The Multiple Sclerosis Modulatory Potential of Natural Multi-Targeting Antioxidants
<p>Structures of referred flavonoids.</p> "> Figure 2
<p>Epigallocatechin gallate, Arbutin and Arctigenin.</p> "> Figure 3
<p>Oleuropein and its secoiridoid derivative, oleacein.</p> "> Figure 4
<p>Ellagic acid, resveratrol and enol-keto form of curcumin.</p> "> Figure 5
<p>alpha-tocopherol, trolox (a widely used in medicinal chemistry synthetic derivative) and TFA-12.</p> "> Figure 6
<p>Vitamin A and Carotenoids with anti-MS potency.</p> "> Figure 7
<p>Punicic acid, general structure of plasmalogens (R’ = R’’ = R’’’ = -CH<sub>3</sub> or -H, R<sub>1</sub> = ω-3 or ω-6 lipid moiety, R<sub>2</sub> = saturated or mono-unsaturated lipid moiety) and linoleic acid with curcumin synthetic derivative Lino-Cur.</p> "> Figure 8
<p>N-acetylcysteine and the properties of its ethyl ester.</p> "> Figure 9
<p>Hydrogen sulfide donating effect of 3H-1,2-ditione-3-thione and its derivative ACDT.</p> "> Figure 10
<p>Structures of S-allyl-L-cysteine, Sulforaphane and Moringin.</p> "> Figure 11
<p>Potential levels of implication of natural antioxidants in the MS initiation and progression.</p> ">
Abstract
:1. Introduction
2. Inflammation and Oxidative Stress in MS
3. Natural Phenolic Antioxidants
3.1. Quercetin
3.2. Isoflavones
3.3. Epigallocatechin Gallate
3.4. Arbutin
3.5. Arctigenin
3.6. Oleuropein
3.7. Ellagic Acid
3.8. Resveratrol
3.9. Curcumin
4. Vitamins
4.1. Vitamin E
4.2. Vitamin A and Carotenoids
5. Polyunsaturated Fatty Acids
6. Sulfur Containing Antioxidants
6.1. N-Acetylcysteine
6.2. S-allyl-L-cysteine
6.3. H-1,2-dithiole-3-thione
6.4. A-Lipoic Acid
6.5. Biotin
6.6. Thiamine
6.7. Sulfarophane and Moringin
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Goldenberg, M.M. Multiple sclerosis review. Pharm. Ther. 2012, 37, 175–184. [Google Scholar]
- Ohl, K.; Tenbrock, K.; Kipp, M. Oxidative stress in multiple sclerosis: Central and peripheral mode of action. Exp. Neurol. 2016, 277, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Kempuraj, D.; Thangavel, R.; Natteru, P.A.; Selvakumar, G.P.; Saeed, D.; Zahoor, H.; Zaheer, S.; Iyer, S.S.; Zaheer, A. Neuroinflammation induces neurodegeneration. J. Neurol. Neurosurg. Spine 2016, 1, 1003. [Google Scholar] [PubMed]
- Lublin, F.D.; Reingold, S.C.; Cohen, J.A.; Cutter, G.R.; Sørensen, S.P.; Thompson, A.J.; Wolinsky, J.S.; Balcer, L.J.; Banwell, B.; Barkhof, F.; et al. Defining the clinical course of multiple sclerosis. The 2013 revisions. Neurology 2014, 83, 278–286. [Google Scholar] [CrossRef] [Green Version]
- Dutta, R.; Trapp, B.D. Relapsing and progressive forms of multiple sclerosis—Insights from pathology. Curr. Opin. Neurol. 2014, 27, 271–278. [Google Scholar] [CrossRef] [Green Version]
- Miller, E.D.; Dziedzic, A.; Saluk-Bijak, J.; Bijak, M. A Review of Various Antioxidant Compounds and their Potential Utility as Complementary Therapy in Multiple Sclerosis. Nutrients 2019, 11, 1528. [Google Scholar] [CrossRef] [Green Version]
- Yadav, S.K.; Mindur, J.E.; Ito, K.; Dhib-Jalbut, S. Advances in the immunopathogenesis of multiple sclerosis. Curr. Opin. Neurol. 2015, 28, 206–219. [Google Scholar] [CrossRef]
- Sun, Y.; Langer, H.F. Platelets, Thromboinflammation and Neurovascular Disease. Front. Immunol. 2022, 13, 843404. [Google Scholar] [CrossRef]
- Raphael, I.; Nalawade, S.; Eagar, T.N.; Forsthuber, T.G. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 2015, 74, 5–17. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.; Du, S.; Zhao, L.; Jain, S.; Sahay, K.; Rizvanov, A.; Lezhnyova, V.; Khaibullin, T.; Martynova, E.; Khaiboullina, S.; et al. Autoreactive lymphocytes in multiple sclerosis: Pathogenesis and treatment target. Front. Immunol. 2022, 13, 996469. [Google Scholar] [CrossRef]
- Van Horssen, J.; Witte, M.E.; Schreibelt, G.; de Vries, H.E. Radical changes in multiple sclerosis pathogenesis. Biochim. Biophys. Acta 2011, 1812, 141–150. [Google Scholar] [CrossRef]
- Covarrubias, A.; Byles, V.; Horng, T. ROS sets the stage for macrophage differentiation. Cell Res. 2013, 23, 984–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Neill, L.A.; Kishton, R.J.; Rathmell, J. A Guide to Immunometabolism for Immunologists. Nat. Rev. Immunol. 2016, 16, 553–565. [Google Scholar] [CrossRef] [Green Version]
- Papagiouvannis, G.; Theodosis-Nobelos, P.; Kourounakis, P.N.; Rekka, E.A. Multi-Target Directed Compounds with Antioxidant and/or Anti- Inflammatory Properties as Potent Agents for Alzheimer’s Disease. Med. Chem. 2021, 17, 1086–1103. [Google Scholar] [CrossRef]
- Radandish, M.; Khalilian, P.; Esmaeil, N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front. Immunol. 2021, 20, 667705. [Google Scholar] [CrossRef] [PubMed]
- Lassmann, H.; van Horssen, J. Oxidative stress and its impact on neurons and glia in multiple sclerosis lesions. Biochim. Biophys. Acta 2016, 1862, 506–510. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burté, F.; Carelli, V.; Chinnery, P.F.; Yu-Wai-Man, P. Disturbed mitochondrial dynamics and neurodegenerative disorders. Nat. Rev. Neurol. 2015, 11, 11–24. [Google Scholar] [CrossRef]
- Sorce, S.; Krause, K.H. NOX enzymes in the central nervous system: From signaling to disease. Antioxid. Redox Signal 2009, 11, 2481–2504. [Google Scholar] [CrossRef]
- Madigan, C.A.; Cambier, C.J.; Kelly-Scumpia, K.M.; Scumpia, P.O.; Cheng, T.Y.; Zailaa, J.; Bloom, B.R.; Moody, D.B.; Smale, S.T.; Sagasti, A.; et al. A macrophage response to Mycobacterium leprae phenolic glycolipid initiates nerve damage in leprosy. Cell 2017, 170, 973–985.e10. [Google Scholar] [CrossRef] [Green Version]
- Hedström, A.K.; Hössjer, O.; Katsoulis, M.; Kockum, I.; Olsson, T.; Alfredsson, L. Organic solvents and MS susceptibility. Interaction with MS risk HLA genes. Neurology 2018, 91, e455–e462. [Google Scholar] [CrossRef] [PubMed]
- Elfawy, H.A.; Das, B. Crosstalk between mitochondrial dysfunction, oxidative stress, and age related neurodegenerative disease: Etiologies and therapeutic strategies. Life Sci. 2019, 218, 165–184. [Google Scholar] [CrossRef] [PubMed]
- Winiarska-Mieczan, A.; Baranowska-Wójcik, E.; Kwiecień, M.; Grela, E.R.; Szwajgier, D.; Kwiatkowska, K.; Kiczorowska, B. The Role of Dietary Antioxidants in the Pathogenesis of Neurodegenerative Diseases and Their Impact on Cerebral Oxidoreductive Balance. Nutrients 2020, 12, 435. [Google Scholar] [CrossRef] [Green Version]
- Ortiz, G.G.; Pacheco-Moisés, F.P.; Bitzer-Quintero, O.K.; Ramírez-Anguiano, A.C.; Flores-Alvarado, L.J.; Ramírez-Ramírez, V.; Macias-Islas, M.A.; Torres-Sánchez, E.D. Immunology and oxidative stress in multiple sclerosis: Clinical and basic approach. Clin. Dev. Immunol. 2013, 2013, 708659. [Google Scholar] [CrossRef] [Green Version]
- Ghonimi, N.A.M.; Elsharkawi, K.A.; Khyal, D.S.M.; Abdelghani, A.A. Serum malondialdehyde as a lipid peroxidation marker in multiple sclerosis patients and its relation to disease characteristics. Mult. Scler. Relat. Disord. 2021, 51, 102941. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.Y.; Gui, L.N.; Liu, Y.Y.; Shi, S.; Cheng, Y. Oxidative Stress Marker Aberrations in Multiple Sclerosis: A Meta-Analysis Study. Front. Neurosci. 2020, 14, 823. [Google Scholar] [CrossRef] [PubMed]
- Zinovkin, R.A.; Romaschenko, V.P.; Galkin, I.I.; Zakharova, V.V.; Pletjushkina, O.Y.; Chernyak, B.V.; Popova, E.N. Role of mitochondrial reactive oxygen species in age-related inflammatory activation of endothelium. Aging 2014, 6, 661–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischer, M.T.; Wimmer, I.; Höftberger, R.; Gerlach, S.; Haider, L.; Zrzavy, T.; Hametner, S.; Mahad, D.; Binder, C.J.; Krumbholz, M.; et al. Disease-specific molecular events in cortical multiple sclerosis lesions. Brain 2013, 136, 1799–1815. [Google Scholar] [CrossRef]
- Adamczyk, B.; Adamczyk-Sowa, M. New Insights into the Role of Oxidative Stress Mechanisms in the Pathophysiology and Treatment of Multiple Sclerosis. Oxid. Med. Cell. Longev. 2016, 2016, 1973834. [Google Scholar] [CrossRef] [Green Version]
- Haider, L. Inflammation, Iron, Energy Failure, and Oxidative Stress in the Pathogenesis of Multiple Sclerosis. Oxid. Med. Cell. Longev. 2015, 2015, 725370. [Google Scholar] [CrossRef]
- Bamm, V.V.; Harauz, G. Hemoglobin as a source of iron overload in multiple sclerosis: Does multiple sclerosis share risk factors with vascular disorders? Cell. Mol. Life Sci. 2014, 71, 1789–1798. [Google Scholar] [CrossRef] [PubMed]
- Karak, P. Biological activities of flavonoids: An overview. IJPSR 2019, 3, 1567–1574. [Google Scholar]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banjarnahor, S.D.S.; Artant, N. Antioxidant properties of flavonoids. Med. J. Indones. 2014, 23, 239–244. [Google Scholar] [CrossRef] [Green Version]
- Ginwala, R.; Bhavsar, R.; Chigbu, D.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [Green Version]
- Dajas, F. Life or death: Neuroprotective and anticancer effects of quercetin. J. Ethnopharmacol. 2012, 143, 383–396. [Google Scholar] [CrossRef]
- Muthian, G.; Bright, J.J. Quercetin, a flavonoid phytoestrogen, ameliorates experimental allergic encephalomyelitis by blocking IL-12 signaling through JAK-STAT pathway in T lymphocyte. J. Clin. Immunol. 2004, 24, 542–552. [Google Scholar] [CrossRef]
- Sternberg, Z.; Chadha, K.; Lieberman, A.; Hojnacki, D.; Drake, A.; Zamboni, P.; Rocco, P.; Grazioli, E.; Weinstock-Guttman, B.; Munschauer, F. Quercetin and interferon-β modulate immune response (s) in peripheral blood mononuclear cells isolated from multiple sclerosis patients. J. Neuroimmunol. 2008, 205, 142–147. [Google Scholar] [CrossRef]
- Jensen, S.N.; Cady, N.M.; Shahi, S.K.; Peterson, S.R.; Gupta, A.; Gibson-Corley, K.N.; Mangalam, A.K. Isoflavone diet ameliorates experimental autoimmune encephalomyelitis through modulation of gut bacteria depleted in patients with multiple sclerosis. Sci. Adv. 2021, 7, eabd4595. [Google Scholar] [CrossRef]
- Choi, E.Y.; Jin, J.Y.; Lee, J.Y.; Choi, J.I.; Choi, I.; Kim, S.J. Anti-inflammatory effects and the underlying mechanisms of action of daidzein in murine macrophages stimulated with Prevotella intermedia lipopolysaccharide. J. Periodontal Res. 2012, 47, 204–211. [Google Scholar] [CrossRef]
- Jahromi, S.R.; Arrefhosseini, S.R.; Ghaemi, A.; Alizadeh, A.; Moradi Tabriz, H.; Togha, M. Alleviation of experimental allergic encephalomyelitis in C57BL/6 mice by soy daidzein. Iran. J. Allergy Asthma Immunol. 2014, 13, 256–264. [Google Scholar]
- El-Deeb, O.S.; Ghanem, H.B.; El-Esawy, R.O.; Sadek, M.T. The modulatory effects of luteolin on cyclic AMP/Ciliary neurotrophic factor signaling pathway in experimentally induced autoimmune encephalomyelitis. IUBMB Life 2019, 71, 1401–1408. [Google Scholar] [CrossRef] [PubMed]
- Contarini, G.; Franceschini, D.; Facci, L.; Barbierato, M.; Giusti, P.; Zusso, M. A co-ultramicronized palmitoylethanolamide/luteolin composite mitigates clinical score and disease-relevant molecular markers in a mouse model of experimental autoimmune encephalomyelitis. J. Neuroinflam. 2019, 16, 126. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Zheng, Y.; Zhang, X.; Hu, X.; Wang, Y.; Zhang, S.; Zhang, D.; Nie, H. Novel immunoregulatory properties of EGCG on reducing inflammation in EAE. Front. Biosci. 2013, 18, 332–342. [Google Scholar]
- Wang, J.; Ren, Z.; Xu, Y.; Xiao, S.; Meydani, S.N.; Wu, D. Epigallocatechin-3-gallate ameliorates experimental autoimmune encephalomyelitis by altering balance among CD4+ T-cell subsets. Am. J. Pathol. 2012, 180, 221–234. [Google Scholar] [CrossRef] [Green Version]
- Jin, S.L.; Zhou, B.R.; Luo, D. Protective effect of epigallocatechin gallate on the immune function of dendritic cells after ultraviolet B irradiation. J. Cosmet. Dermatol. 2009, 8, 174–180. [Google Scholar] [CrossRef]
- Herges, K.; Millward, J.M.; Hentschel, N. Neuroprotective effect of combination therapy of glatiramer acetate and epigallocatechin-3-gallate in neuroinflammation. PLoS ONE 2011, 6, e25456. [Google Scholar] [CrossRef]
- Semnani, M.; Mashayekhi, F.; Azarnia, M.; Salehi, Z. Effects of green tea epigallocatechin-3-gallate on the proteolipid protein and oligodendrocyte transcription factor 1 messenger RNA gene expression in a mouse model of multiple sclerosis. Folia Neuropathol. 2017, 55, 199–205. [Google Scholar] [CrossRef]
- Mähler, A.; Steiniger, J.; Bock, M.; Klug, L.; Parreidt, N.; Lorenz, M.; Zimmermann, B.F.; Krannich, A.; Paul, F.; Boschmann, M. Metabolic response to epigallocatechin-3-gallate in relapsing-remitting multiple sclerosis: A randomized clinical trial. Am. J. Clin. Nutr. 2015, 101, 487–495. [Google Scholar] [CrossRef] [Green Version]
- Ahmadian, S.R.; Ghasemi-Kasman, M.; Pouramir, M.; Sadeghi, F. Arbutin attenuates cognitive impairment and inflammatory response in pentylenetetrazol-induced kindling model of epilepsy. Neuropharmacology 2019, 146, 117–127. [Google Scholar] [CrossRef]
- Ebrahim-Tabar, F.; Nazari, A.; Pouramir, M.; Ashrafpour, M.; Pourabdolhossein, F. Arbutin Improves Functional Recovery and Attenuates Glial Activation in Lysolecethin-Induced Demyelination Model in Rat Optic Chiasm. Mol. Neurobiol. 2020, 57, 3228–3242. [Google Scholar] [CrossRef]
- Wu, R.M.; Sun, Y.Y.; Zhou, T.T.; Zhu, Z.Y.; Zhuang, J.J.; Tang, X.; Chen, J.; Hu, L.H.; Shen, X. Arctigenin enhances swimming endurance of sedentary rats partially by regulation of antioxidant pathways. Acta Pharmacol. Sin. 2014, 35, 1274–1284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Wang, Z.; Chen, H.; Chen, Z.; Tian, Y. Antioxidants: Potential antiviral agents for Japanese encephalitis virus infection. Int. J. Infect. Dis. 2014, 24, 30–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, W.; Zhang, Z.; Zhang, K.; Xue, Z.; Li, Y.; Zhang, Z.; Zhang, L.; Gu, C.; Zhang, Q.; Hao, J.; et al. Arctigenin Suppress Th17 Cells and Ameliorates Experimental Autoimmune Encephalomyelitis Through AMPK and PPAR-γ/ROR-γt Signaling. Mol. Neurobiol. 2016, 53, 5356–5366. [Google Scholar] [CrossRef]
- Butt, M.S.; Tariq, U.; Iahtisham-Ul-Haq; Naz, A.; Rizwan, M. Neuroprotective effects of oleuropein: Recent developments and contemporary research. J. Food Biochem. 2021, 45, e13967. [Google Scholar] [CrossRef] [PubMed]
- Angeloni, C.; Malaguti, M.; Barbalace, M.C.; Hrelia, S. Bioactivity of olive oil phenols in neuroprotection. Int. J. Mol. Sci. 2017, 18, 2230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shibani, F.; Sahamsizadeh, A.; Fatemi, I.; Allahtavakoli, M.; Hasanshahi, J.; Rhmani, M.; Azin, M.; Hassanipour, M.; Mozafari, N.; Kaeidi, A. Effect of oleuropein on morphine-induced hippocampus neurotoxicity and memory impairments in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 1383–1391. [Google Scholar] [CrossRef]
- Giacometti, J.; Grubić-Kezele, T. Olive leaf polyphenols attenuate the clinical course of experimental autoimmune encephalomyelitis and provide neuroprotection by reducing oxidative stress, regulating microglia and SIRT1, and preserving myelin integrity. Oxid. Med. Cell. Longev. 2020, 2020, 6125638. [Google Scholar] [CrossRef]
- Castejón, M.L.; Montoya, T.; Alarcón-De-La-Lastra, C.; Sánchez-Hidalgo, M. Potential Protective Role Exerted by Secoiridoids from Olea europaea L. in Cancer, Cardiovascular, Neurodegenerative, Aging-Related, and Immunoinflammatory Diseases. Antioxidants 2020, 9, 149. [Google Scholar] [CrossRef] [Green Version]
- Gutiérrez-Miranda, B.; Gallardo, I.; Melliou, E.; Cabero, I.; Álvarez, Y.; Magiatis, P.; Hernández, M.; Nieto, M.L. Oleacein attenuates the pathogenesis of experimental autoimmune encephalomyelitis through both antioxidant and anti-inflammatory effects. Antioxidants 2020, 9, 1161. [Google Scholar] [CrossRef]
- Ahmed, T.; Setzer, W.N.; Nabavi, S.F.; Orhan, I.E.; Braidy, N.; Sobarzo-Sanchez, E.; Nabavi, S.M. Insights into effects of ellagic acid on the nervous system: A mini review. Curr. Pharm. Des. 2016, 22, 1350–1360. [Google Scholar] [CrossRef] [PubMed]
- Gianchecchi, E.; Fierabracci, A. Insights on the Effects of Resveratrol and Some of Its Derivatives in Cancer and Autoimmunity: A Molecule with a Dual Activity. Antioxidants 2020, 9, 91. [Google Scholar] [CrossRef] [Green Version]
- Shindler, K.S.; Ventura, E.; Dutt, M.; Elliott, P.; Fitzgerald, D.C.; Rostami, A. Oral resveratrol reduces neuronal damage in a model of multiple sclerosis. J. Neuroophthalmol. 2010, 30, 328–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, N.P.; Hegde, V.L.; Hofseth, L.J.; Nagarkatti, M.; Nagarkatti, P. Resveratrol (trans-3,5,4′-trihydroxystilbene) ameliorates experimental allergic encephalomyelitis, primarily via induction of apoptosis in T cells involving activation of aryl hydrocarbon receptor and estrogen receptor. Mol. Pharmacol. 2007, 72, 1508–1521. [Google Scholar] [CrossRef] [PubMed]
- Fonseca-Kelly, Z.; Nassrallah, M.; Uribe, J.; Khan, R.S.; Dine, K.; Dutt, M.; Shindler, K.S. Resveratrol neuroprotection in a chronic mouse model of multiple sclerosis. Front. Neurol. 2012, 3, 84. [Google Scholar] [CrossRef] [Green Version]
- Makuch, S.; Więcek, K.; Woźniak, M. The Immunomodulatory and Anti-Inflammatory Effect of Curcumin on Immune Cell Populations, Cytokines, and In Vivo Models of Rheumatoid Arthritis. Pharmaceuticals 2021, 14, 309. [Google Scholar] [CrossRef] [PubMed]
- Miller, E.; Markiewicz, Ł.; Kabziński, J.; Odrobina, D.; Majsterek, I. Potential of redox therapies in neuro-degenerative disorders. Front. Biosci. 2017, 9, 214–234. [Google Scholar] [CrossRef] [Green Version]
- Dattilo, S.; Mancuso, C.; Koverech, G.; Di Mauro, P.; Ontario, M.L.; Petralia, C.C.; Petralia, A.; Maiolino, L.; Serra, A.; Calabrese, E.J.; et al. Heat shock proteins and hormesis in the diagnosis and treatment of neurodegenerative diseases. Immun. Ageing 2015, 12, 20. [Google Scholar] [CrossRef] [Green Version]
- Qureshi, M.; Al-Suhaimi, E.A.; Wahid, F.; Shehzad, O.; Shehzad, A. Therapeutic potential of curcumin for multiple sclerosis. Neurol. Sci. 2018, 39, 207–214. [Google Scholar] [CrossRef]
- Xie, L.; Li, X.K.; Funeshima-Fuji, N.; Kimura, H.; Matsumoto, Y.; Isaka, Y.; Takahara, S. Amelioration of experimental autoimmune encephalomyelitis by curcumin treatment through inhibition of IL-17 production. Int. Immunopharmacol. 2009, 9, 575–581. [Google Scholar] [CrossRef]
- Mohajeri, M.; Sadeghizadeh, M.; Najafi, F.; Javan, M. Polymerized nano-curcumin attenuates neurological symptoms in EAE model of multiple sclerosis through down regulation of inflammatory and oxidative processes and enhancing neuroprotection and myelin repair. Neuropharmacology 2015, 99, 156–167. [Google Scholar] [CrossRef] [PubMed]
- Salemi, G.; Gueli, M.C.; Vitale, F.; Battaglieri, F.; Guglielmini, E.; Ragonese, P.; Trentacosti, A.; Massenti, M.F.; Savettieri, G.; Bono, A. Blood lipids, homocysteine, stress factors, and vitamins in clinically stable multiple sclerosis patients. Lipids Health Dis. 2010, 9, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramsaransing, G.S.; Mellema, S.A.; De Keyser, J. Dietary patterns in clinical subtypes of multiple sclerosis: An exploratory study. Nutr. J. 2009, 8, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Løken-Amsrud, K.I.; Myhr, K.-M.; Bakke, S.J.; Beiske, A.G.; Bjerve, K.S.; Bjørnarå, B.T.; Hovdal, H.; Lilleås, F.; Midgard, R.; Pedersen, T.; et al. Alpha-tocopherol and MRI outcomes in multiple sclerosis—Association and prediction. PLoS ONE 2013, 8, e54417. [Google Scholar] [CrossRef]
- Goudarzvand, M.; Javan, M.; Mirnajafi-Zadeh, J.; Mozafari, S.; Tiraihi, T. Vitamins E and D3 attenuate demyelination and potentiate remyelination processes of hippocampal formation of rats following local injection of ethidium bromide. Cell. Mol. Neurobiol. 2010, 30, 289–299. [Google Scholar] [CrossRef]
- Mazzanti, C.M.; Spanevello, R.; Ahmed, M.; Pereira, L.B.; Gonçalves, J.F.; Corrêa, M.; Schmatz, R.; Stefanello, N.; Leal, D.B.; Mazzanti, A.; et al. Pre-treatment with ebselen and vitamin E modulate acetylcholinesterase activity: Interaction with demyelinating agents. Int. J. Dev. Neurosci. 2009, 27, 73–80. [Google Scholar] [CrossRef] [PubMed]
- Theodosis-Nobelos, P.; Papagiouvannis, G.; Rekka, E.A. A Review on Vitamin E Natural Analogues and on the Design of Synthetic Vitamin E Derivatives as Cytoprotective Agents. Mini Rev. Med. Chem. 2021, 21, 10–22. [Google Scholar] [CrossRef] [PubMed]
- Blanchard, B.; Heurtaux, T.; Garcia, C. Tocopherol derivative TFA-12 promotes myelin repair in experimental models of multiple sclerosis. J. Neurosci. 2013, 33, 11633–11642. [Google Scholar] [CrossRef] [Green Version]
- Dao, D.Q.; Ngo, T.C.; Thong, N.M.; Nam, P.C. Is Vitamin A an Antioxidant or a Pro-oxidant? J. Phys. Chem. B 2017, 12, 9348–9357. [Google Scholar] [CrossRef] [PubMed]
- Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushali, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
- Jafarirad, S.; Siassi, F.; Harirchian, M.H.; Sahraian, M.A.; Eshraghian, M.R.; Shokri, F.; Amani, R.; Bitarafan, S.; Mozafari, S.; Saboor-Yaraghi, A. The effect of vitamin A supplementation on stimulated T-cell proliferation with myelin oligodendrocyte glycoprotein in patients with multiple sclerosis. J. Neurosci. Rural Pract. 2012, 3, 294–298. [Google Scholar] [CrossRef] [PubMed]
- Bitarafan, S.; Saboor-Yaraghi, A.; Sahraian, M.-A.; Nafissi, S.; Togha, M.; Moghadam, N.B.; Roostaei, T.; Siassi, F.; Eshraghian, M.-R.; Ghanaati, H.; et al. Impact of vitamin A supplementation on disease progression in patients with multiple sclerosis. Arch. Iran. Med. 2015, 18, 435–440. [Google Scholar]
- Raverdeau, M.; Breen, C.J.; Misiak, A.; Mills, K.H. Retinoic acid suppresses IL-17productionandpathogenic activity of γδT-cells in CNS autoimmunity. Immunol. Cell Biol. 2016, 94, 763–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eriksen, A.B.; Berge, T.; Gustavsen, M.W.; Leikfoss, I.S.; Bos, S.D.; Spurkland, A.; Harbo, H.F.; Blomhoff, H.K. Retinoic acid enhances the levels of IL-10 in TLR-stimulated B cells from patients with relapsing-remitting multiple sclerosis. J. Neuroimmunol. 2015, 278, 11–18. [Google Scholar] [CrossRef] [PubMed]
- Dorosty-Motlagh, A.R.; Honarvar, M.N.; Sedighiyan, M.; Abdolahi, M. The molecular mechanisms of vitamin A deficiency in multiple sclerosis. J. Mol. Neurosci. 2016, 60, 82–90. [Google Scholar] [CrossRef] [PubMed]
- Mizee, M.R.; Nijland, P.G.; van der Pol, S.M.; Drexhage, J.A.; van Het Hof, B.; Mebius, R.; van der Valk, P.; van Horssen, J.; Reijerkerk, A.; de Vries, H.E. Astrocyte-derived retinoic acid: A novel regulator of blood-brain barrier function in multiple sclerosis. Acta Neuropathol. 2014, 128, 691–703. [Google Scholar] [CrossRef] [PubMed]
- de Oliveira, R.G., Jr.; Bonnet, A.; Braconnier, E.; Groult, H.; Prunier, G.; Beaugeard, L.; Grougnet, R.; da Silva Almeida, J.R.G.; Ferraz, C.A.A.; Picot, L. Bixin, an apocarotenoid isolated from Bixa orellana L., sensitizes human melanoma cells to dacarbazine-induced apoptosis through ROS-mediated cytotoxicity. Food Chem. Toxicol. 2019, 125, 549–561. [Google Scholar] [CrossRef]
- Yu, Y.; Wu, D.M.; Li, J.; Deng, S.H.; Liu, T.; Zhang, T.; He, M.; Zhao, Y.Y.; Xu, Y. Bixin Attenuates Experimental Autoimmune Encephalomyelitis by Suppressing TXNIP/NLRP3 Inflammasome Activity and Activating NRF2 Signaling. Front. Immunol. 2020, 11, 593368. [Google Scholar] [CrossRef]
- Fathimoghadam, H.; Farbod, Y.; Ghadiri, A.; Fatemi, R. Moderating effects of crocin on some stress oxidative markers in rat brain following demyelination with ethidium bromide. Heliyon 2019, 5, e01213. [Google Scholar] [CrossRef] [Green Version]
- Ghaffari, S.; Hatami, H.; Dehghan, G. Saffron ethanolic extract attenuates oxidative stress, spatial learning, and memory impairments induced by local injection of ethidium bromide. Res. Pharm. Sci. 2015, 10, 222–232. [Google Scholar]
- Ghiasian, M.; Khamisabadi, F.; Kheiripour, N.; Karami, M.; Haddadi, R.; Ghaleiha, A.; Taghvaei, B.; Oliaie, S.S.; Salehi, M.; Samadi, P.; et al. Effects of crocin in reducing DNA damage, inflammation, and oxidative stress in multiple sclerosis patients: A double-blind, randomized, and placebo-controlled trial. J. Biochem. Mol. Toxicol. 2019, 33, e22410. [Google Scholar] [CrossRef] [PubMed]
- Sadeghnia, H.R.; Shaterzadeh, H.; Forouzanfar, F.; Hosseinzadeh, H. Neuroprotective effect of safranal, an active ingredient of Crocus sativus, in a rat model of transient cerebral ischemia. Folia Neuropathol. 2017, 55, 206–213. [Google Scholar] [CrossRef] [PubMed]
- Alavi, M.S.; Fanoudi, S.; Fard, A.V.; Soukhtanloo, M.; Hosseini, M.; Barzegar, H.; Sadeghnia, H.R. Safranal Attenuates Excitotoxin-Induced Oxidative OLN-93 Cells Injury. Drug Res. 2019, 69, 323–329. [Google Scholar] [CrossRef] [PubMed]
- Sakai, C.; Ishida, M.; Ohba, H.; Yamashita, H.; Uchida, H.; Yoshizumi, M.; Ishida, T. Fish oil omega-3 polyunsaturated fatty acids attenuate oxidative stress-induced DNA damage in vascular endothelial cells. PLoS ONE 2017, 12, e0187934. [Google Scholar] [CrossRef] [Green Version]
- Ramirez-Ramirez, V.; Macias-Islas, M.A.; Ortiz, G.G.; Pacheco-Moises, F.; Torres-Sanchez, E.D.; Sorto-Gomez, T.E.; Cruz-Ramos, J.A.; Orozco-Aviña, G.; Celis de la Rosa, A.J. Efficacy of fish oil on serum of TNF-α, IL-1 β, and IL-6 oxidative stress markers in multiple sclerosis treated with interferon beta-1b. Oxid. Med. Cell. Longev. 2013, 2013, 709493. [Google Scholar] [CrossRef] [Green Version]
- Calder, P.C. n–3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am. J. Clin. Nutr. 2006, 83, 1505S–1519S. [Google Scholar] [CrossRef]
- Shinto, L.; Marracci, G.; Bumgarner, L.; Yadav, V. The Effects of omega-3 fatty acids on matrix metallo-proteinase-9 production and cell migration in human immune cells: Implications for multiple sclerosis. Autoimmune Dis. 2011, 2011, 134592. [Google Scholar]
- Binyamin, O.; Larush, L.; Frid, K.; Keller, G.; Friedman-Levi, Y.; Ovadia, H.; Abramsky, O.; Magdassi, S.; Gabizon, R. Treatment of a multiple sclerosis animal model by a novel nanodrop formulation of a natural antioxidant. Int. J. Nanomed. 2015, 10, 7165–7174. [Google Scholar] [CrossRef] [Green Version]
- Guerra-Vázquez, C.M.; Martínez-Ávila, M.; Guajardo-Flores, D.; Antunes-Ricardo, M. Punicic Acid and Its Role in the Prevention of Neurological Disorders: A Review. Foods 2022, 11, 252. [Google Scholar] [CrossRef]
- Petrou, P.; Ginzberg, A.; Binyamin, O.; Karussis, D. Beneficial effects of a nano formulation of pomegranate seed oil, GranaGard, on the cognitive function of multiple sclerosis patients. Mult. Scler. Relat. Disord. 2021, 54, 103103. [Google Scholar] [CrossRef]
- Messias, M.C.F.; Mecatti, G.C.; Priolli, D.G.; de Oliveira Carvalho, P. Plasmalogen lipids: Functional mechanism and their involvement in gastrointestinal cancer. Lipids Health Dis. 2018, 17, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fuchs, B. Analytical methods for (oxidized) plasmalogens: Methodological aspects and applications. Free Radic. Res. 2015, 49, 599–617. [Google Scholar] [CrossRef] [PubMed]
- Luoma, A.M.; Kuo, F.; Cakici, O.; Crowther, M.N.; Denninger, A.R.; Avila, R.L.; Brites, P.; Kirschner, D.A. Plasmalogen phospholipids protect internodal myelin from oxidative damage. Free Radic. Biol. Med. 2015, 84, 296–310. [Google Scholar] [CrossRef] [PubMed]
- Stadelmann-Ingrand, S.; Pontcharraud, R.; Fauconneau, B. Evidence for the reactivity of fatty aldehydes released from oxidized plasmalogens with phosphatidylethanolamine to form Schiff base adducts in rat brain homogenates. Chem. Phys. Lipids 2004, 131, 93–105. [Google Scholar] [CrossRef] [PubMed]
- Pantzaris, M.C.; Loukaides, G.N.; Ntzani, E.E.; Patrikios, I.S. A novel oral nutraceutical formula of omega-3 and omega-6 fatty acids with vitamins (PLP10) in relapsing remitting multiple sclerosis: A randomised, double-blind, placebo-controlled proof-of-concept clinical trial. BMJ Open 2013, 3, e002170. [Google Scholar] [CrossRef] [Green Version]
- Aristotelous, P.; Stefanakis, M.; Pantzaris, M.; Pattichis, C.S.; Calder, P.C.; Patrikios, I.S.; Sakkas, G.K.; Giannaki, C.D. The Effects of Specific Omega-3 and Omega-6 Polyunsaturated Fatty Acids and Antioxidant Vitamins on Gait and Functional Capacity Parameters in Patients with Relapsing-Remitting Multiple Sclerosis. Nutrients 2021, 13, 3661. [Google Scholar] [CrossRef]
- Torkildsen, O.; Wergeland, S.; Bakke, S.; Beiske, A.G.; Bjerve, K.S.; Hovdal, H.; Midgard, R.; Lilleås, F.; Pedersen, T.; Bjørnarå, B.; et al. ω–3 fatty acid treatment in multiple sclerosis (OFAMS Study): A randomized, double-blind, placebo-controlled trial. Arch. Neurol. 2012, 69, 1044–1051. [Google Scholar] [CrossRef] [Green Version]
- Barzegarzadeh, B.; Hatami, H.; Dehghan, G.; Khajehnasiri, N.; Khoobi, M.; Sadeghian, R. Conjugated Linoleic Acid-Curcumin Attenuates Cognitive Deficits and Oxidative Stress Parameters in the Ethidium Bromide-Induced Model of Demyelination. Neurotox. Res. 2021, 39, 815–825. [Google Scholar] [CrossRef]
- Chiurchiù, V.; Leuti, A.; Smoum, R.; Mechoulam, R.; Maccarrone, M. Bioactive lipids ALIAmides differentially modulate inflammatory responses of distinct subsets of primary human T lymphocytes. FASEB J. 2018, 32, 5716–5723. [Google Scholar] [CrossRef]
- D’Amico, R.; Impellizzeri, D.; Cuzzocrea, S.; Di Paola, R. ALIAmides Update: Palmitoylethanolamide and Its Formulations on Management of Peripheral Neuropathic Pain. Int. J. Mol. Sci. 2020, 21, 5330. [Google Scholar] [CrossRef]
- Landolfo, E.; Cutuli, D.; Petrosini, L.; Caltagirone, C. Effects of Palmitoylethanolamide on Neurodegenerative Diseases: A Review from Rodents to Humans. Biomolecules 2022, 12, 667. [Google Scholar] [CrossRef] [PubMed]
- Holmay, M.J.; Terpstra, M.; Coles, L.D.; Mishra, U.; Ahlskog, M.; Öz, G.; Cloyd, J.C.; Tuite, P.J. N-Acetylcysteine boosts brain and blood glutathione in Gaucher and Parkinson diseases. Clin. Neuropharmacol. 2013, 36, 103–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ljubisavljevic, S.; Stojanovic, I.; Pavlovic, D.; Sokolovic, D.; Stevanovic, I. Aminoguanidine and N-acetyl-cysteine supress oxidative and nitrosative stress in EAE rat brains. Redox Rep. 2011, 16, 166–172. [Google Scholar] [CrossRef] [PubMed]
- Bavarsad Shahripour, R.; Harrigan, M.R.; Alexandrov, A.V. N-acetylcysteine (NAC) in neurological disorders: Mechanisms of action and therapeutic opportunities. Brain Behav. 2014, 4, 108–122. [Google Scholar] [CrossRef]
- Samuni, Y.; Goldstein, S.; Dean, O.M.; Berk, M. The chemistry and biological activities of N-acetylcysteine. Biochim. Biophys. Acta 2013, 1830, 4117–4129. [Google Scholar] [CrossRef]
- Doche, E.; Lecocq, A.; Maarouf, A.; Duhamel, G.; Soulier, E.; Confort-Gouny, S.; Rico, A.; Guye, M.; Audoin, B.; Pelletier, J.; et al. Hypoperfusion of the thalamus is associated with disability in relapsing remitting multiple sclerosis. J. Neuroradiol. 2017, 44, 158–164. [Google Scholar] [CrossRef]
- de la Pena, M.J.; Pena, I.C.; Garcia, P.G.; Gavilán, M.L.; Malpica, N.; Rubio, M.; González, R.A.; de Vega, V.M. Early perfusion changes in multiple sclerosis patients as assessed by MRI using arterial spin labeling. Acta Radiol. Open 2019, 8, 2058460119894214. [Google Scholar] [CrossRef]
- Monti, D.A.; Zabrecky, G.; Leist, T.P.; Wintering, N.; Bazzan, A.J.; Zhan, T.; Newberg, A.B. N-acetyl Cysteine Administration Is Associated with Increased Cerebral Glucose Metabolism in Patients With Multiple Sclerosis: An Exploratory Study. Front. Neurol. 2020, 11, 88. [Google Scholar] [CrossRef]
- Kosuge, Y. Neuroprotective mechanisms of S-allyl-L-cysteine in neurological disease. Exp. Ther. Med. 2020, 19, 1565–1569. [Google Scholar] [CrossRef]
- Baluchnejadmojarad, T.; Kiasalari, Z.; Afshin-Majd, S.; Ghasemi, Z.; Roghani, M. S-allyl cysteine ameliorates cognitive deficits in streptozotocin-diabetic rats via suppression of oxidative stress, inflammation, and acetylcholinesterase. Eur. J. Pharmacol. 2017, 794, 69–76. [Google Scholar] [CrossRef]
- Zeinali, H.; Baluchnejadmojarad, T.; Fallah, S.; Sedighi, M.; Moradi, N.; Roghani, M. S-allyl cysteine improves clinical and neuropathological features of experimental autoimmune encephalomyelitis in C57BL/6 mice. Biomed. Pharmacother. 2018, 97, 557–563. [Google Scholar] [CrossRef] [PubMed]
- Escribano, B.; Agüera, E.; Aguilar-Luque, M.; Luque, E.; Feijóo, M.; LaTorre, M.; Giraldo, A.; Galván-Jurado, A.; Caballero-Villarraso, J.; Garcia-Maceira, F.I.; et al. Neuroprotective effect of S-allyl cysteine on an experimental model of multiple sclerosis: Antioxidant effects. J. Funct. Foods 2018, 42, 281–288. [Google Scholar] [CrossRef]
- Brown, D.A.; Betharia, S.; Yen, J.H.; Tran, Q.; Mistry, H.; Smith, K. Synthesis and structure-activity relationships study of dithiolethiones as inducers of glutathione in the SH-SY5Y neuroblastoma cell line. Bioorgan. Med. Chem. Lett. 2014, 24, 5829–5831. [Google Scholar] [CrossRef] [PubMed]
- Dali, M.M.; Dansette, P.M.; Mansuy, D.; Boucher, J.L. Comparison of Various Aryl-Dithiolethiones and Aryl-Dithiolones As Hydrogen Sulfide Donors in the Presence of Rat Liver Microsomes. Drug Metab. Dispos. 2020, 48, 426–431. [Google Scholar] [CrossRef]
- Kuo, P.C.; Brown, D.A.; Scofield, B.A.; Yu, I.C.; Chang, F.; Wang, P.Y.; Yen, J.H. 3H-1,2-dithiole-3-thione as a novel therapeutic agent for the treatment of experimental autoimmune encephalomyelitis. Brain Behav. Immun. 2016, 57, 173–186. [Google Scholar] [CrossRef]
- Kuo, P.C.; Brown, D.A.; Scofield, B.A.; Paraiso, H.C.; Wang, P.Y.; Yu, I.C.; Yen, J.H. Dithiolethione ACDT suppresses neuroinflammation and ameliorates disease severity in experimental autoimmune encephalomyelitis. Brain Behav. Immun. 2018, 70, 76–87. [Google Scholar] [CrossRef] [Green Version]
- Theodosis-Nobelos, P.; Papagiouvannis, G.; Tziona, P.; Rekka, E.A. Lipoic acid. Kinetics and pluripotent biological properties and derivatives. Mol. Biol. Rep. 2021, 48, 6539–6550. [Google Scholar] [CrossRef]
- Loy, B.D.; Fling, B.W.; Horak, F.B.; Bourdette, D.N.; Spain, R.I. Effects of lipoic acid on walking performance, gait, and balance in secondary progressive multiple sclerosis. Complement. Ther. Med. 2018, 41, 169–174. [Google Scholar] [CrossRef]
- Spain, R.; Powers, K.; Murchison, C.; Heriza, E.; Winges, K.; Yadav, V.; Cameron, M.; Kim, E.; Horak, F.; Simon, J.; et al. Lipoic acid in secondary progressive MS: A randomized controlled pilot trial. Neurol. Neuroimmunol. Neuroinflamm. 2017, 4, e374. [Google Scholar] [CrossRef] [Green Version]
- Fiedler, S.E.; Spain, R.I.; Kim, E.; Salinthone, S. Lipoic acid modulates inflammatory responses of monocytes and monocyte-derived macrophages from healthy and relapsing-remitting multiple sclerosis patients. Immunol. Cell Biol. 2021, 99, 107–115. [Google Scholar] [CrossRef]
- Wang, K.C.; Tsai, C.P.; Lee, C.L.; Chen, S.Y.; Lin, G.J.; Yen, M.H.; Sytwu, H.K.; Chen, S.J. alpha-Lipoic acid enhances endogenous peroxisome-proliferator-activated receptor-gamma to ameliorate experimental autoimmune encephalomyelitis in mice. Clin. Sci. 2013, 125, 329–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khalili, M.; Azimi, A.; Izadi, V.; Eghtesadi, S.; Mirshafiey, A.; Sahraian, M.A.; Motevalian, A.; Norouzi, A.; Sanoobar, M.; Eskandari, G.; et al. Does lipoic acid consumption affect the cytokine profile in multiple sclerosis patients: A double-blind, placebo-controlled, randomized clinical trial. Neuroimmunomodulation 2014, 21, 291–296. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, P.; Marracci, G.; Yu, X.; Galipeau, D.; Morris, B.; Bourdette, D. Lipoic acid decreases inflammation and confers neuroprotection in experimental autoimmune optic neuritis. J. Neuroimmunol. 2011, 233, 90–96. [Google Scholar] [CrossRef] [Green Version]
- Sghaier, R.; Zarrouk, A.; Nury, T.; Badreddine, I.; O’Brien, N.; Mackrill, J.J.; Vejux, A.; Samadi, M.; Nasser, B.; Caccia, C.; et al. Biotin attenuation of oxidative stress, mitochondrial dysfunction, lipid metabolism alteration and 7β-hydroxycholesterol-induced cell death in 158N murine oligodendrocytes. Free Radic. Res. 2019, 53, 535–561. [Google Scholar] [CrossRef] [Green Version]
- Sedel, F.; Papeix, C.; Bellanger, A.; Touitou, V.; Lebrun-Frenay, C.; Galanaud, D.; Gout, O.; Lyon-Caen, O.; Tourbah, A. High doses of biotin in chronic progressive multiple sclerosis: A pilot study. Mult. Scler. Relat. Disord. 2015, 4, 159–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tourbah, A.; Lebrun-Frenay, C.; Edan, G.; Clanet, M.; Papeix, C.; Vukusic, S.; De Sèze, J.; Debouverie, M.; Gout, O.; Clavelou, P.; et al. MD1003 (high-dose biotin) for the treatment of progressive multiple sclerosis: A randomised, double-blind, placebo-controlled study. Mult. Scler. J. 2016, 22, 1719–1731. [Google Scholar] [CrossRef] [Green Version]
- Cree, B.A.C.; Cutter, G.; Wolinsky, J.S.; Freedman, M.S.; Comi, G.; Giovannoni, G.; Hartung, H.P.; Arnold, D.; Kuhle, J.; Block, V.; et al. Safety and efficacy of MD1003 (high-dose biotin) in patients with progressive multiple sclerosis (SPI2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2020, 19, 988–997. [Google Scholar] [CrossRef]
- Nga, N.T.T.; Quang, D.D. Unraveling the antioxidant potential of thiamine: Thermochemical and kineticsstudies in aqueous phase using DFT. Vietnam. J. Chem. 2019, 57, 485–490. [Google Scholar] [CrossRef]
- Costantini, A.; Nappo, A.; Pala, M.I.; Zappone, A. High dose thiamine improves fatigue in multiple sclerosis. BMJ Case Rep. 2013, 2013, bcr2013009144. [Google Scholar] [CrossRef] [Green Version]
- Ji, Z.; Fan, Z.; Zhang, Y.; Yu, R.; Yang, H.; Zhou, C.; Luo, J.; Ke, Z.-J. Thiamine deficiency promotes T cell infiltration in experimental autoimmune encephalomyelitis: The involvement of CCL2. J. Immunol. 2014, 193, 2157–2167. [Google Scholar] [CrossRef]
- Santos, P.W.; Machado, A.R.T.; De Grandis, R.; Ribeiro, D.L.; Tuttis, K.; Morselli, M.; Aissa, A.F.; Pellegrini, M.; Antunes, L.M.G. Effects of sulforaphane on the oxidative response, apoptosis, and the transcriptional profile of human stomach mucosa cells in vitro. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2020, 854–855, 503201. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Gilmour, A.; Ahn, Y.H.; de la Vega, L.; Dinkova-Kostova, A.T. The isothiocyanate sulforaphane inhibits mTOR in an NRF2-independent manner. Phytomedicine 2021, 86, 153062. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Cui, W.; Liu, J.; Li, R.; Liu, Q.; Xie, X.H.; Ge, X.L.; Zhang, J.; Song, X.J.; Wang, Y.; et al. Sulforaphane ameliorates the development of experimental autoimmune encephalomyelitis by antagonizing oxidative stress and Th17-related inflammation in mice. Exp. Neurol. 2013, 250, 239–249. [Google Scholar] [CrossRef] [PubMed]
- Schepici, G.; Bramanti, P.; Mazzon, E. Efficacy of Sulforaphane in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 8637. [Google Scholar] [CrossRef] [PubMed]
- Jaafaru, M.S.; Nordin, N.; Shaari, K.; Rosli, R.; Abdull Razis, A.F. Isothiocyanate from Moringa oleifera seeds mitigates hydrogen peroxide-induced cytotoxicity and preserved morphological features of human neuronal cells. PLoS ONE 2018, 13, e0196403. [Google Scholar] [CrossRef] [Green Version]
- Giacoppo, S.; Soundara Rajan, T.; De Nicola, G.R.; Iori, R.; Bramanti, P.; Mazzon, E. Moringin activates Wnt canonical pathway by inhibiting GSK3β in a mouse model of experimental autoimmune encephalomyelitis. Drug Des. Dev. Ther. 2016, 10, 3291–3304. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. 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
Theodosis-Nobelos, P.; Rekka, E.A. The Multiple Sclerosis Modulatory Potential of Natural Multi-Targeting Antioxidants. Molecules 2022, 27, 8402. https://doi.org/10.3390/molecules27238402
Theodosis-Nobelos P, Rekka EA. The Multiple Sclerosis Modulatory Potential of Natural Multi-Targeting Antioxidants. Molecules. 2022; 27(23):8402. https://doi.org/10.3390/molecules27238402
Chicago/Turabian StyleTheodosis-Nobelos, Panagiotis, and Eleni A. Rekka. 2022. "The Multiple Sclerosis Modulatory Potential of Natural Multi-Targeting Antioxidants" Molecules 27, no. 23: 8402. https://doi.org/10.3390/molecules27238402