Dexmedetomidine decreases the inflammatory response to myocardial surgery under mini-cardiopulmonary bypass

N M H Bulow, E Colpo, R P Pereira, E F M Correa, E P Waczuk, M F Duarte, J B T Rocha, N M H Bulow, E Colpo, R P Pereira, E F M Correa, E P Waczuk, M F Duarte, J B T Rocha

Abstract

Cardiopulmonary bypass (CPB) with extracorporeal circulation produces changes in the immune system accompanied by an increase in proinflammatory cytokines and a decrease in anti-inflammatory cytokines. We hypothesize that dexmedetomidine (DEX) as an anesthetic adjuvant modulates the inflammatory response after coronary artery bypass graft surgery with mini-CPB. In a prospective, randomized, blind study, 12 patients (4 females and 8 males, age range 42-72) were assigned to DEX group and compared with a conventional total intravenous anesthesia (TIVA) group of 11 patients (4 females and 7 males). The endpoints used to assess inflammatory and biochemical responses to mini-CPB were plasma interleukin (IL)-1, IL-6, IL-10, interferon (INF)-γ, tumor necrosis factor (TNF)-α, C-reactive protein, creatine phosphokinase, creatine phosphokinase-MB, cardiac troponin I, cortisol, and glucose levels. These variables were determined before anesthesia, 90 min after beginning CPB, 5 h after beginning CPB, and 24 h after the end of surgery. Endpoints of oxidative stress, including thiobarbituric acid reactive species and delta-aminolevulinate dehydratase activity in erythrocytes were also determined. DEX+TIVA use was associated with a significant reduction in IL-1, IL-6, TNF-α, and INF-γ (P<0.0001) levels compared with TIVA (two-way ANOVA). In contrast, the surgery-induced increase in thiobarbituric acid reactive species was higher in the DEX+TIVA group than in the TIVA group (P<0.01; two-way ANOVA). Delta-aminolevulinate dehydratase activity was decreased after CPB (P<0.001), but there was no difference between the two groups. DEX as an adjuvant in anesthesia reduced circulating IL-1, IL-6, TNF-α, and INF-γ levels after mini-CPB. These findings indicate an interesting anti-inflammatory effect of DEX, which should be studied in different types of surgical interventions.

Figures

Figure 1. Interleukin (IL)-1 (A,B), IL-6 (C,D),…
Figure 1. Interleukin (IL)-1 (A,B), IL-6 (C,D), and IL-10 (E,F) levels were measured in patients who underwent coronary arterial bypass graft surgery under mini-cardiopulmonary bypass, using two different types of anesthesia (conventional total intravenous anesthesia [TIVA; n=11] and TIVA+DEX [n=12]). Two-way ANOVA showed a significant interaction between type of anesthesia and sampling time for IL-1 and IL-6 levels (P
Figure 2. A, B, Plasma γ-interferon (INF-γ)…
Figure 2. A, B, Plasma γ-interferon (INF-γ) and C, D, tumor necrosis factor-α (TNF-α) levels in patients who underwent coronary arterial bypass graft surgery under mini-cardiopulmonary bypass, using two different types of anesthesia (conventional total intravenous anesthesia [TIVA; n=11] and TIVA+DEX [n=12]). Two-way ANOVA showed a significant type of anesthesia versus sampling time interaction for INF-γ and TNF-α levels (P<0.0001). This finding indicated that the increase in INF-γ and TNF-α levels after surgery was lower in patients who were anesthetized with TIVA+DEX than in patients who received only TIVA.
Figure 3. A , B , Thiobarbituric…
Figure 3. A, B, Thiobarbituric reactive substances (TBARS) and C, D, δ-aminolevulinate dehydratase (ALA-D) enzyme activity of erythrocytes in patients who underwent coronary arterial bypass graft surgery under mini-cardiopulmonary bypass, using two different types of anesthesia (conventional total intravenous anesthesia [TIVA; n=11] and TIVA+DEX [n=12]). For TBARS, two-way ANOVA showed a significant sampling time versus type or anesthesia interaction (P<0.001). For ALA-D activity, two-way ANOVA showed only a significant main effect of time (P<0.0001).

References

    1. Sander M, von Heymann C, von Dossow V, Spaethe C, Konertz WF, Jain U, et al. Increased interleukin-6 after cardiac surgery predicts infection. Anesth Analg. 2006;102:1623–1629. doi: 10.1213/01.ane.0000215998.21739.48.
    1. Murkin JM. Panvascular inflammation and mechanisms of injury in perioperative CNS outcomes. Semin Cardiothorac Vasc Anesth. 2010;14:190–195. doi: 10.1177/1089253210378177.
    1. Wan S, LeClerc JL, Vincent JL. Inflammatory response to cardiopulmonary bypass: mechanisms involved and possible therapeutic strategies. Chest. 1997;112:676–692. doi: 10.1378/chest.112.3.676.
    1. Matata BM, Sosnowski AW, Galinanes M. Off-pump bypass graft operation significantly reduces oxidative stress and inflammation. Ann Thorac Surg. 2000;69:785–791. doi: 10.1016/S0003-4975(99)01420-4.
    1. Hall R. Identification of inflammatory mediators and their modulation by strategies for the management of the systemic inflammatory response during cardiac surgery. J Cardiothorac Vasc Anesth. 2015;27:983–1033. doi: 10.1053/j.jvca.2012.09.013.
    1. Mathew JP, Parks R, Savino JS, Friedman AS, Koch C, Mangano DT, et al. Atrial fibrillation following coronary artery bypass graft surgery: predictors, outcomes, and resource utilization. MultiCenter Study of Perioperative Ischemia Research Group. JAMA. 1996;276:300–306. doi: 10.1001/jama.1996.03540040044031.
    1. Rose EA. Off-pump coronary-artery bypass surgery. N Engl J Med. 2003;348:379–380.
    1. Rodemeister S, Duquesne M, Adolph M, Nohr D, Biesalski HK, Unertl K. Massive and long-lasting decrease in vitamin C plasma levels as a consequence of extracorporeal circulation. Nutrition. 2014;30:673–678. doi: 10.1016/j.nut.2013.10.026.
    1. Bulow NMH, Colpo E, Duarte MF, Correa EFM, Schlosser RS, Lauda A, et al. Inflammatory response in patients under coronary artery bypass grafting surgery and clinical implications: a review of the relevance of dexmedetomidine use. ISRN Anesthesiology. 2014 doi: 10.1155/2014/905238.
    1. Aantaa R, Jalonen J. Perioperative use of alpha2-adrenoceptor agonists and the cardiac patient. Eur J Anaesthesiol. 2006;23:361–372. doi: 10.1017/S0265021506000378.
    1. Corcoran TB, Engel A, Sakamoto H, O'Shea A, O'Callaghan-Enright S, Shorten GD. The effects of propofol on neutrophil function, lipid peroxidation and inflammatory response during elective coronary artery bypass grafting in patients with impaired ventricular function. Br J Anaesth. 2006;97:825–831. doi: 10.1093/bja/ael270.
    1. Memis D, Hekimoglu S, Vatan I, Yandim T, Yuksel M, Sut N. Effects of midazolam and dexmedetomidine on inflammatory responses and gastric intramucosal pH to sepsis, in critically ill patients. Br J Anaesth. 2007;98:550–552. doi: 10.1093/bja/aem017.
    1. Nader ND, Ignatowski TA, Kurek CJ, Knight PR, Spengler RN. Clonidine suppresses plasma and cerebrospinal fluid concentrations of TNF-alpha during the perioperative period. Anesth Analg. 2001;93:363–369. doi: 10.1213/00000539-200108000-00026.
    1. Adams R, Brown GT, Davidson M, Fisher E, Mathisen J, Thomson G, et al. Efficacy of dexmedetomidine compared with midazolam for sedation in adult intensive care patients: a systematic review. Br J Anaesth. 2013;111:703–710. doi: 10.1093/bja/aet194.
    1. Abdallah FW, Brull R. Facilitatory effects of perineural dexmedetomidine on neuraxial and peripheral nerve block: a systematic review and meta-analysis. Br J Anaesth. 2013;110:915–925. doi: 10.1093/bja/aet066.
    1. Candiotti KA, Bergese SD, Bokesch PM, Feldman MA, Wisemandle W, Bekker AY. Monitored anesthesia care with dexmedetomidine: a prospective, randomized, double-blind, multicenter trial. Anesth Analg. 2010;110:47–56. doi: 10.1213/ane.0b013e3181ae0856.
    1. Can M, Gul S, Bektas S, Hanci V, Acikgoz S. Effects of dexmedetomidine or methylprednisolone on inflammatory responses in spinal cord injury. Acta Anaesthesiol Scand. 2009;53:1068–1072. doi: 10.1111/j.1399-6576.2009.02019.x.
    1. Gu J, Chen J, Xia P, Tao G, Zhao H, Ma D. Dexmedetomidine attenuates remote lung injury induced by renal ischemia-reperfusion in mice. Acta Anaesthesiol Scand. 2011;55:1272–1278. doi: 10.1111/j.1399-6576.2011.02526.x.
    1. Sleigh J. All hands on dex. Anaesthesia. 2012;67:1193–1197. doi: 10.1111/j.1365-2044.2012.07334.x.
    1. Grotto D, Santa Maria L, Valentini J, Paniz C, Schmitt G, Garcia SC, et al. Importance of the lipid peroxidation biomarkers and methodological aspects for malondialdehyde quantification. Quimica Nova. 2009;32:169–174. doi: 10.1590/S0100-40422009000100032.
    1. Rocha JBT, Saraiva RA, Garcia SC, Gravina FS, Nogueira CW. Aminolevulinate dehydratase (δ-ALA-D) as marker protein of intoxication with metals and other pro-oxidant situations. Toxicol Res. 2012;1:85–102. doi: 10.1039/C2TX20014G.
    1. Rocha JBT, Bulow NMH, Correa EF, Scholze C, Nogueira CW, Barbosa NBV. Dexmedetomidine protects blood δ-minolevulinate dehydratase from inactivation caused by hyperoxygenation in total intravenous anesthesia. Hum Exp Tox. 2011;30:289–295. doi: 10.1177/0960327110372399.
    1. Straub RH, Herrmann M, Berkmiller G, Frauenholz T, Lang B, Scholmerich J, et al. Neuronal regulation of interleukin 6 secretion in murine spleen: adrenergic and opioidergic control. J Neurochem. 1997;68:1633–1639. doi: 10.1046/j.1471-4159.1997.68041633.x.
    1. Wu X, Song X, Li N, Zhan L, Meng Q, Xia Z. Protective effects of dexmedetomidine on blunt chest trauma-induced pulmonary contusion in rats. J Trauma Acute Care Surg. 2013;74:524–530. doi: 10.1097/TA.0b013e31827d5de3.
    1. Taniguchi T, Kidani Y, Kanakura H, Takemoto Y, Yamamoto K. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Crit Care Med. 2004;32:1322–1326. doi: 10.1097/01.CCM.0000128579.84228.2A.
    1. Taniguchi T, Kurita A, Kobayashi K, Yamamoto K, Inaba H. Dose- and time-related effects of dexmedetomidine on mortality and inflammatory responses to endotoxin-induced shock in rats. J Anesth. 2008;22:221–228. doi: 10.1007/s00540-008-0611-9.
    1. Lai YC, Tsai PS, Huang CJ. Effects of dexmedetomidine on regulating endotoxin-induced up-regulation of inflammatory molecules in murine macrophages. J Surg Res. 2009;154:212–219. doi: 10.1016/j.jss.2008.07.010.
    1. Szelenyi J, Kiss JP, Vizi ES. Differential involvement of sympathetic nervous system and immune system in the modulation of TNF-alpha production by alpha2- and beta-adrenoceptors in mice. J Neuroimmunol. 2000;103:34–40. doi: 10.1016/S0165-5728(99)00234-9.
    1. Hofer S, Steppan J, Wagner T, Funke B, Lichtenstern C, Martin E, et al. Central sympatholytics prolong survival in experimental sepsis. Crit Care. 2009;13:R11. doi: 10.1186/cc7709.
    1. Tasdogan M, Memis D, Sut N, Yuksel M. Results of a pilot study on the effects of propofol and dexmedetomidine on inflammatory responses and intraabdominal pressure in severe sepsis. J Clin Anesth. 2009;21:394–400. doi: 10.1016/j.jclinane.2008.10.010.
    1. Peng M, Wang YL, Wang CY, Chen C. Dexmedetomidine attenuates lipopolysaccharide-induced proinflammatory response in primary microglia. J Surg Res. 2013;179:e219–e225. doi: 10.1016/j.jss.2012.05.047.
    1. Maze M, Virtanen R, Daunt D, Banks SJ, Stover EP, Feldman D. Effects of dexmedetomidine, a novel imidazole sedative-anesthetic agent, on adrenal steroidogenesis: in vivo and in vitro studies. Anesth Analg. 1991;73:204–208.
    1. Venn RM, Bryant A, Hall GM, Grounds RM. Effects of dexmedetomidine on adrenocortical function, and the cardiovascular, endocrine and inflammatory responses in post-operative patients needing sedation in the intensive care unit. Br J Anaesth. 2001;86:650–656. doi: 10.1093/bja/86.5.650.
    1. Bulow NM, Barbosa NV, Rocha JB. Opioid consumption in total intravenous anesthesia is reduced with dexmedetomidine: a comparative study with remifentanil in gynecologic videolaparoscopic surgery. J Clin Anesth. 2007;19:280–285. doi: 10.1016/j.jclinane.2007.01.004.
    1. Bekker A, Haile M, Kline R, Didehvar S, Babu R, Martiniuk F, et al. The effect of intraoperative infusion of dexmedetomidine on the quality of recovery after major spinal surgery. J Neurosurg Anesthesiol. 2013;25:16–24. doi: 10.1097/ANA.0b013e31826318af.

Source: PubMed

3
Subskrybuj