Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature
Abstract
:1. Introduction
2. Curcumin as a Potential Neuroprotective Agent
2.1. Chemical and Physical Properties of Curcumin
2.2. Pharmacokinetics and Pharmacodynamics of Curcumin
2.3. Biological Properties of Curcumin
2.4. Molecular and Cellular Neuroprotective Mechanisms of Curcumin in PD
2.4.1. Curcumin Anti-Inflammatory Effects
2.4.2. Curcumin Antioxidant Effects
2.4.3. Curcumin Free Radicals’ Scavenging Activities
2.4.4. Mitochondrial Protection
2.4.5. Curcumin Iron-Chelating Properties
3. Neuroprotective Mechanisms of Curcumin via Nicotinic Acetylcholine Receptors
4. An Update and Current Perspectives on Curcumin
5. Concluding Remarks
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Parkinson, J. An essay on the shaking palsy. 1817. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 223–236, discussion 222. [Google Scholar] [CrossRef] [PubMed]
- Del Tredici, K.; Braak, H. Review: Sporadic Parkinson’s disease: Development and distribution of α-synuclein pathology. Neuropathol. Appl. Neurobiol. 2016, 42, 33–50. [Google Scholar] [CrossRef] [PubMed]
- Franco-Iborra, S.; Vila, M.; Perier, C. The Parkinson Disease Mitochondrial Hypothesis: Where Are We at? Neurosci. Rev. J. Bringing Neurobiol. Neurol. Psychiatry 2016, 22, 266–277. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Song, Y.; Chen, Z.; Leng, S.X. Connection between Systemic Inflammation and Neuroinflammation Underlies Neuroprotective Mechanism of Several Phytochemicals in Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2018, 2018, 1972714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blesa, J.; Trigo-Damas, I.; Quiroga-Varela, A.; Jackson-Lewis, V.R. Oxidative stress and Parkinson’s disease. Front. Neuroanat. 2015, 9, S26–S38. [Google Scholar] [CrossRef] [Green Version]
- Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. Vienna Austria 1996 2017, 124, 901–905. [Google Scholar] [CrossRef]
- Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef] [Green Version]
- Pandey, S.; Srivanitchapoom, P. Levodopa-induced Dyskinesia: Clinical Features, Pathophysiology, and Medical Management. Ann. Indian Acad. Neurol. 2017, 20, 190–198. [Google Scholar] [CrossRef]
- Huot, P.; Johnston, T.H.; Koprich, J.B.; Fox, S.H.; Brotchie, J.M. The pharmacology of L-DOPA-induced dyskinesia in Parkinson’s disease. Pharmacol. Rev. 2013, 65, 171–222. [Google Scholar] [CrossRef] [Green Version]
- Connolly, B.S.; Lang, A.E. Pharmacological treatment of Parkinson disease: A review. JAMA 2014, 311, 1670–1683. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.Y.; Tolleson, C. The role of deep brain stimulation in Parkinson’s disease: An overview and update on new developments. Neuropsychiatr. Dis. Treat. 2017, 13, 723–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rizzone, M.G.; Fasano, A.; Daniele, A.; Zibetti, M.; Merola, A.; Rizzi, L.; Piano, C.; Piccininni, C.; Romito, L.M.; Lopiano, L.; et al. Long-term outcome of subthalamic nucleus DBS in Parkinson’s disease: From the advanced phase towards the late stage of the disease? Parkinsonism Relat. Disord. 2014, 20, 376–381. [Google Scholar] [CrossRef] [PubMed]
- Low, V.; Ben-Shlomo, Y.; Coward, E.; Fletcher, S.; Walker, R.; Clarke, C.E. Measuring the burden and mortality of hospitalisation in Parkinson’s disease: A cross-sectional analysis of the English Hospital Episodes Statistics database 2009–2013. Parkinsonism Relat. Disord. 2015, 21, 449–454. [Google Scholar] [CrossRef] [PubMed]
- Shahgholi, L.; Jesus, S.D.; Wu, S.S.; Pei, Q.; Hassan, A.; Armstrong, M.J.; Martinez-Ramirez, D.; Schmidt, P.; Okun, M.S. Hospitalization and rehospitalization in Parkinson disease patients: Data from the National Parkinson Foundation Centers of Excellence. PLoS ONE 2017, 12, e0180425. [Google Scholar] [CrossRef] [Green Version]
- Macleod, A.D.; Taylor, K.S.M.; Counsell, C.E. Mortality in Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. Off. J. Mov. Disord. Soc. 2014, 29, 1615–1622. [Google Scholar] [CrossRef] [Green Version]
- Gupta, S.C.; Patchva, S.; Koh, W.; Aggarwal, B.B. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin. Exp. Pharmacol. Physiol. 2012, 39, 283–299. [Google Scholar] [CrossRef]
- Amalraj, A.; Pius, A.; Gopi, S.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives—A review. J. Tradit. Complement. Med. 2016, 7, 205–233. [Google Scholar] [CrossRef] [Green Version]
- Stohs, S.J.; Chen, O.; Ray, S.D.; Ji, J.; Bucci, L.R.; Preuss, H.G. Highly Bioavailable Forms of Curcumin and Promising Avenues for Curcumin-Based Research and Application: A Review. Mol. Basel Switz. 2020, 25, 1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Priyadarsini, K.I. The Chemistry of Curcumin: From Extraction to Therapeutic Agent. Molecules 2014, 19, 20091–20112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, A.L.; Hsu, C.H.; Lin, J.K.; Hsu, M.M.; Ho, Y.F.; Shen, T.S.; Ko, J.Y.; Lin, J.T.; Lin, B.R.; Ming-Shiang, W.; et al. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001, 21, 2895–2900. [Google Scholar]
- Wahlström, B.; Blennow, G. A study on the fate of curcumin in the rat. Acta Pharmacol. Toxicol. (Copenh.) 1978, 43, 86–92. [Google Scholar] [CrossRef]
- Perkins, S.; Verschoyle, R.D.; Hill, K.; Parveen, I.; Threadgill, M.D.; Sharma, R.A.; Williams, M.L.; Steward, W.P.; Gescher, A.J. Chemopreventive efficacy and pharmacokinetics of curcumin in the min/+ mouse, a model of familial adenomatous polyposis. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 2002, 11, 535–540. [Google Scholar]
- Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent Developments in Delivery, Bioavailability, Absorption and Metabolism of Curcumin: The Golden Pigment from Golden Spice. Cancer Res. Treat. Off. J. Korean Cancer Assoc. 2014, 46, 2–18. [Google Scholar] [CrossRef] [Green Version]
- Dei Cas, M.; Ghidoni, R. Dietary Curcumin: Correlation between Bioavailability and Health Potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [Green Version]
- Cheng, K.K.; Yeung, C.F.; Ho, S.W.; Chow, S.F.; Chow, A.H.L.; Baum, L. Highly stabilized curcumin nanoparticles tested in an in vitro blood-brain barrier model and in Alzheimer’s disease Tg2576 mice. AAPS J. 2013, 15, 324–336. [Google Scholar] [CrossRef] [Green Version]
- Ferrari, R.; Sponchioni, M.; Morbidelli, M.; Moscatelli, D. Polymer nanoparticles for the intravenous delivery of anticancer drugs: The checkpoints on the road from the synthesis to clinical translation. Nanoscale 2018, 10, 22701–22719. [Google Scholar] [CrossRef]
- Shome, S.; Talukdar, A.D.; Choudhury, M.D.; Bhattacharya, M.K.; Upadhyaya, H. Curcumin as potential therapeutic natural product: A nanobiotechnological perspective. J. Pharm. Pharmacol. 2016, 68, 1481–1500. [Google Scholar] [CrossRef] [PubMed]
- Karthikeyan, A.; Senthil, N.; Min, T. Nanocurcumin: A Promising Candidate for Therapeutic Applications. Front. Pharmacol. 2020, 11, 487. [Google Scholar] [CrossRef] [PubMed]
- Rachmawati, H.; Edityaningrum, C.A.; Mauludin, R. Molecular inclusion complex of curcumin-β-cyclodextrin nanoparticle to enhance curcumin skin permeability from hydrophilic matrix gel. AAPS PharmSciTech 2013, 14, 1303–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siddique, Y.H.; Khan, W.; Singh, B.R.; Naqvi, A.H. Synthesis of alginate-curcumin nanocomposite and its protective role in transgenic Drosophila model of Parkinson’s disease. ISRN Pharmacol. 2013, 2013, 794582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kundu, P.; Das, M.; Tripathy, K.; Sahoo, S.K. Delivery of Dual Drug Loaded Lipid Based Nanoparticles across the Blood-Brain Barrier Impart Enhanced Neuroprotection in a Rotenone Induced Mouse Model of Parkinson’s Disease. ACS Chem. Neurosci. 2016, 7, 1658–1670. [Google Scholar] [CrossRef] [PubMed]
- Hahn, Y.-I.; Kim, S.-J.; Choi, B.-Y.; Cho, K.-C.; Bandu, R.; Kim, K.P.; Kim, D.-H.; Kim, W.; Park, J.S.; Han, B.W.; et al. Curcumin interacts directly with the Cysteine 259 residue of STAT3 and induces apoptosis in H-Ras transformed human mammary epithelial cells. Sci. Rep. 2018, 8, 6409. [Google Scholar] [CrossRef] [PubMed]
- Zhang, N.; Yan, F.; Liang, X.; Wu, M.; Shen, Y.; Chen, M.; Xu, Y.; Zou, G.; Jiang, P.; Tang, C.; et al. Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson’s disease therapy. Theranostics 2018, 8, 2264–2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, C.M.; Pasupuleti, V.; Roman, Y.M.; Li, Y.; Hernandez, A.V. Oral turmeric/curcumin effects on inflammatory markers in chronic inflammatory diseases: A systematic review and meta-analysis of randomized controlled trials. Pharmacol. Res. 2019, 146, 104280. [Google Scholar] [CrossRef]
- Abrahams, S.; Haylett, W.L.; Johnson, G.; Carr, J.A.; Bardien, S. Antioxidant effects of curcumin in models of neurodegeneration, aging, oxidative and nitrosative stress: A review. Neuroscience 2019, 406, 1–21. [Google Scholar] [CrossRef]
- Burapan, S.; Kim, M.; Paisooksantivatana, Y.; Eser, B.E.; Han, J. Thai Curcuma Species: Antioxidant and Bioactive Compounds. Foods Basel Switz. 2020, 9, 1219. [Google Scholar] [CrossRef]
- Zheng, D.; Huang, C.; Huang, H.; Zhao, Y.; Khan, M.R.U.; Zhao, H.; Huang, L. Antibacterial Mechanism of Curcumin: A Review. Chem. Biodivers. 2020, 17, e2000171. [Google Scholar] [CrossRef]
- Šudomová, M.; Hassan, S.T.S. Nutraceutical Curcumin with Promising Protection against Herpesvirus Infections and Their Associated Inflammation: Mechanisms and Pathways. Microorganisms 2021, 9, 292. [Google Scholar] [CrossRef]
- Narayanan, V.S.; Muddaiah, S.; Shashidara, R.; Sudheendra, U.S.; Deepthi, N.C.; Samaranayake, L. Variable antifungal activity of curcumin against planktonic and biofilm phase of different candida species. Indian J. Dent. Res. Off. Publ. Indian Soc. Dent. Res. 2020, 31, 145–148. [Google Scholar] [CrossRef]
- Daily, J.W.; Yang, M.; Park, S. Efficacy of Turmeric Extracts and Curcumin for Alleviating the Symptoms of Joint Arthritis: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J. Med. Food 2016, 19, 717–729. [Google Scholar] [CrossRef] [Green Version]
- Khan, H.; Ullah, H.; Nabavi, S.M. Mechanistic insights of hepatoprotective effects of curcumin: Therapeutic updates and future prospects. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 124, 182–191. [Google Scholar] [CrossRef]
- Keihanian, F.; Saeidinia, A.; Bagheri, R.K.; Johnston, T.P.; Sahebkar, A. Curcumin, hemostasis, thrombosis, and coagulation. J. Cell. Physiol. 2018, 233, 4497–4511. [Google Scholar] [CrossRef]
- Li, H.; Sureda, A.; Devkota, H.P.; Pittalà, V.; Barreca, D.; Silva, A.S.; Tewari, D.; Xu, S.; Nabavi, S.M. Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol. Adv. 2020, 38, 107343. [Google Scholar] [CrossRef]
- Den Hartogh, D.J.; Gabriel, A.; Tsiani, E. Antidiabetic Properties of Curcumin II: Evidence from In Vivo Studies. Nutrients 2019, 12, 58. [Google Scholar] [CrossRef] [Green Version]
- Manarin, G.; Anderson, D.; Silva, J.M.E.; da Coppede, J.S.; Roxo-Junior, P.; Pereira, A.M.S.; Carmona, F. Curcuma longa L. ameliorates asthma control in children and adolescents: A randomized, double-blind, controlled trial. J. Ethnopharmacol. 2019, 238, 111882. [Google Scholar] [CrossRef]
- Shahid, H.; Shahzad, M.; Shabbir, A.; Saghir, G. Immunomodulatory and Anti-Inflammatory Potential of Curcumin for the Treatment of Allergic Asthma: Effects on Expression Levels of Pro-inflammatory Cytokines and Aquaporins. Inflammation 2019, 42, 2037–2047. [Google Scholar] [CrossRef] [PubMed]
- Fereydouni, N.; Darroudi, M.; Movaffagh, J.; Shahroodi, A.; Butler, A.E.; Ganjali, S.; Sahebkar, A. Curcumin nanofibers for the purpose of wound healing. J. Cell. Physiol. 2019, 234, 5537–5554. [Google Scholar] [CrossRef] [PubMed]
- Tomeh, M.A.; Hadianamrei, R.; Zhao, X. A Review of Curcumin and Its Derivatives as Anticancer Agents. Int. J. Mol. Sci. 2019, 20, 1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saleh, M.M.; Darwish, Z.E.; El Nouaem, M.I.; Mourad, G.M.; Ramadan, O.R. Chemopreventive effect of green tea and curcumin in induced oral squamous cell carcinoma: An experimental study. Alex. Dent. J. 2020, 45, 74–80. [Google Scholar] [CrossRef] [Green Version]
- Teng, C.-F.; Yu, C.-H.; Chang, H.-Y.; Hsieh, W.-C.; Wu, T.-H.; Lin, J.-H.; Wu, H.-C.; Jeng, L.-B.; Su, I.-J. Chemopreventive Effect of Phytosomal Curcumin on Hepatitis B Virus-Related Hepatocellular Carcinoma in A Transgenic Mouse Model. Sci. Rep. 2019, 9, 10338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shishodia, S.; Singh, T.; Chaturvedi, M.M. Modulation of transcription factors by curcumin. Adv. Exp. Med. Biol. 2007, 595, 127–148. [Google Scholar] [CrossRef] [PubMed]
- Golonko, A.; Lewandowska, H.; Świsłocka, R.; Jasińska, U.T.; Priebe, W.; Lewandowski, W. Curcumin as tyrosine kinase inhibitor in cancer treatment. Eur. J. Med. Chem. 2019, 181, 111512. [Google Scholar] [CrossRef] [PubMed]
- Soung, Y.H.; Chung, J. Curcumin inhibition of the functional interaction between integrin α6β4 and the epidermal growth factor receptor. Mol. Cancer Ther. 2011, 10, 883–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palizgir, M.T.; Akhtari, M.; Mahmoudi, M.; Mostafaei, S.; Rezaiemanesh, A.; Shahram, F. Curcumin reduces the expression of interleukin 1β and the production of interleukin 6 and tumor necrosis factor alpha by M1 macrophages from patients with Behcet’s disease. Immunopharmacol. Immunotoxicol. 2018, 40, 297–302. [Google Scholar] [CrossRef]
- Mhillaj, E.; Tarozzi, A.; Pruccoli, L.; Cuomo, V.; Trabace, L.; Mancuso, C. Curcumin and Heme Oxygenase: Neuroprotection and Beyond. Int. J. Mol. Sci. 2019, 20, 2419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, H.; Qiao, F.; Wang, Y.; Xu, Y.; Shang, Y. Curcumin inhibits cell proliferation and induces apoptosis of human non-small cell lung cancer cells through the upregulation of miR-192-5p and suppression of PI3K/Akt signaling pathway. Oncol. Rep. 2015, 34. [Google Scholar] [CrossRef] [Green Version]
- Hasima, N.; Aggarwal, B.B. Cancer-linked targets modulated by curcumin. Int. J. Biochem. Mol. Biol. 2012, 3, 328–351. [Google Scholar]
- Rainey, N.; Motte, L.; Aggarwal, B.B.; Petit, P.X. Curcumin hormesis mediates a cross-talk between autophagy and cell death. Cell Death Dis. 2015, 6, e2003. [Google Scholar] [CrossRef]
- Sun, L.-R.; Zhou, W.; Zhang, H.-M.; Guo, Q.-S.; Yang, W.; Li, B.-J.; Sun, Z.-H.; Gao, S.-H.; Cui, R.-J. Modulation of Multiple Signaling Pathways of the Plant-Derived Natural Products in Cancer. Front. Oncol. 2019, 9, 1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the golden nutraceutical: Multitargeting for multiple chronic diseases. Br. J. Pharmacol. 2016, 174, 1325–1348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hassan, F.; Rehman, M.S.; Khan, M.S.; Ali, M.A.; Javed, A.; Nawaz, A.; Yang, C. Curcumin as an Alternative Epigenetic Modulator: Mechanism of Action and Potential Effects. Front. Genet. 2019, 10, 514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khatri, D.K.; Juvekar, A.R. Kinetics of Inhibition of Monoamine Oxidase Using Curcumin and Ellagic Acid. Pharmacogn. Mag. 2016, 12, S116–S120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rabiei, Z.; Solati, K.; Amini-Khoei, H. Phytotherapy in treatment of Parkinson’s disease: A review. Pharm. Biol. 2019, 57, 355–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nguyen, T.T.; Vuu, M.D.; Huynh, M.A.; Yamaguchi, M.; Tran, L.T.; Dang, T.P.T. Curcumin Effectively Rescued Parkinson’s Disease-Like Phenotypes in a Novel Drosophila melanogaster Model with dUCH Knockdown. Oxid. Med. Cell. Longev. 2018, 2018, 2038267. [Google Scholar] [CrossRef] [Green Version]
- Sharma, N.; Nehru, B. Curcumin affords neuroprotection and inhibits α-synuclein aggregation in lipopolysaccharide-induced Parkinson’s disease model. Inflammopharmacology 2018, 26, 349–360. [Google Scholar] [CrossRef] [PubMed]
- Di Martino, R.M.C.; Pruccoli, L.; Bisi, A.; Gobbi, S.; Rampa, A.; Martinez, A.; Pérez, C.; Martinez-Gonzalez, L.; Paglione, M.; Di Schiavi, E.; et al. Novel Curcumin-Diethyl Fumarate Hybrid as a Dualistic GSK-3β Inhibitor/Nrf2 Inducer for the Treatment of Parkinson’s Disease. ACS Chem. Neurosci. 2020, 11, 2728–2740. [Google Scholar] [CrossRef]
- Ramires Júnior, O.V.; da Alves, B.S.; Barros, P.A.B.; Rodrigues, J.L.; Ferreira, S.P.; Monteiro, L.K.S.; de Araújo, G.M.S.; Fernandes, S.S.; Vaz, G.R.; Dora, C.L.; et al. Nanoemulsion Improves the Neuroprotective Effects of Curcumin in an Experimental Model of Parkinson’s Disease. Neurotox. Res. 2021, 39, 787–799. [Google Scholar] [CrossRef]
- Abrahams, S.; Miller, H.C.; Lombard, C.; van der Westhuizen, F.H.; Bardien, S. Curcumin pre-treatment may protect against mitochondrial damage in LRRK2-mutant Parkinson’s disease and healthy control fibroblasts. Biochem. Biophys. Rep. 2021, 27, 101035. [Google Scholar] [CrossRef]
- Motawi, T.K.; Sadik, N.A.H.; Hamed, M.A.; Ali, S.A.; Khalil, W.K.B.; Ahmed, Y.R. Potential therapeutic effects of antagonizing adenosine A2A receptor, curcumin and niacin in rotenone-induced Parkinson’s disease mice model. Mol. Cell. Biochem. 2020, 465, 89–102. [Google Scholar] [CrossRef]
- El Nebrisi, E.; Javed, H.; Ojha, S.K.; Oz, M.; Shehab, S. Neuroprotective Effect of Curcumin on the Nigrostriatal Pathway in a 6-Hydroxydopmine-Induced Rat Model of Parkinson’s Disease is Mediated by α7-Nicotinic Receptors. Int. J. Mol. Sci. 2020, 21, 7329. [Google Scholar] [CrossRef] [PubMed]
- Zbarsky, V.; Datla, K.P.; Parkar, S.; Rai, D.K.; Aruoma, O.I.; Dexter, D.T. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson’s disease. Free Radic. Res. 2005, 39, 1119–1125. [Google Scholar] [CrossRef]
- Agrawal, S.S.; Gullaiya, S.; Dubey, V.; Singh, V.; Kumar, A.; Nagar, A.; Tiwari, P. Neurodegenerative Shielding by Curcumin and Its Derivatives on Brain Lesions Induced by 6-OHDA Model of Parkinson’s Disease in Albino Wistar Rats. Cardiovasc. Psychiatry Neurol. 2012, 2012, 942981. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Song, S.; Li, J.; Liang, T. Neuroprotective effect of curcumin on hippocampal injury in 6-OHDA-induced Parkinson’s disease rat. Pathol. Res. Pract. 2014, 210, 357–362. [Google Scholar] [CrossRef]
- Velmurugan, B.K.; Rathinasamy, B.; Lohanathan, B.P.; Thiyagarajan, V.; Weng, C.-F. Neuroprotective Role of Phytochemicals. Mol. J. Synth. Chem. Nat. Prod. Chem. 2018, 23, 2485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tirthraj, B.; Rohit, S. Role of Curcumin in Regulation of TNF-α Mediated Brain Inflammatory Responses. Recent Pat. Inflamm. Allergy Drug Discov. 2018, 12, 69–77. [Google Scholar]
- Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
- Nosalski, R.; Guzik, T.J. Perivascular adipose tissue inflammation in vascular disease. Br. J. Pharmacol. 2017, 174, 3496–3513. [Google Scholar] [CrossRef] [Green Version]
- Ugalde-Muñiz, P.; Fetter-Pruneda, I.; Navarro, L.; García, E.; Chavarría, A. Chronic Systemic Inflammation Exacerbates Neurotoxicity in a Parkinson’s Disease Model. Oxid. Med. Cell. Longev. 2020, 2020, 4807179. [Google Scholar] [CrossRef] [Green Version]
- Gelders, G.; Baekelandt, V.; Van der Perren, A. Linking Neuroinflammation and Neurodegeneration in Parkinson’s Disease. J. Immunol. Res. 2018, 2018, 4784268. [Google Scholar] [CrossRef] [Green Version]
- Lee, Y.; Lee, S.; Chang, S.-C.; Lee, J. Significant roles of neuroinflammation in Parkinson’s disease: Therapeutic targets for PD prevention. Arch. Pharm. Res. 2019, 42, 416–425. [Google Scholar] [CrossRef]
- Vivekanantham, S.; Shah, S.; Dewji, R.; Dewji, A.; Khatri, C.; Ologunde, R. Neuroinflammation in Parkinson’s disease: Role in neurodegeneration and tissue repair. Int. J. Neurosci. 2015, 125, 717–725. [Google Scholar] [CrossRef]
- Yan, J.; Fu, Q.; Cheng, L.; Zhai, M.; Wu, W.; Huang, L.; Du, G. Inflammatory response in Parkinson’s disease (Review). Mol. Med. Rep. 2014, 10, 2223–2233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Creţu, E.; Trifan, A.; Vasincu, A.; Miron, A. Plant-derived anticancer agents—Curcumin in cancer prevention and treatment. Rev. Med. Chir. Soc. Med. Nat. Iasi 2012, 116, 1223–1229. [Google Scholar] [PubMed]
- Ghasemi, F.; Bagheri, H.; Barreto, G.E.; Read, M.I.; Sahebkar, A. Effects of Curcumin on Microglial Cells. Neurotox. Res. 2019, 36, 12–26. [Google Scholar] [CrossRef] [PubMed]
- Fan, C.; Song, Q.; Wang, P.; Li, Y.; Yang, M.; Yu, S.Y. Neuroprotective Effects of Curcumin on IL-1β-Induced Neuronal Apoptosis and Depression-Like Behaviors Caused by Chronic Stress in Rats. Front. Cell. Neurosci. 2018, 12, 516. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, S.; Banerjee, S.; Sil, P.C. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: A recent update. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2015, 83, 111–124. [Google Scholar] [CrossRef] [PubMed]
- Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18. [Google Scholar] [CrossRef] [Green Version]
- Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef]
- Kumar, H.; Lim, H.-W.; More, S.V.; Kim, B.-W.; Koppula, S.; Kim, I.S.; Choi, D.-K. The Role of Free Radicals in the Aging Brain and Parkinson’s Disease: Convergence and Parallelism. Int. J. Mol. Sci. 2012, 13, 10478–10504. [Google Scholar] [CrossRef] [Green Version]
- Alisi, I.O.; Uzairu, A.; Abechi, S.E.; Idris, S.O. Evaluation of the antioxidant properties of curcumin derivatives by genetic function algorithm. J. Adv. Res. 2018, 12, 47–54. [Google Scholar] [CrossRef]
- Jakubczyk, K.; Drużga, A.; Katarzyna, J.; Skonieczna-Żydecka, K. Antioxidant Potential of Curcumin-A Meta-Analysis of Randomized Clinical Trials. Antioxid. Basel Switz. 2020, 9, 1092. [Google Scholar] [CrossRef] [PubMed]
- Sökmen, M.; Akram Khan, M. The antioxidant activity of some curcuminoids and chalcones. Inflammopharmacology 2016, 24, 81–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, S.; Nie, Q.; Li, Z.; Du, G. Curcumin improves neurofunctions of 6-OHDA-induced parkinsonian rats. Pathol. Res. Pract. 2016, 212, 247–251. [Google Scholar] [CrossRef]
- Khuwaja, G.; Khan, M.M.; Ishrat, T.; Ahmad, A.; Raza, S.S.; Ashafaq, M.; Javed, H.; Khan, M.B.; Khan, A.; Vaibhav, K.; et al. Neuroprotective effects of curcumin on 6-hydroxydopamine-induced Parkinsonism in rats: Behavioral, neurochemical and immunohistochemical studies. Brain Res. 2011, 1368, 254–263. [Google Scholar] [CrossRef]
- Muthian, G.; Mackey, V.; Prasad, K.; Charlton, C. Curcumin and an antioxidant formulation protect C57BL/6J mice from MPTP-induced Parkinson’s disease like changes: Potential neuroprotection for neurodegeneration. Res. Rev. Park. 2018, 8, 49–59. [Google Scholar] [CrossRef] [Green Version]
- Arredondo, S.B.; Valenzuela-Bezanilla, D.; Mardones, M.D.; Varela-Nallar, L. Role of Wnt Signaling in Adult Hippocampal Neurogenesis in Health and Disease. Front. Cell Dev. Biol. 2020, 8, 860. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.-L.; Ju, B.; Zhang, Y.-Z.; Yin, H.-L.; Liu, Y.-J.; Wang, S.-S.; Zeng, Z.-L.; Yang, X.-P.; Wang, H.-T.; Li, J.-F. Protective Effect of Curcumin Against Oxidative Stress-Induced Injury in Rats with Parkinson’s Disease Through the Wnt/ β-Catenin Signaling Pathway. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2017, 43, 2226–2241. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Ahmadi, Z.; Mohamamdinejad, R.; Yaribeygi, H.; Serban, M.-C.; Orafai, H.M.; Sahebkar, A. Curcumin Therapeutic Modulation of the Wnt Signaling Pathway. Curr. Pharm. Biotechnol. 2020, 21, 1006–1015. [Google Scholar] [CrossRef] [PubMed]
- Malik, P.; Mukherjee, T.K. Structure-Function Elucidation of Antioxidative and Prooxidative Activities of the Polyphenolic Compound Curcumin. Chin. J. Biol. 2014, 2014, e396708. [Google Scholar] [CrossRef] [Green Version]
- Davies, J.M.S.; Cillard, J.; Friguet, B.; Cadenas, E.; Cadet, J.; Cayce, R.; Fishmann, A.; Liao, D.; Bulteau, A.-L.; Derbré, F.; et al. The Oxygen Paradox, the French Paradox, and age-related diseases. GeroScience 2017, 39, 499–550. [Google Scholar] [CrossRef] [Green Version]
- Davies, K.J.A. The Oxygen Paradox, Oxidative Stress, and Ageing. Arch. Biochem. Biophys. 2016, 595, 28–32. [Google Scholar] [CrossRef] [Green Version]
- Alam, Z.I.; Daniel, S.E.; Lees, A.J.; Marsden, D.C.; Jenner, P.; Halliwell, B. A generalised increase in protein carbonyls in the brain in Parkinson’s but not incidental Lewy body disease. J. Neurochem. 1997, 69, 1326–1329. [Google Scholar] [CrossRef]
- Dexter, D.; Carter, C.; Agid, F.; Agid, Y.; Lees, A.J.; Jenner, P.; Marsden, C.D. Lipid peroxidation as cause of nigral cell death in Parkinson’s disease. Lancet Lond. Engl. 1986, 2, 639–640. [Google Scholar] [CrossRef]
- Buratta, S.; Chiaradia, E.; Tognoloni, A.; Gambelunghe, A.; Meschini, C.; Palmieri, L.; Muzi, G.; Urbanelli, L.; Emiliani, C.; Tancini, B. Effect of Curcumin on Protein Damage Induced by Rotenone in Dopaminergic PC12 Cells. Int. J. Mol. Sci. 2020, 21, 2761. [Google Scholar] [CrossRef]
- Di Meo, S.; Venditti, P. Evolution of the Knowledge of Free Radicals and Other Oxidants. Oxid. Med. Cell. Longev. 2020, 2020, 9829176. [Google Scholar] [CrossRef]
- Dantuma, N.P.; Bott, L.C. The ubiquitin-proteasome system in neurodegenerative diseases: Precipitating factor, yet part of the solution. Front. Mol. Neurosci. 2014, 7, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Treml, J.; Karel, Š. Flavonoids as Potent Scavengers of Hydroxyl Radicals: Flavonoids versus hydroxyl radical…. Compr. Rev. Food Sci. Food Saf. 2016, 15, 720–738. [Google Scholar] [CrossRef]
- Takahashi, R.; Ono, K.; Takamura, Y.; Mizuguchi, M.; Ikeda, T.; Nishijo, H.; Yamada, M. Phenolic compounds prevent the oligomerization of α-synuclein and reduce synaptic toxicity. J. Neurochem. 2015, 134, 943–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Exner, N.; Lutz, A.K.; Haass, C.; Winklhofer, K.F. Mitochondrial dysfunction in Parkinson’s disease: Molecular mechanisms and pathophysiological consequences. EMBO J. 2012, 31, 3038–3062. [Google Scholar] [CrossRef] [Green Version]
- Son, G.; Han, J. Roles of mitochondria in neuronal development. BMB Rep. 2018, 51, 549–556. [Google Scholar] [CrossRef] [Green Version]
- Grünewald, A.; Kumar, K.R.; Sue, C.M. New insights into the complex role of mitochondria in Parkinson’s disease. Prog. Neurobiol. 2019, 177, 73–93. [Google Scholar] [CrossRef] [PubMed]
- Matsui, H.; Uemura, N.; Yamakado, H.; Takeda, S.; Takahashi, R. Exploring the pathogenetic mechanisms underlying Parkinson’s disease in medaka fish. J. Park. Dis. 2014, 4, 301–310. [Google Scholar] [CrossRef] [PubMed]
- Scarffe, L.A.; Stevens, D.A.; Dawson, V.L.; Dawson, T.M. Parkin and PINK1: Much more than mitophagy. Trends Neurosci. 2014, 37, 315–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khatri, D.K.; Juvekar, A.R. Neuroprotective effect of curcumin as evinced by abrogation of rotenone-induced motor deficits, oxidative and mitochondrial dysfunctions in mouse model of Parkinson’s disease. Pharmacol. Biochem. Behav. 2016, 150–151, 39–47. [Google Scholar] [CrossRef]
- Zia, A.; Farkhondeh, T.; Pourbagher-Shahri, A.M.; Samarghandian, S. The role of curcumin in aging and senescence: Molecular mechanisms. Biomed. Pharmacother. Biomedecine Pharmacother. 2021, 134, 111119. [Google Scholar] [CrossRef]
- Malhotra, A.; Nair, P.; Dhawan, D.K. Premature mitochondrial senescence and related ultrastructural changes during lung carcinogenesis modulation by curcumin and resveratrol. Ultrastruct. Pathol. 2012, 36, 179–184. [Google Scholar] [CrossRef]
- van der Merwe, C.; van Dyk, H.C.; Engelbrecht, L.; van der Westhuizen, F.H.; Kinnear, C.; Loos, B.; Bardien, S. Curcumin Rescues a PINK1 Knock Down SH-SY5Y Cellular Model of Parkinson’s Disease from Mitochondrial Dysfunction and Cell Death. Mol. Neurobiol. 2017, 54, 2752–2762. [Google Scholar] [CrossRef]
- Abbaoui, A.; Gamrani, H. Neuronal, astroglial and locomotor injuries in subchronic copper intoxicated rats are repaired by curcumin: A possible link with Parkinson’s disease. Acta Histochem. 2018, 120, 542–550. [Google Scholar] [CrossRef]
- Ferreira, A.; Neves, P.; Gozzelino, R. Multilevel Impacts of Iron in the Brain: The Cross Talk between Neurophysiological Mechanisms, Cognition, and Social Behavior. Pharmaceuticals 2019, 12, 126. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.-L.; Huang, Y.-J.; Cui, J.-T.; Song, N.; Xie, J. Iron Dysregulation in Parkinson’s Disease: Focused on the Autophagy-Lysosome Pathway. ACS Chem. Neurosci. 2019, 10, 863–871. [Google Scholar] [CrossRef]
- Zucca, F.A.; Segura-Aguilar, J.; Ferrari, E.; Muñoz, P.; Paris, I.; Sulzer, D.; Sarna, T.; Casella, L.; Zecca, L. INTERACTIONS OF IRON, DOPAMINE AND NEUROMELANIN PATHWAYS IN BRAIN AGING AND PARKINSON’S DISEASE. Prog. Neurobiol. 2017, 155, 96–119. [Google Scholar] [CrossRef]
- Thomas, G.E.C.; Leyland, L.A.; Schrag, A.-E.; Lees, A.J.; Acosta-Cabronero, J.; Weil, R.S. Brain iron deposition is linked with cognitive severity in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 2020, 91, 418–425. [Google Scholar] [CrossRef] [Green Version]
- Gröger, A.; Berg, D. Does structural neuroimaging reveal a disturbance of iron metabolism in Parkinson’s disease? Implications from MRI and TCS studies. J. Neural Transm. Vienna Austria 1996 2012, 119, 1523–1528. [Google Scholar] [CrossRef]
- Rainey, N.E.; Moustapha, A.; Saric, A.; Nicolas, G.; Sureau, F.; Petit, P.X. Iron chelation by curcumin suppresses both curcumin-induced autophagy and cell death together with iron overload neoplastic transformation. Cell Death Discov. 2019, 5, 1–15. [Google Scholar] [CrossRef]
- Du, X.-X.; Xu, H.-M.; Jiang, H.; Song, N.; Wang, J.; Xie, J.-X. Curcumin protects nigral dopaminergic neurons by iron-chelation in the 6-hydroxydopamine rat model of Parkinson’s disease. Neurosci. Bull. 2012, 28, 253–258. [Google Scholar] [CrossRef] [Green Version]
- Lv, H.; Liu, J.; Wang, L.; Zhang, H.; Yu, S.; Li, Z.; Jiang, F.; Niu, Y.; Yuan, J.; Cui, X.; et al. Ameliorating Effects of Combined Curcumin and Desferrioxamine on 6-OHDA-Induced Rat Mode of Parkinson’s Disease. Cell Biochem. Biophys. 2014, 70, 1433–1438. [Google Scholar] [CrossRef]
- Mursaleen, L.; Somavarapu, S.; Zariwala, M.G. Deferoxamine and Curcumin Loaded Nanocarriers Protect Against Rotenone-Induced Neurotoxicity. J. Park. Dis. 2020, 10, 99–111. [Google Scholar] [CrossRef]
- Zhang, X.; Chen, Q.; Wang, Y.; Peng, W.; Cai, H. Effects of curcumin on ion channels and transporters. Front. Physiol. 2014, 5, 94. [Google Scholar] [CrossRef] [Green Version]
- El Nebrisi, E.G.; Bagdas, D.; Toma, W.; Al Samri, H.; Brodzik, A.; Alkhlaif, Y.; Yang, K.-H.S.; Howarth, F.C.; Damaj, I.M.; Oz, M. Curcumin Acts as a Positive Allosteric Modulator of α7-Nicotinic Acetylcholine Receptors and Reverses Nociception in Mouse Models of Inflammatory Pain. J. Pharmacol. Exp. Ther. 2018, 365, 190–200. [Google Scholar] [CrossRef] [Green Version]
- Nebrisi, E.E.; Al Kury, L.T.; Yang, K.-H.S.; Jayaprakash, P.; Howarth, F.C.; Kabbani, N.; Oz, M. Curcumin potentiates the function of human α7-nicotinic acetylcholine receptors expressed in SH-EP1 cells. Neurochem. Int. 2018, 114, 80–84. [Google Scholar] [CrossRef]
- Corradi, J.; Bouzat, C. Understanding the Bases of Function and Modulation of α7 Nicotinic Receptors: Implications for Drug Discovery. Mol. Pharmacol. 2016, 90, 288–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uteshev, V.V. The therapeutic promise of positive allosteric modulation of nicotinic receptors. Eur. J. Pharmacol. 2014, 727, 181–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maex, R.; Grinevich, V.P.; Grinevich, V.; Budygin, E.; Bencherif, M.; Gutkin, B. Understanding the Role α7 Nicotinic Receptors Play in Dopamine Efflux in Nucleus Accumbens. ACS Chem. Neurosci. 2014, 5, 1032–1040. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Q.; Yakel, J.L. Presynaptic α7 Nicotinic Acetylcholine Receptors Enhance Hippocampal Mossy Fiber Glutamatergic Transmission via PKA Activation. J. Neurosci. 2014, 34, 124–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Z.-Q.; Zhang, W.-J.; Su, D.-F.; Zhang, G.-Q.; Miao, C.-Y. Cellular responses and functions of α7 nicotinic acetylcholine receptor activation in the brain: A narrative review. Ann. Transl. Med. 2021, 9, 509. [Google Scholar] [CrossRef]
- Kanasaki, H.; Purwana, I.; Oride, A.; Mijiddorj, T.; Miyazaki, K. Extracellular Signal-Regulated Kinase (ERK) Activation and Mitogen-Activated Protein Kinase Phosphatase 1 Induction by Pulsatile Gonadotropin-Releasing Hormone in Pituitary Gonadotrophs. J. Signal Transduct. 2011, 2012, e198527. [Google Scholar] [CrossRef] [Green Version]
- Gu, X.; Liu, L.; Shen, Q.; Xing, D. Photoactivation of ERK/CREB/VMAT2 pathway attenuates MPP+-induced neuronal injury in a cellular model of Parkinson’s disease. Cell. Signal. 2017, 37, 103–114. [Google Scholar] [CrossRef]
- Mohammadi, A.; Amooeian, V.G.; Rashidi, E. Dysfunction in Brain-Derived Neurotrophic Factor Signaling Pathway and Susceptibility to Schizophrenia, Parkinson’s and Alzheimer’s Diseases. Curr. Gene Ther. 2018, 18, 45–63. [Google Scholar] [CrossRef]
- Zhu, R.-L.; Zhi, Y.-K.; Yi, L.; Luo, J.-F.; Li, J.; Bai, S.-S.; Liu, L.; Wang, P.-X.; Zhou, H.; Dong, Y. Sinomenine regulates CD14/TLR4, JAK2/STAT3 pathway and calcium signal via α7nAChR to inhibit inflammation in LPS-stimulated macrophages. Immunopharmacol. Immunotoxicol. 2019, 41, 172–177. [Google Scholar] [CrossRef]
- Lin, Z.; Zhang, P.-W.; Zhu, X.; Melgari, J.-M.; Huff, R.; Spieldoch, R.L.; Uhl, G.R. Phosphatidylinositol 3-kinase, protein kinase C, and MEK1/2 kinase regulation of dopamine transporters (DAT) require N-terminal DAT phosphoacceptor sites. J. Biol. Chem. 2003, 278, 20162–20170. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Li, X.-M.; Meinkoth, J.; Pittman, R.N. Akt Regulates Cell Survival and Apoptosis at a Postmitochondrial Level. J. Cell Biol. 2000, 151, 483–494. [Google Scholar] [CrossRef] [Green Version]
- Long, H.-Z.; Cheng, Y.; Zhou, Z.-W.; Luo, H.-Y.; Wen, D.-D.; Gao, L.-C. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer’s Disease and Parkinson’s Disease. Front. Pharmacol. 2021, 12, 648636. [Google Scholar] [CrossRef]
- Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin Regulates Anti-Inflammatory Responses by JAK/STAT/SOCS Signaling Pathway in BV-2 Microglial Cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef] [Green Version]
- Shrestha, T.; Takahashi, T.; Li, C.; Matsumoto, M.; Maruyama, H. Nicotine-induced upregulation of miR-132-5p enhances cell survival in PC12 cells by targeting the anti-apoptotic protein Bcl-2. Neurol. Res. 2020, 42, 405–414. [Google Scholar] [CrossRef] [PubMed]
- King’s College London. Parkinson’s Disease with Mild Cognitive Impairment Treated With Nicotinic Agonist Drug; clinicaltrials.gov: Online, 2021.
- Aggarwal, M.L.; Chacko, K.M.; Kuruvilla, B.T. Systematic and comprehensive investigation of the toxicity of curcuminoid-essential oil complex: A bioavailable turmeric formulation. Mol. Med. Rep. 2016, 13, 592–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganiger, S.; Malleshappa, H.N.; Krishnappa, H.; Rajashekhar, G.; Ramakrishna Rao, V.; Sullivan, F. A two generation reproductive toxicity study with curcumin, turmeric yellow, in Wistar rats. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2007, 45, 64–69. [Google Scholar] [CrossRef]
- Lao, C.D.; Ruffin, M.T.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef] [Green Version]
- Shankar, T.N.; Shantha, N.V.; Ramesh, H.P.; Murthy, I.A.; Murthy, V.S. Toxicity studies on turmeric (Curcuma longa): Acute toxicity studies in rats, guineapigs & monkeys. Indian J. Exp. Biol. 1980, 18, 73–75. [Google Scholar] [PubMed]
- Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P.S. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef] [Green Version]
- Dadhaniya, P.; Patel, C.; Muchhara, J.; Bhadja, N.; Mathuria, N.; Vachhani, K.; Soni, M.G. Safety assessment of a solid lipid curcumin particle preparation: Acute and subchronic toxicity studies. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2011, 49, 1834–1842. [Google Scholar] [CrossRef]
- Chinese University of Hong Kong. A Pilot Study of Curcumin and Ginkgo for Treating Alzheimer’s Disease; Clinical Trial Registration NCT00164749; clinicaltrials.gov: Online, 2008.
- Huang, D. Using Optical Coherence Tomography and Noninvasive Retinal Amyloid Imaging to Capture Retinal Changes Associated with Dementia; Clinical Trial Registration NCT03761381; clinicaltrials.gov: Online, 2021.
- Jaslok Hospital and Research Centre. Phase II Study of Curcumin Formulation (Longvida) or Placebo on Plasma Biomarkers and Mental State in Moderate to Severe Alzheimer’s Disease or Normal Cognition; Clinical Trial Registration NCT01001637; clinicaltrials.gov: Online, 2009.
- John Douglas French Foundation Phase II. Double-Blind, Placebo-Controlled Study of the Safety and Tolerability of Two Doses of Curcumin C3 Complex Versus Placebo in Patients with Mild to Moderate Alzheimer’s Disease; Clinical Trial Registration NCT00099710; clinicaltrials.gov: Online, 2009.
- Life Extension Foundation Inc. Open Label, Crossover, Pilot Study to Assess the Efficacy & Safety of Perispinal Admin. of Etanercept (Enbrel®) in Comb. w/Nutritional Supplements vs. Nutritional Supplements Alone in Subj. w/Mild to Mod. Alzheimer’s Disease Receiving Std. Care; Clinical Trial Registration NCT01716637; clinicaltrials.gov: Online, 2016.
- Mahidol University A Randomized. Double-Blind, Placebo-Controlled Trial of Curcumin in Leber’s Hereditary Optic Neuropathy (LHON); Clinical Trial Registration NCT00528151; clinicaltrials.gov: Online, 2012.
- Hanna, M. Monitoring of Early Disease Progression in Hereditary Transthyretin Amyloidosis; Clinical Trial Registration NCT03431896; clinicaltrials.gov: Online, 2021.
- Ortí, J.E.R. Impact of the Combined Treatment of Curcumin and Resveratrol Liposomed Polyphenols with G04CB02 on the Clinical Improvement of ALS Patients; Clinical trial registration NCT04654689; clinicaltrials.gov: Online, 2021.
- Bedlack, R. An Open-label, Single-center, 6-month Trial of Theracurmin for Patients with Amyotrophic Lateral Sclerosis (ALS); Clinical trial registration NCT04499963; clinicaltrials.gov: Online, 2021.
- Preventive Medicine Research Institute. Lifestyle Intervention for Early Alzheimer’s Disease; Clinical trial registration NCT04606420; clinicaltrials.gov: Online, 2020.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Nebrisi, E.E. Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature. Int. J. Mol. Sci. 2021, 22, 11248. https://doi.org/10.3390/ijms222011248
Nebrisi EE. Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature. International Journal of Molecular Sciences. 2021; 22(20):11248. https://doi.org/10.3390/ijms222011248
Chicago/Turabian StyleNebrisi, Eslam El. 2021. "Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature" International Journal of Molecular Sciences 22, no. 20: 11248. https://doi.org/10.3390/ijms222011248
APA StyleNebrisi, E. E. (2021). Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature. International Journal of Molecular Sciences, 22(20), 11248. https://doi.org/10.3390/ijms222011248