In Vitro에서 개 말초혈액 백혈구의 순간산소과소비현상에 대한 케타민의 효과

Effect of Ketamine on the Oxidative Burst Activity of Canine Peripheral Blood Leukocytes In Vitro

  • Kim, Min-Jun (Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University) ;
  • Kang, Ji-Houn (Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University) ;
  • Yang, Mhan-Pyo (Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University)
  • 발행 : 2006.12.30

초록

전신마취제인 케타민은 흥분성 아미노산의 활성을 방해하는 N-methyl-D-aspartate (NMDA) 수용체의 비경쟁적인 길항제이다. 본 연구는 개 말초혈액 백혈구의 순간산소과소비현상(Oxidative burst activity; OBA)에 있어서 케타민의 효과를 검토하였다. 탐식세포의 OBA는 유세포 분석기로 분석하였다. 케타민을 말초혈액 다형핵백혈구(peripheral blood polymorphonuclear cells; PMN)와 monocyte-rich cells에 직접처리 하였을 때는 OBA가 감소하였으며, 또한 케타민을 처리한 말초혈액 단핵구세포(peripheral blood mononuclear cells; PBMC) 배양상층액에 의해서도 PMN과 monocyte-rich cells의 OBA가 감소하였다. 그러나 케타민을 처리한 PMN 배양상층액에 의해서는 탐식세포의 OBA에 있어서 아무런 변화가 없었다. 하지만 이러한 OBA의 감소는 latex beads를 넣어 탐식반응이 일어날 때만 측정되었다. 이상의 결과로부터 탐식반응이 일어나는 동안 케타민은 호중구와 단핵구와 같은 개 말초혈액 탐식구의 OBA에 있어 억제효과를 나타내었다.

Ketamine, one of general anesthetics for human and veterinary use, is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist which interferes with the action of excitatory amino acids. It has been reported to impair various leukocyte functions. In this study, the effect of ketamine on the oxidative burst activity (OBA) of canine peripheral blood leukocytes was examined. The OBA of canine peripheral blood phagocytes was analyzed by flow cytometry system. Ketamine at higher concentration such as $1,000{\mu}M$ exhibited a low viability of leukocytes. Thus, ketamine was used at concentration of 10 to $500{\mu}M$ showing no cytotoxic effect and high cell viability. The OBA of leukocytes in the presence or absence of latex beads was analyzed by addition of dihydrorhodamine 123. The direct treatment of ketamine revealed the inhibitory effect on the OBA of peripheral blood polymorphonuclear cells (PMN) and monocyte-rich cells but not peripheral blood mononuclear cells (PBMC) in the presence of latex beads. However, when latex beads were not added to PMN, its OBA was not inhibited by ketamine. The OBA of PMN and monocyte-rich cells but not PBMC in the presence of latex beads was also inhibited by culture supernatant from ketamine-treated- PBMC but not -PMN. But the OBA of PMN in the absence of latex beads was not inhibited by culture supernatant from PBMC treated with ketamine. Therefore, these results suggested that ketamine has the inhibitory effect on the OBA of canine peripheral blood phagocytes such as neutrophils and monocytes during phagocytic response.

키워드

참고문헌

  1. Anis NA, Berry SC, Burton NR, Lodge D. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol 1983; 79: 565-575 https://doi.org/10.1111/j.1476-5381.1983.tb11031.x
  2. Ayala A, Chaudry IH. Immune dysfunction in murine polymicrobial sepsis: mediators, macrophages, lymphocytes and apoptosis. Shock 1996; 6: S27-S38 https://doi.org/10.1097/00024382-199610001-00007
  3. Babior BM, Lambeth JD, Nauseef W. The neutrophil NADPH oxidase. Arch Biochem Biophys 2002; 397: 342-344 https://doi.org/10.1006/abbi.2001.2642
  4. Boyaka PN, McGhee JR. Cytokines as adjuvants for the induction of mucosal immunity. Adv Drug Deliv Rev 2001; 51: 71-79 https://doi.org/10.1016/S0169-409X(01)00170-3
  5. Chang Y, Chen TL, Sheu JR, Chen RM. Suppressive effects of ketamine on macrophage functions. Toxicol Appl Pharmacol 2005; 204: 27-35 https://doi.org/10.1016/j.taap.2004.08.011
  6. DeLeo FR, Quinn MT. Assembly of the phagocyte NADPH oxidase: molecular interaction of oxidase proteins. J Leukoc Biol 1996; 60: 677-691 https://doi.org/10.1002/jlb.60.6.677
  7. Di Virgilio F, Chiozzi P, Ferrari D, Falzoni S, Sanz JM, Morelli A, Torboli M, Bolognesi G, Baricordi OR. Nucleotide receptors: an emerging family of regulatory molecules in blood cells. Blood 2001; 97: 587-600 https://doi.org/10.1182/blood.V97.3.587
  8. Domino EF, Zsigmond EK, Domino LE, Domino KE, Kothary SP, Domino SE. Plasma levels of ketamine and two of its metabolites in surgical patients using a gas chromatographic mass fragmentographic assay. Anesth Analg 1982; 61: 87-92
  9. Emmendorffer A, Hecht M, Lohmann-Matthes ML, Roesler J. A fast and easy method to determine the production of reactive oxygen intermediates by human and murine phagocytes using dihydrorhodamine 123. J Immunol Methods 1990; 131: 269-275 https://doi.org/10.1016/0022-1759(90)90198-5
  10. Faurschou M, Borregaard N. Neutrophil granules and secretory vesicles in inflammation. Microbes Infect 2003; 5: 1317-1327 https://doi.org/10.1016/j.micinf.2003.09.008
  11. Flaminio MJ, Rush BR, Davis EG, Hennessy K, Shuman W, Wilkerson MJ. Simultaneous flow cytometric analysis of phagocytosis and oxidative burst activity in equine leukocytes. Vet Res Commun 2002; 26: 85-92 https://doi.org/10.1023/A:1014033016308
  12. Fredholm BB. Purines and neutrophil leukocytes. Gen Pharmacol 1997; 28: 345-350 https://doi.org/10.1016/S0306-3623(96)00169-3
  13. Grant IS, Nimmo WS, McNicol LR, Clements JA. Ketarnine disposition in children and adults. Br J Anaesth 1983; 55: 1107-1111 https://doi.org/10.1093/bja/55.11.1107
  14. Henson PM, Johnston RB Jr. Tissue injury in inflammation. Oxidants, proteinases, and cationic proteins. J Clin Invest 1987; 79: 669-674 https://doi.org/10.1172/JCI112869
  15. Hill GE, Anderson JL, Lyden ER. Ketamine inhibits the proinflammatory cytokine-induced reduction of cardiac intracellular cAMP accumulation. Anesth Analg 1998; 87: 1015-1019 https://doi.org/10.1097/00000539-199811000-00006
  16. Hofbauer R, Moser D, Hammerschmidt V, Kapiotis S, Frass M. Ketamine significantly reduces the migration of leukocytes through endothelial cell monolayers. Crit Care Med 1998; 26: 1545-1549 https://doi.org/10.1097/00003246-199809000-00022
  17. Kanai AJ, Pearce LL, Clemens PR, Birder LA, VanBibber MM, Choi SY, de Groat WC, Peterson J. Identification of a neuronal nitric oxide synthase in isolated cardiac mitochondria using electrochemical detection. Proc Natl Acad Sci USA 2001; 98: 14126-14131 https://doi.org/10.1073/pnas.241380298
  18. Kang JH, Kim JH, Chung CS, Lee CY, Yang MP. Immuno-enhancing effect of conjugated linoleic acids on phagocytic activity of porcine peripheral blood phagocytes. J Vet Clin 2004; 21: 336-342
  19. Krumholz W, Endrass J, Hempelmann G. Inhibition of phagocytosis and killing of bacteria by anaesthetic agents in vitro. Br J Anaesth 1995; 75: 66-70 https://doi.org/10.1093/bja/75.1.66
  20. Lammas DA, Stober C, Harvey CJ, Kendrick N, Panchalingam S, Kumararatne DS. ATP-induced killing of mycobacteria by human macrophages is mediated by purinergic P2Z(P2X7) receptors. Immunity 1997; 7: 433-444 https://doi.org/10.1016/S1074-7613(00)80364-7
  21. Mullen PG, Windsor AC, Walsh CJ, Fowler AA 3rd, Sugerman HJ. Tumor necrosis factor-alpha and interleukin-6 selectively regulate neutrophil function in vitro. J Surg Res 1995; 58: 124-130 https://doi.org/10.1006/jsre.1995.1020
  22. Oshimi Y, Miyazaki S, Oda S. ATP-induced Ca2+ response mediated by P2U and P2Y purinoceptors in human macrophages: signalling from dying cells to macrophages. Immunology 1999; 98: 220-227 https://doi.org/10.1046/j.1365-2567.1999.00858.x
  23. Peterson PK, Sharp B, Gekker G, Brummit C, Keane WF. Opioid-mediated suppression of interferon-[small gamma, Greek] production by cultured peripheral blood mononuclear cells. J Clin Invest 1987; 80: 824-831 https://doi.org/10.1172/JCI113140
  24. Roos D, van Bruggen R, Meischl C. Oxidative killing of microbes by neutrophils. Microbes Infect 2003; 5: 1307-1315 https://doi.org/10.1016/j.micinf.2003.09.009
  25. Rossano F, Tufano R, Cipollaro De L'Ero G, Servillo G, Baroni A, Tufano MA. Anaesthetic agents induce human mononuclear leukocytes to release cytokines. Immunopharmacol Immunotoxicol 1992; 14: 439-450 https://doi.org/10.3109/08923979209005403
  26. Rothe G, Emmendorffer A, Oser A, Roesler J, Valet G. Flow cytometric measurement of the respiratory burst activity of phagocytes using dihydrorhodamine 123. J Immunol Methods 1991; 138: 133-135 https://doi.org/10.1016/0022-1759(91)90074-P
  27. Royall JA, lschiropoulos H. Evaluation of 2',7'-dichlorofluorescin and dihydrorhodamine 123 as fluorescent probes for intracellular $H_2O_2$ in cultured endothelial cells. Arch Biochem Biophys 1993; 302: 348-355 https://doi.org/10.1006/abbi.1993.1222
  28. Sakai T, Ichiyama T, Whitten CW, Giesecke AH, Lipton JM. Ketamine suppresses endotoxin-induced NF-$\kappa$B expression. Can J Anaesth 2000; 47: 1019-1024 https://doi.org/10.1007/BF03024876
  29. Trinchieri G. Interleukin-12: a pro inflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity. Annu Rev Immunol 1995; 13: 251-276 https://doi.org/10.1146/annurev.iy.13.040195.001343
  30. Tripathi S, Maiti TK. Stimulation of murine macrophages by native and heat-denatured lectin from Abrus precatorius. Int Immunopharmacol 2003; 3: 375-381 https://doi.org/10.1016/S1567-5769(02)00291-6
  31. Weigand MA, Schmidt H, Zhao Q, Plaschke K, Martin E, Bardenheuer HJ. Ketamine modulates the stimulated adhesion molecule expression on human neutrophils in vitro. Anesth Analg 2000; 90: 206-212 https://doi.org/10.1097/00000539-200001000-00041
  32. Yu Y, Zhou Z, Xu J, Liu Z, Wang Y. Ketamine reduces NFkappaB activation and TNF alpha production in rat mononuclear cells induced by lipopolysaccharide in vitro. Ann Clin Lab Sci 2002; 32: 292-298