• 검색 결과가 없습니다.

본 연구는 이독성 약물인 젠타마이신과 시스플라틴에 의해 난청이 유발된 렛트 와우 조직, 청각 유모세포 및 특이 억제제를 사용한 실험결과들을 바탕으로 3 가지 세포사멸 기전 (아포토시스, 오토파지 그리고 네크로토시스)을 조망해 본 바, 종합적으로 젠타마이신은 주로 아포토시스와 오토파지 세포사멸

기전을 활성화시키고, 시스플라틴은 네크로토시스와 일부 오토파지를

활성화시킴으로써 서로 다른 세포사멸 기전이 복합적으로 발생되어 이독성 난청이 야기되는 것을 밝혔다.

참고 문헌

1. Alam SA, Ikeda K, Oshima T, Suzuki M, Kawase T, Kikuchi T, Takasaka T:

Cisplatin-induced apoptotic cell death in Mongolian gerbil cochlea. Hear Res 141:

28-38, 2000

2. Barth S, Glick D, Macleod KF: Autophagy: assays and artifacts. J Pathol 221:

117-124, 2010

3. Bas E, Van De Water TR, Gupta C, Dinh J, Vu L, Martinez-Soriano F, Lainez JM, Marco J: Efficacy of three drugs for protecting against gentamicin-induced hair cell and hearing losses. Br J Pharmacol 166: 1888-1904, 2012

4. Boveris A, Chance B: The mitochondrial generation of hydrogen peroxide.

General properties and effect of hyperbaric oxygen. Biochem J 134: 707-716, 1973 5. Brady HR, Kone BC, Stromski ME, Zeidel ML, Giebisch G, Gullans SR:

Mitochondrial injury: an early event in cisplatin toxicity to renal proximal tubules.

Am J Physiol 258: F1181-1187, 1990

6. Chen FQ, Zheng HW, Schacht J, Sha SH: Mitochondrial peroxiredoxin 3 regulates sensory cell survival in the cochlea. PLoS One 8: e61999, 2013

7. Chirtes F, Albu S: Prevention and restoration of hearing loss associated with the use of cisplatin. Biomed Res Int 2014: 925485, 2014

8. Cho HC, Kashiwakura Y, Marban E: Creation of a biological pacemaker by cell fusion. Circ Res 100: 1112-1115, 2007

9. Clarke PG, Puyal J: Autophagic cell death exists. Autophagy 8: 867-869, 2012 10. Cory S, Adams JM: The Bcl2 family: regulators of the cellular life-or-death switch.

Nat Rev Cancer 2: 647-656, 2002 Wisconsin. The Epidemiology of Hearing Loss Study. Am J Epidemiol 148: 879-886, 1998

13. Cui H, Zhu Y, Yang Q, Zhao W, Zhang S, Zhou A, Jiang D: Necrostatin-1 treatment inhibits osteocyte necroptosis and trabecular deterioration in ovariectomized rats. Sci Rep 6: 33803, 2016

14. Elmore S: Apoptosis: a review of programmed cell death. Toxicol Pathol 35: 495-516, 2007

15. Fiers W, Beyaert R, Boone E, Cornelis S, Declercq W, Decoster E, Denecker G, Depuydt B, De Valck D, De Wilde G, Goossens V, Grooten J, Haegeman G, Heyninck K, Penning L, Plaisance S, Vancompernolle K, Van Criekinge W, Vandenabeele P, Vanden Berghe W, Van de Craen M, Vandevoorde V, Vercammen D: TNF-induced intracellular signaling leading to gene induction or to cytotoxicity by necrosis or by apoptosis. J Inflamm 47: 67-75, 1995

16. Finkel T: Signal transduction by reactive oxygen species. J Cell Biol 194: 7-15, 2011

17. Franklin CA, Franklin TC, Franklin JL: Comparing pure-, pulsed, and warbled tone thresholds in adults at 3000 and 6000 Hz. Am J Audiol 20: 42-47, 2011 18. Garetz SL, Altschuler RA, Schacht J: Attenuation of gentamicin ototoxicity by

glutathione in the guinea pig in vivo. Hear Res 77: 81-87, 1994

19. Giordano P, Lorito G, Ciorba A, Martini A, Hatzopoulos S: Protection against cisplatin ototoxicity in a Sprague-Dawley rat animal model. Acta Otorhinolaryngol Ital 26: 198-207, 2006

20. Goodall ML, Cramer SD, Thorburn A: Autophagy complexes cell death by necroptosis. Oncotarget 7: 50818-50819, 2016

21. Green DR, Reed JC: Mitochondria and apoptosis. Science 281: 1309-1312, 1998 22. Hacker G: The morphology of apoptosis. Cell Tissue Res 301: 5-17, 2000

23. Hawkins E, Jr.: Ototoxic mechanisms. A working hypothesis. Audiology 12: 383-393, 1973

24. He C, Klionsky DJ: Regulation mechanisms and signaling pathways of autophagy.

Annu Rev Genet 43: 67-93, 2009

25. Henderson D, Bielefeld EC, Harris KC, Hu BH: The role of oxidative stress in noise-induced hearing loss. Ear Hear 27: 1-19, 2006

26. Hetz CA, Torres V, Quest AF: Beyond apoptosis: nonapoptotic cell death in physiology and disease. Biochem Cell Biol 83: 579-588, 2005

27. Heyer N, Morata TC, Pinkerton LE, Brueck SE, Stancescu D, Panaccio MP, Kim H, Sinclair JS, Waters MA, Estill CF, Franks JR: Use of historical data and a novel metric in the evaluation of the effectiveness of hearing conservation program components. Occup Environ Med 68: 510-517, 2011

28. Hirsch T, Marchetti P, Susin SA, Dallaporta B, Zamzami N, Marzo I, Geuskens M, Kroemer G: The apoptosis-necrosis paradox. Apoptogenic proteases activated after mitochondrial permeability transition determine the mode of cell death. Oncogene 15: 1573-1581, 1997

29. Hoeppner DJ, Hengartner MO, Schnabel R: Engulfment genes cooperate with ced-3 to promote cell death in Caenorhabditis elegans. Nature 412: 202-206, 2001 30. Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL,

Schneider P, Seed B, Tschopp J: Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol 1: 489-495, 2000

31. Igney FH, Krammer PH: Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer 2: 277-288, 2002

32. Ischiropoulos H, Beckman JS: Oxidative stress and nitration in neurodegeneration:

cause, effect, or association? J Clin Invest 111: 163-169, 2003

33. Johnsson LG, Hawkins JE, Jr., Kingsley TC, Black FO, Matz GJ:

Aminoglycoside-induced cochlear pathology in man. Acta Otolaryngol Suppl 383:

1-19, 1981

34. Kaur T, Mukherjea D, Sheehan K, Jajoo S, Rybak LP, Ramkumar V: Short interfering RNA against STAT1 attenuates cisplatin-induced ototoxicity in the rat by suppressing inflammation. Cell Death Dis 2: e180, 2011

35. Kharbanda S, Ren R, Pandey P, Shafman TD, Feller SM, Weichselbaum RR, Kufe DW: Activation of the c-Abl tyrosine kinase in the stress response to DNA-damaging agents. Nature 376: 785-788, 1995

36. Kim TW, Lee SY, Kim M, Cheon C, Jang BH, Shin YC, Ko SG: DSGOST regulates resistance via activation of autophagy in gastric cancer. Cell Death Dis 9:

649, 2018

37. Kim YJ, Tian C, Kim J, Shin B, Choo OS, Kim YS, Choung YH: Autophagic flux, a possible mechanism for delayed gentamicin-induced ototoxicity. Sci Rep 7:

41356, 2017

38. Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M, Krammer PH, Peter ME: Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491-501, 1998 42. Lin FR, Albert M: Hearing loss and dementia - who is listening? Aging Ment

Health 18: 671-673, 2014

43. Lin JF, Lin YC, Tsai TF, Chen HE, Chou KY, Hwang TI: Cisplatin induces protective autophagy through activation of BECN1 in human bladder cancer cells.

Drug Des Devel Ther 11: 1517-1533, 2017

44. Liu FT, Newland AC, Jia L: Bax conformational change is a crucial step for PUMA-mediated apoptosis in human leukemia. Biochem Biophys Res Commun 310: 956-962, 2003

45. Liu Q, Zhu D, Hao B, Zhang Z, Tian Y: Luteolin promotes the sensitivity of cisplatin in ovarian cancer by decreasing PRPA1-medicated autophagy. Cell Mol Biol (Noisy-le-grand) 64: 17-22, 2018

46. Liu S, Liu H, Johnston A, Hanna-Addams S, Reynoso E, Xiang Y, Wang Z:

MLKL forms disulfide bond-dependent amyloid-like polymers to induce necroptosis. Proc Natl Acad Sci U S A 114: E7450-E7459, 2017

47. Liu W, Staecker H, Stupak H, Malgrange B, Lefebvre P, Van De Water TR:

Caspase inhibitors prevent cisplatin-induced apoptosis of auditory sensory cells.

Neuroreport 9: 2609-2614, 1998

48. Liu Y, Shoji-Kawata S, Sumpter RM, Jr., Wei Y, Ginet V, Zhang L, Posner B, Tran KA, Green DR, Xavier RJ, Shaw SY, Clarke PG, Puyal J, Levine B: Autosis is a Na+,K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc Natl Acad Sci U S A 110: 20364-20371, 2013

49. Locksley RM, Killeen N, Lenardo MJ: The TNF and TNF receptor superfamilies:

integrating mammalian biology. Cell 104: 487-501, 2001

50. Mahoney DJ, Cheung HH, Mrad RL, Plenchette S, Simard C, Enwere E, Arora V, Mak TW, Lacasse EC, Waring J, Korneluk RG: Both cIAP1 and cIAP2 regulate TNFalpha-mediated NF-kappaB activation. Proc Natl Acad Sci U S A 105: 11778-11783, 2008

51. Marino G, Niso-Santano M, Baehrecke EH, Kroemer G: Self-consumption: the interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol 15: 81-94, 2014 52. Matz GJ: Aminoglycoside cochlear ototoxicity. Otolaryngol Clin North Am 26:

705-712, 1993

53. McQuade T, Cho Y, Chan FK: Positive and negative phosphorylation regulates RIP1- and RIP3-induced programmed necrosis. Biochem J 456: 409-415, 2013 54. Mizushima N, Komatsu M: Autophagy: renovation of cells and tissues. Cell 147:

728-741, 2011

55. Muhr P, Mansson B, Hellstrom PA: A study of hearing changes among military conscripts in the Swedish Army. Int J Audiol 45: 247-251, 2006

56. Muyderman H, Sims NR, Tanaka M, Fuku N, Raghupathi R, Thyagarajan D: The mitochondrial T1095C mutation increases gentamicin-mediated apoptosis.

Mitochondrion 12: 465-471, 2012

57. Ofengeim D, Yuan J: Regulation of RIP1 kinase signalling at the crossroads of inflammation and cell death. Nat Rev Mol Cell Biol 14: 727-736, 2013

58. Pierson MG, Moller AR: Prophylaxis of kanamycin-induced ototoxicity by a radioprotectant. Hear Res 4: 79-87, 1981

59. Rybak LP: Mechanisms of cisplatin ototoxicity and progress in otoprotection. Curr Opin Otolaryngol Head Neck Surg 15: 364-369, 2007

60. Rybak LP, Ramkumar V: Ototoxicity. Kidney Int 72: 931-935, 2007

61. Rybak LP, Ravi R, Somani SM: Mechanism of protection by diethyldithiocarbamate against cisplatin ototoxicity: antioxidant system. Fundam Appl Toxicol 26: 293-300, 1995

62. Schacht J: Biochemical basis of aminoglycoside ototoxicity. Otolaryngol Clin North Am 26: 845-856, 1993

63. Schatz A, Bugie E, Waksman SA: Streptomycin, a substance exhibiting antibiotic activity against gram-positive and gram-negative bacteria. 1944. Clin Orthop Relat Res: 3-6, 2005

64. Schuler M, Green DR: Mechanisms of p53-dependent apoptosis. Biochem Soc Trans 29: 684-688, 2001

65. Sha SH, Schacht J: Formation of reactive oxygen species following bioactivation of gentamicin. Free Radic Biol Med 26: 341-347, 1999a

66. Sha SH, Schacht J: Stimulation of free radical formation by aminoglycoside antibiotics. Hear Res 128: 112-118, 1999b

67. Sheth S, Mukherjea D, Rybak LP, Ramkumar V: Mechanisms of Cisplatin-Induced Ototoxicity and Otoprotection. Front Cell Neurosci 11: 338, 2017

68. Sininger YS: Auditory brain stem response for objective measures of hearing. Ear Hear 14: 23-30, 1993

69. Slee EA, Adrain C, Martin SJ: Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem 276:

7320-7326, 2001

70. Stadnicki SW, Fleischman RW, Schaeppi U, Merriam P: Cis-dichlorodiammineplatinum (II) (NSC-119875): hearing loss and other toxic effects in rhesus monkeys. Cancer Chemother Rep 59: 467-480, 1975

71. Stolz A, Ernst A, Dikic I: Cargo recognition and trafficking in selective autophagy.

Nat Cell Biol 16: 495-501, 2014

72. Sugiyama K, Yokoyama T, Ryu H, Uemura K, Miyamoto T, Shimoyama I: [Intra-operative monitoring with ABR during neurovascular decompression for VIIth and VIIIth cranial nerves: a warning sign for surgeons]. No Shinkei Geka 17: 545-553, 1989

73. Talbott EO, Findlay RC, Kuller LH, Lenkner LA, Matthews KA, Day RD, Ishii EK: Noise-induced hearing loss: a possible marker for high blood pressure in older noise-exposed populations. J Occup Med 32: 690-697, 1990

74. Tang LS, Montemayor C, Pereira FA: Sensorineural hearing loss: potential therapies and gene targets for drug development. IUBMB Life 58: 525-530, 2006 75. Tenev T, Bianchi K, Darding M, Broemer M, Langlais C, Wallberg F, Zachariou

A, Lopez J, MacFarlane M, Cain K, Meier P: The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol Cell 43: 432-448, 2011

76. Thornberry NA, Lazebnik Y: Caspases: enemies within. Science 281: 1312-1316, 1998

77. Vanden Berghe T, Hassannia B, Vandenabeele P: An outline of necrosome triggers. Cell Mol Life Sci 73: 2137-2152, 2016

78. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P:

Regulated necrosis: the expanding network of non-apoptotic cell death pathways.

Nat Rev Mol Cell Biol 15: 135-147, 2014

79. Vercammen D, Brouckaert G, Denecker G, Van de Craen M, Declercq W, Fiers W, Vandenabeele P: Dual signaling of the Fas receptor: initiation of both apoptotic and necrotic cell death pathways. J Exp Med 188: 919-930, 1998

80. Wajant H: The Fas signaling pathway: more than a paradigm. Science 296: 1635-1636, 2002

81. Wang H, Meng H, Li X, Zhu K, Dong K, Mookhtiar AK, Wei H, Li Y, Sun SC, Yuan J: PELI1 functions as a dual modulator of necroptosis and apoptosis by regulating ubiquitination of RIPK1 and mRNA levels of c-FLIP. Proc Natl Acad Sci U S A 114: 11944-11949, 2017

82. Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF, Wang FS, Wang X: Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell 54: 133-146, 2014

83. Wood ZA, Poole LB, Karplus PA: Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling. Science 300: 650-653, 2003

84. Wu W, Liu P, Li J: Necroptosis: an emerging form of programmed cell death. Crit Rev Oncol Hematol 82: 249-258, 2012

85. Xia B, Fang S, Chen X, Hu H, Chen P, Wang H, Gao Z: MLKL forms cation channels. Cell Res 26: 517-528, 2016

86. Xu Y, Ma HB, Fang YL, Zhang ZR, Shao J, Hong M, Huang CJ, Liu J, Chen RQ:

Cisplatin-induced necroptosis in TNFalpha dependent and independent pathways.

Cell Signal 31: 112-123, 2017

87. Zong WX, Thompson CB: Necrotic death as a cell fate. Genes Dev 20: 1-15, 2006

ABSTRACT

-Cell death map in drug-induced ototoxicity:

apoptosis, autophagy, and necroptosis

Mi-Jin Choi

BK21 Plus program, Department of Biomedical Sciences, Ajou University The Graduate School of Medicine, Suwon, Republic of Korea

(Supervised by Professor Yun-Hoon Choung, MD, DDS, PhD)

Introduction: Gentamicin (GM) and Cisplatin (CDDP) show various side effects such as sensorineural hearing loss and dizziness in which the mechanisms have not been identified. The purpose of the study is to analyze drug type-induced auditory cell death mechanisms including apoptosis, autophagy, and necroptosis.

Methods: In vitro study, HEI-OC1 auditory cells were cultured and treated with various doses of GM (0, 2.5, 5, and 7.5 mM) or CDDP (0, 3.25, 7.5, and 15 μM), and then analyzed by Western blot. To analyze apoptotic effects by GM- and CDDP-induced ototoxicity, cleaved PARP, and cleaved caspase-3 bands were analyzed in Western blot.

RIP1, RIP3, and MLKL were used as the markers for necroptotic effect. For autophagy markers, Beclin1, p62, and LC3 were used. The measured of GM or CDDP-induced cell death by WST-1 assay under the pre-treatment with specific cell death inhibitors

Chloroquine (10 µM), Necrostatin-1 (10 µM), Z-VAD (0.05 µM) or combinations, respectively. In vivo study, healthy 6~8-week-old male Sprague-Dawley rats (200–350 g) were used. Auditory brainstem responses (ABRs; 8, 16 and 32 kHz) were measured at the time of pre-injection and post-injection, and hearing threshold shifts were analyzed. Each dose of GM (200 mg/kg) or CDDP (16 mg/kg) was intraperitoneally injected and then analyzed by H&E staining, TUNEL, and immunofluorescence for the morphologic study.

Results: On the 6 weeks after GM injection, hearing loss was detected, with thresholds increasing to 26.3 ± 20.0 dB, 23.8 ± 18.9 dB and 27.5 ± 21.9 dB at 8, 16 and 32 kHz, respectively. At 3 days after cisplatin injection, ABR thresholds also increased to 24.4 ± 14.2 dB, 29.7 ± 16.8 dB and 30.8 ± 18.7 dB at 8, 16 and 32 kHz, compared to the control (11.7 ± 4.0 dB). In H&E stain, we found the change of nuclear morphology, such as breakdown and shrinkage, and the decreased number of spiral ganglion cells treated with GM and CDDP. In immunofluorescence, LC3B staining was much more highly expressed than RIP1 and RIP3 in spiral ganglions and lateral ligaments in GM-induced ototoxicity.

Reversely, RIP1 and RIP3 were much higher expressed than LC3B staining in spiral ganglions and lateral ligaments under CDDP. In vitro study showed GM-induced ototoxicity, the intensity of cleaved caspase-3, Bax and LC3B under GM treatment were increased, but the intensity of pMLKL was increased at 6 h single point. In CDDP-induced ototoxicity, the intensity of pMLKL increased in a time-dependent manner. Moreover, LC3B was partially increased under CDDP treatment. Cell death was significantly inhibited by pre-treated with Z-VAD (0.05 µM), CQ (10 µM), Nec-1 (10 µM) or each combinations.

Conclusion: GM and CDDP seem to have different ototoxic mechanisms; apoptosis or autophagy in GM and necroptosis in CDDP.

Keyword: Gentamicin (GM), cisplatin (CDDP), sensorineural hearing loss, drug-induced ototoxicity, apoptosis, autophagy, necroptosis

관련 문서