[1] 王大伟, 周荣斌, 姚咏明. 胆碱能抗炎通路在炎症反应中的作用[J]. 生理科学进展, 2010, 41(3):217-220.
[2] BOUZAT C, LASALA M, NIELSEN B E, et al. Molecular function of α7 nicotinic receptors as drug targets[J]. J Physiol,2018,596(10):1847-1861. doi:  10.1113/JP275101
[3] BERTRAND D, LEE C H, FLOOD D, et al. Therapeutic potential of α7 nicotinic acetylcholine receptors[J]. Pharmacol Rev, 2015, 67(4): 1025-1073.
[4] MACKLIN K D, MAUS A D, PEREIRA E F, et al. Human vascular endothelial cells express functional nicotinic acetylcholine receptors[J]. J Pharmacol Exp Ther,1998,287(1):435-439.
[5] CHEN L Y, LIU Z G, LI Y H, et al. Expression of neuronal acetylcholine receptor alpha 7 (nAChRalpha7) in peripheral blood CD(4)(+) T lymphocytes from asthmatic children[J]. Chin J Tuberc Respir Dis,2008,31(11):803-805.
[6] KALKMAN H O, FEUERBACH D. Modulatory effects of α7 nAChRs on the immune system and its relevance for CNS disorders[J]. Cell Mol Life Sci,2016,73(13):2511-2530. doi:  10.1007/s00018-016-2175-4
[7] CORRADI J, BOUZAT C. Understanding the bases of function and modulation of α7 nicotinic receptors: implications for drug discovery[J]. Mol Pharmacol,2016,90(3):288-299. doi:  10.1124/mol.116.104240
[8] KABBANI N, NICHOLS R A. Beyond the channel: metabotropic signaling by nicotinic receptors[J]. Trends Pharmacol Sci,2018,39(4):354-366. doi:  10.1016/j.tips.2018.01.002
[9] BOROVIKOVA L V, IVANOVA S, ZHANG M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin[J]. Nature,2000,405(6785):458-462. doi:  10.1038/35013070
[10] 余江, 张恒, 张润峰. 胆碱能抗炎通路及其调控心脏炎症反应的作用[J]. 中国心血管病研究, 2021, 19(9):843-848. doi:  10.3969/j.issn.1672-5301.2021.09.017
[11] 马莉, 冷玉芳, 张梦婕, 等. 胆碱能抗炎通路对器官的保护研究进展[J]. 兰州大学学报(医学版), 2020, 46(1):10-15. doi:  10.13885/j.issn.1000-2812.2020.01.003
[12] QIN Z, XIANG K F, SU D F, et al. Activation of the cholinergic anti-inflammatory pathway as a novel therapeutic strategy for COVID-19[J]. Front Immunol,2020,11:595342.
[13] WANG H, YU M, OCHANI M, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation[J]. Nature,2003,421(6921):384-388. doi:  10.1038/nature01339
[14] 陈卓, 何骁, 姚孟维, 等. 胆碱能炎症反射与炎症消退研究进展[J]. 中华烧伤杂志, 2021, 37(9):885-889. doi:  10.3760/cma.j.cn501120-20200609-00299
[15] 李祺, 刘霞. 胆碱能抗炎通路的信号机制研究[J]. 药学实践杂志, 2010, 28(5):325-327,338.
[16] HOOVER D B. Cholinergic modulation of the immune system presents new approaches for treating inflammation[J]. Pharmacol Ther,2017,179:1-16. doi:  10.1016/j.pharmthera.2017.05.002
[17] 王娜, 苏跃. 全身炎症反应综合征治疗新策略: 胆碱能抗炎通路[J]. 医学与哲学, 2010, 31(22):42-43,77.
[18] 资双凤, 李景辉, 刘蕾, 等. 胆碱能抗炎通路及其在脓毒症治疗中的作用[J]. 中南大学学报(医学版), 2020, 45(1):68-73.
[19] BROGNARA F, CASTANIA J A, DIAS D P M, et al. Baroreflex stimulation attenuates central but not peripheral inflammation in conscious endotoxemic rats[J]. Brain Res,2018,1682:54-60. doi:  10.1016/j.brainres.2018.01.003
[20] SAPRONOV N S, NEZHINSKAYA G I, VLADYKIN A L. Effects of cholinergic agonists and antagonists under conditions of spleen denervation in rats with endotoxic shock[J]. Bull Exp Biol Med,2010,149(5):598-600. doi:  10.1007/s10517-010-1002-4
[21] RIVERA C A, WHEELER M D, ENOMOTO N, et al. A choline-rich diet improves survival in a rat model of endotoxin shock[J]. Am J Physiol,1998,275(4):G862-G867.
[22] BENCHERIF M, LIPPIELLO P M, LUCAS R, et al. Alpha7 nicotinic receptors as novel therapeutic targets for inflammation-based diseases[J]. Cell Mol Life Sci,2011,68(6):931-949. doi:  10.1007/s00018-010-0525-1
[23] PAVLOV V A, PARRISH W R, ROSAS-BALLINA M, et al. Brain acetylcholinesterase activity controls systemic cytokine levels through the cholinergic anti-inflammatory pathway[J]. Brain Behav Immun,2009,23(1):41-45. doi:  10.1016/j.bbi.2008.06.011
[24] 郑紫磊, 郭睿, 郜思齐, 等. 迷走神经刺激活化胆碱能抗炎通路对创伤/失血性休克的干预作用[J]. 河北医药, 2021, 43(16):2531-2535. doi:  10.3969/j.issn.1002-7386.2021.16.031
[25] KOHOUTOVA M, HORAK J, JARKOVSKA D, et al. Vagus nerve stimulation attenuates multiple organ dysfunction in resuscitated porcine progressive Sepsis[J]. Crit Care Med,2019,47(6):e461-e469. doi:  10.1097/CCM.0000000000003714
[26] 李建国, 胡正芳, 杜朝晖, 等. 胆碱能抗炎通路对失血性休克大鼠保护作用的研究[J]. 中国危重病急救医学, 2005(1):24-27.
[27] 宫丽荣, 董树安, 阚永星, 等. α7烟碱型乙酰胆碱受体在电针减轻肢体缺血再灌注兔肺损伤中的作用[J]. 中国中西医结合外科杂志, 2019, 25(4):443-447. doi:  10.3969/j.issn.1007-6948.2019.04.004
[28] 田甜, 何君艺, 樊素雄, 等. α7烟碱型乙酰胆碱受体对缺血再灌注损伤影响的研究现状[J]. 中国临床药理学杂志, 2020, 36(18):2914-2917.
[29] MA Z, ZHANG Z L, BAI F H, et al. Electroacupuncture pretreatment alleviates cerebral ischemic injury through α7 nicotinic acetylcholine receptor-mediated phenotypic conversion of microglia[J]. Front Cell Neurosci,2019,13:537.
[30] XIE J L, LI X, ZHANG L M, et al. Genistein-3'-sodium sulfonate ameliorates cerebral ischemia injuries by blocking neuroinflammation through the α7nAChR-JAK2/STAT3 signaling pathway in rats[J]. Phytomedicine,2021,93:153745. doi:  10.1016/j.phymed.2021.153745
[31] MUNYAKA P, RABBI M F, PAVLOV V A, et al. Central muscarinic cholinergic activation alters interaction between splenic dendritic cell and CD4+CD25- T cells in experimental colitis[J]. PLoS One,2014,9(10):e109272. doi:  10.1371/journal.pone.0109272
[32] SHAO B Z, WANG S L, FANG J, et al. Alpha7 nicotinic acetylcholine receptor alleviates inflammatory bowel disease through induction of AMPK-mTOR-p70S6K-mediated autophagy[J]. Inflammation,2019,42(5):1666-1679. doi:  10.1007/s10753-019-01027-9
[33] WAZEA S A, WADIE W, BAHGAT A K, et al. Galantamine anti-colitic effect: role of alpha-7 nicotinic acetylcholine receptor in modulating Jak/STAT3, NF-κB/HMGB1/RAGE and p-AKT/Bcl-2 pathways[J]. Sci Rep,2018,8(1):5110. doi:  10.1038/s41598-018-23359-6
[34] NUNES N S, CHANDRAN P, SUNDBY M, et al. Therapeutic ultrasound attenuates DSS-induced colitis through the cholinergic anti-inflammatory pathway[J]. EBioMedicine,2019,45:495-510. doi:  10.1016/j.ebiom.2019.06.033
[35] HARMYCH S J, KUMAR J, BOUNI M E, et al. Nicotine inhibits MAPK signaling and spheroid invasion in ovarian cancer cells[J]. Exp Cell Res,2020,394(1):112167. doi:  10.1016/j.yexcr.2020.112167
[36] OFFERHAUS G J, TERSMETTE A C, HUIBREGTSE K, et al. Mortality caused by stomach cancer after remote partial gastrectomy for benign conditions: 40 years of follow up of an Amsterdam cohort of 2633 postgastrectomy patients[J]. Gut,1988,29(11):1588-1590. doi:  10.1136/gut.29.11.1588
[37] AHSBERG K, OLSSON H, VON HOLSTEIN C S. Increased mortality in prostate carcinoma and smoking-related disease after parietal cell vagotomy: a long-term follow-up study[J]. Scand J Gastroenterol,2009,44(8):947-951. doi:  10.1080/00365520903039945
[38] OGAWA T, MAKINO T, MIZUMOTO K, et al. Promoting effect of truncal vagotomy on pancreatic carcinogenesis initiated with N-nitrosobis(2-oxopropyl)amine in Syrian golden hamsters[J]. Carcinogenesis,1991,12(7):1227-1230. doi:  10.1093/carcin/12.7.1227
[39] MORGENSTERN L. Vagotomy, gastroenterostomy and experimental gastic cancer[J]. Arch Surg,1968,96(6):920-923. doi:  10.1001/archsurg.1968.01330240066014
[40] KOWALEWSKI K. Relationship between vagotomy, peptic ulcer and gastric adenocarcinoma in rats fed 2, 7-diacetylaminogluorene[J]. Can J Surg,1973,16(3):210-217.
[41] ERIN N, DUYMUŞ O, OZTÜRK S, et al. Activation of vagus nerve by semapimod alters substance P levels and decreases breast cancer metastasis[J]. Regul Pept,2012,179(1-3):101-108. doi:  10.1016/j.regpep.2012.08.001
[42] REIJMEN E, VANNUCCI L, DE COUCK M, et al. Therapeutic potential of the vagus nerve in cancer[J]. Immunol Lett,2018,202:38-43. doi:  10.1016/j.imlet.2018.07.006
[43] WINEK K, SOREQ H, MEISEL A. Regulators of cholinergic signaling in disorders of the central nervous system[J]. J Neurochem,2021,158(6):1425-1438. doi:  10.1111/jnc.15332
[44] SCHUHMANN M K, PAPP L, STOLL G, et al. Mesencephalic electrical stimulation reduces neuroinflammation after photothrombotic stroke in rats by targeting the cholinergic anti-inflammatory pathway[J]. Int J Mol Sci,2021,22(3):1254. doi:  10.3390/ijms22031254
[45] LIU D, LI T, LUO H, et al. The effect of the cholinergic anti-inflammatory pathway on collagen-induced arthritis involves the modulation of dendritic cell differentiation[J]. Arthritis Res Ther,2018,20(1):263. doi:  10.1186/s13075-018-1759-9
[46] LI S, ZHOU B, LIU B, et al. Activation of the cholinergic anti-inflammatory system by nicotine attenuates arthritis via suppression of macrophage migration[J]. Mol Med Rep,2016,14(6):5057-5064. doi:  10.3892/mmr.2016.5904
[47] WU S Y, LUO H, XIAO X Z, et al. Attenuation of collagen induced arthritis via suppression on Th17 response by activating cholinergic anti-inflammatory pathway with nicotine[J]. Eur J Pharmacol,2014,735:97-104. doi:  10.1016/j.ejphar.2014.04.019
[48] WU S Y, ZHAO H J, LUO H, et al. GTS-21, an α7-nicotinic acetylcholine receptor agonist, modulates Th1 differentiation in CD4+ T cells from patients with rheumatoid arthritis[J]. Exp Ther Med,2014,8(2):557-562. doi:  10.3892/etm.2014.1754
[49] VAN MAANEN M A, LEBRE M C, VAN DER POLL T, et al. Stimulation of nicotinic acetylcholine receptors attenuates collagen-induced arthritis in mice[J]. Arthritis Rheum,2009,60(1):114-122. doi:  10.1002/art.24177
[50] YI L, KE J Y, LIU J Y, et al. Sinomenine increases adenosine A 2A receptor and inhibits NF-κB to inhibit arthritis in adjuvant-induced-arthritis rats and fibroblast-like synoviocytes through α7nAChR[J]. J Leukoc Biol,2021,110(6):1113-1120. doi:  10.1002/JLB.3MA0121-024RRRR
[51] KOOPMAN F A, SCHUURMAN P R, VERVOORDELDONK M J, et al. Vagus nerve stimulation: a new bioelectronics approach to treat rheumatoid arthritis? Best Pract Res Clin Rheumatol,2014,28(4):625-635. doi:  10.1016/j.berh.2014.10.015
[52] DREWES A M, BROCK C, RASMUSSEN S E, et al. Short-term transcutaneous non-invasive vagus nerve stimulation may reduce disease activity and pro-inflammatory cytokines in rheumatoid arthritis: results of a pilot study[J]. Scand J Rheumatol,2021,50(1):20-27. doi:  10.1080/03009742.2020.1764617
[53] COURTIES A, BERENBAUM F, SELLAM J. Vagus nerve stimulation in musculoskeletal diseases[J]. Joint Bone Spine,2021,88(3):105149. doi:  10.1016/j.jbspin.2021.105149
[54] LV J Q, JI X X, LI Z, et al. The role of the cholinergic anti-inflammatory pathway in autoimmune rheumatic diseases[J]. Scand J Immunol,2021,94(4):e13092.
[55] ZHOU Y, CHI J W, LV W S, et al. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (Covid-19)[J]. Diabetes Metab Res Rev,2021,37(2):e3377.
[56] KWENANDAR F, JAPAR K V, DAMAY V, et al. Coronavirus disease 2019 and cardiovascular system: a narrative review[J]. Int J Cardiol Heart Vasc,2020,29:100557.
[57] CHEN N S, ZHOU M, DONG X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study[J]. Lancet,2020,395(10223):507-513. doi:  10.1016/S0140-6736(20)30211-7
[58] WEI Y F, SHAN L, QIAO L M, et al. Protective effects of Huang-Lian-Jie-du-Tang against polymicrobial Sepsis induced by cecal ligation and puncture in rats[J]. Evid Based Complement Alternat Med,2013,2013:909624.
[59] XU D Q, LV Y, WANG J S, et al. Deciphering the mechanism of Huang-Lian-Jie-Du-Decoction on the treatment of Sepsis by formula decomposition and metabolomics: enhancement of cholinergic pathways and inhibition of HMGB-1/TLR4/NF-κB signaling[J]. Pharmacol Res,2017,121:94-113. doi:  10.1016/j.phrs.2017.04.016
[60] REN J L, ZHANG A H, WANG X J. Traditional Chinese medicine for COVID-19 treatment[J]. Pharmacol Res,2020,155:104743. doi:  10.1016/j.phrs.2020.104743
[61] MAIZEL' E B, ROZENGART E V, KHAKIMOV I U P, et al. Ephedrine, salsoline and cytisine derivatives as substrates and inhibitirs of cholinesterases[J]. Biokhimiia,1978,43(7):1150-1156.
[62] KINALI M, BEESON D, PITT M C, et al. Congenital myasthenic syndromes in childhood: diagnostic and management challenges[J]. J Neuroimmunol,2008,201-202:6-12. doi:  10.1016/j.jneuroim.2008.06.026
[63] STAATS P, GIANNAKOPOULOS G, BLAKE J, et al. The use of non-invasive vagus nerve stimulation to treat respiratory symptoms associated with COVID-19: a theoretical hypothesis and early clinical experience[J]. Neuromodulation,2020,23(6):784-788. doi:  10.1111/ner.13172
[64] LU F, SELAK M, O'CONNOR J, et al. Oxidative damage to mitochondrial DNA and activity of mitochondrial enzymes in chronic active lesions of multiple sclerosis[J]. J Neurol Sci,2000,177(2):95-103. doi:  10.1016/S0022-510X(00)00343-9
[65] PAVLOV V A, TRACEY K J. The vagus nerve and the inflammatory reflex: linking immunity and metabolism[J]. Nat Rev Endocrinol,2012,8(12):743-754. doi:  10.1038/nrendo.2012.189
[66] WU J, JIAO Z Y, LI R Z, et al. Cholinergic activation suppresses palmitate-induced macrophage activation and improves acylation stimulating protein resistance in co-cultured adipocytes[J]. Exp Biol Med (Maywood),2017,242(9):961-973. doi:  10.1177/1535370217700522
[67] NISHIO T, TAURA K, IWAISAKO K, et al. Hepatic vagus nerve regulates Kupffer cell activation via α7 nicotinic acetylcholine receptor in nonalcoholic steatohepatitis[J]. J Gastroenterol,2017,52(8):965-976. doi:  10.1007/s00535-016-1304-z
[68] DENG J L, WANG M, GUO Y K, et al. Activation of α7nAChR via vagus nerve prevents obesity-induced insulin resistance via suppressing endoplasmic Reticulum stress-induced inflammation in Kupffer cells[J]. Med Hypotheses,2020,140:109671. doi:  10.1016/j.mehy.2020.109671
[69] YKI-JÄRVINEN H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome[J]. Lancet Diabetes Endocrinol,2014,2(11):901-910. doi:  10.1016/S2213-8587(14)70032-4
[70] KATSIKI N, MIKHAILIDIS D P, MANTZOROS C S. Non-alcoholic fatty liver disease and dyslipidemia: an update[J]. Metabolism,2016,65(8):1109-1123. doi:  10.1016/j.metabol.2016.05.003
[71] WOO BAIDAL J A, LAVINE J E. The intersection of nonalcoholic fatty liver disease and obesity[J]. Sci Transl Med,2016,8(323):323rv1.
[72] BIRKENFELD A L, SHULMAN G I. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes[J]. Hepatology,2014,59(2):713-723. doi:  10.1002/hep.26672
[73] BUZZETTI E, PINZANI M, TSOCHATZIS E A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD)[J]. Metabolism,2016,65(8):1038-1048. doi:  10.1016/j.metabol.2015.12.012
[74] KIMURA K, INABA Y, WATANABE H, et al. Nicotinic alpha-7 acetylcholine receptor deficiency exacerbates hepatic inflammation and fibrosis in a mouse model of non-alcoholic steatohepatitis[J]. J Diabetes Investig,2019,10(3):659-666. doi:  10.1111/jdi.12964
[75] TING K K, BREW B, GUILLEMIN G. The involvement of astrocytes and kynurenine pathway in Alzheimer's disease[J]. Neurotox Res,2007,12(4):247-262. doi:  10.1007/BF03033908
[76] O'BRIEN J T, EAGGER S, SYED G M, et al. A study of regional cerebral blood flow and cognitive performance in Alzheimer's disease[J]. J Neurol Neurosurg Psychiatry, 1992, 55(12): 1182-1187. https://pubmed.ncbi.nlm.nih.gov/1479398/https://pubmed.ncbi.nlm.nih.gov/1479398/
[77] DE LA MONTE S M. Brain insulin resistance and deficiency as therapeutic targets in Alzheimer’s disease[J]. Curr Alzheimer Res,2012,9(1):35-66. doi:  10.2174/156720512799015037
[78] TOTA S, KAMAT P K, SHUKLA R, et al. Improvement of brain energy metabolism and cholinergic functions contributes to the beneficial effects of silibinin against streptozotocin induced memory impairment[J]. Behav Brain Res,2011,221(1):207-215. doi:  10.1016/j.bbr.2011.02.041
[79] HOYER S. Glucose metabolism and insulin receptor signal transduction in Alzheimer disease[J]. Eur J Pharmacol,2004,490(1-3):115-125. doi:  10.1016/j.ejphar.2004.02.049
[80] ZEEVALK G D, BERNARD L P, SINHA C, et al. Excitotoxicity and oxidative stress during inhibition of energy metabolism[J]. Dev Neurosci,1998,20(4-5):444-453. doi:  10.1159/000017342
[81] NAVARRO E, GONZALEZ-LAFUENTE L, PÉREZ-LIÉBANA I, et al. Heme-oxygenase I and PCG-1α regulate mitochondrial biogenesis via microglial activation of Alpha7 nicotinic acetylcholine receptors using PNU282987[J]. Antioxid Redox Signal,2017,27(2):93-105. doi:  10.1089/ars.2016.6698
[82] PLASCHKE K, HOYER S. Action of the diabetogenic drug streptozotocin on glycolytic and glycogenolytic metabolism in adult rat brain cortex and Hippocampus[J]. Int J Dev Neurosci,1993,11(4):477-483. doi:  10.1016/0736-5748(93)90021-5
[83] PATHAN A R, VISWANAD B, SONKUSARE S K, et al. Chronic administration of pioglitazone attenuates intrace-rebroventricular streptozotocin induced-memory impairment in rats[J]. Life Sci,2006,79(23):2209-2216. doi:  10.1016/j.lfs.2006.07.018
[84] RAJASEKAR N, NATH C, HANIF K, et al. Intranasal insulin improves cerebral blood flow, Nrf-2 expression and BDNF in STZ (ICV)-induced memory impaired rats[J]. Life Sci,2017,173:1-10. doi:  10.1016/j.lfs.2016.09.020
[85] RAJASEKAR N, DWIVEDI S, NATH C, et al. Protection of streptozotocin induced insulin receptor dysfunction, neuroinflammation and amyloidogenesis in astrocytes by insulin[J]. Neuropharmacology,2014,86:337-352. doi:  10.1016/j.neuropharm.2014.08.013