Generic placeholder image

Current Neurovascular Research

Editor-in-Chief

ISSN (Print): 1567-2026
ISSN (Online): 1875-5739

Research Article

Caffeic Acid Phenethyl Ester (CAPE) Attenuates Paclitaxel-induced Peripheral Neuropathy: A Mechanistic Study

Author(s): Namrata Pramod Kulkarni, Bhupesh Vaidya, Acharan S. Narula and Shyam Sunder Sharma*

Volume 19, Issue 3, 2022

Published on: 19 October, 2022

Page: [293 - 302] Pages: 10

DOI: 10.2174/1567202619666220829104851

Price: $65

Abstract

Background: Chemotherapy-induced peripheral neuropathy is a debilitating pain syndrome produced as a side effect of antineoplastic drugs like paclitaxel. Despite efforts, the currently available therapeutics suffer from serious drawbacks like unwanted side effects and poor efficacy and provide only symptomatic relief. Hence, there is a need to find new therapeutic alternatives for the treatment of chemotherapy-induced peripheral neuropathy.

Objective: The objective of this study was to explore the protective potential of caffeic acid phenethyl ester in paclitaxel-induced neuropathic pain.

Methods: We examined the effects of caffeic acid phenethyl ester by administering paclitaxel (2 mg/kg, intraperitoneal) to female Sprague Dawley rats on four alternate days to induce neuropathic pain, followed by the administration of caffeic acid phenethyl ester (10 and 30 mg/kg, intraperitoneally).

Results: Rats that were administered paclitaxel showed a substantially diminished pain threshold and nerve functions after 28 days. A significantly increased protein expression of Wnt signalling protein (β-catenin), inflammatory marker (matrix metalloproteinase 2) and a decrease in endogenous antioxidant (nuclear factor erythroid 2–related factor 2) levels were found in paclitaxel administered rats in comparison to the naïve control group. Caffeic acid phenethyl ester (10 and 30 mg/kg, intraperitoneal) showed improvements in behavioural and nerve function parameters along with reduced expression of β-catenin, matrix metalloproteinase 2 and an increase in nuclear factor erythroid 2– related factor 2 protein expression.

Conclusion: The present study suggests that caffeic acid phenethyl ester attenuates chemotherapyinduced peripheral neuropathy via inhibition of β-catenin and matrix metalloproteinase 2 and increases nuclear factor erythroid 2–related factor 2 activation.

Keywords: Chemotherapy-induced peripheral neuropathy, paclitaxel, Wnt signalling, β-catenin, MMP2, Nrf2.

[1]
Warwick RA, Hanani M. The contribution of satellite glial cells to chemotherapy-induced neuropathic pain. Eur J Pain 2013; 17(4): 571-80.
[http://dx.doi.org/10.1002/j.1532-2149.2012.00219.x] [PMID: 23065831]
[2]
Woolf CJ, Mannion RJ. Neuropathic pain: Aetiology, symptoms, mechanisms, and management. Lancet 1999; 353(9168): 1959-64.
[http://dx.doi.org/10.1016/S0140-6736(99)01307-0] [PMID: 10371588]
[3]
Otto A, Schmidt C, Luke G, et al. Canonical Wnt signalling induces satellite-cell proliferation during adult skeletal muscle regeneration. J Cell Sci 2008; 121(17): 2939-50.
[http://dx.doi.org/10.1242/jcs.026534] [PMID: 18697834]
[4]
Onyido EK, Sweeney E, Nateri AS. Wnt-signalling pathways and microRNAs network in carcinogenesis: Experimental and bioinformatics approaches. Mol Cancer 2016; 15(1): 56.
[http://dx.doi.org/10.1186/s12943-016-0541-3] [PMID: 27590724]
[5]
Reya T, Duncan AW, Ailles L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 2003; 423(6938): 409-14.
[http://dx.doi.org/10.1038/nature01593] [PMID: 12717450]
[6]
Wodarz A, Nusse R. Mechanisms of Wnt signaling in development. Annu Rev Cell Dev Biol 1998; 14(1): 59-88.
[http://dx.doi.org/10.1146/annurev.cellbio.14.1.59] [PMID: 9891778]
[7]
Huelsken J, Behrens J. The Wnt signalling pathway. J Cell Sci 2002; 115(21): 3977-8.
[http://dx.doi.org/10.1242/jcs.00089] [PMID: 12356903]
[8]
Jin T. The WNT signalling pathway and diabetes mellitus. Diabetologia 2008; 51(10): 1771-80.
[http://dx.doi.org/10.1007/s00125-008-1084-y] [PMID: 18696049]
[9]
Zhang YK, Huang ZJ, Liu S, Liu YP, Song AA, Song XJ. WNT signaling underlies the pathogenesis of neuropathic pain in rodents. J Clin Invest 2013; 123(5): 2268-86.
[http://dx.doi.org/10.1172/JCI65364] [PMID: 23585476]
[10]
Itokazu T, Hayano Y, Takahashi R, Yamashita T. Involvement of Wnt/β-catenin signaling in the development of neuropathic pain. Neurosci Res 2014; 79: 34-40.
[http://dx.doi.org/10.1016/j.neures.2013.12.002] [PMID: 24361264]
[11]
Xu Z, Chen Y, Yu J, et al. TCF4 mediates the maintenance of neuropathic pain through Wnt/β-Catenin signaling following peripheral nerve injury in rats. J Mol Neurosci 2015; 56(2): 397-408.
[http://dx.doi.org/10.1007/s12031-015-0565-y] [PMID: 25963533]
[12]
Feng W, Teng R, Zhao Y, Gao J, Chu H. Epigenetic modulation of Wnt signaling contributes to neuropathic pain in rats. Mol Med Rep 2015; 12(3): 4727-33.
[http://dx.doi.org/10.3892/mmr.2015.3972] [PMID: 26096038]
[13]
Resham K, Sharma SS. Pharmacological interventions targeting Wnt/β-catenin signaling pathway attenuate paclitaxel-induced peripheral neuropathy. Eur J Pharmacol 2019; 864: 172714.
[http://dx.doi.org/10.1016/j.ejphar.2019.172714] [PMID: 31586636]
[14]
Siau C, Xiao W, Bennett G. Paclitaxel- and vincristine-evoked painful peripheral neuropathies: Loss of epidermal innervation and activation of Langerhans cells. Exp Neurol 2006; 201(2): 507-14.
[http://dx.doi.org/10.1016/j.expneurol.2006.05.007] [PMID: 16797537]
[15]
Areti A, Yerra VG, Naidu VGM, Kumar A. Oxidative stress and nerve damage: Role in chemotherapy induced peripheral neuropathy. Redox Biol 2014; 2: 289-95.
[http://dx.doi.org/10.1016/j.redox.2014.01.006] [PMID: 24494204]
[16]
Yardim A, Kandemir FM, Ozdemir S, et al. Quercetin provides protection against the peripheral nerve damage caused by vincristine in rats by suppressing caspase 3, NF-κB, ATF-6 pathways and activating Nrf2, Akt pathways. Neurotoxicology 2020; 81: 137-46.
[http://dx.doi.org/10.1016/j.neuro.2020.10.001] [PMID: 33038355]
[17]
Kaur G, Jaggi AS, Singh N. Exploring the potential effect of Ocimum sanctum in vincristine-induced neuropathic pain in rats. J Brachial Plex Peripher Nerve Inj 2010; 5(1): 3.
[PMID: 20181005]
[18]
Sud’ina GF, Mirzoeva OK, Pushkareva MA, Korshunova GA, Sumbatyan NV, Varfolomeev SD. Caffeic acid phenethyl ester as a lipoxygenase inhibitor with antioxidant properties. FEBS Lett 1993; 329(1-2): 21-4.
[http://dx.doi.org/10.1016/0014-5793(93)80184-V] [PMID: 7689063]
[19]
Cheng H, Zhang Y, Lu W, Gao X, Xu C, Bao H. Caffeic acid phenethyl ester attenuates neuropathic pain by suppressing the p38/NF-κB signal pathway in microglia. J Pain Res 2018; 11: 2709-19.
[http://dx.doi.org/10.2147/JPR.S166274] [PMID: 30464588]
[20]
Russo A, Longo R, Vanella A. Antioxidant activity of propolis: Role of caffeic acid phenethyl ester and galangin. Fitoterapia 2002; 73 (Suppl. 1): S21-9.
[http://dx.doi.org/10.1016/S0367-326X(02)00187-9] [PMID: 12495706]
[21]
Mahmoud NN, Carothers AM, Grunberger D, et al. Plant phenolics decrease intestinal tumors in an animal model of familial adenomatous polyposis. Carcinogenesis 2000; 21(5): 921-7.
[http://dx.doi.org/10.1093/carcin/21.5.921] [PMID: 10783313]
[22]
Chuan G. Effects of caffeic acid phenethyl ester in wnt/β-catenin signaling pathway on human neuroglioma. PhD thesis, Chongqing Medical University, Chongqing, China, 2009.
[23]
Xiang D, Wang D, He Y, et al. Caffeic acid phenethyl ester induces growth arrest and apoptosis of colon cancer cells via the β-catenin/T-cell factor signaling. Anticancer Drugs 2006; 17(7): 753-62.
[http://dx.doi.org/10.1097/01.cad.0000224441.01082.bb] [PMID: 16926625]
[24]
He YJ, Liu BH, Xiang DB, Qiao ZY, Fu T, He YH. Inhibitory effect of caffeic acid phenethyl ester on the growth of SW480 colorectal tumor cells involves β-catenin associated signaling pathway down-regulation. World J Gastroenterol 2006; 12(31): 4981-5.
[http://dx.doi.org/10.3748/wjg.v12.i31.4981] [PMID: 16937493]
[25]
Wang D, Xiang DB, He YJ, et al. Effect of caffeic acid phenethyl ester on proliferation and apoptosis of colorectal cancer cells in vitro. World J Gastroenterol 2005; 11(26): 4008-12.
[http://dx.doi.org/10.3748/wjg.v11.i26.4008] [PMID: 15996024]
[26]
Chen Y, Yang C, Wang ZJ. Proteinase-activated receptor 2 sensitizes transient receptor potential vanilloid 1, transient receptor potential vanilloid 4, and transient receptor potential ankyrin 1 in paclitaxel-induced neuropathic pain. Neuroscience 2011; 193: 440-51.
[http://dx.doi.org/10.1016/j.neuroscience.2011.06.085] [PMID: 21763756]
[27]
Yildiz Y, Serter M, Ek RO, et al. Protective effects of caffeic acid phenethyl ester on intestinal ischemia-reperfusion injury. Dig Dis Sci 2009; 54(4): 738-44.
[http://dx.doi.org/10.1007/s10620-008-0405-9] [PMID: 18683050]
[28]
Ek RO, Serter M, Ergin K, et al. The effects of caffeic acid phenethyl ester (CAPE) on TNBS-induced colitis in ovariectomized rats. Dig Dis Sci 2008; 53(6): 1609-17.
[http://dx.doi.org/10.1007/s10620-007-0056-2] [PMID: 17957471]
[29]
Iraz M. Fadıllıoğlu E, Taşdemir S, Ateş B, Erdoğan S. Dose dependent effects of caffeic acid phenethyl ester on heart rate and blood pressure in rats. Electron J General Med 2005; 2(2): 69-75.
[http://dx.doi.org/10.29333/ejgm/82272]
[30]
Fitzpatrick LR, Wang J, Le T. Caffeic acid phenethyl ester, an inhibitor of nuclear factor-kappaB, attenuates bacterial peptidoglycan polysaccharide-induced colitis in rats. J Pharmacol Exp Ther 2001; 299(3): 915-20.
[PMID: 11714876]
[31]
Hargreaves K, Dubner R, Brown F, Flores C, Joris J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1988; 32(1): 77-88.
[http://dx.doi.org/10.1016/0304-3959(88)90026-7] [PMID: 3340425]
[32]
Joshi RP, Negi G, Kumar A, et al. SNEDDS curcumin formulation leads to enhanced protection from pain and functional deficits associated with diabetic neuropathy: An insight into its mechanism for neuroprotection. Nanomedicine (Lond) 2013; 9(6): 776-85.
[http://dx.doi.org/10.1016/j.nano.2013.01.001] [PMID: 23347896]
[33]
Resham K, Sharma SS. Pharmacologic inhibition of porcupine, disheveled, and β-catenin in Wnt signaling pathway ameliorates diabetic peripheral neuropathy in rats. J Pain 2019; 20(11): 1338-52.
[http://dx.doi.org/10.1016/j.jpain.2019.04.010] [PMID: 31075529]
[34]
Negi G, Kumar A, Kaundal RK, Gulati A, Sharma SS. Functional and biochemical evidence indicating beneficial effect of Melatonin and Nicotinamide alone and in combination in experimental diabetic neuropathy. Neuropharmacology 2010; 58(3): 585-92.
[http://dx.doi.org/10.1016/j.neuropharm.2009.11.018] [PMID: 20005237]
[35]
Negi G, Kumar A, Sharma SS. Nrf2 and NF-κB modulation by sulforaphane counteracts multiple manifestations of diabetic neuropathy in rats and high glucose-induced changes. Curr Neurovasc Res 2011; 8(4): 294-304.
[http://dx.doi.org/10.2174/156720211798120972] [PMID: 22023613]
[36]
Sweitzer SM, Medicherla S, Almirez R, et al. Antinociceptive action of a p38α MAPK inhibitor, SD-282, in a diabetic neuropathy model. Pain 2004; 109(3): 409-19.
[http://dx.doi.org/10.1016/j.pain.2004.02.016] [PMID: 15157702]
[37]
Kumar A, Kaundal RK, Iyer S, Sharma SS. Effects of resveratrol on nerve functions, oxidative stress and DNA fragmentation in experimental diabetic neuropathy. Life Sci 2007; 80(13): 1236-44.
[http://dx.doi.org/10.1016/j.lfs.2006.12.036] [PMID: 17289084]
[38]
Resham K, Khare P, Bishnoi M, Sharma SS. Neuroprotective effects of isoquercitrin in diabetic neuropathy via Wnt/β‐catenin signaling pathway inhibition. Biofactors 2020; 46(3): 411-20.
[http://dx.doi.org/10.1002/biof.1615] [PMID: 31960520]
[39]
Sharma SS, Kumar A, Arora M, Kaundal RK. Neuroprotective potential of combination of resveratrol and 4-amino 1,8 naphthalimide in experimental diabetic neuropathy: Focus on functional, sensorimotor and biochemical changes. Free Radic Res 2009; 43(4): 400-8.
[http://dx.doi.org/10.1080/10715760902801509] [PMID: 19291593]
[40]
Rundquist I, Smith QR, Michel ME, Ask P, Oberg PA, Rapoport SI. Sciatic nerve blood flow measured by laser Doppler flowmetry and [14C]iodoantipyrine. Am J Physiol 1985; 248(3 Pt 2): H311-7.
[PMID: 3156511]
[41]
Al-Brakati A, Alsharif KF, Alzahrani KJ, et al. Using green biosynthesized lycopene-coated selenium nanoparticles to rescue renal damage in glycerol-induced acute kidney injury in rats. Int J Nanomedicine 2021; 16: 4335-49.
[http://dx.doi.org/10.2147/IJN.S306186] [PMID: 34234429]
[42]
Alten B, Yesiltepe M, Bayraktar E, et al. High-fructose corn syrup consumption in adolescent rats causes bipolar-like behavioural phenotype with hyperexcitability in hippocampal CA3-CA1 synapses. Br J Pharmacol 2018; 175(24): 4450-63.
[http://dx.doi.org/10.1111/bph.14500] [PMID: 30221753]
[43]
Das NR, Vaidya B, Khare P, Bishnoi M, Sharma SS. Combination of peroxisome proliferator-activated receptor gamma (PPARγ) agonist and PPAR gamma co-activator 1α (PGC-1α) activator ameliorates cognitive deficits, oxidative stress, and inflammation in rodent model of Parkinson’s disease. Curr Neurovasc Res 2021; 18(5): 497-507.
[http://dx.doi.org/10.2174/1567202619666211217140954] [PMID: 34923943]
[44]
Vaidya B, Kaur H, Thapak P, Sharma SS, Singh JN. Pharmacological modulation of TRPM2 channels via PARP pathway leads to neuroprotection in MPTP-induced Parkinson’s disease in sprague dawley rats. Mol Neurobiol 2022; 59(3): 1528-42.
[http://dx.doi.org/10.1007/s12035-021-02711-4] [PMID: 34997907]
[45]
Polomano RC, Mannes AJ, Clark US, Bennett GJ. A painful peripheral neuropathy in the rat produced by the chemotherapeutic drug, paclitaxel. Pain 2001; 94(3): 293-304.
[http://dx.doi.org/10.1016/S0304-3959(01)00363-3] [PMID: 11731066]
[46]
Mehrotra A, Shanbhag R, Chamallamudi MR, Singh VP, Mudgal J. Ameliorative effect of caffeic acid against inflammatory pain in rodents. Eur J Pharmacol 2011; 666(1-3): 80-6.
[http://dx.doi.org/10.1016/j.ejphar.2011.05.039] [PMID: 21645514]
[47]
Wang H, Shen YJ, Li XJ, et al. DNMT3b SUMOylation mediated MMP-2 upregulation contribute to paclitaxel induced neuropathic pain. Neurochem Res 2021; 46(5): 1214-23.
[http://dx.doi.org/10.1007/s11064-021-03260-x] [PMID: 33550530]
[48]
Sari AN, Dhanjal JK, Elwakeel A, et al. A low dose combination of withaferin A and caffeic acid phenethyl ester possesses anti-metastatic potential in vitro: Molecular targets and mechanisms. Cancers (Basel) 2022; 14(3): 787.
[http://dx.doi.org/10.3390/cancers14030787] [PMID: 35159054]
[49]
Lee HE, Yang G, Kim ND, et al. Targeting ASC in NLRP3 inflammasome by caffeic acid phenethyl ester: A novel strategy to treat acute gout. Sci Rep 2016; 6(1): 38622.
[http://dx.doi.org/10.1038/srep38622] [PMID: 27934918]
[50]
Lee YJ, Liao PH, Chen WK, Yang CC. Preferential cytotoxicity of caffeic acid phenethyl ester analogues on oral cancer cells. Cancer Lett 2000; 153(1-2): 51-6.
[http://dx.doi.org/10.1016/S0304-3835(00)00389-X] [PMID: 10779629]
[51]
Williams B, Tsangari E, Stansborough R, et al. Mixed effects of caffeic acid phenethyl ester (CAPE) on joint inflammation, bone loss and gastrointestinal inflammation in a murine model of collagen antibody-induced arthritis. Inflammopharmacology 2017; 25(1): 55-68.
[http://dx.doi.org/10.1007/s10787-016-0306-z] [PMID: 28044215]
[52]
Soares VEM, do Carmo TIT, dos Anjos F, et al. Role of inflammation and oxidative stress in tissue damage associated with cystic fibrosis: CAPE as a future therapeutic strategy. Mol Cell Biochem 2022; 477(1): 39-51.
[http://dx.doi.org/10.1007/s11010-021-04263-6] [PMID: 34529223]
[53]
Elumalai P, Muninathan N, Megalatha ST, et al. An insight into anticancer effect of propolis and its constituents: A review of molecular mechanisms. J Evid Based Complementary Altern Med 2022. [Epub ahead of print]
[54]
Ameenudeen S, Kashif M, Banerjee S, Srinivasan H, Pandurangan AK, Waseem M. Cellular and molecular machinery of neuropathic pain: An emerging insight. Curr Pharmacol Rep 2022; 8(4): 227-35.
[http://dx.doi.org/10.1007/s40495-022-00294-9] [PMID: 35646513]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy