Transcutaneous auricular vagus nerve stimulation protects endotoxemic rat from lipopolysaccharide-induced inflammation

Yu Xue Zhao, Wei He, Xiang Hong Jing, Jun Ling Liu, Pei Jing Rong, Hui Ben, Kun Liu, Bing Zhu, Yu Xue Zhao, Wei He, Xiang Hong Jing, Jun Ling Liu, Pei Jing Rong, Hui Ben, Kun Liu, Bing Zhu

Abstract

Background. Transcutaneous auricular vagus nerve stimulation (ta-VNS) could evoke parasympathetic activities via activating the brainstem autonomic nuclei, similar to the effects that are produced after vagus nerve stimulation (VNS). VNS modulates immune function through activating the cholinergic anti-inflammatory pathway. Methods. VNS, ta-VNS, or transcutaneous electrical acupoint stimulation (TEAS) on ST36 was performed to modulate the inflammatory response. The concentration of serum proinflammatory cytokines and tissue NF-kappa B p65 (NF-κB p65) were detected in endotoxaemia affected anesthetized rats. Results. Similar to the effect of VNS, ta-VNS suppressed the serum proinflammatory cytokines levels, such as tumour necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) as well as NF-kappa B p65 expressions of lung tissues. ST36 stimulation also decreases LPS-induced high TNF-α level and NF-κB signal, but it did not restrain proinflammatory cytokine IL-1β and IL-6. Neither ta-VNS nor ST36 stimulation could suppress LPS-induced TNF-α and NF-κB after vagotomy or with α7nAChR antagonist injection. Conclusions. The present paper demonstrated that ta-VNS could be utilized to suppress LPS-induced inflammatory responses via α7nAChR-mediated cholinergic anti-inflammatory pathway.

Figures

Figure 1
Figure 1
The time flow chart indicates the precise time for various operations in the present study, from the time point of anesthetic injection to the time point for sampling. (a) Twenty minutes after anesthesia, rats were injected intravenously with LPS or NS. One and a half hour after modeling, treatment (ta-VNS, VNS, or TEAS on ST36) was performed for twenty minutes. Two hours after LPS injection, rats were killed, and samples were collected. (b) Ten minutes after anesthesia, administration of α-BGT or vagotomy was performed. The rest of the operations were the same with the time flow in (a).
Figure 2
Figure 2
Vagus nerve stimulation (VNS) or transcutaneous auricular vagus nerve stimulation (ta-VNS) attenuates the LPS-induced serum cytokine (TNF-α, IL-1β, and IL-6) response. TEAS on ST36 inhibited TNF-α level significantly. Serum TNF-α (a), IL-1β (b), and IL-6 (c) contents were measured by ELISA. The columns represent mean ± SEM for 12 animals in each group. ∆∆P < 0.01 versus the normal saline (NS) group; *P < 0.05 versus LPS group (LPS); **P < 0.01 versus LPS group (LPS); #P < 0.05 versus LPS+VNS group; ##P < 0.01 versus LPS+VNS group.
Figure 3
Figure 3
ta-VNS or TEAS on ST36 with α-bungarotoxin (α-BGT) administration fails to inhibit the LPS-induced serum TNF-α response. Serum TNF-α concentrations were measured by ELISA. Data are expressed as mean ± SEM (n = 12 per group). ∆∆P < 0.01 versus the normal saline (NS) group.
Figure 4
Figure 4
ta-VNS or ST36 stimulation with bilateral cervical vagotomy (VGX) fails to inhibit the LPS-induced serum TNF-α response. TNF-α amounts were measured by ELISA. Data are expressed as mean ± SEM (n = 12 per group). ∆∆P < 0.01 versus the normal saline (NS) group.
Figure 5
Figure 5
VNS or ta-VNS suppresses LPS-induced NF-κB expression; ST36 stimulation did not affect NF-κB in endotoxemia animals significantly. ta-VNS did not significantly affect pulmonary NF-κB expression with normal saline administration. NF-κB expressions were measured by western blot technique. Data are shown by mean ± SEM (n = 12 per group). ∆∆P < 0.01 versus normal saline (NS) group; **P < 0.01 versus LPS group (LPS); ##P < 0.01 versus LPS+VNS group; ▲P < 0.05 versus LPS+ ta-VNS.
Figure 6
Figure 6
Immunohistochemical staining with anti-NF-κB antibodies reveals significant decrease in LPS-induced NF-κB immunoreactivity evoked by interventions as of VNS, ta-VNS, and TEAS on ST36. Data are expressed as mean ± SEM (n = 12 per group). ∆∆P < 0.01 versus the normal saline (NS) group; **P < 0.01 versus LPS group (LPS); #P < 0.05 versus LPS+VNS group. Original magnification: ×400.
Figure 7
Figure 7
ta-VNS or ST36 stimulation with bilateral cervical vagotomy (VGX) fails to inhibit the LPS-induced overexpression of NF-κB. NF-κB distribution was measured by immunohistochemical staining. Data are shown as mean ± SEM (n = 12 per group). ∆∆P < 0.01 versus the normal saline (NS) group. Original magnification: ×400.
Figure 8
Figure 8
The anti-inflammatory mechanisms of the three interventions used in the present study might be as follows: (1) VNS directly activates the cholinergic anti-inflammatory pathway via stimulating efferent vagus nerve. (2) ta-VNS evoked the activity of the auricular branch of vagus nerve (ABVN). The activated signals ascending within afferent vagus nerve are transmitted to the nucleus tractus solitarius. The integrated output is carried by the efferent vagus nerve to inhibit inflammatory responses. (3) TEAS on ST36 activates the somatic fiber endings of the skin around ST36 point, sending signals to the spinal cord via somatic sensory nerve fibers. The nerve impulses were relayed and integrated by NTS by the secondary order neurons in the spinal cord, and the cholinergic anti-inflammatory pathways are activated by the increased efferent vagal output.

References

    1. Tracey KJ. The inflammatory reflex. Nature. 2002;420(6917):853–859.
    1. Tracey KJ, Fong Y, Hesse DG, et al. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature. 1987;330(6149):662–664.
    1. Tracey KJ, Beutler B, Lowry SF. Shock and tissue injury induced by recombinant human cachectin. Science. 1986;234(4775):470–474.
    1. Pavlov VA, Tracey KJ. The cholinergic anti-inflammatory pathway. Brain, Behavior, and Immunity. 2005;19(6):493–499.
    1. Huston JM, Gallowitsch-Puerta M, Ochani M, et al. Transcutaneous vagus nerve stimulation reduces serum high mobility group box 1 levels and improves survival in murine sepsis. Critical Care Medicine. 2007;35(12):2762–2768.
    1. Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405(6785):458–462.
    1. Huang J, Wang Y, Jiang D, Zhou J, Huang X. The sympathetic-vagal balance against endotoxemia. Journal of Neural Transmission. 2010;117(6):729–735.
    1. van Westerloo DJ, Giebelen IAJ, Meijers JCM, et al. Vagus nerve stimulation inhibits activation of coagulation and fibrinolysis during endotoxemia in rats. Journal of Thrombosis and Haemostasis. 2006;4(9):1997–2002.
    1. Bernik TR, Friedman SG, Ochani M, et al. Pharmacological stimulation of the cholinergic antiinflammatory pathway. Journal of Experimental Medicine. 2002;195(6):781–788.
    1. Guarini S, Altavilla D, Cainazzo MM, et al. Efferent vagal fibre stimulation blunts nuclear factor-κB activation and protects against hypovolemic hemorrhagic shock. Circulation. 2003;107(8):1189–1194.
    1. Li M, Zheng C, Sato T, Kawada T, Sugimachi M, Sunagawa K. Vagal nerve stimulation markedly improves long-term survival after chronic heart failure in rats. Circulation. 2004;109(1):120–124.
    1. Mioni C, Bazzani C, Giuliani D, et al. Activation of an efferent cholinergic pathway produces strong protection against myocardial ischemia/reperfusion injury in rats. Critical Care Medicine. 2005;33(11):2621–2628.
    1. Huang ST, Chen GY, Lo HM, Lin JG, Lee YS, Kuo CD. Increase in the vagal modulation by acupuncture at Neiguan point in the healthy subjects. American Journal of Chinese Medicine. 2005;33(1):157–164.
    1. Li Z, Jiao K, Chen M, Wang C. Effect of magnitopuncture on sympathetic and parasympathetic nerve activities in healthy driver—assessment by power spectrum analysis of heart rate variability. European Journal of Applied Physiology. 2003;88(4-5):404–410.
    1. Wu JH, Chen HY, Chang YJ, et al. Study of autonomic nervous activity of night shift workers treated with laser acupuncture. Photomedicine and Laser Surgery. 2009;27(2):273–279.
    1. Hsu CC, Weng CS, Liu TS, Tsai YS, Chang YH. Effects of electrical acupuncture on acupoint BL15 evaluated in terms of heart rate variability, pulse rate variability and skin conductance response. American Journal of Chinese Medicine. 2006;34(1):23–36.
    1. Imai K, Kitakoji H. Comparison of transient heart rate reduction associated with acupuncture stimulation in supine and sitting subjects. Acupuncture in Medicine. 2003;21(4):133–137.
    1. He W, Rong PJ, Li L, Ben H, Zhu B, Litscher G. Auricular acupuncture may suppress epileptic seizures via activating the parasympathetic nervous system: a hypothesis based on innovative methods. Evidence-Based Complementary and Alternative Medicine. 2012;2012:5 pages.615476
    1. Bertók L. Endotoxins and endocrine system. Domestic Animal Endocrinology. 1998;15(5):305–308.
    1. Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation. Nature. 2003;421(6921):384–388.
    1. Deng W, Wang X, Xiao J, et al. Loss of regulator of G protein signaling 5 exacerbates obesity, hepatic steatosis, inflammation and insulin resistance. PLoS ONE. 2012;7(1)e30256
    1. D’Cruz OJ, Uckun FM. Preclinical evaluation of a dual-acting microbicidal prodrug WHI-07 in combination with vanadocene dithiocarbamate in the female reproductive tract of rabbit, pig, and cat. Toxicologic Pathology. 2007;35(7):910–927.
    1. Huston JM, Ochani M, Rosas-Ballina M, et al. Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. Journal of Experimental Medicine. 2006;203(7):1623–1628.
    1. Bernik TR, Friedman SG, Ochani M, et al. Cholinergic antiinflammatory pathway inhibition of tumor necrosis factor during ischemia reperfusion. Journal of Vascular Surgery. 2002;36(6):1231–1236.
    1. Altavilla D, Guarini S, Bitto A, et al. Activation of the cholinergic anti-inflammatory pathway reduces NF-κB activation, blunts TNF-α production, and protects against splanchnic artery occlusion shock. Shock. 2006;25(5):500–506.
    1. Bauerle PA, Baltmore D. NF-κB: ten years after. Cell. 1996;87(1):13–20.
    1. Schmitz ML, Bauerle PA. Multi step activation of NF-κB/rel transcriptional factors. Immunobiology. 1995;193(2–4):116–127.
    1. Schreck R, Rieber P, Baeuerle PA. Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-κB transcription factor and HIV-1. EMBO Journal. 1991;10(8):2247–2258.
    1. Squadrito F, Altavilla D, Deodato B, et al. Early activation of transcription factor NF-κB in splanchnic artery occlusion shock. In: Faist E, editor. Proceedings of the 5th World Congress on Trauma Shock Inflammation and Sepsis Pathophysiology Immune Consequences and Therapy; March 2000; Munich, Germany. Monduzzi; pp. 389–394.
    1. O’Mahony C. Loss of vagal anti-inflammatory effect: in vivo visualization and adoptive transfer. American Journal of Physiology. 2009;297(4):R1118–R1126.
    1. Song XM, Li JG, Wang YL, et al. The protective effect of the cholinergic anti-inflammatory pathway against septic shock in rats. Shock. 2008;30(4):468–472.
    1. Imai K, Ariga H, Chen C, Mantyh C, Pappas TN, Takahashi T. Effects of electroacupuncture on gastric motility and heart rate variability in conscious rats. Autonomic Neuroscience: Basic and Clinical. 2008;138(1-2):91–98.
    1. Ouyang H, Yin J, Wang Z, Pasricha PJ, Chen JDZ. Electroacupuncture accelerates gastric emptying in association with changes in vagal activity. American Journal of Physiology. 2002;282(2):G390–G396.
    1. Wang Q, Wang F, Li X, et al. Electroacupuncture pretreatment attenuates cerebral ischemic injury through α7 nicotinic acetylcholine receptor-mediated inhibition of high-mobility group box 1 release in rats. Journal of Neuroinflammation. 2012;9, article 24
    1. Gao XY, Li YH, Liu K, et al. Acupuncture-like stimulation at auricular point Heart evokes cardiovascular inhibition via activating the cardiac-related neurons in the nucleus tractus solitarius. Brain Research. 2011;1397:19–27.
    1. Gao XY, Zhang SP, Zhu B, Zhang HQ. Investigation of specificity of auricular acupuncture points in regulation of autonomic function in anesthetized rats. Autonomic Neuroscience. 2008;138(1-2):50–56.
    1. Tekdemir I, Asian A, Elhan A. A clinico-anatomic study of the auricular branch of the vagus nerve and Arnold’s ear-cough reflex. Surgical and Radiologic Anatomy. 1998;20(4):253–257.
    1. Gupta D, Verma S, Vishwakarma SK. Anatomic basis of Arnold’s ear-cough reflex. Surgical and Radiologic Anatomy. 1986;8(4):217–220.
    1. Engel D. The gastroauricular phenomenon and related vagus reflexes. Archiv fur Psychiatrie und Nervenkrankheiten. 1979;227(3):271–277.
    1. Pavlov VA, Wang H, Czura CJ, Friedman SG, Tracey KJ. The cholinergic anti-inflammatory pathway: a missing link in neuroimmunomodulation. Molecular Medicine. 2003;9(5–8):125–134.

Source: PubMed

3
Abonnieren