Jiaohong pills attenuate neuroinflammation and amyloid-β protein-induced cognitive deficits by modulating the mitogen-activated protein kinase/nuclear factor kappa-B pathway

Hong Zhang , Weiyan Cai , Lijinchuan Dong , Qing Yang , Qi Li , Qingsen Ran , Li Liu , Yajie Wang , Yujie Li , Xiaogang Weng , Xiaoxin Zhu , Ying Chen

Animal Models and Experimental Medicine ›› 2024, Vol. 7 ›› Issue (3) : 222 -233.

PDF (10255KB)
Animal Models and Experimental Medicine ›› 2024, Vol. 7 ›› Issue (3) : 222 -233. DOI: 10.1002/ame2.12369
ORIGINAL ARTICLE

Jiaohong pills attenuate neuroinflammation and amyloid-β protein-induced cognitive deficits by modulating the mitogen-activated protein kinase/nuclear factor kappa-B pathway

Author information +
History +
PDF (10255KB)

Abstract

Background: Jiaohong pills (JHP) consist of Pericarpium Zanthoxyli (PZ) and Radix Rehmanniae, two herbs that have been extensively investigated over many years due to their potential protective effects against cognitive decline and memory impairment. However, the precise mechanisms underlying the beneficial effects remain elusive. Here, research studies were conducted to investigate and validate the therapeutic effects of JHP on Alzheimer’s disease.

Methods: BV-2 cell inflammation was induced by lipopolysaccharide. AD mice were administered amyloid-β (Aβ). Behavioral experiments were used to evaluate learning and memory ability. The levels of nitric oxide (NO), tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were detected using enzyme-linked immunosorbent assay (ELISA). The protein expressions of inducible nitric oxide synthase (iNOS) and the phosphorylation level of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) were detected using Western blot. Nissl staining was used to detect neuronal degeneration.

Results: The results demonstrated that an alcoholic extract of PZ significantly decreased the levels of NO, IL-1β, TNF-α, and iNOS; increased the expression level of IL-10; and significantly decreased the phosphorylation levels of MAPK and NF-κB. These inhibitory effects were further confirmed in the AD mouse model. Meanwhile, JHP improved learning and memory function in AD mice, reduced neuronal damage, and enriched the Nissl bodies in the hippocampus. Moreover, IL-1β and TNF-α in the cortex were significantly downregulated after JHP administration, whereas IL-10 showed increased expression.

Conclusions: It was found that JHP reduced neuroinflammatory response in AD mice by targeting the MAPK/NF-κB signaling pathway.

Keywords

amyloid-β(Aβ) protein / BV2 / neuroinflammation / Pericarpium Zanthoxyli / Radix Rehmanniae

Cite this article

Download citation ▾
Hong Zhang, Weiyan Cai, Lijinchuan Dong, Qing Yang, Qi Li, Qingsen Ran, Li Liu, Yajie Wang, Yujie Li, Xiaogang Weng, Xiaoxin Zhu, Ying Chen. Jiaohong pills attenuate neuroinflammation and amyloid-β protein-induced cognitive deficits by modulating the mitogen-activated protein kinase/nuclear factor kappa-B pathway. Animal Models and Experimental Medicine, 2024, 7(3): 222-233 DOI:10.1002/ame2.12369

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Calsolaro V, Edison P. Neuroinflammation in Alzheimer’s disease: current evidence and future directions. Alzheimers Dement. 2016;12:719-732.

[2]

Jahn H. Memory loss in Alzheimer’s disease. Dialogues Clin Neuro. 2013;15:445-454.

[3]

Mohamad E, Colombel F, Kapogiannis D, Gallouj K. False memory in Alzheimer’s disease. Behav Neurol. 2020;19:5284504.

[4]

Spangenberg EE, Green KN. Inflammation in Alzheimer’s disease: lessons learned from microglia-depletion models. Brain Behav Immun. 2017;61:1-11.

[5]

Ji J, Xue TF, Guo XD, et al. Antagonizing peroxisome proliferator-activated receptor γfacilitates M1-to-M2 shift of microglia by enhancing autophagy via the LKB1-AMPK signaling pathway. Aging Cell. 2018;17:e12774.

[6]

Crotti A, Ransohoff RM. Microglial physiology and pathophysiology: insights from genome-wide transcriptional profiling. Immunity. 2016;44:505-515.

[7]

Qi Y, Cheng XH, Gong GW, et al. Synergistic neuroprotective effect of schisandrin and nootkatone on regulating inflammation, apoptosis and autophagy via PI3K/AKT pathway. Food Funct. 2020;11:2427-2438.

[8]

Liu N, Zhuang YS, Zhou ZK, Zhao J, Chen Q, Zheng J. NF-κB dependent up-regulation of TRPC6 by Aβin BV-2 microglia cells increases COX-2 expression and contributes to hippocampus neuron damage. Neurosci Lett. 2017;651:1-8.

[9]

Zhao J. The General Record of Saint Francis. Shanghai Science and Technology Press;2015:341.

[10]

Li S. Compendium of Materia Medica, Fruit Department, Volume Heilongjiang Science and Technology Press;2011:664-666.

[11]

State Pharmacopoeia Commission. Pharmacopoeia of the People’s Republic of China: One. China Pharmaceutical Science and Technology Press;2020.

[12]

Shi S, Liang DL, Bao M, et al. Gx-50 inhibits neuroinflammation via α7 nAChR activation of the JAK2/STAT3 and PI3K/AKT pathways. J Alzheimers Dis. 2016;50:859-871.

[13]

Shi S, Liang DL, Chen Y, et al. Gx-50 reduces β-amyloid-induced TNF-α IL-1β NO, and PGE2 expression and inhibits NF-κB signaling in a mouse model of Alzheimer’s disease. Eur J Immunol. 2016;46:665-676.

[14]

Zhang F, You W. Ameliorative effects of pepper polyphenols on memory impairment in aging mice. Chin Mod Appl Pharm. 2011;28:409-411.

[15]

Shi H, Ying L, Shi J, et al. Effects of Rehmannia oligosaccharide on learning and memory ability and hippocampal acetylcholine in vascular dementia rats. Tradit Chin Med Pharmacol Clin. 2008;2:27-29.

[16]

Liu CY, Chen K, Lu YW, Fang Z, Yu G. Catalpol provides a protective effect on fibrillary Aβ1-42-induced barrier disruption in an in vitro model of the blood-brain barrier. Phytother Res. 2018;32:1047-1055.

[17]

MacArthur Clark JA, Sun D. Guidelines for the ethical review of laboratory animal welfare People’s Republic of China National Standard GB/T 35892-2018 [issued 6 February 2018 effective from 1 September 2018]. Animal Model Exp Med. 2020;3:103-113.

[18]

Lucie A, Lucy C, Brenda A. Comparing the effects of isoflurane and alpha chloralose upon mouse physiology. PLoS ONE. 2016;11(5):e0154936.

[19]

Qiang WJ, Cai WY, Yang Q, et al. Artemisinin B improves learning and memory impairment in AD dementia mice by suppressing neuroinflammation. Neuroscience. 2018;395:1-12.

[20]

Yuan HX, Ni XQ, Zheng M, Han X, Song Y, Yu M. Effect of catalpol on behavior and neurodevelopment in an ADHD rat model. Biomed Pharmacother. 2019;118:109033.

[21]

Neuwirth LS, Verrengia MT, Harikinish-Murrary ZI, Orens JE, Lopez OE. Under or absent reporting of light stimuli in testing of anxiety-like behaviors in rodents: the need for standardization. Front Mol Neurosci. 2022;15:912146.

[22]

Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11(1):47-60.

[23]

Archer J. Tests for emotionality in rats and mice: a review. Anim Behav. 1973;21(2):205-235.

[24]

Hu Y, Zhang X, Zhang J, et al. Activated STAT3 signaling pathway by ligature-induced periodontitis could contribute to neuroinflammation and cognitive impairment in rats. J Neuroinflammation. 2021;18(1):80.

[25]

Zhang H, Su Y, Sun Z, et al. Ginsenoside Rg1 alleviates Aβdeposition by inhibiting NADPH oxidase 2 activation in APP/PS1 mice. J Ginseng Res. 2021;45(6):665-675.

[26]

Catorce M, Gevorkian G. LPS-induced murine neuroinflammation model: main features and suitability for pre-clinical assessment of nutraceuticals. Curr Neuropharmacol. 2016;14:155-164.

[27]

Singh SS, Rai SN, Birla H, Zahra W, Rathore AS, Singh SP. NF-κB-mediated neuroinflammation in Parkinson’s disease and potential therapeutic effect of polyphenols. Neurotox Res. 2020;37:491-507.

[28]

Oakes JA, Davies MC, Collins MO. TBK1: a new player in ALS linking autophagy and neuroinflammation. Mol Brain. 2017;10:5.

[29]

Qiu CX, Kivipelto M, Strauss EV. Epidemiology of Alzheimer’s disease:occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci. 2009;11:111-128.

[30]

Alzheimer’s Association. 2016 Alzheimer’s disease facts and figures. Alzheimers Dement. 2016;12:459-509.

[31]

Kant R, Goldstein L, Ossenkoppele R. Amyloid-beta-independent regulators of tau pathology in Alzheimer disease. Nat Rev Neurosci. 2020;21:21-35.

[32]

Benilova I, Karran E, Strooper DB. The toxic Aβoligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;15:1-9.

[33]

Karran E, Strooper DB. The amyloid cascade hypothesis: are we poised for success or failure? J Neurochem. 2016;139:237-252.

[34]

Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, et al. Structure-activity analyses of beta-amyloid peptides: contributions of the beta 25-35 region to aggregation and neurotoxicity. J Neurochem. 1995;64:253-265.

[35]

Banerjee R, Rai A, Iyer SM, et al. Animal models in the study of Alzheimer’s disease and Parkinson’s disease: A historical perspective. Animal Model Exp Med. 2022;5(1):27-37.

[36]

Zhang L, Yu HX, Zhao XC, et al. Neuroprotective effects of salidroside against beta-amyloid-induced oxidative stress in SH-SY5Y human neuroblastoma cells. Neurochem Int. 2010;57:547-555.

[37]

Zhang X, Zhang Y, Zhang L, et al. Overexpression of ACE2 ameliorates Aβ-induced blood-brain barrier damage and angiogenesis by inhibiting NF-κB/VEGF/VEGFR2 pathway. Animal Model Exp Med. 2023;6(3):237-244.

[38]

Diaz A, Rojas K, Espinosa B, et al. Aminoguanidine treatment ameliorates inflammatory responses and memory impairment induced by amyloid-beta 25-35 injection in rats. Neuropeptides. 2014;48:153-159.

[39]

Sambasivam D, Sivanesan S, Sultana S. Conformational preferences of Aβ25-35 and Aβ35-25 in membrane mimicking environments. Protein Pept Lett. 2019;26:386-390.

[40]

Zhou R, Ji B, Kong Y, et al. PET imaging of neuroinflammation in Alzheimer’s disease. Front Immunol. 2021;12:739130.

[41]

Dewald J, Colomb F, Bobowski-Gerard M, Groux-Degroote S, Delannoy P. Role of cytokine-induced glycosylation changes in regulating cell interactions and cell signaling in inflammatory diseases and cancer. Cell. 2016;5:43.

[42]

Zheng C, Zhou XW, Wang JZ. The dual roles of cytokines in Alzheimer’s disease: update on interleukins, TNF-α TGF-βand IFN-γ. Transl Neurodegener. 2016;5:42.

[43]

Zheng J. Hippocampal neurogenesis and pro-neurogenic therapies for Alzheimer’s disease. Animal Model Exp Med. 2022;5(1):3-14.

[44]

Li JJ, Liu SJ, Liu XY, Ling EA. Herbal compounds with special reference to gastrodin as potential therapeutic agents for microglia mediated neuroinflammation. Curr Med Chem. 2018;25:5958-5974.

[45]

Wang JY, Lee CT, Wang JY. Nitric oxide plays a dual role in the oxidative injury of cultured rat microglia but not astroglia. Neuroscience. 2014;281:164-177.

[46]

Carlos CI, Javier MG, Clara AR, et al. Subcellular targeting of nitric oxide synthases mediated by their N-terminal motifs. Adv Protein Chem Struct Biol. 2018;111:165-195.

[47]

Chen HG, Xu YF, Wang JZ, Zhao W, Ruan H. Baicalin ameliorates isoproterenol-induced acute myocardial infarction through iNOS, inflammation and oxidative stress in rat. Int J Clin Exp Pathol. 2015;8:10139-10147.

[48]

Ding X, Wang D, Li LL, Ma H. Dehydroepiandrosterone ameliorates H2O2-induced Leydig cells oxidation damage and apoptosis through inhibition of ROS production and activation of PI3K/Akt pathways. Int J Biochem Biotechnol. 2016;70:126-139.

[49]

Facci L, Barbierato M, Skaper SD. Astrocyte/microglia cocultures as a model to study neuroinflammation. Methods Mol Biol. 2018;1727:127.

[50]

Li L, Xu SF, Liu LF, et al. Multifunctional compound AD-35 improves cognitive impairment and attenuates the production of TNF-αand IL-1βin an Aβ25-35-induced rat model of Alzheimer’s disease. J Alzheimers Dis. 2017;56:1403-1417.

[51]

McFarland AJ, Davey AK, McDermott CM, et al. Differences in statin associated neuroprotection corresponds with either decreased production of IL-1βor TNF-αin an, in vitro, model of neuroinflammation-induced neurodegeneration. Toxicol Appl Pharmacol. 2018;344:56-73.

[52]

Li Z, Qi X, Zhang X, et al. TRDMT1 exhibited protective effects against LPS-induced inflammation in rats through TLR4-NF-κB/MAPK-TNF-αpathway. Animal Model Exp Med. 2022;5(2):172-182.

[53]

McDermott E, O’Neill L. Signal transduction by the lipopolysaccharide receptor, toll-like receptor-4. Immunology. 2004;113:153-162.

[54]

Cunningham C. Microglia and neurodegeneration: the role of systemic inflammation. Glia. 2013;61:71-90.

[55]

Zhu W, Cao FS, Feng J, et al. NLRP3 inflammasome activation contributes to long-term behavioral alterations in mice injected with lipopolysaccharide. Neuroscience. 2017;343:77-84.

[56]

Turrin NP, Gayle D, Ilyin SE, et al. Pro-inflammatory and anti-inflammatory cytokine mRNA induction in the periphery and brain following intraperitoneal administration of bacterial lipopolysaccharide. Brain Res Bull. 2001;54:443-453.

[57]

Xie LS, Zhang N, Zhang Q, et al. Inflammatory factors and amyloid β-induced microglial polarization promote inflammatory crosstalk with astrocytes. Aging (Albany NY). 2020;12:22538-22549.

[58]

Li Y, Liu TT, Li YT, et al. Baicalin ameliorates cognitive impairment and protects microglia from LPS-induced neuroinflammation via the SIRT1/HMGB1 pathway. Oxid Med Cell Longev. 2020;2020:4751349.

[59]

Win S, Than TA, Zhang J, Oo C, Min RWM, Kaplowitz N. New insights into the role and mechanism of c-Jun-N-terminal kinase signaling in the pathobiology of liver diseases. Hepatology. 2018;67:2013-2024.

[60]

Pyo H, Jou I, Jung S, Hong S, Joe EH. Mitogen-activated protein kinases activated by lipopolysaccharide and β-amyloid in cultured rat microglia. Neuroreport. 1998;9:871-874.

[61]

Priyadarshini L, Aggarwal A. Astaxanthin inhibits cytokines production and inflammatory gene expression by suppressing IκB kinase-dependent nuclear factor κB activation in pre and postpartum Murrah buffaloes during different seasons. Vet World. 2018;11:782-788.

[62]

Rhodus NL, Cheng B, Myers S, Miller L, Ho V, Ondrey F. The feasibility of monitoring NF-κB associated cytokines: TNF-α IL-1α IL-6, and IL-8 in whole saliva for the malignant transformation of oral lichen planus. Mol Carcinog. 2005;44:77-82.

[63]

Park J, Min JS, Kim B, et al. Mitochondrial ROS govern the LPS-induced pro-inflammatory response in microglia cells by regulating MAPK and NF-κB pathways. Neurosci Lett. 2015;584:191-196.

RIGHTS & PERMISSIONS

2024 The Author(s). Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for Laboratory Animal Sciences.

AI Summary AI Mindmap
PDF (10255KB)

222

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/