Immunomodulatory natural polysaccharide-based nanoparticles for the treatment of neurodegenerative diseases

Leto-Aikaterini Tziveleka , Mariafrancesca Cascione , Paolo Pellegrino , Annalisa Bianco , Stefano Leporatti , Valeria De Matteis

Ibrain ›› 2025, Vol. 11 ›› Issue (3) : 277 -296.

PDF
Ibrain ›› 2025, Vol. 11 ›› Issue (3) : 277 -296. DOI: 10.1002/ibra.12199
REVIEW

Immunomodulatory natural polysaccharide-based nanoparticles for the treatment of neurodegenerative diseases

Author information +
History +
PDF

Abstract

Polysaccharide-based nanoparticles offer significant potential for the treatment of neurodegenerative diseases and the modulation of inflammatory responses in the central nervous system. These biopolymers, when derived from natural sources, possess inherent immunomodulatory properties, which can be leveraged to regulate immune activity, positioning them as promising candidates for both prophylactic and therapeutic strategies. Furthermore, when integrated with other materials, polysaccharides form nanocomposites with enhanced structural, physicochemical, and biological properties, making them highly versatile platforms for drug delivery in the central nervous system. This review provides a comprehensive analysis of polysaccharide-based nanoparticles, focusing on their application in the treatment of three major neurodegenerative diseases: Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Emphasis is placed on optimizing these nanomaterials for targeted drug delivery and immune modulation, underscoring their potential to improve therapeutic outcomes in neurodegenerative disorders. The review also examines the structural, chemical, and biological characteristics of key polysaccharides, and explores their innovative roles in combating neuroinflammation and neurodegeneration.

Keywords

immunomodulation / nanoparticles / natural polysaccharides / neurodegenerative disorders

Cite this article

Download citation ▾
Leto-Aikaterini Tziveleka, Mariafrancesca Cascione, Paolo Pellegrino, Annalisa Bianco, Stefano Leporatti, Valeria De Matteis. Immunomodulatory natural polysaccharide-based nanoparticles for the treatment of neurodegenerative diseases. Ibrain, 2025, 11(3): 277-296 DOI:10.1002/ibra.12199

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Chaplin DD. Overview of the immune response. J Allergy Clin Immunol. 2010; 125(2 suppl 2): S3-S23.

[2]

Ben-Akiva E, Est Witte S, Meyer RA, Rhodes KR, Green JJ. Polymeric micro- and nanoparticles for immune modulation. Biomater Sci. 2019; 7(1): 14-30.

[3]

Jain NK, Mishra V, Mehra NK. Targeted drug delivery to macrophages. Expert Opin Drug Delivery. 2013; 10(3): 353-367.

[4]

Li J, Jiang X, Li H, Gelinsky M, Gu Z. Tailoring materials for modulation of macrophage fate. Adv Mater. 2021; 33(12):2004172.

[5]

Mason HD, McGavern DB. How the immune system shapes neurodegenerative diseases. Trends Neurosci. 2022; 45(10): 733-748.

[6]

Schwartz M, Cahalon L. The vicious cycle governing the brain-immune system relationship in neurodegenerative diseases. Curr Opin Immunol. 2022; d76:102182.

[7]

Rafati N, Zarepour A, Bigham A. Nanosystems for targeted drug delivery: innovations and challenges inovercoming the blood-brain barrier for neurodegenerative disease and cancer therapy. IntJPharm. 2024; 666:124800.

[8]

Yuan C, Liu Y, Wang T, Sun M, Chen X. Nanomaterials as smart immunomodulator delivery system for enhanced cancer therapy. ACS Biomater Sci Eng. 2020; 6(9): 4774-4798.

[9]

Duncan R, Vicent MJ. Polymer therapeutics-prospects for 21st century: the end of the beginning. Adv Drug Deliv Rev. 2013; 65(1): 60-70.

[10]

Li J, Yu F, Chen Y, Oupický D. Polymeric drugs: advances in the development of pharmacologically active polymers. J Controlled Release. 2015; 219: 369-382.

[11]

Prasher P, Sharma M, Mehta M, et al. Current-status and applications of polysaccharides in drug delivery systems. Colloid Interface Sci Commun. 2021; 42:100418.

[12]

Torres FG, Troncoso OP, Pisani A, Gatto F, Bardi G. Natural polysaccharide nanomaterials: an overview of their immunological properties. Int J Mol Sci. 2019; 20(20):5092.

[13]

Kajiwara K, Miyamoto T. Progress in structural characterization of functional polysaccharides. In: Dumitriu S, ed. Polysaccharides Structural Diversity and Functional Versatility. CRC Press; 2004.

[14]

Heinze T. Cellulose: Structure and properties. In: Rojas OJ, ed. Cellulose Chemistry and Properties: Fibers, Nanocelluloses and Advanced Materials. Advances in Polymer Science. Switzerland, Springer International Publishing; 2015: 1-52.

[15]

Szekalska M, Puciłowska A, Szymańska E, Ciosek P, Winnicka K. Alginate: current use and future perspectives in pharmaceutical and biomedical applications. Int J Polym Sci. 2016; 2016: 1-17.

[16]

Dicker KT, Gurski LA, Pradhan-Bhatt S, Witt RL, Farach-Carson MC, Jia X. Hyaluronan: a simple polysaccharide with diverse biological functions. Acta Biomater. 2014; 10(4): 1558-1570.

[17]

Wu GJ, Shiu SM, Hsieh MC, Tsai GJ. Anti-inflammatory activity of a sulfated polysaccharide from the brown alga Sargassum cristaefolium. Food Hydrocolloids. 2016; 53: 16-23.

[18]

Guo L, Ma R, Sun H, Raza A, Tang J, Li Z. Anti-inflammatory activities and related mechanism of polysaccharides isolated from Sargentodoxa cuneata. Chem Biodiversity. 2018; 15(11):e1800343.

[19]

Zhang W, Zhang X, Zou K, et al. Seabuckthorn berry polysaccharide protects against carbon tetrachloride-induced hepatotoxicity in mice via anti-oxidative and anti-inflammatory activities. Food Funct. 2017; 8(9): 3130-3138.

[20]

Li N, Wang C, Georgiev MI, et al. Advances in dietary polysaccharides as anticancer agents: structure-activity relationship. Trends Food Sci Technol. 2021; 111: 360-377.

[21]

Ljungman AG, Leanderson P, Tagesson C. (1 → 3)-β-d-Glucan stimulates nitric oxide generation and cytokine mRNA expression in macrophages. Environ Toxicol Pharmacol. 1998; 5(4): 273-281.

[22]

Gupta P, Nayak KK. Characteristics of protein-based biopolymer and its application. Polym Eng Sci. 2015; 55(3): 485-498.

[23]

Diener M, Adamcik J, Sánchez-Ferrer A, Jaedig F, Schefer L, Mezzenga R. Primary, secondary, tertiary and quaternary structure levels in linear polysaccharides: from random coil, to single helix to supramolecular assembly. Biomacromolecules. 2019; 20(4): 1731-1739.

[24]

Yang L, Zhang LM. Chemical structural and chain conformational characterization of some bioactive polysaccharides isolated from natural sources. Carbohydr Polym. 2009; 76(3): 349-361.

[25]

El Seoud O, Nawaz H, Arêas E. Chemistry and applications of polysaccharide solutions in strong electrolytes/dipolar aprotic solvents: an overview. Molecules. 2013; 18(1): 1270-1313.

[26]

Seidi F, Yazdi MK, Jouyandeh M, et al. Crystalline polysaccharides: a review. Carbohydr Polym. 2022; 275:118624.

[27]

Forget A, Christensen J, Lüdeke S, et al. Polysaccharide hydrogels with tunable stiffness and provasculogenic properties via α-helix to β-sheet switch in secondary structure. Proc Natl Acad Sci2013; 110(32): 12887-12892.

[28]

Loh CCJ. Exploiting non-covalent interactions in selective carbohydrate synthesis. Nat Rev Chem. 2021; 5(11): 792-815.

[29]

Ferreira SS, Passos CP, Madureira P, Vilanova M, Coimbra MA. Structure-function relationships of immunostimulatory polysaccharides: a review. Carbohydr Polym. 2015; 132: 378-396.

[30]

Hou C, Chen L, Yang L, Ji X. An insight into anti-inflammatory effects of natural polysaccharides. Int J Biol Macromol. 2020; 153: 248-255.

[31]

Ji X, Peng Q, Wang M. Anti-colon-cancer effects of polysaccharides: a minireview of the mechanisms. Int J Biol Macromol. 2018; 114: 1127-1133.

[32]

Meng LZ, Feng K, Wang LY, et al. Activation of mouse macrophages and dendritic cells induced by polysaccharides from a novel Cordyceps sinensis fungus UM01. J Funct Foods. 2014; 9(1): 242-253.

[33]

Zhang X, Qi C, Guo Y, Zhou W, Zhang Y. Toll-like receptor 4-related immunostimulatory polysaccharides: primary structure, activity relationships, and possible interaction models. Carbohydr Polym. 2016; 149: 186-206.

[34]

Du B, Lin C, Bian Z, Xu B. An insight into anti-inflammatory effects of fungal beta glucans. Trends Food Sci Technol. 2015; 41(1): 49-59.

[35]

Javed S, Li WM, Zeb M, et al. Anti-inflammatory activity of the wild mushroom, Echinodontiumtinctorium, in RAW264.7 macrophage cells and mouse microcirculation. Molecules. 2019; 24:3509.

[36]

Lee JS, Cho JY, Hong EK. Study on macrophage activation and structural characteristics of purified polysaccharides from the liquid culture broth of Hericium erinaceus. Carbohydr Polymers. 2009; 78(1): 162-168.

[37]

Lee JS. Study of macrophage activation and structural characteristics of purified polysaccharides from the fruiting body of Hericium erinaceus. J Microbiol Biotechnol. 2009; 19(9): 951-959.

[38]

Liu J, Wu C, Li X, Yan Q, Reaney MJT, Jiang Z. Xylose rich heteroglycan from flaxseed gum mediates the immunostimulatory effects on macrophages via TLR2 activation. Carbohydr Polym. 2019; 213: 59-69.

[39]

Biao Y, Jiannan H, Yaolan C, et al. Identification and characterization of antioxidant and immune-stimulatory polysaccharides in flaxseed hull. Food Chem. 2020; 315:126266.

[40]

Lee SH, Ko CI, Jee Y, et al. Anti-inflammatory effect of fucoidan extracted from Ecklonia cava in zebrafish model. Carbohydr Polym. 2013; 92(1): 84-89.

[41]

Sanjeewa KKA, Fernando IPS, Kim SY, et al. In vitro and in vivo anti-inflammatory activities of high molecular weight sulfated polysaccharide; containing fucose separated from Sargassum horneri: short communication. Int J Biol Macromol. 2018; 107(Pt A): 803-807.

[42]

Wang X-Y, Zhang D, Yin J-Y, Nie SP, Xie MY. Recent developments in Hericiumerinaceus polysaccharides: extraction, purification, structural characteristics and biological activities. Crit Rev Food Sci Nutr. 2019; 59(suppl 1): S96-S115.

[43]

Xiong Q, Hao H, He L, et al. Anti-inflammatory and anti-angiogenic activities of a purified polysaccharide from flesh of Cipangopaludinachinensis. Carbohydr Polym. 2017; 176: 152-159.

[44]

Zeng Y-J, Yang H-R, Wu X-L, et al. Structure and immunomodulatory activity of polysaccharides from Fusarium solani DO7 by solid-state fermentation. Int J Biiol Macromol. 2019; 137: 568-575.

[45]

Tang S, Wang T, Huang C, Lai C, Fan Y, Yong Q. Arabinogalactans from Larix principis-rupprechtii: an investigation into the structure-function contribution of side-chain structures. Carbohydr Polymers. 2020; 227:115354.

[46]

Han Y, Ouyang K, Li J, et al. Sulfated modification, characterization, immunomodulatory activities and mechanism of the polysaccharides from Cyclocaryapaliurus on dendritic cells. Int J Biiol Macromol. 2020; 159: 108-116.

[47]

Kidgell JT, Glasson CRK, Magnusson M, et al. The molecular weight of ulvan affects the in vitro inflammatory response of a murine macrophage. Int J Biiol Macromol. 2020; 150: 839-848.

[48]

Liu X, Chen X, Xie L, Xie J, Shen M. Sulfated Chinese yam polysaccharide enhances the immunomodulatory activity of RAW 264.7 cells via the TLR4-MAPK/NF-κB signaling pathway. Food Funct. 2022; 13: 1316-1326.

[49]

Jing Y, Liu D, Zhang Y, et al. Structural characterization and immunomodulatory activity of a polysaccharide from Salvia miltiorrhiza and its sulfated derivative. J Funct Foods. 2024; 121:106452.

[50]

Xu X, Chang Y, Xue C, Wang J, Shen J. Gastric protective activities of sea cucumber fucoidans with different molecular weight and chain conformations: a structure activity relationship investigation. J Agricult Food Chem. 2018; 66(32): 8615-8622.

[51]

Dunjic BS, Svensson I, Axelson J, et al. Green banana protection of gastric mucosa against experimentally induced injuries in rats. A multicomponent mechanism? Scand J Gastroenterol. 1993; 28(10): 894-898.

[52]

Palleschi A, Bocchinfuso G, Coviello T, Alhaique F. Molecular dynamics investigations of the polysaccharide scleroglucan: first study on the triple helix structure. Carbohydr Res. 2005; 340(13): 2154-2162.

[53]

Meng Y, Lyu F, Xu X, Zhang L. Recent advances in chain conformation and bioactivities of triple-helix polysaccharides. Biomacromolecules. 2020; 21(5): 1653-1677.

[54]

Mueller A, Raptis J, Rice PJ, et al. The influence of glucan polymer structure and solution conformation on binding to (1->3)- -D-glucan receptors in a human monocyte-like cell line. Glycobiology. 2000; 10(4): 339-346.

[55]

Olatunji O. Classification of natural polymers. Natural Polymers. Springer International Publishing; 2016: 1-17.

[56]

Alvarez F. The effect of chitin size, shape, source and purification method on immune recognition. Molecules. 2014; 19(4): 4433-4451.

[57]

Elieh Ali Komi D, Sharma L, Dela Cruz CS. Chitin and its effects on inflammatory and immune responses. Clin Rev Allergy Immunol. 2018; 54(2): 213-223.

[58]

Lee CG, Da Silva CA, Dela Cruz CS, et al. Role of chitin and chitinase/chitinase-like proteins in inflammation, tissue remodeling, and injury. Annu Rev Physiol. 2011; 73: 479-501.

[59]

Da Silva CA, Hartl D, Liu W, Lee CG, Elias JA. TLR-2 and IL-17A in chitin-induced macrophage activation and acute inflammation. J Immunol. 2008; 181(6): 4279-4286.

[60]

Van Dyken SJ, Mohapatra A, Nussbaum JC, et al. Chitin activates parallel immune modules that direct distinct inflammatory responses via innate lymphoid type 2 and γδ T cells. Immunity. 2014; 40(3): 414-424.

[61]

Reese TA, Liang H-E, Tager AM, et al. Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature. 2007; 447: 92-96.

[62]

Cuesta A, Esteban , Meseguer J. In vitro effect of chitin particles on the innate cellular immune system of gilthead seabream (Sparus aurata L.). Fish Shellfish Immunol. 2003; 15(1): 1-11.

[63]

Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer's disease. J Cell Biol. 2018; 217(2): 459-472.

[64]

Turano E, Busetto G, Marconi S, et al. Neurotoxicity and synaptic plasticity impairment of N-acetylglucosamine polymers: implications for Alzheimer's disease. Neurobiol Aging. 2015; 36(5): 1780-1791.

[65]

Russo C, Valle MS, Casabona A, Malaguarnera L. Chitinase signature in the plasticity of neurodegenerative diseases. Int J Mol Sci. 2023; 24(7):6301.

[66]

Rinaudo M. Chitin and chitosan: properties and applications. Prog Polym Sci. 2006; 31(7): 603-632.

[67]

Sarvaiya J, Agrawal YK. Chitosan as a suitable nanocarrier material for anti-Alzheimer drugdelivery. Int J Biol Macromol. 2015; 72: 454-465.

[68]

Chang S-H, Lin Y-Y, Wu G-J, Huang CH, Tsai GJ. Effect of chitosan molecular weight on anti-inflammatory activity in the RAW 264.7 macrophage model. Int J Biiol Macromol. 2019; 131: 167-175.

[69]

Zhang P, Liu W, Peng Y, Han B, Yang Y. Toll like receptor 4 (TLR4) mediates the stimulating activities of chitosan oligosaccharide on macrophages. Int Immunopharmacol. 2014; 23(1): 254-261.

[70]

Chakrabarti A, Talukdar D, Pal A, Ray M. Immunomodulation of macrophages by methylglyoxal conjugated with chitosan nanoparticles against Sarcoma-180 tumor in mice. Cell Immunol. 2014; 287(1): 27-35.

[71]

Feng X, Lu X, Huang D, et al. 3D porous chitosan scaffolds suit survival and neural differentiation of dental pulp stem cells. Cell Mol Neurobiol. 2014; 34(6): 859-870.

[72]

Azuma K, Osaki T, Minami S, Okamoto Y. Anticancer and anti-inflammatory properties of chitin and chitosan oligosaccharides. J Funct Biomater. 2015; 6(1): 33-49.

[73]

Guan G, Azad MAK, Lin Y, et al. Biological effects and applications of chitosan and chito-oligosaccharides. Front Physiol. 2019; 10:516.

[74]

Kim M-S, Sung M-J, Seo S-B, Yoo SJ, Lim WK, Kim HM. Water-soluble chitosan inhibits the production of pro-inflammatory cytokine in human astrocytoma cells activated by amyloid β peptide and interleukin-1β. Neurosci Lett. 2002; 321(1-2): 105-109.

[75]

Vasvani S, Kulkarni P, Rawtani D. Hyaluronic acid: a review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int J Biol Macromol. 2020; 151: 1012-1029.

[76]

Litwiniuk M, Krejner A, Speyrer MS, Gauto AR, Grzela T. Hyaluronic acid in inflammation and tissue regeneration. Wounds: Compendium Clin Res Pract. 2016; 28(3): 78-88.

[77]

Ruppert SM, Hawn TR, Arrigoni A, Wight TN, Bollyky PL. Tissue integrity signals communicated by high-molecular weight hyaluronan and the resolution of inflammation. Immunol Res. 2014; 58: 186-192.

[78]

Lee BM, Park SJ, Noh I, Kim CH. The effects of the molecular weights ofhyaluronic acid on the immune responses. Biomater Res. 2021; 25: 27.

[79]

Austin JW, Gilchrist C, Fehlings MG. High molecular weight hyaluronan reduces lipopolysaccharide mediated microglial activation. J Neurochem. 2012; 122(2): 344-355.

[80]

Remminghorst U, Rehm BHA. Bacterial alginates: from biosynthesis to applications. Biotechnol Lett. 2006; 28(21): 1701-1712.

[81]

Yang D, Jones KS. Effect of alginate on innate immune activation of macrophages. J Biomed Mater Res A. 2009; 90(2): 411-418.

[82]

Ge F, Zhu L, Yang L, et al. The soluble and particulate form of alginates positively regulate immune response. Iran J Immunol: IJI. 2018; 15(3): 228-238.

[83]

Iwamoto Y, Xu X, Tamura T, ODA T, MURAMATSU T. Enzymatically depolymerized alginate oligomers that cause cytotoxic cytokine production in human mononuclear cells. Biosci Biotechnol Biochem. 2003; 67(2): 258-263.

[84]

Yamamoto Y, Kurachi M, Yamaguchi K, ODA T. Induction of multiple cytokine secretion from RAW264.7 cells by alginate oligosaccharides. Biosci Biotechnol Biochem. 2007; 71(1): 238-241.

[85]

Eftekharzadeh B, Khodagholi F, Abdi A, Maghsoudi N. Alginate protects NT2 neurons against H2O2-induced neurotoxicity. Carbohydr Polym. 2010; 79(4): 1063-1072.

[86]

Zhou R, Shi X-Y, Bi D-C, Fang WS, Wei GB, Xu X. Alginate-derived oligosaccharide inhibits neuroinflammation and promotes microglial phagocytosis of β-amyloid. Mar Drugs. 2015; 13: 5828-5846.

[87]

Campo VL, Kawano DF, Silva DB, Carvalho I. Carrageenans: biological properties, chemical modifications and structural analysis-a review. Carbohydr Polym. 2009; 77(2): 167-180.

[88]

Thangaraj P. Pharmacological assays of plant-based natural products. In: Rainsford KD, ed. Progress in Drug Research. 71. Switzerland, Springer International Publishing; 2016: 103-111.

[89]

Lahrsen E, Schoenfeld A-K, Alban S. Degradation of eight sulfated polysaccharides extracted from red and brown algae and its impact on structure and pharmacological activities. ACS Biomater Sci Eng. 2019; 5(3): 1200-1214.

[90]

Stephanie B, Eric D, Sophie FM, Christian B, Yu G. Carrageenan from Solieriachordalis (Gigartinales): structural analysis and immunological activities of the low molecular weight fractions. Carbohydr Polym. 2010; 81(2): 448-460.

[91]

Sun H, Xu L, Wang K, et al. κ-Carrageenan oligosaccharides protect nerves by regulating microglial autophagy in Alzheimer's disease. ACS Chem Neurosci. 2023; 14(18): 3540-3550.

[92]

Yao Z, Xu L, Jin L, et al. κ-Carrageenan oligosaccharides inhibit the inflammation of lipopolysaccharide-activated microglia via TLR4/NF-κB and p38/JNK MAPKs pathways. Neurochem Res. 2022; 47(2): 295-304.

[93]

Li B, Lu F, Wei X, Zhao R. Fucoidan: structure and bioactivity. Molecules. 2008; 13(8): 1671-1695.

[94]

Ale MT, Mikkelsen JD, Meyer AS. Important determinants for fucoidan bioactivity: a critical review of structure-function relations and extraction methods for fucose-containing sulfated polysaccharides from brown seaweeds. Mar Drugs. 2011; 9(10): 2106-2130.

[95]

Wang Y, Xing M, Cao Q, Ji A, Liang H, Song S. Biological activities of fucoidan and the factors mediating its therapeutic effects: a review of recent studies. Mar Drugs. 2019; 17(3):183.

[96]

Apostolova E, Lukova P, Baldzhieva A, et al. Immunomodulatory and anti-inflammatory effects of fucoidan: a review. Polymers. 2020; 12(10):2338.

[97]

Jeong J-W, Hwang SJ, Han MH, et al. Fucoidan inhibits lipopolysaccharide-induced inflammatory responses in RAW 264.7 macrophages and zebrafish larvae. Mol Cell Toxicol. 2017; 13: 405-417.

[98]

Sanjeewa KKA, Fernando IPS, Kim E-A, Ahn G, Jee Y, Jeon YJ. Anti-inflammatory activity of a sulfated polysaccharide isolated from an enzymatic digest of brown seaweed Sargassum horneri in RAW 264.7 cells. Nutr Res Pract. 2017; 11(1): 3-10.

[99]

Ni L, Wang L, Fu X, et al. In vitro and in vivo anti-inflammatory activities of a fucose-rich fucoidan isolated from Saccharina japonica. Int J Biol Macromol. 2020; 156: 717-729.

[100]

Cui YQ, Jia YJ, Zhang T, Zhang QB, Wang XM. Fucoidan protects against lipopolysaccharide-induced rat neuronal damage and inhibits the production of proinflammatory mediators in primary microglia. CNS Neurosci Ther. 2012; 18(10): 827-833.

[101]

Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010; 7(4): 354-365.

[102]

Do H, Pyo S, Sohn E-H. Suppression of iNOS expression by fucoidan is mediated by regulation of p38 MAPK, JAK/STAT, AP-1 and IRF-1, and depends on up-regulation of scavenger receptor B1 expression in TNF-alpha- and IFN-gamma-stimulated C6 glioma cells. J Nutr Biochem. 2010; 21(8): 671-679.

[103]

Choi Y-S, Eom S-Y, Kim I-S, et al. Fucoidan extracted from hijiki protects brain microvesselendothelial cells against diesel exhaust particle exposure-induced disruption. J Med Food. 2016; 19(5): 466-471.

[104]

Borazjani NJ, Tabarsa M, You S, Rezaei M. Purification, molecular properties, structural characterization, and immunomodulatory activities of water soluble polysaccharides from Sargassum angustifolium. Int J Biol Macromol. 2018; 109: 793-802.

[105]

Hadavi D, Poot AA. Biomaterials for the treatment of Alzheimer's disease. Front Bioeng Biotechnol. 2016; 4:49.

[106]

Tosi G, Duskey JT, Kreuter J. Nanoparticles as carriers for drug delivery of macromolecules across the blood-brain barrier. Expert Opin Drug Delivery. 2020; 17(1): 23-32.

[107]

Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview. Neurobiol Dis. 2004; 16(1): 1-13.

[108]

Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010; 37(1): 13-25.

[109]

Saeedi M, Eslamifar M, Khezri K, Dizaj SM. Applications of nanotechnology in drug delivery to the central nervoussystem. Biomed Pharmacother Biomed Pharmacother. 2019; 111: 666-675.

[110]

Duan L, Li X, Ji R, et al. Nanoparticle-based drug delivery systems: an inspiring therapeutic strategy for neurodegenerative diseases. Polymers. 2023; 15(9):2196.

[111]

Chakrabarty P, Jansen-West K, Beccard A, et al. Massive gliosis induced by interleukin-6 suppresses Aβ deposition in vivo: evidence against inflammation as a driving force for amyloid deposition. FASEB J. 2010; 24(2): 548-559.

[112]

Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther. 2023; 8(1): 359.

[113]

Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther. 2023; 8(1): 267.

[114]

Pardridge WM. Treatment of Alzheimer's disease and blood-brain barrier drug delivery. Pharmaceuticals. 2020; 13(11):394.

[115]

Pandit R, Chen L, Götz J. The blood-brain barrier: physiology and strategies for drug delivery. Adv Drug Deliv Rev. 2020; 165-166: 1-14.

[116]

Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L. Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Controlled Release. 2016; 235(10): 34-47.

[117]

Ibrahim RM, Teaima M, El-Nabarawi M, Badawi NM. Intranasal delivery of chitosan-based nanoparticles as an innovative way for management of neurodegenerative disorders: A comprehensive review of advanced strategies for CNS targeting. J Drug Delivery Sci Technol. 2024; 99(2):105885.

[118]

Yu S, Xu X, Feng J, Liu M, Hu K. Chitosan and chitosan coating nanoparticles for the treatment of brain disease. Int J Pharm. 2019; 560: 282-293.

[119]

Elnaggar YSR, Etman SM, Abdelmonsif DA, Abdallah OY. Intranasal piperine-loaded chitosan nanoparticles as brain-targeted therapy in Alzheimer's disease: optimization, biological efficacy, and potential toxicity. J Pharm Sci. 2015; 104(10): 3544-3556.

[120]

Samaridou E, Walgrave H, Salta E, et al. Nose-to-brain delivery of enveloped RNA - cell permeating peptide nanocomplexes for the treatment of neurodegenerative diseases. Biomaterials. 2020; 230:119657.

[121]

Stucky EC, Schloss RS, Yarmush ML, Shreiber DI. Alginate micro-encapsulation of mesenchymal stromal cells enhances modulation of the neuro-inflammatory response. Cytotherapy. 2015; 17(10): 1353-1364.

[122]

Batista P, Cunha SA, Ribeiro T, et al. Fucoidans: exploring its neuroprotective mechanisms and therapeutic applications in brain disorders. Trends Food Sci Technol. 2024; 143:104300.

[123]

Yang J, Zhao H, Qu S. Therapeutic potential of fucoidan in central nervous system disorders: a systematic review. Int J Biiol Macromol. 2024; 277(3):134397.

[124]

Mensah EO, Bulya ET, Minin AS, Mironov MA. Submicron polymer particles loaded with piperine: preparation from fucoidan and evaluation of morphology, release profile, and antioxidant activity. Food Hydrocolloids. 2023; 145:109147.

[125]

Sun S, Zhang X, Li J, et al. Preparation and evaluation of ovalbumin-fucoidan nanoparticles for nicotinamide mononucleotide encapsulation with enhanced stability and anti-aging activity. Food Chem. 2023; 418:135982.

[126]

Arena G, Sharma K, Agyeah G, Krüger R, Grünewald A, Fitzgerald JC. Neurodegeneration and neuroinflammation in Parkinson's disease: a self-sustained loop. Curr Neurol Neurosci Rep. 2022; 22(8): 427-440.

[127]

Alotaibi BS, Abdel-Rahman mohamed A, Abd-Elhakim YM, et al. Exploring the link between pyrethroids exposure and dopaminergic degeneration through morphometric, immunofluorescence, and in-silico approaches: the therapeutic role of chitosan-encapsulated curcumin nanoparticles. Front Pharmacol. 2024; 15:1388784.

[128]

Clementino AR, Marchi C, Pozzoli M, Bernini F, Zimetti F, Sonvico F. Anti-inflammatory properties of statin-loaded biodegradable lecithin/chitosan nanoparticles: a step toward nose-to-brain treatment of neurodegenerative diseases. Front Pharmacol. 2021; 12:716380.

[129]

Sridhar V, Gaud R, Bajaj A, Wairkar S. Pharmacokinetics and pharmacodynamics of intranasally administered selegiline nanoparticles with improved brain delivery in Parkinson's disease. Nanomed: Nanotechnol Biol Med. 2018; 14(8): 2609-2618.

[130]

Raj R, Wairkar S, Sridhar V, Gaud R. Pramipexole dihydrochloride loaded chitosan nanoparticles for nose to brain delivery: development, characterization and in vivo anti-Parkinson activity. Int J Biol Macromol. 2018; 109: 27-35.

[131]

Zohri M, Arefian E, Akbari Javar H, et al. Potential of chitosan/alginate nanoparticles as a non-viral vector for gene delivery: formulation and optimization using D-optimal design. Mater Sci Eng: C. 2021; 128:112262.

[132]

Chen Y, Zhang B, Yu L, et al. A novel nanoparticle system targeting damaged mitochondria for the treatment of Parkinson's disease. Biomater Adv. 2022; 138:212876.

[133]

Grishko V, Xu M, Ho R, et al. Effects of hyaluronic acid on mitochondrial function and mitochondria-driven apoptosis following oxidative stress in human chondrocytes. J Biol Chem. 2009; 284(14): 9132-9139.

[134]

Zhao H, Tanaka T, Mitlitski V, Heeter J, Balazs EA, Darzynkiewicz Z. Protective effect of hyaluronate on oxidative DNA damage in WI-38 and A549 cells. Int J Oncol. 2008; 32(6): 1159-1167.

[135]

Koyano F, Yamano K, Kosako H, et al. Parkin-mediated ubiquitylation redistributes MITOL/March5 from mitochondria to peroxisomes. EMBO Rep. 2019; 20(12):e47728.

[136]

Han M, Yi B, Song R, et al. Fucoidan-derived carbon dots as nanopenetrants of blood-brain barrier for Parkinson's disease treatment. J Colloid Interface Sci. 2025; 680(Pt A): 516-527.

[137]

Dhaiban S, Al-Ani M, Elemam NM, Al-Aawad MH, Al-Rawi Z, Maghazachi AA. Role of peripheral immune cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Sci. 2021; 3(1):12.

[138]

Charabati M, Wheeler MA, Weiner HL, Quintana FJ. Multiple sclerosis: neuroimmune crosstalk and therapeutic targeting. Cell. 2023; 186(7): 1309-1327.

[139]

Adamu A, Li S, Gao F, Xue G. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets. Front Aging Neurosci. 2024; 16:1347987.

[140]

Bianco A, Antonacci Y, Liguori M. Sex and gender differences in neurodegenerative diseases: challenges for therapeutic opportunities. Int J Mol Sci. 2023; 24(7):6354.

[141]

Pei W, Wan X, Shahzad KA, et al. Direct modulation of myelin-autoreactive CD4+ and CD8+ T cells in EAE mice by a tolerogenic nanoparticle co-carrying myelin peptide-loaded major histocompatibility complexes, CD47 and multiple regulatory molecules. Int J Nanomed. 2018; 13: 3731-3750.

[142]

Zhao X, Sun L, Wang J, et al. Nose to brain delivery of Astragaloside IV by β-Asarone modified chitosan nanoparticles for multiple sclerosis therapy. Int J Pharm. 2023; 644:123351.

[143]

Li M, Fan YN, Chen ZY, et al. Optimized nanoparticle-mediated delivery of CRISPR-Cas9 system for B cell intervention. Nano Res. 2018; 11(12): 6270-6282.

[144]

Kong F, Liu G, Zhou S, Guo J, Chen S, Wang Z. Superior transfection efficiency of phagocytic astrocytes by large chitosan/DNA nanoparticles. Int J Biol Macromol. 2017; 105(Pt 2): 1473-1481.

RIGHTS & PERMISSIONS

2025 The Author(s). Ibrain published by Affiliated Hospital of Zunyi Medical University (AHZMU) and Wiley-VCH GmbH.

AI Summary AI Mindmap
PDF

54

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/