Korean J Physiol Pharmacol.  2016 May;20(3):279-286. 10.4196/kjpp.2016.20.3.279.

Neuroprotective effect of caffeic acid phenethyl ester in 3-nitropropionic acid-induced striatal neurotoxicity

Affiliations
  • 1Department of Pharmaceutical Science and Technology, College of Health and Medical Science, Catholic University of Daegu, Gyeongsan 38430, Korea. tiana@cu.ac.kr
  • 2Department of Physiology, College of Medicine, Dankook University, Cheonan 31116, Korea.
  • 3Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.

Abstract

Caffeic acid phenethyl ester (CAPE), derived from honeybee hives, is a bioactive compound with strong antioxidant activity. This study was designed to test the neuroprotective effect of CAPE in 3-nitropropionic acid (3NP)-induced striatal neurotoxicity, a chemical model of Huntington's disease (HD). Initially, to test CAPE's antioxidant activity, a 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulfonic acid (ABTS) antioxidant assay was employed, and CAPE showed a strong direct radical-scavenging eff ect. In addition, CAPE provided protection from 3NP-induced neuronal cell death in cultured striatal neurons. Based on these observations, the in vivo therapeutic potential of CAPE in 3NP-induced HD was tested. For this purpose, male C57BL/6 mice were repeatedly given 3NP to induce HD-like pathogenesis, and 30 mg/kg of CAPE or vehicle (5% dimethyl sulfoxide and 95% peanut oil) was administered daily. CAPE did not cause changes in body weight, but it reduced mortality by 29%. In addition, compared to the vehicle-treated group, robustly reduced striatal damage was observed in the CAPE-treated animals, and the 3NP-induced behavioral defi cits on the rotarod test were signifi cantly rescued after the CAPE treatment. Furthermore, immunohistochemical data showed that immunoreactivity to glial fibrillary acidic protein (GFAP) and CD45, markers for astrocyte and microglia activation, respectively, were strikingly reduced. Combined, these data unequivocally indicate that CAPE has a strong antioxidant eff ect and can be used as a potential therapeutic agent against HD.

Keyword

3-nitropropionic acid; Antioxidant; Caff eic acid phenethyl ester; Huntington's disease; Striatum

MeSH Terms

Animals
Astrocytes
Body Weight
Cell Death
Dimethyl Sulfoxide
Glial Fibrillary Acidic Protein
Humans
Huntington Disease
Male
Mice
Microglia
Models, Chemical
Mortality
Neurons
Neuroprotective Agents*
Rotarod Performance Test
Urticaria
Dimethyl Sulfoxide
Glial Fibrillary Acidic Protein
Neuroprotective Agents

Figure

  • Fig. 1 Neuroprotective effect of CAPE in cultured striatal neurons.(A) The radical-scavenging activity of CAPE was measured with the ABTS antioxidant assay (n=4 for each group). (B) The neuroprotective effect of CAPE against 3NP intoxication was tested. For this study, 3NP and CAPE were treated and an LDH-release assay was performed two days later (n=4 for each group). Data are presented as mean±SEM, and significance was assessed using one-way analysis of variance (ANOVA). Post-hoc tests were performed using the Bonferroni correction. **p<0.01.

  • Fig. 2 Body-weight change and survival rates after 3NP injection in mice.(A) Body weight was measured at 12-h intervals during the 3NP injections (60, 60, 80, and 80 mg/kg) and daily for the remaining three days. There was no significant difference between the two groups. Data are presented as mean±SEM, and significance was assessed using Student's t-test. (B) The survival rate was 60% in the vehicle-treated group (6 out of 10 mice survived) and 89% in the CAPE-treated group (8 out of 9 mice survived).

  • Fig. 3 Rotarod analysis of 3NP-evoked fall latency.Motor activity was tested three times per day on an accelerating rotarod apparatus (4~40 rpm for 3 min), and the longest latency to fall off the rotarod during the three daily trials was used for analysis. The data from the surviving animals until three days after the last 3NP injection were included (n=6 and n=8 for the vehicle- and CAPE-treated groups, respectively). The data are presented as mean±SEM, and significance was assessed using Student's t-test. *p<0.05.

  • Fig. 4 CAPE reduced 3NP-evoked striatal damage.(A) The mice were perfused three days after the last 3NP injection and striatal damage was assessed with Cresyl Violet staining. The representative image shows distinct striatal damage in a vehicle-treated animal. The scale bar indicates 400 µm. (B) Quantitative analysis of striatal damage in the vehicle- and CAPE-treated groups. Quantitative measurements of striatal damage were analyzed with the χ2 test. *p<0.05.

  • Fig. 5 Astrocyte activation in the striatum.Representative images of GFAP immunoreactivity were shown from vehicle-treated animals (A and a1) and CAPE-treated animals (B and b1). Animals were perfused at 3 days after the last 3NP injection. Size bars are 400 and 50 µm in B and b1, respectively.

  • Fig. 6 Microglia/macrophage activation in the striatum.Representative images of immunoreactivity to CD45, a marker of activated microglia/macrophages, were shown from vehicle-treated animals (A and a1) and CAPE-treated animals (B and b1). Animals were perfused at 3 days after the last 3NP injection. Size bars are 400 and 50 µm in B and b1, respectively.


Reference

1. Gil JM, Rego AC. Mechanisms of neurodegeneration in Huntington's disease. Eur J Neurosci. 2008; 27:2803–2820. PMID: 18588526.
Article
2. Semaka A, Creighton S, Warby S, Hayden MR. Predictive testing for Huntington disease: interpretation and significance of intermediate alleles. Clin Genet. 2006; 70:283–294. PMID: 16965319.
Article
3. Ahmed I, Sbodio JI, Harraz MM, Tyagi R, Grima JC, Albacarys LK, Hubbi ME, Xu R, Kim S, Paul BD, Snyder SH. Huntington's disease: neural dysfunction linked to inositol polyphosphate multikinase. Proc Natl Acad Sci U S A. 2015; 112:9751–9756. PMID: 26195796.
Article
4. Sepers MD, Raymond LA. Mechanisms of synaptic dysfunction and excitotoxicity in Huntington's disease. Drug Discov Today. 2014; 19:990–996. PMID: 24603212.
Article
5. Ueda M, Li S, Itoh M, Hayakawa-Yano Y, Wang MX, Hayakawa M, Hasebe-Matsubara R, Ohta K, Ohta E, Mizuno A, Hida Y, Matsumoto M, Chen H, Nakagawa T. Polyglutamine expansion disturbs the endoplasmic reticulum formation, leading to caspase-7 activation through Bax. Biochem Biophys Res Commun. 2014; 443:1232–1238. PMID: 24388981.
Article
6. Quintanilla RA, Jin YN, von Bernhardi R, Johnson GV. Mitochondrial permeability transition pore induces mitochondria injury in Huntington disease. Mol Neurodegener. 2013; 8:45. PMID: 24330821.
Article
7. Jenkins BG, Koroshetz WJ, Beal MF, Rosen BR. Evidence for impairment of energy metabolism in vivo in Huntington's disease using localized 1H NMR spectroscopy. Neurology. 1993; 43:2689–2695. PMID: 8255479.
8. Feigin A, Leenders KL, Moeller JR, Missimer J, Kuenig G, Spetsieris P, Antonini A, Eidelberg D. Metabolic network abnormalities in early Huntington's disease: an [(18)F]FDG PET study. J Nucl Med. 2001; 42:1591–1595. PMID: 11696626.
9. Costa V, Scorrano L. Shaping the role of mitochondria in the pathogenesis of Huntingtons disease. EMBO J. 2012; 31:1853–1864. PMID: 22446390.
Article
10. Choi YS, Lee B, Cho HY, Reyes IB, Pu XA, Saido TC, Hoyt KR, Obrietan K. CREB is a key regulator of striatal vulnerability in chemical and genetic models of Huntington's disease. Neurobiol Dis. 2009; 36:259–268. PMID: 19632326.
Article
11. Bhat AH, Dar KB, Anees S, Zargar MA, Masood A, Sofi MA, Ganie SA. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed Pharmacother. 2015; 74:101–110. PMID: 26349970.
Article
12. Jung YJ, Suh EC, Lee KE. Oxygen/glucose deprivation and reperfusion cause modifications of postsynaptic morphology and activity in the CA3 area of organotypic hippocampal slice cultures. Korean J Physiol Pharmacol. 2012; 16:423–429. PMID: 23269905.
Article
13. Wang X, Stavchansky S, Bowman PD, Kerwin SM. Cytoprotective effect of caffeic acid phenethyl ester (CAPE) and catechol ring-fluorinated CAPE derivatives against menadione-induced oxidative stress in human endothelial cells. Bioorg Med Chem. 2006; 14:4879–4887. PMID: 16580214.
Article
14. Michaluart P, Masferrer JL, Carothers AM, Subbaramaiah K, Zweifel BS, Koboldt C, Mestre JR, Grunberger D, Sacks PG, Tanabe T, Dannenberg AJ. Inhibitory effects of caffeic acid phenethyl ester on the activity and expression of cyclooxygenase-2 in human oral epithelial cells and in a rat model of inflammation. Cancer Res. 1999; 59:2347–2352. PMID: 10344742.
15. Lee KW, Chun KS, Lee JS, Kang KS, Surh YJ, Lee HJ. Inhibition of cyclooxygenase-2 expression and restoration of gap junction intercellular communication in H-ras-transformed rat liver epithelial cells by caffeic acid phenethyl ester. Ann N Y Acad Sci. 2004; 1030:501–507. PMID: 15659835.
Article
16. Juman S, Yasui N, Ikeda K, Ueda A, Sakanaka M, Negishi H, Miki T. Caffeic acid phenethyl ester suppresses the production of proinflammatory cytokines in hypertrophic adipocytes through lipopolysaccharide-stimulated macrophages. Biol Pharm Bull. 2012; 35:1941–1946. PMID: 23123466.
Article
17. Murtaza G, Karim S, Akram MR, Khan SA, Azhar S, Mumtaz A, Bin Asad MH. Caffeic acid phenethyl ester and therapeutic potentials. Biomed Res Int. 2014; DOI: 10.1155/2014/145342.
Article
18. Kokoszko-Bilska A, Stepniak J, Lewinski A, Karbownik-Lewinska M. Protective antioxidative effects of caffeic acid phenethyl ester (CAPE) in the thyroid and the liver are similar to those caused by melatonin. Thyroid Res. 2014; 7:5. PMID: 25009581.
Article
19. Kassim M, Mansor M, Kamalden TA, Shariffuddin II, Hasan MS, Ong G, Sekaran SD, Suhaimi A, Al-Abd N, Yusoff KM. Caffeic acid phenethyl ester (CAPE): scavenger of peroxynitrite in vitro and in sepsis models. Shock. 2014; 42:154–160. PMID: 24667629.
20. Saulle E, Gubellini P, Picconi B, Centonze D, Tropepi D, Pisani A, Morari M, Marti M, Rossi L, Papa M, Bernardi G, Calabresi P. Neuronal vulnerability following inhibition of mitochondrial complex II: a possible ionic mechanism for Huntington\'s disease. Mol Cell Neurosci. 2004; 25:9–20. PMID: 14962736.
Article
21. Huang QY, Wei C, Yu L, Coelho JE, Shen HY, Kalda A, Linden J, Chen JF. Adenosine A2A receptors in bone marrow-derived cells but not in forebrain neurons are important contributors to 3-nitropropionic acid-induced striatal damage as revealed by cell-type-selective inactivation. J Neurosci. 2006; 26:11371–11378. PMID: 17079665.
Article
22. Lee B, Butcher GQ, Hoyt KR, Impey S, Obrietan K. Activity-dependent neuroprotection and cAMP response element-binding protein (CREB): kinase coupling, stimulus intensity, and temporal regulation of CREB phosphorylation at serine 133. J Neurosci. 2005; 25:1137–1148. PMID: 15689550.
Article
23. Koh JY, Choi DW. Quantitative determination of glutamate mediated cortical neuronal injury in cell culture by lactate dehydrogenase efflux assay. J Neurosci Methods. 1987; 20:83–90. PMID: 2884353.
Article
24. Jones BJ, Roberts DJ. The quantitative measurement of motor incoordination in naive mice using an accelerating rotarod. J Pharm Pharmacol. 1968; 20:302–304. PMID: 4384609.
Article
25. Ranju V, Sathiya S, Kalaivani P, Priya RJ, Saravana Babu C. Memantine exerts functional recovery by improving BDNF and GDNF expression in 3-nitropropionic acid intoxicated mice. Neurosci Lett. 2015; 586:1–7. PMID: 25475686.
Article
26. Lobsiger CS, Cleveland DW. Glial cells as intrinsic components of non-cell-autonomous neurodegenerative disease. Nat Neurosci. 2007; 10:1355–1360. PMID: 17965655.
Article
27. Jang M, Lee MJ, Cho IH. Ethyl pyruvate ameliorates 3-nitropropionic acid-induced striatal toxicity through anti-neuronal cell death and anti-inflammatory mechanisms. Brain Behav Immun. 2014; 38:151–165. PMID: 24576481.
Article
28. Yasui N, Nishiyama E, Juman S, Negishi H, Miki T, Yamori Y, Ikeda K. Caffeic acid phenethyl ester suppresses oxidative stress in 3T3-L1 adipocytes. J Asian Nat Prod Res. 2013; 15:1189–1196. PMID: 23927014.
Article
29. 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. PMID: 23313590.
Article
30. Kurauchi Y, Hisatsune A, Isohama Y, Mishima S, Katsuki H. Caffeic acid phenethyl ester protects nigral dopaminergic neurons via dual mechanisms involving haem oxygenase-1 and brain-derived neurotrophic factor. Br J Pharmacol. 2012; 166:1151–1168. PMID: 22224485.
Article
31. Ilhan A, Iraz M, Gurel A, Armutcu F, Akyol O. Caffeic acid phenethyl ester exerts a neuroprotective effect on CNS against pentylenetetrazol-induced seizures in mice. Neurochem Res. 2004; 29:2287–2292. PMID: 15672552.
Article
32. Eser O, Cosar M, Sahin O, Mollaoglu H, Sezer M, Yaman M, Songur A. The neuroprotective effects of caffeic acid phenethyl ester (CAPE) in the hippocampal formation of cigarette smoke exposed rabbits. Pathology. 2007; 39:433–437. PMID: 17676486.
Article
33. Akyol S, Erdemli HK, Armutcu F, Akyol O. In vitro and in vivo neuroprotective effect of caffeic acid phenethyl ester. J Intercult Ethnopharmacol. 2015; 4:192–193. PMID: 26401406.
Article
34. Kim H, Kim W, Yum S, Hong S, Oh JE, Lee JW, Kwak MK, Park EJ, Na DH, Jung Y. Caffeic acid phenethyl ester activation of Nrf2 pathway is enhanced under oxidative state: structural analysis and potential as a pathologically targeted therapeutic agent in treatment of colonic inflammation. Free Radic Biol Med. 2013; 65:552–562. PMID: 23892357.
Article
35. 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. PMID: 21499987.
Article
36. Zhang H, Davies KJ, Forman HJ. Oxidative stress response and Nrf2 signaling in aging. Free Radic Biol Med. 2015; 88:314–336. PMID: 26066302.
Article
37. Wang W, Wu Y, Zhang G, Fang H, Wang H, Zang H, Xie T, Wang W. Activation of Nrf2-ARE signal pathway protects the brain from damage induced by epileptic seizure. Brain Res. 2014; 1544:54–61. PMID: 24333359.
Article
38. Wang R, Tu J, Zhang Q, Zhang X, Zhu Y, Ma W, Cheng C, Brann DW, Yang F. Genistein attenuates ischemic oxidative damage and behavioral deficits via eNOS/Nrf2/HO-1 signaling. Hippocampus. 2013; 23:634–647. PMID: 23536494.
Article
39. Stack C, Ho D, Wille E, Calingasan NY, Williams C, Liby K, Sporn M, Dumont M, Beal MF. Triterpenoids CDDO-ethyl amide and CDDO-trifluoroethyl amide improve the behavioral phenotype and brain pathology in a transgenic mouse model of Huntington's disease. Free Radic Biol Med. 2010; 49:147–158. PMID: 20338236.
Article
40. Chen YJ, Huang AC, Chang HH, Liao HF, Jiang CM, Lai LY, Chan JT, Chen YY, Chiang J. Caffeic acid phenethyl ester, an antioxidant from propolis, protects peripheral blood mononuclear cells of competitive cyclists against hyperthermal stress. J Food Sci. 2009; 74:H162–H167. PMID: 19723200.
41. Wu W, Lu L, Long Y, Wang T, Liu L, Chen Q, Wang R. Free radical scavenging and antioxidative activities of caffeic acid phenethyl ester (CAPE) and its related compounds in solution and membranes: a structure-activity insight. Food Chem. 2007; 105:107–115.
Article
42. Bacsi A, Woodberry M, Widger W, Papaconstantinou J, Mitra S, Peterson JW, Boldogh I. Localization of superoxide anion production tomitochondrial electron transport chain in 3-NPA-treated cells. Mitochondrion. 2006; 6:235–244. PMID: 17011837.
43. Akashiba H, Ikegaya Y, Nishiyama N, Matsuki N. Differential involvement of cell cycle reactivation between striatal and cortical neurons in cell death induced by 3-nitropropionic acid. J Biol Chem. 2008; 283:6594–6606. PMID: 18182390.
Article
Full Text Links
  • KJPP
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