Targeting the pathogenesis and boosting the therapeutic efficacy of Parkinson's disease by advanced nanoparticles
Hanghang Liu
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang, China
Contribution: Conceptualization (supporting), Investigation (lead), Writing - original draft (lead)
Search for more papers by this authorMenglong Hua
Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang, China
Contribution: Investigation (supporting)
Search for more papers by this authorQing Zheng
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Contribution: Investigation (supporting)
Search for more papers by this authorYifan Gao
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Contribution: Investigation (supporting)
Search for more papers by this authorCorresponding Author
Zhen Li
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Correspondence Zhen Li, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, 215123 Suzhou, China.
Email: [email protected]
Contribution: Conceptualization (lead), Funding acquisition (lead), Writing - review & editing (lead)
Search for more papers by this authorHanghang Liu
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang, China
Contribution: Conceptualization (supporting), Investigation (lead), Writing - original draft (lead)
Search for more papers by this authorMenglong Hua
Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang, China
Contribution: Investigation (supporting)
Search for more papers by this authorQing Zheng
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Contribution: Investigation (supporting)
Search for more papers by this authorYifan Gao
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Contribution: Investigation (supporting)
Search for more papers by this authorCorresponding Author
Zhen Li
State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou, China
Correspondence Zhen Li, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Center for Molecular Imaging and Nuclear Medicine, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, 215123 Suzhou, China.
Email: [email protected]
Contribution: Conceptualization (lead), Funding acquisition (lead), Writing - review & editing (lead)
Search for more papers by this authorAbstract
With the aging of global population, the early diagnosis and treatment of neurodegenerative diseases such as Parkinson's disease (PD) have attracted considerable attention. Despite great advances achieved during the past decades, PD as the second largest neurodegenerative disease is still incurable. In the clinical practice, PD patients are mainly treated by drugs, and supplemented with deep brain stimulation or nerve nucleus destruction. The existing drugs can only relieve the symptoms of motor disorder, and cannot stop the progression of PD. Compared with small molecular drugs, nanoparticles exhibit multiple functions in the neuroprotection and neurorepair due to their tunable physical and chemical properties, easy modification and functionalization. Herein, we first briefly review the characteristics of nanoparticles crossing the blood–brain barrier, which is a primary challenge for the treatment of PD. Then, we summarize the pathologic mechanisms of PD and comprehensively discuss the novel PD therapy based on diverse nanoparticles, including alleviating oxidative stress, scavenging α-synuclein aggregates, chelating metal ions, delivering neurotrophic factors and genes, and transplanting stem cells. This review aims to highlight the great potential of advanced nanoparticles in the therapy of PD.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest.
Open Research
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this review because no new data were created or analyzed in this manuscript.
REFERENCES
- 1Dorsey ER, Elbaz A, Nichols E, et al. Global, regional, and national burden of Parkinson's disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018; 17(11): 939-953.
- 2Vijiaratnam N, Simuni T, Bandmann O, Morris HR, Foltynie T. Progress towards therapies for disease modification in Parkinson's disease. Lancet Neurol. 2021; 20(7): 559-572.
- 3Poewe W, Seppi K, Tanner CM, et al. Parkinson disease. Nat Rev Dis Primers. 2017; 3(1):17013.
- 4Charvin D, Medori R, Hauser RA, Rascol O. Therapeutic strategies for Parkinson disease: beyond dopaminergic drugs. Nat Rev Drug Discov. 2018; 17(11): 804-822.
- 5Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson disease: a review. JAMA. 2020; 323(6): 548-560.
- 6Armstrong MJ, Okun MS. Choosing a Parkinson disease treatment. JAMA. 2020; 323(14): 1420.
- 7Elkouzi A, Vedam-Mai V, Eisinger RS, Okun MS. Emerging therapies in Parkinson disease—repurposed drugs and new approaches. Nat Rev Neurol. 2019; 15(4): 204-223.
- 8Cheng G, Liu Y, Ma R, et al. Anti-Parkinsonian therapy: strategies for crossing the blood–brain barrier and nano-biological effects of nanomaterials. Nanomicro Lett. 2022; 14(1): 105.
- 9Wang J, Li Y, Nie G. Multifunctional biomolecule nanostructures for cancer therapy. Nat Rev Mater. 2021; 6(9): 766-783.
- 10Liu P, Jiang C. Brain-targeting drug delivery systems. WIREs Nanomed Nanobiotechnol. 2022; 14(5):e1818.
- 11Mukherjee S, Madamsetty VS, Bhattacharya D, Roy Chowdhury S, Paul MK, Mukherjee A. Recent advancements of nanomedicine in neurodegenerative disorders theranostics. Adv Funct Mater. 2020; 30(35):2003054.
- 12Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021; 20(2): 101-124.
- 13Zhang P, Meng J, Li Y, et al. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation. 2021; 2(4):100174.
- 14Zhong Y, Zeng X, Zeng Y, et al. Nanomaterials-based imaging diagnosis and therapy of cardiovascular diseases. Nano Today. 2022; 45:101554.
- 15Parnetti L, Gaetani L, Eusebi P, et al. CSF and blood biomarkers for Parkinson's disease. Lancet Neurol. 2019; 18(6): 573-586.
- 16Gleichman AJ, Carmichael ST. Glia in neurodegeneration: drivers of disease or along for the ride? Neurobiol Dis. 2020; 142:104957.
- 17Liddelow SA, Guttenplan KA, Clarke LE, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017; 541(7638): 481-487.
- 18Bloem BR, Okun MS, Klein C. Parkinson's disease. Lancet. 2021; 397(10291): 2284-2303.
- 19Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha–synuclein gene identified in families with Parkinson's disease. Science. 1997; 276(5321): 2045-2047.
- 20Kalia LV, Lang AE. Parkinson's disease. Lancet. 2015; 386(9996): 896-912.
- 21Rocha EM, De Miranda B, Sanders LH. Alpha–synuclein: pathology, mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurobiol Dis. 2018; 109: 249-257.
- 22Fusco G, Chen SW, Williamson PTF, et al. Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomers. Science. 2017; 358(6369): 1440-1443.
- 23Dehay B, Bourdenx M, Gorry P, et al. Targeting α-synuclein for treatment of Parkinson's disease: mechanistic and therapeutic considerations. Lancet Neurol. 2015; 14(8): 855-866.
- 24González-Rodríguez P, Zampese E, Stout KA, et al. Disruption of mitochondrial complex I induces progressive Parkinsonism. Nature. 2021; 599(7886): 650-656.
- 25Moors T, Paciotti S, Chiasserini D, et al. Lysosomal dysfunction and α-synuclein aggregation in Parkinson's disease: diagnostic links. Mov Disord. 2016; 31(6): 791-801.
- 26Oftedal L, Maple-Grødem J, Dalen I, et al. Association of CSF glucocerebrosidase activity with the risk of incident dementia in patients with Parkinson disease. Neurology. 2023; 100(4): e388-e395.
- 27Xie YX, Naseri NN, Fels J, et al. Lysosomal exocytosis releases pathogenic α-synuclein species from neurons in synucleinopathy models. Nat Commun. 2022; 13(1): 4918.
- 28Panicker N, Kam T-I, Wang H, et al. Neuronal NLRP3 is a parkin substrate that drives neurodegeneration in Parkinson's disease. Neuron. 2022; 110(15): 2422-2437.
- 29Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017; 35(1): 441-468.
- 30Nguyen PT, Dorman LC, Pan S, et al. Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell. 2020; 182(2): 388-403.e15.
- 31Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010; 7(4): 354-365.
- 32Li G, Gong J, Lei H, Liu J, Xu XZS. Promotion of behavior and neuronal function by reactive oxygen species in C. elegans. Nat Commun. 2016; 7(1):13234.
- 33Adeluyi A, Guerin L, Fisher ML, et al. Microglia morphology and proinflammatory signaling in the nucleus accumbens during nicotine withdrawal. Sci Adv. 2019; 5(10):eaax7031.
- 34Qin Y, Qiu J, Wang P, et al. Impaired autophagy in microglia aggravates dopaminergic neurodegeneration by regulating NLRP3 inflammasome activation in experimental models of Parkinson's disease. Brain Behav Immun. 2021; 91: 324-338.
- 35Xia Y, Zhang G, Kou L, et al. Reactive microglia enhance the transmission of exosomal α-synuclein via toll-like receptor 2. Brain. 2021; 144(7): 2024-2037.
- 36Fellner L, Irschick R, Schanda K, et al. Toll-like receptor 4 is required for α-synuclein dependent activation of microglia and astroglia. GLIA. 2013; 61(3): 349-360.
- 37Haque ME, Akther M, Jakaria M, Kim I-S, Azam S, Choi D-K. Targeting the microglial NLRP3 inflammasome and its role in Parkinson's disease. Mov Disord. 2020; 35(1): 20-33.
- 38Liu H, Han Y, Wang T, et al. Targeting microglia for therapy of Parkinson's disease by using biomimetic ultrasmall nanoparticles. J Am Chem Soc. 2020; 142(52): 21730-21742.
- 39Yuan J, Liu H, Zhang H, Wang T, Zheng Q, Li Z. Controlled activation of TRPV1 channels on microglia to boost their autophagy for clearance of alpha-synuclein and enhance therapy of Parkinson's disease. Adv Mater. 2022; 34(11):2108435.
- 40Scheiblich H, Dansokho C, Mercan D, et al. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell. 2021; 184(20): 5089-5106.e21.
- 41Harms AS, Cao S, Rowse AL, et al. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J Neurosci. 2013; 33(23): 9592-9600.
- 42Lezmy J, Arancibia-Cárcamo IL, Quintela-López T, Sherman DL, Brophy PJ, Attwell D. Astrocyte Ca2+-evoked ATP release regulates myelinated axon excitability and conduction speed. Science. 2021; 374(6565):eabh2858.
- 43Lee H-G, Wheeler MA, Quintana FJ. Function and therapeutic value of astrocytes in neurological diseases. Nat Rev Drug Discov. 2022; 21(5): 339-358.
- 44Phatnani H, Maniatis T. Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect Biol. 2015; 7(6):a020628.
- 45Yun SP, Kam T-I, Panicker N, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease. Nat Med. 2018; 24(7): 931-938.
- 46Li T, Liu T, Chen X, et al. Microglia induce the transformation of A1/A2 reactive astrocytes via the CXCR7/PI3K/Akt pathway in chronic post-surgical pain. J Neuroinflamm. 2020; 17(1): 211.
- 47Clarke LE, Liddelow SA, Chakraborty C, Münch AE, Heiman M, Barres BA. Normal aging induces A1-like astrocyte reactivity. Proc Natl Acad Sci USA. 2018; 115(8): E1896-E1905.
- 48Miyazaki I, Asanuma M. Neuron-astrocyte interactions in Parkinson's disease. Cells. 2020; 9(12): 2623.
- 49Rizor A, Pajarillo E, Johnson J, Aschner M, Lee E. Astrocytic oxidative/nitrosative stress contributes to Parkinson's disease pathogenesis: the dual role of reactive astrocytes. Antioxidants. 2019; 8(8): 265.
- 50Zong X, Li Y, Liu C, et al. Theta-burst transcranial magnetic stimulation promotes stroke recovery by vascular protection and neovascularization. Theranostics. 2020; 10(26): 12090-12110.
- 51Booth HDE, Hirst WD, Wade-Martins R. The role of astrocyte dysfunction in Parkinson's disease pathogenesis. Trends Neurosci. 2017; 40(6): 358-370.
- 52Choi D-J, An J, Jou I, Park SM, Joe E-H. A Parkinson's disease gene, DJ-1, regulates anti-inflammatory roles of astrocytes through prostaglandin D2 synthase expression. Neurobiol Dis. 2019; 127: 482-491.
- 53Strobbe D, Robinson AA, Harvey K, et al. Distinct mechanisms of pathogenic DJ-1 mutations in mitochondrial quality control. Front Mol Neurosci. 2018; 11: 68.
- 54Qiao C, Yin N, Gu H-Y, et al. Atp13a2 deficiency aggravates astrocyte-mediated neuroinflammation via NLRP3 inflammasome activation. CNS Neurosci Ther. 2016; 22(6): 451-460.
- 55Liu Y, Cao L, Song Y, et al. Mitochondrial glutamine transporter SLC1A5_var, a potential target to suppress astrocyte reactivity in Parkinson's disease. Cell Death Dis. 2022; 13(11): 946.
- 56Morales I, Sanchez A, Puertas-Avendaño R, Rodriguez-Sabate C, Perez-Barreto A, Rodriguez M. Neuroglial transmitophagy and Parkinson's disease. GLIA. 2020; 68(11): 2277-2299.
- 57Hayakawa K, Esposito E, Wang X, et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature. 2016; 535(7613): 551-555.
- 58Burdett TC, Freeman MR. Astrocytes eyeball axonal mitochondria. Science. 2014; 345(6195): 385-386.
- 59Asanuma M, Miyazaki I, Diaz-Corrales FJ, et al. Neuroprotective effects of zonisamide target astrocyte. Ann Neurol. 2010; 67(2): 239-249.
- 60Wei Z-YD, Shetty AK. Treating Parkinson's disease by astrocyte reprogramming: progress and challenges. Sci Adv. 2021; 7(26):eabg3198.
- 61Lee Y-K, Uchida H, Smith H, Ito A, Sanchez T. The isolation and molecular characterization of cerebral microvessels. Nat Protoc. 2019; 14(11): 3059-3081.
- 62Ding S, Khan AI, Cai X, et al. Overcoming blood–brain barrier transport: advances in nanoparticle-based drug delivery strategies. Mater Today. 2020; 37: 112-125.
- 63Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood–brain barrier. Nat Med. 2013; 19(12): 1584-1596.
- 64Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood–brain barrier: from physiology to disease and back. Physiol Rev. 2019; 99(1): 21-78.
- 65Fan K, Jia X, Zhou M, et al. Ferritin nanocarrier traverses the blood–brain barrier and kills glioma. ACS Nano. 2018; 12(5): 4105-4115.
- 66dos Santos Rodrigues B, Oue H, Banerjee A, Kanekiyo T, Singh J. Dual functionalized liposome-mediated gene delivery across triple co-culture blood–brain barrier model and specific in vivo neuronal transfection. J Control Release. 2018; 286: 264-278.
- 67Ren F, Liu H, Zhang H, et al. Engineering NIR-IIb fluorescence of Er-based lanthanide nanoparticles for through-skull targeted imaging and imaging-guided surgery of orthotopic glioma. Nano Today. 2020; 34:100905.
- 68Xie R, Wu Z, Zeng F, et al. Retro-enantio isomer of angiopep-2 assists nanoprobes across the blood–brain barrier for targeted magnetic resonance/fluorescence imaging of glioblastoma. Signal Transduct Target Ther. 2021; 6(1): 309.
- 69Min HS, Kim HJ, Naito M, et al. Systemic brain delivery of antisense oligonucleotides across the blood–brain barrier with a glucose-coated polymeric nanocarrier. Angew Chem Int Ed. 2020; 59(21): 8173-8180.
- 70Sun R, Liu M, Xu Z, Song B, He Y, Wang H. Silicon-based nanoprobes cross the blood–brain barrier for photothermal therapy of glioblastoma. Nano Res. 2022; 15(8): 7392-7401.
- 71Wu T, Liu Y, Cao Y, Liu Z. Engineering macrophage exosome disguised biodegradable nanoplatform for enhanced sonodynamic therapy of glioblastoma. Adv Mater. 2022; 34(15):2110364.
- 72Yuan D, Zhao Y, Banks WA, et al. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials. 2017; 142: 1-12.
- 73Mistry A, Stolnik S, Illum L. Nanoparticles for direct nose-to-brain delivery of drugs. Int J Pharm. 2009; 379(1): 146-157.
- 74Zhang H, Wang T, Qiu W, et al. Monitoring the opening and recovery of the blood-brain barrier with noninvasive molecular imaging by biodegradable ultrasmall Cu2–xSe nanoparticles. Nano Lett. 2018; 18(8): 4985-4992.
- 75Meng Y, Reilly RM, Pezo RC, et al. MR-guided focused ultrasound enhances delivery of trastuzumab to Her2-positive brain metastases. Sci Transl Med. 2021; 13(615):eabj4011.
- 76Wei KC, Chu PC, Wang HYJ, et al. Focused ultrasound-induced blood–brain barrier opening to enhance temozolomide delivery for glioblastoma treatment: a preclinical study. PLoS One. 2013; 8(3):e58995.
- 77Burgess A, Huang Y, Querbes W, Sah DW, Hynynen K. Focused ultrasound for targeted delivery of siRNA and efficient knockdown of Htt expression. J Control Release. 2012; 163(2): 125-129.
- 78Lin CY, Tsai CH, Feng LY, et al. Focused ultrasound-induced blood brain–barrier opening enhanced vascular permeability for GDNF delivery in Huntington's disease mouse model. Brain Stimul. 2019; 12(5): 1143-1150.
- 79Huang Q, Deng J, Wang F, et al. Targeted gene delivery to the mouse brain by MRI-guided focused ultrasound-induced blood–brain barrier disruption. Exp Neurol. 2012; 233(1): 350-356.
- 80Zhu H, Ren F, Wang T, Jiang Z, Sun Q, Li Z. Targeted immunoimaging of tumor-associated macrophages in orthotopic glioblastoma by the NIR-IIb nanoprobes. Small. 2022; 18(30):2202201.
- 81Yun B, Gu Z, Liu Z, Han Y, Sun Q, Li Z. Reducing chemo-/radioresistance to boost the therapeutic efficacy against temozolomide-resistant glioblastoma. ACS Appl Mater Interfaces. 2022; 14(34): 38617-38630.
- 82Xu Q, Zhang H, Liu H, Han Y, Qiu W, Li Z. Inhibiting autophagy flux and DNA repair of tumor cells to boost radiotherapy of orthotopic glioblastoma. Biomaterials. 2022; 280:121287.
- 83Wang T, Zhang H, Qiu W, Han Y, Liu H, Li Z. Biomimetic nanoparticles directly remodel immunosuppressive microenvironment for boosting glioblastoma immunotherapy. Bioact Mater. 2022; 16: 418-432.
- 84Jankovic J, Tan EK. Parkinson's disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020; 91(8): 795-808.
- 85Kish SJ, Morito C, Hornykiewicz O. Glutathione peroxidase activity in Parkinson's disease brain. Neurosci Lett. 1985; 58(3): 343-346.
- 86Abraham S, Soundararajan CC, Vivekanandhan S, Behari M. Erythrocyte antioxidant enzymes in Parkinson's disease. Indian J Med Res. 2005; 121(2): 111-115.
- 87Dionísio PA, Amaral JD, Rodrigues CMP. Oxidative stress and regulated cell death in Parkinson's disease. Ageing Res Rev. 2021; 67:101263.
- 88Deas E, Cremades N, Angelova PR, et al. Alpha-synuclein oligomers interact with metal ions to induce oxidative stress and neuronal death in Parkinson's disease. Antioxid Redox Signal. 2016; 24(7): 376-391.
- 89Manoharan S, Guillemin GJ, Abiramasundari RS, Essa MM, Akbar M, Akbar MD. The Role of reactive oxygen species in the pathogenesis of Alzheimer's disease, Parkinson's disease, and Huntington's disease: a mini review. Oxid Med Cell Longev. 2016; 2016: 1-15.
- 90Cai X, Jia H, Liu Z, et al. Polyhydroxylated fullerene derivative C60(OH)24 prevents mitochondrial dysfunction and oxidative damage in an MPP+-induced cellular model of Parkinson's disease. J Neurosci Res. 2008; 86(16): 3622-3634.
- 91Ren C, Hu X, Zhou Q. Graphene oxide quantum dots reduce oxidative stress and inhibit neurotoxicity in vitro and in vivo through catalase-like activity and metabolic regulation. Adv Sci. 2018; 5(5):1700595.
10.1002/advs.201700595 Google Scholar
- 92Lei L, Tu Q, Jiao L, et al. Reactive oxygen species scavenging by hemin-based nanosheets reduces Parkinson's disease symptoms in an animal model. Chem Eng J. 2022; 432:134356.
- 93Ma X, Hao J, Wu J, Li Y, Cai X, Zheng Y. Prussian blue nanozyme as a pyroptosis inhibitor alleviates neurodegeneration. Adv Mater. 2022; 34(15):2106723.
- 94Hao C, Qu A, Xu L, et al. Chiral molecule-mediated porous CuxO nanoparticle clusters with antioxidation activity for ameliorating Parkinson's disease. J Am Chem Soc. 2019; 141(2): 1091-1099.
- 95Liu Y-Q, Mao Y, Xu E, et al. Nanozyme scavenging ROS for prevention of pathologic α-synuclein transmission in Parkinson's disease. Nano Today. 2021; 36:101027.
- 96Zhang Y, Wang Z, Li X, et al. Dietary iron oxide nanoparticles delay aging and ameliorate neurodegeneration in Drosophila. Adv Mater. 2016; 28(7): 1387-1393.
- 97Zeng F, Wu Y, Li X, et al. Custom-made ceria nanoparticles show a neuroprotective effect by modulating phenotypic polarization of the microglia. Angew Chem Int Ed. 2018; 57(20): 5808-5812.
- 98Li Y, Li Y, Wang H, Liu R. Yb3+, Er3+ codoped cerium oxide upconversion nanoparticles enhanced the enzymelike catalytic activity and antioxidative activity for Parkinson's disease treatment. ACS Appl Mater Interfaces. 2021; 13(12): 13968-13977.
- 99Liu X, Zhang H, Hao C, Kuang H, Xu C, Xu L. Chiral Se@CeO2 superparticles for ameliorating Parkinson's disease. Nanoscale. 2023; 15(9): 4367-4377.
- 100Singh N, Savanur MA, Srivastava S, D'Silva P, Mugesh G. A redox modulatory Mn3O4 nanozyme with multi-enzyme activity provides efficient cytoprotection to human cells in a Parkinson's disease model. Angew Chem Int Ed. 2017; 56(45): 14267-14271.
- 101Li L, Lu Y, Xu X, et al. Catalytic-enhanced lactoferrin-functionalized Au-Bi2Se3 nanodots for Parkinson's disease therapy via reactive oxygen attenuation and mitochondrial protection. Adv Healthc Mater. 2021; 10(13):2100316.
- 102Wang W, Zheng J, Zhou H, et al. Polydopamine-based nanocomposite as a biomimetic antioxidant with a variety of enzymatic activities for Parkinson's disease. ACS Appl Mater Interfaces. 2022; 14(29): 32901-32913.
- 103Chen Y-B, Qiao T, Wang Y-Q, Cui Y-L, Wang Q-S. Hydrogen bond-enhanced nanogel delivery system for potential intranasal therapy of Parkinson's disease. Mater Des. 2022; 219:110741.
- 104Zhang W, Chen H, Ding L, et al. Trojan horse delivery of 4,4′-dimethoxychalcone for Parkinsonian neuroprotection. Adv Sci. 2021; 8(9):2004555.
- 105Wang Q, Li T, Yang J, et al. Engineered exosomes with independent module/cascading function for therapy of Parkinson's disease by multistep targeting and multistage intervention method. Adv Mater. 2022; 34(27):2201406.
- 106Liu Y, Zhu D, Luo J, et al. NIR-II-activated Yolk–shell nanostructures as an intelligent platform for Parkinsonian therapy. ACS Appl Bio Mater. 2020; 3(10): 6876-6887.
- 107Yao J, Cheng Y, Zhou M, et al. ROS scavenging Mn3O4 nanozymes for in vivo anti-inflammation. Chem Sci. 2018; 9(11): 2927-2933. doi:10.1039/C7SC05476A
- 108Zhang N, Yan F, Liang X, et al. Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson's disease therapy. Theranostics. 2018; 8(8): 2264-2277.
- 109Liu J, Liu C, Zhang J, et al. A self-assembled α-synuclein nanoscavenger for Parkinson's disease. ACS Nano. 2020; 14(2): 1533-1549.
- 110Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol. 2010; 11(4): 301-307.
- 111Mahul-Mellier A-L, Burtscher J, Maharjan N, et al. The process of Lewy body formation, rather than simply α-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc Natl Acad Sci. 2020; 117(9): 4971-4982.
- 112Bendor JT, Logan TP, Edwards RH. The function of α-synuclein. Neuron. 2013; 79(6): 1044-1066.
- 113Martinez-Vicente M, Cuervo AM. Autophagy and neurodegeneration: when the cleaning crew goes on strike. Lancet Neurol. 2007; 6(4): 352-361.
- 114Haque ME, Akther M, Azam S, et al. Targeting α-synuclein aggregation and its role in mitochondrial dysfunction in Parkinson's disease. Br J Pharmacol. 2022; 179(1): 23-45.
- 115Chu Y, Dodiya H, Aebischer P, Olanow CW, Kordower JH. Alterations in lysosomal and proteasomal markers in Parkinson's disease: relationship to alpha-synuclein inclusions. Neurobiol Dis. 2009; 35(3): 385-398.
- 116Ryan T, Bamm VV, Stykel MG, et al. Cardiolipin exposure on the outer mitochondrial membrane modulates α-synuclein. Nat Commun. 2018; 9(1): 817.
- 117Albert K, Raymundo DP, Panhelainen A, et al. Cerebral dopamine neurotrophic factor reduces α-synuclein aggregation and propagation and alleviates behavioral alterations in vivo. Mol Ther. 2021; 29(9): 2821-2840.
- 118Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008; 132(1): 27-42.
- 119Sahoo S, Padhy AA, Kumari V, Mishra P. Role of ubiquitin–proteasome and autophagy-lysosome pathways in α-synuclein aggregate clearance. Mol Neurobiol. 2022; 59(9): 5379-5407.
- 120Li T, Feng Y, Yang R, et al. Salidroside promotes the pathological α-synuclein clearance through ubiquitin-proteasome system in SH-SY5Y cells. Front Pharmacol. 2018; 9: 377.
- 121Ebrahimi-Fakhari D, Wahlster L, McLean PJ. Protein degradation pathways in Parkinson's disease: curse or blessing. Acta Neuropathol. 2012; 124(2): 153-172.
- 122Xilouri M, Brekk OR, Landeck N, et al. Boosting chaperone-mediated autophagy in vivo mitigates α-synuclein-induced neurodegeneration. Brain. 2013; 136(7): 2130-2146.
- 123Wang L, Klionsky DJ, Shen H-M. The emerging mechanisms and functions of microautophagy. Nat Rev Mol Cell Biol. 2023; 24(3): 186-203.
- 124Schneider MM, Gautam S, Herling TW, et al. The Hsc70 disaggregation machinery removes monomer units directly from α-synuclein fibril ends. Nat Commun. 2021; 12(1): 5999.
- 125Wentink AS, Nillegoda NB, Feufel J, et al. Molecular dissection of amyloid disaggregation by human HSP70. Nature. 2020; 587(7834): 483-488.
- 126Burmann BM, Gerez JA, Matečko-Burmann I, et al. Regulation of α-synuclein by chaperones in mammalian cells. Nature. 2020; 577(7788): 127-132.
- 127Falsone SF, Kungl AJ, Rek A, Cappai R, Zangger K. The molecular chaperone Hsp90 modulates intermediate steps of amyloid assembly of the Parkinson-related protein α-synuclein. J Biol Chem. 2009; 284(45): 31190-31199.
- 128Jia C, Ma X, Liu Z, et al. Different heat shock proteins bind α-synuclein with distinct mechanisms and synergistically prevent its amyloid aggregation. Front Neurosci. 2019; 13: 1124.
- 129Banks SL, Medeiros A, Sousa R, Lafer E, Morgan J. Chaperone proteins as ameliorators of α-synuclein-induced synaptic pathologies: insights into Parkinson's disease. Neural Regen Res. 2021; 16(6): 1198-1199.
- 130Cox D, Selig E, Griffin MDW, Carver JA, Ecroyd H. Small heat-shock proteins prevent α-synuclein aggregation via transient interactions and their efficacy is affected by the rate of aggregation. J Biol Chem. 2016; 291(43): 22618-22629.
- 131Wu X, Ma F, Pan B-B, et al. Tailoring a nanochaperone to regulate α-synuclein assembly. Angew Chem Int Ed. 2022; 61(19):e202200192.
- 132Butler YR, Liu Y, Kumbhar R, et al. α-Synuclein fibril-specific nanobody reduces prion-like α-synuclein spreading in mice. Nat Commun. 2022; 13(1): 4060.
- 133Kim D, Yoo JM, Hwang H, et al. Graphene quantum dots prevent α-synucleinopathy in Parkinson's disease. Nat Nanotechnol. 2018; 13(9): 812-818.
- 134Liu H, Zheng Q, Yuan J, et al. Modulating SQSTM1/p62-dependent selective autophagy of neurons by activating Nrf2 with multifunctional nanoparticles to eliminate α-synuclein aggregates and boost therapy of Parkinson's disease. Nano Today. 2023; 49:101770.
- 135Cao K, Zhu Y, Hou Z, et al. α-Synuclein as a target for metallo-anti-neurodegenerative agents. Angew Chem Int Ed. 2023; 62(1):e202215360.
- 136Lei L, Tu Q, Zhang X, et al. Stimulus-responsive curcumin-based polydopamine nanoparticles for targeting Parkinson's disease by modulating α-synuclein aggregation and reactive oxygen species. Chem Eng J. 2023; 461:141606.
- 137Jiang H, Wang J, Rogers J, Xie J. Brain iron metabolism dysfunction in Parkinson's disease. Mol Neurobiol. 2017; 54(4): 3078-3101.
- 138Mahoney-Sánchez L, Bouchaoui H, Ayton S, Devos D, Duce JA, Devedjian J-C. Ferroptosis and its potential role in the physiopathology of Parkinson's disease. Prog Neurobiol. 2021; 196:101890.
- 139Bi M, Du X, Jiao Q, Liu Z, Jiang H. α-Synuclein regulates iron homeostasis via preventing Parkin-mediated DMT1 ubiquitylation in Parkinson's disease models. ACS Chem Neurosci. 2020; 11(11): 1682-1691.
- 140Zeng X, An H, Yu F, et al. Benefits of iron chelators in the treatment of Parkinson's disease. Neurochem Res. 2021; 46(5): 1239-1251.
- 141Masaldan S, Bush AI, Devos D, Rolland AS, Moreau C. Striking while the iron is hot: iron metabolism and ferroptosis in neurodegeneration. Free Radic Biol Med. 2019; 133: 221-233.
- 142Devos D, Moreau C, Devedjian JC, et al. Targeting chelatable iron as a therapeutic modality in Parkinson's disease. Antioxid Redox Signal. 2014; 21(2): 195-210.
- 143Kaur D, Yantiri F, Rajagopalan S, et al. Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo. Neuron. 2003; 37(6): 899-909.
- 144Wang N, Jin X, Guo D, Tong G, Zhu X. Iron chelation nanoparticles with delayed saturation as an effective therapy for Parkinson disease. Biomacromolecules. 2017; 18(2): 461-474.
- 145You L, Wang J, Liu T, et al. Targeted brain delivery of rabies virus glycoprotein 29-modified deferoxamine-loaded nanoparticles reverses functional deficits in Parkinsonian mice. ACS Nano. 2018; 12(5): 4123-4139.
- 146Xiao G, Zhao M, Liu Z, Du F, Zhou B. Zinc antagonizes iron-regulation of tyrosine hydroxylase activity and dopamine production in Drosophila melanogaster. BMC Biol. 2021; 19(1): 236.
- 147Teil M, Doudnikoff E, Thiolat M-L, Bohic S, Bezard E, Dehay B. The zinc ionophore clioquinol reduces Parkinson's disease patient-derived brain extracts-induced neurodegeneration. Mol Neurobiol. 2022; 59(10): 6245-6259.
- 148Budinger D, Barral S, Soo AKS, Kurian MA. The role of manganese dysregulation in neurological disease: emerging evidence. Lancet Neurol. 2021; 20(11): 956-968.
- 149Bouabid S, Tinakoua A, Lakhdar-Ghazal N, Benazzouz A. Manganese neurotoxicity: behavioral disorders associated with dysfunctions in the basal ganglia and neurochemical transmission. J Neurochem. 2016; 136(4): 677-691.
- 150Lin M, Colon-Perez LM, Sambo DO, et al. Mechanism of manganese dysregulation of dopamine neuronal activity. J Neurosci. 2020; 40(30): 5871-5891.
- 151Xin Y, Gao H, Wang J, et al. Manganese transporter Slc39a14 deficiency revealed its key role in maintaining manganese homeostasis in mice. Cell Discov. 2017; 3(1):17025.
- 152Sampaio T, Savall A, Gutierrez MZ, Pinton S. Neurotrophic factors in Alzheimer's and Parkinson's diseases: implications for pathogenesis and therapy. Neural Regen Res. 2017; 12(4): 549-557.
- 153Lindholm P, Saarma M. Cerebral dopamine neurotrophic factor protects and repairs dopamine neurons by novel mechanism. Mol Psychiatry. 2022; 27(3): 1310-1321.
- 154Castrén E, Antila H. Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry. 2017; 22(8): 1085-1095.
- 155Bondarenko O, Saarma M. Neurotrophic factors in Parkinson's disease: clinical trials, open challenges and nanoparticle-mediated delivery to the brain. Front Cell Neurosci. 2021; 15:682597.
- 156Thorne RG, Frey WH. Delivery of neurotrophic factors to the central nervous system. Clin Pharmacokinet. 2001; 40(12): 907-946.
- 157Huttunen HJ, Saarma M. CDNF protein therapy in Parkinson's disease. Cell Transplant. 2019; 28(4): 349-366.
- 158Tan J, Wang Y, Yip X, Glynn F, Shepherd RK, Caruso F. Nanoporous peptide particles for encapsulating and releasing neurotrophic factors in an animal model of neurodegeneration. Adv Mater. 2012; 24(25): 3362-3366.
- 159Xu D, Wu D, Qin M, et al. Efficient delivery of nerve growth factors to the central nervous system for neural regeneration. Adv Mater. 2019; 31(33):1900727.
- 160Wu X, Wang L, Wang K, et al. SC79 promotes efficient entry of GDNF liposomes into brain parenchyma to repair dopamine neurons through reversible regulation of tight junction proteins. Nano Res. 2023; 16(2): 2695-2705.
- 161Garbayo E, Ansorena E, Lana H, et al. Brain delivery of microencapsulated GDNF induces functional and structural recovery in Parkinsonian monkeys. Biomaterials. 2016; 110: 11-23.
- 162Li Y, Li Y, Ji W, et al. Positively charged polyprodrug amphiphiles with enhanced drug loading and reactive oxygen species-responsive release ability for traceable synergistic therapy. J Am Chem Soc. 2018; 140(11): 4164-4171.
- 163Corti O, Lesage S, Brice A. What genetics tells us about the causes and mechanisms of Parkinson's disease. Physiol Rev. 2011; 91(4): 1161-1218.
- 164Fan CH, Lin CY, Liu HL, Yeh CK. Ultrasound targeted CNS gene delivery for Parkinson's disease treatment. J Control Release. 2017; 261: 246-262.
- 165Cortes H, Alcala-Alcala S, Avalos-Fuentes A, et al. Nanotechnology as potential tool for siRNA delivery in Parkinson's disease. Curr Drug Targets. 2017; 18(16): 1866-1879.
- 166Helmschrodt C, Höbel S, Schöniger S, et al. Polyethylenimine nanoparticle-mediated siRNA delivery to reduce α-synuclein expression in a model of Parkinson's disease. Mol Ther Nucleic Acids. 2017; 9: 57-68.
- 167Liu L, Li Y, Peng H, et al. Targeted exosome coating gene-chem nanocomplex as “nanoscavenger” for clearing α-synuclein and immune activation of Parkinson's disease. Sci Adv. 2020; 6(50):eaba3967.
- 168Meijer HA, Kong YW, Lu WT, et al. Translational repression and eIF4A2 activity are critical for microRNA-mediated gene regulation. Science. 2013; 340(6128): 82-85.
- 169Angelopoulou E, Paudel YN, Piperi C. miR-124 and Parkinson's disease: a biomarker with therapeutic potential. Pharmacol Res. 2019; 150:104515.
- 170Esteves M, Abreu R, Fernandes H, et al. MicroRNA-124-3p-enriched small extracellular vesicles as a therapeutic approach for Parkinson's disease. Mol Ther. 2022; 30(10): 3176-3192.
- 171Saraiva C, Paiva J, Santos T, Ferreira L, Bernardino L. MicroRNA-124 loaded nanoparticles enhance brain repair in Parkinson's disease. J Control Release. 2016; 235: 291-305.
- 172Gan L, Li Z, Lv Q, Huang W. Rabies virus glycoprotein (RVG29)-linked microRNA-124-loaded polymeric nanoparticles inhibit neuroinflammation in a Parkinson's disease model. Int J Pharm. 2019; 567:118449.
- 173Wu Y, Wang W, Qiu X, et al. A STIR nucleic acid drug delivery system for stirring phenotypic switch of microglia in Parkinson's disease treatments. Nano Res. 2023; 16: 7216-7226. doi:10.1007/s12274-022-5353-5
- 174Peng H, Li Y, Ji W, et al. Intranasal administration of self-oriented nanocarriers based on therapeutic exosomes for synergistic treatment of Parkinson's disease. ACS Nano. 2022; 16(1): 869-884.
- 175Lin D, Li M, Gao Y, Yin L, Guan Y. Brain-targeted gene delivery of ZnO quantum dots nanoplatform for the treatment of Parkinson disease. Chem Eng J. 2022; 429:132210.
- 176Gao Y, Cheng Y, Chen J, et al. NIR-assisted MgO-based polydopamine nanoparticles for targeted treatment of Parkinson's disease through the blood–brain barrier. Adv Healthc Mater. 2022; 11(23):2201655.
- 177Liu L, Li M, Xu M, et al. Actively targeted gold nanoparticle composites improve behavior and cognitive impairment in Parkinson's disease mice. Mater Sci Eng C Mater Biol Appl. 2020; 114:111028.
- 178Hu K, Chen X, Chen W, et al. Neuroprotective effect of gold nanoparticles composites in Parkinson's disease model. Nanomed Nanotechnol Biolo Med. 2018; 14(4): 1123-1136.
- 179Miao Y-B, Ren H-X, Zhong Q, Song F-X. Tailoring a luminescent metal–organic framework precise inclusion of Pt-Aptamer nanoparticle for noninvasive monitoring Parkinson's disease. Chem Eng J. 2022; 441:136009.
- 180Xue Y, Wang N, Zeng Z, Huang J, Xiang Z, Guan Y-Q. Neuroprotective effect of chitosan nanoparticle gene delivery system grafted with acteoside (ACT) in Parkinson's disease models. J Mater Sci Technol. 2020; 43: 197-207.
- 181Mead BP, Kim N, Miller GW, et al. Novel focused ultrasound gene therapy approach noninvasively restores dopaminergic neuron function in a rat Parkinson's disease model. Nano Lett. 2017; 17(6): 3533-3542.
- 182Lin C-Y, Lin Y-C, Huang C-Y, Wu S-R, Chen C-M, Liu H-L. Ultrasound-responsive neurotrophic factor-loaded microbubble–liposome complex: preclinical investigation for Parkinson's disease treatment. J Control Release. 2020; 321: 519-528.
- 183Xiao B, Zhang S-C, Tan E-K. Combination therapy using GDNF and cell transplant in Parkinson's disease. Mol Neurodegener. 2022; 17(1): 49.
- 184Lindvall O. Balancing expectations for success in stem cell-based clinical trials for Parkinson's disease. Cell Stem Cell. 2020; 27(4): 519-522.
- 185Vissers C, Ming G, Song H. Nanoparticle technology and stem cell therapy team up against neurodegenerative disorders. Adv Drug Deliv Rev. 2019; 148: 239-251.
- 186Carradori D, Labrak Y, Miron VE, et al. Retinoic acid-loaded NFL-lipid nanocapsules promote oligodendrogenesis in focal white matter lesion. Biomaterials. 2020; 230:119653.
- 187Barbuti PA, Barker RA, Brundin P, et al. Recent advances in the development of stem-cell-derived dopaminergic neuronal transplant therapies for Parkinson's disease. Mov Disord. 2021; 36(8): 1772-1780.
- 188Sonntag K-C, Song B, Lee N, et al. Pluripotent stem cell-based therapy for Parkinson's disease: current status and future prospects. Prog Neurobiol. 2018; 168: 1-20.
- 189Urrutia-Cabrera D, Hsiang-Chi Liou R, Lin J, et al. Combinatorial approach of binary colloidal crystals and CRISPR activation to improve induced pluripotent stem cell differentiation into neurons. ACS Appl Mater Interfaces. 2022; 14(7): 8669-8679.
- 190Fattorelli N, Martinez-Muriana A, Wolfs L, Geric I, De Strooper B, Mancuso R. Stem-cell-derived human microglia transplanted into mouse brain to study human disease. Nat Protoc. 2021; 16(2): 1013-1033.
- 191Pous L, Deshpande SS, Nath S, et al. Fibrinogen induces neural stem cell differentiation into astrocytes in the subventricular zone via BMP signaling. Nat Commun. 2020; 11(1): 630.
- 192Adil MM, Rao AT, Ramadoss GN, et al. Dopaminergic neurons transplanted using cell-instructive biomaterials alleviate Parkinsonism in rodents. Adv Funct Mater. 2018; 28(41):1804144.
- 193He F, Liu Z, Xu J, et al. Black phosphorus nanosheets suppress oxidative damage of stem cells for improved neurological recovery. Chem Eng J. 2023; 451:138737.
- 194Shi B, Zhao J, Xu Z, et al. Chiral nanoparticles force neural stem cell differentiation to alleviate Alzheimer's disease. Adv Sci. 2022; 9(29):2202475.
- 195Huang D, Cao Y, Yang X, et al. A nanoformulation-mediated multifunctional stem cell therapy with improved beta-amyloid clearance and neural regeneration for Alzheimer's disease. Adv Mater. 2021; 33(13):2006357.
- 196Shen Y, Liu F, Duan J, et al. Biomaterial cues regulated differentiation of neural stem cells into GABAergic neurons through Ca2+/c-Jun/TLX3 signaling promoted by hydroxyapatite nanorods. Nano Lett. 2021; 21(17): 7371-7378.
- 197Perets N, Betzer O, Shapira R, et al. Golden exosomes selectively target brain pathologies in neurodegenerative and neurodevelopmental disorders. Nano Lett. 2019; 19(6): 3422-3431.
- 198Deweerdt S. Parkinson's disease: 4 big questions. Nature. 2016; 538(7626): S17.