Fields RD. The Brain Learns in Unexpected Ways: Neuroscientists have discovered a set of unfamiliar cellular mechanisms for making fresh memories. Sci Am. 2020;322(3):74.
Google Scholar
Bramlett HM, Dietrich WD. Long-term consequences of traumatic brain injury: current status of potential mechanisms of injury and neurological outcomes. J Neurotrauma. 2015;32(23):1834–48.
Article
Google Scholar
Nagappan PG, Chen H, Wang DY. Neuroregeneration and plasticity: a review of the physiological mechanisms for achieving functional recovery postinjury. Mil Med Res. 2020;7(1):30.
Google Scholar
Council NR. From neurons to neighborhoods: The science of early childhood development. 2000.
Freitas RA Jr. Nanotechnology, nanomedicine and nanosurgery. Int J Surg (London, England). 2005;3(4):243–6.
Article
Google Scholar
Feynman RP. There’s plenty of room at the bottom: An invitation to enter a new field of physics. New York: CRC Press; 2018.
Book
Google Scholar
Jin C, et al. Application of nanotechnology in cancer diagnosis and therapy - a mini-review. Int J Med Sci. 2020;17(18):2964–73.
Article
Google Scholar
Patil M, Mehta DS, Guvva S. Future impact of nanotechnology on medicine and dentistry. J Indian Soc Periodontol. 2008;12(2):34–40.
Article
Google Scholar
Jain K. Role of nanotechnology in developing new therapies for diseases of the nervous system. 2006.
Rostamizadeh S, et al. Silica supported sodium hydrogen sulfate (NaHSO4/SiO2): A mild and efficient reusable catalyst for the synthesis of aryl-14-H-dibenzo [a, j] xanthenes under solvent-free conditions. Chin Chem Lett. 2008;19(10):1151–5.
Article
Google Scholar
Gwinn MR, Vallyathan V. Nanoparticles: health effects–pros and cons. Environ Health Perspect. 2006;114(12):1818–25.
Article
Google Scholar
Mahdavinia GH, et al. Fast and efficient method for the synthesis of 2-arylbenzimidazoles using MCM-41-SO3H. 2012.
Modi G, Pillay V, Choonara YE. Advances in the treatment of neurodegenerative disorders employing nanotechnology. Ann N Y Acad Sci. 2010;1184(1):154–72.
Article
Google Scholar
Silva GA. Neuroscience nanotechnology: progress, opportunities and challenges. Nat Rev Neurosci. 2006;7(1):65–74.
Article
Google Scholar
Mahdavinia GH, Amani AM, Sepehrian H. MCM-41-SO3H as a highly efficient sulfonic acid nanoreactor for the rapid and green synthesis of some novel highly substituted imidazoles under solvent-free condition. Chin J Chem. 2012;30(3):703–8.
Article
Google Scholar
Vidu R, et al. Nanostructures: a platform for brain repair and augmentation. Front Syst Neurosci. 2014;8:91–91.
Article
Google Scholar
Shah S. The nanomaterial toolkit for neuroengineering. Nano Convergence. 2016;3(1):25.
Article
Google Scholar
Jamwal D, et al. The multifaceted dimensions of potent nanostructures: a comprehensive review. Mater Chem Front. 2021;5(7):2967–95.
Article
Google Scholar
Gao W, et al. Liposome-like nanostructures for drug delivery. J Mater Chem. 2013;1:48. https://0-doi-org.brum.beds.ac.uk/10.1039/C3TB21238F.
Article
Google Scholar
Chenthamara D, et al. Therapeutic efficacy of nanoparticles and routes of administration. Biomater Res. 2019;23(1):20.
Article
Google Scholar
Pampaloni NP, et al. Advances in nano neuroscience: from nanomaterials to nanotools. Front Neurosci. 2019;12:953–953.
Article
Google Scholar
Rostamizadeh S, et al. Aqueous NaHSO4 catalyzed regioselective and versatile synthesis of 2-thiazolamines. Monatshefte für Chemie-Chemical Monthly. 2008;139(10):1241–5.
Article
Google Scholar
Rostamizadeh S, et al. Silica supported ammonium dihydrogen phosphate (NH4H2PO4/SiO2): A mild, reusable and highly efficient heterogeneous catalyst for the synthesis of 14-aryl-14-H-dibenzo [a, j] xanthenes. Chin Chem Lett. 2009;20(7):779–83.
Article
Google Scholar
Bayda S, et al. The history of nanoscience and nanotechnology: from chemical-physical applications to nanomedicine. Molecules. 2019;25:1.
Article
Google Scholar
Jeevanandam J, et al. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050–74.
Article
Google Scholar
Hardy N, et al. Neuro-SWARM3: system-on-a-nanoparticle for wireless recording of brain activity. IEEE Photonics Technol Lett. 2021;33(16):900–3.
Article
Google Scholar
Chen R, et al. Wireless magnetothermal deep brain stimulation. Science. 2015;347(6229):1477–80.
Article
Google Scholar
Taniguchi N. On the basic concept of nanotechnology. Proceeding of the ICPE, 1974.
Garcia-Etxarri A, Yuste R. Time for NanoNeuro. Nat Methods. 2021;18(11):1287–93.
Article
Google Scholar
Wood T, Nance E. Disease-directed engineering for physiology-driven treatment interventions in neurological disorders. APL Bioeng. 2019;3(4): 040901.
Article
Google Scholar
Markett S, Montag C, Reuter M. Network Neuroscience and Personality. Personal Neurosci. 2018;1: e14.
Article
Google Scholar
Kumar R, et al. Advances in nanotechnology and nanomaterials based strategies for neural tissue engineering. J Drug Deliv Sci Technol. 2020;57: 101617.
Article
Google Scholar
Chen S, et al. Electrospinning: An enabling nanotechnology platform for drug delivery and regenerative medicine. Adv Drug Deliv Rev. 2018;132:188–213.
Article
Google Scholar
Ojha S, Kumar B. A review on nanotechnology based innovations in diagnosis and treatment of multiple sclerosis. J Cell Immunother. 2018;4(2):56–64.
Article
Google Scholar
Das S, et al. Bio-inspired nano tools for neuroscience. Prog Neurobiol. 2016;142:1–22.
Article
Google Scholar
Kumar A, et al. Nanotechnology for neuroscience: promising approaches for diagnostics, therapeutics and brain activity mapping. Adv Func Mater. 2017;27(39):1700489.
Article
Google Scholar
Sharma D, Hussain CM. Smart nanomaterials in pharmaceutical analysis. Arab J Chem. 2020;13(1):3319–43.
Article
Google Scholar
Anjum S, et al. Emerging applications of nanotechnology in healthcare systems: grand challenges and perspectives. Pharmaceuticals (Basel). 2021;14(8):8.
Article
Google Scholar
Ouyang J, et al. Ultrasound mediated therapy: recent progress and challenges in nanoscience. Nano Today. 2020;35: 100949.
Article
Google Scholar
Krol S, et al. Therapeutic benefits from nanoparticles: the potential significance of nanoscience in diseases with compromise to the blood brain barrier. Chem Rev. 2013;113(3):1877–903.
Article
Google Scholar
Tindle J, Tadi P. Neuroanatomy, parasympathetic nervous system, in StatPearls. 2021, StatPearls Publishing.
Kadry H, Noorani B, Cucullo L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS. 2020;17(1):1–24.
Article
Google Scholar
Han L, Jiang C. Evolution of blood–brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharmaceutica Sinica B. 2021;11(8):2306–25.
Article
Google Scholar
Suárez LE, et al. Linking structure and function in macroscale brain networks. Trends Cogn Sci. 2020;24(4):302–15.
Article
Google Scholar
Lam E, Hemraz UD. Preparation and surface functionalization of carboxylated cellulose nanocrystals. Nanomaterials. 2021;11(7):1641.
Article
Google Scholar
Welch EC, et al. Advances in biosensors and diagnostic technologies using nanostructures and nanomaterials. Adv Func Mater. 2021;31(44):2104126.
Article
MathSciNet
Google Scholar
Baig N, Kammakakam I, Falath W. Nanomaterials: a review of synthesis methods, properties, recent progress, and challenges. Mater Adv. 2021;2(6):1821–71.
Article
Google Scholar
Mitchell MJ, et al. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discovery. 2021;20(2):101–24.
Article
Google Scholar
Yetisgin AA, et al. Therapeutic nanoparticles and their targeted delivery applications. Molecules. 2020;25:9.
Article
Google Scholar
Chakravarty M, Vora A. Nanotechnology-based antiviral therapeutics. Drug Deliv Transl Res. 2021;11(3):748–87.
Article
Google Scholar
Gilmore JL, et al. Novel nanomaterials for clinical neuroscience. J Neuroimmune Pharmacol. 2008;3(2):83–94.
Article
MathSciNet
Google Scholar
Maurer N, Fenske DB, Cullis PR. Developments in liposomal drug delivery systems. Expert Opin Biol Ther. 2001;1(6):923–47.
Article
Google Scholar
Wilson B, et al. Significant delivery of tacrine into the brain using magnetic chitosan microparticles for treating Alzheimer’s disease. J Neurosci Methods. 2009;177(2):427–33.
Article
Google Scholar
Lu CT, et al. Current approaches to enhance CNS delivery of drugs across the brain barriers. Int J Nanomedicine. 2014;9:2241–57.
Article
Google Scholar
Shao K, et al. Angiopep-2 modified PE-PEG based polymeric micelles for amphotericin B delivery targeted to the brain. J Control Release. 2010;147(1):118–26.
Article
Google Scholar
Lu YM, et al. Targeted therapy of brain ischaemia using Fas ligand antibody conjugated PEG-lipid nanoparticles. Biomaterials. 2014;35(1):530–7.
Article
Google Scholar
Huang R, et al. Solid lipid nanoparticles enhanced the neuroprotective role of curcumin against epilepsy through activation of Bcl-2 family and P38 MAPK pathways. ACS Chem Neurosci. 2020;11(13):1985–95.
Article
Google Scholar
Rakotoarisoa M, et al. Liquid crystalline lipid nanoparticles for combined delivery of curcumin, fish oil and BDNF: In vitro neuroprotective potential in a cellular model of tunicamycin-induced endoplasmic reticulum stress. Smart Mater Med. 2022;3:274–88.
Article
Google Scholar
Maier MA, et al. Biodegradable lipids enabling rapidly eliminated lipid nanoparticles for systemic delivery of RNAi therapeutics. Mol Ther. 2013;21(8):1570–8.
Article
Google Scholar
Kulkarni JA, Cullis PR, Van Der Meel R. Lipid nanoparticles enabling gene therapies: from concepts to clinical utility. Nucleic Acid Ther. 2018;28(3):146–57.
Article
Google Scholar
Sarode A, et al. Predictive high-throughput screening of PEGylated lipids in oligonucleotide-loaded lipid nanoparticles for neuronal gene silencing. Nanoscale Adv. 2022;4(9):2107–23.
Article
Google Scholar
Liao HC, et al. Dopant-free hole transporting polymers for high efficiency, environmentally stable perovskite solar cells. Adv Energy Mater. 2016;6(16):1600502.
Article
Google Scholar
Ramires Júnior OV, et al. Nanoemulsion improves the neuroprotective effects of curcumin in an experimental model of Parkinson’s Disease. Neurotox Res. 2021;39(3):787–99.
Article
Google Scholar
Mahajan HS, et al. Nanoemulsion-based intranasal drug delivery system of saquinavir mesylate for brain targeting. Drug Deliv. 2014;21(2):148–54.
Article
Google Scholar
Parikh H, Patel RJ. Nanoemulsions for intranasal delivery of riluzole to improve brain bioavailability: formulation development and pharmacokinetic studies. Curr Drug Deliv. 2016;13(7):1130–43.
Article
Google Scholar
Arora A, et al. Intranasal delivery of tetrabenazine nanoemulsion via olfactory region for better treatment of hyperkinetic movement associated with Huntington’s disease: Pharmacokinetic and brain delivery study. Chem Phys Lipid. 2020;230: 104917.
Article
Google Scholar
Azadi A, Hamidi M, Rouini MR. Methotrexate-loaded chitosan nanogels as “Trojan Horses” for drug delivery to brain: preparation and in vitro/in vivo characterization. Int J Biol Macromol. 2013;62:523–30.
Article
Google Scholar
Picone P, et al. Ionizing radiation-engineered nanogels as insulin nanocarriers for the development of a new strategy for the treatment of Alzheimer’s disease. Biomaterials. 2016;80:179–94.
Article
Google Scholar
Rodrigues SF, et al. Lipid-core nanocapsules act as a drug shuttle through the blood brain barrier and reduce glioblastoma after intravenous or oral administration. J Biomed Nanotechnol. 2016;12(5):986–1000.
Article
Google Scholar
Ghosh S, et al. Triphenyl phosphonium coated nano-quercetin for oral delivery: Neuroprotective effects in attenuating age related global moderate cerebral ischemia reperfusion injury in rats. Nanomedicine. 2017;13(8):2439–50.
Article
Google Scholar
Frozza RL, et al. Lipid-core nanocapsules improve the effects of resveratrol against Abeta-induced neuroinflammation. J Biomed Nanotechnol. 2013;9(12):2086–104.
Article
Google Scholar
Veetil AT, et al. Cell-targetable DNA nanocapsules for spatiotemporal release of caged bioactive small molecules. Nat Nanotechnol. 2017;12(12):1183–9.
Article
Google Scholar
Guerrero S, et al. Improving the brain delivery of gold nanoparticles by conjugation with an amphipathic peptide. Nanomedicine (Lond). 2010;5(6):897–913.
Article
Google Scholar
Morshed RA, et al. Cell-penetrating peptide-modified gold nanoparticles for the delivery of doxorubicin to brain metastatic breast cancer. Mol Pharm. 2016;13(6):1843–54.
Article
Google Scholar
Papastefanaki F, et al. Intraspinal delivery of polyethylene glycol-coated gold nanoparticles promotes functional recovery after spinal cord injury. Mol Ther. 2015;23(6):993–1002.
Article
Google Scholar
Cartwright AN, Nicolau DV. Nanoscale Imaging, Sensing, and Actuation for Biomedical Applications XII. in Proc. of SPIE Vol. 2015.
Corezzi S, et al. Synchrotron-based X-ray fluorescence imaging of human cells labeled with CdSe quantum dots. Anal Biochem. 2009;388(1):33–9.
Article
Google Scholar
Janjic JM, Gorantla VS. Peripheral nerve nanoimaging: monitoring treatment and regeneration. AAPS J. 2017;19(5):1304–16.
Article
Google Scholar
Liu L, et al. A new method for preparing mesenchymal stem cells and labeling with ferumoxytol for cell tracking by MRI. Sci Rep. 2016;6:26271.
Article
Google Scholar
Bulte JW. In vivo MRI cell tracking: clinical studies. AJR Am J Roentgenol. 2009;193(2):314–25.
Article
Google Scholar
Mousavi M, et al. Erythrosine adsorption from aqueous solution via decorated graphene oxide with magnetic iron oxide nano particles: kinetic and equilibrium studies. Acta Chim Slov. 2018;65(4):882–94.
Article
MathSciNet
Google Scholar
Madsen SJ, et al. Increased nanoparticle-loaded exogenous macrophage migration into the brain following PDT-induced blood–brain barrier disruption. Lasers Surg Med. 2013;45(8):524–32.
Article
Google Scholar
Walling MA, Novak JA, Shepard JR. Quantum dots for live cell and in vivo imaging. Int J Mol Sci. 2009;10(2):441–91.
Article
Google Scholar
Chan WC, Nie S. Quantum dot bioconjugates for ultrasensitive nonisotopic detection. Science. 1998;281(5385):2016–8.
Article
Google Scholar
Tokuraku K, Ikezu T. Chapter 11 - Imaging of Amyloid-β Aggregation Using a Novel Quantum dot Nanoprobe and its Advanced Applications. In: Uversky VN, Lyubchenko YL, editors. Bio-nanoimaging. Boston: Academic Press; 2014. p. 121–31.
Chapter
Google Scholar
Karatum O, et al. Electrical stimulation of neurons with quantum dots via near-infrared light. ACS Nano. 2022;16(5):8233–43.
Article
Google Scholar
Dahan M, et al. Diffusion dynamics of glycine receptors revealed by single-quantum dot tracking. Science. 2003;302(5644):442–5.
Article
Google Scholar
Kumar VB, et al. One-pot hydrothermal synthesis of elements (B, N, P)-doped fluorescent carbon dots for cell labelling, differentiation and outgrowth of neuronal cells. ChemistrySelect. 2019;4(14):4222–32.
Article
Google Scholar
Modi S, et al. Quantum dot conjugated nanobodies for multiplex imaging of protein dynamics at synapses. Nanoscale. 2018;10(21):10241–9.
Article
Google Scholar
Sun B, et al. Nerve growth factor-conjugated mesoporous silica nanoparticles promote neuron-like PC12 cell proliferation and neurite growth. J Nanosci Nanotechnol. 2016;16(3):2390–3.
Article
Google Scholar
Zhang L, et al. High MRI performance fluorescent mesoporous silica-coated magnetic nanoparticles for tracking neural progenitor cells in an ischemic mouse model. Nanoscale. 2013;5(10):4506–16.
Article
Google Scholar
Schmidt N, et al. Long-term delivery of brain-derived neurotrophic factor (BDNF) from nanoporous silica nanoparticles improves the survival of spiral ganglion neurons in vitro. PLoS ONE. 2018;13(3): e0194778.
Article
Google Scholar
Cheng SH, et al. Dynamic in vivo SPECT imaging of neural stem cells functionalized with radiolabeled nanoparticles for tracking of glioblastoma. J Nucl Med. 2016;57(2):279–84.
Article
Google Scholar
Keefer EW, et al. Carbon nanotube coating improves neuronal recordings. Nat Nanotechnol. 2008;3(7):434–9.
Article
Google Scholar
Fabbro A, Prato M, Ballerini L. Carbon nanotubes in neuroregeneration and repair. Adv Drug Deliv Rev. 2013;65(15):2034–44.
Article
Google Scholar
Shaw LM, et al. Biomarkers of neurodegeneration for diagnosis and monitoring therapeutics. Nat Rev Drug Discov. 2007;6(4):295–303.
Article
Google Scholar
Chen RJ, et al. Noncovalent functionalization of carbon nanotubes for highly specific electronic biosensors. Proc Natl Acad Sci U S A. 2003;100(9):4984–9.
Article
Google Scholar
Park KH, et al. Single-walled carbon nanotubes are a new class of ion channel blockers. J Biol Chem. 2003;278(50):50212–6.
Article
Google Scholar
Fabbro A, et al. Spinal cord explants use carbon nanotube interfaces to enhance neurite outgrowth and to fortify synaptic inputs. ACS Nano. 2012;6(3):2041–55.
Article
Google Scholar
Srinivasan M, Rajabi M, Mousa SA. Multifunctional nanomaterials and their applications in drug delivery and cancer therapy. Nanomaterials. 2015;5(4):1690–703.
Article
Google Scholar
Kim D, et al. Synthesis and biomedical applications of multifunctional nanoparticles. Adv Mater. 2018;30(49):1802309.
Article
Google Scholar
Seleci M, et al. Smart multifunctional nanoparticles in nanomedicine. BioNanoMaterials. 2016;17(1–2):33–41.
Google Scholar
Lee D-E, et al. Multifunctional nanoparticles for multimodal imaging and theragnosis. Chem Soc Rev. 2012;41(7):2656–72.
Article
Google Scholar
Cheng Q, Liu Y. Multifunctional platinum-based nanoparticles for biomedical applications. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9:2.
Article
Google Scholar
Chenthamara D, et al. Therapeutic efficacy of nanoparticles and routes of administration. Biomaterials research. 2019;23(1):1–29.
Article
Google Scholar
Ahlawat J, et al. Nanocarriers as potential drug delivery candidates for overcoming the blood–brain barrier: challenges and possibilities. ACS Omega. 2020;5(22):12583–95.
Article
Google Scholar
Hoshyar N, et al. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine. 2016;11(6):673–92.
Article
Google Scholar
Auría-Soro C, et al. Interactions of nanoparticles and biosystems: microenvironment of nanoparticles and biomolecules in nanomedicine. Nanomaterials (Basel). 2019;9:10.
Article
Google Scholar
Ridolfo R, et al. Exploring the impact of morphology on the properties of biodegradable nanoparticles and their diffusion in complex biological medium. Biomacromol. 2020;22(1):126–33.
Article
Google Scholar
Gulumian M, et al. Importance of surface topography in both biological activity and catalysis of nanomaterials: can catalysis by design guide safe by design? Int J Mol Sci. 2021;22:15.
Article
Google Scholar
Dresselhaus MS. Fifty years in studying carbon-based materials. Phys Scr. 2012;2012(T146): 014002.
Article
Google Scholar
Chae SH, Lee YH. Carbon nanotubes and graphene towards soft electronics. Nano Convergence. 2014;1(1):1–26.
Article
MathSciNet
Google Scholar
Herbert R, Jeong J-W, Yeo W-H. Soft material-enabled electronics for medicine, healthcare, and human-machine interfaces. Materials. 2020;13(3):517.
Article
Google Scholar
Hanif A, et al. Aptamer based nanobiosensors: promising healthcare devices. Saudi Pharma J. 2019;27(3):312–9.
Article
Google Scholar
Rauti R, et al. Properties and behavior of carbon nanomaterials when interfacing neuronal cells: How far have we come? Carbon. 2019;143:430–46.
Article
Google Scholar
Wang P, et al. Superhydrophobic flexible supercapacitors formed by integrating hydrogel with functional carbon nanomaterials. Chin J Chem. 2021;39(5):1153–8.
Article
Google Scholar
Alagarsamy KN, et al. Carbon nanomaterials for cardiovascular theranostics: Promises and challenges. Bioactive Materials. 2021;6(8):2261–80.
Article
Google Scholar
Gul A, et al. Carbon nanotubes for neural cell growth. In: Nanomedicine Manufacturing and Applications. Elsevier; 2021. p. 337–53.
Chapter
Google Scholar
Farokhi M, et al. Conductive biomaterials as substrates for neural stem cells differentiation towards neuronal lineage cells. Macromol Biosci. 2021;21(1):2000123.
Article
MathSciNet
Google Scholar
Cellot G, et al. Carbon nanotubes might improve neuronal performance by favouring electrical shortcuts. Nat Nanotechnol. 2009;4(2):126–33.
Article
Google Scholar
Pampaloni NP, et al. Sculpting neurotransmission during synaptic development by 2D nanostructured interfaces. Nanomedicine. 2018;14(7):2521–32.
Article
Google Scholar
Fabbro A, et al. Graphene-based interfaces do not alter target nerve cells. ACS Nano. 2016;10(1):615–23.
Article
Google Scholar
Zhang PX, et al. Tissue engineering for the repair of peripheral nerve injury. Neural Regen Res. 2019;14(1):51–8.
Article
Google Scholar
Koehne JE, et al. Carbon nanofiber electrode array for electrochemical detection of dopamine using fast scan cyclic voltammetry. Analyst. 2011;136(9):1802–5.
Article
Google Scholar
Meyyappan M, Sunkara MK. Inorganic nanowires: applications, properties, and characterization. New York: CRC Press; 2018.
Book
Google Scholar
Ren Z, et al. Heterogeneous wafer bonding technology and thin-film transfer technology-enabling platform for the next generation applications beyond 5G. Micromachines. 2021;12(8):946.
Article
Google Scholar
Gahl TJ, Kunze A. Force-mediating magnetic nanoparticles to engineer neuronal cell function. Front Neurosci. 2018;12:299.
Article
Google Scholar
Moretti D, et al. Biocompatibility of a magnetic tunnel junction sensor array for the detection of neuronal signals in culture. Front Neurosci. 2018;12:909.
Article
Google Scholar
Srivastava AK, et al. Advances in using MRI probes and sensors for in vivo cell tracking as applied to regenerative medicine. Dis Model Mech. 2015;8(4):323–36.
Article
Google Scholar
Lin B, et al. Nanomedicine directs neuronal differentiation of neural stem cells via silencing long noncoding RNA for Stroke Therapy. Nano Lett. 2021;21(1):806–15.
Article
Google Scholar
Li J, et al. Magnetic nanobubble mechanical stress induces the Piezo1-Ca2+-BMP2/Smad pathway to modulate neural stem cell fate and MRI/ultrasound dual imaging surveillance for ischemic stroke. Small. 2022;18(23):2201123.
Article
Google Scholar
Loeb C, et al. Liposome-entrapped γ-aminobutyric acid inhibits isoniazid-induced epileptogenic activity in rats. Epilepsia. 1986;27(2):98–102.
Article
Google Scholar
Loeb C, et al. Liposome-entrapped GABA modifies behavioral and electrographic changes of penicillin-induced epileptic activity. Neurology. 1982;32(11):1234–8.
Article
Google Scholar
Nsairat H, et al. Liposomes: structure, composition, types, and clinical applications. Heliyon. 2022;8(5): e09394.
Article
Google Scholar
Vaz GC, et al. Cardiovascular and behavioral effects produced by administration of liposome-entrapped GABA into the rat central nervous system. Neuroscience. 2015;285:60–9.
Article
Google Scholar
Silva-Barcellos NM, et al. Long-lasting cardiovascular effects of liposome-entrapped angiotensin-(1–7) at the rostral ventrolateral medulla. Hypertension. 2001;38(6):1266–71.
Article
Google Scholar
Frézard F, dos Santos RA, Fontes MA. Liposome-encapsulated neuropeptides for site-specific microinjection. In: Neuropeptides. Springer; 2011. p. 343–55.
Chapter
Google Scholar
Vaz GC, et al. Liposome-entrapped GABA modulates the expression of nNOS in NG108-15 cells. J Neurosci Methods. 2016;273:55–63.
Article
Google Scholar
Han L, Jiang C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B. 2021;11(8):2306–25.
Article
Google Scholar
Saraiva C, et al. Nanoparticle-mediated brain drug delivery: Overcoming blood–brain barrier to treat neurodegenerative diseases. J Control Release. 2016;235:34–47.
Article
Google Scholar
Archie SR, AlShoyaib A, Cucullo L. Blood-Brain Barrier Dysfunction in CNS disorders and putative therapeutic targets: an overview. Pharmaceutics. 2021;13:11.
Article
Google Scholar
Rip J. Liposome technologies and drug delivery to the CNS. Drug Discov Today Technol. 2016;20:53–8.
Article
Google Scholar
D’Agata F, et al. Magnetic nanoparticles in the central nervous system: targeting principles, applications and safety issues. Molecules. 2017;23(1):9.
Article
Google Scholar
Dahiya S, Dahiya R. 10 - Liposomes in drug targeting to brain tumors. In: Kumar L, Pathak YY, editors. Nanocarriers for drug-targeting brain tumors. Elsevier: New York; 2022. p. 299–327.
Chapter
Google Scholar
Ewert KK, et al. Cationic liposomes as vectors for nucleic acid and hydrophobic drug therapeutics. Pharmaceutics. 2021;13(9):1365.
Article
Google Scholar
Joshi S, et al. Cationic surface charge enhances early regional deposition of liposomes after intracarotid injection. J Neurooncol. 2014;120(3):489–97.
Article
Google Scholar
Tagalakis AD, et al. PEGylation improves the receptor-mediated transfection efficiency of peptide-targeted, self-assembling, anionic nanocomplexes. J Control Release. 2014;174:177–87.
Article
Google Scholar
Pulgar VM. Transcytosis to cross the blood brain barrier, new advancements and challenges. Front Neurosci. 2019;12:1019.
Article
Google Scholar
Spuch C, Navarro C. Liposomes for targeted delivery of active agents against neurodegenerative diseases (Alzheimer’s disease and Parkinson’s disease). J Drug Deliv. 2011;2011:99.
Article
Google Scholar
Ying X, et al. Dual-targeting daunorubicin liposomes improve the therapeutic efficacy of brain glioma in animals. J Control Release. 2010;141(2):183–92.
Article
Google Scholar
Pandey PK, Sharma AK, Gupta U. Blood brain barrier: An overview on strategies in drug delivery, realistic in vitro modeling and in vivo live tracking. Tissue Barriers. 2016;4(1): e1129476.
Article
Google Scholar
Marianecci C, et al. Drug delivery in overcoming the blood–brain barrier: role of nasal mucosal grafting. Drug Des Dev Ther. 2017;11:325.
Article
Google Scholar
Pashirova TN, et al. Mixed cationic liposomes for brain delivery of drugs by the intranasal route: the acetylcholinesterase reactivator 2-PAM as encapsulated drug model. Colloids Surf B Biointerfaces. 2018;171:358–67.
Article
Google Scholar
Dhaliwal HK, et al. Intranasal delivery and transfection of mRNA therapeutics in the brain using cationic liposomes. Mol Pharm. 2020;17(6):1996–2005.
Article
Google Scholar
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomed. 2016;11:5381–414.
Article
Google Scholar
Pehlivan SB. Nanotechnology-based drug delivery systems for targeting, imaging and diagnosis of neurodegenerative diseases. Pharm Res. 2013;30(10):2499–511.
Article
Google Scholar
Bors LA, Erdo F. Overcoming the blood–brain barrier challenges and tricks for CNS drug delivery. Sci Pharm. 2019;87(1):6.
Article
Google Scholar
Wen H, Jung H, Li X. Drug delivery approaches in addressing clinical pharmacology-related issues: opportunities and challenges. Aaps j. 2015;17(6):1327–40.
Article
Google Scholar
El-Boghdadly K, Pawa A, Chin KJ. Local anesthetic systemic toxicity: current perspectives. Local Reg Anesth. 2018;11:35–44.
Article
Google Scholar
During MJ, et al. Biochemical and behavioral recovery in a rodent model of Parkinson’s disease following stereotactic implantation of dopamine-containing liposomes. Exp Neurol. 1992;115(2):193–9.
Article
Google Scholar
Sercombe L, et al. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.
Article
Google Scholar
Ghosh S, Carter KA, Lovell JF. Liposomal formulations of photosensitizers. Biomaterials. 2019;218: 119341.
Article
Google Scholar
Eme-Scolan E, Dando SJ. Tools and approaches for studying microglia in vivo. Front Immunol. 2020;11: 583647.
Article
Google Scholar
Bachiller S, et al. Microglia in neurological diseases: a road map to brain-disease dependent-inflammatory response. Front Cell Neurosci. 2018;12:488.
Article
Google Scholar
Waltl I, Kalinke U. Beneficial and detrimental functions of microglia during viral encephalitis. Trends Neurosci. 2021;78:5.
Google Scholar
Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000.
Article
Google Scholar
Kraft AD, Harry GJ. Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity. Int J Environ Res Public Health. 2011;8(7):2980–3018.
Article
Google Scholar
Zhao N, et al. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng. 2020;4(3): 030902.
Article
Google Scholar
De SR, et al. Expression of phosphatidylserine receptor and down-regulation of pro-inflammatory molecule production by its natural ligand in rat microglial cultures. J Neuropathol Exp Neurol. 2002;61(3):237–44.
Article
Google Scholar
Naeini MB, et al. The role of phosphatidylserine recognition receptors in multiple biological functions. Cell Mol Biol Lett. 2020;25:23.
Article
Google Scholar
Ajmone-Cat MA, et al. Effects of phosphatidylserine on p38 mitogen activated protein kinase, cyclic AMP responding element binding protein and nuclear factor-kappaB activation in resting and activated microglial cells. J Neurochem. 2003;84(2):413–6.
Article
Google Scholar
Hashioka S, et al. Phospholipids modulate superoxide and nitric oxide production by lipopolysaccharide and phorbol 12-myristate-13-acetate-activated microglia. Neurochem Int. 2007;50(3):499–506.
Article
Google Scholar
Hashioka S, et al. Phosphatidylserine and phosphatidylcholine-containing liposomes inhibit amyloid beta and interferon-gamma-induced microglial activation. Free Radic Biol Med. 2007;42(7):945–54.
Article
Google Scholar
Patel T, et al. Polymeric nanoparticles for drug delivery to the central nervous system. Adv Drug Deliv Rev. 2012;64(7):701–5.
Article
Google Scholar
Lu Q, et al. Synthetic polymer nanoparticles functionalized with different ligands for receptor-mediated transcytosis across the blood–brain barrier. ACS Appl Bio Mater. 2018;1(5):1687–94.
Article
Google Scholar
Su S, Kang PM. Systemic review of biodegradable nanomaterials in nanomedicine. Nanomaterials (Basel). 2020;10:4.
Article
Google Scholar
Khiev D, et al. Emerging nano-formulations and nanomedicines applications for ocular drug delivery. Nanomaterials (Basel). 2021;11:1.
Article
Google Scholar
Mikušová V, Mikuš P. Advances in chitosan-based nanoparticles for drug delivery. Int J Mol Sci. 2021;22(17):45.
Article
Google Scholar
Zhang T-T, et al. Strategies for transporting nanoparticles across the blood–brain barrier. Biomater Sci. 2016;4(2):219–29.
Article
Google Scholar
Trujillo-de Santiago G, et al. Ocular adhesives: design, chemistry, crosslinking mechanisms, and applications. Biomaterials. 2019;197:345–67.
Article
Google Scholar
Couvreur P, et al. Polycyanoacrylate nanocapsules as potential lysosomotropic carriers: preparation, morphological and sorptive properties. J Pharm Pharmacol. 1979;31(1):331–2.
Article
Google Scholar
Vauthier C. A journey through the emergence of nanomedicines with poly(alkylcyanoacrylate) based nanoparticles. J Drug Target. 2019;27(5–6):502–24.
Article
Google Scholar
Zhang W, et al. Development of polymeric nanoparticles for blood–brain barrier transfer—strategies and challenges. Advan Sci. 2021;8(10):2003937.
Article
Google Scholar
He Y, et al. The effect of side chain engineering on conjugated polymers in organic electrochemical transistors for bioelectronic applications. J Mater Chem C. 2022;34:56.
Google Scholar
Nicolas J, Vauthier C. Poly (Alkyl Cyanoacrylate) Nanosystems. In: Intracellular Delivery. Springer; 2011. p. 225–50.
Chapter
Google Scholar
Jin X, et al. Chitosan–glutathione conjugate-coated poly (butyl cyanoacrylate) nanoparticles: promising carriers for oral thymopentin delivery. Carbohyd Polym. 2011;86(1):51–7.
Article
Google Scholar
Cabeza L, et al. Enhanced antitumor activity of doxorubicin in breast cancer through the use of poly (butylcyanoacrylate) nanoparticles. Int J Nanomed. 2015;10:1291.
Google Scholar
Arias J, et al. Preparation and characterization of carbonyl iron/poly (butylcyanoacrylate) core/shell nanoparticles. J Colloid Interface Sci. 2006;299(2):599–607.
Article
Google Scholar
Soppimath KS, et al. Biodegradable polymeric nanoparticles as drug delivery devices. J Control Release. 2001;70(1–2):1–20.
Article
Google Scholar
Andrieux K, Couvreur P. Polyalkylcyanoacrylate nanoparticles for delivery of drugs across the blood-brain barrier. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2009;1(5):463–74.
Article
Google Scholar
Alyautdin R, et al. Significant entry of tubocurarine into the brain of rats by adsorption to polysorbate 80–coated polybutylcyanoacrylate nanoparticles: an in situ brain perfusion study. J Microencapsul. 1998;15(1):67–74.
Article
Google Scholar
Van Der Kant R, Goldstein LS. Cellular functions of the amyloid precursor protein from development to dementia. Dev Cell. 2015;32(4):502–15.
Article
Google Scholar
Rayasa M, Reddy L. Poly(Alkyl Cyanoacrylate) Nanoparticles for Delivery of Anti-Cancer Drugs. 2006. p. 251–88.
Vauthier C, Labarre D, Ponchel G. Design aspects of poly(alkylcyanoacrylate) nanoparticles for drug delivery. J Drug Target. 2008;15:641–63.
Article
Google Scholar
Caraway CA, et al. Polymeric nanoparticles in brain cancer therapy: a review of current approaches. Polymers (Basel). 2022;14(14):56.
Article
Google Scholar
Vauthier C. A journey through the emergence of nanomedicines with poly (alkylcyanoacrylate) based nanoparticles. J Drug Target. 2019;27(5–6):502–24.
Article
Google Scholar
Wang C-X, et al. Antitumor effects of polysorbate-80 coated gemcitabine polybutylcyanoacrylate nanoparticles in vitro and its pharmacodynamics in vivo on C6 glioma cells of a brain tumor model. Brain Res. 2009;1261:91–9.
Article
Google Scholar
Merle P, et al. Safety and efficacy of intra-arterial hepatic chemotherapy with doxorubicin-loaded nanoparticles in hepatocellular carcinoma. ESMO open. 2017;2(4): e000238.
Article
MathSciNet
Google Scholar
Duan J, et al. Reversion of multidrug resistance by co-encapsulation of doxorubicin and curcumin in chitosan/poly(butyl cyanoacrylate) nanoparticles. Int J Pharm. 2012;426(1–2):193–201.
Article
Google Scholar
Gentile P, et al. An overview of poly(lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. Int J Mol Sci. 2014;15(3):3640–59.
Article
Google Scholar
Zhao D, et al. Poly(lactic-co-glycolic acid)-based composite bone-substitute materials. Bioactive Materials. 2021;6(2):346–60.
Article
Google Scholar
Mi F-L, et al. Chitin/PLGA blend microspheres as a biodegradable drug delivery system: a new delivery system for protein. Biomaterials. 2003;24(27):5023–36.
Article
Google Scholar
Makadia HK, Siegel SJ. Poly Lactic-co-Glycolic Acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers (Basel). 2011;3(3):1377–97.
Article
Google Scholar
Zeng F-R, Liang Y, Li Z-L. Precision aliphatic polyesters via segmer assembly polymerization. Molecules. 2018;23(2):452.
Article
Google Scholar
Lu Y, et al. Chirality-directed regioselectivity: an approach for the synthesis of alternating poly (lactic-co-glycolic acid). J Am Chem Soc. 2021;143(11):4119–24.
Article
Google Scholar
Tran KT, Nguyen TD. Lithography-based methods to manufacture biomaterials at small scales. J Sci. 2017;2(1):1–14.
Google Scholar
Leong J, et al. Engineering polymersomes for diagnostics and therapy. Adv Healthc Mater. 2018;7(8): e1701276.
Article
Google Scholar
Cao J, et al. Development of PLGA micro- and nanorods with high capacity of surface ligand conjugation for enhanced targeted delivery. Asian J Pharm Sci. 2019;14(1):86–94.
Article
Google Scholar
Rezvantalab S, et al. PLGA-based nanoparticles in cancer treatment. Front Pharmacol. 2018;9:1260.
Article
Google Scholar
Hoyos-Ceballos GP, et al. PLGA-PEG-ANG-2 nanoparticles for blood-brain barrier crossing: proof-of-concept study. Pharmaceutics. 2020;12(1):72.
Article
Google Scholar
Fornaguera C, et al. PLGA nanoparticles prepared by nano-emulsion templating using low-energy methods as efficient nanocarriers for drug delivery across the blood–brain barrier. J Control Release. 2015;211:134–43.
Article
Google Scholar
Li J, et al. Targeting the brain with PEG–PLGA nanoparticles modified with phage-displayed peptides. Biomaterials. 2011;32(21):4943–50.
Article
Google Scholar
Huang N, et al. PLGA nanoparticles modified with a BBB-penetrating peptide co-delivering Aβ generation inhibitor and curcumin attenuate memory deficits and neuropathology in Alzheimer’s disease mice. Oncotarget. 2017;8(46):81001–13.
Article
Google Scholar
Abbasi E, et al. Dendrimers: synthesis, applications, and properties. Nanoscale Res Lett. 2014;9(1):247.
Article
Google Scholar
Sato K, Anzai J-I. Dendrimers in layer-by-layer assemblies: synthesis and applications. Molecules. 2013;18(7):8440–60.
Article
Google Scholar
Yamashita S, et al. Development of PEGylated carboxylic acid-modified polyamidoamine dendrimers as bone-targeting carriers for the treatment of bone diseases. J Control Release. 2017;262:10–7.
Article
Google Scholar
Yellepeddi VK, Ghandehari H. Poly(amido amine) dendrimers in oral delivery. Tissue Barriers. 2016;4(2): e1173773.
Article
Google Scholar
Thakur S, et al. Impact of pegylation on biopharmaceutical properties of dendrimers. Polymer. 2015;59:67–92.
Article
Google Scholar
Igartúa DE, et al. PAMAM dendrimers as a carbamazepine delivery system for neurodegenerative diseases: A biophysical and nanotoxicological characterization. Int J Pharm. 2018;544(1):191–202.
Article
Google Scholar
Zhu Y, Liu C, Pang Z. Dendrimer-based drug delivery systems for brain targeting. Biomolecules. 2019;9(12):790.
Article
Google Scholar
Nance E, et al. Nanoscale effects in dendrimer-mediated targeting of neuroinflammation. Biomaterials. 2016;101:96–107.
Article
Google Scholar
Sarin H, et al. Effective transvascular delivery of nanoparticles across the blood-brain tumor barrier into malignant glioma cells. J Transl Med. 2008;6:80.
Article
Google Scholar
Sharma R, et al. Dendrimer mediated targeted delivery of sinomenine for the treatment of acute neuroinflammation in traumatic brain injury. J Control Release. 2020;323:361–75.
Article
Google Scholar
Moscariello P, et al. Brain delivery of multifunctional dendrimer protein bioconjugates. Advanced Science. 2018;5(5):1700897.
Article
Google Scholar
Srinageshwar B, et al. Surface-modified G4 PAMAM dendrimers cross the blood-brain barrier following multiple tail-vein injections in C57BL/6J Mice. ACS Chem Neurosci. 2019;10(9):4145–50.
Article
Google Scholar
Cook AB, Perrier S. Branched and dendritic polymer architectures: functional nanomaterials for therapeutic delivery. Adv Func Mater. 2020;30(2):1901001.
Article
Google Scholar
Zhang H, Cheng J, Ao Q. Preparation of alginate-based biomaterials and their applications in biomedicine. Mar Drugs. 2021;19(5):78.
Article
Google Scholar
Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog Polym Sci. 2012;37(1):106–26.
Article
Google Scholar
Yang J-S, Xie Y-J, He W. Research progress on chemical modification of alginate: a review. Carbohyd Polym. 2011;84(1):33–9.
Article
Google Scholar
Li Y, et al. Biodegradable polymer nanogels for drug/nucleic acid delivery. Chem Rev. 2015;115(16):8564–608.
Article
Google Scholar
Paques JP, et al. Preparation methods of alginate nanoparticles. Adv Coll Interface Sci. 2014;209:163–71.
Article
Google Scholar
Cai H, Ni C, Zhang L. Preparation of complex nano-particles based on alginic acid/poly [(2-dimethylamino) ethyl methacrylate] and a drug vehicle for doxorubicin release controlled by ionic strength. Eur J Pharm Sci. 2012;45(1–2):43–9.
Article
Google Scholar
Zheng S, et al. Development of high drug-loading nanomicelles targeting steroids to the brain. Int J Nanomed. 2014;9:55.
Google Scholar
Haque S, et al. Development and evaluation of brain targeted intranasal alginate nanoparticles for treatment of depression. J Psychiatr Res. 2014;48(1):1–12.
Article
Google Scholar
Hefnawy A, Khalil IA, El-Sherbiny IM. Facile development of nanocomplex-in-nanoparticles for enhanced loading and selective delivery of doxorubicin to brain. Nanomedicine. 2017;12(24):2737–61.
Article
Google Scholar
Qiu A, et al. Natural polysaccharide-based nanodrug delivery systems for treatment of diabetes. Polymers. 2022;14(15):3217.
Article
Google Scholar
Katsarov P, et al. Polysaccharide-based micro- and nanosized drug delivery systems for potential application in the pediatric dentistry. Polymers (Basel). 2021;13:19.
Article
Google Scholar
Younes I, et al. Chitin extraction from shrimp shell using enzymatic treatment. Antitumor, antioxidant and antimicrobial activities of chitosan. Int J Biol Macromol. 2014;69:489–98.
Article
Google Scholar
Aranaz I, et al. Chitosan: an overview of its properties and applications. Polymers. 2021;13(19):3256.
Article
Google Scholar
Jiménez-Gómez CP, Cecilia JA. Chitosan: a natural biopolymer with a wide and varied range of applications. Molecules. 2020;25(17):3981.
Article
Google Scholar
Aljawish A, et al. Enzymatic synthesis of chitosan derivatives and their potential applications. J Mol Catal B Enzym. 2015;112:25–39.
Article
Google Scholar
Ribeiro EF, et al. Chitosan and crosslinked chitosan nanoparticles: Synthesis, characterization and their role as Pickering emulsifiers. Carbohyd Polym. 2020;250: 116878.
Article
Google Scholar
Tarhini M, et al. Chapter 12 - Nanoparticles/nanoplatform to carry and deliver the drug molecules to the target site. In: Chappel E, editor., et al., Drug Delivery Devices and Therapeutic Systems. Academic Press: New York; 2021. p. 249–66.
Chapter
Google Scholar
Monsalve Y, et al. PEG-g-chitosan nanoparticles functionalized with the monoclonal antibody OX26 for brain drug targeting. Nanomedicine. 2015;10(11):1735–50.
Article
Google Scholar
Yu S, et al. Chitosan and chitosan coating nanoparticles for the treatment of brain disease. Int J Pharm. 2019;560:282–93.
Article
Google Scholar
Kotta S, et al. Progress in polymeric micelles for drug delivery applications. Pharmaceutics. 2022;14(8):78.
Article
Google Scholar
Kumar V, et al. Pharmacokinetics and biodistribution of polymeric micelles containing miRNA and small-molecule drug in orthotopic pancreatic tumor-bearing mice. Theranostics. 2018;8(15):4033–49.
Article
Google Scholar
Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm Res. 2007;24(1):1–16.
Article
Google Scholar
Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-based drug delivery systems in cancer therapy: what is available and what is yet to come. Pharmacol Rev. 2016;68(3):701–87.
Article
Google Scholar
Batrakova EV, Kabanov AV. Pluronic block copolymers: evolution of drug delivery concept from inert nanocarriers to biological response modifiers. J Control Release. 2008;130(2):98–106.
Article
Google Scholar
Yi X, Kabanov AV. Brain delivery of proteins via their fatty acid and block copolymer modifications. J Drug Target. 2013;21(10):940–55.
Article
Google Scholar
Kabanov AV, et al. The neuroleptic activity of haloperidol increases after its solubilization in surfactant micelles: micelles as microcontainers for drug targeting. FEBS Lett. 1989;258(2):343–5.
Article
Google Scholar
Zhang P, et al. Transferrin-modified c [RGDfK]-paclitaxel loaded hybrid micelle for sequential blood-brain barrier penetration and glioma targeting therapy. Mol Pharm. 2012;9(6):1590–8.
Article
Google Scholar
Miele D, et al. Chitosan oleate coated poly lactic-glycolic acid (PLGA) nanoparticles versus chitosan oleate self-assembled polymeric micelles, loaded with resveratrol. Mar Drugs. 2019;17(9):515.
Article
Google Scholar
Mishra V, et al. Solid lipid nanoparticles: emerging colloidal nano drug delivery systems. Pharmaceutics. 2018;10(4):7.
Article
Google Scholar
Mukherjee S, Ray S, Thakur RS. Solid lipid nanoparticles: a modern formulation approach in drug delivery system. Indian J Pharm Sci. 2009;71(4):349–58.
Article
Google Scholar
Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application. Adv Pharm Bull. 2015;5(3):305–13.
Article
Google Scholar
Wong HL, et al. Chemotherapy with anticancer drugs encapsulated in solid lipid nanoparticles. Adv Drug Deliv Rev. 2007;59(6):491–504.
Article
Google Scholar
Blasi P, et al. Solid lipid nanoparticles for targeted brain drug delivery. Adv Drug Deliv Rev. 2007;59(6):454–77.
Article
Google Scholar
Laquintana V, et al. New strategies to deliver anticancer drugs to brain tumors. Expert Opin Drug Deliv. 2009;6(10):1017–32.
Article
Google Scholar
Ayloo S, Gu C. Transcytosis at the blood-brain barrier. Curr Opin Neurobiol. 2019;57:32–8.
Article
Google Scholar
Stamatovic SM, Keep RF, Andjelkovic AV. Brain endothelial cell-cell junctions: how to “open” the blood brain barrier. Curr Neuropharmacol. 2008;6(3):179–92.
Article
Google Scholar
Aggarwal P, et al. Nanoparticle interaction with plasma proteins as it relates to particle biodistribution, biocompatibility and therapeutic efficacy. Adv Drug Deliv Rev. 2009;61(6):428–37.
Article
Google Scholar
Kreuter J. Nanoparticulate systems for brain delivery of drugs. Adv Drug Deliv Rev. 2001;47(1):65–81.
Article
Google Scholar
Manjunath K, Venkateswarlu V. Pharmacokinetics, tissue distribution and bioavailability of nitrendipine solid lipid nanoparticles after intravenous and intraduodenal administration. J Drug Target. 2006;14(9):632–45.
Article
Google Scholar
Dhawan S, Kapil R, Singh B. Formulation development and systematic optimization of solid lipid nanoparticles of quercetin for improved brain delivery. J Pharm Pharmacol. 2011;63(3):342–51.
Article
Google Scholar
Chattopadhyay N, et al. Solid lipid nanoparticles enhance the delivery of the HIV protease inhibitor, atazanavir, by a human brain endothelial cell line. Pharm Res. 2008;25(10):2262–71.
Article
Google Scholar
Bondì ML, et al. Brain-targeted solid lipid nanoparticles containing riluzole: preparation, characterization and biodistribution. Nanomedicine. 2010;5(1):25–32.
Article
Google Scholar
Bhalla N, et al. Opportunities and challenges for biosensors and nanoscale analytical tools for pandemics: COVID-19. ACS Nano. 2020;14(7):7783–807.
Article
Google Scholar
Rasooly A, Herold KE, Herold KE. Biosensors and biodetection. Berlin: Springer; 2009.
Book
Google Scholar
Moerner WE, Shechtman Y, Wang Q. Single-molecule spectroscopy and imaging over the decades. Faraday Discuss. 2015;184:9–36.
Article
Google Scholar
Lai M, Slaughter G. Label-Free MicroRNA Optical Biosensors. Nanomaterials (Basel, Switzerland). 2019;9(11):1573.
Article
Google Scholar
Kaisti M. Detection principles of biological and chemical FET sensors. Biosens Bioelectron. 2017;98:437–48.
Article
Google Scholar
Vu C-A, Chen W-Y. Field-effect transistor biosensors for biomedical applications: recent advances and future prospects. Sensors. 2019;19(19):4214.
Article
Google Scholar
Banica F-G. Chemical sensors and biosensors: fundamentals and applications. New York: Wiley; 2012.
Book
Google Scholar
Rim T, et al. Silicon nanowire biologically sensitive field effect transistors: electrical characteristics and applications. J Nanosci Nanotechnol. 2014;14(1):273–87.
Article
Google Scholar
Matte H, et al. Synthesis of inorganic fullerene-like nanostructures by concentrated solar and artificial light. Angew Chem Int Ed. 2010;122:4153–5.
Google Scholar
Lovinger DM. Communication networks in the brain: neurons, receptors, neurotransmitters, and alcohol. Alcohol Res Health. 2008;31(3):196–214.
Google Scholar
Spitzer NC. Neurotransmitter switching in the developing and adult brain. Annu Rev Neurosci. 2017;40:1–19.
Article
Google Scholar
Chvilicek MM, Titos I, Rothenfluh A. The neurotransmitters involved in drosophila alcohol-induced behaviors. Front Behav Neurosci. 2020;14:237.
Article
Google Scholar
Nabovati E, et al. Drug-drug interactions in inpatient and outpatient settings in Iran: a systematic review of the literature. DARU J Pharma Sci. 2014;22(1):1–13.
Google Scholar
McDonald AJ, Mott DD. Functional neuroanatomy of amygdalohippocampal interconnections and their role in learning and memory. J Neurosci Res. 2017;95(3):797–820.
Article
Google Scholar
Siddique SA, et al. Evaluation of neurotransmitter alterations in four distinct brain regions after rapid eye movement sleep deprivation (REMSD) induced mania-like behaviour in Swiss albino mice. Neurochem Res. 2018;43(6):1171–81.
Article
MathSciNet
Google Scholar
Chauhan N, et al. Recent advancement in nanosensors for neurotransmitters detection: Present and future perspective. Process Biochem. 2020;91:241–59.
Article
Google Scholar
Gallelli L, et al. Drug-Drug Interactions in cocaine-users and their clinical implications. Curr Drug Abuse Rev. 2017;10(1):25–30.
Article
Google Scholar
Robinson SL, Thiele TE. The role of neuropeptide Y (NPY) in alcohol and drug abuse disorders. Int Rev Neurobiol. 2017;136:177–97.
Article
Google Scholar
Birtwistle J, Baldwin D. Role of dopamine in schizophrenia and Parkinson’s disease. Br J Nurs. 1998;7(14):832–41.
Article
Google Scholar
Meyyappan M. Nano biosensors for neurochemical monitoring. Nano Convergence. 2015;2(1):18.
Article
Google Scholar
Lee KH, et al. Neurotransmitter release from high-frequency stimulation of the subthalamic nucleus. J Neurosurg. 2004;101(3):511–7.
Article
Google Scholar
Lin S-H, Lee L-T, Yang YK. Serotonin and mental disorders: a concise review on molecular neuroimaging evidence. Clin Psychopharmacol Neurosci. 2014;12(3):196.
Article
Google Scholar
Padayatty SJ, Levine M. Vitamin C: the known and the unknown and Goldilocks. Oral Dis. 2016;22(6):463–93.
Article
Google Scholar
Wei F, Lillehoj PB, Ho C-M. DNA diagnostics: nanotechnology-enhanced electrochemical detection of nucleic acids. Pediatr Res. 2010;67(5):458–68.
Article
Google Scholar
Kapur S, Remington G. Serotonin-dopamine interaction and its relevance to schizophrenia. Am J Psychiatry. 1996;153(4):466–76.
Article
Google Scholar
Wightman RM, May LJ, Michael AC. Detection of dopamine dynamics in the brain. Anal Chem. 1988;60(13):769A-793A.
Article
Google Scholar
Ni Y, Malarkey EB, Parpura V. Vesicular release of glutamate mediates bidirectional signaling between astrocytes and neurons. J Neurochem. 2007;103(4):1273–84.
Article
Google Scholar
Power AC, et al. Carbon nanomaterials and their application to electrochemical sensors: a review. Nanotechnol Rev. 2018;7(1):19–41.
Article
Google Scholar
Sekar M, et al. Carbon fiber based electrochemical sensor for sweat cortisol measurement. Sci Rep. 2019;9(1):403.
Article
Google Scholar
Zhang C, Du X. Electrochemical sensors based on carbon nanomaterial used in diagnosing metabolic disease. Front Chem. 2020;8:651.
Article
Google Scholar
Vitale F, et al. Neural stimulation and recording with bidirectional, soft carbon nanotube fiber microelectrodes. ACS Nano. 2015;9(4):4465–74.
Article
Google Scholar
Chen R, Canales A, Anikeeva P. Neural recording and modulation technologies. Nat Rev Mater. 2017;2(2):1–16.
Article
Google Scholar
Lee KH, et al. Evolution of deep brain stimulation: human electrometer and smart devices supporting the next generation of therapy. Neuromodulation. 2009;12(2):85–103.
Article
Google Scholar
Hartmann CJ, et al. An update on best practice of deep brain stimulation in Parkinson’s disease. Ther Adv Neurol Disord. 2019;12:1756286419838096.
Article
Google Scholar
Robinson DL, et al. Monitoring rapid chemical communication in the brain. Chem Rev. 2008;108(7):2554–84.
Article
Google Scholar
Swami V, Vijayaraghavan V, Swami V. Current trends to measure implant stability. J Indian Prosthodontic Soc. 2016;16(2):124.
Article
Google Scholar
Li Y, et al. Electrocatalytic detection of dopamine in the presence of ascorbic acid and uric acid using single-walled carbon nanotubes modified electrode. Colloids Surf, B. 2012;97:32–6.
Article
Google Scholar
Jeon SK, et al. Mechanical test method and properties of a carbon nanomaterial with a high aspect ratio. Nano Convergence. 2016;3(1):1–10.
Article
Google Scholar
Komathi S, Gopalan AI, Lee K-P. Nanomolar detection of dopamine at multi-walled carbon nanotube grafted silica network/gold nanoparticle functionalised nanocomposite electrodes. Analyst. 2010;135(2):397–404.
Article
Google Scholar
Yang S, et al. Nano-sized copper oxide/multi-wall carbon nanotube/Nafion modified electrode for sensitive detection of dopamine. J Electroanal Chem. 2013;703:45–51.
Article
Google Scholar
Aravind SJ, Ramaprabhu S. Noble metal dispersed multiwalled carbon nanotubes immobilized ss-DNA for selective detection of dopamine. Sens Actuators, B Chem. 2011;155(2):679–86.
Article
Google Scholar
Swamy BK, Venton BJ. Carbon nanotube-modified microelectrodes for simultaneous detection of dopamine and serotonin in vivo. Analyst. 2007;132(9):876–84.
Article
Google Scholar
Kim Y-R, et al. Electrochemical detection of dopamine in the presence of ascorbic acid using graphene modified electrodes. Biosens Bioelectron. 2010;25(10):2366–9.
Article
Google Scholar
Alwarappan S, et al. Probing the electrochemical properties of graphene nanosheets for biosensing applications. J Phys Chem C. 2009;113(20):8853–7.
Article
Google Scholar
Sun C-L, et al. The simultaneous electrochemical detection of ascorbic acid, dopamine, and uric acid using graphene/size-selected Pt nanocomposites. Biosens Bioelectron. 2011;26(8):3450–5.
Article
Google Scholar
Tan L, et al. Nanomolar detection of dopamine in the presence of ascorbic acid at β-cyclodextrin/graphene nanocomposite platform. Electrochem Commun. 2010;12(4):557–60.
Article
Google Scholar
Han D, et al. Simultaneous determination of ascorbic acid, dopamine and uric acid with chitosan-graphene modified electrode. Electroanalysis. 2010;22(17–18):2001–8.
Article
Google Scholar
Gao F, et al. Highly sensitive and selective detection of dopamine in the presence of ascorbic acid at graphene oxide modified electrode. Sens Actuators, B Chem. 2013;186:380–7.
Article
Google Scholar
Thomas T, et al. Graphite oxide bulk modified carbon paste electrode for the selective detection of dopamine: a voltammetric study. J Electroanal Chem. 2011;659(1):113–9.
Article
Google Scholar
Nurzulaikha R, et al. Graphene/SnO2 nanocomposite-modified electrode for electrochemical detection of dopamine. Sens Bio-Sens Res. 2015;5:42–9.
Article
Google Scholar
Tsai H-Y, Lin Z-H, Chang H-T. Tellurium-nanowire-coated glassy carbon electrodes for selective and sensitive detection of dopamine. Biosens Bioelectron. 2012;35(1):479–83.
Article
Google Scholar
Minnikanti S, et al. In vivo electrochemical characterization and inflammatory response of multiwalled carbon nanotube-based electrodes in rat hippocampus. J Neural Eng. 2010;7(1): 016002.
Article
Google Scholar
Raina S, et al. Electrochemical biosensor utilizing nitrogen incorporated nanodiamond ultra-microelectrode array. ECS Trans. 2019;28(34):21–8.
Article
Google Scholar
Li D-C, Yang P-H, Lu MS-C. CMOS open-gate ion-sensitive field-effect transistors for ultrasensitive dopamine detection. IEEE Trans Electron Devices. 2010;57(10):2761–7.
Article
Google Scholar
Balogh EP, Miller BT, Ball JR. Improving diagnosis in health care. 2015.
Mount CW, Monje M. Wrapped to adapt: experience-dependent myelination. Neuron. 2017;95(4):743–56.
Article
Google Scholar
Trachtenberg JT, et al. Long-term in vivo imaging of experience-dependent synaptic plasticity in adult cortex. Nature. 2002;420(6917):788–94.
Article
Google Scholar
Bansal R, et al. Anatomical brain images alone can accurately diagnose chronic neuropsychiatric illnesses. PLoS ONE. 2012;7(12): e50698.
Article
Google Scholar
McGhee DJ, et al. A systematic review of biomarkers for disease progression in Parkinson’s disease. BMC Neurol. 2013;13(1):1–13.
Article
Google Scholar
Vaghela V, Kesavadas C, Thomas B. Functional magnetic resonance imaging of the brain: A quick review. Neurol India. 2010;58(6):879.
Article
Google Scholar
He B, Liu Z. Multimodal functional neuroimaging: integrating functional MRI and EEG/MEG. IEEE Rev Biomed Eng. 2008;2008(1):23–40.
Google Scholar
Sheng ZH, Cai Q. Mitochondrial transport in neurons: impact on synaptic homeostasis and neurodegeneration. Nat Rev Neurosci. 2012;13(2):77–93.
Article
Google Scholar
Svoboda K, Yasuda R. Principles of two-photon excitation microscopy and its applications to neuroscience. Neuron. 2006;50(6):823–39.
Article
Google Scholar
Misgeld T, Kerschensteiner M. In vivo imaging of the diseased nervous system. Nat Rev Neurosci. 2006;7(6):449–63.
Article
Google Scholar
Nune SK, et al. Nanoparticles for biomedical imaging. Expert Opin Drug Deliv. 2009;6(11):1175–94.
Article
Google Scholar
Nowostawska M, et al. Porphyrin-magnetite nanoconjugates for biological imaging. J Nanobiotechnol. 2011;9(1):1–12.
Article
Google Scholar
Naqvi S, Panghal A, Flora SJS. Nanotechnology: a promising approach for delivery of neuroprotective drugs. Front Neurosci. 2020;14:494.
Article
Google Scholar
Papadimitriou L, et al. Biofabrication for neural tissue engineering applications. Materials Today Bio. 2020;6: 100043.
Article
Google Scholar
Wang W, et al. Carbon nanomaterials for electro-active structures: a review. Polymers. 2020;12(12):2946.
Article
Google Scholar
Narayanan G, et al. Poly (lactic acid)-based biomaterials for orthopaedic regenerative engineering. Adv Drug Deliv Rev. 2016;107:247–76.
Article
Google Scholar
Silva GA, et al. Selective differentiation of neural progenitor cells by high-epitope density nanofibers. Science. 2004;303(5662):1352–5.
Article
Google Scholar
Tunggal P, et al. Laminins: structure and genetic regulation. Microsc Res Tech. 2000;51(3):214–27.
Article
Google Scholar
Nomizu M, et al. Structure-activity study of a laminin α1 chain active peptide segment Ile-Lys-Val-Ala-Val (IKVAV). FEBS Lett. 1995;365(2–3):227–31.
Article
Google Scholar
Powell SK, et al. Neural cell response to multiple novel sites on laminin-1. J Neurosci Res. 2000;61(3):302–12.
Article
Google Scholar
Droge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002;82(1):47–95.
Article
Google Scholar
Choi SW, Kim J. Recent progress in autocatalytic ceria nanoparticles-based translational research on brain diseases. ACS Applied Nano Materials. 2020;3(2):1043–62.
Article
Google Scholar
Roy P, et al. Exploring the inhibitory and antioxidant effects of fullerene and fullerenol on ribonuclease A. ACS Omega. 2018;3(9):12270–83.
Article
Google Scholar
Belov Kirdajova D, et al. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front Cell Neurosci. 2020;14:51.
Article
Google Scholar
Dugan LL, et al. Buckminsterfullerenol free radical scavengers reduce excitotoxic and apoptotic death of cultured cortical neurons. Neurobiol Dis. 1996;3(2):129–35.
Article
Google Scholar
Fortina P, et al. Nanobiotechnology: the promise and reality of new approaches to molecular recognition. Trends Biotechnol. 2005;23(4):168–73.
Article
Google Scholar
Jin H, et al. Polyhydroxylated C60, fullerenols, as glutamate receptor antagonists and neuroprotective agents. J Neurosci Res. 2000;62(4):600–7.
Article
Google Scholar
Grebowski J, Kazmierska P, Krokosz A. Fullerenols as a new therapeutic approach in nanomedicine. Biomed Res Int. 2013;2013:45.
Article
Google Scholar
Foran E, Trotti D. Glutamate transporters and the excitotoxic path to motor neuron degeneration in amyotrophic lateral sclerosis. Antioxid Redox Signal. 2009;11(7):1587–602.
Article
Google Scholar
Van Damme P, et al. GluR2 deficiency accelerates motor neuron degeneration in a mouse model of amyotrophic lateral sclerosis. J Neuropathol Exp Neurol. 2005;64(7):605–12.
Article
Google Scholar
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res Int. 2014;2014:45.
Article
Google Scholar
Gil ES, et al. β-Cyclodextrin-poly (β-amino ester) nanoparticles for sustained drug delivery across the blood–brain barrier. Biomacromol. 2012;13(11):3533–41.
Article
Google Scholar
Vinogradov SV, Batrakova EV, Kabanov AV. Nanogels for oligonucleotide delivery to the brain. Bioconjug Chem. 2004;15(1):50–60.
Article
Google Scholar
Schroeder U, et al. Nanoparticle technology for delivery of drugs across the blood–brain barrier. J Pharm Sci. 1998;87(11):1305–7.
Article
Google Scholar
Alyaudtin RN, et al. Interaction of poly (butylcyanoacrylate) nanoparticles with the blood-brain barrier in vivo and in vitro. J Drug Target. 2001;9(3):209–21.
Article
Google Scholar
Garcia-Garcia E, et al. A relevant in vitro rat model for the evaluation of blood-brain barrier translocation of nanoparticles. Cell Mol Life Sci. 2005;62(12):1400–8.
Article
Google Scholar
Brigger I, et al. Poly (ethylene glycol)-coated hexadecylcyanoacrylate nanospheres display a combined effect for brain tumor targeting. J Pharmacol Exp Ther. 2002;303(3):928–36.
Article
Google Scholar
Kreuter J, et al. Apolipoprotein-mediated transport of nanoparticle-bound drugs across the blood-brain barrier. J Drug Target. 2002;10(4):317–25.
Article
Google Scholar
Zhang ZG, Chopp M. Neurorestorative therapies for stroke: underlying mechanisms and translation to the clinic. Lancet Neurol. 2009;8(5):491–500.
Article
Google Scholar
Zagrean A-M, et al. Multicellular crosstalk between exosomes and the neurovascular unit after cerebral ischemia, therapeutic implications. Front Neurosci. 2018;12:811.
Article
Google Scholar
Council, N.R., Opportunities in biology. 1989.
Nguyen PQ, et al. Engineered living materials: prospects and challenges for using biological systems to direct the assembly of smart materials. Adv Mater. 2018;30(19): e1704847.
Article
Google Scholar
Simon T, Bromberg JS. Regulation of the immune system by laminins. Trends Immunol. 2017;38(11):858–71.
Article
Google Scholar
Schéele S, et al. Laminin isoforms in development and disease. J Mol Med. 2007;85(8):825–36.
Article
Google Scholar
Miner JH, et al. The laminin alpha chains: expression, developmental transitions, and chromosomal locations of alpha1-5, identification of heterotrimeric laminins 8–11, and cloning of a novel alpha3 isoform. J Cell Biol. 1997;137(3):685–701.
Article
Google Scholar
Nelson J, et al. Facing addiction in America: The surgeon general’s report on alcohol, drugs, and health: A commentary. Alcohol Treat Q. 2017;35(4):445–54.
Article
Google Scholar
Silva GA. Neuroscience nanotechnology: progress, opportunities and challenges. Nanosci Technol. 2010;34:251–60.
Google Scholar
Patra JK, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol. 2018;16(1):71.
Article
Google Scholar
van Haasteren J, Hyde SC, Gill DR. Lessons learned from lung and liver in-vivo gene therapy: implications for the future. Expert Opin Biol Ther. 2018;18(9):959–72.
Article
Google Scholar