Naringin and Naringenin: Potential Multi-Target Agents for Alzheimer’s Disease

Jing Lu, Jie Chen, Shu-yue Li, Guang-jie Pan, Yi Ou, Li-fu Yuan, Jian-ping Jiang, Ling-hui Zeng, Jie Zhao

Current Medical Science ›› DOI: 10.1007/s11596-024-2921-z
Review

Naringin and Naringenin: Potential Multi-Target Agents for Alzheimer’s Disease

Author information +
History +

Abstract

Alzheimer’s disease (AD) is one of the most common forms of neurodegenerative dementia. The etiology of AD is multifactorial, and its complex pathophysiology involves tau and amyloid-β deposition, increased oxidative stress, neuroinflammation, metabolic disorders, and massive neuronal loss. Due to its complex pathology, no effective cure for AD has been found to date. Therefore, there is an unmet clinical need for the development of new drugs against AD. Natural products are known to be good sources of compounds with pharmacological activity and have potential for the development of new therapeutic agents. Naringin, a naturally occurring flavanone glycoside, is predominantly found in citrus fruits and Chinese medicinal herbs. Mounting evidence shows that naringin and its aglycone, naringenin, have direct neuroprotective effects on AD, such as anti-amyloidogenic, antioxidant, anti-acetylcholinesterase, and anti-neuroinflammatory effects, as well as metal chelation. Furthermore, they are known to improve disordered glucose/lipid metabolism, which is a high risk factor for AD. In this review, we summarize the latest data on the impact of naringin and naringenin on the molecular mechanisms involved in AD pathophysiology. Additionally, we provide an overview of the current clinical applications of naringin and naringenin. The novel delivery systems for naringin and naringenin, which can address their widespread pharmacokinetic limitations, are also discussed. The literature indicates that naringin and naringenin could be multilevel, multitargeted, and multifaceted for preventing and treating AD.

Cite this article

Download citation ▾
Jing Lu, Jie Chen, Shu-yue Li, Guang-jie Pan, Yi Ou, Li-fu Yuan, Jian-ping Jiang, Ling-hui Zeng, Jie Zhao. Naringin and Naringenin: Potential Multi-Target Agents for Alzheimer’s Disease. Current Medical Science, https://doi.org/10.1007/s11596-024-2921-z

References

[1]
2023 Alzheimer’s disease facts and figures. Alzheimers Dement, 2023,19(4):1598–1695
[2]
Forrest SL, Kovacs GG. Current concepts of mixed pathologies in neurodegenerative diseases. Can J Neurol Sci, 2023, 50(3): 329-345.
CrossRef Google scholar
[3]
Wimo A, Guerchet M, Ali GC, et al. . The worldwide costs of dementia 2015 and comparisons with 2010. Alzheimers Dement, 2017, 13(1): 1-7.
CrossRef Google scholar
[4]
Ma C, Hong F, Yang S. Amyloidosis in Alzheimer’s Disease: Pathogeny, Etiology, and Related Therapeutic Directions. Molecules, 2022, 27(4): 1210.
CrossRef Google scholar
[5]
Gao Y, Tan L, Yu JT, et al. . Tau in Alzheimer’s Disease: Mechanisms and Therapeutic Strategies. Curr Alzheimer Res, 2018, 15(3): 283-300.
CrossRef Google scholar
[6]
Bai R, Guo J, Ye XY, et al. . Oxidative stress: The core pathogenesis and mechanism of Alzheimer’s disease. Aging Res Rev, 2022, 77: 101619.
CrossRef Google scholar
[7]
Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here?. Nat Rev Neurol, 2021, 17(3): 157-172.
CrossRef Google scholar
[8]
Song Y, Liu J, Zhao K, et al. . Cholesterol-induced toxicity: An integrated view of the role of cholesterol in multiple diseases. Cell Metab, 2021, 33(10): 1911-1925.
CrossRef Google scholar
[9]
Biessels GJ, Despa F. Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications. Nat Rev Endocrinol, 2018, 14(10): 591-604.
CrossRef Google scholar
[10]
Picone P, Di Carlo M, Nuzzo D. Obesity and Alzheimer’s disease: Molecular bases. Eur J Neurosci, 2020, 52(8): 3944-3950.
CrossRef Google scholar
[11]
Vinciguerra F, Graziano M, Hagnäs M, et al. . Influence of the mediterranean and ketogenic diets on cognitive status and decline: a narrative review. Nutrients, 2020, 12(4): 1019.
CrossRef Google scholar
[12]
Glynn-Servedio BE, Ranola TS. AChE Inhibitors and NMDA Receptor Antagonists in Advanced Alzheimer’s Disease. Consult Pharm, 2017, 32(9): 511-518.
CrossRef Google scholar
[13]
Terao I, Kodama W. Comparative efficacy, tolerability and acceptability of donanemab, lecanemab, aducanumab and lithium on cognitive function in mild cognitive impairment and Alzheimer’s disease: A systematic review and network metaanalysis. Aging Res Rev, 2024, 94: 102203.
CrossRef Google scholar
[14]
Kumar MS, Khan S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease. Curr Neuropharmacol, 2020, 18(11): 1106-1125.
CrossRef Google scholar
[15]
Patel D, Shukla S, Gupta S. Apigenin and cancer chemoprevention: progress, potential and promise. Int J Oncol, 2007, 30(1): 233-245
[16]
Miccadei S, Di Venere D, Cardinali A, et al. . Antioxidative and apoptotic properties of polyphenolic extracts from edible part of artichoke (Cynara scolymus L.) on cultured rat hepatocytes and on human hepatoma cells. Nutr Cancer, 2008, 60(2): 276-283.
CrossRef Google scholar
[17]
Madunić J, Madunić IV, Gajski G, et al. . Apigenin: A dietary flavonoid with diverse anticancer properties. Cancer Lett, 2018, 413: 11-22.
CrossRef Google scholar
[18]
Abenavoli L, Izzo AA, Milić N, et al. . Milk thistle (Silybum marianum): A concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother Res, 2018, 32(11): 2202-2213.
CrossRef Google scholar
[19]
Azzini E, Maiani G, Garaguso I, et al. . The potential health benefits of polyphenol-rich extracts from Cichorium intybus L. studied on Caco-2 cells model. Oxid Med Cell Longev, 2016, 2016: 1594616.
CrossRef Google scholar
[20]
Calderaro A, Patanè GT, Tellone E, et al. . The Neuroprotective Potentiality of Flavonoids on Alzheimer’s Disease. Int J Mol Sci, 2022, 23(23): 14835.
CrossRef Google scholar
[21]
Yin L, Cheng W, Qin Z, et al. . Effects of naringin on proliferation and osteogenic differentiation of human periodontal ligament stem cells in vitro and in vivo. Stem Cells Int, 2015, 2015: 758760.
CrossRef Google scholar
[22]
Wong KC, Pang WY, Wang XL, et al. . Drynaria fortunei-derived total flavonoid fraction and isolated compounds exert oestrogen-like protective effects in bone. Br J Nutr, 2013, 110(3): 475-485.
CrossRef Google scholar
[23]
Lee S, Youn K, Lim G, et al. . In silico docking and in vitro approaches towards BACE1 and cholinesterases inhibitory effect of citrus flavanones. Molecules, 2018, 23(7): 1509.
CrossRef Google scholar
[24]
Garcez ML, Mina F, Bellettini-Santos T, et al. . The Involvement of NLRP3 on the Effects of Minocycline in an AD-Like Pathology Induced by β-Amyloid Oligomers Administered to Mice. Mol Neurobiol, 2018, 56(4): 2606-2617.
CrossRef Google scholar
[25]
Ahsan AU, Sharma VL, Wani A, et al. . Naringenin upregulates AMPK-mediated autophagy to rescue neuronal cells from β-amyloid (1–42) evoked neurotoxicity. Mol Neurobiol, 2020, 57(8): 3589-3602.
CrossRef Google scholar
[26]
Wang K, Chen Z, Huang L, et al. . Naringenin reduces oxidative stress and improves mitochondrial dysfunction via activation of the Nrf2/ARE signaling pathway in neurons. Int J Mol Med, 2017, 40(5): 1582-1590.
CrossRef Google scholar
[27]
Kumar A, Prakash A, Dogra S. Naringin alleviates cognitive impairment, mitochondrial dysfunction and oxidative stress induced by D-galactose in mice. Food Chem Toxicol, 2010, 48(2): 626-632.
CrossRef Google scholar
[28]
Sachdeva AK, Kuhad A, Chopra K. Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol Biochem Behav, 2014, 127: 101-110.
CrossRef Google scholar
[29]
Zaidun NH, Thent ZC, Abd Latiff A. Combating oxidative stress disorders with citrus flavonoid: Naringenin. Life Sci, 2018, 208: 111-122.
CrossRef Google scholar
[30]
Heo HJ, Kim M-J, Lee J-M, et al. . Naringenin from Citrus junos has an inhibitory effect on acetylcholinesterase and a mitigating effect on amnesia. Dement Geriatr Cogn Disord, 2004, 17(3): 151-157.
CrossRef Google scholar
[31]
Umukoro S, Kalejaye HA, Ben-Azu B, et al. . Naringenin attenuates behavioral derangements induced by social defeat stress in mice via inhibition of acetylcholinesterase activity, oxidative stress and release of pro-inflammatory cytokines. Biomed Pharmacother, 2018, 105: 714-723.
CrossRef Google scholar
[32]
Haider S, Liaquat L, Ahmad S, et al. . Naringenin protects AlCl3/D-galactose induced neurotoxicity in rat model of AD via attenuation of acetylcholinesterase levels and inhibition of oxidative stress. PLoS One, 2020, 15(1): e0227631.
CrossRef Google scholar
[33]
Wu LH, Lin C, Lin HY, et al. . Naringenin suppresses neuroinflammatory responses through inducing suppressor of cytokine signaling 3 expression. Mol Neurobiol, 2016, 53(2): 1080-1091.
CrossRef Google scholar
[34]
Khajevand-Khazaei MR, Ziaee P, Motevalizadeh SA, et al. . Naringenin ameliorates learning and memory impairment following systemic lipopolysaccharide challenge in the rat. Eur J Pharmacol, 2018, 826: 114-122.
CrossRef Google scholar
[35]
Chen C, Wei YZ, He XM, et al. . Naringenin produces neuroprotection against LPS-induced dopamine neurotoxicity via the inhibition of microglial NLRP3 inflammasome activation. Front Immunol, 2019, 10: 936.
CrossRef Google scholar
[36]
Yang Z, Kuboyama T, Tohda C. Naringenin promotes microglial M2 polarization and Aβ degradation enzyme expression. Phytother Res, 2019, 33(4): 1114-1121.
CrossRef Google scholar
[37]
Jahanshahi M, Khalili M, Margedari A. Naringin chelates excessive iron and prevents the formation of amyloid-beta plaques in the hippocampus of iron-overloaded mice. Front Pharmacol, 2021, 12: 651156.
CrossRef Google scholar
[38]
Guo LX, Sun B. N, N′-1, 10-Bis (Naringin) Triethylenetetraamine, Synthesis and as a Cu (II) Chelator for Alzheimer’s Disease Therapy. Biol Pharm Bull, 2021, 44(1): 51-56.
CrossRef Google scholar
[39]
Jagetia GC, Reddy TK, Venkatesha V, et al. . Influence of naringin on ferric iron induced oxidative damage in vitro. Clin Chim Acta, 2004, 347(1–2): 189-197.
CrossRef Google scholar
[40]
Yang Y, Trevethan M, Wang S, et al. . Beneficial effects of citrus flavanones naringin and naringenin and their food sources on lipid metabolism: An update on bioavailability, pharmacokinetics, and mechanisms. J Nutr Biochem, 2022, 104: 108967.
CrossRef Google scholar
[41]
Nyane NA, Tlaila TB, Malefane TG, et al. . Metformin-like antidiabetic, cardio-protective and non-glycemic effects of naringenin: Molecular and pharmacological insights. Eur J Pharmacol, 2017, 803: 103-111.
CrossRef Google scholar
[42]
Stevens Y, Rymenant EV, Grootaert C, et al. . The Intestinal Fate of Citrus Flavanones and Their Effects on Gastrointestinal Health. Nutrients, 2019, 11(7): 1464.
CrossRef Google scholar
[43]
Panche AN, Diwan AD, Chandra SR. Flavonoids: an overview. J Nutr Sci, 2016, 5: e47.
CrossRef Google scholar
[44]
Kay CD. The future of flavonoid research. Br J Nutr, 2010, 104(S3): S91-S95.
CrossRef Google scholar
[45]
Kasote DM, Katyare SS, Hegde MV, et al. . Significance of antioxidant potential of plants and its relevance to therapeutic applications. Int J Biol Sci, 2015, 11(8): 982-991.
CrossRef Google scholar
[46]
Manchope MF, Casagrande R, Verri WA Jr. Naringenin: an analgesic and anti-inflammatory citrus flavanone. Oncotarget, 2017, 8(3): 3766-3767.
CrossRef Google scholar
[47]
Memariani Z, Abbas SQ, Ul Hassan SS, et al. . Naringin and naringenin as anticancer agents and adjuvants in cancer combination therapy: Efficacy and molecular mechanisms of action, a comprehensive narrative review. Pharmacol Res, 2021, 171: 105264.
CrossRef Google scholar
[48]
Barreca D, Gattuso G, Bellocco E, et al. . Flavanones: Citrus phytochemical with health-promoting properties. Biofactors, 2017, 43(4): 495-506.
CrossRef Google scholar
[49]
Di Majo D, Giammanco M, La Guardia M, et al. . Flavanones in Citrus fruit: Structure-antioxidant activity relationships. Food Res Int, 2005, 38(10): 1161-1166.
CrossRef Google scholar
[50]
Hsiu SL, Huang TY, Hou YC, et al. . Comparison of metabolic pharmacokinetics of naringin and naringenin in rabbits. Life Sci, 2002, 70(13): 1481-1489.
CrossRef Google scholar
[51]
Liu M, Zou W, Yang C, et al. . Metabolism and excretion studies of oral administered naringin, a putative antitussive, in rats and dogs. Biopharm Drug Dispos, 2012, 33(3): 123-134.
CrossRef Google scholar
[52]
Orrego-Lagarón N, Martínez-Huélamo M, Vallverdú-Queralt A, et al. . High gastrointestinal permeability and local metabolism of naringenin: influence of antibiotic treatment on absorption and metabolism. Br J Nutr, 2015, 114(2): 169-180.
CrossRef Google scholar
[53]
Zeng X, Bai Y, Peng W, et al. . Identification of naringin metabolites in human urine and feces. Eur J Drug Metab Pharmacokinet, 2017, 42: 647-656.
CrossRef Google scholar
[54]
Li SQ, Dong S, Su ZH, et al. . Comparative pharmacokinetics of naringin in rat after oral administration of chaihu-shugan-san aqueous extract and naringin alone. Metabolites, 2013, 3(4): 867-880.
CrossRef Google scholar
[55]
Zeng X, Yao H, Zheng Y, et al. . Tissue distribution of naringin and derived metabolites in rats after a single oral administration. J Chromatogr B Analyt Technol Biomed Life Sci, 2020, 1136: 121846.
CrossRef Google scholar
[56]
Manach C, Williamson G, Morand C, et al. . Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr, 2005, 81(1): 230S-242S.
CrossRef Google scholar
[57]
Zou W, Luo Y, Liu M, et al. . Human intestinal microbial metabolism of naringin. Eur J Drug Metab Pharmacokinet, 2015, 40: 363-367.
CrossRef Google scholar
[58]
Joshi R, Kulkarni YA, Wairkar S. Pharmacokinetic, pharmacodynamic and formulations aspects of Naringenin: An update. Life Sci, 2018, 215: 43-56.
CrossRef Google scholar
[59]
Rebello CJ, Beyl RA, Lertora JJ, et al. . Safety and pharmacokinetics of naringenin: A randomized, controlled, single-ascending-dose clinical trial. Diabetes Obes Metab, 2020, 22(1): 91-98.
CrossRef Google scholar
[60]
Zeng X, Su W, Zheng Y, et al. . Pharmacokinetics, tissue distribution, metabolism, and excretion of naringin in aged rats. Front Pharmacol, 2019, 10: 34.
CrossRef Google scholar
[61]
Zou W, Yang C, Liu M, et al. . Tissue distribution study of naringin in rats by liquid chromatography-tandem mass spectrometry. Arzneimittelforschung, 2012, 62(4): 181-186.
CrossRef Google scholar
[62]
Khan MB, Khan MM, Khan A, et al. . Naringenin ameliorates Alzheimer’s disease (AD)-type neurodegeneration with cognitive impairment (AD-TNDCI) caused by the intracerebroventricular-streptozotocin in rat model. Neurochem Int, 2012, 61(7): 1081-1093.
CrossRef Google scholar
[63]
Kay CD, Pereira-Caro G, Ludwig IA, et al. . Anthocyanins and flavanones are more bioavailable than previously perceived: A review of recent evidence. Annu Rev Food Sci Technol, 2017, 8: 155-180.
CrossRef Google scholar
[64]
Zhang J, Brodbelt JS. Screening flavonoid metabolites of naringin and narirutin in urine after human consumption of grapefruit juice by LC-MS and LC-MS/MS. Analyst, 2004, 129(12): 1227-1233.
CrossRef Google scholar
[65]
Pereira-Caro G, Ludwig IA, Polyviou T, et al. . Identification of plasma and urinary metabolites and catabolites derived from orange juice (poly) phenols: analysis by high-performance liquid chromatography-high-resolution mass spectrometry. J Agric Food Chem, 2016, 64(28): 5724-5735.
CrossRef Google scholar
[66]
Additives EPo Feed PoSuiA. Scientific Opinion on the safety and efficacy of naringin when used as a sensory additive for all animal species. EFSA J, 2011, 9(11): 2416
[67]
Bacanli M, Başaran AA, Başaran N. The major flavonoid of grapefruit: naringin. Polyphenols: prevention and treatment of human disease, 2018 37-44.
CrossRef Google scholar
[68]
Li P, Wang S, Guan X, et al. . Acute and 13 weeks subchronic toxicological evaluation of naringin in Sprague-Dawley rats. Food Chem Toxicol, 2013, 60: 1-9.
CrossRef Google scholar
[69]
Li D, Lu L, Zhang J, et al. . Mitigating the effects of Xuebijing injection on hematopoietic cell injury induced by total body irradiation with γ rays by decreasing reactive oxygen species levels. Int J Mol Sci, 2014, 15(6): 10541-10553.
CrossRef Google scholar
[70]
Fuhr U, Kummert AL. The fate of naringin in humans: a key to grapefruit juice-drug interactions?. Clin Pharmacol Ther, 1995, 58(4): 365-373.
CrossRef Google scholar
[71]
Tiwari MK, Kepp KP. β-Amyloid pathogenesis: Chemical properties versus cellular levels. Alzheimers Dement, 2016, 12(2): 184-194.
CrossRef Google scholar
[72]
Hardy J, Higgins G. Disease: Alzheimer’s cascade hypothesis amyloid. Science, 1992, 256: 184-185.
CrossRef Google scholar
[73]
Hardy J, Selkoe DJ. The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics. Science, 2002, 297(5580): 353-356.
CrossRef Google scholar
[74]
Zhou T, Liu L, Wang Q, et al. . Naringenin alleviates cognition deficits in high-fat diet-fed SAMP8 mice. J Food Biochem, 2020, 44(9): e13375.
CrossRef Google scholar
[75]
Meng X, Fu M, Wang S, et al. . Naringin ameliorates memory deficits and exerts neuroprotective effects in a mouse model of Alzheimer’s disease by regulating multiple metabolic pathways. Mol Med Rep, 2021, 23(5): 332.
CrossRef Google scholar
[76]
Adewole KE, Ishola AA. BACE1 and cholinesterase inhibitory activities of compounds from Cajanus cajan and Citrus reticulata: an in silico study. LIn Silico Pharmacol, 2021, 9(1): 14.
CrossRef Google scholar
[77]
Zhang N, Hu Z, Zhang Z, et al. . Protective Role Of Naringenin Against Aβ(25–35)-Caused Damage via ER and PI3K/Akt-Mediated Pathways. Cell Mol Neurobiol, 2018, 38(2): 549-557.
CrossRef Google scholar
[78]
Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell, 2017, 66(6): 789-800.
CrossRef Google scholar
[79]
Ghofrani S, Joghataei MT, Mohseni S, et al. . Naringenin improves learning and memory in an Alzheimer’s disease rat model: Insights into the underlying mechanisms. Eur J Pharmacol, 2015, 764: 195-201.
CrossRef Google scholar
[80]
Choi GY, Kim HB, Hwang ES, et al. . Naringin enhances long-term potentiation and recovers learning and memory deficits of amyloid-beta induced Alzheimer’s disease-like behavioral rat model. Neurotoxicology, 2023, 95: 35-45.
CrossRef Google scholar
[81]
Onyango IG. Modulation of mitochondrial bioenergetics as a therapeutic strategy in Alzheimer’s disease. Neural Regen Res, 2018, 13(1): 19-25.
CrossRef Google scholar
[82]
Varshney V, Garabadu D. Naringin Exhibits Mas Receptor–Mediated Neuroprotection Against Amyloid Beta–Induced Cognitive Deficits and Mitochondrial Toxicity in Rat Brain. Neurotox Res, 2021, 39(4): 1023-1043.
CrossRef Google scholar
[83]
Wang DM, Yang YJ, Zhang L, et al. . Naringin Enhances CaMKII Activity and Improves Long-Term Memory in a Mouse Model of Alzheimer’s Disease. Int J Mol Sci, 2013, 14(3): 5576-86.
CrossRef Google scholar
[84]
Lei P, Ayton S, Bush AI. The essential elements of Alzheimer’s disease. J Biol Chem, 2021, 296: 100105.
CrossRef Google scholar
[85]
Chen LL, Fan YG, Zhao LX, et al. . The metal ion hypothesis of Alzheimer’s disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem, 2023, 131: 106301.
CrossRef Google scholar
[86]
Ban XX, Wan H, Wan XX, et al. . Copper Metabolism and Cuproptosis: Molecular Mechanisms and Therapeutic Perspectives in Neurodegenerative Diseases. Curr Med Sci, 2024, 44(1): 28-50.
CrossRef Google scholar
[87]
Eleutherio ECA, Silva Magalhães RS, de Araújo Brasil A, et al. . SOD1, more than just an antioxidant. Arch Biochem Biophys, 2021, 697: 108701.
CrossRef Google scholar
[88]
Rios M, Habecker B, Sasaoka T, et al. . Catecholamine synthesis is mediated by tyrosinase in the absence of tyrosine hydroxylase. J Neurosci, 1999, 19(9): 3519-3526.
CrossRef Google scholar
[89]
Squitti R. Copper dysfunction in Alzheimer’s disease: from meta-analysis of biochemical studies to new insight into genetics. J Trace Elem Med Biol, 2012, 26(2–3): 93-96.
CrossRef Google scholar
[90]
Rembach A, Hare DJ, Lind M, et al. . Decreased copper in Alzheimer’s disease brain is predominantly in the soluble extractable fraction. Int J Alzheimers Dis, 2013, 2013: 623241.
[91]
Bayer TA, Schafer S, Simons A, et al. . Dietary Cu stabilizes brain superoxide dismutase 1 activity and reduces amyloid Abeta production in APP23 transgenic mice. Proc Natl Acad Sci U S A, 2003, 100(24): 14187-14192.
CrossRef Google scholar
[92]
Matheou CJ, Younan ND, Viles JH. Cu2+ accentuates distinct misfolding of Aβ (1–40) and Aβ (1–42) peptides, and potentiates membrane disruption. Biochem J, 2015, 466(2): 233-242.
CrossRef Google scholar
[93]
Guilloreau L, Combalbert S, Sournia-Saquet A, et al. . Redox chemistry of copper-amyloid-β: The generation of hydroxyl radical in the presence of ascorbate is linked to redox-potentials and aggregation state. Chembiochem, 2007, 8(11): 1317-1325.
CrossRef Google scholar
[94]
Multhaup G, Schlicksupp A, Hesse L, et al. . The amyloid precursor protein of Alzheimer’s disease in the reduction of copper (II) to copper (I). Science, 1996, 271(5254): 1406-1409.
CrossRef Google scholar
[95]
Fernandez MT, Mira ML, Florêncio MH, et al. . Iron and copper chelation by flavonoids: an electrospray mass spectrometry study. J Inorg Biochem, 2002, 92(2): 105-111.
CrossRef Google scholar
[96]
Celiz G, Suarez SA, Arias A, et al. . Synthesis, structural elucidation and antiradical activity of a copper (II) naringenin complex. Biometals, 2019, 32(4): 595-610.
CrossRef Google scholar
[97]
Tarkowski E, Andreasen N, Tarkowski A, et al. . Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry, 2003, 74(9): 1200-1205.
CrossRef Google scholar
[98]
Ransohoff RM, Brown MA. Innate immunity in the central nervous system. J Clin Invest, 2012, 122(4): 1164-1171.
CrossRef Google scholar
[99]
Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol, 2016, 173(4): 649-665.
CrossRef Google scholar
[100]
Cao J, Hou J, Ping J, et al. . Advances in developing novel therapeutic strategies for Alzheimer’s disease. Mol Neurodegener, 2018, 13(1): 1-20.
CrossRef Google scholar
[101]
Subhramanyam CS, Wang C, Hu Q, et al. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin Cell Dev Biol: Elsevier, 2019,112–120.
[102]
Xu Y, Gao W, Sun Y, et al. . New insight on microglia activation in neurodegenerative diseases and therapeutics. Front Neurosci, 2023, 17: 1308345.
CrossRef Google scholar
[103]
Cai Z, Hussain MD, Yan LJ. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci, 2014, 124(5): 307-321.
CrossRef Google scholar
[104]
Vepsäläinen S, Helisalmi S, Mannermaa A, et al. . Combined risk effects of IDE and NEP gene variants on Alzheimer disease. J Neurol Neurosurg Psychiatry, 2009, 80(11): 1268-1270.
CrossRef Google scholar
[105]
Park HY, Kim GY, Choi YH. Naringenin attenuates the release of pro-inflammatory mediators from lipopolysaccharide-stimulated BV2 microglia by inactivating nuclear factor-κB and inhibiting mitogen-activated protein kinases. Int J Mol Med, 2012, 30(1): 204-210
[106]
Zhang B, Wei YZ, Wang GQ, et al. . Targeting MAPK pathways by naringenin modulates microglia M1/M2 polarization in lipopolysaccharide-stimulated cultures. Front Cell Neurosci, 2019, 12: 531.
CrossRef Google scholar
[107]
Bai J, Li S, Wu G, et al. . Naringin inhibits lipopolysaccharide-induced activation of microglia cells. Cell Mol Biol (Noisy-le-grand), 2019, 65(5): 38-42.
CrossRef Google scholar
[108]
Qin H, Roberts KL, Niyongere SA, et al. . Molecular mechanism of lipopolysaccharide-induced SOCS-3 gene expression in macrophages and microglia. J Immunol, 2007, 179(9): 5966-5976.
CrossRef Google scholar
[109]
Ahmed NY, Knowles R, Dehorter N. New Insights Into Cholinergic Neuron Diversity. Front Mol Neurosci, 2019, 12: 204.
CrossRef Google scholar
[110]
Darvesh S, Hopkins DA, Geula C. Neurobiology of butyrylcholinesterase. Nat Rev Neurosci, 2003, 4(2): 131-138.
CrossRef Google scholar
[111]
Hampel H, Mesulam MM, Cuello AC, et al. . The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain, 2018, 141(7): 1917-1933.
CrossRef Google scholar
[112]
Goyal A, Verma A, Dubey N, et al. . Naringenin: A prospective therapeutic agent for Alzheimer’s and Parkinson’s disease. J Food Biochem, 2022, 46(12): e14415.
CrossRef Google scholar
[113]
Al-Rajhi AMH, Qanash H, Almashjary MN, et al. . Anti-Helicobacter pylori, Antioxidant, Antidiabetic, and Anti-Alzheimer’s Activities of Laurel Leaf Extract Treated by Moist Heat and Molecular Docking of Its Flavonoid Constituent, Naringenin, against Acetylcholinesterase and Butyrylcholinesterase. Life (Basel), 2023, 13(7): 1512
[114]
Remya C, Dileep KV, Tintu I, et al. . Flavanone glycosides as acetylcholinesterase inhibitors: computational and experimental evidence. Indian J Pharm Sci, 2014, 76(6): 567-570.
[115]
Zaki HF, Abd-El-Fattah MA, Attia AS. Naringenin protects against scopolamine-induced dementia in rats. Bulletin of Faculty of Pharmacy, Cairo University, 2014, 52(1): 15-25.
CrossRef Google scholar
[116]
Mahdavinia M, Ahangarpour A, Zeidooni L, et al. . Protective effect of naringin on bisphenol A-induced cognitive dysfunction and oxidative damage in rats. Int J Mol Cell Med, 2019, 8(2): 141-153.
[117]
Sachdeva AK, Chopra K. Naringin mitigate okadaic acid-induced cognitive impairment in an experimental paradigm of Alzheimer’s disease. J Funct Foods, 2015, 19: 110-125.
CrossRef Google scholar
[118]
Ionescu-Tucker A, Cotman CW. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol Aging, 2021, 107: 86-95.
CrossRef Google scholar
[119]
Sies H, Berndt C, Jones DP. Oxidative Stress. Annu Rev Biochem, 2017, 86(1): 715-748.
CrossRef Google scholar
[120]
Andersen JK. Oxidative stress in neurodegeneration: cause or consequence?. Nat Med, 2004, 10(Suppl7): S18-S25.
CrossRef Google scholar
[121]
Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev, 2013, 2013: 316523.
CrossRef Google scholar
[122]
Chen R, Qi QL, Wang MT, et al. . Therapeutic potential of naringin: an overview. Pharm Biol, 2016, 54(12): 3203-3210.
CrossRef Google scholar
[123]
Rajadurai M, Prince PS. Preventive effect of naringin on isoproterenol-induced cardiotoxicity in Wistar rats: an in vivo and in vitro study. Toxicology, 2007, 232(3): 216-225.
CrossRef Google scholar
[124]
Cavia-Saiz M, Busto MD, Pilar-Izquierdo MC, et al. . Antioxidant properties, radical scavenging activity and biomolecule protection capacity of flavonoid naringenin and its glycoside naringin: a comparative study. J Sci Food Agric, 2010, 90(7): 1238-1244.
CrossRef Google scholar
[125]
Muthaiah VP, Venkitasamy L, Michael FM, et al. . Neuroprotective role of naringenin on carbaryl induced neurotoxicity in mouse neuroblastoma cells. Pharmacol Pharmacother, 2013, 4(3): 192-197.
CrossRef Google scholar
[126]
de Oliveira MR, Custódio de Souza IC, Fürstenau CR. Promotion of mitochondrial protection by naringenin in methylglyoxal-treated SH-SY5Y cells: Involvement of the Nrf2/GSH axis. Chem Biol Interact, 2019, 310: 108728.
CrossRef Google scholar
[127]
de Oliveira MR, Brasil FB, Andrade CMB. Naringenin Attenuates H2O2-Induced Mitochondrial Dysfunction by an Nrf2-Dependent Mechanism in SH-SY5Y Cells. Neurochem Res, 2017, 42(11): 3341-3350.
CrossRef Google scholar
[128]
Kumar A, Dogra C, Prakash A. Protective Effect of Naringin, a Citrus Flavonoid, Against Colchicine-Induced Cognitive Dysfunction and Oxidative Damage in Rats. J Med Food, 2010, 13(4): 976-984.
CrossRef Google scholar
[129]
Hassan HM, Elnagar MR, Abdelrazik E, et al. . Neuroprotective effect of naringin against cerebellar changes in Alzheimer’s disease through modulation of autophagy, oxidative stress and tau expression: An experimental study. Front Neuroanat, 2022, 16: 1012422.
CrossRef Google scholar
[130]
Anstey KJ, Ashby-Mitchell K, Peters R. Updating the Evidence on the Association between Serum Cholesterol and Risk of Late-Life Dementia: Review and Meta-Analysis. Alzheimers Dis, 2017, 56(1): 215-228.
CrossRef Google scholar
[131]
Popp J, Meichsner S, Kölsch H, et al. . Cerebral and extracerebral cholesterol metabolism and CSF markers of Alzheimer’s disease. Biochem Pharmacol, 2013, 86(1): 37-42.
CrossRef Google scholar
[132]
Souza JPF, Povala G, Brum WS, et al. . Hypercholesterolemia accelerates aβ deposition in regions associated with early amyloidosis: Neuroimaging/optimal neuroimaging measures for tracking disease progression. Alzheimers Dement, 2020, 16: e044114.
CrossRef Google scholar
[133]
Raulin AC, Doss SV, Trottier ZA, et al. . ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol Neurodegener, 2022, 17(1): 72.
CrossRef Google scholar
[134]
Nabizadeh F, Valizadeh P, Balabandian M. Does statin use affect amyloid beta deposition and brain metabolism?. CNS Neurosci Ther, 2023, 29(5): 1434-1443.
CrossRef Google scholar
[135]
Chen JM, Chang CW, Chang TH, et al. . Effects of statins on incident dementia in patients with type 2 DM: a population-based retrospective cohort study in Taiwan. PLoS One, 2014, 9(2): e88434.
CrossRef Google scholar
[136]
Taghibiglou C, Martin HG, Lai TW, et al. . Role of NMDA receptor-dependent activation of SREBP1 in excitotoxic and ischemic neuronal injuries. Nat Med, 2009, 15(12): 1399-1406.
CrossRef Google scholar
[137]
Yu X, Meng X, Yan Y, et al. . Extraction of Naringin from Pomelo and Its Therapeutic Potentials against Hyperlipidemia. Molecules, 2022, 27(24): 9033.
CrossRef Google scholar
[138]
Cai X, Wang S, Wang H, et al. . Naringenin inhibits lipid accumulation by activating the AMPK pathway in vivo and vitro. Food Sci Hum Wellness, 2023, 12(4): 1174-1183.
CrossRef Google scholar
[139]
Allister EM, Mulvihill EE, Barrett PH, et al. . Inhibition of apoB secretion from HepG2 cells by insulin is amplified by naringenin, independent of the insulin receptor. J Lipid Res, 2008, 49(10): 2218-2229.
CrossRef Google scholar
[140]
Sui G, Xiao H, Lu X, et al. . Naringin Activates AMPK Resulting in Altered Expression of SREBPs, PCSK9, and LDLR To Reduce Body Weight in Obese C57BL/6J Mice. J Agric Food Chem, 2018, 66(34): 8983-8990.
CrossRef Google scholar
[141]
Choi H, Kim HJ, Yang J, et al. . Acetylation changes tau interactome to degrade tau in Alzheimer’s disease animal and organoid models. Aging Cell, 2020, 19(1): e13081.
CrossRef Google scholar
[142]
Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci, 2016, 17(1): 22-35.
CrossRef Google scholar
[143]
Qiu Q, Lei X, Wang Y, et al. . Naringin Protects against Tau Hyperphosphorylation in Aβ (25–35)-Injured PC12 Cells through Modulation of ER, PI3K/AKT, and GSK-3β Signaling Pathways. Behav Neurol, 2023, 2023: 1857330.
CrossRef Google scholar
[144]
Md S, Gan SY, Haw YH, et al. . in vitro neuroprotective effects of naringenin nanoemulsion against β-amyloid toxicity through the regulation of amyloidogenesis and tau phosphorylation. Int J Biol Macromol, 2018, 118: 1211-1219. Pt A
CrossRef Google scholar
[145]
Kuji M, Becer E, Vatansever HS, et al. . Neuroprotective Effects of Hesperidin and Naringin in SK-N-AS Cell as an In Vitro Model for Alzheimer’s Disease. J Am Nutr Assoc, 2023, 42(4): 418-426
[146]
Jia YR, Guo ZQ, Guo Q, et al. . Glycogen Synthase Kinase-3β, NLRP3 Inflammasome, and Alzheimer’s Disease. Curr Med Sci, 2023, 43(5): 847-854.
CrossRef Google scholar
[147]
Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochem Res, 2007, 32: 577-595.
CrossRef Google scholar
[148]
Iwaloye O, Elekofehinti OO, Oluwarotimi EA, et al. . Insight into glycogen synthase kinase-3β inhibitory activity of phytoconstituents from Melissa officinalis: in silico studies. In Silico Pharmacol, 2020, 8(1): 2.
CrossRef Google scholar
[149]
Abd-Elmawla MA, Essam RM, Ahmed KA, et al. . Implication of Wnt/GSK-3β/β-Catenin Signaling in the Pathogenesis of Mood Disturbances Associated with Hyperthyroidism in Rats: Potential Therapeutic Effect of Naringin. ACS Chem Neurosci, 2023, 14(11): 2035-2048.
CrossRef Google scholar
[150]
Guo X, Ji Q, Wu M, et al. . Naringin attenuates acute myocardial ischemia-reperfusion injury via miR-126/GSK-3β/β-catenin signaling pathway. Acta Cir Bras, 2022, 37(1): e370102.
CrossRef Google scholar
[151]
Huang XT, Liu X, Ye CY, et al. . Iron-induced energy supply deficiency and mitochondrial fragmentation in neurons. J Neurochem, 2018, 147(6): 816-830.
CrossRef Google scholar
[152]
Hare D, Ayton S, Bush A, et al. . A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci, 2013, 5: 34.
CrossRef Google scholar
[153]
Levi S, Finazzi D. Neurodegeneration with brain iron accumulation: update on pathogenic mechanisms. Front Pharmacol, 2014, 5: 99.
CrossRef Google scholar
[154]
Gleason A, Bush AI. Iron and Ferroptosis as Therapeutic Targets in Alzheimer’s Disease. Neurotherapeutics, 2021, 18(1): 252-264.
CrossRef Google scholar
[155]
Shamsi A, Shahwan M, Khan MS, et al. . Elucidating the interaction of human ferritin with quercetin and naringenin: Implication of natural products in neurodegenerative diseases: Molecular docking and dynamics simulation insight. ACS omega, 2021, 6(11): 7922-7930.
CrossRef Google scholar
[156]
Chtourou Y, Fetoui H, Gdoura R. Protective effects of naringenin on iron-overload-induced cerebral cortex neurotoxicity correlated with oxidative stress. Biol Trace Elem Res, 2014, 158(3): 376-383.
CrossRef Google scholar
[157]
Talbot K. Brain insulin resistance in Alzheimer’s disease and its potential treatment with GLP-1 analogs. Neurodegener Dis Manag, 2014, 4(1): 31-40.
CrossRef Google scholar
[158]
Ashton NJ, Leuzy A, Karikari TK, et al. . The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur J Nucl Med Mol Imaging, 2021, 48(7): 2140-2156.
CrossRef Google scholar
[159]
Talbot K, Wang HY, Kazi H, et al. . Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest, 2012, 122(4): 1316-1338.
CrossRef Google scholar
[160]
Rivera EJ, Goldin A, Fulmer N, et al. . Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: link to brain reductions in acetylcholine. J Alzheimers Dis, 2005, 8(3): 247-268.
CrossRef Google scholar
[161]
Wang D, Yan J, Chen J, et al. . Naringin Improves Neuronal Insulin Signaling, Brain Mitochondrial Function, and Cognitive Function in High-Fat Diet-Induced Obese Mice. Cell Mol Neurobiol, 2015, 35(7): 1061-1071.
CrossRef Google scholar
[162]
Wang D, Gao K, Li X, et al. . Long-term naringin consumption reverses a glucose uptake defect and improves cognitive deficits in a mouse model of Alzheimer’s disease. Pharmacol Biochem Behav, 2012, 102(1): 13-20.
CrossRef Google scholar
[163]
Yang W, Ma J, Liu Z, et al. . Effect of naringenin on brain insulin signaling and cognitive functions in ICV-STZ induced dementia model of rats. Neurol Sci, 2014, 35(5): 741-751.
CrossRef Google scholar
[164]
Variya BC, Bakrania AK, Patel SS. Antidiabetic potential of gallic acid from Emblica officinalis: Improved glucose transporters and insulin sensitivity through PPAR-γ and Akt signaling. Phytomedicine, 2020, 73: 152906.
CrossRef Google scholar
[165]
Liu X, Liu M, Mo Y, et al. . Naringin ameliorates cognitive deficits in streptozotocin-induced diabetic rats. Iran J Basic Med Sci, 2016, 19(4): 417-422.
[166]
Amawi H, Ashby CR Jr, Tiwari AK. Cancer chemoprevention through dietary flavonoids: what’s limiting?. Chin J Cancer, 2017, 36(1): 50.
CrossRef Google scholar
[167]
Salehi B, Fokou PVT, Sharifi-Rad M, et al. . The Therapeutic Potential of Naringenin: A Review of Clinical Trials. Pharmaceuticals, 2019, 12(1): 11.
CrossRef Google scholar
[168]
Barajas-Vega JL, Raffoul-Orozco AK, Hernandez-Molina D, et al. . Naringin reduces body weight, plasma lipids and increases adiponectin levels in patients with dyslipidemia. Int J Vitam Nutr Res, 2022, 92(3–4): 292-298.
CrossRef Google scholar
[169]
Jung UJ, Kim HJ, Lee JS, et al. . Naringin supplementation lowers plasma lipids and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic subjects. Clin Nutr, 2003, 22(6): 561-268.
CrossRef Google scholar
[170]
Nouri Z, Fakhri S, El-Senduny FF, et al. . On the Neuroprotective Effects of Naringenin: Pharmacological Targets, Signaling Pathways, Molecular Mechanisms, and Clinical Perspective. Biomolecules, 2019, 9(11): 690.
CrossRef Google scholar
[171]
Bhandari R, Paliwal JK, Kuhad A. Enhanced Bioavailability and Higher Uptake of Brain-Targeted Surface Engineered Delivery System of Naringenin developed as a Therapeutic for Autism Spectrum Disorder. Curr Drug Deliv, 2023, 20(2): 158-182.
CrossRef Google scholar
[172]
Hanna DMF, Youshia J, Fahmy SF, et al. Nose to brain delivery of naringin-loaded chitosan nanoparticles for potential use in oxaliplatin-induced chemobrain in rats: impact on oxidative stress, cGAS/STING and HMGB1/RAGE/TLR2/MYD88 inflammatory axes. Expert Opin Drug Deliv, 2023,1–15
[173]
Yan C, Gu J, Yin S, et al. . Design and preparation of naringenin loaded functional biomimetic nano-drug delivery system for Alzheimer’s disease. J Drug Target, 2024, 32(1): 80-92.
CrossRef Google scholar
[174]
Shi X, Ma R, Lu Y, et al. . iRGD and TGN co-modified PAMAM for multi-targeted delivery of ATO to gliomas. Biochem Biophys Res Commun, 2020, 527(1): 117-123.
CrossRef Google scholar
[175]
Gandhi S, Shastri DH, Shah J, et al. . Nasal Delivery to the Brain: Harnessing Nanoparticles for Effective Drug Transport. Pharmaceutics, 2024, 16(4): 481.
CrossRef Google scholar
[176]
Gaba B, Khan T, Haider MF, et al. . Vitamin E Loaded Naringenin Nanoemulsion via Intranasal Delivery for the Management of Oxidative Stress in a 6-OHDA Parkinson’s Disease Model. Biomed Res Int, 2019, 2019: 2382563.
CrossRef Google scholar
[177]
Nagaraja S, Basavarajappa GM, Karnati RK, et al. . Ion-Triggered In Situ Gelling Nanoemulgel as a Platform for Nose-to-Brain Delivery of Small Lipophilic Molecules. Pharmaceutics, 2021, 13(8): 1216.
CrossRef Google scholar
[178]
Cunha S, Costa CP, Loureiro JA, et al. . Double Optimization of Rivastigmine-Loaded Nanostructured Lipid Carriers (NLC) for Nose-to-Brain Delivery Using the Quality by Design (QbD) Approach: Formulation Variables and Instrumental Parameters. Pharmaceutics, 2020, 12(7): 599.
CrossRef Google scholar
[179]
Qizilbash FF, Ashhar MU, Zafar A, et al. . Thymoquinone-Enriched Naringenin-Loaded Nanostructured Lipid Carrier for Brain Delivery via Nasal Route: In Vitro Prospect and In Vivo Therapeutic Efficacy for the Treatment of Depression. Pharmaceutics, 2022, 14(3): 656.
CrossRef Google scholar

Accesses

Citations

Detail

Sections
Recommended

/