DOI QR코드

DOI QR Code

Protective role of caffeic acid in an Aβ25-35-induced Alzheimer's disease model

  • Kim, Ji Hyun (Department of Food Science and Nutrition, and Kimchi Research Institute, Pusan National University) ;
  • Wang, Qian (Department of Food Science and Nutrition, and Kimchi Research Institute, Pusan National University) ;
  • Choi, Ji Myung (Department of Food Science and Nutrition, and Kimchi Research Institute, Pusan National University) ;
  • Lee, Sanghyun (Department of Integrative Plant Science, Chung-Ang University) ;
  • Cho, Eun Ju (Department of Food Science and Nutrition, and Kimchi Research Institute, Pusan National University)
  • Received : 2015.01.27
  • Accepted : 2015.04.14
  • Published : 2015.10.01

Abstract

BACKGROUND/OBJECTIVES: Alzheimer's disease (AD) is characterized by deficits in memory and cognitive functions. The accumulation of amyloid beta peptide ($A{\beta}$) and oxidative stress in the brain are the most common causes of AD. MATERIALS/METHODS: Caffeic acid (CA) is an active phenolic compound that has a variety of pharmacological actions. We studied the protective abilities of CA in an $A{\beta}_{25-35}$-injected AD mouse model. CA was administered at an oral dose of 10 or 50 mg/kg/day for 2 weeks. Behavioral tests including T-maze, object recognition, and Morris water maze were carried out to assess cognitive abilities. In addition, lipid peroxidation and nitric oxide (NO) production in the brain were measured to investigate the protective effect of CA in oxidative stress. RESULTS: In the T-maze and object recognition tests, novel route awareness and novel object recognition were improved by oral administration of CA compared with the $A{\beta}_{25-35}$-injected control group. These results indicate that administration of CA improved spatial cognitive and memory functions. The Morris water maze test showed that memory function was enhanced by administration of CA. In addition, CA inhibited lipid peroxidation and NO formation in the liver, kidney, and brain compared with the $A{\beta}_{25-35}$-injected control group. In particular, CA 50 mg/kg/day showed the stronger protective effect from cognitive impairment than CA 10 mg/kg/day. CONCLUSIONS: The present results suggest that CA improves $A{\beta}_{25-35}$-induced memory deficits and cognitive impairment through inhibition of lipid peroxidation and NO production.

Keywords

References

  1. Tsunekawa H, Noda Y, Mouri A, Yoneda F, Nabeshima T. Synergistic effects of selegiline and donepezil on cognitive impairment induced by amyloid beta (25-35). Behav Brain Res 2008;190:224-32. https://doi.org/10.1016/j.bbr.2008.03.002
  2. Doraiswamy PM. Non-cholinergic strategies for treating and preventing Alzheimer's disease. CNS Drugs 2002;16:811-24. https://doi.org/10.2165/00023210-200216120-00003
  3. Selkoe DJ. Alzheimer's disease: a central role for amyloid. J Neuropathol Exp Neurol 1994;53:438-47. https://doi.org/10.1097/00005072-199409000-00003
  4. Takahata K, Minami A, Kusumoto H, Shimazu S, Yoneda F. Effects of selegiline alone or with donepezil on memory impairment in rats. Eur J Pharmacol 2005;518:140-4. https://doi.org/10.1016/j.ejphar.2005.06.024
  5. Rottkamp CA, Raina AK, Zhu X, Gaier E, Bush AI, Atwood CS, Chevion M, Perry G, Smith MA. Redox-active iron mediates amyloid-beta toxicity. Free Radic Biol Med 2001;30:447-50. https://doi.org/10.1016/S0891-5849(00)00494-9
  6. Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G, Smith MA. Involvement of oxidative stress in Alzheimer disease. J Neuropathol Exp Neurol 2006;65:631-41. https://doi.org/10.1097/01.jnen.0000228136.58062.bf
  7. Avdulov NA, Chochina SV, Igbavboa U, O'Hare EO, Schroeder F, Cleary JP, Wood WG. Amyloid beta-peptides increase annular and bulk fluidity and induce lipid peroxidation in brain synaptic plasma membranes. J Neurochem 1997;68:2086-91.
  8. Butterfield DA. Beta-amyloid-associated free radical oxidative stress and neurotoxicity: implications for Alzheimer's disease. Chem Res Toxicol 1997;10:495-506. https://doi.org/10.1021/tx960130e
  9. Butterfield DA. Amyloid beta-peptide (1-42)-induced oxidative stress and neurotoxicity: implications for neurodegeneration in Alzheimer's disease brain. A review. Free Radic Res 2002;36:1307-13. https://doi.org/10.1080/1071576021000049890
  10. Markesbery WR, Carney JM. Oxidative alterations in Alzheimer's disease. Brain Pathol 1999;9:133-46.
  11. Behl C, Davis J, Cole GM, Schubert D. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun 1992;186:944-50. https://doi.org/10.1016/0006-291X(92)90837-B
  12. Kim J, Lee HJ, Lee KW. Naturally occurring phytochemicals for the prevention of Alzheimer's disease. J Neurochem 2010;112:1415-30. https://doi.org/10.1111/j.1471-4159.2009.06562.x
  13. Butterfield D, Castegna A, Pocernich C, Drake J, Scapagnini G, Calabrese V. Nutritional approaches to combat oxidative stress in Alzheimer's disease. J Nutr Biochem 2002;13:444-61. https://doi.org/10.1016/S0955-2863(02)00205-X
  14. Moridani MY, Scobie H, Jamshidzadeh A, Salehi P, O'Brien PJ. Caffeic acid, chlorogenic acid, and dihydrocaffeic acid metabolism: glutathione conjugate formation. Drug Metab Dispos 2001;29:1432-9.
  15. Scalbert A, Manach C, Morand C, Remesy C, Jimenez L. Dietary polyphenols and the prevention of diseases. Crit Rev Food Sci Nutr 2005;45:287-306. https://doi.org/10.1080/1040869059096
  16. Touaibia M, Jean-Francois J, Doiron J. Caffeic acid, a versatile pharmacophore: an overview. Mini Rev Med Chem 2011;11:695-713. https://doi.org/10.2174/138955711796268750
  17. U Rehman M, Sultana S. Attenuation of oxidative stress, inflammation and early markers of tumor promotion by caffeic acid in Fe-NTA exposed kidneys of Wistar rats. Mol Cell Biochem 2011;357:115-24. https://doi.org/10.1007/s11010-011-0881-7
  18. Roos TU, Heiss EH, Schwaiberger AV, Schachner D, Sroka IM, Oberan T, Vollmar AM, Dirsch VM. Caffeic acid phenethyl ester inhibits PDGF-induced proliferation of vascular smooth muscle cells via activation of p38 MAPK, HIF-$1\alpha$, and heme oxygenase-1. J Nat Prod 2011;74:352-6. https://doi.org/10.1021/np100724f
  19. Scapagnini G, Vasto S, Abraham NG, Caruso C, Zella D, Fabio G. Modulation of Nrf2/ARE pathway by food polyphenols: a nutritional neuroprotective strategy for cognitive and neurodegenerative disorders. Mol Neurobiol 2011;44:192-201. https://doi.org/10.1007/s12035-011-8181-5
  20. Eom TK, Ryu B, Lee JK, Byun HG, Park SJ, Kim SK. $\beta$-secretase inhibitory activity of phenolic acid conjugated chitooligosaccharides. J Enzyme Inhib Med Chem 2013;28:214-7. https://doi.org/10.3109/14756366.2011.629197
  21. Sul D, Kim HS, Lee D, Joo SS, Hwang KW, Park SY. Protective effect of caffeic acid against beta-amyloid-induced neurotoxicity by the inhibition of calcium influx and tau phosphorylation. Life Sci 2009;84:257-62. https://doi.org/10.1016/j.lfs.2008.12.001
  22. Khan KA, Kumar N, Nayak PG, Nampoothiri M, Shenoy RR, Krishnadas N, Rao CM, Mudgal J. Impact of caffeic acid on aluminium chloride-induced dementia in rats. J Pharm Pharmacol 2013;65:1745-52. https://doi.org/10.1111/jphp.12126
  23. Muthaiyah B, Essa MM, Lee M, Chauhan V, Kaur K, Chauhan A. Dietary supplementation of walnuts improves memory deficits and learning skills in transgenic mouse model of Alzheimer's disease. J Alzheimers Dis 2014;42:1397-405. https://doi.org/10.3233/JAD-140675
  24. Maurice T, Lockhart BP, Privat A. Amnesia induced in mice by centrally administered $\beta$-amyloid peptides involves cholinergic dysfunction. Brain Res 1996;706:181-93. https://doi.org/10.1016/0006-8993(95)01032-7
  25. Laursen SE, Belknap JK. Intracerebroventricular injections in mice. Some methodological refinements. J Pharmacol Methods 1986;16:355-7. https://doi.org/10.1016/0160-5402(86)90038-0
  26. Montgomery KC. A test of two explanations of spontaneous alternation. J Comp Physiol Psychol 1952;45:287-93. https://doi.org/10.1037/h0058118
  27. Bevins RA, Besheer J. Object recognition in rats and mice: a one-trial non-matching-to-sample learning task to study 'recognition memory'. Nat Protoc 2006;1:1306-11. https://doi.org/10.1038/nprot.2006.205
  28. Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 1984;11:47-60. https://doi.org/10.1016/0165-0270(84)90007-4
  29. Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979;95:351-8. https://doi.org/10.1016/0003-2697(79)90738-3
  30. Schmidt HH, Warner TD, Nakane M, Forstermann U, Murad F. Regulation and subcellular location of nitrogen oxide synthases in RAW264.7 macrophages. Mol Pharmacol 1992;41:615-24.
  31. Bachman DL, Wolf PA, Linn RT, Knoefel JE, Cobb JL, Belanger AJ, White LR, D'Agostino RB. Incidence of dementia and probable Alzheimer's disease in a general population: the Framingham Study. Neurology 1993;43:515-9. https://doi.org/10.1212/WNL.43.3_Part_1.515
  32. Lu P, Mamiya T, Lu L, Mouri A, Ikejima T, Kim HC, Zou LB, Nabeshima T. Xanthoceraside attenuates amyloid $\beta$ peptide(25-35)-induced learning and memory impairments in mice. Psychopharmacology (Berl) 2012;219:181-90. https://doi.org/10.1007/s00213-011-2386-1
  33. Kubo T, Nishimura S, Kumagae Y, Kaneko I. In vivo conversion of racemized beta-amyloid ([D-Ser 26]A beta 1-40) to truncated and toxic fragments ([D-Ser 26]A beta 25-35/40) and fragment presence in the brains of Alzheimer's patients. J Neurosci Res 2002;70:474-83. https://doi.org/10.1002/jnr.10391
  34. Mattson MP, Begley JG, Mark RJ, Furukawa K. Abeta25-35 induces rapid lysis of red blood cells: contrast with A$\beta$1-42 and examination of underlying mechanisms. Brain Res 1997;771:147-53. https://doi.org/10.1016/S0006-8993(97)00824-X
  35. Barnham KJ, Masters CL, Bush AI. Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov 2004;3:205-14. https://doi.org/10.1038/nrd1330
  36. Wypijewska A, Galazka-Friedman J, Bauminger ER, Wszolek ZK, Schweitzer KJ, Dickson DW, Jaklewicz A, Elbaum D, Friedman A. Iron and reactive oxygen species activity in parkinsonian substantia nigra. Parkinsonism Relat Disord 2010;16:329-33. https://doi.org/10.1016/j.parkreldis.2010.02.007
  37. Choi SJ, Kim MJ, Heo HJ, Kim JK, Jun WJ, Kim HK, Kim EK, Kim MO, Cho HY, Hwang HJ, Kim YJ, Shin DH. Ameliorative effect of 1,2-benzenedicarboxylic acid dinonyl ester against amyloid beta peptide-induced neurotoxicity. Amyloid 2009;16:15-24. https://doi.org/10.1080/13506120802676997
  38. Salah N, Miller NJ, Paganga G, Tijburg L, Bolwell GP, Rice-Evans C. Polyphenolic flavanols as scavengers of aqueous phase radicals and as chain-breaking antioxidants. Arch Biochem Biophys 1995;322:339-46. https://doi.org/10.1006/abbi.1995.1473
  39. Ono K, Hasegawa K, Naiki H, Yamada M. Anti-amyloidogenic activity of tannic acid and its activity to destabilize Alzheimer's betaamyloid fibrils in vitro. Biochim Biophys Acta 2004;1690:193-202. https://doi.org/10.1016/j.bbadis.2004.06.008
  40. Ono K, Yoshiike Y, Takashima A, Hasegawa K, Naiki H, Yamada M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: implications for the prevention and therapeutics of Alzheimer's disease. J Neurochem 2003;87:172-81.
  41. Chung TW, Moon SK, Chang YC, Ko JH, Lee YC, Cho G, Kim SH, Kim JG, Kim CH. Novel and therapeutic effect of caffeic acid and caffeic acid phenyl ester on hepatocarcinoma cells: complete regression of hepatoma growth and metastasis by dual mechanism. FASEB J 2004;18:1670-81. https://doi.org/10.1096/fj.04-2126com
  42. Huang Y, Jin M, Pi R, Zhang J, Chen M, Ouyang Y, Liu A, Chao X, Liu P, Liu J, Ramassamy C, Qin J. Protective effects of caffeic acid and caffeic acid phenethyl ester against acrolein-induced neurotoxicity in HT22 mouse hippocampal cells. Neurosci Lett 2013;535:146-51. https://doi.org/10.1016/j.neulet.2012.12.051
  43. Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci Biobehav Rev 2002;26:91-104. https://doi.org/10.1016/S0149-7634(01)00041-0
  44. Roher AE, Esh CL, Kokjohn TA, Castano EM, Van Vickle GD, Kalback WM, Patton RL, Luehrs DC, Daugs ID, Kuo YM, Emmerling MR, Soares H, Quinn JF, Kaye J, Connor DJ, Silverberg NB, Adler CH, Seward JD, Beach TG, Sabbagh MN. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer's disease. Alzheimers Dement 2009;5:18-29. https://doi.org/10.1016/j.jalz.2008.10.004
  45. Selkoe DJ, Podlisny MB, Joachim CL, Vickers EA, Lee G, Fritz LC, Oltersdorf T. Beta-amyloid precursor protein of Alzheimer disease occurs as 110- to 135-kilodalton membrane-associated proteins in neural and nonneural tissues. Proc Natl Acad Sci U S A 1988;85:7341-5. https://doi.org/10.1073/pnas.85.19.7341
  46. Sandbrink R, Masters CL, Beyreuther K. Beta A4-amyloid protein precursor mRNA isoforms without exon 15 are ubiquitously expressed in rat tissues including brain, but not in neurons. J Biol Chem 1994;269:1510-7.
  47. Butterfield DA, Boyd-Kimball D. Amyloid beta-peptide(1-42) contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol 2004;14:426-32.
  48. Cotman CW, Su JH. Mechanisms of neuronal death in Alzheimer's disease. Brain Pathol 1996;6:493-506. https://doi.org/10.1111/j.1750-3639.1996.tb00878.x
  49. Choi JY, Cho EJ, Lee HS, Lee JM, Yoon YH, Lee S. Tartary buckwheat improves cognition and memory function in an in vivo amyloid-$\beta$-induced Alzheimer model. Food Chem Toxicol 2013;53:105-11. https://doi.org/10.1016/j.fct.2012.11.002
  50. Lee AY, Yamabe N, Kang KS, Kim HY, Lee S, Cho EJ. Cognition and memory function of Taraxacum coreanum in an in vivo amyloid-$\beta$-induced mouse model of Alzheimer's disease. Arch Biol Sci 2014;66:1357-66. https://doi.org/10.2298/ABS1404357L
  51. Choi YY, Maeda T, Fujii H, Yokozawa T, Kim HY, Cho EJ, Shibamoto T. Oligonol improves memory and cognition under an amyloid $\beta$ (25-35)-induced Alzheimer's mouse model. Nutr Res 2014;34:595-603. https://doi.org/10.1016/j.nutres.2014.06.008
  52. Soliman KF, Mazzio EA. In vitro attenuation of nitric oxide production in C6 astrocyte cell culture by various dietary compounds. Proc Soc Exp Biol Med 1998;218:390-7. https://doi.org/10.3181/00379727-218-44309
  53. Yan JJ, Cho JY, Kim HS, Kim KL, Jung JS, Huh SO, Suh HW, Kim YH, Song DK. Protection against $\beta$-amyloid peptide toxicity in vivo with long-term administration of ferulic acid. Br J Pharmacol 2001;133:89-96. https://doi.org/10.1038/sj.bjp.0704047
  54. Manach C, Scalbert A, Morand C, Remesy C, Jimenez L. Polyphenols: food sources and bioavailability. Am J Clin Nutr 2004;79:727-47. https://doi.org/10.1093/ajcn/79.5.727
  55. National Institute of Food and Drug Safety Evaluation (KR). Tox-Info. Caffeic acid. [internet]. Cheongju: National Institute of Food and Drug Safety Evaluation; 2015 [cited 2015 March 22]. Available from: http://www.nifds.go.kr/toxinfo/index.
  56. Shinomiya K, Omichi J, Ohnishi R, Ito H, Yoshida T, Kamei C. Effects of chlorogenic acid and its metabolites on the sleep-wakefulness cycle in rats. Eur J Pharmacol 2004;504:185-9. https://doi.org/10.1016/j.ejphar.2004.09.054

Cited by

  1. Antioxidative Properties of Asparagus cochinchinensis Root vol.45, pp.4, 2016, https://doi.org/10.3746/jkfn.2016.45.4.524
  2. The Polyherbal Wattana Formula Displays Anti-Amyloidogenic Properties by Increasing α-Secretase Activities vol.12, pp.1, 2017, https://doi.org/10.1371/journal.pone.0170360
  3. SOD3 Ameliorates Aβ25–35-Induced Oxidative Damage in SH-SY5Y Cells by Inhibiting the Mitochondrial Pathway vol.37, pp.3, 2017, https://doi.org/10.1007/s10571-016-0390-z
  4. by Near-infrared Spectroscopy vol.50, pp.16, 2017, https://doi.org/10.1080/00032719.2017.1307384
  5. Quantitative determination, principal component analysis and discriminant analysis of eight marker compounds in crude and sweated Dipsaci Radix by HPLC-DAD vol.55, pp.1, 2017, https://doi.org/10.1080/13880209.2017.1297469
  6. Design, Synthesis, and Biological Evaluation of Novel Tetramethylpyrazine Derivatives as Potential Neuroprotective Agents vol.65, pp.1, 2017, https://doi.org/10.1248/cpb.c16-00699
  7. Nutritional and Pharmacological Strategies to Regulate Microglial Polarization in Cognitive Aging and Alzheimer’s Disease vol.9, pp.1663-4365, 2017, https://doi.org/10.3389/fnagi.2017.00175
  8. Essential Roles of Natural Products and Gaseous Mediators on Neuronal Cell Death or Survival vol.17, pp.10, 2016, https://doi.org/10.3390/ijms17101652
  9. ) on Alzheimer's disease vol.42, pp.10, 2018, https://doi.org/10.1039/C7NJ04223J
  10. An outlook on the role of decaffeinated coffee in neurodegenerative diseases pp.1549-7852, 2019, https://doi.org/10.1080/10408398.2018.1550384
  11. Tadalafil improves bladder dysfunction and object recognition in rats with pelvic venous congestion pp.09198172, 2019, https://doi.org/10.1111/iju.13932
  12. Influence of theMelissa officinalisLeaf Extract on Long-Term Memory in Scopolamine Animal Model with Assessment of Mechanism of Action vol.2016, pp.None, 2015, https://doi.org/10.1155/2016/9729818
  13. Glutamine Improves Oxidative Stress through the Wnt3a/ β -Catenin Signaling Pathway in Alzheimer's Disease In Vitro and In Vivo vol.2019, pp.None, 2015, https://doi.org/10.1155/2019/4690280
  14. Effects of Enteric Environmental Modification by Coffee Components on Neurodegeneration in Rotenone-Treated Mice vol.8, pp.3, 2015, https://doi.org/10.3390/cells8030221
  15. Protective Effects of Active Compounds from Salviae miltiorrhizae Radix against Glutamate-Induced HT-22 Hippocampal Neuronal Cell Death vol.8, pp.8, 2015, https://doi.org/10.3390/pr8080914
  16. Destabilization potential of phenolics on Aβ fibrils: mechanistic insights from molecular dynamics simulation vol.22, pp.35, 2015, https://doi.org/10.1039/d0cp02459g
  17. Evaluation of the effectiveness of the fungicidal effect of drugs against pathogens of cultivated grapes (Vitis vinifera L.) vol.39, pp.None, 2015, https://doi.org/10.1051/bioconf/20213904003
  18. Neuroprotective Effect of Intrastriatal Caffeic Acid Phenethyl Ester Treatment in 6-OH Dopamine Model of Parkinson’s Disease in Rats vol.2021, pp.None, 2015, https://doi.org/10.1155/2021/5553480
  19. Reduced Cerebral Glucose Uptake in an Alzheimer’s Rat Model With Glucose-Weighted Chemical Exchange Saturation Transfer Imaging vol.13, pp.None, 2015, https://doi.org/10.3389/fnagi.2021.618690
  20. Potential of Sorghum Polyphenols to Prevent and Treat Alzheimer’s Disease: A Review Article vol.13, pp.None, 2015, https://doi.org/10.3389/fnagi.2021.729949
  21. 5-Lipoxygenase as an emerging target against age-related brain disorders vol.69, pp.None, 2015, https://doi.org/10.1016/j.arr.2021.101359
  22. Neuroprotective Potential of Mung Bean (Vigna radiata L.) Polyphenols in Alzheimer’s Disease: A Review vol.69, pp.39, 2015, https://doi.org/10.1021/acs.jafc.1c04049
  23. Assessment of lipophilic fluorescence products in β-amyloid-induced cognitive decline: A parallel track in hippocampus, CSF, plasma and erythrocytes vol.157, pp.None, 2015, https://doi.org/10.1016/j.exger.2021.111645
  24. Panax notoginseng saponins protect PC12 cells against Aβ induced injury via promoting parkin-mediated mitophagy vol.285, pp.None, 2022, https://doi.org/10.1016/j.jep.2021.114859