Effects of perioperative transcutaneous electrical acupoint stimulation on monocytic HLA-DR expression in patients undergoing coronary artery bypass grafting with cardiopulmonary bypass: study protocol for a double-blind randomized controlled trial

Wen-Ting Chen, Jin-Feng Wei, Lan Wang, Deng-Wen Zhang, Wei Tang, Jian Wang, Yue Yong, Jing Wang, Ya-Lan Zhou, Lan Yuan, Guo-Qiang Fu, Sheng Wang, Jian-Gang Song, Wen-Ting Chen, Jin-Feng Wei, Lan Wang, Deng-Wen Zhang, Wei Tang, Jian Wang, Yue Yong, Jing Wang, Ya-Lan Zhou, Lan Yuan, Guo-Qiang Fu, Sheng Wang, Jian-Gang Song

Abstract

Background: Cardiac surgery involving cardiopulmonary bypass (CPB) is known to be associated with a transient postoperative immunosuppression. When severe and persistent, this immune dysfunction predisposes patients to infectious complications, which contributes to a prolonged stay in the intensive care unit (ICU), and even mortality. Effective prevention and treatment methods are still lacking. Recent studies revealed that acupuncture-related techniques, such as electroacupuncture and transcutaneous electrical acupoint stimulation (TEAS), are able to produce effective cardioprotection and immunomodulation in adult and pediatric patients undergoing cardiac surgery with CPB, which leads to enhanced recovery. However, whether perioperative application of TEAS, a non-invasive technique, is able to improve immunosuppression of the patients with post-cardiosurgical conditions is unknown. Thus, as a preliminary study, the main objective is to evaluate the effects of TEAS on the postoperative expression of monocytic human leukocyte antigen (-D related) (mHLA-DR), a standardized "global" biomarker of injury or sepsis-associated immunosuppression, in patients receiving on-pump coronary artery bypass grafting (CABG).

Methods: This study is a single-center clinical trial. The 88 patients scheduled to receive CABG under CPB will be randomized into two groups: the group receiving TEAS, and the group receiving transcutaneous acupoint pseudo-electric stimulation (Sham TEAS). Expression of mHLA-DR serves as a primary endpoint, and other laboratory parameters (e.g., interleukin [IL]-6, IL-10) and clinical outcomes (e.g., postoperative infectious complications, ICU stay time, and mortality) as the secondary endpoints. In addition, immune indicators, such as high mobility group box 1 protein and regulatory T cells will also be measured.

Discussion: The current study is a preliminary monocentric clinical trial with a non-clinical primary endpoint, expression of mHLA-DR, aiming at determining whether perioperative application of TEAS has a potential to reverse CABG-associated immunosuppression. Although the immediate clinical impact of this study is limited, its results would inform further large-sample clinical trials using relevant patient-centered clinical outcomes as primary endpoints.

Trial registration: ClinicalTrials.gov, NCT02933996. Registered on 13 October 2016.

Keywords: CABG; Immunosuppression; TEAS; mHLA-DR.

Conflict of interest statement

The authors declare that they have no competing interests. The Ethics Committee of Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine is independent from the sponsor and competing interests.

Figures

Fig. 1
Fig. 1
Flowchart of the study
Fig. 2
Fig. 2
Zusanli location: outside of the shank, 3 cun(10 cm) below Dubi acupoint and a finger’s width (middle finger) to tibial front edge
Fig. 3
Fig. 3
Shenshu location: below the spinous process of the second lumbar vertebra, 1.5 cun (5 cm) to the central line
Fig. 4
Fig. 4
The patients will receive TEAS therapy 30 min before anesthesia (one stimulation for 30 min), during the period of surgery (stimulation for the whole course) and within 24h after surgery (four times of stimulation, each for 30min)
Fig. 5
Fig. 5
We will use Low-frequency electronic pulse therapeutic device G6805-2 (Huayi, Shanghai, China) for TEAS therapy, the frequency will be 2/100 Hz alternating and the intensity will be 15mA
Fig. 6
Fig. 6
a: Patient received TEAS therapy at bilateral Zusanli acupoint (ST36) and Shenshu acupoint (BL23) during the perioperative period. b: Zusanli acupoint (ST36) is identified. c: Shenshu acupoint (BL23) is identified

References

    1. Butler J, Rocker GM, Westaby S. Inflammatory response to cardiopulmonary bypass. Ann Thorac Surg. 1993;55(2):552–559. doi: 10.1016/0003-4975(93)91048-R.
    1. Kollef MH, et al. The impact of nosocomial infections on patient outcomes following cardiac surgery. Chest. 1997;112(3):666–675. doi: 10.1378/chest.112.3.666.
    1. Hortal J, et al. Ventilator-associated pneumonia in patients undergoing major heart surgery: an incidence study in Europe. Crit Care. 2009;13(3):R80. doi: 10.1186/cc7896.
    1. Bouza E, et al. Infections following major heart surgery in European intensive care units: there is room for improvement (ESGNI 007 Study) J Hosp Infect. 2006;63(4):399–405. doi: 10.1016/j.jhin.2006.03.007.
    1. Leal-Noval SR, et al. Nosocomial pneumonia in patients undergoing heart surgery. Crit Care Med. 2000;28(4):935–940. doi: 10.1097/00003246-200004000-00004.
    1. Kirklin JK, et al. Complement and the damaging effects of cardiopulmonary bypass. J Thorac Cardiovasc Surg. 1983;86(6):845–857. doi: 10.1016/S0022-5223(19)39061-0.
    1. Sawa Y, et al. Attenuation of cardiopulmonary bypass-derived inflammatory reactions reduces myocardial reperfusion injury in cardiac operations. J Thorac Cardiovasc Surg. 1996;111(1):29–35. doi: 10.1016/S0022-5223(96)70398-7.
    1. Kharazmi A, et al. Endotoxemia and enhanced generation of oxygen radicals by neutrophils from patients undergoing cardiopulmonary bypass. J Thorac Cardiovasc Surg. 1989;98(3):381–385. doi: 10.1016/S0022-5223(19)34384-3.
    1. Landow L, Andersen LW. Splanchnic ischaemia and its role in multiple organ failure. Acta Anaesthesiol Scand. 1994;38(7):626–639. doi: 10.1111/j.1399-6576.1994.tb03969.x.
    1. Hall RI, Smith MS, Rocker G. The systemic inflammatory response to cardiopulmonary bypass: pathophysiological, therapeutic, and pharmacological considerations. Anesth Analg. 1997;85(4):766–782. doi: 10.1213/00000539-199710000-00011.
    1. Butler J, et al. Effect of cardiopulmonary bypass on systemic release of neutrophil elastase and tumor necrosis factor. J Thorac Cardiovasc Surg. 1993;105(1):25–30. doi: 10.1016/S0022-5223(19)33843-7.
    1. Hill GE. Cardiopulmonary bypass-induced inflammation: is it important? J Cardiothorac Vasc Anesth. 1998;12(2 Suppl 1):21–25.
    1. Stoppelkamp S, et al. Identification of predictive early biomarkers for sterile-SIRS after cardiovascular surgery. PLoS One. 2015;10(8):e0135527. doi: 10.1371/journal.pone.0135527.
    1. Hadley JS, et al. Alterations in inflammatory capacity and TLR expression on monocytes and neutrophils after cardiopulmonary bypass. Shock. 2007;27(5):466–473. doi: 10.1097/01.shk.0000245033.69977.c5.
    1. Naldini A, et al. In vitro cytokine production and T-cell proliferation in patients undergoing cardiopulmonary by-pass. Cytokine. 1995;7(2):165–170. doi: 10.1006/cyto.1995.1022.
    1. Lee H, et al. Acupuncture for symptom management of rheumatoid arthritis: a pilot study. Clin Rheumatol. 2008;27(5):641–5. doi: 10.1007/s10067-007-0819-3.
    1. Ng DK, et al. A double-blind, randomized, placebo-controlled trial of acupuncture for the treatment of childhood persistent allergic rhinitis. Pediatrics. 2004;114(5):1242–1247. doi: 10.1542/peds.2004-0744.
    1. Biernacki W, Peake MD. Acupuncture in treatment of stable asthma. Respir Med. 1998;92(9):1143–1145. doi: 10.1016/S0954-6111(98)90409-7.
    1. Yang L, et al. Cardioprotective effects of electroacupuncture pretreatment on patients undergoing heart valve replacement surgery: a randomized controlled trial. Ann Thorac Surg. 2010;89(3):781–786. doi: 10.1016/j.athoracsur.2009.12.003.
    1. Ni X, et al. Cardioprotective effect of transcutaneous electric acupoint stimulation in the pediatric cardiac patients: a randomized controlled clinical trial. Paediatr Anaesth. 2012;22(8):805–811. doi: 10.1111/j.1460-9592.2012.03822.x.
    1. Wang J, et al. Electroacupuncture suppresses surgical trauma stress-induced lymphocyte apoptosis in rats. Neurosci Lett. 2005;383(1–2):68–72. doi: 10.1016/j.neulet.2005.03.068.
    1. Cheng XD, Wu GC, et al. Dynamic observation on regulation of electroacupuncture on the proliferation responses of spleen lymphocytes from traumatized rats. Chin J Immunol. 1997;13(2):68–70.
    1. Cheng XD, et al. Effect of continued electroacupuncture on induction of interleukin-2 production of spleen lymphocytes from the injured rats. Acupunct Electrother Res. 1997;22(1):1–8. doi: 10.3727/036012997816356770.
    1. Xiaoding C. Involvement of orphanin FQ in electroacupuncture modulation on immunosuppression by trauma. Zhen Ci Yan Jiu. 2001;26:219–220.
    1. Yu Y, et al. Role of endogenous interferon-gamma on the enhancement of splenic NK cell activity by electroacupuncture stimulation in mice. J Neuroimmunol. 1998;90(2):176–186. doi: 10.1016/S0165-5728(98)00143-X.
    1. Wang K, et al. The effects of electroacupuncture on TH1/TH2 cytokine mRNA expression and mitogen-activated protein kinase signaling pathways in the splenic T cells of traumatized rats. Anesth Analg. 2009;109(5):1666–1673. doi: 10.1213/ANE.0b013e3181b5a234.
    1. Zhang Q, et al. The effect of pre-treatment with transcutaneous electrical acupoint stimulation on the quality of recovery after ambulatory breast surgery: a prospective, randomised controlled trial. Anaesthesia. 2014;69(8):832–839. doi: 10.1111/anae.12639.
    1. Man KM, et al. Transcutaneous electrical nerve stimulation on ST36 and SP6 acupoints prevents hyperglycaemic response during anaesthesia: a randomised controlled trial. Eur J Anaesthesiol. 2011;28(6):420–6. doi: 10.1097/EJA.0b013e32833fad52.
    1. Wang H, et al. Transcutaneous electric acupoint stimulation reduces intra-operative remifentanil consumption and alleviates postoperative side-effects in patients undergoing sinusotomy: a prospective, randomized, placebo-controlled trial. Br J Anaesth. 2014;112(6):1075–1082. doi: 10.1093/bja/aeu001.
    1. Yeh ML, et al. Effect of transcutaneous acupoint electrical stimulation on post-hemorrhoidectomy-associated pain, anxiety, and heart rate variability: a randomized-controlled study. Clin Nurs Res. 2018;27(4):450–466. doi: 10.1177/1054773816685745.
    1. Grech D, et al. Intraoperative low-frequency electroacupuncture under general anesthesia improves postoperative recovery in a randomized trial. J Acupunct Meridian Stud. 2016;9(5):234–241. doi: 10.1016/j.jams.2016.03.009.
    1. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol. 2015;15(4):203–216. doi: 10.1038/nri3818.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–268. doi: 10.1016/S1473-3099(13)70001-X.
    1. Monneret G, et al. Monitoring immune dysfunctions in the septic patient: a new skin for the old ceremony. Mol Med. 2008;14(1–2):64–78. doi: 10.2119/2007-00102.Monneret.
    1. Boomer JS, et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011;306(23):2594–2605. doi: 10.1001/jama.2011.1829.
    1. Cheron A, et al. Lack of recovery in monocyte human leukocyte antigen-DR expression is independently associated with the development of sepsis after major trauma. Crit Care. 2010;14(6):R208. doi: 10.1186/cc9331.
    1. Venet F, et al. Decreased monocyte human leukocyte antigen-DR expression after severe burn injury: correlation with severity and secondary septic shock. Crit Care Med. 2007;35(8):1910–1917. doi: 10.1097/01.CCM.0000275271.77350.B6.
    1. Ho YP, et al. A strong association between down-regulation of HLA-DR expression and the late mortality in patients with severe acute pancreatitis. Am J Gastroenterol. 2006;101(5):1117–1124. doi: 10.1111/j.1572-0241.2006.00495.x.
    1. Satoh A, et al. Human leukocyte antigen-DR expression on peripheral monocytes as a predictive marker of sepsis during acute pancreatitis. Pancreas. 2002;25(3):245–250. doi: 10.1097/00006676-200210000-00006.
    1. Reinke P, et al. Late acute rejection in long-term renal allograft recipients. Diagnostic and predictive value of circulating activated T cells. Transplantation. 1994;58(1):35–41. doi: 10.1097/00007890-199407000-00007.
    1. Antoniades CG, et al. Reduced monocyte HLA-DR expression: a novel biomarker of disease severity and outcome in acetaminophen-induced acute liver failure. Hepatology. 2006;44(1):34–43. doi: 10.1002/hep.21240.
    1. Ahlstrom A, et al. Predictive value of interleukins 6, 8 and 10, and low HLA-DR expression in acute renal failure. Clin Nephrol. 2004;61(2):103–110. doi: 10.5414/CNP61103.
    1. Meisel A, et al. Predicting post-stroke infections and outcome with blood-based immune and stress markers. Cerebrovasc Dis. 2012;33(6):580–588. doi: 10.1159/000338080.
    1. Venet F, et al. Decreased monocyte HLA-DR expression in patients after non-shockable out-of-hospital cardiac arrest. Shock. 2016;46(1):33–36. doi: 10.1097/SHK.0000000000000561.
    1. Haeusler KG, et al. Immune responses after acute ischemic stroke or myocardial infarction. Int J Cardiol. 2012;155(3):372–377. doi: 10.1016/j.ijcard.2010.10.053.
    1. Schefold JC, et al. Heart failure and kidney dysfunction: epidemiology, mechanisms and management. Nat Rev Nephrol. 2016;12(10):610–623. doi: 10.1038/nrneph.2016.113.
    1. von Haehling S, et al. Leukocyte redistribution: effects of beta blockers in patients with chronic heart failure. PLoS One. 2009;4(7):e6411. doi: 10.1371/journal.pone.0006411.
    1. Monneret G, et al. Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med. 2006;32(8):1175–1183. doi: 10.1007/s00134-006-0204-8.
    1. Franke A, et al. Delayed recovery of human leukocyte antigen-DR expression after cardiac surgery with early non-lethal postoperative complications: only an epiphenomenon? Interact Cardiovasc Thorac Surg. 2008;7(2):207–211. doi: 10.1510/icvts.2007.158899.
    1. Allen ML, et al. Early postoperative monocyte deactivation predicts systemic inflammation and prolonged stay in pediatric cardiac intensive care. Crit Care Med. 2002;30(5):1140–5. doi: 10.1097/00003246-200205000-00031.
    1. Bianchi ME, Manfredi AA. High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity. Immunol Rev. 2007;220:35–46. doi: 10.1111/j.1600-065X.2007.00574.x.
    1. Zhang J, et al. Vagal modulation of high mobility group box-1 protein mediates electroacupuncture-induced cardioprotection in ischemia-reperfusion injury. Sci Rep. 2015;5:15503. doi: 10.1038/srep15503.
    1. Wild CA, et al. HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. Int Immunol. 2012;24(8):485–494. doi: 10.1093/intimm/dxs051.
    1. Zhang Y, et al. The potential effect and mechanism of high-mobility group box 1 protein on regulatory T cell-mediated immunosuppression. J Interf Cytokine Res. 2011;31(2):249–257. doi: 10.1089/jir.2010.0019.
    1. Shevach EM, et al. Control of T-cell activation by CD4+ CD25+ suppressor T cells. Immunol Rev. 2001;182:58–67. doi: 10.1034/j.1600-065X.2001.1820104.x.
    1. Allan SE, et al. CD4+ T-regulatory cells: toward therapy for human diseases. Immunol Rev. 2008;223:391–421. doi: 10.1111/j.1600-065X.2008.00634.x.
    1. Yeh CH, et al. Induced interleukin-19 contributes to cell-mediated immunosuppression in patients undergoing coronary artery bypass grafting with cardiopulmonary bypass. Ann Thorac Surg. 2011;92(4):1252–1259. doi: 10.1016/j.athoracsur.2011.04.061.
    1. Horan Teresa C., Andrus Mary, Dudeck Margaret A. CDC/NHSN surveillance definition of health care–associated infection and criteria for specific types of infections in the acute care setting. American Journal of Infection Control. 2008;36(5):309–332. doi: 10.1016/j.ajic.2008.03.002.

Source: PubMed

3
購読する