–84.
48. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8.
49. Kim Y-K, Kim VN. Processing of intronic microRNAs. EMBO J. 2007;26(3):775–83.
50. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.
51. Wan G, Liu Y, Han C, Zhang X, Lu X. Noncoding RNAs in DNA repair and genome integrity. Antioxid Redox Signal. 2014;20(4):655–77.
52. Gooderham NJ, Koufaris C. Using microRNA profiles to predict and evaluate hepatic carcinogenic potential. Toxicol Lett. 2014;228(2):127–32.
53. Han C, Wan G, Langley RR, Zhang X, Lu X. Crosstalk between the DNA damage response pathway and microRNAs. Cell Mol Life Sci. 2012;69(17):2895–906.
54. Crosby ME, Kulshreshtha R, Ivan M, Glazer PM. MicroRNA regulation of DNA repair gene expression in hypoxic stress. Cancer Res. 2009;69(3):1221–9.
55. Moskwa P, Buffa FM, Pan Y, Panchakshari R, Gottipati P, Muschel RJ, et al. miR-182-mediated downregulation of BRCA1 impacts DNA repair and sensitivity to PARP inhibitors. Mol Cell. 2011;41(2):210–20.
56. Valeri N, Gasparini P, Fabbri M, Braconi C, Veronese A, Lovat F, et al. Modulation of mismatch repair and genomic stability by miR-155. Proc Natl Acad Sci. 2010;107(15):6982–7.
57. Lin Y, Liu X, Cheng Y, Yang J, Huo Y, Zhang C. Involvement of MicroRNAs in hydrogen peroxide-mediated gene regulation and cellular injury response in vascular smooth muscle cells. J Biol Chem. 2009;284(12):7903–13.
58. Lin J, Chuang C-C, Zuo L. Potential roles of microRNAs and ROS in colorectal cancer: diagnostic biomarkers and therapeutic targets. Oncotarget. 2017;8(10):17328.
59. He J, Jiang B-H. Interplay between reactive oxygen species and microRNAs in cancer. Curr Pharmacol Rep. 2016;2(2):82–90.
60. Kwon J-E, Kim B-Y, Kwak S-Y, Bae I-H, Han Y-H. Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis. 2013;18(7):896–909.
61. Lizarraga D, Gaj S, Brauers KJ, Timmermans L, Kleinjans JC, van Delft JH. Benzo [a] pyrene-induced changes in MicroRNA–mRNA networks. Chem Res Toxicol. 2012;25(4):838–49.
62. Perrone S, Lotti F, Geronzi U, Guidoni E, Longini M, Buonocore G. Oxidative stress in cancer-prone genetic diseases in pediatric age: the role of mitochondrial dysfunction. Oxid Med Cell Longev. 2016
63. Hammons GJ, Milton D, Stepps K, Guengerich FP, Tukey RH, Kadlubar FF. Metabolism of carcinogenic heterocyclic and aromatic amines by recombinant human cytochrome P450 enzymes. Carcinogenesis. 1997;18(4):851–4.
64. Shibutani S, Suzuki N, Tan X, Johnson F, Grollman AP. Influence of flanking sequence context on the mutagenicity of acetylaminofluorene-derived DNA adducts in mammalian cells. Biochemistry. 2001;40(12):3717–22.
65. Mu H, Kropachev K, Wang L, Zhang L, Kolbanovskiy A, Kolbanovskiy M, et al. Nucleotide excision repair of 2-acetylaminofluorene-and 2-aminofluorene-(C8)-guanine adducts: molecular dynamics simulations elucidate how lesion structure and base sequence context impact repair efficiencies. Nucleic Acids Res. 2012;40(19):9675–90.
66. Giglia-Mari G, Zotter A, Vermeulen W. DNA damage response. Cold Spring Harb Perspect Biol. 2011;3(1):a000745.
67. Harper JW, Elledge SJ. The DNA damage response: ten years after. Mol Cell. 2007;28(5):739–45.
68. Podhorecka M, Skladanowski A, Bozko P. H2AX phosphorylation: its role in DNA damage response and cancer therapy. J Nucleic Acids. 2010.
69. Weber S. Light-driven enzymatic catalysis of DNA repair: a review of recent biophysical studies on photolyase. Biochim Biophys Acta BBA-Bioenerg. 2005;1707(1):1–23.
70. Pegg AE. Multifaceted roles of alkyltransferase and related proteins in DNA repair, DNA damage, resistance to chemotherapy, and research tools. Chem Res Toxicol. 2011;24(5):618–39.
71. Mao Z, Bozzella M, Seluanov A, Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle. 2008;7(18):2902–6.
72. Weterings E, Van Gent DC. The mechanism of non-homologous end-joining: a synopsis of synapsis. DNA Repair. 2004;3(11):1425–35.
73. Rodgers K, McVey M. Error‐prone repair of DNA double‐strand breaks. J Cell Physiol. 2016;231(1):15–24.
74. Zhang J. The role of BRCA1 in homologous recombination repair in response to replication stress: significance in tumorigenesis and cancer therapy. Cell Biosci. 2013;3(1):11.
75. Li X, Heyer W-D. Homologous recombination in DNA repair and DNA damage tolerance. Cell Res. 2008 Jan;18(1):99–113.
76. Bhatti S, Kozlov S, Farooqi AA, Naqi A, Lavin M, Khanna KK. ATM protein kinase: the linchpin of cellular defenses to stress. Cell Mol Life Sci. 2011;68(18):2977.
77. Altmeyer M, Lukas J. To spread or not to spread—chromatin modifications in response to DNA damage. Curr Opin Genet Dev. 2013;23(2):156–65.
78. Chapman JR, Taylor MR, Boulton SJ. Playing the end game: DNA double-strand break repair pathway choice. Mol Cell. 2012;47(4):497–510.
79. Nimonkar AV, Genschel J, Kinoshita E, Polaczek P, Campbell JL, Wyman C, et al. BLM–DNA2–RPA–MRN and EXO1–BLM–RPA–MRN constitute two DNA end resection machineries for human DNA break repair. Genes Dev. 2011;25(4):350–62.
80. Zhao W, Vaithiyalingam S, San Filippo J, Maranon DG, Jimenez-Sainz J, Fontenay GV, et al. Promotion of BRCA2-dependent homologous recombination by DSS1 via RPA targeting and DNA mimicry. Mol Cell. 2015;59(2):176–87.
81. Holloman WK. Unraveling the mechanism of BRCA2 in homologous recombination. Nat Struct Mol Biol. 2011;18(7):748.
82. Sebesta M, Burkovics P, Juhasz S, Zhang S, Szabo JE, Lee MY, et al. Role of PCNA and TLS polymerases in D-loop extension during homologous recombination in humans. DNA Repair. 2013;12(9):691–8.
83. Ashcroft M, Kubbutat MH, Vousden KH. Regulation of p53 function and stability by phosphorylation. Mol Cell Biol. 1999;19(3):1751–8.
84. Helton ES, Chen X. p53 modulation of the DNA damage response. J Cell Biochem. 2007;100(4):883–96.
85. Pellegata NS, Antoniono RJ, Redpath JL, Stanbridge EJ. DNA damage and p53-mediated cell cycle arrest: a reevaluation. Proc Natl Acad Sci. 1996;93(26):15209–14.
86. Chen J. The cell-cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harb Perspect Med. 2016;6(3):a026104.
87. Williams AB, Schumacher B. p53 in the DNA-damage-repair process. Cold Spring Harb Perspect Med. 2016;6(5):a026070.
88. Haupt S, Berger M, Goldberg Z, Haupt Y. Apoptosis-the p53 network. J Cell Sci. 2003;116(20):4077–85.
89. Liu Y, Qiang W, Xu X, Dong R, Karst AM, Liu Z, et al. Role of miR-182 in response to oxidative stress in the cell fate of human fallopian tube epithelial cells. Oncotarget. 2015;6(36):38983.
90. Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR‐200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9(6):582–9.
91. Natsume T, Nishimura K, Minocherhomji S, Bhowmick R, Hickson ID, Kanemaki MT. Acute inactivation of the replicative helicase in human cells triggers MCM8–9-dependent DNA synthesis. Genes Dev. 2017;31(8):816–29.
92. Park J, Long DT, Lee KY, Abbas T, Shibata E, Negishi M, et al. The MCM8-MCM9 Complex Promotes RAD51 Recruitment at DNA Damage Sites To Facilitate Homologous Recombination. Mol Cell Biol. 201;33(8):1632–44.
93. Lutzmann M, Grey C, Traver S, Ganier O, Maya-Mendoza A, Ranisavljevic N, et al. MCM8- and MCM9-Deficient Mice Reveal Gametogenesis Defects and Genome Instability Due to Impaired Homologous Recombination. Mol Cell. 2012;47(4):523–34.
94. Lutzmann M, Bernex F, de Jesus C da C, Hodroj D, Marty C, Plo I, et al. MCM8-and MCM9 deficiencies cause lifelong increased hematopoietic DNA damage driving p53-dependent myeloid tumors. Cell Rep. 2019;28(11):2851-2865. e4.
95. Bosch FX, Ribes J, Díaz M, Cléries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127(5):S5–16.
96. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet‐Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.
97. Lee J-S, Thorgeirsson SS. Genome-scale profiling of gene expression in hepatocellular carcinoma: classification, survival prediction, and identification of therapeutic targets. Gastroenterology. 2004;127(5):S51–5.
98. Duan M, Hao J, Cui S, Worthley DL, Zhang S, Wang Z, et al. Diverse modes of clonal evolution in HBV-related hepatocellular carcinoma revealed by single-cell genome sequencing. Cell Res. 2018;28(3):359–73.
99. Couri T, Pillai A. Goals and targets for personalized therapy for HCC. Hepatol Int. 2019;13(2):125–37.
100. Zhang Y-J. Interactions of chemical carcinogens and genetic variation in hepatocellular carcinoma. World J Hepatol. 2010;2(3):94–102.
101. SUGIMURA T, NAGAO M, WAKABAYASHI K. How we should deal with unavoidable exposure of man to environmental mutagens : cooked food mutagen discovery, facts and lessons for cancer prevention. We Should Deal Unavoidable Expo Man Environ Mutagens Cooked Food Mutagen Discov Facts Lessons Cancer Prev. 2000;447(1):15–25.
102. Turesky RJ. Heterocyclic Aromatic Amine Metabolism, Dna Adduct Formation, Mutagenesis, and Carcinogenesis. Drug Metab Rev. 2002;34(3):625–50.
103. Autrup H. Genetic polymorphisms in human xenobiotica metabolizing enzymes as susceptibility factors in toxic response. Mutat Res Toxicol Environ. 2000;464(1):65–76.
104. Pluchino LA, Liu AK-Y, Wang H-CR. Reactive oxygen species-mediated breast cell carcinogenesis enhanced by multiple carcinogens and intervened by dietary ergosterol and mimosine. Free Radic Biol Med. 2015;80:12–26.
105. Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem J. 1996;313(1):17–29.
106. Chiba T, Marusawa H, Ushijima T. Inflammation-Associated Cancer Development in Digestive Organs: Mechanisms and Roles for Genetic and Epigenetic Modulation. Gastroenterology. 2012;143(3):550–63.
107. Marra M, Sordelli IM, Lombardi A, Lamberti M, Tarantino L, Giudice A, et al. Molecular targets and oxidative stress biomarkers in hepatocellular carcinoma: an overview. J Transl Med. 2011;9(1):171.
108. Wang Z, Li Z, Ye Y, Xie L, Li W. Oxidative Stress and Liver Cancer: Etiology and Therapeutic Targets. Oxid Med Cell Longev. 2016.
109. Vafa O, Wade M, Kern S, Beeche M, Pandita TK, Hampton GM, et al. c-Myc Can Induce DNA Damage, Increase Reactive Oxygen Species, and Mitigate p53 Function: A Mechanism for Oncogene-Induced Genetic Instability. Mol Cell. 2002;9(5):1031–44.
110. Hanczko R, Fernandez DR, Doherty E, Qian Y, Vas G, Niland B, et al. Prevention of hepatocarcinogenesis and increased susceptibility to acetaminophen-induced liver failure in transaldolase-deficient mice by N-acetylcysteine. J Clin Invest. 2009;119(6):1546–57.
111. Majumder S, Roy S, Kaffenberger T, Wang B, Costinean S, Frankel W, et al. Loss of Metallothionein Predisposes Mice to Diethylnitrosamine-Induced Hepatocarcinogenesis by Activating NF-κB Target Genes. Cancer Res. 2010 Dec 15;70(24):10265–76.
112. Marcellin P, Kutala BK. Liver diseases: A major, neglected global public health problem requiring urgent actions and large‐scale screening. Liver Int. 2018;38:2–6.
113. Garcia GR, Noyes PD, Tanguay RL. Advancements in zebrafish applications for 21st century toxicology. Pharmacol Ther. 2016;161:11–21.
114. Goessling W, Sadler KC. Zebrafish: an important tool for liver disease research. Gastroenterology. 2015;149(6):1361–77.
115. White R, Rose K, Zon L. Zebrafish cancer: the state of the art and the path forward. Nat Rev Cancer. 2013;13(9):624–36.
116. Wilkins BJ, Pack M. Zebrafish models of human liver development and disease. Compr Physiol. 2013;3(3):1213–30.
117. Phillips JB, Westerfield M. Zebrafish models in translational research: tipping the scales toward advancements in human health. Dis Model Mech. 2014;7(7):739–43.
118. Zhang B, Xuan C, Ji Y, Zhang W, Wang D. Zebrafish xenotransplantation as a tool for in vivo cancer study. Fam Cancer. 2015;14(3):487–93.
119. Mirbahai L, Williams TD, Zhan H, Gong Z, Chipman JK. Comprehensive profiling of zebrafish hepatic proximal promoter CpG island methylation and its modification during chemical carcinogenesis. BMC Genomics. 2011;12:3.
120. Basten SG, Davis EE, Gillis AJM, van Rooijen E, Stoop H, Babala N, et al. Mutations in LRRC50 predispose zebrafish and humans to seminomas. PLoS Genet. 2013;9(4):e1003384.
121. Spitsbergen JM, Tsai H-W, Reddy A, Miller T, Arbogast D, Hendricks JD, et al. Neoplasia in Zebrafish (Danio rerio) Treated with N-methyl-N’nitro-N-nitrosoguanidine by Three Exposure Routes at ifferent Developmental Stages. Toxicol Pathol. 2000;28(5):716–25.
122. Nguyen AT, Emelyanov A, Koh CHV, Spitsbergen JM, Lam SH, Mathavan S, et al. A high level of liver-specific expression of oncogenic Kras(V12) drives robust liver tumorigenesis in transgenic zebrafish. Dis Model Mech. 2011;4(6):801–13.
123. Liu W, Chen J-R, Hsu C-H, Li Y-H, Chen Y-M, Lin C-Y, et al. A zebrafish model of intrahepatic cholangiocarcinoma by dual expression of hepatitis B virus X and hepatitis C virus core protein in liver. Hepatology. 2012;56(6):2268–76.
124. Lin H-S, Huang Y-L, Wang Y-RS, Hsiao E, Hsu T-A, Shiao H-Y, et al. Identification of Novel Anti-Liver Cancer Small Molecules with Better Therapeutic Index Than Sorafenib via Zebrafish Drug Screening Platform. Cancers. 2019;11(6):739.
125. Yang W-Y, Rao P-S, Luo Y-C, Lin H-K, Huang S-H, Yang J-M, et al. Omics-based Investigation of Diet-induced Obesity Synergized with HBx, Src, and p53 Mutation Accelerating Hepatocarcinogenesis in Zebrafish Model. Cancers. 2019;11(12):1899.
126. Brown HK, Schiavone K, Tazzyman S, Heymann D, Chico TJ. Zebrafish xenograft models of cancer and metastasis for drug discovery. Expert Opin Drug Discov. 2017;12(4):379–89.
127. Chen L, Groenewoud A, Tulotta C, Zoni E, Kruithof-de Julio M, van der Horst G, et al. Chapter 17 - A zebrafish xenograft model for studying human cancer stem cells in distant metastasis and therapy response. Methods in Cell Biology. Academic Press; 201. p. 471–96.
128. Zhang B, Shimada Y, Kuroyanagi J, Nishimura Y, Umemoto N, Nomoto T, et al. Zebrafish xenotransplantation model for cancer stem-like cell study and high-throughput screening of inhibitors. Tumor Biol. 2014;35(12):11861–9.
129. Butler MA, Iwasaki M, Guengerich FP, Kadlubar FF. Human cytochrome P-450PA (P-450IA2), the phenacetin O-deethylase, is primarily responsible for the hepatic 3-demethylation of caffeine and N-oxidation of carcinogenic arylamines. Proc Natl Acad Sci. 1989;86(20):7696–700.
130. Orzechowski A, Schrenk D, Schut HAJ, Bock KW. Consequences of 3-methylcholanthrene-type induction for the metabolism of 4-aminobiphenyl in isolated rat hepatocytes. Carcinogenesis. 1994;15(3):489–94.
131. Lin H-D, Wang F-Z, Lee C-Y, Nien C-Y, Tseng Y-K, Yao C-L, et al. 4-Aminobiphenyl inhibits the DNA homologous recombination repair in human liver cells: The role of miR-630 in downregulating RAD18 and MCM8. Toxicology. 2020;440:152441.
132. Lin H-D, Yao C-L, Ou W-J, Luo Y-H, Chen SC. 4-Aminobiphenyl suppresses homologous recombination repair by a reactive oxygen species-dependent p53/miR-513a-5p/p53 loop. Toxicology. 2020;444:152580.
133. Sie Z-L, Li R-Y, Sampurna BP, Hsu P-J, Liu S-C, Wang H-D, et al. WNK1 Kinase Stimulates Angiogenesis to Promote Tumor Growth and Metastasis. Cancers. 2020;12(3).
134. Mišík M, Nersesyan A, Ropek N, Huber WW, Haslinger E, Knasmueller S. Use of human derived liver cells for the detection of genotoxins in comet assays. Mutat Res Toxicol Environ Mutagen. 2019;845:402995.
135. Di Gennaro A, Damiano V, Brisotto G, Armellin M, Perin T, Zucchetto A, et al. A p53/miR-30a/ZEB2 axis controls triple negative breast cancer aggressiveness. Cell Death Differ. 2018;25(12):2165–80.
136. Chen Y, Li Z, Dong Z, Beebe J, Yang K, Fu L, et al. 14-3-3σ contributes to radioresistance by regulating DNA repair and cell cycle via PARP1 and CHK2. Mol Cancer Res. 2017;15(4):418–28.
137. Zhang X, Xu R, Zhang C, Xu Y, Han M, Huang B, et al. Trifluoperazine, a novel autophagy inhibitor, increases radiosensitivity in glioblastoma by impairing homologous recombination. J Exp Clin Cancer Res. 2017;36(1):118.
138. Hartford SA, Luo Y, Southard TL, Min IM, Lis JT, Schimenti JC. Minichromosome maintenance helicase paralog MCM9 is dispensible for DNA replication but functions in germ-line stem cells and tumor suppression. Proc Natl Acad Sci. 2011;108(43):17702–7.
139. Yang S, Liu G. Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma. Oncol Lett. 2017;13(3):1041–7.
140. Wang Y, Pan T, Li L, Wang H, Zhang D, Yang H. Benzo(a)pyrene promotes Hep-G2 cell migration and invasion by upregulating phosphorylated extracellular signal-regulated kinase expression. Oncol Lett. 2018;15(6):8325–32.
141. Prasad S, Gupta SC, Tyagi AK. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 2017;387:95–105.
142. Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med. 2017;104:144–64.
142. Hill D, Chen L, Snaar-Jagalska E, Chaudhry B. Embryonic zebrafish xenograft assay of human cancer metastasis. F1000Research. 2018.
144. Chen H-J, Hu M-H, Xu F-G, Xu H-J, She J-J, Xia H-P. Understanding the inflammation-cancer transformation in the development of primary liver cancer. Hepatoma Res. 2018;4(7):29.
145. Refolo MG, Messa C, Guerra V, Carr BI, D’Alessandro R. Inflammatory Mechanisms of HCC Development. Cancers. 2020;12(3):641.
146. Shi J, Wang X, Lyu L, Jiang H, Zhu H-J. Comparison of protein expression between human livers and the hepatic cell lines HepG2, Hep3B, and Huh7 using SWATH and MRM-HR proteomics: Focusing on drug-metabolizing enzymes. Drug Metab Pharmacokinet. 2018;33(2):133–40.
147. Lançon A, Hanet N, Jannin B, Delmas D, Heydel J-M, Lizard G, et al. Resveratrol in human hepatoma HepG2 cells: metabolism and inducibility of detoxifying enzymes. Drug Metab Dispos. 2007;35(5):699–703.
148. Hu X, Yang T, Li C, Zhang L, Li M, Huang W, et al. Human fetal hepatocyte line, L-02, exhibits good liver function in vitro and in an acute liver failure model. In: Transplantation Proceedings. 2013. p. 695–700.
149. Wu Y, Geng X, Wang J, Miao Y, Lu Y, Li B. The HepaRG cell line, a superior in vitro model to L-02, HepG2 and hiHeps cell lines for assessing drug-induced liver injury. Cell Biol Toxicol. 2016;32(1):37–59.
150. LeCluyse EL, Madan A, Hamilton G, Carroll K, DeHaan R, Parkinson A. Expression and regulation of cytochrome P450 enzymes in primary cultures of human hepatocytes. J Biochem Mol Toxicol. 2000;14(4):177–88.
151. Gilot D, Loyer P, Corlu A, Glaise D, Lagadic-Gossmann D, Atfi A, et al. Liver protection from apoptosis requires both blockage of initiator caspase activities and inhibition of ASK1/JNK pathway via glutathione S-transferase regulation. J Biol Chem. 2002;277(51):49220–9.
152. Gerets HHJ, Tilmant K, Gerin B, Chanteux H, Depelchin Bo, Dhalluin S, et al. Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biol Toxicol. 2012;28(2):69–87.
153. He X, He L, Hannon GJ. The Guardian’s Little Helper: MicroRNAs in the p53 Tumor Suppressor Network. Cancer Res. 2007;67(23):11099–101.
154. Raver-Shapira N, Oren M. Tiny Actors, Great Roles: microRNAs in p53’s Service. Cell Cycle. 2007;6(21):2656–61.
155. Li S, Xu Y-N, Niu X, Li Z, Wang J-F. miR-513a-5p targets Bcl-2 to promote dichlorvos induced apoptosis in HK-2 cells. Biomed Pharmacother. 2018;108:876–82.
156. Hu W, Chan CS, Wu R, Zhang C, Sun Y, Song JS, et al. Negative regulation of tumor suppressor p53 by microRNA miR-504. Mol Cell. 2010;38(5):689–99.
157. Luo Z, Cui R, Tili E, Croce C. Friend or Foe: MicroRNAs in the p53 network. Cancer Lett. 2018;419:96–102.
158. Sullivan KD, Galbraith MD, Andrysik Z, Espinosa JM. Mechanisms of transcriptional regulation by p53. Cell Death Differ. 2018;25(1):133–43.
159. Hafner A, Bulyk ML, Jambhekar A, Lahav G. The multiple mechanisms that regulate p53 activity and cell fate. Nat Rev Mol Cell Biol. 2019;20(4):199–210.
160. Huang Y, Chuang A, Hao H, Talbot C, Sen T, Trink B, et al. Phospho-ΔNp63 α is a key regulator of the cisplatin-induced microRNAome in cancer cells. Cell Death Differ. 2011;18(7):1220–30.
161. Cao J-X, Lu Y, Qi J-J, An G-S, Mao Z-B, Jia H-T, et al. MiR-630 inhibits proliferation by targeting CDC7 kinase, but maintains the apoptotic balance by targeting multiple modulators in human lung cancer A549 cells. Cell Death Dis. 2014;5(9):e1426–e1426.
162. Zhang Y, Yu J, Liu H, Ma W, Yan L, Wang J, et al. Novel Epigenetic CREB-miR-630 Signaling Axis Regulates Radiosensitivity in Colorectal Cancer. PLoS One. 2015;10(8):e0133870.
163. Wang Y-W, Chen X, Ma R, Gao P. Understanding the CREB1-miRNA feedback loop in human malignancies. Tumor Biol. 2016;37(7):8487–502.
164. Shi Y, Venkataraman SL, Dodson GE, Mabb AM, LeBlanc S, Tibbetts RS. Direct regulation of CREB transcriptional activity by ATM in response to genotoxic stress. Proc Natl Acad Sci. 2004;101(16):5898–903.
165. Martin NT, Nakamura K, Davies R, Nahas SA, Brown C, Tunuguntla R, et al. ATM–Dependent MiR-335 Targets CtIP and Modulates the DNA Damage Response. PLoS Genet. 2013;9(5):e1003505.
166. Tripathi K, Mani C, Clark DW, Palle K. Rad18 is required for functional interactions between FANCD2, BRCA2, and Rad51 to repair DNA topoisomerase 1-poisons induced lesions and promote fork recovery. Oncotarget. 2016;7(11):12537–53.
167. Deriano L, Roth DB. Modernizing the Nonhomologous End-Joining Repertoire: Alternative and Classical NHEJ Share the Stage. Annu Rev Genet. 2013;47(1):433–55.
168. Jasin M, Rothstein R. Repair of Strand Breaks by Homologous Recombination. Cold Spring Harb Perspect Biol. 2013;5(11):a012740.
169. Meschini R, Berni A, Marotta E, Filippi S, Fiore M, Mancinelli P, et al. DNA Repair Mechanisms Involved in the Removal of DBPDE-Induced Lesions Leading to Chromosomal Alterations in CHO Cells. Cytogenet Genome Res. 2010;128(1–3):124–30.
170. Niziolek-Kierecka M, Dreij K, Lundstedt S, Stenius U. γH2AX, pChk1, and Wip1 as Potential Markers of Persistent DNA Damage Derived from Dibenzo[a,l]pyrene and PAH-Containing Extracts from Contaminated Soils. Chem Res Toxicol. 2012;25(4):862–72.
171. Stellas D, Souliotis VL, Bekyrou M, Smirlis D, Kirsch-Volders M, Degrassi F, et al. Benzo[a]pyrene-induced cell cycle arrest in HepG2 cells is associated with delayed induction of mitotic instability. Mutat Res Mol Mech Mutagen. 2014;769:59–68.
173. Black KA, McFarland RD, Grisham JW, Smith GJ. S-phase block and cell death in human lymphoblasts exposed to benzo[a]pyrene diol epoxide or N-acetoxy-2-acetylaminofluorene. Toxicol Appl Pharmacol. 1989;97(3):463–72.
174. Jeffy BD, Chen EJ, Gudas JM, Romagnolo DF. Disruption of Cell Cycle Kinetics by Benzo[a]pyrene: Inverse Expression Patterns of BRCA-1 and p53 in MCF-7 Cells Arrested in S and G2. Neoplasia N Y N. 2000;2(5):460–70.
175. Solhaug A, Refsnes M, Låg M, Schwarze PE, Husøy T, Holme JA. Polycyclic aromatic hydrocarbons induce both apoptotic and anti-apoptotic signals in Hepa1c1c7 cells. Carcinogenesis. 2004;25(5):809–19.
176. Dansen TB, Burgering BM. Unravelling the tumor-suppressive functions of FOXO proteins. Trends Cell Biol. 2008;18(9):421–9.
177. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49(11):1603–16.
178. Akyüz M, Ata S. Determination of aromatic amines in hair dye and henna samples by ion-pair extraction and gas chromatography-mass spectrometry. J Pharm Biomed Anal. 2008;47(1):68–80.
179. Ledda C, Loreto C, Zammit C, Marconi A, Fago L, Matera S, et al. Non‑infective occupational risk factors for hepatocellular carcinoma: A review (Review). Mol Med Rep. 2017;15(2):511–33.
180. Banerjee R, Caruccio L, Zhang YJ, McKercher S, Santella RM. Effects of carcinogen-induced transcription factors on the activation of hepatitis B virus expression in human hepatoblastoma HepG2 cells and its implication on hepatocellular carcinomas. Hepatol Baltim Md. 2000;32(2):367–74.
181. Jiangning C, Hongxia Y, Ying L, Wei J, Jie J, Junfeng Z, et al. Ecotoxicological evaluation of 4-aminobiphenyl using a test battery. Ecotoxicol Environ Saf. 2004;58(1):104–9.
182. Saletta F, Matullo G, Manuguerra M, Arena S, Bardelli A, Vineis P. Exposure to the Tobacco Smoke Constituent 4-Aminobiphenyl Induces Chromosomal Instability in Human Cancer Cells. Cancer Res. 2007;67(15):7088–94.
183. Wang S, Sugamori KS, Brenneman D, Hsu I, Calce A, Grant DM. Influence of arylamine n-acetyltransferase, sex, and age on 4-aminobiphenyl-induced in vivo mutant frequencies and spectra in mouse liver. Environ Mol Mutagen. 2012;53(5):350–7.
184. Siriwardhana N, Wang H-CR. Precancerous carcinogenesis of human breast epithelial cells by chronic exposure to benzo[a]pyrene. Mol Carcinog. 2008;47(5):338–48.
185. Li L, Zhao G-D, Shi Z, Qi L-L, Zhou L-Y, Fu Z-X. The Ras/Raf/MEK/ERK signaling pathway and its role in the occurrence and development of HCC (Review). Oncol Lett. 2016;12(5):3045–50.
186. Parsons BL, Beland FA, Von Tungeln LS, Delongchamp RR, Fu PP, Heflich RH. Levels of 4‐aminobiphenyl‐induced somatic H‐ras mutation in mouse liver DNA correlate with potential for liver tumor development. Mol Carcinog Publ Coop Univ Tex MD Anderson Cancer Cent. 2005;42(4):193–201.
187. Stanton MF. Diethylnitrosamine-induced hepatic degeneration and neoplasia in the aquarium fish, Brachydanio rerio. J Natl Cancer Inst. 1965;34(1):117–30.
188. Laale HW. Ethanol induced notochord and spinal cord duplications in the embryo of the zebrafish, Brachydanio rerio. J Exp Zool. 1971;177(1):51–64.
189. Rennekamp AJ, Peterson RT. 15 years of zebrafish chemical screening. Curr Opin Chem Biol. 2015;24:58–70.
190. Goldstone JV, McArthur AG, Kubota A, Zanette J, Parente T, Jönsson ME, et al. Identification and developmental expression of the full complement of Cytochrome P450 genes in Zebrafish. BMC Genomics. 2010;11(1):643.
191. Scornaienchi ML, Thornton C, Willett KL, Wilson JY. Functional differences in the cytochrome P450 1 family enzymes from zebrafish (Danio rerio) using heterologously expressed proteins. Arch Biochem Biophys. 2010;502(1):17–22.
192. de Souza Anselmo C, Sardela VF, de Sousa VP, Pereira HMG. Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of xenobiotics in humans? Comp Biochem Physiol Part C Toxicol Pharmacol. 2018;212:34–46.
193. Nakayama J, Gong Z. Transgenic zebrafish for modeling hepatocellular carcinoma. MedComm. 2020;1(2):140–56.
194. Berghmans S, Murphey RD, Wienholds E, Neuberg D, Kutok JL, Fletcher CDM, et al. tp53 mutant zebrafish develop malignant peripheral nerve sheath tumors. Proc Natl Acad Sci. 2005;102(2):407–12.
195. Lu J-W, Yang W-Y, Tsai S-M, Lin Y-M, Chang P-H, Chen J-R, et al. Liver-Specific Expressions of HBx and src in the p53 Mutant Trigger Hepatocarcinogenesis in Zebrafish. PLoS One. 2013 Oct 9;8(10):e76951.
196. Lam SH, Wu YL, Vega VB, Miller LD, Spitsbergen J, Tong Y, et al. Conservation of gene expression signatures between zebrafish and human liver tumors and tumor progression. Nat Biotechnol. 2006;24(1):73–5.
指導教授 陳師慶 王紹文(Ssu-Ching Chen Shao-Win Wang) 審核日期 2021-1-20
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明