Glycative Stress and the progression of Alzheimer-type Dementia: From the Perspective of Amyloid-beta Clearance

Main Article Content

Yoshikazu Yonei Masayuki Yagi A.NM Mamun-or-Rashid A.NM Mamun-or-Rashid

Abstract

Glycative stress is a conception of a state that causes an excess of carbohydrate-derived or fatty acid-derived short-chain aldehydes in the body. Short-chain aldehydes, which are highly reactive, undergo a carbonylation process with lysine and arginine residues in proteins and form advanced glycation endproducts (AGEs). When postprandial hyperglycemia occurs, open-ring glucose with exposed aldehyde groups (-CHO) increases and various carbohydrate-derived aldehydes are formed simultaneously (this phenomenon is named “aldehyde sparks”). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is an enzyme abundant intracellularly as the keystone of defense against glycative stress and plays a role in metabolizing highly toxic glyceraldehyde, however, under diabetic conditions, the enzyme activity is markedly reduced by forming S-(2-succinyl) cysteine-GAPDH. We also found that a high-fat diet reduced GAPDH activity and increased glyceraldehyde and methylglyoxal. High animal fat diets in particular require caution because they increase preference dependence on animal fat. In the lipid-rich brain, they promotes the formation of lipid-derived AGEs formed by methylglyoxal and acrolein providing evidence of their close association with amyloid cascade, ultimately leading to the onset of Alzheimer-type dementia. Through the process of amylod beta (Aβ) glycation modification and cross-linking, Aβ polymerization is promoted and deposited in the brain. Thus, neurotoxicity is aggravated. Furthermore, Aβ progresses toward being persistent and Aβ clearance is reduced. Tau proteins similarly undergo glycation modification and trigger polymerization. methylglyoxal or acrolein-derived glycated Aβ clearance by primary microglia cultured cells reduced significantly compared to the unglycated Aβ provides evidence of the reduced clearance upon glycation. It was also shown that melatonin, which promotes glycolytic cross-linking degradation, may promote microglial Aβ phagocytosis. We next plan to examine the potential of plant extracts that promote glycation cross-linking degradation to improve Aβ phagocytosis. Advanced Alzheimer-type dementia with a disoriented neural network hardly recovers with treatment. In this review article, we discussed possible future therapeutic strategies for Alzheimer-type dementia by increasing Aβ clearance from the early stage such as the prevention of Aβ-glycation and the promotion of glycated-Aβ degradation, which may be a paradigm shift.


 

Keywords: Glycative Stress, Stress, Dementia, Alzheimer-type Dementia, Amyloid-beta Clearance

Article Details

How to Cite
YONEI, Yoshikazu et al. Glycative Stress and the progression of Alzheimer-type Dementia: From the Perspective of Amyloid-beta Clearance. Medical Research Archives, [S.l.], v. 10, n. 12, dec. 2022. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/3374>. Date accessed: 25 apr. 2024. doi: https://doi.org/10.18103/mra.v10i12.3374.
Section
Review Articles

References

1. Nagai R, Mori T, Yamamoto Y, Kaji Y, Yonei Y. Significance of advanced glycation end products in aging-related disease. Anti-Aging Med. 2010;7:112-9. https://www.jstage.jst.go.jp/article/jaam/7/10/7_10_112/_pdf/-char/ja
2. Ichihashi M, Yagi M, Nomoto K, Yonei Y. Glycation stress and photo-aging in skin. Anti-Aging Med. 2011;8:23-9.
https://www.jstage.jst.go.jp/article/jaam/8/3/8_3_23/_pdf
3. Yonei Y, Yagi M, Takabe W. Stop the "Vicious Cycle" induced by glycative stress. Glycative Stress Res. 2020;7:13-21.
https://doi.org/10.24659/gsr.7.1_13
4. Yonei Y, Yagi M, Takabe W, Kon M. Skin aging: Oxidative stress and glycative stress. Journal of Society Cosmetic Chemists Japan. 2019;53:83-90. (in Japanese) https://www.jstage.jst.go.jp/article/sccj/53/2/53_83/_pdf
5. Maessen DE, Hanssen NM, Scheijen JL, van der Kallen CJ, van Greevenbroek MM, Stehouwer CD, Schalkwijk CG. Post-glucose load plasma α-dicarbonyl concentrations are increased in individuals with impaired glucose metabolism and type 2 diabetes: The CODAM study. Diabetes Care. 2015;38:913-20.
6. Sato K, Zheng Y, Martin-Morales A, Taira T, Yonei Y.Generation of short chain aldehydes and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Glycative Stress Res. 2022;9:129-34.
https://doi.org/10.24659/gsr.9.3_129
7. Nakajima H, Itakura M, Kubo T, Kaneshige A, Harada N, Izawa T, Azuma Y, Kuwamura M, Yamaji R, Takeuchi T. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) aggregation causes mitochondrial dysfunction during oxidative stress-induced cell death. J Biol Chem. 2017;292:4727-42.
8. Nakano T, Goto S, Takaoka Y, Tseng HP, Fujimura T, Kawamoto S, Ono K, Chen CL. A novel moonlight function of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) for immunomodulation. Biofactors. 2018;44: 597 -608.
9. Miwa I, Kito Y, Okuda J. Purification and characterization of triokinase from porcine kidney. Prep Biochem. 1994;24:203-23.
10. Nagai R, Brock JW, Blatnik M, Baatz JE, Bethard J, Walla MD, Thorpe SR, Baynes JW, Frizzell N. Succination of protein thiols during adipocyte maturation: A biomarker of mitochondrial stress. J Biol Chem. 2007;282:34219-28.
11. Frizzell N, Rajesh M, Jepson MJ, Nagai N, Carson JA, Thorpe SR, Baynes JW. Succination of thiol groups in adipose tissue proteins in diabetes: Succination inhibits polymerization and secretion of adiponectin. J Biol Chem. 2009;284: 25772-81.
12. Blatnik M, Thorpe SR, Baynes JW. Succination of proteins by fumarate: mechanism of inactivation of glyceraldehyde-3-phosphate dehydrogenase in diabetes. Ann N Y Acad Sci. 2008;1126:272-5.
13. Adam J, Ramracheya R, Chibalina MV, Ternette N, Hamilton A, Tarasov AI, Zhang Q, Rebelato E, Rorsman NJG, Martín-Del-Río R, Lewis A, Özkan G, Do HW, Spégel P, Saitoh K, Kato K, Igarashi K, Kessler BM, Pugh CW, Tamarit-Rodriguez J, Mulder H, Clark A, Frizzell N, Soga T, Ashcroft FM, Silver A, Pollard PJ, Rorsman P. Fumarate hydratase deletion in pancreatic β cells leads to progressive diabetes. Cell Rep. 2017;20:3135-48.
14. Martin-Morales A, Arakawa T, Sato M, Matsumura Y, Mano-Usui F, Ikeda K, Inagaki N, Sato K. Development of a method for quantitation of glyceraldehyde in various body compartments of rodents and humans. J Agric Food Chem. 2021;69:13246-54.
15. Schalkwijk CG, Stehouwer CDA. Methylglyoxal, a highly reactive dicarbonyl compound, in diabetes, its vascular complications, and other age-related diseases. Physiol Rev. 2020;100: 407-61.
16. Masuzaki H, Kozuka C, Yonamine M, Shimabukuro M. Brown rice-specific γ-oryzanol-based novel approach toward lifestyle-related dysfunction of brain and impaired glucose metabolism. Glycative Stress Res. 2017;4:58-66.
https://doi.org/10.24659/gsr.4.1_058
17. Masuzaki H, Kozuka C, Okamoto S, Yonamine M, Tanaka H, Shimabukuro M. Brown rice-specific γ-oryzanol as a promising prophylactic avenue to protect against diabetes mellitus and obesity in humans. J Diabetes Investig. 2019;10:18-25.
18. Schwartz M, Barush K. The resolution of neuro-inflammation in neuro-degeneration: Leukocyte recruitment via the choroid plexus. EMBO J. 2014;33:7-22.
19. Yi CX, Al-Massadi O, Donelan E, Lehti M, Weber J, Ress C, Trivedi C, Müller TD, Woods SC, Hofmann SM. Exercise protects against high-fat diet-induced hypothalamic inflammation. Physiol Behav. 2012;106:485-90.
20. Epstein DH, Shaham Y. Cheesecake-eating rats and the question of food addiction. Nat Neurosci. 2010;13: 529-31.
21. Kozuka C, Yabiku K, Sunagawa S, Ueda R, Taira S, Ohshiro H, Ikema T, Yamakawa K, Higa M, Tanaka H, Takayama C, Matsushita M, Oyadomari S, Shimabukuro M, Masuzaki H. Brown rice and its component, γ-oryzanol, attenuate the preference for high-fat diet by decreasing hypothalamic endoplasmic reticulum stress in mice. Diabetes. 2012;61:3084-93.
22. Masuzaki H, Fukuda K, Ogata M, Kinjo A, MatsuoT, Nishihira J. Safety and efficacy of nanoparticulated brown rice germ extract on reduction of body fat mass and improvement of fuel metabolism in both pre-obese and mild obese subjects without excess of visceral fat accumulation. Glycative Stress Res. 2020;7:1-12.
https://doi.org/10.24659/gsr.7.1_1
23. Nunomura A, Tamaoki T, Motohashi N, Nakamura M, McKeel Jr DW, Tabaton N, Lee HG, Smith MA, Perry G, Zhu X. The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation invulnerable neurons. J Neuropathol Exp Neurol. 2012;71:233-41.
24. Takeuchi M, Yamagishi S. Possible involvement of advanced glycation end products (AGEs) in the pathogenesis of Alzheimer’s disease. Curr Pharm Des. 2008;14:973-8.
25. Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides. 2015;52:1-18.
26. Cline EN, Bicca MA, Viola KL, Klein WL. The amyloid-β oligomer hypothesis: Beginning of the third decade. J Alzheimers Dis. 2018;64:S567-610.
27. Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: The amyloid hypothesis revised. Nat Rev Neurosci. 2022;23:53-66.
28. Woltjer RL, Maezawa I, Ou JJ, Montine KS, Montine TJ. Advanced glycation endproduct precursor alters intracellular amyloid-beta/A beta PP carboxy-terminal fragment aggregation and cytotoxicity. J Alzheimers Dis. 2003;5:467-76.
29. Barić N. Role of advanced glycation end products (AGEs) on the reactive oxygen species (ROS) generation in Alzheimer's disease amyloid plaque. Glycative Stress Res. 2015;2:140-55.
https://doi.org/10.24659/gsr.2.3_140
30. Muronetz VI, Melnikova AK, Seferbekova ZN, Barinova KV, Schmalhausen EV. Glycation, glycolysis, and neurodegenerative diseases: Is there any connection? Biochemistry (Mosc). 2017; 82:874-86.
31. Yagi M, Yonei Y. Glycative stress and anti-aging: 12. Glycative stress and dementia. Glycative Stress Res. 2019;6:87-91.
https://doi.org/10.24659/gsr.6.2_87
32. Vitek MP, Bhattacharya K, Glendening JM, Stopa E, Vlassara H, Bucala R, Manogue K, Cerami A. Advanced glycation end products contribute to amyloidosis in Alzheimer disease. Proc Natl Acad Sci USA. 1994;91:4766-70.
33. Kuhla B, Lüth HJ, Haferburg D, Boeck K, Arendt T, Münch G. Methylglyoxal, glyoxal, and their detoxification in Alzheimer's disease. Ann N Y Acad Sci. 2005;1043:211-6.
34. Ahmed N, Ahmed U, Thornalley PJ, Hager K, Fleischer G, Münch G. Protein glycation, oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer’s disease and link to cognitive impairment. J Neurochem. 2005;92:255-63.
35. Beeri MS, Moshier E, Schmeidler J, Godbold J, Uribarri J, Reddy S, Sano M, Grossman HT, Cai W, Vlassara H, Silverman JM. Serum concentration of an inflammatory glycotoxin, methylglyoxal, is associated with increased cognitive decline in elderly individuals. Mech Ageing Dev. 2011;132:583-7.
36. Angeloni C, Zambonin L, Hrelia S. Role of methylglyoxal in Alzheimer's disease. Biomed Res Int. 2014;2014:238485.
37. Dang TN, Arseneault M, Murthy V, Ramassamy C. Potential role of acrolein in neurodegeneration and in Alzheimer's disease. Curr Mol Pharmacol. 2010;3:66-78.
38. Waragai M, Yoshida M, Mizoi M, Saiki R, Kashiwagi K, Takagi K, Arai H, Tashiro J, Hashimoto M, Iwai N, Uemura K, Igarashi K. Increased protein-conjugated acrolein and amyloid-β40/42 ratio in plasma of patients with mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2012;32:33-41.
39. Huang YJ, Jin MH, Pi RB, Zhang JJ, Ouyang Y, Chao XJ, Chen MH, Liu PQ, Yu JC, Ramassamy C, Dou J, Chen XH, Jiang YM, Qin J. Acrolein induces Alzheimer's disease-like pathologies in vitro and in vivo. Toxicol Lett. 2013;217:184-91.
40. Igarashi K, Yoshida M, Waragai M, Kashiwagi K. Evaluation of dementia by acrolein, amyloid-β and creatinine. Clin Chim Acta. 2015;450:56-63.
41. Tsou HH, Hsu WC, Fuh JL, Chen SP, Liu TY, Wang HT. Alterations in acrolein metabolism contribute to Alzheimer's disease. J Alzheimers Dis. 2018;61:571-80.
42. Chen C, Chen Y, Lu J, Chen Z, Wang C, Pi R. Acrolein-conjugated proteomics in brains of adult C57BL/6 mice chronically exposed to acrolein and aged APP/PS1 transgenic AD mice. Toxicol Lett. 2021; 344:11-7.
43. Chen C, Lu J, Peng W, Mak MS, Yang Y, Zhu Z, Wang S, Hou J, Zhou X, Xin W, Hu Y, Tsim KWK, Han Y, Liu Q, Pi R. Acrolein, an endogenous aldehyde induces Alzheimer's disease-like pathologies in mice: A new sporadic AD animal model. Pharmacol Res. 2022;175:106003.
44. Alghamdi A, Forbes S, Birch DJS, Vyshemirsky V, Rolinski OJ. Detecting beta-amyloid glycation by intrinsic fluorescence: Understanding the link between diabetes and Alzheimer's disease. Arch Biochem Biophys. 2021; 704:108886.
45. Emendato A, Milordini G, Zacco E, Sicorello A, Dal Piaz F, Guerrini R, Thorogate R, Picone D, Pastore A. Glycation affects fibril formation of Aβ peptides. J Biol Chem. 2018;293:13100-11
46. Smith MA, Taneda S, Richey PL, Miyata S, Yan SD, Stern D, Sayre LM, Monnier VM, Perry G. Advanced Maillard reaction end products are associated with Alzheimer disease pathology. Proc Natl Acad Sci USA. 1994;91:5710-4.
47. Horie K, Miyata T, Yasuda T, Takeda A, Yasuda Y, Maeda K, Sobue G, Kurokawa K. Immunohistochemical localization of advanced glycation end products, pentosidine, and carboxymethyllysine in lipofuscin pigments of Alzheimer's disease and aged neurons. Biochem Biophys Res Commun. 1997;236:327-32.
48. Nacharaju P, Ko L, Yen SH. Characterization of in vitro glycation sites of tau. J Neurochem. 1997;69:1709-19.
49. Yan SD, Chen X, Schmidt AM, Brett J, Godman G, Zou YS, Scott CW, Caputo C, Frappier T, Smith MA. Glycated tau protein in Alzheimer disease: A mechanism for induction of oxidant stress. Proc Natl Acad Sci USA. 1994;91:7787-91.
50. Ledesma MD, Bonay P, Colaço C, Avila J. Analysis of microtubule-associated protein tau glycation in paired helical filaments. J Biol Chem. 1994;269:21614-9.
51. Srikanth V, Maczurek A, Phan T, Steele M, Westcott B, Juskiw D, Münch G. Advanced glycation endproducts and their receptor RAGE in Alzheimer's disease. Neurobiol Aging. 2011;32:763-77.
52. Takeda A, Yasuda T, Miyata T, Goto Y, Wakai M, Watanabe M, Yasuda Y, Horie K, Inagaki T, Doyu M, Maeda K, Sobue G. Advanced glycation end products co-localized with astrocytes and microglial cells in Alzheimer's disease brain. Acta Neuropathol. 1998;95:555-8.
53. Suzumura A. Neurotoxicity by microglia: The mechanisms and potential therapeutic strategy. Fukuoka Acta Medica. 2009;100;243-7. (in Japanese)
54. Hamby ME, Sofroniew MV. Reactive astrocytes as therapeutic targets for CNS disorders. Neurotherapeutics. 2010;7:494-506.
55. Dukic-Stefanovic S, Gasic-Milenkovic J, Deuther-Conrad W, Münch G. Signal transduction pathways in mouse microglia N-11 cells activated by advanced glycation endproducts (AGEs). J Neurochem. 2003;87:44-55.
56. Takeuchi M, Bucala R, Suzuki T, Ohkubo T, Yamazaki M, Koike T, Kameda Y, Makita Z. Neurotoxicity of advanced glycation end-products for cultured cortical neurons. J Neuropathol Exp Neurol. 2000;59:1094-105.
57. Adav SS, Sze SK. Insight of brain degenerative protein modifications in the pathology of neurodegeneration and dementia by proteomic profiling. Mol Brain. 2016;9:92.
58. Akiyama H, Arai T, Kondo H, Tanno E, Haga C, Ikeda K. Cell mediators of inflammation in the Alzheimer disease brain. Alzheimer Dis Assoc Disord. 2000; 14:S47-53.
59. Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: The amyloid hypothesis revised. Nat Rev Neurosci. 2022;23:53-66.
60. Yonei Y, Taira T, Otaka S, Sekiguchi S, Mamun-or-Rashid ANM, Yagi M, Masuzaki H. Amyloid beta clearance and microglia: Effects of glycative stress and melatonin. Glycative Stress Res. 2022;9:135-45.
https://doi.org/10.24659/gsr.9.3_135
61. Matsumura A, Suzuki S, Iwahara N, Hisahara S, Kawamata J, Suzuki H, Yamauchi A, Takata K, Kitamura Y, Shimohama S. Temporal changes of CD68 and α7 nicotinic acetylcholine receptor expression in microglia in Alzheimer's disease-like mouse models. J Alzheimers Dis. 2015;44:409-23.
62. Pan RY, Ma J, Kong XX, Wang XF, Li SS, Qi XL, Yan YH, Cheng J, Liu Q, Jin W, Tan CH, Yuan Z. Sodium rutin ameliorates Alzheimer's disease-like pathology by enhancing microglial amyloid-β clearance. Sci Adv. 2019;5:eaau6328.
63. Yagi M, Yonei Y. Glycative stress and anti-aging: 14. Regulation of Glycative stress. 2. Inhibition of the AGE production and accumulation. Glycative Stress Res. 2019; 6:212-8.
https://doi.org/10.24659/gsr.6.4_212
64. Takeshita S, Yagi M, Uemura T, Yamada M, Yonei Y. Peel extract of water chestnut (Trapa bispinosa Roxb.) inhibits glycation, degrades α-dicarbonyl compound, and breaks advanced glycation end product crosslinks. Glycative Stress Res. 2015;2:72-9.
https://doi.org/10.24659/gsr.2.2_072
65. Jean D, Pouligon M, Dalle C. Evaluation in vitro of AGE-crosslinks breaking ability of rosmarinic acid. Glycative Stress Res. 2015;2:204-7.
https://doi.org/10.24659/gsr.2.4_204
66. Yagi M, Takabe W, Matsumi S, Shimode A, Maruyama T, Yonei Y. Biochemistry of Kuromoji (Lindera umbellata) extract: Anti-oxidative and anti-glycative actions. Glycative Stress Res. 2017;4:329-40.
https://doi.org/10.24659/gsr.4.4_329
67. Yagi M, Inoue K, Sato Y, Ishizaki K, Sakiyama C, Yonei Y. Antiglycative effect of black galangal, Kaempferia parviflora Wall. Ex. Baker (Zingiberaceae). Glycative Stress Res. 2021;8:1-7.
https://doi.org/10.24659/gsr.8.1_1
68. Noguchi-Shinohara M, Ono K, Hamaguchi T, Nagai T, Kobayashi S, Komatsu J, Samuraki-Yokohama M, Iwasa K, Yokoyama K, Nakamura H, Yamada M. Safety and efficacy of Melissa officinalis extract containing rosmarinic acid in the prevention of Alzheimer's disease progression. Sci Rep. 2020;10:18627.
69. Takabe W, Mitsuhashi R, Parengkuan L, Yagi M, Yonei Y. Cleaving effect of melatonin on crosslinks in advanced glycation end products. Glycative Stress Res. 2016;3:38-43.
https://doi.org/10.24659/gsr.3.1_038
70. Vasan S, Zhang X, Zhang X. Kapurniotu A, Bernhagen J, Teichberg S, Basgen J, Wagle D, Shih D, Terlecky I, Bucala R, Cerami A, Egan J, Ulrich P. An agent cleaving glucose-derived protein crosslinks in vitro and in vivo. Nature. 1996; 382: 275-278.
71. Wu H, Dunnett S, Ho YS, Chang RC. The role of sleep deprivation and circadian rhythm disruption as risk factors of Alzheimer's disease. Front Neuroendocrinol. 2019;54: 100764.
72. Shukla M, Govitrapong P, Boontem P, Reiter RJ, Satayavivad J. Mechanisms of melatonin in alleviating Alzheimer's disease. Curr Neuropharmacol. 2017;15:1010-31.
73. Rebelo S, Vieira SI, Esselmann H, Wiltfang J, da Cruz e Silva EF, da Cruz e Silva OA. Tyrosine 687 phosphorylated Alzheimer’s amyloid precursor protein is retained intracellularly and exhibits a decreased turnover rate. Neurodegener Dis. 2007;4: 78-87.
74. Robakis NK, Ramakrishna N, Wolfe G, Wisniewski HM. Molecular cloning and characterization of a cDNA encoding the cerebrovascular and the neuritic plaque amyloid peptides. Proc Natl Acad Sci USA. 1987;84:4190-4.
75. Rogaev EI, Sherrington R, Rogaeva EA, Levesque G, Ikeda M, Liang Y, Chi H, Lin C, Holman K, Tsuda T, Mar L, Sorbi S, Nacmias B, Piacentini S, Amaducci L, Chumakov I, Cohen D, Lannfelt L, Fraser PE, Rommens JM, St George-Hyslop PH. Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature. 1995;376:775-8.
76. Balmik AA, Chinnathambi S. Multi-faceted role of melatonin in neuroprotection and amelioration of Tau aggregates in Alzheimer's disease. J Alzheimers Dis. 2018;62:1481-93.
77. Hoppe JB, Frozza RL, Horn AP, Comiran RA, Bernardi A, Campos MM, Battastini AMO, Salbego C. Amyloid-beta neurotoxicity in organotypic culture is attenuated by melatonin: Involvement of GSK-3beta, tau and neuroinflammation. J Pineal Res. 2010; 48:230-8.
78. Ionov M, Burchell V, Klajnert B, Bryszewska M, Abramov AY. Mechanism of neuroprotection of melatonin against beta-amyloid neurotoxicity. Neuroscience. 2011; 180:229-37.
79. Pappolla MA, Matsubara E, Vidal R, Pacheco-Quinto J, Poeggeler B, Zagorski M, Sambamurti K. Melatonin treatment enhances Aβ lymphatic clearance in a transgenic mouse model of amyloidosis. Curr Alzheimer Res. 2018;15:637-42.
80. 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.
81. Zhang S, Wang P, Ren L, Hu C, Bi J. Protective effect of melatonin on soluble Aβ1-42-induced memory impairment, astrogliosis, and synaptic dysfunction via the Musashi1/Notch1/Hes1 signaling pathway in the rat hippocampus. Alzheimers Res Ther. 2016;8:40.