Generic placeholder image

Current Neuropharmacology

Editor-in-Chief

ISSN (Print): 1570-159X
ISSN (Online): 1875-6190

Review Article

Current Drugs and Potential Future Neuroprotective Compounds for Parkinson’s Disease

Author(s): Iván Carrera* and Ramón Cacabelos

Volume 17, Issue 3, 2019

Page: [295 - 306] Pages: 12

DOI: 10.2174/1570159X17666181127125704

Price: $65

Abstract

The research progress of understanding the etiology and pathogenesis of Parkinson's disease (PD) has yet lead to the development of some clinical approaches intended to treat cognitive and behavioral symptoms, such as memory and perception disorders. Despite the major advances in different genetic causes and risk factors for PD, which share common pathways to cell dysfunction and death, there is not yet a complete model of PD that can be used to accurately predict the effect of drugs on disease progression. Clinical trials are also important to test any novel neuro-protective agent, and recently there have been great advances in the use of anti-inflammatory drugs and plant flavonoid antioxidants to protect against specific neuronal degeneration and its interference with lipid and cholesterol metabolism. The increasing knowledge of the molecular events underlying the degenerative process of PD has stimulated research to identify natural compounds capable of halting or slowing the progress of neural deterioration. Polyphenols and flavonoids, which play a neuroprotective role in a wide array of in vitro and in vivo models of neurological disorders, emerged from among the multi-target bio-agents found mainly in plants and microorganisms. This review presents a detailed overview of the multimodal activities of neuroprotective bio-agents tested so far, emphasizing their neurorescue/neuroregenerative activity. The brain-penetrating property of bioagents may make these compounds an important class of natural drugs for the treatment of neurodegenerative diseases. Although there are numerous studies demonstrating beneficial effects in the laboratory by identifying critical molecular targets, the clinical efficacy of these neuroprotective treatments remains to be proven accurately.

Keywords: Parkinson's disease, nutraceuticals, neuroprotection, phytobioactivity, degeneration, neuroscience.

« Previous
Graphical Abstract
[1]
Yuan H, Zhang ZW, Liang LW, et al. Treatment strategies for Parkinson’s disease. Neurosci Bull 2010; 26(1): 66-76.
[http://dx.doi.org/dx.doi. org/10.1007/s12264-010-0302-z] [PMID: 20101274]
[2]
Cacabelos R. Parkinson’s disease: From pathogenesis to pharmacogenomics. Int J Mol Sci 2017; 18(3): E551.
[http://dx.doi.org/ 10.3390/ijms18030551] [PMID: 28273839]
[3]
Cacabelos, R. Parkinson’s disease: Old concepts and new challenges. Sci. Pages Alzheimers Dis. Dement., 2016, 1, 001.
[4]
Cacabelos R, Carrera I, Fernández-Novoa L, et al. Parkinson’s Disease: New solutions to old problems. Euro Espes J 2017; 11: 74-96.
[5]
Tang SW, Helmeste DM, Leonard BE. Neurodegeneration, neuroregeneration, and neuroprotection in psychiatric disorders. Mod Trends Pharmacopsychiatry 2017; 31: 107-23.
[http://dx.doi.org/dx. doi.org/10.1159/000470811] [PMID: 28738379]
[6]
Cummings J. Disease modification and neuroprotection in neurodegenerative disorders. Transl Neurodegener 2017; 6: 25.
[http://dx.doi.org/ dx.doi.org/10.1186/s40035-017-0096-2] [PMID: 29021896]
[7]
Seidl SE, Potashkin JA. The promise of neuroprotective agents in Parkinson’s disease. Front Neurol 2011; 2: 68.
[http://dx.doi.org/dx. doi.org/10.3389/fneur.2011.00068] [PMID: 22125548]
[8]
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2018; 38(4): 1126-58.
[http://dx.doi.org/10.1002/med.21458] [PMID: 28697282]
[9]
Francardo V, Schmitz Y, Sulzer D, Cenci MA. Neuroprotection and neurorestoration as experimental therapeutics for Parkinson’s disease 2017.
[10]
Rangasamy SB, Soderstrom K, Bakay RA, Kordower JH. Neurotrophic factor therapy for Parkinson’s disease. 2010.
[11]
Ibáñez, C.F.; Andressoo, J.O. Biology of GDNF and its receptors - relevance for disorders of the central nervous system. Neurobiol. Dis., 2017, 97(Pt B), 80-89. [http://dx.doi.org/10.1016/j.nbd. 2016.01.021] 26829643]
[12]
Vilar M, Mira H. Regulation of neurogenesis by neurotrophins during adulthood: expected and unexpected roles. Front Neurosci 2016; 10: 26.
[http://dx.doi.org/10.3389/fnins.2016.00026] [PMID: 26903794]
[13]
Zhao C, Deng W, Gage FH. Mechanisms and functional implications of adult neurogenesis. Cell 2008; 132(4): 645-60.
[http://dx.doi.org/10.1016/j.cell.2008.01.033] [PMID: 18295581]
[14]
Carbon M, Reetz K, Ghilardi MF, Dhawan V, Eidelberg D. Early Parkinson’s disease: Longitudinal changes in brain activity during sequence learning. Neurobiol Dis 2010; 37(2): 455-60.
[http://dx.doi.org/10.1016/j.nbd.2009.10.025] [PMID: 19900556]
[15]
Cheng HC, Ulane CM, Burke RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol 2010; 67(6): 715-25.
[http://dx.doi.org/10.1002/ana.21995] [PMID: 20517933]
[16]
Song DD, Haber SN. Striatal responses to partial dopaminergic lesion: Evidence for compensatory sprouting. J Neurosci 2000; 20(13): 5102-14.
[http://dx.doi.org/10.1523/JNEUROSCI.20-13-05102.2000] [PMID: 10864967]
[17]
Sossi V, de la Fuente-Fernández R, Holden JE, Schulzer M, Ruth TJ, Stoessl J. Changes of dopamine turnover in the progression of Parkinson’s disease as measured by positron emission tomography: their relation to disease-compensatory mechanisms. J Cereb Blood Flow Metab 2004; 24(8): 869-76.
[http://dx.doi.org/dx. doi.org/10.1097/01.WCB.0000126563.85360.75] [PMID: 15362717]
[18]
Brotchie J, Fitzer-Attas C. Mechanisms compensating for dopamine loss in early Parkinson disease. Neurology 2009; 72(7)(Suppl.): S32-8.
[http://dx.doi.org/10.1212/WNL.0b013e318198 e0e9] [PMID: 19221312]
[19]
Maneuf YP, Mitchell IJ, Crossman AR, Brotchie JM. On the role of enkephalin cotransmission in the GABAergic striatal efferents to the globus pallidus. Exp Neurol 1994; 125(1): 65-71.
[http://dx.doi.org/10.1006/exnr.1994.1007] [PMID: 8307125]
[20]
Vila M, Périer C, Féger J, et al. Evolution of changes in neuronal activity in the subthalamic nucleus of rats with unilateral lesion of the substantia nigra assessed by metabolic and electrophysiological measurements. Eur J Neurosci 2000; 12(1): 337-44.
[http://dx.doi.org/10.1046/j.1460-9568.2000.00901.x] [PMID: 10651888]
[21]
Bezard E, Gross CE, Brotchie JM. Presymptomatic compensation in Parkinson’s disease is not dopamine-mediated. Trends Neurosci 2003; 26(4): 215-21.
[http://dx.doi.org/10.1016/S0166-2236 (03)00038-9] [PMID: 12689773]
[22]
Liu CY, Lee B, Boulis N, Rezai AR. Introduction: Neurorestoration: re-animating the CNS. Neurosurg Focus 2016; 40(5): E1.
[http://dx.doi.org/10.3171/2016.2.FOCUS1688] [PMID: 27132522]
[23]
Maruyama W, Youdim MB, Naoi M. Antiapoptotic properties of rasagiline, N-propargylamine-1(R)-aminoindan, and its optical (S)-isomer, TV1022. Ann N Y Acad Sci 2001; 939: 320-9.
[http://dx.doi.org/10.1111/j.1749-6632.2001.tb03641.x] [PMID: 11462787]
[24]
Sagi Y, Mandel S, Amit T, Youdim MB. Activation of tyrosine kinase receptor signaling pathway by rasagiline facilitates neurorescue and restoration of nigrostriatal dopamine neurons in post-MPTP-induced parkinsonism. Neurobiol Dis 2007; 25(1): 35-44.
[http://dx.doi.org/10.1016/j.nbd.2006.07.020] [PMID: 17055733]
[25]
Kupsch A, Sautter J, Götz ME, et al. Monoamine oxidase-inhibition and MPTP-induced neurotoxicity in the non-human primate: Comparison of rasagiline (TVP 1012) with selegiline. J Neural Transm (Vienna) 2001; 108(8-9): 985-1009.
[http://dx.doi.org/10.1007/s007020170018] [PMID: 11716151]
[26]
Weintraub D, Hauser RA, Elm JJ, Pagan F, Davis MD, Choudhry A. Rasagiline for mild cognitive impairment in Parkinson’s disease: A placebo-controlled trial. Mov Disord 2016; 31(5): 709-14.
[http://dx.doi.org/10.1002/mds.26617] [PMID: 27030249]
[27]
Cronin A, Grealy M. Neuroprotective and neuro-restorative effects of minocycline and rasagiline in a zebrafish 6-hydroxydopamine model of parkinson’s disease. Neuroscience 2017; 367: 34-46.
[http://dx.doi.org/10.1016/j.neuroscience.2017. 10.018] [PMID: 29079063]
[28]
Hoyles L, Vulevic J. Diet, immunity and functional foods. Adv Exp Med Biol 2008; 635: 79-92.
[http://dx.doi.org/10.1007/978-0-387-09550-9_7] [PMID: 18841705]
[29]
Hang L, Basil AH, Lim KL. Nutraceuticals in Parkinson’s Disease. Neuromolecular Med 2016; 18(3): 306-21.
[http://dx.doi.org/
dx.doi.org/10.1007/s12017-016-8398-6] [PMID: 27147525]
[30]
Chao J, Leung Y, Wang M, Chang RC. Nutraceuticals and their preventive or potential therapeutic value in Parkinson’s disease. Nutr Rev 2012; 70(7): 373-86.
[http://dx.doi.org/10.1111/j.1753-4887.2012.00484.x] [PMID: 22747840]
[31]
Boskabady MH, Farkhondeh T. Antiinflammatory, antioxidant, and immunomodulatory effects of Crocus sativus L. and its main constituents. Phytother Res 2016; 30(7): 1072-94.
[http://dx.doi.org/dx. doi.org/10.1002/ptr.5622] [PMID: 27098287]
[32]
Tseng TH, Chu CY, Huang JM, Shiow SJ, Wang CJ. Crocetin protects against oxidative damage in rat primary hepatocytes. Cancer Lett 1995; 97(1): 61-7.
[http://dx.doi.org/10.1016/0304-3835(95)03964-X] [PMID: 7585479]
[33]
Bhandari PR. Crocus sativus L. (saffron) for cancer chemoprevention: A mini review. J Tradit Complement Med 2015; 5(2): 81-7.
[http://dx.doi.org/10.1016/j.jtcme.2014.10.009] [PMID: 26151016]
[34]
Premkumar K, Thirunavukkarasu C, Abraham SK, Santhiya ST, Ramesh A. Protective effect of saffron (Crocus sativus L.) aqueous extract against genetic damage induced by anti-tumor agents in mice. Hum Exp Toxicol 2006; 25(2): 79-84.
[http://dx.doi.org/dx. doi.org/10.1191/0960327106ht589oa] [PMID: 16539212]
[35]
Yousefi E, Eskandari A, Gharavi MJ, Khademvatan S. In vitro activity and cytotoxicity of Crocus sativus extract against leihmania major (MRHO/IR/75/ER). Infect Disord Drug Targets 2014; 14(1): 56-60.
[http://dx.doi.org/10.2174/1871526514666140827 101901] [PMID: 25159304]
[36]
Kianbakht S, Mozaffari K. Effects of saffron and its active constituents, crocin and safranal, on prevention of indomethacin induced gastric ulcers in diabetic and nondiabetic rats. Faslnamah-i Giyahan-i Daruyi 2009; 8: 30-8.
[37]
El-Maraghy SA, Rizk SM, Shahin NN. Gastroprotective effect of crocin in ethanol-induced gastric injury in rats. Chem Biol Interact 2015; 229: 26-35.
[http://dx.doi.org/10.1016/j.cbi.2015. 01.015] [PMID: 25637687]
[38]
Boskabady MH, Ghasemzadeh Rahbardar M, Nemati H, Esmaeilzadeh M. Inhibitory effect of Crocus sativus (saffron) on histamine (H1) receptors of guinea pig tracheal chains. Pharmazie 2010; 65(4): 300-5.
[PMID: 20432629]
[39]
Hazman Ö, Bozkurt MF. Anti-inflammatory and antioxidative activities of safranal in the reduction of renal dysfunction and damage that occur in diabetic nephropathy. Inflammation 2015; 38(4): 1537-45.
[http://dx.doi.org/10.1007/s10753-015-0128-y] [PMID: 25667012]
[40]
Rezaee R, Hosseinzadeh H. Safranal: from an aromatic natural product to a rewarding pharmacological agent. Iran J Basic Med Sci 2013; 16(1): 12-26.
[PMID: 23638289]
[41]
Abdullaev F, Ortega CH, Miranda PR. HPLC quantification of major active components from 11 different saffron (Crocus sativus L.) sources. Food Chem 2007; 100: 1126-31.
[http://dx.doi.org/dx.doi.
org/10.1016/j.foodchem.2005.11.020]
[42]
Zhang C, Ma J, Fan L, et al. Neuroprotective effects of safranal in a rat model of traumatic injury to the spinal cord by anti-apoptotic, anti-inflammatory and edema-attenuating. Tissue Cell 2015; 47(3): 291-300.
[http://dx.doi.org/dx. doi.org/10.1016/j.tice.2015.03.007] [PMID: 25891268]
[43]
Wang K, Zhang L, Rao W, et al. Neuroprotective effects of crocin against traumatic brain injury in mice: Involvement of notch signaling pathway. Neurosci Lett 2015; 591: 53-8.
[http://dx.doi.org/dx.doi.
org/10.1016/j.neulet.2015.02.016] [PMID: 25681620]
[44]
Nam KN, Park YM, Jung HJ, et al. Anti-inflammatory effects of crocin and crocetin in rat brain microglial cells. Eur J Pharmacol 2010; 648(1-3): 110-6.
[http://dx.doi.org/10.1016/j.ejphar.2010.09.003] [PMID: 20854811]
[45]
Hatziagapiou K, Kakouri E, Lambrou GI, Bethanis K, Tarantilis PA. Antioxidant Properties of Crocus Sativus L. and its Constituents and Relevance to Neurodegenerative Diseases; Focus on Alzheimer’s And Parkinson’s disease. Curr Neuropharmacol 2018. Epub ahead of print
[http://dx.doi.org/10.2174/1570159 X16666180321095705] [PMID: 29564976]
[46]
Mythri RB, Bharath MM. Curcumin: a potential neuroprotective agent in Parkinson’s disease. Curr Pharm Des 2012; 18(1): 91-9.
[http://dx.doi.org/10.2174/138161212798918995] [PMID: 22211691]
[47]
Lee WH, Loo CY, Bebawy M, Luk F, Mason RS, Rohanizadeh R. Curcumin and its derivatives: their application in neuropharmacology and neuroscience in the 21st century. Curr Neuropharmacol 2013; 11(4): 338-78.
[http://dx.doi.org/10. 2174/1570159X11311040002] [PMID: 24381528]
[48]
Wang J, Du XX, Jiang H, Xie JX. Curcumin attenuates 6-hydroxydopamine-induced cytotoxicity by anti-oxidation and nuclear factor-kappa B modulation in MES23.5 cells. Biochem Pharmacol 2009; 78(2): 178-83.
[http://dx.doi.org/10.1016/j.bcp.2009.03.031] [PMID: 19464433]
[49]
Rajeswari A, Sabesan M. Inhibition of monoamine oxidase-B by the polyphenolic compound, curcumin and its metabolite tetrahydrocurcumin, in a model of Parkinson’s disease induced by MPTP neurodegeneration in mice. Inflammopharmacology 2008; 16(2): 96-9.
[http://dx.doi.org/10.1007/s10787-007-1614-0] [PMID: 18408903]
[50]
Xie CJ, Gu AP, Cai J, Wu Y, Chen RC. Curcumin protects neural cells against ischemic injury in N2a cells and mouse brain with ischemic stroke. Brain Behav 2018; 8(2): e00921.
[http://dx.doi.org/dx. doi.org/10.1002/brb3.921] [PMID: 29484272]
[51]
More SV, Choi DK. Promising cannabinoid-based therapies for Parkinson’s disease: Motor symptoms to neuroprotection. Mol Neurodegener 2015; 10: 17.
[http://dx.doi.org/10.1186/s13024-015-0012-0] [PMID: 25888232]
[52]
Berger C, Schmid PC, Schabitz WR, Wolf M, Schwab S, Schmid HH. Massive accumulation of N-acylethanolamines after stroke. Cell signalling in acute cerebral ischemia? J Neurochem 2004; 88(5): 1159-67.
[http://dx.doi.org/10.1046/j.1471-4159. 2003.02244.x] [PMID: 15009671]
[53]
Stampanoni BM, Sancesario A, Morace R, Centonze D, Iezzi E. Cannabinoids in parkinson’s disease. Cannabis Cannabinoid Res 2017; 2(1): 21-9.
[http://dx.doi.org/10.1089/can.2017.0002] [PMID: 28861502]
[54]
García-Arencibia M, González S, de Lago E, Ramos JA, Mechoulam R, Fernández-Ruiz J. Evaluation of the neuroprotective effect of cannabinoids in a rat model of Parkinson’s disease: importance of antioxidant and cannabinoid receptor-independent properties. Brain Res 2007; 1134(1): 162-70.
[http://dx.doi.org/
10.1016/j.brainres.2006.11.063] [PMID: 17196181]
[55]
Fernández-Ruiz J. The endocannabinoid system as a target for the treatment of motor dysfunction. Br J Pharmacol 2009; 156(7): 1029-40.
[http://dx.doi.org/10.1111/j.1476-5381.2008.00088.x] [PMID: 19220290]
[56]
Hassell KJ, Ezzati M, Alonso-Alconada D, Hausenloy DJ, Robertson NJ. New horizons for newborn brain protection: enhancing endogenous neuroprotection. Arch Dis Child Fetal Neonatal Ed 2015; 100(6): F541-52.
[http://dx.doi.org/10.1136/archdischild-2014-306284] [PMID: 26063194]
[57]
Chung ES, Bok E, Chung YC, Baik HH, Jin BK. Cannabinoids prevent lipopolysaccharide-induced neurodegeneration in the rat substantia nigra in vivo through inhibition of microglial activation and NADPH oxidase. Brain Res 2012; 1451: 110-6.
[http://dx.doi.org/
dx.doi.org/10.1016/j.brainres.2012.02.058] [PMID: 22436849]
[58]
Pazos MR, Mohammed N, Lafuente H, et al. Mechanisms of cannabidiol neuroprotection in hypoxic-ischemic newborn pigs: role of 5HT(1A) and CB2 receptors. Neuropharmacology 2013; 71: 282-91.
[http://dx.doi.org/10.1016/j.neuropharm. 2013.03.027] [PMID: 23587650]
[59]
Lastres-Becker I, Fernández-Ruiz J. An overview of Parkinson’s disease and the cannabinoid system and possible benefits of cannabinoid-based treatments. Curr Med Chem 2006; 13(30): 3705-18.
[http://dx.doi.org/10.2174/092986706779026156] [PMID: 17168732]
[60]
Wang Y, Catana F, Yang Y, Roderick R, van Breemen RB. An LC-MS method for analyzing total resveratrol in grape juice, cranberry juice, and in wine. J Agric Food Chem 2002; 50(3): 431-5.
[http://dx.doi.org/10.1021/jf010812u] [PMID: 11804508]
[61]
Renaud S, de Lorgeril M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992; 339(8808): 1523-6.
[http://dx.doi.org/10.1016/0140-6736(92)91277-F] [PMID: 1351198]
[62]
Jang M, Cai L, Udeani GO, et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997; 275(5297): 218-20.
[http://dx.doi.org/10.1126/science. 275.5297.218] [PMID: 8985016]
[63]
Farooqi AA, Khalid S, Ahmad A. Regulation of cell signaling pathways and miRNAs by resveratrol in different cancers. Int J Mol Sci 2018; 19(3): E652.
[http://dx.doi.org/10.3390/ijms 19030652] [PMID: 29495357]
[64]
Kosmeder JW II, Pezzuto JM, Pezzuto JM. Biological effects of resveratrol. Antioxid Redox Signal 2001; 3(6): 1041-64.
[http://dx.doi.org/10.1089/152308601317203567] [PMID: 11813979]
[65]
Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov 2006; 5(6): 493-506.
[http://dx.doi.org/10.1038/nrd2060] [PMID: 16732220]
[66]
Potdar S, Parmar MS, Ray SD, Cavanaugh JE. Protective effects of the resveratrol analog piceid in dopaminergic SH-SY5Y cells. Arch Toxicol 2018; 92(2): 669-77.
[http://dx.doi.org/10. 1007/s00204-017-2073-z] [PMID: 28980048]
[67]
Zhang J, Fan W, Wang H, et al. Resveratrol protects PC12 cell against 6-OHDA damage via CXCR4 signaling pathway. Evid Based Complement Alternat Med 2015; 2015: 730121.
[http://dx.doi.org/10.1155/
2015/730121] [PMID: 26681969]
[68]
Zeng W, Zhang W, Lu F, Gao L, Gao G. Resveratrol attenuates MPP+-induced mitochondrial dysfunction and cell apoptosis via AKT/GSK-3β pathway in SN4741 cells. Neurosci Lett 2017; 637: 50-6.
[http://dx.doi.org/10.1016/j.neulet.2016.11.054] [PMID: 27894919]
[69]
Gaballah HH, Zakaria SS, Elbatsh MM, Tahoon NM. Modulatory effects of resveratrol on endoplasmic reticulum stress-associated apoptosis and oxido-inflammatory markers in a rat model of rotenone-induced Parkinson’s disease. Chem Biol Interact 2016; 251: 10-6.
[http://dx.doi.org/10.1016/j.cbi.2016.03.023] [PMID: 27016191]
[70]
Khan MA, Chen HC, Wan XX, et al. Regulatory effects of resveratrol on antioxidant enzymes: A mechanism of growth inhibition and apoptosis induction in cancer cells. Mol Cells 2013; 35(3): 219-25.
[http://dx.doi.org/dx. doi.org/10.1007/s10059-013-2259-z] [PMID: 23456297]
[71]
Wang ZH, Zhang JL, Duan YL, Zhang QS, Li GF, Zheng DL. MicroRNA-214 participates in the neuroprotective effect of Resveratrol via inhibiting α-synuclein expression in MPTP-induced Parkinson’s disease mouse. Biomed Pharmacother 2015; 74: 252-6.
[http://dx.doi.org/10.1016/j.biopha.2015.08.025] [PMID: 26349993]
[72]
Singh G, Pai RS. In-vitro/in-vivo characterization of trans-resveratrol-loaded nanoparticulate drug delivery system for oral administration. J Pharm Pharmacol 2014; 66(8): 1062-76.
[PMID: 24779896]
[73]
Singh G, Pai RS. Trans-resveratrol self-nano-emulsifying drug delivery system (SNEDDS) with enhanced bioavailability potential: optimization, pharmacokinetics and in situ single pass intestinal perfusion (SPIP) studies. Drug Deliv 2015; 22(4): 522-30.
[http://dx.doi.org/10.3109/10717544.2014.885616] [PMID: 24512464]
[74]
Tellone E, Galtieri A, Russo A, Giardina B, Ficarra S. Resveratrol: A focus on several neurodegenerative diseases. Oxid Med Cell Longev 2015; 2015: 392169.
[http://dx.doi.org/10. 1155/2015/392169] [PMID: 26180587]
[75]
Blanchet J, Longpré F, Bureau G, et al. Resveratrol, a red wine polyphenol, protects dopaminergic neurons in MPTP-treated mice. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32(5): 1243-50.
[http://dx.doi.org/10.1016/j.pnpbp.2008.03.024] [PMID: 18471948]
[76]
Donmez G, Arun A, Chung CY, McLean PJ, Lindquist S, Guarente L. SIRT1 protects against α-synuclein aggregation by activating molecular chaperones. J Neurosci 2012; 32(1): 124-32.
[http://dx.doi.org/10.1523/JNEUROSCI.3442-11.2012] [PMID: 22219275]
[77]
Wang H, Dong X, Liu Z, et al. Resveratrol suppresses rotenone-induced neurotoxicity through activation of SIRT1/Akt1 signaling pathway. Anat, Rec, (Hoboken)., 2018. In:
[78]
Bounda GA, Feng YU. Review of clinical studies of Polygonum multiflorum Thunb. and its isolated bioactive compounds. Pharmacognosy Res 2015; 7(3): 225-36.
[http://dx.doi.org/10.4103/0974-8490.157957] [PMID: 26130933]
[79]
Lin L, Ni B, Lin H, et al. Traditional usages, botany, phytochemistry, pharmacology and toxicology of Polygonum multiflorum Thunb.: a review. J Ethnopharmacol 2015; 159: 158-83.
[http://dx.doi.org/10.1016/j.jep. 2014.11.009] [PMID: 25449462]
[80]
Zhang F, Wang YY, Yang J, Lu YF, Liu J, Shi JS. Tetrahydroxystilbene glucoside attenuates neuroinflammation through the inhibition of microglia activation. Oxid Med Cell Longev 2013; 2013: 680545.
[http://dx.doi.org/10.1155/2013/680545] [PMID: 24349614]
[81]
Huang C, Lin F, Wang G, et al. Tetrahydroxystilbene glucoside produces neuroprotection against 6-OHDA-induced dopamine neurotoxicity. Oxid Med Cell Longev 2018; 2018: 7927568.
[http://dx.doi.org/10.1155/
2018/7927568] [PMID: 29576855]
[82]
Shen C, Sun FL, Zhang RY, et al. Tetrahydroxystilbene glucoside ameliorates memory and movement functions, protects synapses and inhibits α-synuclein aggregation in hippocampus and striatum in aged mice. Restor Neurol Neurosci 2015; 33(4): 531-41.
[http://dx.doi.org/10. 3233/RNN-150514] [PMID: 26409411]
[83]
Wang T, Gu J, Wu PF, et al. Protection by tetrahydroxystilbene glucoside against cerebral ischemia: involvement of JNK, SIRT1, and NF-kappaB pathways and inhibition of intracellular ROS/RNS generation. Free Radic Biol Med 2009; 47(3): 229-40.
[http://dx.doi.org/10.1016/j.freeradbiomed.2009.02.027] [PMID: 19272442]
[84]
Nash KM, Shah ZA. Current perspectives on the beneficial role of ginkgo biloba in neurological and cerebrovascular disorders. Integr Med Insights 2015; 10: 1-9.
[http://dx.doi.org/10.4137/I MI.S25054] [PMID: 26604665]
[85]
Yoshikawa T, Naito Y, Kondo M. Ginkgo biloba leaf extract: review of biological actions and clinical applications. Antioxid Redox Signal 1999; 1(4): 469-80.
[http://dx.doi.org/10.1089/ars. 1999.1.4-469] [PMID: 11233145]
[86]
Yin B, Xu Y, Wei R, Luo B. Ginkgo biloba on focal cerebral ischemia: a systematic review and meta-analysis. Am J Chin Med 2014; 42(4): 769-83.
[http://dx.doi.org/10.1142/S0192415X14500499] [PMID: 25004874]
[87]
Riederer P, Jellinger K. Neurochemical insights into monoamine oxidase inhibitors, with special reference to deprenyl (selegiline). Acta Neurol Scand Suppl 1983; 95: 43-55.
[http://dx.doi.org/dx.doi.
org/10.1111/j.1600-0404.1983.tb01516.x] [PMID: 6145282]
[88]
Myllylä VV, Sotaniemi KA, Vuorinen JA, Heinonen EH. Selegiline as initial treatment in de novo parkinsonian patients. Neurology 1992; 42(2): 339-43.
[http://dx.doi.org/10.1212/WNL.42.2.339] [PMID: 1736162]
[89]
Baez S, Segura-Aguilar J, Widersten M, Johansson AS, Mannervik B. Glutathione transferases catalyse the detoxication of oxidized metabolites (o-quinones) of catecholamines and may serve as an antioxidant system preventing degenerative cellular processes. Biochem J 1997; 324(Pt 1): 25-8.
[http://dx.doi.org/
10.1042/bj3240025] [PMID: 9164836]
[90]
Wu WR, Zhu XZ. Involvement of monoamine oxidase inhibition in neuroprotective and neurorestorative effects of Ginkgo biloba extract against MPTP-induced nigrostriatal dopaminergic toxicity in C57 mice. Life Sci 1999; 65(2): 157-64.
[http://dx.doi.org/dx.doi.
org/10.1016/S0024-3205(99)00232-5] [PMID: 10416821]
[91]
Ahmad M, Saleem S, Ahmad AS, et al. Ginkgo biloba affords dose-dependent protection against 6-hydroxydopamine-induced parkinsonism in rats: neurobehavioural, neurochemical and immunohistochemical evidences. J Neurochem 2005; 93(1): 94-104.
[http://dx.doi.org/10.1111/j.1471-4159.2005.03000.x] [PMID: 15773909]
[92]
Sur S, Panda CK. Molecular aspects of cancer chemopreventive and therapeutic efficacies of tea and tea polyphenols. Nutrition 2017; 43-44: 8-15.
[http://dx.doi.org/10.1016/j.nut.2017.06.006] [PMID: 28935149]
[93]
Singh NA, Mandal AK, Khan ZA. Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG). Nutr J 2016; 15(1): 60.
[http://dx.doi.org/10.1186/s12937-016-0179-4] [PMID: 27268025]
[94]
Hang L, Basil AH, Lim KL. Nutraceuticals in Parkinson’s Disease. Neuromol Med 2016; 18(3): 306-21.
[http://dx.doi.org/dx.doi.
org/10.1007/s12017-016-8398-6] [PMID: 27147525]
[95]
Kim J, Shin J, Ha J. Screening methods for AMP-activated protein kinase modulators: a patent review. Expert Opin Ther Pat 2015; 25(3): 261-77.
[http://dx.doi.org/10.1517/13543776. 2014.995626] [PMID: 25535089]
[96]
Whitworth AJ. Drosophila models of Parkinson’s disease. Adv Genet 2011; 73: 1-50.
[http://dx.doi.org/10.1016/B978-0-12-380860-8.00001-X] [PMID: 21310293]
[97]
Ng CH, Guan MS, Koh C, et al. AMP kinase activation mitigates dopaminergic dysfunction and mitochondrial abnormalities in Drosophila models of Parkinson’s disease. J Neurosci 2012; 32(41): 14311-7.
[http://dx.doi.org/10.1523/JNEUROSCI. 0499-12.2012] [PMID: 23055502]
[98]
Ng CH, Basil AH, Hang L, et al. Genetic or pharmacological activation of the Drosophila PGC-1α ortholog spargel rescues the disease phenotypes of genetic models of Parkinson’s disease. Neurobiol Aging 2017; 55: 33-7.
[http://dx.doi.org/10.1016/j.neurobiolaging. 2017.03.017] [PMID: 28407521]
[99]
Choi JY, Park CS, Kim DJ, et al. Prevention of nitric oxide-mediated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson’s disease in mice by tea phenolic epigallocatechin 3-gallate. Neurotoxicology 2002; 23(3): 367-74.
[http://dx.doi.org/10.1016/S0161-813X (02)00079-7] [PMID: 12387363]
[100]
Zhou T, Zhu M, Liang Z. (-)-Epigallocatechin-3-gallate modulates peripheral immunity in the MPTP-induced mouse model of Parkinson’s disease. Mol Med Rep 2018; 17(4): 4883-8.
[PMID: 29363729]
[101]
Caruana M, Vassallo N. Tea Polyphenols in Parkinson’s Disease. Adv Exp Med Biol 2015; 863: 117-37.
[http://dx.doi.org/
10.1007/978-3-319-18365-7_6] [PMID: 26092629]
[102]
Cacabelos R. 2016.
[103]
Cotzias GC, Papavasiliou PS, Gellene R. L-dopa in parkinson’s syndrome. N Engl J Med 1969; 281(5): 272.
[http://dx.doi.org/dx. doi.org/10.1056/NEJM196907312810517] [PMID: 5791298]
[104]
Oertel WH. Recent advances in treating Parkinson’s disease. F1000 Res 2017; 6: 260.
[http://dx.doi.org/10.12688/f1000 research.10100.1] [PMID: 28357055]
[105]
Cacabelos R, Lombardi V, Fernandez-Novoa L, et al. 2018.
[106]
Romero A, Parada E, González-Lafuente L, et al. Neuroprotective effects of E-PodoFavalin-15999 (Atremorine®). CNS Neurosci Ther 2017; 23(5): 450-2.
[http://dx.doi.org/10.1111/cns.12693] [PMID: 28371323]
[107]
Cacabelos, R. Parkinson’s Disease: From Pathogenesis to Pharmacogenomics. Angelini S, Ed. Int. J. Mol. Sci., 2017, 18, 551.
[108]
Carrera I, Fernandez-Novoa L, Sampedro C, Cacabelos R, Aliev G. Dopaminergic neuroprotection with Atremorine in Parkinson’s disease. Curr Med Chem 2018. Epub ahead of print
[http://dx.doi.org/10.2174/0929867325666180410100559] [PMID: 29637853]
[109]
Carrera I, Fernandez-Novoa L, Sampedro C, Cacabelos R. Neuroprotective effect of atremorine in an experimental model of parkinson’s disease. Curr Pharm Des 2017; 23(18): 2673-84.
[http://dx.doi.org/10.2174/1381612823666170210143530] [PMID: 28190394]
[110]
Cacabelos R, Fernández-Novoa L, Alejo R, et al. E-PodoFavalin-15999 (Atremorine®)-induced dopamine response in Parkinson’s Disease: Pharmacogenetics-related effects. J Genomic Med Pharmacogenomics 2016; 1: 1-26.
[111]
Cacabelos R, Fernández-Novoa L, Alejo R, et al. E-podofavalin-15999 (Atremorine®)-Induced neurotransmitter and hormonal response in parkinson’s diseasE. J Exploratory Res in Pharmacology 2016; 1: 1-12.
[http://dx.doi.org/10.14218/JERP. 2016.00031]
[112]
Liu H, Deng Y, Gao J, et al. Sodium hydrosulfide attenuates beta-amyloid-induced cognitive deficits and neuroinflammation via modulation of MAPK/NF-κB pathway in rats. Curr Alzheimer Res 2015; 12(7): 673-83.
[http://dx.doi.org/10.2174/1567205012666150713102326] [PMID: 26165866]
[113]
Jeong YH, Oh YC, Cho WK, Yim NH, Ma JY. Anti-inflammatory effect of rhapontici radix ethanol extract via inhibition of NF-κB and MAPK and induction of HO-1 in macrophages. Mediators Inflamm 2016; 2016: 13.
[http://dx.doi.org/10.1155/2016/7216912]

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