$\require{mediawiki-texvc}$

연합인증

연합인증 가입 기관의 연구자들은 소속기관의 인증정보(ID와 암호)를 이용해 다른 대학, 연구기관, 서비스 공급자의 다양한 온라인 자원과 연구 데이터를 이용할 수 있습니다.

이는 여행자가 자국에서 발행 받은 여권으로 세계 각국을 자유롭게 여행할 수 있는 것과 같습니다.

연합인증으로 이용이 가능한 서비스는 NTIS, DataON, Edison, Kafe, Webinar 등이 있습니다.

한번의 인증절차만으로 연합인증 가입 서비스에 추가 로그인 없이 이용이 가능합니다.

다만, 연합인증을 위해서는 최초 1회만 인증 절차가 필요합니다. (회원이 아닐 경우 회원 가입이 필요합니다.)

연합인증 절차는 다음과 같습니다.

최초이용시에는
ScienceON에 로그인 → 연합인증 서비스 접속 → 로그인 (본인 확인 또는 회원가입) → 서비스 이용

그 이후에는
ScienceON 로그인 → 연합인증 서비스 접속 → 서비스 이용

연합인증을 활용하시면 KISTI가 제공하는 다양한 서비스를 편리하게 이용하실 수 있습니다.

유기인 계열 독성물질 분해를 위한 바이오스캐빈저 최신 연구 동향
Recent Trend in Bioscavengers for Inactivation of Toxic Organophosphorus Compounds 원문보기

공업화학 = Applied chemistry for engineering, v.31 no.2, 2020년, pp.125 - 137  

김희정 (육군사관학교 물리화학과) ,  정근홍 (육군사관학교 물리화학과) ,  계영식 (육군사관학교 물리화학과)

초록
AI-Helper 아이콘AI-Helper

최근 몇 년간 유기인 계열 독성물질이 민간인을 대상으로 사용되어 전 세계적으로 큰 위협이 되고 있다. 독성물질에 대한 예방이 불가능한 현 치료대책 대신, 보다 개선된 치료 대책으로서의 바이오스캐빈저에 대한 연구가 활발히 진행됐다. 바이오스캐빈저는 유기인 계열 독성물질이 인체 내 표적 기관에 도달하기 전, 독성물질 자체를 비활성 상태로 전환하거나 독성물질과 기질 간의 결합을 차단함으로써 중독을 예방하는 단백질 및 효소를 일컫는다. 특히 독성물질을 분해하는 과정에서 활성 상태를 유지함으로써 적은 양의 단백질로도 독성물질의 중독을 빠르게 치료하는 촉매성 바이오스캐빈저 개발에 많은 노력이 투여되어 왔다. 본 리뷰에서는 촉매성 바이오스캐빈저 개발을 위해 분자진화 및 단백질 공학 기술을 적용한 최신 연구들에 대해 소개하고, 끝으로 이러한 효소들을 임상적으로 승인된 약으로 개발하기 위해 남은 몇 가지 과제들을 간단히 제시할 것이다.

Abstract AI-Helper 아이콘AI-Helper

In recent years, toxic organophosphorus compounds (OPs) have been used for civilians, becoming a great threat to the world. Alternative to the current treatment policy unpredictable for any prevention, researches on bioscavenger as an improved treatment have been actively conducted. Bioscavengers re...

주제어

표/그림 (7)

AI 본문요약
AI-Helper 아이콘 AI-Helper

* AI 자동 식별 결과로 적합하지 않은 문장이 있을 수 있으니, 이용에 유의하시기 바랍니다.

문제 정의

  • 소만에 대해서는 Y337A/F338A-HuAChE 돌연변이가 가장 높은 가수분해 효율을 보였으며, 이를 폴리에틸렌글리콜(polyethylene glycol,PEG)로 전처리(PEGylated)하고 HI-6 옥심으로 후처리 한 경우, 쥐는 고용량의 소만에 의한 2번의 순차적 중독에서도 생존하는 것으로 나타났다[42-44]. Y337A/F338A-HuAChE 돌연변이를 다수의 독성물질에 대해서도 효율을 높이기 위해 타분을 활용하여 추가 연구하였고,이 연구에서는 재활성제로서 가장 적합한 옥심을 선별해내는 실험이 시도되었다. 시험된 새로운 옥심 중에는 2-PAM보다 긴 알킬 사슬(alkyl chain)을 갖고 있는 hexyl 2-PAM이 재활성제로 사용되었을 경우, Y337A/F338A-HuAChE가 타분에 대해 높은 효율을 가질 수 있었다[45].
  • 정제된 PTE는 소만 및 사이클로 사린 가수분해의 경우 중간 정도의 효율을 보였지만[84] VX에 대해서는 효율이 훨씬 낮았으며[85], 파라옥손에 대한 가수분해 효율과 비교했을 때에도 작용제에 대한 가수분해 효율은 현저히 낮은 수준이었다(Table 3)[83]. 따라서 PTE를 기반으로 한 효율적이고 넓은 스펙트럼을 갖는 촉매성 바이오스캐빈저를 개발하기 위해 유도 진화 및 전산 설계 방법의 적용이 필요했다.
  • DFPase는 비포유류 단백질이기 때문에 포유동물의 면역원성으로 인해 혈액 내에서 제거될 확률이 높다. 따라서 혈액 내 단백질 반감기 시간을 늘리기 위한 방법으로 비활성 폴리머로 단백질을 변형시켜 면역원성 감소, 단백질 분해와 세포독성으로부터의 보호, 분자량 증가로 신장에서의 제거율 감소 등의 효과를 얻고자 했다[102,103]. 이 모든 과정을 거쳐 만들어진 PEGylated DFPase 변이체를 3 × LD50 용량의 소만에 노출시키기 5 min 전 쥐에 투여한 결과, 무처리의 대조군은 중독 후 15 min 내에 사망한 반면 PEGylated DFPase 변이체를 주사한 쥐는 중독 후 24 h가 경과한 이후에도 생존하였다(Table 2)[103].
  • 따라서 독성물질에 대한 천연 효소의 활성 및 선택성을 모두 향상시킴으로써 효과적인 치료물질로 사용될 수 있는 촉매성 바이오스캐빈저를 개발하기 위해 분자 진화(molecular evolution)와 단백질 공학 기술(protein engineering techniques)이 사용되어 왔다[21]. 우리는 여기서 촉매성 바이오스캐빈저의 후보 단백질들과, 그 단백질들의 활동 및 독성물질 가수분해 기능을 높이기 위해 개발하고 있는 단백질 효소에 대해서 논의하고자 한다.
  • 사람, 박테리아, 해조류 등으로부터 발견된 천연 유기인산 가수분해효소들은 인공 제노바이오틱(xenobiotic) 독성물질에 대해서는 높은 촉매 효율을 나타내지 않을 것이라는 인식이 확대된 가운데, 유도 진화(directed evolution) 및 전산 설계 방법(computational design)의 진보는 이러한 천연 효소들을 재설계함으로써 그 촉매 효율을 상당히 향상시키는 데 많은 기여를 해오고 있다. 이번 장에서는 천연 효소들을 어떻게 재설계하였고, 또 그로 인해 어느 정도의 촉매 효율 향상이 이루어졌는지에 대하여 다뤄보겠다.
본문요약 정보가 도움이 되었나요?

질의응답

핵심어 질문 논문에서 추출한 답변
OPs란 무엇인가? 최근 영국의 노비촉(Novichok) 및 말레이시아 공항의 VX 테러 사건에서 보았듯이 독성물질 피독 시 초기 처치는 매우 중요한 과정이다. 두 사건에서 쓰인 독성물질은 유기인 화합물(organophosphorus coumpounds, OPs)이라는 큰 범주에 포함되는데, OPs는 인과 탄소가 결합되어 있는 화합물로 농약, 비료, 화학작용제 등 그 쓰임이 다양하다. 최초 살충제나 제초제로 널리 사용되던 OPs는 그 강한 독성으로 인해 화학작용제로 합성 및 무기화되었으며, 사린(sarin, GB), 소만(soman, GD), 사이클로 사린(cyclosarin, GF), 타분(tabun, GA), VX, 노비촉 등이 이에 포함된다(Figure 1).
기존에 사용되었던 유기인산 독성물질 중독 치료제의 한계는 무엇인가? 이를 초기에 또한 적시에 적용한다면, 치사율을 줄일 수 있고 중독 증상을 완화할 수 있다. 하지만 앞서 언급한 치료물질들은 사후 조치에 해당하는 치료제이기 때문에 회복 이후에도 의식손실이나 영구적인 뇌의 손상을 야기하는 콜린성 위기(cholinergic crisis)를 예방하는 데는 한계가 있다[2]. 또한 현재까지 개발된 치료법들은 유기인산 독성물질 중독 이전에 적용할 경우에 중추신경계 손상, 혈압 및 심박수 증가 등과 같은 심각한 부작용을 야기하기 때문에 중독되기 전 예방 대책으로는 적합하지 않다[3].
AChE의 특징은 무엇인가? 통상적으로 촉매성 세린으로부터 억제제가 자발적으로 분리되는 속도는 효소 및 유기인 화합물의 유형에 따라 다르지만, 유기인산 독성물질과 상호작용하는 B-에스테라제의 경우는 일반적으로 그 속도가 매우 느리기 때문에 외부 친핵체(옥심 재활성화제)가 필요한 것이다[10,11]. 유사촉매 바이오스캐빈저의 경우, 그 구성성분이 통상 혈액 내에 존재하며(인간혈청의 BChE 및 적혈구의 AChE), AChE는 모든 유형의 유기인산 독성물질을 신속하게 유리시킨다는 장점을 갖고 있다[12]. 뿐만 아니라, 옥심이 말초 신경 조직 및 일부의 경우 중추신경계에까지 도달하여 억제된 AChE을 다시 활성화 할 수 있다는 점 또한 큰 장점이다[13-15].
질의응답 정보가 도움이 되었나요?

참고문헌 (131)

  1. I. Koplovitz, S. M. Schulz, R. F. Railer, M. Sigler, and R. B. Lee, Effect of atropine and diazepam on the efficacy of oxime treatment of nerve agent intoxication, J. Med. CBR. Def., 5, 1-15 (2007). 

  2. J. Newmark, Seminars in Neurology, 24, 185-196, Thieme Medical Publishers, Inc., New York, USA (2004). 

  3. P. Taylor, Anticholinesterase agents, In: L. L. Brunton, B. A. Chabner, B. C. Knollmann (eds.), Goodman & Gilman's the Pharmacological Basis of Therapeutics, 239-254, McGraw-Hill, New York, USA (2011). 

  4. M. Trovaslet-Leroy, L. Musilova, F. Renault, X. Brazzolotto, J. Misik, L. Novotny, M. T. Froment, E. Gillon, M. Loiodice, L. Verdier, and P. Masson, Organophosphate hydrolases as catalytic bioscavengers of organophosphorus nerve agents, Toxicol. Lett., 206, 14-23 (2011). 

  5. F. Nachon, X.Brazzolotto, M. Trovaslet, and P. Masson, Progress in the development of enzyme-based nerve agent bioscavengers, Chem. Biol. Interact., 206, 536-544 (2013). 

  6. J. Descotes, Immunotoxicology of Drugs and Chemicals: An Experimental and Clinical Approach, 2-18, Elsevier, Amsterdam, Netherlands (2004). 

  7. N. Aurbek, H. Thiermann, F. Eyer, P. Eyer, and F. Worek, Suitability of human butyrylcholinesterase as therapeutic marker and pseudo catalytic scavenger in organophosphate poisoning: A kinetic analysis, Toxicology, 259, 133-139 (2009). 

  8. K. G. McGarry, R. F. Lalisse, R. A. Moyer, K. M. Johnson, A. M. Tallan, T. P. Winters, J. E. Taris, C. A. McElroy, E. E. Lemmon, H. S. Shafaat, Y. Fan, A. Deal, S. C. Marguet, J. A. Harvilchuck, C. M. Hadad, and D. W. Wood, A novel, modified human butyrylcholinesterase catalytically degrades the chemical warfare nerve agent, sarin, Toxicol. Sci., 174, 133-146 (2019). 

  9. J. Estevez and E. Vilanova, Model equations for the kinetics of covalent irreversible enzyme inhibition and spontaneous reactivation: Esterases and organophosphorus compounds, Crit. Rev. Toxicol., 39, 427-448 (2009). 

  10. W. N. Aldridge, Organophosphorus compounds: Molecular basis for their biological properties, Sci. Prog., 67, 138-139 (1981). 

  11. F. Worek, N. Aurbek, T. Wille, P. Eyer, and H. Thiermann, Kinetic prerequisites of oximes as effective reactivators of organophosphate-inhibited acetylcholinesterase: A theoretical approach, J. Enzyme Inhib., 26, 303-308 (2010). 

  12. R. L. Maynard and F. W. Beswick, Clinical and Experimental Toxicology of Organophosphates and Carbamates, 373-385, Elsevier, Oxford, UK (1992). 

  13. G. E. Garcia, A. J. Campbell, J. Olson, D. Moorad-Doctor, and V. I. Morthole, Novel oximes as blood-brain barrier penetrating cholinesterase reactivators, Chem. Biol. Interact., 187, 199-206 (2010). 

  14. G. Mercey, T. Verdelet, G. Saint-Andre, E. Gillon, A. Wagner, R. Baati, L. Jean, F. Nachon, and P. Y. Renard, First efficient uncharged reactivators for the dephosphylation of poisoned human acetylcholinesterase, Chem. Commun., 47, 5295-5297 (2011). 

  15. J. Kalisiak, E. C. Ralph, and J. R. Cashman, Nonquaternary reactivators for organophosphate-inhibited cholinesterases, J. Med. Chem., 55, 465-474 (2011). 

  16. M. Katalinic, N. Macek Hrvat, K. Baumann, S. Morasi Pipercic, S. Makaric, S. Tomic, O. Jovic, T. Hrenar, A. Milicevic, D. Jelic, S. Zunec, I. Primozic, and Z. Kovarik, A comprehensive evaluation of novel oximes in creation of butyrylcholinesterase-based nerve agent bioscavengers, Toxicol. Appl. Pharmacol., 310, 195-204 (2016). 

  17. J. G. Clement, D. G. Bailey, H. D. Madill, L. T. Tran, and J. D. Spence, The acetylcholinesterase oxime reactivator HI-6 in man: Pharmacokinetics and tolerability in combination with atropine, Biopharm. Drug Dispos., 16, 415-425 (1995). 

  18. D. Josse, O. Lockridge, W. Xie, C. F. Bartels, L. M. Schopfer, and P. Masson, The active site of human paraoxonase (PON1), J. Appl. Toxicol., 21, S7-S11 (2001). 

  19. B. Mackness, M. Mackness, M. Aviram, and G. Paragh (eds.). The Paraoxonases: Their Role In Disease Development And Xenobiotic Metabolism, 3-32, Springer Science & Business Media (2007). 

  20. L. Briseno-Roa, J. Hill, S. Notman, D. Sellers, A. P. Smith, C. M. Timperley, J. Wetherell, N. H. Williams, G. R. Williams, A. R. Fersht, and A. D. Griffiths, Analogues with fluorescent leaving groups for screening and selection of enzymes that efficiently hydrolyze organophosphorus nerve agents, J. Med. Chem., 49, 246-255 (2006). 

  21. M. Goldsmith, N. Aggarwal, Y. Ashani, H. Jubran, P. J. Greisen, S. Ovchinnikov, H. Leader, D. Baker, J. L. Sussman, A. Goldenzweig, S. J. Fleishman, and D. S. Tawfik, Overcoming an optimization plateau in the directed evolution of highly efficient nerve agent bioscavengers, Protein Eng., 30, 333-345 (2017). 

  22. T. Imai and K. Ohura, The role of intestinal carboxylesterase in the oral absorption of prodrugs, Curr. Drug Metab., 11, 793-805 (2010). 

  23. U. T. Bornscheuer, Microbial carboxylesterases: Classification, properties and application in biocatalysis, FEMS Microbiol. Rev., 26, 73-81 (2002). 

  24. T. Imai, Human carboxylesterase isozymes: Catalytic properties and rational drug design, Drug Metab. Pharm., 21, 173-185 (2006). 

  25. M. J. Hatfield, R. A. Umans, J. L. Hyatt, C. C. Edwards, M. Wierdl, L. Tsurkan, M. R. Taylor, and P. M. Potter, Carboxylesterases: General detoxifying enzymes, Chem. Biol. Interact., 259, 327-331 (2016). 

  26. T. Satoh and M. Hosokawa, Molecular aspects of carboxylesterase isoforms in comparison with other esterases, Toxicol. Lett., 82, 439-445 (1995). 

  27. A. C. Hemmert, T. C. Otto, M. Wierdl, C. C. Edwards, C. D. Fleming, M. MacDonald, J. R. Cashman, P. M. Potter, D. M. Cerasoli, and M. R. Redinbo, Human carboxylesterase 1 stereoselectively binds the nerve agent cyclosarin and spontaneously hydrolyzes the nerve agent sarin, Mol. Pharmacol., 77, 508-516 (2010). 

  28. P. Masson and S. V. Lushchekina, Emergence of catalytic bioscavengers against organophosphorus agents, Chem. Biol. Interact., 259, 319-326 (2016). 

  29. A. C. Hemmert, T. C. Otto, R. A. Chica, M. Wierdl, J. S. Edwards, S. L. Lewis, C. C. Edwards, L. Tsurkan, C. L. Cadieux, S. A. Kasten, J. R. Cashman, S. L. Mayo, P. M. Potter, D. M. Cerasoli, and M. R. Redinbo, Nerve agent hydrolysis activity designed into a human drug metabolism enzyme, PLoS One, 6, e17441 (2011). 

  30. P. Masson, P. L. Fortier, C. Albaret, M. T. Froment, C. F. Bartels, and O. Lockridge, Aging of di-isopropyl-phosphorylated human butyrylcholinesterase, Biochem. J., 327, 601-607 (1997). 

  31. P. M. Legler, S. M. Boisvert, J. R. Compton, and C. B. Millard, Development of organophosphate hydrolase activity in a bacterial homolog of human cholinesterase, Front Chem., 2, 46 (2014). 

  32. F. Soto-Mancera, J. M. Arellano, and M. G. Albendin, Carboxylesterase in Sparus aurata: characterisation and sensitivity to organophosphorus pesticides and pharmaceutical products, Ecol. Indic., 109, 105603 (2020). 

  33. V. V. Frolkis, V. V. Bezrukov, Y. K. Duplenko, I. V. Shchegoleva, V. G. Shevtchuk, and N. S. Verkhratsky, Acetyl- choline metabolism and cholinergic regulation of functions in aging, Gerontology, 19, 45-57 (1973). 

  34. F. Wore, H. Thiermann, L. Szinicz, and P. Eyer, Kinetic analysis of interactions between human acetylcholinesterase, structurally different organophosphorus compounds and oximes, Biochem. Pharmacol., 68, 2237-2248 (2004). 

  35. K. G. McGarry, K. E. Schill, T. P. Winters, E. E. Lemmon, C. L. Sabourin, J. A. Harvilchuck, and R. A. Moyer, Characterization of cholinesterases from multiple large animal species for medical countermeasure development against chemical warfare nerve agents, Toxicol. Sci., 174, 124-132 (2019). 

  36. Z. Chen, R. Newcomb, E. Forbes, J. McKenzie, and P. Batterham, The acetylcholinesterase gene and organophosphorus resistance in the Australian sheep blowfly, Lucilia cuprina, Insect Biochem. Mol. Biol., 31, 805-816 (2001). 

  37. P. Menozzi, M. Shi, A. Lougarre, Z. H. Tang, and D. Fournier, Mutations of acetylcholinesterase which confer insecticide resistance in Drosophila melanogaster populations, BMC Evol. Biol., 4, 4 (2004). 

  38. Y. Ashani, Z. Radic, I. Tsigelny, D. C. Vellom, N. A. Pickering, D. M. Quinn, B. P. Doctor, and P. Taylor, Amino acid residues controlling reactivation of organophosphonyl conjugates of acetylcholinesterase by mono-and bisquaternary oximes, J. Biol. Chem., 270, 6370-6380 (1995). 

  39. L. Wong, Z. Radic, R. J. Bruggemann, N. Hosea, H. A. Berman, and P. Taylor, Mechanism of oxime reactivation of acetylcholinesterase analyzed by chirality and mutagenesis, Biochemistry, 39, 5750-5757 (2000). 

  40. T. Kucukkilinc, R. Cochran, J. Kalisiak, E. Garcia, A. Valle, G. Amitai, Z. Radica, and P. Taylor, Investigating the structural influence of surface mutations on acetylcholinesterase inhibition by organophosphorus compounds and oxime reactivation, Chem. Biol. Interact., 187, 238-240 (2010). 

  41. M. Katalinic, G. Sinko, M. N. Hrvat, T. Zorbaz, A. Bosak, and Z. Kovarik, Oxime-assisted reactivation of tabun-inhibited acetylcholinesterase analysed by active site mutations, Toxicology, 406-407, 104-113 (2018). 

  42. O. Mazor, O. Cohen, C. Kronman, L. Raveh, D. Stein, A. Ordentlich, and A. Shafferman, Aging-resistant organophosphate bioscavenger based on polyethylene glycol-conjugated F338A human acetylcholinesterase, Mol. Pharmacol., 74, 755-763 (2008). 

  43. C. Kronman, O. Cohen, O. Mazor, A. Ordentlich, L. Raveh, B. Velan, and A. Shafferman, Next generation OP-bioscavengers: A circulatory long-lived 4-PEG hypolysine mutant of F338A-HuAChE with optimal pharmacokinetics and pseudo-catalytic characteristics, Chem. Biol. Interact., 187, 253-258 (2010). 

  44. N. M. Hrvat, S. Zunec, P. Taylor, Z. Radic, and Z. Kovarik, HI-6 assisted catalytic scavenging of VX by acetylcholinesterase choline binding site mutants, Chem. Biol. Interact., 259, 148-153 (2016). 

  45. Z. Kovarik, N. M. Hrvat, J. Kalisiak, M. Katalinic, R. K. Sit, T. Zorbaz, Z. Radic, V. V. Fokin, K. B. Sharpless, and P. Taylor, Counteracting tabun inhibition by reactivation by pyridinium aldoximes that interact with active center gorge mutants of acetylcholinesterase, Toxicol. Appl. Pharmacol., 372, 40-46 (2019). 

  46. J. Massoulie, J. Sussman, S. Bon, and I. Silman, Structure and function of acetylcholinesterase and butyrylcholinesterase, Brain Res., 98, 139-146 (1993). 

  47. L. Raveh, J. Grunwald, D. Marcus, Y. Papier, E. Cohen, and Y. Ashani, Human butyrylcholinesterase as a general prophylactic antidote for nerve agent toxicity, Biochem. Pharmacol., 45, 2465-2474 (1993). 

  48. V. P. Chen, Y. Gao, L. Geng, and S. Brimijoin, Butyrylcholinesterase gene transfer in obese mice prevents postdieting body weight rebound by suppressing ghrelin signaling, Proc. Natl. Acad. Sci., 114, 10960-10965 (2017). 

  49. S. V. Lushchekina, B. L. Grigorenko, D. I. Morozov, I. V. Polyakov, A. V. Nemukhin, and S. D. Varfolomeev, Modeling of the mechanism of hydrolysis of succinylcholine in the active site of native and modified (Asp70Gly) human butyrylcholinesterase, Russ. Chem. Bull., 59, 55-60 (2010). 

  50. K. A. Gonzalez, E. H. Viana, and R. V. Duhalt, Enzymatic detoxification of organophosphorus pesticides and related toxicants, J. Pestic. Sci., 43, 1-9 (2018). 

  51. Y. Cai, S. Zhou, M. J. Stewart, F. Zheng, and C. G. Zhan, Dimerization of human butyrylcholinesterase expressed in bacterium for development of a thermally stable bioscavenger of organophosphorus compounds, Chem. Biol. Interact., 310, 108756 (2019). 

  52. B. Mackness, P. N. Durrington, and M. I. Mackness, Human serum paraoxonase, Gen. Pharmacol., 31, 329-336 (1998). 

  53. M. Aviram, M. Rosenblat, C. L. Bisgaier, R. S. Newton, S. L. Primo-Parmo, and B. N. La Du, Paraoxonase inhibits high-density lipoprotein oxidation and preserves its functions. A possible peroxidative role for paraoxonase, J. Clin. Invest., 101, 1581-1590 (1998). 

  54. P. N. Durrington, B. Mackness, and M. I. Mackness, Paraoxonase and atherosclerosis, Arter. Thromb. Vasc. Biol., 21, 473-480 (2001). 

  55. R. C. Stevens, S. M. Suzuki, T. B. Cole, S. S. Park, R. J. Richter, and C. E. Furlong, Engineered recombinant human paraoxonase 1 (rHuPON1) purified from Escherichia coli protects against organophosphate poisoning, Proc. Natl. Acad. Sci., 105, 12780-12784 (2008). 

  56. L. G. Costa, G. Giordano, T. B. Cole, J. Marsillach, and C. E. Furlong, Paraoxonase 1 (PON1) as a genetic determinant of susceptibility to organophosphate toxicity, Toxicology, 307, 115-122 (2013). 

  57. G. Kaur, A. K. Jain, and S. Singh, CYP/PON genetic variations as determinant of organophosphate pesticides toxicity, J. Genet., 96, 187-201 (2017). 

  58. H. G. Davies, R. J. Richter, M. Keifer, C. A. Broomfield, J. Sowalla, and C. E. Furlong, The effect of the human serum paraoxonase polymorphism is reversed with diazoxon, soman and sarin, Nat. Genet., 14, 334-336 (1996). 

  59. M. W. Peterson, S. Z. Fairchild, T. C. Otto, M. Mohtashemi, D. M. Cerasoli, and W. E. Chang, VX hydrolysis by human serum paraoxonase 1: A comparison of experimental and computational results, PLoS One, 6, e20335 (2011). 

  60. D. G. Mata, P. Sabnekar, C. A. Watson, P. E. Rezk, and N. Chilukuri, Assessing the stoichiometric efficacy of mammalian expressed paraoxonase-1 variant I-F11 to afford protection against G-type nerve agents, Chem. Biol. Interact., 259, 233-241 (2016). 

  61. M. Valiyaveettil, Y. Alamneh, P. Rezk, L. Biggemann, M. W. Perkins, A. M. Sciuto, B. P. Doctor, and M. P. Nambiar, Protective efficacy of catalytic bioscavenger, paraoxonase 1 against sarin and soman exposure in guinea pigs, Biochem. Pharmacol., 81, 800-809 (2011). 

  62. M. Valiyaveettil, Y. Alamneh, P. Rezk, M. W. Perkins, A. M. Sciuto, B. P. Doctor, and M. P. Nambiar, Recombinant paraoxonase 1 protects against sarin and soman toxicity following microinstillation inhalation exposure in guinea pigs, Toxicol. Lett., 202, 203-208 (2011). 

  63. S. M. Hodgins, S. A. Kasten, J. Harrison, T. C. Otto, Z. P. Oliver, P. Rezk, T. E. Reeves, N. Chilukuri, and D. M. Cerasoli, Assessing protection against OP pesticides and nerve agents provided by wild-type HuPON1 purified from Trichoplusia ni larvae or induced via adenoviral infection, Chem. Biol. Interact., 203, 177-180 (2013). 

  64. A. Aharoni, L. Gaidukov, S. Yagur, L. Toker, I. Silman, and D. S. Tawfik, Directed evolution of mammalian paraoxonases PON1 and PON3 for bacterial expression and catalytic specialization, Proc. Natl. Acad. Sci., 101, 482-487 (2003). 

  65. M. Harel, A. Aharoni, L. Gaidukov, B. Brumshtein, O. Khersonsky, R. Meged, H. Dvir, R. B. Ravelli, A. McCarthy, L. Toker, I. Silman, J. Sussman, and D. S. Tawfik, Structure and evolution of the serum paraoxonase family of detoxifying and anti-atherosclerotic enzymes, Nat. Struct. Biol., 11, 412-419 (2004). 

  66. R. D. Gupta, M. Goldsmith, Y. Ashani, Y. Simo, G. Mullokandov, H. Bar, M. B. David, H. Leader, R. Margalit, I. Silman, J. L. Sussman, and D. S. Tawfik, Directed evolution of hydrolases for prevention of G-type nerve agent intoxication, Nat. Chem. Biol., 7, 120-125 (2011). 

  67. M. Goldsmith, Y. Ashani, Y. Simo, M. B. David, H. Leader, I. Silman, J. L. Sussman, and D. S. Tawfik, Evolved stereoselective hydrolases for broad-spectrum G-type nerve agent detoxification, Chem. Biol., 19, 456-466 (2012). 

  68. F. Worek, T. Seeger, M. Goldsmith, Y. Ashani, H. Leader, J. S. Sussman, D. S. Tawfik, H. Thiermann, and T. Wille, Efficacy of the rePON1 mutant IIG1 to prevent cyclosarin toxicity in vivo and to detoxify structurally different nerve agents in vitro, Arch. Toxicol., 88, 1257-1266 (2014). 

  69. M. Goldsmith, Y. Ashani, R. Margalit, A. Nyska, D. Mirelman, and D. S. Tawfik, A new post-intoxication treatment of paraoxon and parathion poisonings using an evolved PON1 variant and recombinant GOT1, Chem. Biol. Interact., 259, 242-251 (2016). 

  70. A. Zlotnik, S. E. Gruenbaum, A. A. Artru, I. Rozet, M. Dubilet, S. Tkachov, E. Brotfain, Y. Klin, Y. Shapira, and V. I. Teichberg, The neuroprotective effects of oxaloacetate in closed head injury in rats is mediated by its blood glutamate scavenging activity, J. Neurosurg. Anesthesiol., 21, 235-241 (2009). 

  71. A. Ruban, B. Mohar, G. Jona, and V. I. Teichberg, Blood glutamate scavenging as a novel neuroprotective treatment for paraoxon intoxication, J. Cerebr. Blood. F. Met., 34, 221-227 (2013). 

  72. A. Ruban, I. Biton, A. Markovich, and D. Mirelman, MRS of brain metabolite levels demonstrates the ability of scavenging of excess brain glutamate to protect against nerve agent induced seizures, Int. J. Mol. Sci., 16, 3226-3236 (2015). 

  73. J. Cowan, C. M. Sinton, A. W. Varley, F. H. Wians, R. W. Haley, and R. S. Munford, Gene therapy to prevent organophosphate intoxication, Toxicol. Appl. Pharmacol., 173, 1-6 (2001). 

  74. L. G. Costa, R. J. Richter, W. F. Li, T. Cole, M. Guizzetti, and C. E. Furlong, Paraoxonase (PON1) as a biomarker of susceptibility for organophosphate toxicity, Biomarkers, 8, 1-12 (2003). 

  75. A. L. Fu, Y. X. Wang, and M. J. Sun, Naked DNA prevents soman intoxication, Biochem. Biophys. Res. Commun., 328, 901-905 (2005). 

  76. D. G. Mata, P. E. Rezk, P. Sabnekar, D. M. Cerasoli, and N. Chilukuri, Investigation of evolved paraoxonase-1 variants for prevention of organophosphorous pesticide compound intoxication, J. Pharmacol. Exp. Ther., 349, 549-558 (2014). 

  77. G. Amitai, L. Gaidukov, R. Adani, S. Yishay, G. Yacov, M. Kushnir, S. Teitlboim, M. Lindenbaum, P. Bel, O. Khersonsky, and D. S. Tawfik, Enhanced stereoselective hydrolysis of toxic organophosphates by directly evolved variants of mammalian serum paraoxonase, FEBS J., 273, 1906-1919 (2006). 

  78. S. D. Kirby, J. R. Norris, J. R. Smith, B. J. Bahnson, and D. M. Cerasoli, Human paraoxonase double mutants hydrolyze V and G class organophosphorus nerve agents, Chem. Biol. Interact., 203, 181-185 (2013). 

  79. W. W. Mulbry, J. S. Karns, P. C. Kearney, J. O. Nelson, C. S. McDaniel, and J. R. Wild, Identification of a plasmid-borne parathion hydrolase gene from Flavobacterium sp. by southern hybridization with opd from Pseudomonas diminuta, Appl. Environ. Microbiol., 51, 926-930 (1986). 

  80. D. P. Dumas, S. R. Caldwell, J. R. Wild, and F. M. Raushel, Purification and properties of the phosphotriesterase from Pseudomonas diminuta, J. Biol. Chem., 264, 19659-19665 (1989). 

  81. E. Ghanem and F. M. Raushel, Detoxification of organophosphate nerve agents by bacterial phosphotriesterase, Toxicol. Appl. Pharmacol., 207, 459-470 (2005). 

  82. C. M. Theriot and A. M. Grunden, Hydrolysis of organophosphorus compounds by microbial enzymes, Appl. Microbiol. Biotechnol., 89, 35-43 (2011). 

  83. P. Masson, Handbook of Toxicology of Chemical Warfare Agents, 2nd ed., 1107-1123, Elsevier, Kentucky, USA (2015). 

  84. D. P. Dumas, H. D. Durst, and W. G. Landis, Inactivation of organophosphorus nerve agents by the phosphotriesterase from Pseudomonas diminuta, Arch. Biochem. Biophys., 277, 155-159 (1990). 

  85. V. K. Rastogi, J. J. Defranck, and T. C. Cheng, Enzymatic hydrolysis of Russian-VX by organophosphorus hydrolase, Biochem. Biophys. Res. Commun., 241, 294-296 (1997). 

  86. A. D. Griffiths and D. S. Tawfik, Directed evolution of an extremely fast phosphotriesterase by in vitro compartmentalization, EMBO J., 22, 24-35 (2003). 

  87. C. Roodveldt and D. S. Tawfik, Directed evolution of phosphotriesterase from Pseudomonas diminuta for heterologous expression in Escherichia coli results in stabilization of the metal-free state, Protein Eng. Des. Select., 18, 51-58 (2005). 

  88. C. M. H. Cho, A. Mulchandani, and W. Chen, Bacterial cell surface display of organophosphorus hydrolase for selective screening of improved hydrolysis of organophosphate nerve agents, Appl. Environ. Microbiol., 68, 2026-2030 (2002). 

  89. C. M. Hill, W. S. Li, J. B. Thoden, H. M. Holden, and F. M. Raushel, Enhanced degradation of chemical warfare agents through molecular engineering of the phosphotriesterase active site, J. Am. Chem. Soc., 125, 8990-8991 (2003). 

  90. I. Cherny, P. Greisen, Y. Ashani, S. D. Khare, G. Oberdorfer, H. Leader, D. Baker, and D. S. Tawfik, Engineering V-type nerve agents detoxifying enzymes using computationally focused libraries, ACS Chem. Biol., 8, 2394-2403 (2013). 

  91. P. Jacquet, J. Hiblot, D. Daude, C. Bergonzi, G. Gotthard, N. Armstrong, E. Chabriere, and M. Elias, Rational engineering of a native hyperthermostable lactonase into a broad spectrum phosphotriesterase, Sci. Rep., 7, 1-15 (2017). 

  92. T. Wille, K. Neumaier, M. Koller, C. Ehinger, N. Aggarwal, Y. Ashani, M. Goldsmith, J. L. Sussman, D. S. Tawfik, H. Thiermann, and F. Worek, Single treatment of VX poisoned guinea pigs with the phosphotriesterase mutant C23AL: Intraosseous versus intravenous injection, Toxicol. Lett., 258, 198-206 (2016). 

  93. F. Ely, K. S. Hadler, N. Mitic, L. R. Gahan, D. L. Ollis, N. M. Plugis, M. T. Russo, J. A. Larrabee, and G. Schenk, Electronic and geometric structures of the organophosphate-degrading enzyme from Agrobacterium radiobacter (OpdA), J. Biol. Inorg. Chem., 16, 777-787 (2011). 

  94. S. B. Bird, T. D. Sutherland, C. Gresham, J. Oakeshott, C. Scott, and M. Eddleston, OpdA, a bacterial organophosphorus hydrolase, prevents lethality in rats after poisoning with highly toxic organophosphorus pesticides, Toxicology, 247, 88-92 (2008). 

  95. J. Hiblot, G. Gotthard, E. Chabriere, and M. Elias, Structural and enzymatic characterization of the lactonase SisLac from Sulfolobus islandicus, PLoS One, 7, e47028 (2012). 

  96. J. Hiblot, G. Gotthard, E. Chabriere, and M. Elias, Characterisation of the organophosphate hydrolase catalytic activity of SsoPox, Sci. Rep., 2, 779 (2012). 

  97. M. M. Meier, C. Rajendran, C. Malisi, N. G. Fox, C. Xu, S. Schlee, D. P. Barondeau, B. Hocker, R. Sterner, and F. M. Raushel, Molecular engineering of organophosphate hydrolysis activity from a weak promiscuous lactonase template, J. Am. Chem. Soc., 135, 11670-11677 (2013). 

  98. J. Bzdrenga, J. Hiblot, G. Gotthard, C. Champion, M. Elias, and E. Chabriere, SacPox from the thermoacidophilic crenarchaeon Sulfolobus acidocaldarius is a proficient lactonase, BMC Research Notes, 7, 333 (2014). 

  99. L. Merone, L. Mandrich, E. Porzio, M. Rossi, S. Muller, G. Reiter, F. Worek, and G. Manco, Improving the promiscuous nerve agent hydrolase activity of a thermostable archaeal lactonase, Bioresour. Technol., 101, 9204-9212 (2010). 

  100. E. I. Scharff, J. Koepke, G. Fritzsch, C. Lucke, and H. Ruterjans, Crystal structure of diisopropylfluorophosphatase from Loligo vulgaris, Structure, 9, 493-502 (2001). 

  101. H. Allahyari and A. M. Latifi, Diisopropyl-fluorophosphatase as a catalytic bioscavenger, J. Appl. Biotechnol. Rep., 3, 477-482 (2016). 

  102. R. Webster, E. Didier, P. Harris, N. Siegel, J. Stadler, L. Tilbury, and D. Smith, PEGylated Proteins: Evaluation of their safety in the absence of definitive metabolism studies, Drug Metab. Dispos., 35, 9-16 (2006). 

  103. M. Melzer, A. Heidenreich, F. Dorandeu, J. Gab, K. Kehe, H. Thiermann, T. Letzel, and M. M. Blum, In vitro and in vivo efficacy of PEGylated diisopropyl fluorophosphatase (DFPase), Drug Test. Anal., 4, 262-270 (2011). 

  104. D. Zhou, D. Yin, F. Xiao, and J. Hao, Expressions of senescence-associated ${\beta}$ -galactosidase and senescence marker protein-30 are associated with lens epithelial cell apoptosis, Med. Sci. Monit., 21, 3728-3735 (2015). 

  105. J. S. Little, C. A. Broomfield, M. K. Fox-Talbot, L. J. Boucher, B. MacIver, and D. E. Lenz, Partial characterization of an enzyme that hydrolyzes sarin, soman, tabun, and diisopropyl phosphorofluoridate (DFP), Biochem. Pharmacol., 38, 23-29 (1989). 

  106. R. C. diTargiani, L. Chandrasekaran, T. Belinskaya, and A. Saxena, In search of a catalytic bioscavenger for the prophylaxis of nerve agent toxicity, Chem. Biol. Interact., 187, 349-354 (2010). 

  107. M. S. Choi, A. Saxena, and N. Chilukuri, A strategy for the production of soluble human senescence marker protein-30 in Escherichia coli, Biochem. Biophys. Res. Commun., 393, 509-513 (2010). 

  108. T. C. Cheng, S. Harvey, and A. N. Stroup, Purification and properties of a highly active organophosphorus acid anhydrolase from Alteromonas undina, Appl. Environ. Microbiol., 59, 3138-3140 (1993). 

  109. T. C. Cheng, S. P. Harvey, and G. L. Chen, Cloning and expression of a gene encoding a bacterial enzyme for decontamination of organophosphorus neve agents and nucleotide sequence of the enzyme, Appl. Environ. Microbiol., 62, 1636-1641 (1996). 

  110. S. P. Harvey, R. M. Leslie, and J. B. Frederic, Hydrolysis and enzymatic degradation of Novichok nerve agents, Heliyon, 6, e03153 (2020). 

  111. C. M. Daczkowski, S. D. Pegan, and S. P. Harvey, Engineering the organophosphorus acid anhydrolase enzyme for increased catalytic efficiency and broadened stereospecificity on russian VX, Biochemistry, 54, 6423-6433 (2015). 

  112. I. Petrikovics, Long circulating liposomes encapsulating organophosphorus acid Anhydrolase in diisopropylfluorophosphate antagonism, Toxicol. Sci., 57, 16-21 (2000). 

  113. P. Li, S. Y. Moon, M. A. Guelta, L. Lin, D. A. Gomez-Gualdron, R. Q. Snurr, S. P. Harvey, J. T. Hupp, and O. K. Farha, Nanosizing a metal-organic framework enzyme carrier for accelerating nerve agent hydrolysis, ACS Nano, 10, 9174-9182 (2016). 

  114. G. Zanaboni, K. M. Dyne, A. Rossi, V. Monafo, and G. Cetta, Prolidase deficiency: Biochemical study of erythrocyte and skin fibroblast prolidase activity in italian patients, Haematologica, 79, 13-18 (1994). 

  115. L. Chandrasekaran, T. Belinskaya, and A. Saxena, In vitro characterization of organophosphorus compound hydrolysis by native and recombinant human prolidase, Toxicology in vitro, 27, 499-506 (2013). 

  116. V. Aleti, G. B. Reddy, K. Parikh, P. Arun, and N. Chilukuri, Persistent and high-level expression of human liver prolidase in vivo in mice using adenovirus, Chem. Biol. Interact., 203, 191-195 (2013). 

  117. P. E. Rezk, P. Zdenka, P. Sabnekar, T. Kajih, D. G. Mata, C. Wrobel, D. M. Cerasoli, and N. Chilukuri, An in vitro and in vivo evaluation of the efficacy of recombinant human liver prolidase as a catalytic bioscavenger of chemical warfare nerve agents, Drug Chem. Toxicol., 38, 37-43 (2014). 

  118. H. Liu, J. J. Zhang, S. J. Wang, X. E. Zhang, and N. Y. Zhou, Plasmid-borne catabolism of methyl parathion and p-nitrophenol in Pseudomonas sp. strain WBC-3, Biochem. Biophys. Res. Commun., 334, 1107-1114 (2005). 

  119. Y. J. Dong, M. Bartlam, L. Sun, Y. F. Zhou, Z. P. Zhang, C. G. Zhang, Z. Rao, and X. E. Zhang, Crystal structure of methyl parathion hydrolase from Pseudomonas sp. WBC-3, J. Mol. Biol., 353, 655-663 (2005). 

  120. T. K. Ng, L. R. Gahan, G. Schenk, and D. L. Ollis, Altering the substrate specificity of methyl parathion hydrolase with directed evolution, Arch. Biochem. Biophys., 573, 59-68 (2015). 

  121. A. Ozgur and Y. Tutar, Therapeutic proteins: A to Z, Protein Pept. Lett., 20, 1365-1372 (2013). 

  122. H. D. Lagasse, A. Alexaki, V. L. Simhadri, N. H. Katagiri, W. Jankowski, Z. E. Sauna, and C. Kimchi-Sarfaty, Recent advances in (therapeutic protein) drug development, F1000Res., 6, 113 (2017). 

  123. J. L. Sussman and I. Silman, Acetylcholinesterase: Structure and use as a model for specific cation-protein interactions, Curr. Opin. Struct. Biol., 2, 721-729 (1992). 

  124. O. Lockridge, C. F. Bartels, T. A. Vaughan, C. K. Wong, S. E. Norton, and L. L. Johnson, Complete amino acid sequence of human serum cholinesterase, J. Biol. Chem., 262, 549-557 (1987). 

  125. M. M. Benning, J. M. Kuo, F. M. Raushel, and H. M. Holden, Three-dimensional structure of phosphotriesterase: An enzyme capable of detoxifying organophosphate nerve agents, Biochemistry, 33, 15001-15007 (1994). 

  126. I. Horne, T. D. Sutherland, R. L. Harcourt, R. J. Russell, and J. G. Oakeshott, Identification of an opd (Organophosphate Degradation) gene in an agrobacterium isolate, Appl. Environ. Microbiol., 68, 3371-3376 (2002). 

  127. S. Chakraborti and B. J. Bahnson, Crystal structure of human senescence marker protein 30: Insights linking structural, enzymatic, and physiological functions, Biochemistry, 49, 3436-3444 (2010). 

  128. N. K. Vyas, A. Nickitenko, V. K. Rastogi, S. S. Shah, and F. A. Quiocho, Structural insights into the dual activities of the nerve agent degrading organophosphate anhydrolase/prolidase, Biochemistry, 49, 547-559 (2010). 

  129. A. Lupi, R. Tenni, A. Rossi, G. Cetta, and A. Forlino, Human prolidase and prolidase deficiency: An overview on the characterization of the enzyme involved in proline recycling and on the effects of its mutations, Amino Acids, 35, 739-752 (2008). 

  130. C. Zhongli, L. Shunpeng, and F. Guoping, Isolation of methyl parathion-degrading strain M6 and cloning of the methyl parathion hydrolase gene, Appl. Environ. Microbiol., 67, 4922-4925 (2001). 

  131. M. Goldsmith and Y. Ashani, Catalytic bioscavengers as countermeasures against organophosphate nerve agents, Chem. Biol. Interact., 292, 50-64 (2018). 

저자의 다른 논문 :

관련 콘텐츠

오픈액세스(OA) 유형

BRONZE

출판사/학술단체 등이 한시적으로 특별한 프로모션 또는 일정기간 경과 후 접근을 허용하여, 출판사/학술단체 등의 사이트에서 이용 가능한 논문

저작권 관리 안내
섹션별 컨텐츠 바로가기

AI-Helper ※ AI-Helper는 오픈소스 모델을 사용합니다.

AI-Helper 아이콘
AI-Helper
안녕하세요, AI-Helper입니다. 좌측 "선택된 텍스트"에서 텍스트를 선택하여 요약, 번역, 용어설명을 실행하세요.
※ AI-Helper는 부적절한 답변을 할 수 있습니다.

선택된 텍스트

맨위로