Nutr Res Pract.  2015 Oct;9(5):480-488. 10.4162/nrp.2015.9.5.480.

Protective role of caffeic acid in an Abeta25-35-induced Alzheimer's disease model

Affiliations
  • 1Department of Food Science and Nutrition, and Kimchi Research Institute, Pusan National University, Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 609-735, Korea. ejcho@pusan.ac.kr
  • 2Department of Integrative Plant Science, Chung-Ang University, Seodong-daero 4726, Daedeok-myeon, Anseong 456-756, Korea.

Abstract

BACKGROUND/OBJECTIVES
Alzheimer's disease (AD) is characterized by deficits in memory and cognitive functions. The accumulation of amyloid beta peptide (Abeta) 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 Abeta25-35-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 Abeta25-35-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 Abeta25-35-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 Abeta25-35-induced memory deficits and cognitive impairment through inhibition of lipid peroxidation and NO production.

Keyword

Caffeic acid; Alzheimer's disease; amyloid beta; oxidative stress; nitric oxide

MeSH Terms

Administration, Oral
Alzheimer Disease*
Amyloid beta-Peptides
Animals
Brain
Kidney
Lipid Peroxidation
Liver
Memory
Memory Disorders
Mice
Nitric Oxide
Oxidative Stress
Phenol
Water
Amyloid beta-Peptides
Nitric Oxide
Phenol
Water

Figure

  • Fig. 1 Behavioral experimental schedule for mice injected with Aβ25-35.

  • Fig. 2 Spatial alternation test in the T-maze test. Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~bThe different letters among groups represent significant differences (P < 0.05) by Duncan's multiple range test. * The space perceptive abilities for old and new routes are significantly different as determined by Student's t-test (P < 0.05).

  • Fig. 3 Effect of CA on objective recognition test. Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~cThe different letters among groups represent significant differences (P < 0.05) by Duncan's multiple range test. * The object cognitive abilities for original and novel objects are significantly different as determined by Student's t-test (P < 0.05).

  • Fig. 4 Effect of CA on spatial learning and memory impairment after injection of Aβ25-35 in the Morris water maze test. Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~bThe different letters represent significant differences (P < 0.05) by Duncan's multiple range test.

  • Fig. 5 Effect of CA on memory impairment induced by injection of Aβ25-35 in the Morris water maze test. Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~bThe different letters represent significant differences (P < 0.05) by Duncan's multiple range test.

  • Fig. 6 Effects of CA on the performance of Aβ25-35 treated mice in finding the hidden (A) and exposed (B) platforms in the Morris water maze test. Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~bThe different letters represent significant differences (P < 0.05) by Duncan's multiple range test.

  • Fig. 7 Effect of CA administration on lipid peroxidation in mouse brain (A), kidney (B), and liver (C). Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~dThe different letters represent significant differences (P < 0.05) by Duncan's multiple range test.

  • Fig. 8 Effect of CA administration on Aβ25-35 induced NO production in mouse brain (A), kidney (B), and liver (C). Normal = 0.9% NaCl injection + oral administration of water; Control = Aβ25-35 injection + oral administration of water; CA10 = Aβ25-35 injection + oral administration of CA (10 mg/kg/day); CA50 = Aβ25-35 injection + oral administration of CA (50 mg/kg/day). Values are mean ± SD. a~dThe different letters represent significant differences (P < 0.05) by Duncan's multiple range test.


Reference

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; 190:224–232.
Article
2. Doraiswamy PM. Non-cholinergic strategies for treating and preventing Alzheimer's disease. CNS Drugs. 2002; 16:811–824.
Article
3. Selkoe DJ. Alzheimer's disease: a central role for amyloid. J Neuropathol Exp Neurol. 1994; 53:438–447.
Article
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; 518:140–144.
Article
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; 30:447–450.
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; 65:631–641.
Article
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; 68:2086–2091.
Article
8. Butterfield DA. Beta-amyloid-associated free radical oxidative stress and neurotoxicity: implications for Alzheimer's disease. Chem Res Toxicol. 1997; 10:495–506.
Article
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; 36:1307–1313.
10. Markesbery WR, Carney JM. Oxidative alterations in Alzheimer's disease. Brain Pathol. 1999; 9:133–146.
Article
11. Behl C, Davis J, Cole GM, Schubert D. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun. 1992; 186:944–950.
12. Kim J, Lee HJ, Lee KW. Naturally occurring phytochemicals for the prevention of Alzheimer's disease. J Neurochem. 2010; 112:1415–1430.
Article
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; 13:444–461.
Article
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; 29:1432–1439.
15. Scalbert A, Manach C, Morand C, Rémésy C, Jiménez L. Dietary polyphenols and the prevention of diseases. Crit Rev Food Sci Nutr. 2005; 45:287–306.
Article
16. Touaibia M, Jean-François J, Doiron J. Caffeic acid, a versatile pharmacophore: an overview. Mini Rev Med Chem. 2011; 11:695–713.
Article
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; 357:115–124.
Article
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-1α, and heme oxygenase-1. J Nat Prod. 2011; 74:352–356.
Article
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.
Article
20. Eom TK, Ryu B, Lee JK, Byun HG, Park SJ, Kim SK. β-secretase inhibitory activity of phenolic acid conjugated chitooligosaccharides. J Enzyme Inhib Med Chem. 2013; 28:214–217.
Article
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; 84:257–262.
Article
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; 65:1745–1752.
Article
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; 42:1397–1405.
Article
24. Maurice T, Lockhart BP, Privat A. Amnesia induced in mice by centrally administered β-amyloid peptides involves cholinergic dysfunction. Brain Res. 1996; 706:181–193.
Article
25. Laursen SE, Belknap JK. Intracerebroventricular injections in mice. Some methodological refinements. J Pharmacol Methods. 1986; 16:355–357.
26. Montgomery KC. A test of two explanations of spontaneous alternation. J Comp Physiol Psychol. 1952; 45:287–293.
Article
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; 1:1306–1311.
Article
28. Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984; 11:47–60.
Article
29. Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 1979; 95:351–358.
Article
30. Schmidt HH, Warner TD, Nakane M, Förstermann U, Murad F. Regulation and subcellular location of nitrogen oxide synthases in RAW264.7 macrophages. Mol Pharmacol. 1992; 41:615–624.
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; 43:515–519.
Article
32. Lu P, Mamiya T, Lu L, Mouri A, Ikejima T, Kim HC, Zou LB, Nabeshima T. Xanthoceraside attenuates amyloid β peptide(25-35)-induced learning and memory impairments in mice. Psychopharmacology (Berl). 2012; 219:181–190.
Article
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; 70:474–483.
Article
34. Mattson MP, Begley JG, Mark RJ, Furukawa K. Abeta25-35 induces rapid lysis of red blood cells: contrast with Aβ1-42 and examination of underlying mechanisms. Brain Res. 1997; 771:147–153.
Article
35. Barnham KJ, Masters CL, Bush AI. Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov. 2004; 3:205–214.
Article
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; 16:329–333.
Article
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.
Article
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; 322:339–346.
Article
39. Ono K, Hasegawa K, Naiki H, Yamada M. Anti-amyloidogenic activity of tannic acid and its activity to destabilize Alzheimer's beta-amyloid fibrils in vitro. Biochim Biophys Acta. 2004; 1690:193–202.
Article
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; 87:172–181.
Article
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; 18:1670–1681.
Article
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; 535:146–151.
Article
43. Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci Biobehav Rev. 2002; 26:91–104.
Article
44. Roher AE, Esh CL, Kokjohn TA, Castaño 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.
Article
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; 85:7341–7345.
Article
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; 269:1510–1517.
Article
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; 14:426–432.
Article
48. Cotman CW, Su JH. Mechanisms of neuronal death in Alzheimer's disease. Brain Pathol. 1996; 6:493–506.
Article
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-β-induced Alzheimer model. Food Chem Toxicol. 2013; 53:105–111.
Article
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-β-induced mouse model of Alzheimer's disease. Arch Biol Sci. 2014; 66:1357–1366.
Article
51. Choi YY, Maeda T, Fujii H, Yokozawa T, Kim HY, Cho EJ, Shibamoto T. Oligonol improves memory and cognition under an amyloid β(25-35)-induced Alzheimer's mouse model. Nutr Res. 2014; 34:595–603.
Article
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; 218:390–397.
Article
53. Yan JJ, Cho JY, Kim HS, Kim KL, Jung JS, Huh SO, Suh HW, Kim YH, Song DK. Protection against β-amyloid peptide toxicity in vivo with long-term administration of ferulic acid. Br J Pharmacol. 2001; 133:89–96.
Article
54. Manach C, Scalbert A, Morand C, Rémésy C, Jiménez L. Polyphenols: food sources and bioavailability. Am J Clin Nutr. 2004; 79:727–747.
Article
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; 504:185–189.
Article
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr