Marine Bioactive Components and Chronic Neuroinflammation: Focus on Neurodegenerative Disease
Abstract
1. Introduction
2. Neuroinflammation
2.1. Pathophysiology
2.2. Neuroinflammation and Neurodegenerative Disease
2.2.1. Alzheimer’s Disease (AD)
2.2.2. Parkinson’s Disease (PD)
2.2.3. Multiple Sclerosis (MD)
2.2.4. Amyotrophic Lateral Sclerosis (ALS)
3. Marine-Derived Bioactive Components and AD, PD, MD, and ALS
3.1. Proteins, Peptides and Amino Acids
3.2. Astaxanthin
3.3. Polysaccharides
3.4. Polyphenols
3.5. Clinical Evidence and Translational Gaps for Marine-Derived Bioactive Compounds in AD, PD, MS and ALS
4. Materials and Methods
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Engelhardt, E.; Vajkoczy, P.; Weller, R.O. The movers and shapers in immune privilege of the CNS. Nat. Immunol. 2017, 18, 123–131. [Google Scholar] [CrossRef] [PubMed]
- Leunig, A.; Gianeselli, M.; Russo, S.J.; Swirski, F.K. Connection and communication between the nervous and immune systems. Nat. Rev. Immunol. 2025; online ahead of print. [Google Scholar] [CrossRef]
- Billingham, R.E.; Boswell, T. Studies on the problem of corneal homografts. Proc. R. Soc. Lond. B Biol. Sci. 1953, 141, 392–406. [Google Scholar] [CrossRef]
- Galea, I.; Bechmann, I.; Perry, V.H. What is immune privilege (not)? Trends Immunol. 2007, 28, 12–18. [Google Scholar] [CrossRef]
- Louvea, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.-S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef]
- Smyth, L.C.D.; Kipnis, J. Redefining CNS immune privilege. Nat. Rev. Immunol. 2025; online ahead of print. [Google Scholar] [CrossRef]
- Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef]
- Parolini, C. Marine n-3 polyunsaturated fatty acids: Efficacy on inflammatory-based disorders. Life Sci. 2020, 263, 118591. [Google Scholar] [CrossRef] [PubMed]
- Yang, Q.; Wang, G.; Zhang, F. Role of Peripheral Immune Cells-Mediated Inflammation on the Process of Neurodegenerative Diseases. Front. Immunol. 2020, 11, 582825. [Google Scholar] [CrossRef] [PubMed]
- Chiesa, G.; Busnelli, M.; Manzini, S.; Parolini, C. Nutraceuticals and Bioactive Components from Fish for Dyslipidemia and Cardiovascular Risk Reduction. Mar. Drugs 2016, 14, 113. [Google Scholar] [CrossRef]
- Lamminpää, I.; Amedei, A.; Parolini, C. Effects of Marine-Derived Components on Cardiovascular Disease Risk Factors and Gut Microbiota Diversity. Mar. Drugs 2024, 22, 523. [Google Scholar] [CrossRef]
- Parolini, C. The Role of Marine n-3 Polyunsaturated Fatty Acids in Inflammatory-Based Disease: The Case of Rheumatoid Arthritis. Mar. Drugs 2023, 22, 17. [Google Scholar] [CrossRef]
- Amedei, A.; Lamminpää, I.; Parolini, C. Potential and Future Therapeutic Applications of Eicosapentaenoic acid/Docosahexaenoic Acid and Probiotics in Chronic Low-Grade Inflammation. Biomedicines 2025, 13, 2428. [Google Scholar] [CrossRef] [PubMed]
- Jensen, I.-J.; Maehre, H.K. Preclinical and Clinical Studies on Antioxidative, Antihypertensive and Cardioprotective Effect of Marine Proteins and Peptides-A Review. Mar. Drugs 2016, 14, 211. [Google Scholar] [CrossRef] [PubMed]
- Tørris, C.; Småstuen, M.C.; Molin, M. Nutrients in Fish and Possible Associations with Cardiovascular Disease Risk Factors in Metabolic Syndrome. Nutrients 2018, 10, 952. [Google Scholar] [CrossRef] [PubMed]
- Catanesi, M.; Caioni, G.; Castelli, V.; Benedetti, E.; d’Angelo, M.; Cimini, A. Benefits under the Sea: The Role of Marine Compounds in Neurodegenerative Disorders. Mar. Drugs 2021, 19, 24. [Google Scholar] [CrossRef]
- Hannan, M.A.; Dash, R.; Haque, M.N.; Mohibbullah, M.; Sohag, A.A.M.; Rahman, M.A.; Uddin, M.J.; Alam, M.; Moon, I.S. Neuroprotective Potentials of Marine Algae and Their Bioactive Metabolites: Pharmacological Insights and Therapeutic Advances. Mar. Drugs 2020, 18, 347. [Google Scholar] [CrossRef]
- Cooke, J.P. Inflammation and Its Role in Regeneration and Repair. Circ. Res. 2019, 124, 1166–1168. [Google Scholar] [CrossRef]
- Streit, W.J. Microglia as neuroprotective, immunocompetent cells of the CNS. Glia 2002, 40, 133–139. [Google Scholar] [CrossRef]
- Serhan, C.N. Treating inflammation and infection in the 21st century: New hints from decoding resolution mediators and mechanisms. FASEB J. 2017, 31, 1273–1288. [Google Scholar] [CrossRef]
- Sugimoto, M.A.; Vago, J.P.; Perretti, M.; Teixeira, M.M. Mediators of the Resolution of the Inflammatory Response. Trends Immunol. 2019, 40, 212–227. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Gelb, S.; Lehtinen, M.K. Snapshot: Choroid plexus brain barrier. Cell 2023, 186, 3522.e1. [Google Scholar] [CrossRef]
- Cui, J.; Xu, H.; Lehtinen, M.K. Macrophages on the margin: Choroid plexus immune responses. Trends Neurosci. 2021, 44, 864–875. [Google Scholar] [CrossRef]
- Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef]
- Parolini, C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim. Biophys. Acta Mol. Basis Dis. 2025, 1871, 167761. [Google Scholar] [CrossRef]
- Angus, D.C.; van der Poll, T. Severe sepsis and septic shock. N. Engl. J. Med. 2013, 369, 840–851. [Google Scholar] [CrossRef]
- Van der Poll, T.; Shankar-Hari, M.; Wiersinga, W.J. The immunology of sepsis. Immunity 2021, 54, 2450–2464. [Google Scholar] [CrossRef] [PubMed]
- Parolini, C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem. Pharmacol. 2025, 235, 116844. [Google Scholar] [CrossRef] [PubMed]
- Sharp, F.R.; Bernaudin, M. HIF1 and oxygen sensing in the brain. Nat. Rev. Neurosci. 2004, 5, 437–448. [Google Scholar] [CrossRef]
- Manzini, S.; Pinna, C.; Busnelli, M.; Cinquanta, P.; Rigamonti, E.; Ganzetti, S.G.; Dellera, F.; Sala, A.; Calabresi, L.; Franceschini, G.; et al. Beta2-adrenergic activity modulates vascular tone regulation in lecithin:cholesterol acyltransferase knockout mice. Vascul. Pharmacol. 2015, 74, 114–121. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve 2002, 26, 459–470. [Google Scholar] [CrossRef] [PubMed]
- McGeer, P.L.; McGeer, E.G. Glial reactions in Parkinson’s disease. Mov. Disord. 2008, 23, 474–483. [Google Scholar] [CrossRef] [PubMed]
- Brambilla, R. The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol. 2019, 137, 757–783. [Google Scholar] [CrossRef]
- Bayraktaroglu, I.; Ortí-Casañ, N.; Van Dam, D.; De Deyn, P.P.; Eisel, U.L.M. Systemic inflammation as a central player in the initiation and development of Alzheimer’s disease. Immun. Ageing 2025, 22, 33. [Google Scholar] [CrossRef] [PubMed]
- Ertekin-Taner, N. Genetics of Alzheimer’s disease: A centennial review. Neurol. Clin. 2007, 25, 611–667. [Google Scholar] [CrossRef]
- Andrews, S.J.; Renton, A.E.; Fulton-Howard, B.; Podlesny-Drabiniok, A. Marcora E, Goate AM: The complex genetic architecture of Alzheimer’s disease: Novel insights and future directions. EBioMedicine 2023, 90, 104511. [Google Scholar] [CrossRef]
- Campion, D.; Dumanchin, C.; Hannequin, D.; Dubois, B.; Belliard, S.; Puel, M.; Thomas-Anterion, C.; Michon, A.; Martin, C.; Charbonnier, F.; et al. Early-onset autosomal dominant Alzheimer disease: Prevalence, genetic heterogeneity, and mutation spectrum. Am. J. Hum. Genet. 1999, 65, 664–670. [Google Scholar] [CrossRef]
- Ayodele, T.; Rogaeva, E.; Kurup, J.T.; Beecham, G.; Reitz, C. Early-Onset Alzheimer’s Disease: What is Missing in Research? Curr. Neurol. Nerosci. Rep. 2021, 21, 4. [Google Scholar] [CrossRef]
- Selkoe, D.J. Alzheimer’s disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a004457. [Google Scholar] [CrossRef]
- Haass, C.; Selkoe, D.J. Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev. Mol. Cell Biol. 2007, 8, 101–112. [Google Scholar] [CrossRef]
- Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef] [PubMed]
- Fedele, E. Anti-Amyloid Therapies for Alzheimer’s Disease and the Amyloid Cascade Hypothesis. Int. J. Mol. Sci. 2023, 24, 14499. [Google Scholar] [CrossRef]
- Behl, C. In 2024, the amyloid-cascade-hypothesis still remains a working hypothesis, no less but certainly no more. Front. Aging Neurosci. 2024, 16, 1459224. [Google Scholar] [CrossRef]
- Ingelsson, M.; Fukumoto, H.; Newell, K.L.; Growdon, J.H.; Hedley-whyte, E.T.; Frosch, M.P.; Albert, M.S. Early Abeta accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain. Neurology 2004, 62, 925–931. [Google Scholar] [CrossRef]
- Eikelenboom, P.; Veerhuis, R.; Scheper, W.; Rozemuller, A.J.M.; van Gool, W.A.; Hoozemans, J.J.M. The significance of neuroinflammation in understanding Alzheimer’s disease. J. Neural. Transm. 2006, 113, 1685–1695. [Google Scholar] [CrossRef]
- Lutshumba, J.; Nikolajczyk, B.S.; Bachstetter, A.D. Dysregulation of Systemic Immunity in Aging and Dementia. Front. Cell Neurosci. 2021, 15, 652111. [Google Scholar] [CrossRef] [PubMed]
- Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G.M.; Cooper, N.R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B.L.; et al. Inflammation and Alzheimer’s disease. Neurobiol. Aging 2000, 21, 383–421. [Google Scholar] [CrossRef] [PubMed]
- McCoy, M.K.; Taney, M.G. TNF signaling inhibition in the CNS: Implications for normal brain function and neurodegenerative disease. J. Neuroinflamm. 2008, 5, 45. [Google Scholar] [CrossRef]
- Elbandy, M. Anti-Inflammatory Effects of Marine Bioactive Compounds and Their Potential as Functional Food Ingredients in the Prevention and Treatment of Neuroinflammatory Disorders. Molecules 2002, 28, 2. [Google Scholar] [CrossRef] [PubMed]
- Bu, G. Apolipoprotein E and its receptors in Alzheimer’s disease: Pathways, pathogenesis and therapy. Nat. Rev. Neurosci. 2009, 10, 333–344. [Google Scholar] [CrossRef]
- GBD 2016 Neurology Collaborators. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef]
- Braak, H.; Tredici, D.K.; Rub, U.; de Vos, R.A.I.; Steur, E.N.H.J.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef] [PubMed]
- Perez-Pardo, P.; Kliest, T.; Dodiya, H.B.; Broersen, L.M.; Garssen, J.; Keshavarzian, A.; Kraneveld, A.D. The gut-brain axis in Parkinson’s disease: Possibilities for food-based therapies. Eur. J. Pharmacol. 2017, 817, 86–95. [Google Scholar] [CrossRef]
- Chen, H.; Zhao, E.J.; Zhang, W.; Lu, Y.; Liu, R.; Huang, X.; Ciesielski-Jones, A.J.; Justice, M.A.; Cousins, D.S.; Peddada, S. Meta-analyses on prevalence of selected Parkinson’s nonmotor symptoms before and after diagnosis. Transl. Neurodegener. 2015, 4, 1. [Google Scholar] [CrossRef]
- Gao, X.; Chen, H.; Schwarzchild, M.A.; Ascherio, A. A prospective study of bowel movement frequency and risk of Parkinson’s disease. Am. J. Epidemiol. 2011, 174, 546–551. [Google Scholar] [CrossRef]
- Abbott, R.D.; Petrovitch, H.; White, L.R.; Masaki, K.H.; Tanner, C.M.; Curb, J.D.; Grandinetti, A.; Blanchette, P.L.; Popper, J.-S.; Ross, J.W. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 2001, 57, 456–462. [Google Scholar] [CrossRef]
- Ponsen, M.M.; Stoffers, D.; Twisk, J.W.R.; Wolters, E.C.; Berendse, H.W. Hyposmia and executive dysfunction as predictors of future Parkinson’s disease: A prospective study. Mov. Disord. 2009, 24, 1060.1065. [Google Scholar] [CrossRef]
- Westenberger, A.; Skrahina, V.; Usnich, T.; Beetz, C.; Vollstedt, E.-J.; Laabs, B.-H.; Paul, J.J.; Curado, F.; Skobalj, S.; Gaber, H.; et al. Relevance of genetic testing in the gene-targeted trial era: The Rostock Parkinson’s disease study. Brain 2024, 147, 2652–2667. [Google Scholar] [CrossRef]
- Chopra, A.; Lang, A.E.; Höglinger, G.; Outeiro, T.F. Towards a biological diagnosis of PD. Park. Relat. Disord. 2024, 122, 106078. [Google Scholar] [CrossRef] [PubMed]
- Buniello, A.; MacArthur, J.A.L.; Cerezo, M.; Harris, L.W.; Hayhurst, J.; Malangone, C.; McMahon, A.; Morales, J.; Mountjoy, E.; Sollis, E.; et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic. Acids Res. 2019, 47, D1005–D1012. [Google Scholar] [CrossRef]
- Marchesi, M.; Parolini, C.; Valetti, C.; Mangione, P.; Obici, L.; Giorgetti, S.; Raimondi, S.; Donadei, S.; Gregorini, G.; Merlini, G.; et al. The intracellular quality control system down-regulates the secretion of amyloidogenic apolipoprotein A-I variants: A possible impact on the natural history of the disease. Biochim. Biophys. Acta 2011, 1812, 87–93. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Benito, M.; Granado, N.; García-Sanz, P.; Michel, P.; Dumoulin, M.; Moratalla, R. Modeling Parkinson’s Disease with the Alpha-Synuclein Protein. Front. Pharmacol. 2020, 11, 356. [Google Scholar] [CrossRef]
- Pauwels, E.K.J.; Boer, G.J. Parkinson’s Disease: A Tale of Many Players. Med. Princ. Pract. 2023, 32, 155–165. [Google Scholar] [CrossRef]
- Park, J.-Y.; Kim, K.S.; Lee, S.-B.; Ryu, J.-S.; Chung, K.C.; Choo, Y.-K.; Jou, I.; Kim, J.; Park, S.M. On the mechanism of internalization of alpha-synuclein into microglia: Roles of ganglioside GM1 and lipid raft. J. Neurochem. 2009, 110, 400–411. [Google Scholar] [CrossRef]
- Kuhn, D.M.; Francescutti-Verbeem, D.M.; Thomas, D.M. Dopamine quinones activate microglia and induce a neurotoxic gene expression profile: Relationship to methamphetamine-induced nerve ending damage. Ann. N. Y. Acad. Sci. 2006, 1074, 31–41. [Google Scholar] [CrossRef] [PubMed]
- Ito, M.; Komai, K.; Mise-Omata, S.; Iizuka-Koga, M.; Nogushi, Y.; Kondo, T.; Sakai, R.; Matsuo, K.; Nakayama, T.; Yoshie, O.; et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 2019, 565, 246–250. [Google Scholar] [CrossRef]
- Saijo, K.; Winner, B.; Carson, C.T.; Collier, J.G.; Boyer, L.; Rosenfeld, M.G.; Gafe, F.H.; Glass, C.K. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell 2009, 137, 47–59. [Google Scholar] [CrossRef] [PubMed]
- Nylander, A.; Hafler, D.A. Multiple sclerosis. J. Clin. Investig. 2012, 122, 1180–1188. [Google Scholar] [CrossRef] [PubMed]
- Sospedra, M.; Martin, R. Immunology of multiple sclerosis. Annu. Rev. Immunol. 2005, 23, 683–747. [Google Scholar] [CrossRef]
- Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef]
- Eisen, A. Amyotrophic lateral sclerosis: A 40-year personal perspective. J. Clin. Neurosci. 2009, 16, 505–512. [Google Scholar] [CrossRef]
- Feldman, E.L.; Goutman, S.A.; Petri, S.; Mazzini, L.; Savelieff, M.G.; Saw, P.J.; Sobue, G. Amyotrophic lateral sclerosis. Lancet 2022, 400, 1363–1380. [Google Scholar] [CrossRef]
- Mejzini, R.; Flynn, L.L.; Pitout, I.L.; Fletcher, S.; Wilton, S.D.; Akkari, P.A. ALS Genetics, Mechanisms, and Therapeutics: Where Are We Now? Front. Neurosci. 2019, 13, 1310. [Google Scholar] [CrossRef] [PubMed]
- Appel, S.H.; Zhao, W.; Beers, D.R.; Henkel, J.S. The microglial-motoneuron dialogue in ALS. Acta. Myol. 2011, 30, 4–8. [Google Scholar] [PubMed]
- Chiu, I.M.; Morimoto, E.T.A.; Goodarzi, H.; Liao, J.T.; O’Keeffe, S.; Phatnani, H.P.; Muratet, M.; Carroll, M.C.; Levy, S.; Tavazoie, S.; et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 2013, 4, 385–401. [Google Scholar] [CrossRef] [PubMed]
- Gravel, M.; Beland, L.-C.; Soucy, G.; Abdelhamid, E.; Rahimian, R.; Gravel, C.; Kriz, J. IL-10 Controls Early Microglial Phenotypes and Disease Onset in ALS Caused by Misfolded Superoxide Dismutase 1. J. Neurosci. 2016, 36, 1031–1048. [Google Scholar] [CrossRef] [PubMed]
- Beers, D.R.; Zhao, W.; Liao, B.; Kano, O.; Wang, J.; Huang, A.; Appel, S.H.; Henkel, J.S. Neuroinflammation modulates distinct regional and temporal clinical responses in ALS mice. Brain Behav. Immun. 2011, 25, 1025–1035. [Google Scholar] [CrossRef]
- Zhao, W.; Beers, D.R.; Liao, B.; Henkel, J.S.; Appel, S.H. Regulatory T lymphocytes from ALS mice suppress microglia and effector T lymphocytes through different cytokine-mediated mechanisms. Neurobiol. Dis. 2012, 48, 418–428. [Google Scholar] [CrossRef]
- Kang, J.; Rivest, S. MyD88-deficient bone marrow cells accelerate onset and reduce survival in a mouse model of amyotrophic lateral sclerosis. J. Cell Biol. 2007, 179, 12191230. [Google Scholar] [CrossRef]
- Hailman, E.; Lichenstein, H.S.; Wurfel, M.M.; Miller, D.S.; Johnson, D.A.; Kelley, M.; Busse, L.A.; Zukowski, M.M.; Wright, S.D. Lipopolysaccharide (LPS)-binding protein accelerates the binding of LPS to CD14. J. Exp. Med. 1994, 179, 269–277. [Google Scholar] [CrossRef]
- Sun, H.; Yang, Y.; Gu, M.; Li, Y.; Jiao, Z.; Lu, C.; Li, B.; Jiang, Y.; Jiang, L.; Chu, F.; et al. The role of Fas-FasL-FADD signaling pathway in arsenic-mediated neuronal apoptosis in vivo and in vitro. Toxicol. Lett. 2022, 356, 143–150. [Google Scholar] [CrossRef]
- Liu, Y.; Hao, W.; Dawson, A.; Liu, S.; Fassbender, K. Expression of amyotrophic lateral sclerosis-linked SOD1 mutant increases the neurotoxic potential of microglia via TLR2. J. Biol. Chem. 2009, 284, 3691–3699. [Google Scholar] [CrossRef]
- Turner, M.R.; Cagnin, A.; Turkheimer, F.E.; Miller, C.C.J.; Shaw, C.E.; Brooks, D.J.; Leigh, P.N.; Banati, R.B. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: An [11C](R)-PK11195 positron emission tomography study. Neurobiol. Dis. 2004, 15, 601–609. [Google Scholar] [CrossRef] [PubMed]
- Nutma, E.; Fancy, N.; Weinert, M.; Tsartsalis, S.; Marzin, M.C.; Muirhead, R.C.J.; Falk, I.; Breur, M.; de Bruin, J.; Hollaus, D.; et al. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases. Nat. Commun. 2023, 14, 5247. [Google Scholar] [CrossRef] [PubMed]
- Weiner, H.L. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat. Rev. Neurol. 2025, 21, 67–85. [Google Scholar] [CrossRef]
- McGill, R.B.; Steyn, F.J.; Ngo, S.T.; Thorpe Ka Heggie, S.; Ruitenberg, M.J.; Henderson, R.D.; McCombe, P.A.; Woodruff, T.M. Monocytes and neutrophils are associated with clinical features in amyotrophic lateral sclerosis. Brain Commun. 2020, 2, fcaa013. [Google Scholar] [CrossRef]
- Yazdani, S.; Seitz, C.; Cui, C.; Lovik, A.; Piehl, F.; Pawitan, Y.; Klappe, U.; Press, R.; Samuelsson, K.; Yin, L.; et al. T cell responses at diagnosis of amyotrophic lateral sclerosis predict disease progression. Nat. Commun. 2022, 13, 6733. [Google Scholar] [CrossRef]
- Yangou, Y.; Facer, P.; Durrenberger, P.; Chessell, I.P.; Naylor, A.; Bountra, C.; Banati, R.R.; Anand, P. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006, 6, 12. [Google Scholar] [CrossRef] [PubMed]
- Mohd Sairazi, N.S.; Sirajudeen, K.N.S. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases. Evid. Based Complement. Altern. Med. 2020, 2020, 6565396. [Google Scholar] [CrossRef]
- Jia, C.; Chai, J.; Zhang, S.; Sun, Y.; He, L.; Sang, Z.; Chen, D.; Zheng, X. The Advancements of Marine Natural Products in the Treatment of Alzheimer’s Disease: A Study Based on Cell and Animal Experiments. Mar. Drugs 2025, 23, 91. [Google Scholar] [CrossRef]
- Silva, J.; Alves, C.; Soledade, F.; Martins, A.; Pinteus, S.; Gaspar, H.; Alfonso, A.; Pedrosa, R. Marine-Derived Components: Can They Be a Potential Therapeutic Approach to Parkinson’s Disease? Mar. Drugs 2023, 21, 451. [Google Scholar] [CrossRef]
- Kemp, D.C.; Ewon, J.Y. Fish and Shellfish-Derived Anti-Inflammatory Protein Products: Properties and Mechanisms. Molecules 2021, 26, 3225. [Google Scholar] [CrossRef]
- Ripps, H.; Shen, W. Review: Taurine: A "very essential" amino acid. Mol. Vis. 2012, 18, 2673. [Google Scholar] [PubMed]
- Choi, J.Y.; Jang, J.S.; Son, D.J.; Im, H.-S.; Kim, J.Y.; Park, J.E.; Choi, W.R.; Han, S.-B.; Hong, J.T. Antarctic Krill Oil Diet Protects against Lipopolysaccharide-Induced Oxidative Stress, Neuroinflammation and Cognitive Impairment. Int. J. Mol. Sci. 2017, 18, 2554. [Google Scholar] [CrossRef] [PubMed]
- Parolini, C.; Bjorndal, B.; Busnelli, M.; Manzini, M.; Ganzetti, G.S.; Dellera, F.; Ramsvik, M.; Bruheim, I.; Berge, R.F.; Chiesa, G. Effect of Dietary Components from Antarctic Krill on Atherosclerosis in apoE-Deficient Mice. Mol. Nutr. Food Res. 2017, 61, 1700098. [Google Scholar] [CrossRef]
- Wang, K.; Li, Y.; Dai, Y.; Han, L.; Zhu, Y.; Xue, C.; Wang, P.; Wang, J. Peptides from Antarctic Krill (Euphausia superba) Improve Osteoarthritis via Inhibiting HIF-2α-Mediated Death Receptor Apoptosis and Metabolism Regulation in Osteoarthritic Mice. J. Agric. Food Chem. 2019, 67, 3125–3133. [Google Scholar] [CrossRef] [PubMed]
- Yue, H.; Cai, W.; Bai, X.; Dong, P.; Wang, J. Antarctic krill peptide alleviates liver fibrosis via downregulating the secondary bile acid mediated NLRP3 signaling pathway. Food Funct. 2022, 13, 7740–7749. [Google Scholar] [CrossRef]
- Zheng, J.; Gao, Y.; Ding, J.; Sun, N.; Lin, S. Antarctic krill peptides improve scopolamine-induced memory impairment in mice. Food Biosci. 2022, 49, 101987. [Google Scholar] [CrossRef]
- Yang, J.; Qi, Y.; Zhu, B.; Lin, S. A Novel Tetrapeptide Ala-Phe-Phe-Pro (AFFP) Derived from Antarctic Krill Prevents Scopolamine-Induced Memory Disorder by Balancing Lipid Metabolism of Mice Hippocampus. Nutrients 2024, 16, 1019. [Google Scholar] [CrossRef]
- Yang, T.; Meng, Y.; Li, S.; Zhao, X.; Hou, H. Preparation, unique structural characteristics and immunomodulatory effects of peptides from Antarctic krill (Euphausia superba). Food Res. Int. 2025, 214, 116592. [Google Scholar] [CrossRef]
- Xu, X.; Liang, R.; Li, D.; Jiang, C.; Lin, S. Evaluation of sea cucumber peptides-assisted memory activity and acetylation modification in hippocampus of test mice based on scopolamine-induced experimental animal model of memory disorder. J. Funct. Foods 2020, 68, 103909. [Google Scholar] [CrossRef]
- Lu, X.; Xu, X.; Li, D.; Sun, N.; Lin, S. Sea Cucumber Peptides Attenuated the Scopolamine-Induced Memory Impairment in Mice and Rats and the Underlying Mechanism. J. Agric. Food Chem. 2022, 70, 157–170. [Google Scholar] [CrossRef] [PubMed]
- Gong, H.; Zhao Mao, X. Sea Cucumber Hydrolysates Alleviate Cognitive Deficits in D-Galactose-Induced C57BL/6J Aging Mice Associated with Modulation of Gut Microbiota. Foods 2025, 14, 1938. [Google Scholar] [CrossRef]
- Ren, Q.; Jiang, X.; Zhang, S.; Gao, X.; Paudel, Y.N.; Zhang, P.; Wang, R.; Liu, K.; Jin, M. Neuroprotective effect of YIAEDAER peptide against Parkinson’s disease like pathology in zebrafish. Biomed. Pharmacother. 2022, 147, 112629. [Google Scholar] [CrossRef]
- Fakhri, S.; Dargahi, L.; Abbaszadeh, F.; Jorjani, M. Astaxanthin attenuates neuroinflammation contributed to the neuropathic pain and motor dysfunction following compression spinal cord injury. Brain Res. Bull. 2018, 143, 217–224. [Google Scholar] [CrossRef]
- Rahman, S.O.; Panda, B.P.; Parvez, S.; Kaundal, M.; Hussain, S.; Akhtar, M.; Najmi, A.K. Neuroprotective role of astaxanthin in hippocampal insulin resistance induced by Aβ peptides in animal model of Alzheimer’s disease. Biomed. Pharmacother. 2019, 110, 47–58. [Google Scholar] [CrossRef]
- Liu, N.; Lyiu, X.; Zhang, X.; Zhang, F.; Chen, Y.; Li, G. Astaxanthin attenuates cognitive deficits in Alzheimer’s disease models by reducing oxidative stress via the SIRT1/PGC-1α signaling pathway. Cell Biosci. 2023, 13, 173. [Google Scholar] [CrossRef]
- Bidaran, S.; Ahmadi, A.R.; Yaghmaei, P.; Sanati, M.H.; Ebrahim-Habibi, A. Astaxanthin effectiveness in preventing multiple sclerosis in animal model. Bratisl. Lek. Listy 2018, 119, 160–166. [Google Scholar] [CrossRef]
- Lofti, A.; Soleimani, M.; Ghasemi, N. Astaxanthin Reduces Demyelination and Oligodendrocytes Death in A Rat Model of Multiple Sclerosis. Cell J. 2021, 22, 565–571. [Google Scholar] [CrossRef]
- Wei, H.; Gao, Z.; Zheng, L.; Zhang, C.; Liu, Z.; Yang, Y.; Teng, H.; Hou, L.; Yin, Y.; Zou, X. Protective Effects of Fucoidan on Aβ25-35 and d-Gal-Induced Neurotoxicity in PC12 Cells and d-Gal-Induced Cognitive Dysfunction in Mice. Mar. Drugs 2017, 15, 77. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Li, C.; Yin, J.; Shen, J.; Wang, H.; Wu, Y.; Jin, H. Fucoidan, a sulfated polysaccharide from brown algae, improves cognitive impairment induced by infusion of Aβ peptide in rats. Environ. Toxicol. Pharmacol. 2012, 33, 304–311. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Wang, X.; Pan, Y.; Wang, G.; Mao, G. The degraded polysaccharide from Pyropia haitanensis represses amyloid beta peptide-induced neurotoxicity and memory in vivo. Int. J. Biol. Macromol. 2020, 146, 725–729. [Google Scholar] [CrossRef]
- Park, S.K.; Kang, J.Y.; Kim, J.M.; Yoo, S.K.; Han, H.J.; Chung, D.H.; Kim, D.-O.; Kim, G.-H.; Heo, H.J. Fucoidan-Rich Substances from Ecklonia cava Improve Trimethyltin-Induced Cognitive Dysfunction via Down-Regulation of Amyloid β Production/Tau Hyperphosphorylation. Mar. Drugs 2019, 17, 591. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yi, K.; Zhao, Y. Fucoidan inhibits amyloid-β-induced toxicity in transgenic Caenorhabditis elegans by reducing the accumulation of amyloid-β and decreasing the production of reactive oxygen species. Food Funct. 2018, 9, 552–560. [Google Scholar] [CrossRef]
- Zhang, L.; Hao, J.; Zheng, Y.; Su, R.; Liao, Y.; Gong, X.; Liu, L.; Wang, X. Fucoidan Protects Dopaminergic Neurons by Enhancing the Mitochondrial Function in a Rotenone-induced Rat Model of Parkinson’s Disease. Aging Dis. 2018, 9, 590–604. [Google Scholar] [CrossRef] [PubMed]
- Luo, D.; Zhang, Q.; Wang, H.; Cui, Y.; Sun, Z.; Yang, J.; Zheng, Y.; Jia, J.; Yu, F.; Wang, X.; et al. Fucoidan protects against dopaminergic neuron death in vivo and in vitro. Eur. J. Pharmacol. 2009, 617, 33–40. [Google Scholar] [CrossRef]
- Zhang, F.-L.; He, Y.; Zheng, Y.; Zhang, W.-J.; Wang, Q.; Jia, Y.-J.; Song, H.-L.; An, H.-T.; Zhang, H.-B.; Qian, Y.-J.; et al. Therapeutic effects of fucoidan in 6-hydroxydopamine-lesioned rat model of Parkinson’s disease: Role of NADPH oxidase-1. CNS Neurosci. Ther. 2014, 20, 1036–1044. [Google Scholar] [CrossRef]
- Cui, Y.Q.; Jia, Y.-J.; Zhang, T.; Zhang, Q.-B.; Wang, X.-M. Fucoidan protects against lipopolysaccharide-induced rat neuronal damage and inhibits the production of proinflammatory mediators in primary microglia. CNS Neurosci. Ther. 2012, 18, 827–833. [Google Scholar] [CrossRef]
- Meenakshi, S.; Umayaparvathi, S.; Saravanan, R.; Manivasagam, T.; Balasubramanian, T. Neuroprotective effect of fucoidan from Turbinaria decurrens in MPTP intoxicated Parkinsonic mice. Int. J. Biol. Macromol. 2016, 86, 425–433. [Google Scholar] [CrossRef] [PubMed]
- Xing, M.; Li, G.; Liu, Y.; Yang, L.; Zhang, Y.; Zhang, Y.; Ding, J.; Lu, M.; Yu, G.; Hu, G. Fucoidan from Fucus vesiculosus prevents the loss of dopaminergic neurons by alleviating mitochondrial dysfunction through targeting ATP5F1a. Carbohydr. Polym. 2023, 303, 120470. [Google Scholar] [CrossRef]
- Yang, X.; Zhang, X.; Ma, Y.; Li, S.; Wang, Q.; Hong, J.-S.; Yu, G.; Qi, B.; Wang, J.; Liu, C.; et al. Fucoidan ameliorates rotenone-induced Parkinsonism in mice by regulating the microbiota-gut-brain axis. Int. J. Biol. Macromol. 2024, 283, 137373. [Google Scholar] [CrossRef]
- Kim, H.; Moon, C.; Park, E.; Jee, Y.; Ahn, M.; Wie, M.B.; Shin, T. Amelioration of experimental autoimmune encephalomyelitis in Lewis rats treated with fucoidan. Phytother. Res. 2010, 24, 399–403. [Google Scholar] [CrossRef]
- Um, M.Y.; Lim, D.W.; Son, H.J.; Cho, S.; Lee, C. hlorotannin-rich fraction from Ishige foliacea brown seaweed prevents the scopolamine-induced memory impairment via regulation of ERK-CREB-BDNF pathway. J. Funct. Foods 2018, 40, 110–116. [Google Scholar] [CrossRef]
- Kim, T.-E.; Son, H.J.; Lim, D.W.; Yoon, M.; Lee, J.; Kim, Y.T.; Han, D.; Lee, C.; Um, M.Y. Memory-enhancing effects of Ishige foliacea extract: In Vitro and in vivo study. J. Food Biochem. 2020, 44, e13162. [Google Scholar] [CrossRef] [PubMed]
- Kwon, O.Y.; Lee, S.H. Ishige okamurae Suppresses Trimethyltin-Induced Neurodegeneration and Glutamate-Mediated Excitotoxicity by Regulating MAPKs/Nrf2/HO-1 Antioxidant Pathways. Antioxidants 2021, 10, 440. [Google Scholar] [CrossRef]
- Yasuda, Y.; Tokumatsu, T.; Ueda, C.; Sakai, M.; Sasaki, Y.; Norikura, T.; Matsui-Yuasa, I.; Kojima-Yuasa, A. Ecklonia cava Polyphenols Have a Preventive Effect on Parkinson’s Disease through the Activation of the Nrf2-ARE Pathway. Nutrients 2024, 16, 2076. [Google Scholar] [CrossRef]
- Houser, M.C.; Chang, J.; Factor, S.A.; Molho, E.S.; Zabetian, C.P.; Hill-Burns, E.M.; Payami, H.; Hertzberg, V.S.; Tansey, M.G. Stool Immune Profiles Evince Gastrointestinal Inflammation in Parkinson’s Disease. Mov. Disord. 2018, 33, 793–804. [Google Scholar] [CrossRef] [PubMed]
- Illiano, P.; Brambilla, R.; Parolini, C. The mutual interplay of gut microbiota, diet and human disease. FEBS J. 2020, 287, 833–855. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-F.; Fan, Z.-K.; Gao, X.; Zhou, F.; Guo, X.-F.; Sinclair, A.J.; Li, D. n-3 polyunsaturated fatty acids in phospholipid or triacylglycerol form attenuate nonalcoholic fatty liver disease via mediating cannabinoid receptor 1/adiponectin/ceramide pathway. J. Nutr. Biochem. 2024, 123, 109484. [Google Scholar] [CrossRef]
- Li, K.; Song, X.; Li, H.; Kuang, X.; Liu, S.; Liu, R.; Li, D. Mussel oil is superior to fish oil in preventing atherosclerosis of ApoE-/-mice. Front. Nutr. 2024, 11, 1326421. [Google Scholar] [CrossRef]
- Alugoju, P.; Krishna Swamy, V.K.D.; Anthikapalli, N.V.A.; Tencomnao, T. Health benefits of astaxanthin against age-related diseases of multiple organs: A comprehensive review. Crit. Rev. Food Sci. Nutr. 2023, 63, 10709–10774. [Google Scholar] [CrossRef]
- Wang, C.-C.; Shi, H.-H.; Xu, J.; Yanagita, T.; Xue, C.-H.; Zhang, T.-T.; Wang, Y.-M. Docosahexaenoic acid-acylated astaxanthin ester exhibits superior performance over non-esterified astaxanthin in preventing behavioral deficits coupled with apoptosis in MPTP-induced mice with Parkinson’s disease. Food Funct. 2010, 11, 8038–8050. [Google Scholar] [CrossRef]
- Wang, L.; Lu, K.; Lou, X.; Zhang, S.; Song, W.; Li, R.; Geng, L.; Cheng, B. Astaxanthin ameliorates dopaminergic neuron damage in paraquat-induced SH-SY5Y cells and mouse models of Parkinson’s disease. Brain Res. Bull. 2023, 202, 110762. [Google Scholar] [CrossRef]
- Park, J.S.; Chyn, J.H.; Kim, Y.K.; Line, L.L.; Chew, B.P. Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr. Metab. 2010, 7, 18. [Google Scholar] [CrossRef] [PubMed]
- Thomas, N.V.; Kim, S.-W. Beneficial effects of marine algal compounds in cosmeceuticals. Mar. Drugs 2013, 11, 146–164. [Google Scholar] [CrossRef]
- Holdt, S.L.; Kraan, S. Bioactive compounds in seaweed: Functional food applications and legislation. J. Appl. Phycol. 2011, 23, 543–597. [Google Scholar] [CrossRef]
- Dimitrova-Shumkovska, J.; Krstanoski, L.; Veenman, L. Potential Beneficial Actions of Fucoidan in Brain and Liver Injury, Disease, and Intoxication-Potential Implication of Sirtuins. Mar. Drugs 2020, 18, 242. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Wang, Q.; Han, X.; Ma, Y.; Zhang, Z.; Zhao, L.; Guan, F.; Ma, S. Fucoidan: A promising agent for brain injury and neurodegenerative disease intervention. Food Funct. 2021, 12, 3820–3830. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, C.-H.; Lu, C.-H.; Kuo, Y.-Y.; Lin, G.-B.; Chao, C.-Y. The protective effect of non-invasive low intensity pulsed electric field and fucoidan in preventing oxidative stress-induced motor neuron death via ROCK/Akt pathway. PLoS ONE 2019, 14, e0214100. [Google Scholar] [CrossRef]
- Heffernan, N.; Brunton, N.P.; FtzGerald, R.J.; Smyth, T.J. Profiling of the molecular weight and structural isomer abundance of macroalgae-derived phlorotannins. Mar. Drugs 2015, 13, 509–528. [Google Scholar] [CrossRef]
- Murugan, A.C.; Karim, M.R.; Yusoff, M.B.; Tan, S.H.; Asras, M.F.; Rashid, S.S. New insights into seaweed polyphenols on glucose homeostasis. Pharm. Biol. 2015, 53, 1087–1097. [Google Scholar] [CrossRef]
- Murray, M.; Dordevic, A.L.; Ryan, L.; Bonham, M.P. An emerging trend in functional foods for the prevention of cardiovascular disease and diabetes: Marine algal polyphenols. Crit. Rev. Food Sci. Nutr. 2018, 58, 1342–1358. [Google Scholar] [CrossRef]
- Kang, S.-M.; Cha, S.-H.; Ko, J.-Y.; Kang, M.-C.; Kim, D.; Heo, S.-J.; Kim, J.-S.; Heu, M.S.; Kim, Y.-T.; Jung, W.-K.; et al. Neuroprotective effects of phlorotannins isolated from a brown alga, Ecklonia cava, against H2O2-induced oxidative stress in murine hippocampal HT22 cells. Environ. Toxicol. Pharmacol. 2012, 34, 96–105. [Google Scholar] [CrossRef] [PubMed]
- Heo, S.-J.; Cha, S.-H.; Kim, K.-N.; Lee, S.-H.; Ahn, G.; Kang, D.-H.; Oh, C.; Choi, Y.-U.; Affan, A.; Kim, D.; et al. Neuroprotective effect of phlorotannin isolated from Ishige okamurae against H2O2-induced oxidative stress in murine hippocampal neuronal cells, HT22. Appl. Biochem. Biotechnol. 2012, 166, 1520–1532. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Amarsanaa, K.; Lee, J.H.; Rhim, J.-K.; Kwon, J.M.; Kim, S.-H.; Park, J.M.; Jung, S.-C.; Eun, S.-Y. Neuroprotective mechanisms of dieckol against glutamate toxicity through reactive oxygen species scavenging and nuclear factor-like 2/heme oxygenase-1 pathway. Korean J. Physiol. Pharmacol. 2019, 23, 121–130. [Google Scholar] [CrossRef]
- Yang, Y.-I.; Jung, S.-H.; Lee, K.-T.; Choi, J.-H. 8,8′-Bieckol, isolated from edible brown algae, exerts its anti-inflammatory effects through inhibition of NF-κB signaling and ROS production in LPS-stimulated macrophages. Int. Immunopharmacol. 2014, 23, 460–468. [Google Scholar] [CrossRef]
- Ahmad, F.; Sachdeva, P.; Sachdeva, B.; Singh, G.; Soni, H.; Tandon, S.; Rafeeq, M.M.; Alam, Z.M.; Baeissa, H.M.; Khalid, M. Dioxinodehydroeckol: A Potential Neuroprotective Marine Compound Identified by In Silico Screening for the Treatment and Management of Multiple Brain Disorders. Mol. Biotechnol. 2024, 66, 663–686. [Google Scholar] [CrossRef] [PubMed]
- Gao, C.; Gong, N.; Chen, F.; Hu, S.; Zhou, Q.; Gao, X. The Effects of Astaxanthin on Metabolic Syndrome: A Comprehensive Review. Mar. Drugs 2024, 23, 9. [Google Scholar] [CrossRef]
- Nouchi, R.; Suiko, T.; Kimura, E.; Takenada, H.; Murakoshi, M.; Uchiyama, A.; Aono, M.; Kawashima, R. Effects of Lutein and Astaxanthin Intake on the Improvement of Cognitive Functions among Healthy Adults: A Systematic Review of Randomized Controlled Trials. Nutrients 2020, 12, 617. [Google Scholar] [CrossRef]
- Hecht, K.A.; Marwah, M.; Wood, V.; Nishida, Y.; Bach, A.E.; Gerson, J.; Hom, M.M.; Schnackenberg, J.; Raote, S.; Srivastava, S.; et al. Astaxanthin (AstaReal®) Improved Acute and Chronic Digital Eye Strain in Children: A Randomized Double-Blind Placebo-Controlled Trial. Adv. Ther. 2025, 42, 1811–1833. [Google Scholar] [CrossRef]
- Li, P.; Song, C. Potential treatment of Parkinson’s disease with omega-3 polyunsaturated fatty acids. Nutr. Neurosci. 2022, 25, 180–191. [Google Scholar] [CrossRef]


| Compound | Species | Models | Effects/Mode of Action | Refs |
|---|---|---|---|---|
Peptides![]() | Euphasia superba | Scopolamine-treated mice Immunosuppressed mice | ↓neuronal cell harm ↓SOD activity ↓ROS levels Immunomodulation | [99,100,101] |
| Stichopus japonicus | Scopolamine-treated mice and rats | ↓acetylcholinesterase activity ↑acetylcholine content ↑LTP pathway ↑unsaturated lipid levels | [102,103] | |
| Stichopus japonicus | D-galactose-induced aging mice | ↓neuroinflammation ↑BDNF/TrkB ↓NF-kB signaling ↓gut dysbiosis ↓fecal short-chain fatty acids levels | [104] | |
| Neptunea arthritica cumingii | Zebrafish | ↓locomotor impairment ↓dopaminergic neuron loss ↓loss of cerebral vessels | [105] | |
Astaxanthin![]() | Heamatococcus pluvialis | SCI mice | ↓post-SCI motor dysfunction ↓NF-kB signaling ↓pro-inflammatory cytokine ↑antioxidant defenses | [106] |
| Heamatococcus pluvialis | Abeta-injected mice | ↓cognitive deficits ↓GSK-3beta activity | [107] | |
| Heamatococcus pluvialis | Abeta-injected mice | ↓cognitive deficits ↓GSK-3beta activity | [108] | |
| Heamatococcus pluvialis | EAE mice | ↑disease prevention/progression ↓pro-inflammatory cytokine ↑Treg differentiation | [109] | |
| Heamatococcus pluvialis | MS rats | ↓demyelination ↓oligodendrocyte death | [110] | |
Fucoidan![]() | Undaria Pinnatifida | Abeta-injected mice | ↓learning memory impairment ↑cholinergic system ↓oxidation ↓mitochondrial apoptosis ↓caspase pathway | [111] |
| Laminaria japonica | D-galactose mice | Vlearning memory impairment ↑cholinergic system ↓oxidation ↓mitochondrial apoptosis ↓caspase pathway | [112] | |
| Fucoidan | Pyropia haitanensis | Abeta-injected mice | ↓learning memory impairment ↓oxidation ↓mitochondrial apoptosis | [113] |
| Ecklonia cava | TMT-treated mice | ↓cognitive dysfunctions ↓Abeta production ↓tau phosphorylation | [114] | |
| Brown algae | AD transgenic C. elegans | ↓cognitive dysfunctions ↓Abeta production ↓tau phosphorylation | [115] | |
| Laminaria japonica | Rotenone-treated rats MPTP-treated mice 6-OHDA-treated rats | ↓mitochondrial dysfunction by PGC-1alpha/NRF2 pathway ↑antioxidant activity ↓oxidation ↓microglial activation | [116,117,118] | |
| Brown algae | LPS-treated rats | ↓behavioral deficits ↓microglial activation | [119] | |
| Turbinaria decurrens | MPTP-treated mice | ↓behavioral deficits ↓microglial activation | [120] | |
| Fucus vesiculosus | MPTP-treated mice | ↓mitochondrial dysfunction ↓motor deficits ↓neuronal apoptosis ↓dopaminergic neuron loss | [121] | |
| Laminaria japonica | Rotenone-treated mice | ↓gut dysbiosis ↓permeability of the intestinal barrier ↓pro-inflammatory levels | [122] | |
| -- | EAE rats | ↓demyelination ↓autoreactive T cell response ↓pro-inflammatory levels | [123] | |
Polyphenols![]() | Ishige foliacea | Scopolamine-treated mice | ↓cognitive dysfunctions ↓acetylcholinesterase activity ↓lipid peroxidation ↑ROS scavenging ↑BDNF | [124] |
| Ishige foliacea | Scopolamine-treated mice | ↓cognitive dysfunctions ↓acetylcholinesterase activity ↓lipid peroxidation ↑ROS scavenging ↑BDNF | [125] | |
| Ishige okamurae | TMT-treated mice | ↓memory impairments ↓cellular apoptosis ↑BDNF ↑Nrf2 and HO-1 | [126] | |
| Ecklonia cava | PD mice | ↓dopaminergic neuronal death ↓motor deficits ↑intestinal motility ↑colon tissue morphology | [127] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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
Favari, E.; Parolini, C. Marine Bioactive Components and Chronic Neuroinflammation: Focus on Neurodegenerative Disease. Mar. Drugs 2025, 23, 446. https://doi.org/10.3390/md23110446
Favari E, Parolini C. Marine Bioactive Components and Chronic Neuroinflammation: Focus on Neurodegenerative Disease. Marine Drugs. 2025; 23(11):446. https://doi.org/10.3390/md23110446
Chicago/Turabian StyleFavari, Elda, and Cinzia Parolini. 2025. "Marine Bioactive Components and Chronic Neuroinflammation: Focus on Neurodegenerative Disease" Marine Drugs 23, no. 11: 446. https://doi.org/10.3390/md23110446
APA StyleFavari, E., & Parolini, C. (2025). Marine Bioactive Components and Chronic Neuroinflammation: Focus on Neurodegenerative Disease. Marine Drugs, 23(11), 446. https://doi.org/10.3390/md23110446





