Therapeutic Potential of Ascorbic Acid in the Management of Alzheimer's Disease: An Update


Cite item

Full Text

Abstract

Background:Ascorbic acid is a potent natural antioxidant that protects against oxidative stress and performs various bodily functions. It is commonly found in fruits and vegetables.

Objective:The manuscript has been written to provide valuable insights into ascorbic acid in managing Alzheimer's disease.

Methods:The data has been gathered from web sources, including PubMed, Science Direct, Publons, Web of Science, and Scopus from 2000-2022 using AA, ascorbic acid, Alzheimer’s diseases, memory, dementia, and antioxidant Keywords.

Results:In the present manuscript, we have summarized the impact of ascorbic acid and its possible mechanism in Alzheimer's disease by, outlining the information currently available on the behavioral and biochemical effects of ascorbic acid in animal models of Alzheimer's disease as well as its usage as a therapeutic agent to slow down the progression of Alzheimer disease in human beings. Oxidative stress plays a significant role in the advancement of AD. AA is a wellknown antioxidant that primarily reduces oxidative stress and produces protein aggregates, which may help decrease cognitive deficits in Alzheimer's disease. The current paper analyses of ascorbic acid revealed that deficiency of ascorbic acid adversely affects the central nervous system and leads to cognitive defects. However, the results of clinical studies are conflicting, but some of the studies suggested that supplementation of ascorbic acid improved cognitive deficits and decreased disease progression.

Conclusion:Based on clinical and preclinical studies, it is observed that ascorbic acid supplementation improves cognitive deficits and protects the neurons from oxidative stress injury

About the authors

Bhupesh Semwal

Department of Pharmacy, Institute of Pharmaceutical Research GLA University

Author for correspondence.
Email: info@benthamscience.net

Bhoopendra Singh

Department of Pharmacy, Institute of Pharmaceutical Research GLA University,

Email: info@benthamscience.net

Yogesh Murti

Department of Pharmacy, Institute of Pharmaceutical Research GLA University

Email: info@benthamscience.net

Sonia Singh

Department of Pharmacy, Institute of Pharmaceutical Research GLA University

Email: info@benthamscience.net

References

  1. Nichols, E.; Steinmetz, J.D.; Vollset, S.E.; Fukutaki, K.; Chalek, J.; Abd-Allah, F.; Abdoli, A.; Abualhasan, A.; Abu-Gharbieh, E.; Akram, T.T.; Al Hamad, H.; Alahdab, F.; Alanezi, F.M.; Alipour, V.; Almustanyir, S.; Amu, H.; Ansari, I.; Arabloo, J.; Ashraf, T.; Astell-Burt, T.; Ayano, G.; Ayuso-Mateos, J.L.; Baig, A.A.; Barnett, A.; Barrow, A.; Baune, B.T.; Béjot, Y.; Bezabhe, W.M.M.; Bezabih, Y.M.; Bhagavathula, A.S.; Bhaskar, S.; Bhattacharyya, K.; Bijani, A.; Biswas, A.; Bolla, S.R.; Boloor, A.; Brayne, C.; Brenner, H.; Burkart, K.; Burns, R.A.; Cámera, L.A.; Cao, C.; Carvalho, F.; Castro-de-Araujo, L.F.S.; Catalá-López, F.; Cerin, E.; Chavan, P.P.; Cherbuin, N.; Chu, D-T.; Costa, V.M.; Couto, R.A.S.; Dadras, O.; Dai, X.; Dandona, L.; Dandona, R.; De la Cruz-Góngora, V.; Dhamnetiya, D.; Dias da Silva, D.; Diaz, D.; Douiri, A.; Edvardsson, D.; Ekholuenetale, M.; El Sayed, I.; El-Jaafary, S.I.; Eskandari, K.; Eskandarieh, S.; Esmaeilnejad, S.; Fares, J.; Faro, A.; Farooque, U.; Feigin, V.L.; Feng, X.; Fereshtehnejad, S-M.; Fernandes, E.; Ferrara, P.; Filip, I.; Fillit, H.; Fischer, F.; Gaidhane, S.; Galluzzo, L.; Ghashghaee, A.; Ghith, N.; Gialluisi, A.; Gilani, S.A.; Glavan, I-R.; Gnedovskaya, E.V.; Golechha, M.; Gupta, R.; Gupta, V.B.; Gupta, V.K.; Haider, M.R.; Hall, B.J.; Hamidi, S.; Hanif, A.; Hankey, G.J.; Haque, S.; Hartono, R.K.; Hasaballah, A.I.; Hasan, M.T.; Hassan, A.; Hay, S.I.; Hayat, K.; Hegazy, M.I.; Heidari, G.; Heidari-Soureshjani, R.; Herteliu, C.; Househ, M.; Hussain, R.; Hwang, B-F.; Iacoviello, L.; Iavicoli, I.; Ilesanmi, O.S.; Ilic, I.M.; Ilic, M.D.; Irvani, S.S.N.; Iso, H.; Iwagami, M.; Jabbarinejad, R.; Jacob, L.; Jain, V.; Jayapal, S.K.; Jayawardena, R.; Jha, R.P.; Jonas, J.B.; Joseph, N.; Kalani, R.; Kandel, A.; Kandel, H.; Karch, A.; Kasa, A.S.; Kassie, G.M.; Keshavarz, P.; Khan, M.A.B.; Khatib, M.N.; Khoja, T.A.M.; Khubchandani, J.; Kim, M.S.; Kim, Y.J.; Kisa, A.; Kisa, S.; Kivimäki, M.; Koroshetz, W.J.; Koyanagi, A.; Kumar, G.A.; Kumar, M.; Lak, H.M.; Leonardi, M.; Li, B.; Lim, S.S.; Liu, X.; Liu, Y.; Logroscino, G.; Lorkowski, S.; Lucchetti, G.; Lutzky Saute, R.; Magnani, F.G.; Malik, A.A.; Massano, J.; Mehndiratta, M.M.; Menezes, R.G.; Meretoja, A.; Mohajer, B.; Mohamed Ibrahim, N.; Mohammad, Y.; Mohammed, A.; Mokdad, A.H.; Mondello, S.; Moni, M.A.A.; Moniruzzaman, M.; Mossie, T.B.; Nagel, G.; Naveed, M.; Nayak, V.C.; Neupane Kandel, S.; Nguyen, T.H.; Oancea, B.; Otstavnov, N.; Otstavnov, S.S.; Owolabi, M.O.; Panda-Jonas, S.; Pashazadeh Kan, F.; Pasovic, M.; Patel, U.K.; Pathak, M.; Peres, M.F.P.; Perianayagam, A.; Peterson, C.B.; Phillips, M.R.; Pinheiro, M.; Piradov, M.A.; Pond, C.D.; Potashman, M.H.; Pottoo, F.H.; Prada, S.I.; Radfar, A.; Raggi, A.; Rahim, F.; Rahman, M.; Ram, P.; Ranasinghe, P.; Rawaf, D.L.; Rawaf, S.; Rezaei, N.; Rezapour, A.; Robinson, S.R.; Romoli, M.; Roshandel, G.; Sahathevan, R.; Sahebkar, A.; Sahraian, M.A.; Sathian, B.; Sattin, D.; Sawhney, M.; Saylan, M.; Schiavolin, S.; Seylani, A.; Sha, F.; Shaikh, M.A.; Shaji, K.S.; Shannawaz, M.; Shetty, J.K.; Shigematsu, M.; Shin, J.I.; Shiri, R.; Silva, D.A.S.; Silva, J.P.; Silva, R.; Singh, J.A.; Skryabin, V.Y.; Skryabina, A.A.; Smith, A.E.; Soshnikov, S.; Spurlock, E.E.; Stein, D.J.; Sun, J.; Tabarés-Seisdedos, R.; Thakur, B.; Timalsina, B.; Tovani-Palone, M.R.; Tran, B.X.; Tsegaye, G.W.; Valadan Tahbaz, S.; Valdez, P.R.; Venketasubramanian, N.; Vlassov, V.; Vu, G.T.; Vu, L.G.; Wang, Y-P.; Wimo, A.; Winkler, A.S.; Yadav, L.; Yahyazadeh Jabbari, S.H.; Yamagishi, K.; Yang, L.; Yano, Y.; Yonemoto, N.; Yu, C.; Yunusa, I.; Zadey, S.; Zastrozhin, M.S.; Zastrozhina, A.; Zhang, Z-J.; Murray, C.J.L.; Vos, T. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: An analysis for the Global Burden of Disease Study 2019. Lancet Public Health, 2022, 7(2), e105-e125. doi: 10.1016/S2468-2667(21)00249-8 PMID: 34998485
  2. Varshney, V.; Garabadu, D. Ang(1–7) exerts Nrf2-mediated neuroprotection against amyloid beta-induced cognitive deficits in rodents. Mol. Biol. Rep., 2021, 48(5), 4319-4331. doi: 10.1007/s11033-021-06447-1 PMID: 34075536
  3. World Health Organization. Global status report on the public health response to dementia. 2021. Available From: https://www.who.int/publications/i/item/9789240033245
  4. Bloom, D.E. 7 billion and counting. Science, 2011, 333(6042), 562-569. doi: 10.1126/science.1209290 PMID: 21798935
  5. Kaitelidou, D.; Kalogeropoulou, M.; Mougias, A.; Galanis, P.; Kontodimopoulos, N.; Pasaloglou, S.; Siskou, O. Socio-economic impact of Alzheimer’s disease in Greece: Pilot Study. Value Health, 2013, 16(7), A545. doi: 10.1016/j.jval.2013.08.1394
  6. Thomas, P.; Lalloué, F.; Preux, P.M.; Hazif-Thomas, C.; Pariel, S.; Inscale, R.; Belmin, J.; Clément, J.P. Dementia patients caregivers quality of life: The PIXEL study. Int. J. Geriatr. Psychiatry, 2006, 21(1), 50-56. doi: 10.1002/gps.1422 PMID: 16323256
  7. Romero-Mas, M.; Ramon-Aribau, A.; Souza, D.L.B.; Cox, A.M.; Gómez-Zúñiga, B. Improving the quality of life of family caregivers of people with Alzheimer’s disease through virtual communities of practice: A quasi-experimental study. Int. J. Alzheimers Dis., 2021, 2021, 1-10. doi: 10.1155/2021/8817491 PMID: 33884204
  8. Sheppard, O.; Coleman, M. Alzheimer’s disease: Etiology, neuropathology and pathogenesis. Exon Publications., 2020, 19, 1-21. PMID: 33400468
  9. Liu, S.; Liu, Y.; Hao, W.; Wolf, L.; Kiliaan, A.J.; Penke, B.; Rübe, C.E.; Walter, J.; Heneka, M.T.; Hartmann, T.; Menger, M.D.; Fassbender, K. TLR2 is a primary receptor for Alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J. Immunol., 2012, 188(3), 1098-1107. doi: 10.4049/jimmunol.1101121 PMID: 22198949
  10. Kametani, F.; Hasegawa, M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front. Neurosci., 2018, 12, 25. doi: 10.3389/fnins.2018.00025 PMID: 29440986
  11. Harrison, F.E.; May, J.M. Vitamin C function in the brain: Vital role of the ascorbate transporter SVCT2. Free Radic. Biol. Med., 2009, 46(6), 719-730. doi: 10.1016/j.freeradbiomed.2008.12.018 PMID: 19162177
  12. Figueroa-Méndez, R.; Rivas-Arancibia, S. Vitamin C in health and disease: Its role in the metabolism of cells and redox state in the brain. Front. Physiol., 2015, 6, 397. doi: 10.3389/fphys.2015.00397 PMID: 26779027
  13. Padayatty, S.J.; Levine, M.; Vitamin, C. The known and the unknown and Goldilocks. Oral Dis., 2016, 22(6), 463-493. doi: 10.1111/odi.12446 PMID: 26808119
  14. Travica, N.; Ried, K.; Sali, A.; Scholey, A.; Hudson, I.; Pipingas, A. Vitamin C status and cognitive function: A systematic review. Nutrients, 2017, 9(9), 960. doi: 10.3390/nu9090960 PMID: 28867798
  15. Harrison, F.; Bowman, G.; Polidori, M. Ascorbic acid and the brain: Rationale for the use against cognitive decline. Nutrients, 2014, 6(4), 1752-1781. doi: 10.3390/nu6041752 PMID: 24763117
  16. Walcher, T.; Haenle, M.M.; Kron, M.; Hay, B.; Mason, R.A.; Walcher, D.; Steinbach, G.; Kern, P.; Piechotowski, I.; Adler, G.; Boehm, B.O.; Koenig, W.; Kratzer, W. Vitamin C supplement use may protect against gallstones: An observational study on a randomly selected population. BMC Gastroenterol., 2009, 9(1), 74. doi: 10.1186/1471-230X-9-74 PMID: 19814821
  17. Gupta, P.; Tiwari, S.; Haria, J. Relationship between depression and vitamin C status: A study on rural patients from western Uttar Pradesh in India. Int. J. Sci. Res., 2014, 1(4), 37-39.
  18. Hansen, S.; Tveden-Nyborg, P.; Lykkesfeldt, J. Does vitamin C deficiency affect cognitive development and function? Nutrients, 2014, 6(9), 3818-3846. doi: 10.3390/nu6093818 PMID: 25244370
  19. Nazari, H.; Heydarpoor, S.; Mohamadi Mofrad, A.; Nazari, Y.; Nazari, A. Effect of vitamin c on serum concentration of brain-derived neurotrophic factor among healthy inactive young men. Neurosci J Shefaye Khatam, 2016, 4(2), 27-32. doi: 10.18869/acadpub.shefa.4.2.27
  20. Nishikimi, M.; Yagi, K. Biochemistry and molecular biology of ascorbic acid biosynthesis. Subcellular Biochemistry; Springer: Cham, 1996. doi: 10.1007/978-1-4613-0325-1_2
  21. Devaki, S.J.; Raveendran, R.L. Vitamin C: Sources, functions, sensing and analysis. Vitamin C; Vitamin, C; Ed.; Intech Open: London, 2017. doi: 10.5772/intechopen.70162
  22. Siqueira, I.R.; Elsner, V.R.; Leite, M.C.; Vanzella, C.; Moysés, F.S.; Spindler, C.; Godinho, G.; Battú, C.; Wofchuk, S.; Souza, D.O.; Gonçalves, C.A.; Netto, C.A. Ascorbate uptake is decreased in the hippocampus of ageing rats. Neurochem. Int., 2011, 58(4), 527-532. doi: 10.1016/j.neuint.2011.01.011 PMID: 21238526
  23. Rice, M.E. Ascorbate regulation and its neuroprotective role in the brain. Trends Neurosci., 2000, 23(5), 209-216. doi: 10.1016/S0166-2236(99)01543-X PMID: 10782126
  24. Ashor, A.W.; Siervo, M.; Mathers, J.C. Vitamin C, antioxidant status, and cardiovascular aging. Molecular basis of nutrition and aging; Academic Press: Massachusetts, 2016, pp. 609-619. doi: 10.1016/B978-0-12-801816-3.00043-1
  25. Ma, N.; Jie, H.; Liu, Y.; Dana, B.; Siegfried, C.J.; Beebe, D.C.; Shui, Y.B. Ascorbic acid related transporters SVCT2 and GLUT1 in human and mouse eyes. Invest. Ophthalmol. Vis. Sci., 2015, 56(7), 4671.
  26. May, J.M. Vitamin C transport and its role in the central nervous system. Subcell. Biochem., 2012, 56, 85-103. doi: 10.1007/978-94-007-2199-9_6 PMID: 22116696
  27. Bowman, G.L.; Dodge, H.; Frei, B.; Calabrese, C.; Oken, B.S.; Kaye, J.A.; Quinn, J.F. Ascorbic acid and rates of cognitive decline in Alzheimer’s disease. J. Alzheimers Dis., 2009, 16(1), 93-98. doi: 10.3233/JAD-2009-0923 PMID: 19158425
  28. Frei, B. Ascorbic acid protects lipids in human plasma and low-density lipoprotein against oxidative damage. Am. J. Clin. Nutr., 1991, 54(6)(Suppl.), 1113S-1118S. doi: 10.1093/ajcn/54.6.1113s PMID: 1962556
  29. Huang, W.J.; Zhang, X.; Chen, W.W. Role of oxidative stress in Alzheimer’s disease. Biomed. Rep., 2016, 4(5), 519-522. doi: 10.3892/br.2016.630 PMID: 27123241
  30. Mosoni, L.; Breuillé, D.; Buffière, C.; Obled, C.; Mirand, P.P. Age-related changes in glutathione availability and skeletal muscle carbonyl content in healthy rats. Exp. Gerontol., 2004, 39(2), 203-210. doi: 10.1016/j.exger.2003.10.014 PMID: 15036413
  31. Qiu, S.; Li, L.; Weeber, E.J.; May, J.M. Ascorbate transport by primary cultured neurons and its role in neuronal function and protection against excitotoxicity. J. Neurosci. Res., 2007, 85(5), 1046-1056. doi: 10.1002/jnr.21204 PMID: 17304569
  32. Lykkesfeldt, J.; Tveden-Nyborg, P. The pharmacokinetics of vitamin C. Nutrients, 2019, 11(10), 2412. doi: 10.3390/nu11102412 PMID: 31601028
  33. Lindblad, M.; Tveden-Nyborg, P.; Lykkesfeldt, J. Regulation of vitamin C homeostasis during deficiency. Nutrients, 2013, 5(8), 2860-2879. doi: 10.3390/nu5082860 PMID: 23892714
  34. Levine, M.; Conry-Cantilena, C.; Wang, Y.; Welch, R.W.; Washko, P.W.; Dhariwal, K.R.; Park, J.B.; Lazarev, A.; Graumlich, J.F.; King, J.; Cantilena, L.R. Vitamin C pharmacokinetics in healthy volunteers: Evidence for a recommended dietary allowance. Proc. Natl. Acad. Sci. USA, 1996, 93(8), 3704-3709. doi: 10.1073/pnas.93.8.3704 PMID: 8623000
  35. Levine, M.; Wang, Y.; Padayatty, S.J.; Morrow, J. A new recommended dietary allowance of vitamin C for healthy young women. Proc. Natl. Acad. Sci. USA, 2001, 98(17), 9842-9846. doi: 10.1073/pnas.171318198 PMID: 11504949
  36. Rajat, S.; Thanawala, S.; Abiraamasundari, R. Pharmacokinetics of a novel sustained-release vitamin C oral tablet: a single dose, randomized, double-blind, placebo-controlled trial. J. Pharmacol. Pharmacother., 2022, 13(2), 167-174. doi: 10.1177/0976500X221111669
  37. Nielsen, T.K.; Højgaard, M.; Andersen, J.T.; Poulsen, H.E.; Lykkesfeldt, J.; Mikines, K.J. Elimination of ascorbic acid after high-dose infusion in prostate cancer patients: A pharmacokinetic evaluation. Basic Clin. Pharmacol. Toxicol., 2015, 116(4), 343-348. doi: 10.1111/bcpt.12323 PMID: 25220574
  38. Carr, A.C.; Lykkesfeldt, J. Discrepancies in global vitamin C recommendations: A review of RDA criteria and underlying health perspectives. Crit. Rev. Food Sci. Nutr., 2021, 61(5), 742-755. doi: 10.1080/10408398.2020.1744513 PMID: 32223303
  39. Padayatty, S.J.; Sun, A.Y.; Chen, Q.; Espey, M.G.; Drisko, J.; Levine, M.; Vitamin, C. Intravenous use by complementary and alternative medicine practitioners and adverse effects. PLoS One, 2010, 5(7), e11414. doi: 10.1371/journal.pone.0011414 PMID: 20628650
  40. Knight, J.; Madduma-Liyanage, K.; Mobley, J.A.; Assimos, D.G.; Holmes, R.P. Ascorbic acid intake and oxalate synthesis. Urolithiasis, 2016, 44(4), 289-297. doi: 10.1007/s00240-016-0868-7 PMID: 27002809
  41. Traxer, O.; Huet, B.; Poindexter, J.; Pak, C.C.; Pearle, M.S. Effect of ascorbic acid consumption on urinary stone risk factors. J. Urol., 2003, 170(2), 397-401. doi: 10.1097/01.ju.0000076001.21606.53 PMID: 12853784
  42. Taylor, E.N.; Stampfer, M.J.; Curhan, G.C. Dietary factors and the risk of incident kidney stones in men: New insights after 14 years of follow-up. J. Am. Soc. Nephrol., 2004, 15(12), 3225-3232. doi: 10.1097/01.ASN.0000146012.44570.20 PMID: 15579526
  43. Quinn, J.; Gerber, B.; Fouche, R.; Kenyon, K.; Blom, Z. Effect of high-dose vitamin C infusion in a glucose-6-phosphate dehydrogenase-deficient patient. Case Rep. Med., 2017, 2017, 5202606.
  44. Pizzino, G.; Irrera, N.; Cucinotta, M.; Palio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxid. Med. Cell. Longev., 2017, 2017, 8416763. doi: 10.1155/2017/8416763
  45. Sharifi-Rad, M.; Anil Kumar, N.V.; Zucca, P.; Varoni, E.M.; Dini, L.; Panzarini, E.; Rajkovic, J.; Tsouh Fokou, P.V.; Azzini, E.; Peluso, I.; Prakash Mishra, A.; Nigam, M.; El Rayess, Y.; Beyrouthy, M.E.; Polito, L.; Iriti, M.; Martins, N.; Martorell, M.; Docea, A.O.; Setzer, W.N.; Calina, D.; Cho, W.C.; Sharifi-Rad, J. Lifestyle, oxidative stress, and antioxidants: Back and forth in the pathophysiology of chronic diseases. Front. Physiol., 2020, 11, 694. doi: 10.3389/fphys.2020.00694 PMID: 32714204
  46. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules, 2019, 24(8), 1583. doi: 10.3390/molecules24081583 PMID: 31013638
  47. Akbar, M.; Essa, M.M.; Daradkeh, G.; Abdelmegeed, M.A.; Choi, Y.; Mahmood, L.; Song, B.J. Mitochondrial dysfunction and cell death in neurodegenerative diseases through nitroxidative stress. Brain Res., 2016, 1637, 34-55. doi: 10.1016/j.brainres.2016.02.016 PMID: 26883165
  48. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol., 2018, 14, 450-464. doi: 10.1016/j.redox.2017.10.014 PMID: 29080524
  49. Zhang, Y.; Thompson, R.; Zhang, H.; Xu, H. APP processing in Alzheimer’s disease. Mol. Brain, 2011, 4(1), 3. doi: 10.1186/1756-6606-4-3 PMID: 21214928
  50. Haass, C.; Kaether, C.; Thinakaran, G.; Sisodia, S. Trafficking and proteolytic processing of APP. Cold Spring Harb. Perspect. Med., 2012, 2(5), a006270. doi: 10.1101/cshperspect.a006270 PMID: 22553493
  51. Phillips, J.C. Why Aβ42 is much more toxic than Aβ40. ACS Chem. Neurosci., 2019, 10(6), 2843-2847. doi: 10.1021/acschemneuro.9b00068 PMID: 31042351
  52. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; Abete, P. Oxidative stress, aging, and diseases. Clin. Interv. Aging, 2018, 13, 757-772. doi: 10.2147/CIA.S158513 PMID: 29731617
  53. Salim, S. Oxidative stress and the central nervous system. J. Pharmacol. Exp. Ther., 2017, 360(1), 201-205. doi: 10.1124/jpet.116.237503 PMID: 27754930
  54. Kregel, K.C.; Zhang, H.J. An integrated view of oxidative stress in aging: Basic mechanisms, functional effects, and pathological considerations. Am. J. Physiol. Regul. Integr. Comp. Physiol., 2007, 292(1), R18-R36. doi: 10.1152/ajpregu.00327.2006 PMID: 16917020
  55. Metcalfe, M.J.; Figueiredo-Pereira, M.E. Relationship between tau pathology and neuroinflammation in Alzheimer’s disease. Mt. Sinai J. Med., 2010, 77(1), 50-58. doi: 10.1002/msj.20163 PMID: 20101714
  56. Karapetyan, G.; Fereshetyan, K.; Harutyunyan, H.; Yenkoyan, K. The synergy of β amyloid 1-42 and oxidative stress in the development of Alzheimer’s disease-like neurodegeneration of hippocampal cells. Sci. Rep., 2022, 12(1), 17883. doi: 10.1038/s41598-022-22761-5 PMID: 36284177
  57. Cole, S.L.; Vassar, R. The Alzheimer’s disease β-secretase enzyme, BACE1. Mol. Neurodegener., 2007, 2(1), 22. doi: 10.1186/1750-1326-2-22 PMID: 18005427
  58. Jo, D.G.; Arumugam, T.V.; Woo, H.N.; Park, J.S.; Tang, S.C.; Mughal, M.; Hyun, D.H.; Park, J.H.; Choi, Y.H.; Gwon, A.R.; Camandola, S.; Cheng, A.; Cai, H.; Song, W.; Markesbery, W.R.; Mattson, M.P. Evidence that γ-secretase mediates oxidative stress-induced β-secretase expression in Alzheimer’s disease. Neurobiol. Aging, 2010, 31(6), 917-925. doi: 10.1016/j.neurobiolaging.2008.07.003 PMID: 18687504
  59. Scherz-Shouval, R.; Elazar, Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem. Sci., 2011, 36(1), 30-38. doi: 10.1016/j.tibs.2010.07.007 PMID: 20728362
  60. Ighodaro, O.M.; Akinloye, O.A. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alex. J. Med., 2018, 54(4), 287-293. doi: 10.1016/j.ajme.2017.09.001
  61. Dong, Y.; Wang, S.; Zhang, T.; Zhao, X.; Liu, X.; Cao, L.; Chi, Z. Ascorbic acid ameliorates seizures and brain damage in rats through inhibiting autophagy. Brain Res., 2013, 1535, 115-123. doi: 10.1016/j.brainres.2013.08.039 PMID: 23994218
  62. Naseer, M.I.; Ullah, N.; Ullah, I.; Koh, P.O.; Lee, H.Y.; Park, M.S.; Kim, M.O. Vitamin C protects against ethanol and PTZ-induced apoptotic neurodegeneration in prenatal rat hippocampal neurons. Synapse, 2011, 65(7), 562-571. doi: 10.1002/syn.20875 PMID: 20963815
  63. Robea, M.A.; Jijie, R.; Nicoara, M.; Plavan, G.; Ciobica, A.S.; Solcan, C.; Audira, G.; Hsiao, C.D.; Strungaru, S.A. Vitamin C attenuates oxidative stress and behavioral abnormalities triggered by fipronil and pyriproxyfen insecticide chronic exposure on zebrafish juvenile. Antioxidants, 2020, 9(10), 944. doi: 10.3390/antiox9100944 PMID: 33019596
  64. Hsueh, Y.J.; Meir, Y.J.J.; Yeh, L.K.; Wang, T.K.; Huang, C.C.; Lu, T.T.; Cheng, C.M.; Wu, W.C.; Chen, H.C. Topical ascorbic acid ameliorates oxidative stress-induced corneal endothelial damage via suppression of apoptosis and autophagic flux blockage. Cells, 2020, 9(4), 943. doi: 10.3390/cells9040943 PMID: 32290365
  65. Chang, B.J.; Jang, B.J.; Son, T.G.; Cho, I.H.; Quan, F.S.; Choe, N.H.; Nahm, S.S.; Lee, J.H. Ascorbic acid ameliorates oxidative damage induced by maternal low-level lead exposure in the hippocampus of rat pups during gestation and lactation. Food Chem. Toxicol., 2012, 50(2), 104-108. doi: 10.1016/j.fct.2011.09.043 PMID: 22056337
  66. Parle, M.; Dhingra, D. Ascorbic Acid: A promising memory-enhancer in mice. J. Pharmacol. Sci., 2003, 93(2), 129-135. doi: 10.1254/jphs.93.129 PMID: 14578579
  67. Rosales-Corral, S.; Tan, D.X.; Reiter, R.J.; Valdivia-Velázquez, M.; Martínez-Barboza, G.; Pablo Acosta-Martínez, J.; Ortiz, G.G. Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid- β peptide in rat brain: A comparative, in vivo study versus vitamin C and E. J. Pineal Res., 2003, 35(2), 80-84. doi: 10.1034/j.1600-079X.2003.00057.x PMID: 12887649
  68. Ballaz, S.; Morales, I.; Rodríguez, M.; Obeso, J.A. Ascorbate prevents cell death from prolonged exposure to glutamate in an in vitro model of human dopaminergic neurons. J. Neurosci. Res., 2013, 91(12), 1609-1617. doi: 10.1002/jnr.23276 PMID: 23996657
  69. Lee, K.H.; Kim, U.J.; Cha, M.; Lee, B.H. Chronic treatment of ascorbic acid leads to age-dependent neuroprotection against oxidative injury in hippocampal slice cultures. Int. J. Mol. Sci., 2021, 22(4), 1608. doi: 10.3390/ijms22041608 PMID: 33562628
  70. Iqbal, K.; Grundke-Iqbal, I. Alzheimer’s disease, a multifactorial disorder seeking multitherapies. Alzheimers Dement., 2010, 6(5), 420-424. doi: 10.1016/j.jalz.2010.04.006 PMID: 20813343
  71. Feng, Y.; Wang, X. Antioxidant therapies for Alzheimer’s disease. Oxid. Med. Cell. Longev., 2012, 2012, 472932. doi: 10.1155/2012/472932
  72. Ahmad, A.; Shah, S.A.; Badshah, H.; Kim, M.J.; Ali, T.; Yoon, G.H.; Kim, T.H.; Abid, N.B.; Rehman, S.U.; Khan, S.; Kim, M.O. Neuroprotection by vitamin C against ethanol-induced neuroinflammation associated neurodegeneration in the developing rat brain. CNS Neurol. Disord. Drug Targets, 2016, 15(3), 360-370. doi: 10.2174/1871527315666151110130139 PMID: 26831257
  73. Moretti, M.; Fraga, D.B.; Rodrigues, A.L.S. Preventive and therapeutic potential of ascorbic acid in neurodegenerative diseases. CNS Neurosci. Ther., 2017, 23(12), 921-929. doi: 10.1111/cns.12767 PMID: 28980404
  74. Alhusaini, A.M.; Fadda, L.M.; Alsharafi, H.; Alshamary, A.F.; Hasan, I.H. L-ascorbic acid and curcumin prevents brain damage induced via lead acetate in rats: Possible mechanisms. Dev. Neurosci., 2022, 44(2), 59-66. doi: 10.1159/000521619 PMID: 34942627
  75. Covarrubias-Pinto, A.; Acuña, A.; Beltrán, F.; Torres-Díaz, L.; Castro, M. Old things new view: Ascorbic acid protects the brain in neurodegenerative disorders. Int. J. Mol. Sci., 2015, 16(12), 28194-28217. doi: 10.3390/ijms161226095 PMID: 26633354
  76. Jha, M.K.; Jeon, S.; Suk, K. Glia as a link between neuroinflammation and neuropathic pain. Immune Netw., 2012, 12(2), 41-47. doi: 10.4110/in.2012.12.2.41 PMID: 22740789
  77. Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener., 2020, 9(1), 42. doi: 10.1186/s40035-020-00221-2 PMID: 33239064
  78. Olajide, O.J.; Yawson, E.O.; Gbadamosi, I.T.; Arogundade, T.T.; Lambe, E.; Obasi, K.; Lawal, I.T.; Ibrahim, A.; Ogunrinola, K.Y. Ascorbic acid ameliorates behavioural deficits and neuropathological alterations in rat model of Alzheimer’s disease. Environ. Toxicol. Pharmacol., 2017, 50, 200-211. doi: 10.1016/j.etap.2017.02.010 PMID: 28192749
  79. Wang, S.F.; Liu, X.; Ding, M.Y.; Ma, S.; Zhao, J.; Wang, Y.; Li, S. 2-O-β-d-glucopyranosyl- -ascorbic acid, a novel vitamin C derivative from Lycium barbarum, prevents oxidative stress. Redox Biol., 2019, 24, 101173. doi: 10.1016/j.redox.2019.101173 PMID: 30903981
  80. Dhingra, D.; Parle, M.; Kulkarni, S.K. Comparative brain cholinesterase-inhibiting activity of Glycyrrhiza glabra, Myristica fragrans, ascorbic acid, and metrifonate in mice. J. Med. Food, 2006, 9(2), 281-283. doi: 10.1089/jmf.2006.9.281 PMID: 16822217
  81. Kara, Y.; Doguc, D.K.; Kulac, E.; Gultekin, F. Acetylsalicylic acid and ascorbic acid combination improves cognition; via antioxidant effect or increased expression of NMDARs and nAChRs? Environ. Toxicol. Pharmacol., 2014, 37(3), 916-927. doi: 10.1016/j.etap.2014.02.019 PMID: 24699240
  82. Huang, J.; May, J.M. Ascorbic acid protects SH-SY5Y neuroblastoma cells from apoptosis and death induced by β-amyloid. Brain Res., 2006, 1097(1), 52-58. doi: 10.1016/j.brainres.2006.04.047 PMID: 16725131
  83. Harrison, F.E.; May, J.M.; McDonald, M.P. Vitamin C deficiency increases basal exploratory activity but decreases scopolamine-induced activity in APP/PSEN1 transgenic mice. Pharmacol. Biochem. Behav., 2010, 94(4), 543-552. doi: 10.1016/j.pbb.2009.11.009 PMID: 19941887
  84. Murakami, K.; Murata, N.; Ozawa, Y.; Kinoshita, N.; Irie, K.; Shirasawa, T.; Shimizu, T. Vitamin C restores behavioral deficits and amyloid-β oligomerization without affecting plaque formation in a mouse model of Alzheimer’s disease. J. Alzheimers Dis., 2011, 26(1), 7-18. doi: 10.3233/JAD-2011-101971 PMID: 21558647
  85. Kook, S-Y.; Lee, K-M.; Kim, Y.; Cha, M-Y.; Kang, S.; Baik, S.H.; Lee, H.; Park, R.; Mook-Jung, I. High-dose of vitamin C supplementation reduces amyloid plaque burden and ameliorates pathological changes in the brain of 5XFAD mice. Cell Death Dis., 2014, 5(2), e1083. doi: 10.1038/cddis.2014.26 PMID: 24577081
  86. Tveden-Nyborg, P.; Johansen, L.K.; Raida, Z.; Villumsen, C.K.; Larsen, J.O.; Lykkesfeldt, J. Vitamin C deficiency in early postnatal life impairs spatial memory and reduces the number of hippocampal neurons in guinea pigs. Am. J. Clin. Nutr., 2009, 90(3), 540-546. doi: 10.3945/ajcn.2009.27954 PMID: 19640959
  87. Hansen, S.; Jørgensen, J.; Nyengaard, J.; Lykkesfeldt, J.; Tveden-Nyborg, P. Early life vitamin C deficiency does not alter morphology of hippocampal CA1 pyramidal neurons or markers of synaptic plasticity in a guinea pig model. Nutrients, 2018, 10(6), 749. doi: 10.3390/nu10060749 PMID: 29890692
  88. Sangeetha, A.; Mohan, S.K.; Kumaresan, M. Chronic administration of vitamin C increases cognitive function in chronic stress-induced rats. Natl. J. Physiol. Pharm. Pharmacol., 2017, 7(11), 1190-1194.
  89. Basambombo, L.L.; Carmichael, P.H.; Côté, S.; Laurin, D. Use of vitamin E and C supplements for the prevention of cognitive decline. Ann. Pharmacother., 2017, 51(2), 118-124. doi: 10.1177/1060028016673072 PMID: 27708183
  90. Scheff, S.; Price, D.A. Synaptic pathology in Alzheimer’s disease: A review of ultrastructural studies. Neurobiol. Aging, 2003, 24(8), 1029-1046. doi: 10.1016/j.neurobiolaging.2003.08.002 PMID: 14643375
  91. Skaper, S.D.; Facci, L.; Zusso, M.; Giusti, P. Synaptic Plasticity, Dementia and Alzheimer Disease. CNS Neurol. Disord. Drug Targets, 2017, 16(3), 220-233. doi: 10.2174/1871527316666170113120853 PMID: 28088900
  92. Sattari, S.; Vaezi, G.; Shahidi, S.; Hojati, V.; Komaki, A. Protective effects of oral vitamin c on memory and learning impairment and attenuation of synaptic plasticity induced by intracerebroventricular injection of beta-amyloid peptide in male rats. Res. Sq., 2021, 1, 587881. doi: 10.21203/rs.3.rs-587881/v1
  93. Heruye, S.H.; Warren, T.J.; Kostansek, J.A., IV; Draves, S.B.; Matthews, S.A.; West, P.J.; Simeone, K.A.; Simeone, T.A. Ascorbic acid reduces neurotransmission, synaptic plasticity, and spontaneous hippocampal rhythms in in vitro slices. Nutrients, 2022, 14(3), 613. doi: 10.3390/nu14030613 PMID: 35276972
  94. Craig, A.; Guest, R.; Tran, Y.; Middleton, J. Cognitive impairment and mood states after spinal cord injury. J. Neurotrauma, 2017, 34(6), 1156-1163. doi: 10.1089/neu.2016.4632 PMID: 27717295
  95. Chen, C; Yang, Q; Ma, X Synergistic effect of ascorbic acid and taurine in the treatment of a spinal cord injury-induced model in rats. 3 Biotech, 2020, 10, 1-8.
  96. Adebiyi, O.; Adigun, K.; David-Odewumi, P.; Akindele, U.; Olayemi, F. Gallic and ascorbic acids supplementation alleviate cognitive deficits and neuropathological damage exerted by cadmium chloride in Wistar rats. Sci. Rep., 2022, 12(1), 14426. doi: 10.1038/s41598-022-18432-0 PMID: 36002551
  97. Singh, N.K.; Garabadu, D. Quercetin exhibits α7nAChR/Nrf2/HO-1-mediated neuroprotection against STZ-induced mitochondrial toxicity and cognitive impairments in experimental rodents. Neurotox. Res., 2021, 39(6), 1859-1879. doi: 10.1007/s12640-021-00410-5 PMID: 34554409
  98. Yamamoto, M.; Kensler, T.W.; Motohashi, H. The KEAP1-NRF2 system: A thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol. Rev., 2018, 98(3), 1169-1203. doi: 10.1152/physrev.00023.2017 PMID: 29717933
  99. He, F.; Ru, X.; Wen, T. NRF2, a transcription factor for stress response and beyond. Int. J. Mol. Sci., 2020, 21(13), 4777. doi: 10.3390/ijms21134777 PMID: 32640524
  100. Jaganjac, M.; Milkovic, L.; Sunjic, S.B.; Zarkovic, N. The NRF2, thioredoxin, and glutathione system in tumorigenesis and anticancer therapies. Antioxidants, 2020, 9(11), 1151. doi: 10.3390/antiox9111151 PMID: 33228209
  101. Gan, L.; Johnson, J.A. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases. Biochim. Biophys. Acta Mol. Basis Dis., 2014, 1842(8), 1208-1218. doi: 10.1016/j.bbadis.2013.12.011 PMID: 24382478
  102. Zhang, L.; Ma, Q.; Zhou, Y. Strawberry leaf extract treatment alleviates cognitive impairment by activating Nrf2/HO-1 signaling in rats with streptozotocin-induced diabetes. Front. Aging Neurosci., 2020, 12, 201. doi: 10.3389/fnagi.2020.00201 PMID: 32792939
  103. Wu, L.; Xu, W.; Li, H.; Dong, B.; Geng, H.; Jin, J.; Han, D.; Liu, H.; Zhu, X.; Yang, Y.; Xie, S. Vitamin C attenuates oxidative stress, inflammation, and apoptosis induced by acute hypoxia through the Nrf2/Keap1 signaling pathway in gibel carp (Carassiusgibelio). Antioxidants, 2022, 11(5), 935. doi: 10.3390/antiox11050935 PMID: 35624798
  104. Majchrzak, D.; Mitter, S.; Elmadfa, I. The effect of ascorbic acid on total antioxidant activity of black and green teas. Food Chem., 2004, 88(3), 447-451. doi: 10.1016/j.foodchem.2004.01.058
  105. Intra, J.; Kuo, S.M. Physiological levels of tea catechins increase cellular lipid antioxidant activity of vitamin C and vitamin E in human intestinal Caco-2 cells. Chem. Biol. Interact., 2007, 169(2), 91-99. doi: 10.1016/j.cbi.2007.05.007 PMID: 17603031
  106. Traber, M.G.; Stevens, J.F. Vitamins C and E: Beneficial effects from a mechanistic perspective. Free Radic. Biol. Med., 2011, 51(5), 1000-1013. doi: 10.1016/j.freeradbiomed.2011.05.017 PMID: 21664268
  107. Zhao, Y.; Pan, Y.; Tang, M.; Lin, W. Blocking p38 signaling reduces the activation of pro-inflammatory cytokines and the phosphorylation of p38 in the habenula and reverses depressive-like behaviors induced by neuroinflammation. Front. Pharmacol., 2018, 9, 511. doi: 10.3389/fphar.2018.00511 PMID: 29867510
  108. Zhang, X.Y.; Xu, Z.P.; Wang, W.; Cao, J.B.; Fu, Q.; Zhao, W.X.; Li, Y.; Huo, X.L.; Zhang, L.M.; Li, Y.F.; Mi, W.D. Vitamin C alleviates LPS-induced cognitive impairment in mice by suppressing neuroinflammation and oxidative stress. Int. Immunopharmacol., 2018, 65, 438-447. doi: 10.1016/j.intimp.2018.10.020 PMID: 30388518
  109. Moretti, M.; Budni, J.; Freitas, A.E.; Neis, V.B.; Ribeiro, C.M.; de Oliveira Balen, G.; Rieger, D.K.; Leal, R.B.; Rodrigues, A.L.S. TNF-α-induced depressive-like phenotype and p38MAPK activation are abolished by ascorbic acid treatment. Eur. Neuropsychopharmacol., 2015, 25(6), 902-912. doi: 10.1016/j.euroneuro.2015.03.006 PMID: 25836357
  110. Radi, A.M.; Mohammed, E.T.; Abushouk, A.I.; Aleya, L.; Abdel-Daim, M.M. The effects of abamectin on oxidative stress and gene expression in rat liver and brain tissues: Modulation by sesame oil and ascorbic acid. Sci. Total Environ., 2020, 701, 134882. doi: 10.1016/j.scitotenv.2019.134882 PMID: 31739238
  111. Xiao, Y.; Su, C.; Zhang, G.; Liang, L.; Jin, T.; Bradley, J.; Ornato, J.P.; Tang, W.; Vitamin, C. Vitamin C improves the outcomes of cardiopulmonary resuscitation and alters shedding of syndecan-1 and p38/MAPK phosphorylation in a rat model. J. Am. Heart Assoc., 2022, 11(7), e023787. doi: 10.1161/JAHA.121.023787 PMID: 35289183
  112. Cárcamo, J.M.; Pedraza, A.; Bórquez-Ojeda, O.; Golde, D.W. Vitamin C suppresses TNF alpha-induced NF kappa B activation by inhibiting I kappa B alpha phosphorylation. Biochemistry, 2002, 41(43), 12995-13002. doi: 10.1021/bi0263210 PMID: 12390026
  113. Cárcamo, J.M.; Pedraza, A.; Bórquez-Ojeda, O.; Zhang, B.; Sanchez, R.; Golde, D.W. Vitamin C is a kinase inhibitor: Dehydroascorbic acid inhibits IkappaBalpha kinase β. Mol. Cell. Biol., 2004, 24(15), 6645-6652. doi: 10.1128/MCB.24.15.6645-6652.2004 PMID: 15254232
  114. Dang, W. The controversial world of sirtuins. Drug Discov. Today. Technol., 2014, 12, e9-e17. doi: 10.1016/j.ddtec.2012.08.003 PMID: 25027380
  115. Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of neuroprotection by quercetin: Counteracting oxidative stress and more. Oxid. Med. Cell. Longev., 2016, 12016, 2986796. doi: 10.1155/2016/2986796
  116. Wei, W.; Li, L.; Zhang, Y. Geriletu; Yang, J.; Zhang, Y.; Xing, Y. Vitamin C protected human retinal pigmented epithelium from oxidant injury depending on regulating SIRT1. ScientificWorldJournal, 2014, 2014, 1-8. doi: 10.1155/2014/750634 PMID: 25147862
  117. Nam, S.; Seo, M.; Seo, J.S.; Rhim, H.; Nahm, S.S.; Cho, I.H.; Chang, B.J.; Kim, H.J.; Choi, S.H.; Nah, S.Y. Ascorbic acid mitigates D-galactose-induced brain aging by increasing hippocampal neurogenesis and improving memory function. Nutrients, 2019, 11(1), 176. doi: 10.3390/nu11010176 PMID: 30650605
  118. Martin, A.; Joseph, J.A.; Cuervo, A.M. Stimulatory effect of vitamin C on autophagy in glial cells. J. Neurochem., 2002, 82(3), 538-549. doi: 10.1046/j.1471-4159.2002.00978.x PMID: 12153478
  119. Chen, Y.; Luo, G.; Yuan, J.; Wang, Y.; Yang, X.; Wang, X.; Li, G.; Liu, Z.; Zhong, N. Vitamin C mitigates oxidative stress and tumor necrosis factor-alpha in severe community-acquired pneumonia and LPS-induced macrophages. Mediators Inflamm., 2014, 2014, 426740.
  120. Huang, Y.N.; Yang, L.Y.; Wang, J.Y.; Lai, C.C.; Chiu, C.T.; Wang, J.Y. L-Ascorbate protects against methamphetamine-induced neurotoxicity of cortical cells via inhibiting oxidative stress, autophagy, and apoptosis. Mol. Neurobiol., 2017, 54(1), 125-136. doi: 10.1007/s12035-015-9561-z PMID: 26732595
  121. Morris, M.C.; Evans, D.A.; Tangney, C.C.; Bienias, J.L.; Wilson, R.S.; Aggarwal, N.T.; Scherr, P.A. Relation of the tocopherol forms to incident Alzheimer disease and to cognitive change. Am. J. Clin. Nutr., 2005, 81(2), 508-514. doi: 10.1093/ajcn.81.2.508 PMID: 15699242
  122. Arlt, S.; Müller-Thomsen, T.; Beisiegel, U.; Kontush, A. Effect of one-year vitamin C- and E-supplementation on cerebrospinal fluid oxidation parameters and clinical course in Alzheimer’s disease. Neurochem. Res., 2012, 37(12), 2706-2714. doi: 10.1007/s11064-012-0860-8 PMID: 22878647
  123. Galasko, D.R.; Peskind, E.; Clark, C.M.; Quinn, J.F.; Ringman, J.M.; Jicha, G.A.; Cotman, C.; Cottrell, B.; Montine, T.J.; Thomas, R.G.; Aisen, P. Antioxidants for Alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol., 2012, 69(7), 836-841. doi: 10.1001/archneurol.2012.85 PMID: 22431837
  124. Ulstein, I.; Bøhmer, T. Normal vitamin levels and nutritional indices in Alzheimer’s disease patients with mild cognitive impairment or dementia with normal body mass indexes. J. Alzheimers Dis., 2016, 55(2), 717-725. doi: 10.3233/JAD-160393 PMID: 27716664
  125. Polidori, M.; Nelles, G. Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease - challenges and perspectives. Curr. Pharm. Des., 2014, 20(18), 3083-3092. doi: 10.2174/13816128113196660706 PMID: 24079767
  126. Li, Y.; Liu, S.; Man, Y.; Li, N.; Zhou, Y. Effects of vitamins E and C combined with β-carotene on cognitive function in the elderly. Exp. Ther. Med., 2015, 9(4), 1489-1493. doi: 10.3892/etm.2015.2274 PMID: 25780457
  127. Travica, N.; Ried, K.; Sali, A.; Hudson, I.; Scholey, A.; Pipingas, A. Plasma vitamin C concentrations and cognitive function: A cross-sectional study. Front. Aging Neurosci., 2019, 11, 72. doi: 10.3389/fnagi.2019.00072 PMID: 31001107
  128. Khodaie, M.; Alibeigi, N.; Mirzaei, V.G. The effect of Ascorbic Acid as supplementary treatment with risperidone in controlling the symptoms of schizophrenia: A double-blind, placebo-controlled clinical trial. J Basic Clin Pathophysiol., 2019, 7(1), 7-14.
  129. Sharma, Y.; Popescu, A.; Horwood, C.; Hakendorf, P.; Thompson, C. Relationship between vitamin C deficiency and cognitive impairment in older hospitalised patients: A cross-sectional study. Antioxidants, 2022, 11(3), 463. doi: 10.3390/antiox11030463 PMID: 35326113
  130. Sim, M.; Hong, S.; Jung, S.; Kim, J.S.; Goo, Y.T.; Chun, W.Y.; Shin, D.M. Vitamin C supplementation promotes mental vitality in healthy young adults: Results from a cross-sectional analysis and a randomized, double-blind, placebo-controlled trial. Eur. J. Nutr., 2022, 61(1), 447-459. doi: 10.1007/s00394-021-02656-3 PMID: 34476568
  131. Lanyau-Domínguez, Y.; Macías-Matos, C.; Jesús, J.; María, G.; Suárez-Medina, R.; Eugenia, M.; Noriega-Fernández, L.; Guerra-Hernández, M.; Calvo-Rodríguez, M.; Sánchez-Gil, Y.; García-Klibanski, M.; Herrera-Javier, D.; Arocha-Oriol, C.; Díaz-Domínguez, M. Levels of vitamins and homocysteine in older adults with Alzheimer disease or mild cognitive impairment in Cuba. MEDICC Rev., 2020, 22(4), 40-47. doi: 10.37757/MR2020.V22.N4.14 PMID: 33295319
  132. Uabundit, N.; Wattanathorn, J.; Mucimapura, S.; Ingkaninan, K. Cognitive enhancement and neuroprotective effects of Bacopa monnieri in Alzheimer’s disease model. J. Ethnopharmacol., 2010, 127(1), 26-31. doi: 10.1016/j.jep.2009.09.056 PMID: 19808086
  133. Akter, F.; Haque, M.; Islam, J.; Rahaman, A.; Bhowmick, S.; Hossain, S. Chronic administration of Curcuma longa extract improves spatial memory-related learning ability in aged rats by inhibiting brain cortico-hippocampal oxidative stress and TNFα. Adv. Alzheimer Dis., 2015, 4(3), 78-89. doi: 10.4236/aad.2015.43008
  134. Lee, J.; Torosyan, N.; Silverman, D.H. Examining the impact of grape consumption on brain metabolism and cognitive function in patients with mild decline in cognition: A double-blinded placebo controlled pilot study. Exp. Gerontol., 2017, 87(Pt A), 121-128. doi: 10.1016/j.exger.2016.10.004 PMID: 27856335
  135. Alaei, H.; Pilehvarian, A.A.; Siahmard, Z.; Reisi, P. The effect of red grape juice on Alzheimer′s disease in rats. Adv. Biomed. Res., 2012, 1(1), 63. doi: 10.4103/2277-9175.100188 PMID: 23326794
  136. Najwa, F.R.; Azrina, A. Comparison of vitamin C content in citrus fruits by titration and high performance liquid chromatography (HPLC) methods. Int. Food Res. J., 2017, 24(2), 726.
  137. Semwal, B.C.; Verma, M.; Murti, Y.; Yadav, H.N. Neuroprotective activity of Sesbania grandifolara seeds extract against celecoxib induced amnesia in mice. Pharmacogn. J., 2018, 10(4), 747-752. doi: 10.5530/pj.2018.4.125
  138. Abu Almaaty, A.H.; Mosaad, R.M.; Hassan, M.K.; Ali, E.H.A.; Mahmoud, G.A.; Ahmed, H.; Anber, N.; Alkahtani, S.; Abdel-Daim, M.M.; Aleya, L.; Hammad, S. Urtica dioica extracts abolish scopolamine-induced neuropathies in rats. Environ. Sci. Pollut. Res. Int., 2021, 28(14), 18134-18145. doi: 10.1007/s11356-020-12025-y PMID: 33405105
  139. Jayachitra, A.; Padma, P.R. Non-enzymic antioxidant activity of Clitoria ternatea leaf extracts in vitro. Biosci. Biotechnol. Res. Asia, 2016, 7(1), 209-218.
  140. Damodaran, T.; Tan, B.W.L.; Liao, P.; Ramanathan, S.; Lim, G.K.; Hassan, Z. Clitoria ternatea L. root extract ameliorated the cognitive and hippocampal long-term potentiation deficits induced by chronic cerebral hypoperfusion in the rat. J. Ethnopharmacol., 2018, 224, 381-390. doi: 10.1016/j.jep.2018.06.020 PMID: 29920356
  141. Tan, M.A.; Sharma, N.; An, S.S.A. Multi-target approach of Murraya koenigii leaves in treating neurodegenerative diseases. Pharmaceuticals (Basel), 2022, 15(2), 188. doi: 10.3390/ph15020188 PMID: 35215300
  142. Valšíková, M.; Mezeyová, I.; Rehuš, M.; Šlosár, M. Changes of vitamin C content in celery and parsley herb after processing. Potravinárstvo, 2016.
  143. Chonpathompikunlert, P.; Boonruamkaew, P.; Sukketsiri, W.; Hutamekalin, P.; Sroyraya, M. The antioxidant and neurochemical activity of Apium graveolens L. and its ameliorative effect on MPTP-induced Parkinson-like symptoms in mice. BMC Complement. Altern. Med., 2018, 18(1), 103. doi: 10.1186/s12906-018-2166-0 PMID: 29558946
  144. Ahmad, L.; Mujahid, M.; Mishra, A.; Rahman, M.A. Protective role of hydroalcoholic extract of Cajanus cajan Linn leaves against memory impairment in sleep deprived experimental rats. J. Ayurveda Integr. Med., 2020, 11(4), 471-477. doi: 10.1016/j.jaim.2018.08.003 PMID: 30661946
  145. Ercisli, S.; Orhan, E. Chemical composition of white (Morus alba), red (Morus rubra) and black (Morus nigra) mulberry fruits. Food Chem., 2007, 103(4), 1380-1384. doi: 10.1016/j.foodchem.2006.10.054
  146. Kaewkaen, P.; Tong-un, T.; Wattanathorn, J.; Muchimapura, S.; Kaewrueng, W.; Wongcharoenwanakit, S. Mulberry fruit extract protects against memory impairment and hippocampal damage in animal model of vascular dementia. Evid. Based Complement. Alternat. Med., 2012, 2012, 1-9. doi: 10.1155/2012/263520 PMID: 22952555
  147. Sadeghi-Aliabadi, H.; Momtazi-borojeni, A.A.; Rabbani, M.; Ghannadi, A.; Abdollahi, E. Cognitive enhancing of pineapple extract and juice in scopolamine-induced amnesia in mice. Res. Pharm. Sci., 2017, 12(3), 257-264. doi: 10.4103/1735-5362.207198 PMID: 28626484
  148. Sarkar, T.; Salauddin, M.; Hazra, S.K.; Chakraborty, R. The impact of raw and differently dried pineapple (Ananas comosus) fortification on the vitamins, organic acid and carotene profile of dairy rasgulla (sweetened cheese ball). Heliyon, 2020, 6(10), e05233. doi: 10.1016/j.heliyon.2020.e05233 PMID: 33102856
  149. Santana, L.F.; Inada, A.C.; Espirito Santo, B.L.S.; Filiú, W.F.O.; Pott, A.; Alves, F.M.; Guimarães, R.C.A.; Freitas, K.C.; Hiane, P.A. Nutraceutical potential of Carica papaya in metabolic syndrome. Nutrients, 2019, 11(7), 1608. doi: 10.3390/nu11071608 PMID: 31315213
  150. Bindhu, K.H.; Vijayalakshmi, A. Neuroprotective effect of Carica papaya leaf extract against aluminium toxicity: An experimental study on cognitive dysfunction and biochemical alterations in rats. Indian J Pharmaceut Edu Res, 2019, 53(3s), s392-s398. doi: 10.5530/ijper.53.3s.111
  151. Chiteva, R.; Wairagu, N. Chemical and nutritional content of Opuntiaficus-indica (L.). Afr. J. Biotechnol., 2013, 12(21)
  152. Han, E.H.; Lim, M.K.; Lee, S.; Lee, S.H.; Yun, S.M.; Yu, H.J.; Ryu, S.H.; Lim, Y.H. Efficacy of ethanolic extract of Opuntiaficus-indica var. saboten stems for improving cognitive function in elderly subjects 55–85 years of age: A randomized, double-blind, placebo-controlled study. J. Med. Food, 2020, 23(11), 1146-1154. doi: 10.1089/jmf.2019.4678 PMID: 33006504
  153. Jang, H.; Srichayet, P.; Park, W.J.; Heo, H.J.; Kim, D.O.; Tongchitpakdee, S.; Kim, T.J.; Jung, S.H.; Lee, C.Y. Phyllanthus emblica L. (Indian gooseberry) extracts protect against retinal degeneration in a mouse model of amyloid beta-induced Alzheimer’s disease. J. Funct. Foods, 2017, 37, 330-338. doi: 10.1016/j.jff.2017.07.056
  154. Kandeda, A.K.; Nguedia, D.; Ayissi, E.R.; Kouamouo, J.; Dimo, T. Ziziphus jujuba (rhamnaceae) alleviates working memory impairment and restores neurochemical alterations in the prefrontal cortex of D-galactose-treated rats. Evid. Based Complement. Alternat. Med., 2021, 2021, 1-15. doi: 10.1155/2021/6610864 PMID: 34194520
  155. Otong, E.S.; Musa, S.A.; Danborno, B.; Sambo, S.J. Adansoniadigitata ameliorates lead-induced memory impairments in rats by reducing glutamate concentration and oxidative stress. Egypt J Basic Appl Sci., 2022, 9(1), 1-0.
  156. Azevêdo, J.C.S.; Borges, K.C.; Genovese, M.I.; Correia, R.T.P.; Vattem, D.A. Neuroprotective effects of dried camu-camu (Myrciaria dubia HBK McVaugh) residue in C. elegans. Food Res. Int., 2015, 73, 135-141. doi: 10.1016/j.foodres.2015.02.015
  157. Li, H.; Lei, T.; Zhang, J.; Yan, Y.; Wang, N.; Song, C.; Li, C.; Sun, M.; Li, J.; Guo, Y.; Yang, J.; Kang, T. Longan (Dimocarpus longan Lour.) Aril ameliorates cognitive impairment in AD mice induced by combination of D-gal/AlCl3 and an irregular diet via RAS/MEK/ERK signaling pathway. J. Ethnopharmacol., 2021, 267, 113612. doi: 10.1016/j.jep.2020.113612 PMID: 33249246
  158. Zhang, S.; Yu, Z.; Sun, L.; Ren, H.; Zheng, X.; Liang, S.; Qi, X. An overview of the nutritional value, health properties, and future challenges of Chinese bayberry. PeerJ, 2022, 10, e13070. doi: 10.7717/peerj.13070 PMID: 35265403
  159. Cho, C.H.; Jung, Y.S.; Kim, J.M.; Nam, T.G.; Lee, S.H.; Cho, H.S.; Song, M.C.; Heo, H.J.; Kim, D.O. Neuroprotective effects of Actinidia eriantha cv. Bidan kiwifruit on amyloid beta-induced neuronal damages in PC-12 cells and ICR mice. J. Funct. Foods, 2021, 79, 104398. doi: 10.1016/j.jff.2021.104398
  160. Jones, J.R.; Lebar, M.D.; Jinwal, U.K.; Abisambra, J.F.; Koren, J., III; Blair, L.; O’Leary, J.C.; Davey, Z.; Trotter, J.; Johnson, A.G.; Weeber, E.; Eckman, C.B.; Baker, B.J.; Dickey, C.A. The diarylheptanoid (+)-aR,11S-myricanol and two flavones from bayberry (Myrica cerifera) destabilize the microtubule-associated protein tau. J. Nat. Prod., 2011, 74(1), 38-44. doi: 10.1021/np100572z PMID: 21141876
  161. Sato, A.; Tagai, N.; Ogino, Y.; Uozumi, H.; Kawakami, S.; Yamamoto, T.; Tanuma, S.; Maruki-Uchida, H.; Mori, S.; Morita, M. Passion fruit seed extract protects beta-amyloid-induced neuronal cell death in a differentiated human neuroblastoma SH‐SY5Y cell model. Food Sci. Nutr., 2022, 10(5), 1461-1468. doi: 10.1002/fsn3.2757 PMID: 35592293
  162. Temviriyanukul, P.; Kittibunchakul, S.; Trisonthi, P.; Kunkeaw, T.; Inthachat, W.; Siriwan, D.; Suttisansanee, U. Mangifera indica ‘Namdokmai’ prevents neuronal cells from amyloid peptide toxicity and inhibits BACE-1 activities in a Drosophila model of Alzheimer’s amyloidosis. Pharmaceuticals (Basel), 2022, 15(5), 591. doi: 10.3390/ph15050591 PMID: 35631418

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2024 Bentham Science Publishers