The gut-brain axis: Effect of electroacupuncture pretreatment on learning, memory, and JNK signaling in D-galactose-induced AD-like rats

Document Type : Original Article

Authors

1 College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China

2 Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, Hubei, China

3 Medical Department,Wuhan Red Cross Hospital, Wuhan, Hubei, China

4 School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China

Abstract

Objective(s): To examine the effect and potential mechanism of electroacupuncture (EA) pretreatment in spatial learning, memory, gut microbiota, and JNK signaling in D-galactose-induced AD-like rats. 
Materials and Methods: The AD-like rat model was generated by intraperitoneal injection of D-galactose. Morris water maze was used to determine spatial learning and memory ability, Real-time PCR to determine intestinal flora levels, ELISA to determine tryptophan (Trp) and 5-HT levels in the colon and hippocampal tissues, immunofluorescence to determine 5-HT levels in enterochromaffin cells (ECs), and immunoblotting to determine JNK signaling protein levels in hippocampal tissues.
Results: Electroacupuncture pretreatment significantly reduced escape latency and prolonged exploration time in the target quadrant, and significantly increased the relative DNA abundance of Lactobacillus and Bifidobacterium. Meanwhile, electroacupuncture pretreatment also reduced colonic 5-HT levels and increased hippocampal 5-HT levels. Moreover, electroacupuncture pretreatment significantly inhibited hippocampal JNK pathway-related protein expression, including 5-HT6R, JNK, p-JUNK, c-JUN, and p-c-Jun. And the combination of GV20 and ST36 was more effective than single acupoints.
Conclusion: Electroacupuncture pretreatment improved the learning and memory ability of D-galactose-induced AD-like model rats, changed the gut microbiota composition, and the mechanism may be related to the gut-brain axis and the JNK signaling pathway.   In addition, the combination of GV20 and ST36 could further enhance the efficacy.

Keywords


1. Gaugler J, James B, Johnson T, Reimer J, Solis M, Weuve J, et al. 2022 Alzheimer’s disease facts and figures. Alzheimers Dement 2022; 18:700-789.
2. Mokdad AH, Ballestros K, Echko M, Glenn S, Olsen HE, Mullany E, et al. The state of US health, 1990-2016: burden of diseases, injuries, and risk factors among US states. JAMA 2018; 319:1444-1472.
3. Ciaccio M, Lo Sasso B, Scazzone C, Gambino CM, Ciaccio AM, Bivona G, et al. COVID-19 and Alzheimer’s disease. Brain Sci 2021; 11:305-315.
4. Rajan KB, Weuve J, Barnes LL, McAninch EA, Wilson RS, Evans DA. Population estimate of people with clinical Alzheimer’s disease and mild cognitive impairment in the United States (2020–2060). Alzheimers Dement 2021; 17:1966-1975.
5. China Dementia and Cognitive Disorders Writing Group, Cognitive Disorders Committee of the Neurologist Branch of the Chinese Medical Association. 2018 Chinese guidelines for the diagnosis and treatment of dementia and cognitive impairment (II): Guidelines for the diagnosis and treatment of Alzheimer’s disease. National Medical Journal of China 2018; 98:971-977.
6. Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. New Eng J Med 2018; 378:321-330.
7. McDade E, Bateman RJ. Stop Alzheimer’s before it starts. Nature 2017; 547:153-155.
8. Zhao Y, Dua P, Lukiw W. Microbial sources of amyloid and relevance to amyloidogenesis and Alzheimer’s disease (AD). J Alzheimers Dis Parkinsonism 2015; 5:177-190.
9. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010; 464:59-65.
10. O’Toole PW, Jeffery IB. Gut microbiota and aging. Science 2015; 350:1214-1215.
11. Bischoff SC. Microbiota and aging. Curr Opin Clin Nutr Metab Care 2016; 19:26-30.
12. Woodmansey EJ. Intestinal bacteria and ageing. J Apll Microbiol 2007; 102:1178-1186.
13. Thevaranjan N, Puchta A, Schulz C, Naidoo A, Szamosi J, Verschoor CP, et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 2017; 21:455-466.
14. Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and butyrate-producing colon bacteria: importance and strategies for their stimulation in the human gut. Front Microbiol 2016; 7:979-1000.
15. Sharon G, Sampson TR, Geschwind DH, Mazmanian SK. The central nervous system and the gut microbiome. Cell 2016; 167:915-932.
16. Scott KA, Ida M, Peterson VL, Prenderville JA, Moloney GM, Izumo T, et al. Revisiting metchnikoff: age-related alterations in microbiota-gut-brain axis in the mouse. Brain Behav Immun 2017; 65:20-32.
17. Jiang C, Li G, Huang P, Liu Z, Zhao B. The gut microbiota and Alzheimer’s disease. J Alzheimers Dis 2017; 58:1-15.
18. Wang XS, Li JJ, Wang YS, Yu CC, He C, Huang ZS, et al. Acupuncture and related therapies for the cognitive function of alzheimer’s disease: a network meta-analysis. Iran J Public Health 2021; 50:2411-2426.
19. Wu C, Xu Y, Guo H, Tang C, Chen D, Zhu M. Effects of aerobic exercise and mind-body exercise in parkinson’s disease: a mixed-treatment comparison analysis. Front Aging Neurosci 2021; 13:739115-739125.
20. Zhang ZJ, Chen HY, Yip KC, Ng R, Wong VT. The effectiveness and safety of acupuncture therapy in depressive disorders: systematic review and meta-analysis. J Affect Disord 2010; 124:9-21.
21. Wang X, Shi X, Lv J, Zhang J, Huo Y, Zuo G, et al. Acupuncture and related therapies for the anxiety and depression in irritable bowel syndrome with diarrhea (IBS-D): a network meta-analysis of randomized controlled trials. Front Psychiatry 2022; 13:1067329-1067347.
22. Kwon CY, Ko SJ, Lee B, Cha JM, Yoon JY, Park JW. Acupuncture as an add-on treatment for functional dyspepsia: a systematic review and meta-analysis. Front Med (Lausanne) 2021; 8:682783-682804.
23. Jin Y, Hu F, Zhu J. Exploration of acupuncture therapy in the treatment of mild cognitive impairment based on the brain-gut axis theory. Front Hum Neurosci 2022; 16:891411-891420.
24. He C, Huang Z-S, Yu C-C, Wang X-S, Jiang T, Wu M, et al. Preventive electroacupuncture ameliorates D-galactose-induced Alzheimer’s disease-like inflammation and memory deficits, probably via modulating the microbiota–gut–brain axis. Iran J Basic Medi Sci 2021; 24:341-348.
25. He C, Huang C, Chen H, Yu C, Wang X, Jiang T, et al. Effect of pretreatment of acupuncture on learning-memory ability and related protein of NLRP3 inflammasome in hippocampus in Alzheimer’s disease like rats. Zhongguo Zhen Jiu 2021; 38:4-5.
26. Gao J, Zhou R, You X, Luo F, He H, Chang X, et al. Salidroside suppresses inflammation in a D-galactose-induced rat model of Alzheimer’s disease via SIRT1/NF-κB pathway. Metab Brain Dis 2016; 31:771-778.
27. Wang C, Cai Z, Wang W, Wei M, Si X, Shang Y, et al. Piperine regulates glycogen synthase kinase-3β-related signaling and attenuates cognitive decline in D-galactose-induced aging mouse model. J Nutr Biochem 2020; 75:108261-108304.
28. Sun R, Li S, Shao Y, Li W, Ding C, Zhang J, et al. Method optimization of D-galactose induced mice subacute aging model. Zhongguo Zhong Yao Za Zhi 2015; 31:293-297.
29. Yu C-C, He C, Du Y-J, Gao S, Lin Y-F, Wang S-Q, et al. Preventive electroacupuncture reduces cognitive deficits in a rat model of D-galactose-induced aging. Neural Regen Res 2021; 16:916-926.
30. Azman KF, Zakaria R. D-Galactose-induced accelerated aging model: an overview. Biogerontology 2019; 20:763-782.
31. Shwe T, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Role of D-galactose-induced brain aging and its potential used for therapeutic interventions. Exp Gerontol 2018; 101:13-36.
32. Tsai SJ, Chiu CP, Yang HT, Yin MC. S-Allyl cysteine, s-ethyl cysteine, and s-propyl cysteine alleviate β-amyloid, glycative, and oxidative injury in brain of mice treated by D-galactose. J Agric Food Chem 2011; 59:6319-6326.
33. Ali T, Badshah H, Kim TH, Kim MO. Melatonin attenuates D-galactose-induced memory impairment, neuroinflammation and neurodegeneration via RAGE/NF-K B/JNK signaling pathway in aging mouse model. J Pineal Res 2015; 58:71-85.
34. Haider S, Liaquat L, Shahzad S, Sadir S, Madiha S, Batool Z, et al. A high dose of short term exogenous D-galactose administration in young male rats produces symptoms simulating the natural aging process. Life Sci 2015; 124:110-119.
35. Hong XP, Chen T, Yin NN, Han YM, Yuan F, Duan YJ, et al. Puerarin ameliorates D-galactose induced enhanced hippocampal neurogenesis and tau hyperphosphorylation in rat brain. J Alzheimers Dis 2016; 51:605-617.
36. Yu C-C, Wang J, Ye S-S, Gao S, Li J, Wang L, et al. Preventive electroacupuncture ameliorates D-galactose-induced Alzheimer’s disease-like pathology and memory deficits probably via inhibition of GSK3β/mTOR signaling pathway. Evid Based Complement Alternat Med 2020; 2020: 1428752.
37. Yuk-Yu R. A network meta-analysis of acupuncture and moxibustion for Alzheimer’s disease(AD) and discussion on the principles of acupoints [Doctoral Thesis]: Guangzhou University of Chinese Medicine; 2019.
38. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol 2015; 28:203-209.
39. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s disease microbiome is associated with dysregulation of the anti-inflammatory P-glycoprotein pathway. mBio 2019; 10:1-14.
40. Meneses A. 5-HT system and cognition. Neurosci Biobehav Rev 1999; 23:1111-1125.
41. Ouchi Y, Yoshikawa E, Futatsubashi M, Yagi S, Ueki T, Nakamura K. Altered brain serotonin transporter and associated glucose metabolism in Alzheimer disease. J Nucl Med 2009; 50:1260-1266.
42. Bowen D, Allen S, Benton J, Goodhardt M, Haan E, Palmer A, et al. Biochemical assessment of serotonergic and cholinergic dysfunction and cerebral atrophy in Alzheimer’s disease. J Neurochem 1983; 41:266-272.
43. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis communication. Adv Exp Med Biol 2014; 817:115-133.
44. O’Mahony SM, Clarke G, Borre Y, Dinan TG, Cryan J. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res 2015; 277:32-48.
45. Kohen R, Metcalf MA, Khan N, Druck T, Huebner K, Lachowicz JE, et al. Cloning, characterization, and chromosomal localization of a human 5‐HT6 serotonin receptor. J Neurochem 1996; 66:47-56.
46. de Jong IE, Mørk A. Antagonism of the 5-HT6 receptor–preclinical rationale for the treatment of Alzheimer’s disease. Neuropharmacology 2017; 125:50-63.
47. Bokare AM, Bhonde M, Goel R, Nayak Y. 5-HT6 receptor agonist and antagonist modulates ICV-STZ-induced memory impairment in rats. Psychopharmacology 2018; 235:1557-1570.
48. Yun H-M, Baik J-H, Kang I, Jin C, Rhim H. Physical interaction of Jab1 with human serotonin 6 G-protein-coupled receptor and their possible roles in cell survival. J  Biol Chem 2010; 285:10016-10029.
49. Cui Y, Jiang L, Shao Y, Mei L, Tao Y. Anti-alcohol liver disease effect of gentianae macrophyllae extract through MAPK/JNK/p38 pathway. J Pharm Pharmacol 2019; 71:240-250.
50. Liu C, Wen Y, Huang H, Lin W, Huang M, Lin R, et al. Over-expression of 5-HT6 receptor and activated Jab-1/pc-Jun play important roles in pilocarpine-induced seizures and learning-memory impairment. J Mol Neurosci 2019; 67:388-399.