Somatotopic Organization and Intensity Dependence in Driving Distinct NPY-Expressing Sympathetic Pathways by Electroacupuncture

Shenbin Liu, Zhi-Fu Wang, Yang-Shuai Su, Russell S Ray, Xiang-Hong Jing, Yan-Qing Wang, Qiufu Ma, Shenbin Liu, Zhi-Fu Wang, Yang-Shuai Su, Russell S Ray, Xiang-Hong Jing, Yan-Qing Wang, Qiufu Ma

Abstract

The neuroanatomical basis behind acupuncture practice is still poorly understood. Here, we used intersectional genetic strategy to ablate NPY+ noradrenergic neurons and/or adrenal chromaffin cells. Using endotoxin-induced systemic inflammation as a model, we found that electroacupuncture stimulation (ES) drives sympathetic pathways in somatotopy- and intensity-dependent manners. Low-intensity ES at hindlimb regions drives the vagal-adrenal axis, producing anti-inflammatory effects that depend on NPY+ adrenal chromaffin cells. High-intensity ES at the abdomen activates NPY+ splenic noradrenergic neurons via the spinal-sympathetic axis; these neurons engage incoherent feedforward regulatory loops via activation of distinct adrenergic receptors (ARs), and their ES-evoked activation produces either anti- or pro-inflammatory effects due to disease-state-dependent changes in AR profiles. The revelation of somatotopic organization and intensity dependency in driving distinct autonomic pathways could form a road map for optimizing stimulation parameters to improve both efficacy and safety in using acupuncture as a therapeutic modality.

Keywords: ST25 and ST36 acupoints; adrenal chromaffin cells; adrenergic receptors; electroacupuncture; neuropeptide Y; somatosensory autonomic pathways; spleen; sympathetic neurons; systemic inflammation; vagal-adrenal axis.

Conflict of interest statement

Declaration of Interests The authors declare no competing interests.

Copyright © 2020 Elsevier Inc. All rights reserved.

Figures

Figure 1.. Intersectional marking of NPY Cre…
Figure 1.. Intersectional marking of NPYCre-expressing sympathetic cells.
(A-C) Colocalization of tdTomato with NPY, DBH orVAChT mRNA or the TH protein in sections through sympathetic ganglia (“g.”) and adrenal glands of adultNPYDBH-tdTomato mice. (D) Schematic illustration of splenic subcapsular regions. RP: red pulp, MZ: marginal zone, WP: white pulp. (E) Splenic innervations by NPYDBH-tdTomato+ fibers. Arrows and arrowheads in WPs: fibers along the arterioles and the periarteriolar lymphatic sheath, respectively. “*”: fibers innervating the trabeculae. (F) A representative image showing tdTomato overlapping with TH in a WP. n = 5–6 mice for all groups. Scale bars, 100 μm.
Figure 2.. Intersectional ablation of peripheral and…
Figure 2.. Intersectional ablation of peripheral and adrenal NPYDBH cells.
(A) Intraperitoneal injection (i.p.) of PEGyDT to create NPYDBH (peri.)-Abl [“(peri.)-Abl”] mice. “WT”: wild type littermates. All NPYCre cells were labeled by tdTomato from an unshown reporter allele. (B) Ablation of NPYCre-tdTomato+ cells in suprarenal ganglia (t8 = 13.1, ***P < 0.001) and adrenal glands (t8 = 14.96, ***P < 0.001). (C) Intra-adrenal medulla injection of PEGyDT to create NPYDBH (adrenal)-Abl mice [“(adrenal)-Abl”]. (D) Ablation of NPYCre-tdTomato+ cells in adrenal glands, but not in suprarenal ganglia compared with WT mice (adrenal,t8 = 11.68, ***P < 0.001; suprarenal, t8 = 0.659, NS, no significant,P = 0.528). n = 5 mice for all groups. Two-side student’s unpairedt-test in B and D. Data are shown as mean ± SEM. Scale bars, 100 μm.
Figure 3.. Ablation of NPY DBH sympathetic…
Figure 3.. Ablation of NPYDBH sympathetic cells caused an increase in LPS-induced TNF-α production.
(A-B) LPS induced a larger increase of TNF-α levels in NPYDBH (peri.)-Abl mice compared with WT mice in serum and in spleen. Two-way ANOVA plus post-hoc Tukey test. Serum:F1, 20 = 13.457, P = 0.002; spleen: F1, 16 = 13.174, P < 0.001. ***P < 0.001; NS, not significant,P = 0.913 in A and 0.892 in B. (C) TNF-α immunostaining images through splenic red pulps (“RP”) and white pulps (“WP”). (D) TNF-α expression (by immunostaining) in splenic macrophages with indicated markers in LPS-treated wild type (WT) versus NPYDBH (peri.)-Abl mice (two-side student’s unpaired t test;t8 = 1.528 for F4/80, NS, not significant,P = 0.165; t8 = 13.36 for CD169 and 12.22 for CD68, ***P < 0.001). Arrowheads indicate co-localized cells. N = 5 mice for all groups. Data are shown as mean ± SEM. Scale bars, 100 μm.
Figure 4.. Requirement of NPY DBH cells…
Figure 4.. Requirement of NPYDBH cells for preST25 ES-evoked anti-inflammatory effects.
(A) Schematics showing the abdominal ST25 acupoint. (B) Modulation of LPS-induced TNF-α by ST25 ES (F3, 20 = 43.772, P < 0.001; ***P < 0.001; ###P< 0.001; NS, P = 0.406). (C) c-Fos induction by ST25 ES in ChAT+ neurons in the spinal intermediolateral nuclei (“IML”). F2, 14 = 86.02, P < 0.001; ***P< 0.001; NS, P = 0.519. (D) c-Fos induction in suprarenal ganglia (“g.”) by 3.0 mA compared with sham 0 mA ES (two-side student’s unpairedt-test, t8 = 4.97, **P = 0.001). (E-F) Loss of 3.0 mA preST25 ES-evoked reduction of TNF-α in NPYDBH(peri.)-Abl mice compared with wild type (WT) mice, both in serum (F1, 20 = 19.113, P < 0.001;***P < 0.001; NS, P = 0.715) and in spleen (F1, 16 = 13.147, P = 0.012;***P < 0.001; NS, P = 0.783). (G, H) 3.0 mA preST25 ES (compared with 0 mA ES) reduced LPS-induced fatality in WT mice but not in (peri.)-Abl mice (n = 24–25 mice per group; log-rank test, **P = 0.007; NS, P = 0.418). (I) Loss of 3.0 mA preST25 ES-induced splenic noradrenaline release in (peri.)-Abl mice compared with WT mice (F1, 16 = 79.677, P < 0.001; *P < 0.05, ***P < 0.001; NS, P = 0.915). (J) Splenic TNF-α immunostaining in LPS-treated mice receiving 3.0 mA preST25 ES. Increased signals in NPYDBH (peri.)-Abl mice compared with WT mice (two-side student’s unpairedt-test, t8 = 6.856, ***P < 0.001). (K) A blockage of 3.0 mA preST25 ES-evoked reduction in serum TNF-α in LPS-treated C57BL/6 mice by ICI 118,551 (a β2-AR antagonist) (F1, 16 = 61.757, P< 0.001; ***P < 0.001; **P = 0.006; NS, P = 0.254). (L) preST25 ES evokes NPYDBH cell- and β2 AR-dependent anti-inflammatory pathways, via segmental and/or spinal-supra-spinal pathways (“dashed”, to be determined). “Sym. g.”: sympathetic ganglia. n = 5–6 mice for all mouse groups (except G and H). One-way (B, C) or two-way (E, F, I, K) ANOVA plus post hoc Tukey test. NS, not significant. Data are shown as mean ± SEM. Scale bars, 100 μm.
Figure 5.. Activation of NPY DBH sympathetic…
Figure 5.. Activation of NPYDBH sympathetic cells switched to promote inflammation following LPS pre-exposure.
(A) 3.0 mA postST25 ES, performed 1.5 hours after LPS exposure, increased lethality in LPS-treated C57BL/6J mice (log-rank test; 0 mA, n = 23; 3.0 mA, n = 22; **P = 0.006). (B) Loss of 3.0 mA postST25 ES-evoked increase in serum TNF-α in NPYDBH (peri.)-Abl mice compared with wild type (WT) mice (F1, 20 = 5.602, P < 0.05; ***P < 0.001; NS, P = 0.271). (C) Splenectomy, compared with sham surgery, blocked postST25 ES-evoked increase in serum TNF-α (F1, 16 = 39.871,P < 0.001; ***P < 0.001; NS,P = 0.125). (D) Heat map of quantitative RT-PCR results, showing changes in various adrenergic receptor (AR) expression 1 hour after LPS injection (two-side student’s unpaired t-test; β1:t8 = −0.049, P = 0.962; β2: t8 = −3.840, **P= 0.015; β3: t8 = −0.671,P = 0.521; α1a: t8 = −3.764, **P = 0.016; α1b:t8 = −0.876, P = 0.406; a1d: t8 = −0.270, P = 0.794; α2a: t8 = −2.957, *P= 0.018; α2c: t8 = −6.328, ***P < 0.001; α2b: Mann-Whitney Rank Sum test, *P = 0.016). (E) α2-AR antagonist Yohimbine blocked postST25 ES-evoked increase of TNF-α (F1, 20 = 59.709,P < 0.001;***P < 0.001, *P = 0.016). (F) Co-treatment with Yohimbine allowed 3.0 mA postST25 ES to promote survival for LPS-treated C57BL/6J mice (log-rank test, *P = 0.045). (G) postST25 ES drive NPYDBH cell-dependent pro-inflammatory pathways, via activation of α2 ARs. (H) Schematic description of incoherent feed-forward loops. ES-evoked noradrenaline release from NPYDBH cells modulates LPS-induced TNF-α production (via activation of TLR4) in splenic cells, via activation of β2 and a2 ARs that produce the negative and positive modulatory legs, respectively. n = 22–24 mice (A, F) and 5–6 mice for all other groups. NS: not significant. Data are shown as mean ± SEM.
Figure 6.. 0.5 mA ES at ST36…
Figure 6.. 0.5 mA ES at ST36 attenuated ongoing systemic inflammation.
(A) Schematic showing the hindlimb ST36 acupoint. (B) ES induced c-Fos in ChAT+ neurons in the dorsal motor nuclei of the vagus (“DMV”). Arrowheads: co-expression. Two-side student’s unpaired t-tests,t9 = 7.09, ***P < 0.001. (C) 0.5 mA ST36 ES increased catecholamine release (“NA” for noradrenaline, “A” for adrenaline, “DA” for dopamine) in WT, but not in NPYDBH (adrenal)-Abl mice [F1, 20 = 40.560 (“NA”), 69.780 (“A”), 60.912 (“DA”), P < 0.001; ***P < 0.001; NS, P = 0.483 (“NA”), 0.734 (“A”), and 0.757 (“DA”)]. (D) 0.5 mA preST36 ES reduced LPS-induced TNF-α in WT, but not in (adrenal)-Abl mice (F1,20 = 20.457, P < 0.001; ***P < 0.001; NS, left, P = 0.064, right, P =0.893). (E) 0.5 mA preST36 ES reduced LPS-induced TNF-α in mice with sham surgery, but not with subdiaphragmatic vagotomy (“sVX”) (F1, 20 = 72.188, P < 0.001;**P < 0.01; NS, P = 0.628). (F) 0.5 mA preST36 ES promoted survival compared with 0 mA ES (0 mA, n = 28; 0.5 mA, n = 33, log-rank test, *P = 0.019). (G) 0.5 mA postST36 ES reduced LPS-induced TNF-α in WT, but not in (adrenal)-Abl mice (F1, 20 = 23.155, P < 0.001; ***P < 0.001; NS, left, P = 0.147, right, P =0.550). (H) Loss of 0.5 mA postST36 ES-evoked TNF-α reduction by subdiaphragmatic vagotomy (“sVX”) compared with sham surgery (F1, 20 = 81.237, P < 0.001; ***P < 0.001; *P = 0.025; NS,P = 0.315). (I) 0.5 mA postST36 ES improved survival compared with 0 mA ES (0 mA, n = 23; 0.5 mA, n = 28, log-rank test, **P = 0.001). (J) ES drives two autonomic pathways and modulates LPS-induced inflammation in somatotopy-, intensity-, and disease state-dependent manners. Sym. g.: sympathetic ganglia. IML: spinal intermediolateral nuclei. Adrenal g.: adrenal gland. Noradren.: Noradrenergic. Chroma.: Chromaffin. n = 5–6 mice for all groups (except F and I). Two-way ANOVA pluspost hoc Tukey’s test (C, D, E, G, and H). NS, not significant. Data are shown as mean ± SEM. Scale bars, 100 μm.

References

    1. Abe C, Inoue T, Inglis MA, Viar KE, Huang L, Ye H, Rosin DL, Stornetta RL, Okusa MD, and Guyenet PG (2017). C1 neurons mediate a stress-induced anti-inflammatory reflex in mice. Nat Neurosci 20, 700–707.
    1. Agac D, Estrada LD, Maples R, Hooper LV, and Farrar JD (2018). The beta2-adrenergic receptor controls inflammation by driving rapid IL-10 secretion. Brain Behav Immun 74, 176–185.
    1. Alon U (2007). Network motifs: theory and experimental approaches. Nat Rev Genet 8, 450–461.
    1. Archer T, and Fredriksson A (2000). Effects of clonidine and alpha-adrenoceptor antagonists on motor activity in DSP4-treated mice I: dose-, time- and parameter-dependency. Neurotoxicity research 1, 235–247.
    1. Armbruster BN, Li X, Pausch MH, Herlitze S, and Roth BL (2007). Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A 104, 5163–5168.
    1. Beutler B (2000). Endotoxin, toll-like receptor 4, and the afferent limb of innate immunity. Curr Opin Microbiol 3, 23–28.
    1. Borges da Silva H, Fonseca R, Pereira RM, Cassado Ados A, Alvarez JM, and D’Imperio Lima MR (2015). Splenic Macrophage Subsets and Their Function during Blood-Borne Infections. Front Immunol 6, 480.
    1. Borovikova L, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, Eaton JW, and Tracey KJ (2000). Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 405, 458–462.
    1. Bourane S, Grossmann KS, Britz O, Dalet A, Del Barrio MG, Stam FJ, Garcia-Campmany L, Koch S, and Goulding M (2015). Identification of a spinal circuit for light touch and fine motor control. Cell 160, 503–515.
    1. Bratton BO, Martelli D, McKinley MJ, Trevaks D, Anderson CR, and McAllen RM (2012). Neural regulation of inflammation: no neural connection from the vagus to splenic sympathetic neurons. Exp Physiol 97, 1180–1185.
    1. Bronte V, and Pittet MJ (2013). The spleen in local and systemic regulation of immunity. Immunity 39, 806–818.
    1. Deuchars SA, and Lall VK (2015). Sympathetic preganglionic neurons: properties and inputs. Compr Physiol 5, 829–869.
    1. Duan B, Cheng L, Bourane S, Britz O, Padilla C, Garcia-Campmany L, Krashes M, Knowlton W, Velasquez T, Ren X, et al. (2014). Identification of spinal circuits transmitting and gating mechanical pain. Cell 159, 1417–1432.
    1. Fernandes ES, Liang L, Smillie SJ, Kaiser F, Purcell R, Rivett DW, Alam S, Howat S, Collins H, Thompson SJ, et al. (2012). TRPV1 deletion enhances local inflammation and accelerates the onset of systemic inflammatory response syndrome. J Immunol 188, 5741–5751.
    1. Flierl MA, Rittirsch D, Nadeau BA, Chen AJ, Sarma JV, Zetoune FS, McGuire SR, List RP, Day DE, Hoesel LM, et al. (2007). Phagocyte-derived catecholamines enhance acute inflammatory injury. Nature 449, 721–725.
    1. Furlan A, La Manno G, Lubke M, Haring M, Abdo H, Hochgerner H, Kupari J, Usoskin D, Airaksinen MS, Oliver G, et al. (2016). Visceral motor neuron diversity delineates a cellular basis for nipple- and pilo-erection muscle control. Nat Neurosci 19, 1331–1340.
    1. Gao X, Zhao Y, Su Y, Liu K, Yu X, Cui C, Yang Z, Shi H, Jing X, and Zhu B (2016). beta1/2 or M2/3 Receptors Are Required for Different Gastrointestinal Motility Responses Induced by Acupuncture at Heterotopic or Homotopic Acupoints. PLoS One 11, e0168200.
    1. Gerfen CR, Paletzki R, and Heintz N (2013). GENSAT BAC cre-recombinase driver lines to study the functional organization of cerebral cortical and basal ganglia circuits. Neuron 80, 1368–1383.
    1. Gu G, Zhang Z, Wang G, Han F, Han L, Wang K, Liu J, and Li W (2011). Effects of electroacupuncture pretreatment on inflammatory response and acute kidney injury in endotoxaemic rats. J Int Med Res 39, 1783–1797.
    1. Huang CL, Tsai PS, Wang TY, Yan LP, Xu HZ, and Huang CJ (2007). Acupuncture stimulation of ST36 (Zusanli) attenuates acute renal but not hepatic injury in lipopolysaccharide-stimulated rats. Anesth Analg 104, 646–654.
    1. Huang JL, Zhang YL, Wang CC, Zhou JR, Ma Q, Wang X, Shen XH, and Jiang CL (2012). Enhanced phosphorylation of MAPKs by NE promotes TNF-alpha production by macrophage through alpha adrenergic receptor. Inflammation 35, 527–534.
    1. Huang S, Ziegler CGK, Austin J, Mannoun N, Vukovic M, Ordovas-Montanes J, Shalek AK, and von Andrian UH (2019). Sensory Neurons Innervate Peripheral Lymph Nodes and Locally Regulate Gene Expression in Postsynaptic Endothelium, Stromal Cells, and Innate Leukocytes. bioRxiv. doi: 10.1101/833509
    1. Hunt SP, Pini A, and Evan G (1987). Induction of c-fos-like protein in spinal cord neurons following sensory stimulation. Nature 328, 632–634.
    1. Huston JM, Ochani M, Rosas-Ballina M, Liao H, Ochani K, Pavlov VA, Gallowitsch-Puerta M, Ashok M, Czura CJ, Foxwell B, et al. (2006). Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J Exp Med 203, 1623–1628.
    1. Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ, Kurosawa S, Stepien D, Valentine C, and Remick DG (2013). Sepsis: multiple abnormalities, heterogeneous responses, and evolving understanding. Physiological reviews 93, 1247–1288.
    1. Janig W, and Habler HJ (2000). Specificity in the organization of the autonomic nervous system: a basis for precise neural regulation of homeostatic and protective body functions. Prog Brain Res 122, 351–367.
    1. Ji RR, Baba H, Brenner GJ, and Woolf CJ (1999). Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci 2, 1114–1119.
    1. John L, Nastaran H & Dean M Electrically evoked compound action potential recording in peripheral nerves. Bioelectronics in medicine 2018. 1(1), 71–83.
    1. Johnson EM Jr., and O’Brien F (1976). Evaluation of the permanent sympathectomy produced by the administration of guanethidine to adult rats. J Pharmacol Exp Ther 196, 53–61.
    1. Kadl A, Pontiller J, Exner M, and Leitinger N (2007). Single bolus injection of bilirubin improves the clinical outcome in a mouse model of endotoxemia. Shock 28, 582–588.
    1. Kolibal-Pegher S, Edwards DJ, Meyers-Schoy SA, and Vollmer RR (1994). Adrenal medullary adaptations and cardiovascular regulation after 6-hydroxydopamine treatment in rats. J Auton Nerv Syst 48, 113–120.
    1. Komegae EN, Farmer DGS, Brooks VL, McKinley MJ, McAllen RM, and Martelli D (2018). Vagal afferent activation suppresses systemic inflammation via the splanchnic antiinflammatory pathway. Brain Behav Immun 73, 441–449.
    1. Li YQ, Zhu B, Rong PJ, Ben H, and Li YH (2007). Neural mechanism of acupuncture-modulated gastric motility. World J Gastroenterol 13, 709–716.
    1. Lim HD, Kim MH, Lee CY, and Namgung U (2016). Anti-Inflammatory Effects of Acupuncture Stimulation via the Vagus Nerve. PLoS One 11, e0151882.
    1. Lima D (2008). Ascending Pathways: Anatomy and Physiology. The Senses: A Comprehensive Reference 5, 477–526.
    1. Liu Y, Abdel Samad O, Duan B, Zhang L, Tong Q, Ji RR, Lowell B, and Ma Q (2010). VGLUT2-dependent glutamate release from peripheral nociceptors is required to sense pain and suppress itch. Neuron 68, 543–556.
    1. Liu Y, Yang FC, Okuda T, Dong X, Zylka MJ, Chen CL, Anderson DJ, Kuner R, and Ma Q (2008). Mechanisms of compartmentalized expression of Mrg class G-protein-coupled sensory receptors. J Neurosci 28, 125–132.
    1. Longhurst JC (2010). Defining meridians: a modern basis of understanding. J Acupunct Meridian Stud 3, 67–74.
    1. Martelli D, Farmer DG, and Yao ST (2016). The splanchnic anti-inflammatory pathway: could it be the efferent arm of the inflammatory reflex? Exp Physiol 101, 1245–1252.
    1. Martelli D, Farmer DGS, McKinley MJ, Yao ST, and McAllen RM (2019). Antiinflammatory reflex action of splanchnic sympathetic nerves is distributed across abdominal organs. Am J Physiol Regul Integr Comp Physiol 316, R235–R242.
    1. Martelli D, Yao ST, McKinley MJ, and McAllen RM (2014). Reflex control of inflammation by sympathetic nerves, not the vagus. J Physiol 592, 1677–1686.
    1. Molinoff PB (1984). Alpha- and beta-adrenergic receptor subtypes properties, distribution and regulation. Drugs 28 Suppl 2, 1–15.
    1. Nielsen GD (1977). Guanethidine induced sympathectomy in the adult rat. II. Functional effects following chronic administration. Acta Pharmacol Toxicol (Copenh) 41, 209–217.
    1. O’Donnell SR, and Wanstall JC (1980). Evidence that ICI 118, 551 is a potent, highly Beta 2-selective adrenoceptor antagonist and can be used to characterize Beta-adrenoceptor populations in tissues. Life Sci 27, 671–677.
    1. Pavlov VA, and Tracey KJ (2017). Neural regulation of immunity: molecular mechanisms and clinical translation. Nat Neurosci 20, 156–166.
    1. Pereira MM, Mahu I, Seixas E, Martinez-Sanchez N, Kubasova N, Pirzgalska RM, Cohen P, Dietrich MO, Lopez M, Bernardes GJ, et al. (2017). A brain-sparing diphtheria toxin for chemical genetic ablation of peripheral cell lineages. Nat Commun 8, 14967.
    1. Pinho-Ribeiro FA, Baddal B, Haarsma R, O’Seaghdha M, Yang NJ, Blake KJ, Portley M, Verri WA, Dale JB, Wessels MR, et al. (2018). Blocking Neuronal Signaling to Immune Cells Treats Streptococcal Invasive Infection. Cell 173, 1083–1097 e1022.
    1. Remick DG, Newcomb DE, Bolgos GL, and Call DR (2000). Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock 13, 110–116.
    1. Rosas-Ballina M, Ochani M, Parrish WR, Ochani K, Harris YT, Huston JM, Chavan S, and Tracey KJ (2008). Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc Natl Acad Sci U S A 105, 11008–11013.
    1. Rouot B, Quennedey MC, and Schwartz J (1982). Characteristics of the [3H]-yohimbine binding on rat brain alpha2-adrenoceptors. Naunyn Schmiedebergs Arch Pharmacol 321, 253259.
    1. Salomao R, Martins PS., Brunialti MK, Fernandes Mda, L., Martos LS, Mendes ME., Gomes NE, and Rigato O. (2008). TLR signaling pathway in patients with sepsis. Shock 30 Suppl 1, 73–77.
    1. Sato A (1997). Neural mechanisms of autonomic responses elicited by somatic sensory stimulation. Neurosci Behav Physiol 27, 610–621.
    1. Sell R, Erdelyi A, and Schaefer H (1958). [Effect of peripheral nerve stimulation on sympathetic activity]. Pflugers Arch Gesamte Physiol Menschen Tiere 267, 566–581.
    1. Song JG, Li HH, Cao YF, Lv X, Zhang P, Li YS, Zheng YJ, Li Q, Yin PH, Song SL, et al. (2012). Electroacupuncture improves survival in rats with lethal endotoxemia via the autonomic nervous system. Anesthesiology 116, 406–414.
    1. Spengler RN, Chensue SW, Giacherio DA, Blenk N, and Kunkel SL (1994). Endogenous norepinephrine regulates tumor necrosis factor-alpha production from macrophages in vitro. J Immunol 152, 3024–3031.
    1. Staedtke V, Bai RY, Kim K, Darvas M, Davila ML, Riggins GJ, Rothman PB., Papadopoulos N, Kinzler KW, Vogelstein B, et al. (2018). Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome. Nature 564, 273–277.
    1. Sun JJ, and Ray R (2016). Generation of Two Noradrenergic-Specific Dopamine-Beta-Hydroxylase-FLPo Knock-In Mice Using CRISPR/Cas9-Mediated Targeting in Embryonic Stem Cells. PLoS One 11, e0159474.
    1. Szelenyi J, Kiss JP, and Vizi ES (2000). 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 103, 34–40.
    1. Thoenen H, and Tranzer JP (1973). The pharmacology of 6-hydroxydopamine. Annu Rev Pharmacol 13, 169–180.
    1. Torres-Rosas R, Yehia G, Pena G, Mishra P, del Rocio Thompson-Bonilla M, Moreno-Eutimio MA, Arriaga-Pizano LA, Isibasi A, and Ulloa L (2014). Dopamine mediates vagal modulation of the immune system by electroacupuncture. Nat Med 20, 291–295.
    1. Travagli RA, and Anselmi L (2016). Vagal neurocircuitry and its influence on gastric motility. Nat Rev Gastroenterol Hepatol 13, 389–401.
    1. Ulloa L, Quiroz-Gonzalez S, and Torres-Rosas R (2017). Nerve Stimulation: Immunomodulation and Control of Inflammation. Trends Mol Med 23, 1103–1120.
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, and Netea MG (2017). The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol 17, 407–420.
    1. Vida G, Pena G, Kanashiro A, Thompson-Bonilla Mdel R, Palange D, Deitch EA, and Ulloa L (2011). beta2-Adrenoreceptors of regulatory lymphocytes are essential for vagal neuromodulation of the innate immune system. FASEB J 25, 4476–4485.
    1. Villegas-Bastida A, Torres-Rosas R, Arriaga-Pizano LA, Flores-Estrada J, Gustavo-Acosta A, and Moreno-Eutimio MA (2014). Electrical Stimulation at the ST36 Acupoint Protects against Sepsis Lethality and Reduces Serum TNF Levels through Vagus Nerve- and Catecholamine-Dependent Mechanisms. Evid Based Complement Alternat Med 2014, 451674.
    1. Vranjkovic O, Hang S, Baker DA, and Mantsch JR (2012). beta-adrenergic receptor mediation of stress-induced reinstatement of extinguished cocaine-induced conditioned place preference in mice: roles for betal and beta2 adrenergic receptors. J Pharmacol Exp Ther 342, 541–551.
    1. Yaffe D, Forrest LR, and Schuldiner S (2018). The ins and outs of vesicular monoamine transporters. J Gen Physiol 150, 671–682.
    1. Zhao YX, He W, Jing XH, Liu JL, Rong PJ, Ben H, Liu K, and Zhu B (2012). Transcutaneous auricular vagus nerve stimulation protects endotoxemic rat from lipopolysaccharide-induced inflammation. 2012 Evid Based Complement Altemat Med. 2012;2012:627023, 627023.

Source: PubMed

3
Předplatit