Xenon triggers pro-inflammatory effects and suppresses the anti-inflammatory response compared to sevoflurane in patients undergoing cardiac surgery

Thomas Breuer, Christoph Emontzpohl, Mark Coburn, Carina Benstoem, Rolf Rossaint, Gernot Marx, Gereon Schälte, Juergen Bernhagen, Christian S Bruells, Andreas Goetzenich, Christian Stoppe, Thomas Breuer, Christoph Emontzpohl, Mark Coburn, Carina Benstoem, Rolf Rossaint, Gernot Marx, Gereon Schälte, Juergen Bernhagen, Christian S Bruells, Andreas Goetzenich, Christian Stoppe

Abstract

Introduction: Cardiac surgery encompasses various stimuli that trigger pro-inflammatory mediators, reactive oxygen species and mobilization of leucocytes. The aim of this study was to evaluate the effect of xenon on the inflammatory response during cardiac surgery.

Methods: This randomized trial enrolled 30 patients who underwent elective on-pump coronary-artery bypass grafting in balanced anaesthesia of either xenon or sevoflurane. For this secondary analysis, blood samples were drawn prior to the operation, intra-operatively and on the first post-operative day to measure the pro- and anti-inflammatory cytokines interleukin-6 (IL-6), interleukin-8/C-X-C motif ligand 8 (IL-8/CXCL8), and interleukin-10 (IL-10). Chemokines such as C-X-C motif ligand 12/ stromal cell-derived factor-1α (CXCL12/SDF-1α) and macrophage migration inhibitory factor (MIF) were measured to characterize xenon's perioperative inflammatory profile and its impact on migration of peripheral blood mononuclear cells (PBMC).

Results: Xenon enhanced the postoperative increase of IL-6 compared to sevoflurane (Xenon: 90.7 versus sevoflurane: 33.7 pg/ml; p = 0.035) and attenuated the increase of IL-10 (Xenon: 127.9 versus sevoflurane: 548.3 pg/ml; p = 0.028). Both groups demonstrated a comparable intraoperative increase of oxidative stress (intra-OP: p = 0.29; post-OP: p = 0.65). While both groups showed an intraoperative increase of the cardioprotective mediators MIF and CXCL12/SDF-1α, only MIF levels decreased in the xenon group on the first postoperative day (50.0 ng/ml compared to 23.3 ng/ml; p = 0.012), whereas it remained elevated after sevoflurane anaesthesia (58.3 ng/ml to 53.6 ng/ml). Effects of patients' serum on chemotactic migration of peripheral mononuclear blood cells taken from healthy volunteers indicated a tendency towards enhanced migration after sevoflurane anaesthesia (p = 0.07).

Conclusions: Compared to sevoflurane, balanced xenon anaesthesia triggers pro-inflammatory effects and suppresses the anti-inflammatory response in cardiac surgery patients even though the clinical significance remains unknown.

Trial registration: This clinical trial was approved by the European Medicines Agency (EudraCT-number: 2010-023942-63) and at ClinicalTrials.gov ( NCT01285271 ; first received: January 24, 2011).

Figures

Fig. 1
Fig. 1
Perioperative time course of interleukin (IL)-6 (IL-6) (a), IL-8 (b) and IL-10 (c). Circulating serum levels of IL-6, IL-8, and IL-10 were measured perioperatively in serum samples of patients who underwent cardiac surgery. Values are depicted in pg/ml. IL-6, IL-8 and IL-10 increased during the surgical intervention. While IL-6 levels increased postoperatively in the xenon group compared to the sevoflurane group, IL-10 levels only increased significantly in the sevoflurane group during surgery. Serum levels of IL-8 levels did not differ between both groups during the observation. Data are shown as boxplots with means and maximal to minimal values (p values are indicated within the figure). Pre-OP baseline, before induction of anaesthesia, intra-op immediately before termination of surgery, post-OP 24 h after surgery
Fig. 2
Fig. 2
a Perioperative time course of CXCL12/stromal cell-derived factor 1α (SDF-1α) serum concentrations. Measurement of CXCL12/SDF-1α levels in the serum of patients who underwent cardiac surgery. Values are depicted in pg/ml. CXCL12 levels increased in both groups during surgical intervention. However, there were no significant differences between groups. Shown as boxplots with means and maximal to minimal values (p values are indicated). Pre-OP baseline, before induction of anaesthesia, intra-op immediately before termination of surgery, post-OP 24 h after surgery. b Perioperative time course of macrophage migration inhibitory factor (MIF) serum concentrations. The measurement of perioperative circulating MIF levels in cardiac surgery patients was performed as described previously [25]. Values are depicted in ng/ml. There was a strong intraoperative increase in serum MIF levels in both groups, which decreased again postoperatively. Postoperative measured MIF levels were significantly reduced after xenon anaesthesia compared to sevoflurane. Data are shown as interleaved boxes with means and maximal to minimal values (p values are indicated)
Fig. 3
Fig. 3
Migration assay of peripheral blood mononuclear cells (PBMCs) in serum samples of cardiac surgery patients after xenon or sevoflurane anaesthesia. a In vitro migration of PBMCs is increased by serum samples of patients after sevoflurane anaesthesia. Extent of PBMC migration (received from healthy volunteers) towards serum samples from cardiac surgery patients is demonstrated in both groups. While PBMC migration towards serum samples was increased during surgery in the sevoflurane group, PBMC migration was not affected in serum samples from the xenon group. Data are shown in boxplots with means and maximal to minimal values (p values are indicated). Pre-OP baseline, before induction of anaesthesia, intra-op immediately before termination of surgery, post-OP 24 h after surgery. b Serum samples from patients after xenon anaesthesia show no chemokinetic effect on PBMCs. Migration of PBMCs from healthy volunteers, which were pre-incubated in serum samples elicited by recombinant CCL2/monocyte chemoattractant protein-1 (MCP-1), is demonstrated. No significant difference was measured between the groups during surgery. Data are shown as boxplots with means and maximal to minimal values (p values are indicated)

References

    1. Dingley J, Tooley J, Porter H, Thoresen M. Xenon provides short-term neuroprotection in neonatal rats when administered after hypoxia-ischemia. Stroke. 2006;37:501–6. doi: 10.1161/.
    1. Coburn M, Maze M, Franks NP. The neuroprotective effects of xenon and helium in an in vitro model of traumatic brain injury. Crit Care Med. 2008;36:588–95. doi: 10.1097/01.CCM.0B013E3181611F8A6.
    1. Schmidt M, Marx T, Glöggl E, Reinelt H, Schirmer U. Xenon attenuates cerebral damage after ischemia in pigs. Anesthesiology. 2005;102:929–36. doi: 10.1097/00000542-200505000-00011.
    1. de Sousa SL, Dickinson R, Lieb WR, Franks NP. Contrasting synaptic actions of the inhalational general anesthetics isoflurane and xenon. Anesthesiology. 2000;92:1055–66. doi: 10.1097/00000542-200004000-00024.
    1. Franks NP, Dickinson R, de Sousa SL, Hall AC, Lieb WR. How does xenon produce anaesthesia? Nature. 1998;396:324–4. doi: 10.1038/24525.
    1. Saravanan P, Exley AR, Valchanov K, Casey ND, Falter F. Impact of xenon anaesthesia in isolated cardiopulmonary bypass on very early leucocyte and platelet activation and clearance: a randomized, controlled study. Br J Anaesth. 2009;103:805–10. doi: 10.1093/bja/aep297.
    1. de Rossi LW, Horn NA, Stevanovic A, Buhre W, Hutschenreuter G, Rossaint R. Xenon modulates neutrophil adhesion molecule expression in vitro. Eur J Anaesthesiol. 2004;21:139–43. doi: 10.1097/00003643-200402000-00010.
    1. Crumrine RC, LaManna JC. Regional cerebral metabolites, blood flow, plasma volume, and mean transit time in total cerebral ischemia in the rat. J Cereb Blood Flow Metab. 1991;11:272–82. doi: 10.1038/jcbfm.1991.59.
    1. Petito CK, Feldmann E, Pulsinelli WA, Plum F. Delayed hippocampal damage in humans following cardiorespiratory arrest. Neurology. 1987;37:1281–6. doi: 10.1212/WNL.37.8.1281.
    1. Levine B, Kalman J, Mayer L, Fillit HM, Packer M. Elevated circulating levels of tumor necrosis factor in severe chronic heart failure. N Engl J Med. 1990;323:236–41. doi: 10.1056/NEJM199007263230405.
    1. Suzuki H, Sato R, Sato T, Shoji M, Iso Y, Kondo T, et al. Time-course of changes in the levels of interleukin 6 in acutely decompensated heart failure. Int J Cardiol. 2005;100:415–20. doi: 10.1016/j.ijcard.2004.08.041.
    1. Franke A, Lante W, Fackeldey V, Becker HP, Kurig E, Zöller LG, et al. Pro-inflammatory cytokines after different kinds of cardio-thoracic surgical procedures: is what we see what we know? Eur J Cardiothorac Surg. 2005;28:569–75. doi: 10.1016/j.ejcts.2005.07.007.
    1. Petzelbauer P, Zacharowski PA, Miyazaki Y, Friedl P, Wickenhauser G, Castellino FJ, et al. The fibrin-derived peptide Bbeta15-42 protects the myocardium against ischemia-reperfusion injury. Nat Med. 2005;11:298–304. doi: 10.1038/nm1198.
    1. Stoppe C, Cremer J, Rex S, Schälte G, Fahlenkamp AV, Rossaint R, et al. Xenon anaesthesia for laparoscopic cholecystectomy in a patient with multiple chemical sensitivity. Br J Anaesth. 2011;107:645–7. doi: 10.1093/bja/aer285.
    1. Hirai S. Systemic inflammatory response syndrome after cardiac surgery under cardiopulmonary bypass. Ann Thorac Cardiovasc Surg. 2003;9:365–70.
    1. Schilling T, Kozian A, Senturk M, Huth C, Reinhold A, Hedenstierna G, et al. Effects of volatile and intravenous anesthesia on the alveolar and systemic inflammatory response in thoracic surgical patients. Anesthesiology. 2011;115:65–74. doi: 10.1097/ALN.0b013e318214b9de.
    1. Tandon M, Pandey CK. Myocardial oxidative stress protection with sevoflurane versus propofol. Eur J Anaesthesiol. 2012;29:296–7. doi: 10.1097/EJA.0b013e328351660a.
    1. Rossaint R, Reyle-Hahn M. Schulte am Esch J, Scholz J, Scherpereel P, Vallet B, et al. Multicenter randomized comparison of the efficacy and safety of xenon and isoflurane in patients undergoing elective surgery. Anesthesiology. 2003;98:6–13. doi: 10.1097/00000542-200301000-00005.
    1. Coburn M, Kunitz O, Baumert J-H, Hecker K, Haaf S, Zühlsdorff A, et al. Randomized controlled trial of the haemodynamic and recovery effects of xenon or propofol anaesthesia. Br J Anaesth. 2005;94:198–202. doi: 10.1093/bja/aei023.
    1. Wappler F, Rossaint R, Baumert J, Scholz J, Tonner PH, van Aken H, et al. Multicenter randomized comparison of xenon and isoflurane on left ventricular function in patients undergoing elective surgery. Anesthesiology. 2007;106:463–71. doi: 10.1097/00000542-200703000-00010.
    1. Baumert J-H, Hein M, Hecker KE, Satlow S, Neef P, Rossaint R. Xenon or propofol anaesthesia for patients at cardiovascular risk in non-cardiac surgery. Br J Anaesth. 2008;100:605–11. doi: 10.1093/bja/aen050.
    1. Preckel B, Müllenheim J, Moloschavij A, Thämer V, Schlack W. Xenon administration during early reperfusion reduces infarct size after regional ischemia in the rabbit heart in vivo. Anesth Analg. 2000;91:1327–32. doi: 10.1097/00000539-200012000-00003.
    1. Hartlage MAG, Berendes E, van Aken H, Fobker M, Theisen M, Weber TP. Xenon improves recovery from myocardial stunning in chronically instrumented dogs. Anesth Analg. 2004;99:655–64. doi: 10.1213/01.ANE.0000129999.74324.4E.
    1. Stoppe C, Fahlenkamp AV, Rex S, Veeck NC, Gozdowsky SC, Schälte G, et al. Feasibility and safety of xenon compared with sevoflurane anaesthesia in coronary surgical patients: a randomized controlled pilot study. Br J Anaesth. 2013;111:406–16. doi: 10.1093/bja/aet072.
    1. Stoppe C, Werker T, Rossaint R, Dollo F, Lue H, Wonisch W, et al. What is the significance of perioperative release of macrophage migration inhibitory factor in cardiac surgery? Antioxid Redox Signal. 2013;19:231–9. doi: 10.1089/ars.2012.5015.
    1. Stoppe C, Grieb G, Rossaint R, Simons D, Coburn M, Götzenich A, et al. High postoperative blood levels of macrophage migration inhibitory factor are associated with less organ dysfunction in patients after cardiac surgery. Mol Med. 2012;18:843–50. doi: 10.2119/molmed.2012.00071.
    1. Rael LT, Bar-Or R, Aumann RM, Slone DS, Mains CW, Bar-Or D. Oxidation-reduction potential and paraoxonase-arylesterase activity in trauma patients. Biochem Biophys Res Commun. 2007;361:561–5. doi: 10.1016/j.bbrc.2007.07.078.
    1. Zakkar M, Guida G, Suleiman M-S, Angelini GD. Cardiopulmonary Bypass and Oxidative Stress. Oxid Med Cell Longev. 2015;2015:189863–8. doi: 10.1155/2015/189863.
    1. Palmieri B, Sblendorio V. Oxidative stress tests: overview on reliability and use. Part I. Eur Rev Med Pharmacol Sci. 2007;11:309–42.
    1. Rael LT, Bar-Or R, Mains CW, Slone DS, Levy AS, Bar-Or D. Plasma oxidation-reduction potential and protein oxidation in traumatic brain injury. J Neurotrauma. 2009;26:1203–11. doi: 10.1089/neu.2008.0816.
    1. Laffey JG, Boylan JF, Cheng DCH. The systemic inflammatory response to cardiac surgery: implications for the anesthesiologist. Anesthesiology. 2002;97:215–52. doi: 10.1097/00000542-200207000-00030.
    1. Miller EJ, Li J, Leng L, McDonald C, Atsumi T, Bucala R, et al. Macrophage migration inhibitory factor stimulates AMP-activated protein kinase in the ischaemic heart. Nature. 2008;451:578–82. doi: 10.1038/nature06504.
    1. Koga K, Kenessey A, Powell SR, Sison CP, Miller EJ, Ojamaa K. Macrophage migration inhibitory factor provides cardioprotection during ischemia/reperfusion by reducing oxidative stress. Antioxid Redox Signal. 2011;14:1191–202. doi: 10.1089/ars.2010.3163.
    1. Kawamura T, Kadosaki M, Nara N, Kaise A, Suzuki H, Endo S, et al. Effects of sevoflurane on cytokine balance in patients undergoing coronary artery bypass graft surgery. J Cardiothorac Vasc Anesth. 2006;20:503–8. doi: 10.1053/j.jvca.2006.01.011.
    1. Nader ND, Karamanoukian HL, Reedy RL, Salehpour F, Knight PR. Inclusion of sevoflurane in cardioplegia reduces neutrophil activity during cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 2006;20:57–62. doi: 10.1053/j.jvca.2005.07.030.
    1. Heindl B, Conzen PF, Becker BF. The volatile anesthetic sevoflurane mitigates cardiodepressive effects of platelets in reperfused hearts. Basic Res Cardiol. 1999;94:102–11. doi: 10.1007/s003950050132.
    1. Gregory JL, Morand EF, McKeown SJ, Ralph JA, Hall P, Yang YH, et al. Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J Immunol. 2006;177:8072–9. doi: 10.4049/jimmunol.177.11.8072.
    1. Baggiolini M, Clark-Lewis I. Interleukin-8, a chemotactic and inflammatory cytokine. FEBS Lett. 1992;307:97–101. doi: 10.1016/0014-5793(92)80909-Z.
    1. Lin TJ, Issekutz TB, Marshall JS. Human mast cells transmigrate through human umbilical vein endothelial monolayers and selectively produce IL-8 in response to stromal cell-derived factor-1 alpha. J Immunol. 2000;165:211–20. doi: 10.4049/jimmunol.165.1.211.
    1. Meiron M, Zohar Y, Anunu R, Wildbaum G, Karin N. CXCL12 (SDF-1alpha) suppresses ongoing experimental autoimmune encephalomyelitis by selecting antigen-specific regulatory T cells. J Exp Med. 2008;205:2643–55. doi: 10.1084/jem.20080730.
    1. Seeger FH, Rasper T, Fischer A, Muhly-Reinholz M, Hergenreider E, Leistner DM, et al. Heparin disrupts the CXCR4/SDF-1 axis and impairs the functional capacity of bone marrow-derived mononuclear cells used for cardiovascular repair. Circ Res. 2012;111:854–62. doi: 10.1161/CIRCRESAHA.112.265678.
    1. Roth E, Manhart N, Wessner B. Assessing the antioxidative status in critically ill patients. Curr Opin Clin Nutr Metab Care. 2004;7:161–8. doi: 10.1097/00075197-200403000-00010.
    1. Hearse DJ, Humphrey SM, Bullock GR. The oxygen paradox and the calcium paradox: two facets of the same problem? J Mol Cell Cardiol. 1978;10:641–68. doi: 10.1016/S0022-2828(78)80004-2.
    1. Dhalla NS, Elmoselhi AB, Hata T, Makino N. Status of myocardial antioxidants in ischemia-reperfusion injury. Cardiovasc Res. 2000;47:446–56. doi: 10.1016/S0008-6363(00)00078-X.
    1. Fahlenkamp AV, Coburn M, Rossaint R, Stoppe C, Haase H. Comparison of the effects of xenon and sevoflurane anaesthesia on leucocyte function in surgical patients: a randomized trial. Br J Anaesth. 2014;112:272–80. doi: 10.1093/bja/aet330.
    1. Boeken U, Feindt P, Zimmermann N, Kalweit G, Petzold T, Gams E. Increased preoperative C-reactive protein (CRP)-values without signs of an infection and complicated course after cardiopulmonary bypass (CPB)-operations. Eur J Cardiothorac Surg. 1998;13:541–5. doi: 10.1016/S1010-7940(98)00062-1.
    1. Clark JA, Ma D, Homi HM, Maze M, Grocott HP. Xenon and the inflammatory response to cardiopulmonary bypass in the rat. J Cardiothorac Vasc Anesth. 2005;19:488–93. doi: 10.1053/j.jvca.2005.05.007.

Source: PubMed

3
Suscribir