943
Views
51
CrossRef citations to date
0
Altmetric
Review Article

Nanoneurotherapeutics approach intended for direct nose to brain delivery

, , &
Pages 1922-1934 | Received 12 Jan 2015, Accepted 07 May 2015, Published online: 09 Jun 2015

References

  • Verma R, Nehru B. Effect of centrophenoxine against rotenone-induced oxidative stress in an animal model of Parkinson's disease. Neurochem Int 2009;55:369–75
  • Pardridge WM. Blood–brain barrier delivery. Drug Discov Today 2007;12:55–61
  • Dhuria SV, Hanson LR, Frey WH. Intranasal delivery to the central nervous system: mechanism and experimental considerations. J Pharm Sci 2010;99:1654–73
  • Illum L. Is nose-to-brain transport of drugs in man a reality? J Pharm Pharmacol 2004;56:3–17
  • Mistry A, Stolnik S, Illum L. Nanoparticles for direct nose-to-brain delivery of drugs. Int J Pharm 2009;379:146–57
  • Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 2011;147:728–41
  • Jellinger KA. General aspects of neurodegeneration. J Neural Trans 2003;65:101–44
  • Jellinger KA. Basic mechanisms of neurodegeneration: a critical update. J Cell Mol Med 2010;14:457–87
  • Mizushima N, Yamamoto A, Matsui M, et al. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 2004;15:1101–11
  • Nixon RA, Wegiel J, Kumar A, et al. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol 2005;64:113–22
  • Boland B, Nixon RA. Neuronal macroautophagy: from development to degeneration. Mol Aspects Med 2006;27:503–19
  • Lee JA, Beigneux A, Ahmad ST, et al. ESCRT-III dysfunction causes autophagosome accumulation and neurodegeneration. Curr Biol 2007;17:1561–7
  • Yue Z, Friedman L, Komatsu M, Tanaka K. The cellular pathways of neuronal autophagy and their implication in neurodegenerative diseases. Biochim Biophys Acta 2009;1793:1496–507
  • Komatsu M, Waguri S, Koike M, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007;131:1149–63
  • Pankiv S. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 2007;282:24131–45
  • Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 2002;11:1107–17
  • Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006;443:780–6
  • Fortun J, Dunn WA Jr, Joy S, et al. Emerging role for autophagy in the removal of aggresomes in Schwann cells. J Neurosci 2003;23:10672–80
  • Bjørkøy G, Lamark T, Brech A, et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 2005;171:603–14
  • Kabuta T, Suzuki Y, Wada K. Degradation of amyotrophic lateral sclerosis-linked mutant Cu/Zn-superoxide dismutase proteins by macroautophagy and the proteasome. J Biol Chem 2006;281:30524–33
  • Berger Z, Ravikumar B, Menzies FM, et al. Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet 2006;15:433–42
  • Hara T, Nakamura K, Matsui M, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006;441:885–9
  • Komatsu M. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 2006;441:880–4
  • Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008;132:27–42
  • Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature 2008;451:1069–75
  • Pardridge WM. The blood–brain barrier: bottleneck in brain drug development. NeuroRx 2005;2:3–14
  • Misra A, Ganesh S, Shahiwala A. Drug delivery to the central nervous system: a review. J Pharm Sci 2003;6:252–73
  • Neuwelt EA. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 2011;12:169–82
  • Wong HL, Wu XY, Bendayan R. Nanotechnological advances for the delivery of CNS therapeutics. Adv Drug Deliv Rev 2012;64:686–700
  • Segal MB. The choroid plexuses and the barriers between the blood and the cerebro spinal fluid. Cell Mol Neurobiol 2000;20:183–96
  • Enting RH, Hoetelmans RM, Lange JM, et al. Anti-retroviral drugs and the central nervous system. AIDS 1998;12:1941–55
  • Lee G, Dallas S, Hong M, Bendayan R. Drug transporters in the central nervous system: brain barriers and brain parenchyma considerations. Pharmacol Rev 2001;53:569–96
  • Wohlfart S, Gelperina S, Kreuter J. Transport of drugs across the blood–brain barrier by nanoparticles. J Control Release 2012;161:264–73
  • Nutt JG, Burchiel KJ, Comella CL, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 2003;60:69–73
  • Lesniak MS, Brem H. Targeted therapy for brain tumours. Nat Rev Drug Discov 2004;3:499–508
  • Ashby LS, Ryken TC. Management of malignant glioma: steady progress with multimodal approaches. Neurosurg Focus 2006;20:E3
  • Biddlestone-Thorpe L, Marchi N, Guo K, et al. Nanomaterial-mediated CNS delivery of diagnostic and therapeutic agents. Adv Drug Deliv Rev 2012;64:605–13
  • Illum L. Nasal drug delivery – possibilities, problems and solutions. J Control Release 2003;87:187–98
  • Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev 2012;64:614–28
  • Dhuria SV, Hanson LR, Frey WH. Novel vasoconstrictor formulation to enhance intranasal targeting of neuropeptide therapeutics to the central nervous system. J Pharmacol Exp Ther 2009;328:312–20
  • Pires A, Fortuna A, Alves G, Falcão A. Intranasal drug delivery: how, why and what for? J Pharm Pharmaceut Sci 2009;12:288–311
  • Ying W. The nose may help the brain: intranasal drug delivery for treating neurological diseases. Future Neurol 2008;3:1–4
  • Mittal D, Ali A, Md S, et al. Insights into direct nose to brain delivery: current status and future perspective. Drug Deliv 2014;21:75–86
  • Chen XQ, Fawcett JR, Rahman YE, et al. Delivery of nerve growth factor to the brain via the olfactory pathway. J Alzheimers Dis 1998;1:35–44
  • Md S, Kumar M, Baboota S, et al. Preparation, characterization and evaluation of bromocriptine loaded chitosan nanoparticles for intranasal delivery. Sci Adv Mater 2012;4:949–60
  • Chugh Y, Kapoor P, Kapoor AK. Intranasal drug delivery: a novel approach. Indian J Otolaryngol Head Neck Surg 2009;61:90–4
  • Gao X, Tao W, Lu W, et al. Lectin conjugated PEG–PLA nanoparticles: preparation and brain delivery after intranasal administration. Biomaterials 2006;27:3482–90
  • Gao X, Chen J, Tao W, et al. Uea I bearing nanoparticles for brain delivery following intranasal administration. Int J Pharm 2007;340:207–15
  • Mustafa G, Ahuja A, Ali J, et al. Nose to brain targeting potential of a chitosan coated nano-formulation: pharmacodynamic and pharmacoscintigraphic evaluation. Sci Adv Mater 2014;5:1236–49
  • Md S, Khan RA, Mustafa G, et al. Bromocriptine loaded chitosan nanoparticles intended for direct nose to brain delivery: pharmacodynamic, pharmacokinetic and scintigraphy study in mice model. Eur J Pharm Sci 2012;48:393–405
  • Haque S, Md S, Fazil M, et al. Venlafaxine loaded chitosan NPs for brain targeting: pharmacokinetic and pharmacodynamic evaluation. Carbohydr Polym 2012;89:72–9
  • Alam S, Khan ZI, Mustafa G, et al. Development and evaluation of thymoquinone-encapsulated chitosan nanoparticles for nose-to-brain targeting: a pharmacoscintigraphic study. Int J Nanomed 2012;5705–18
  • Labuzek K, Gorki K, Jaroszek H, et al. Highly organized nanostructures for brain drug delivery – new hope or just a fad? CNS Neurol Disord Drug Targets 2013;12:1271–85
  • Webb MS, Rebstein P, Lamson W, Bally MB. Liposomal drug delivery: recent patents and emerging opportunities. Recent Pat Drug Deliv Formul 2007;1:185–94
  • Barenholz Y. Liposome application: problems and prospects. Curr Opin Colloid Inter Sci 2001;6:66–77
  • Bertrand N, Simard P, Leroux JC. Serum-stable, long-circulating, pH-sensitive PEGylated liposomes. Methods Mol Biol 2010;605:545–58
  • Yue X, Dai Z. Recent advances in liposomal nanohybrid cerasomes as promising drug nanocarriers. Adv Colloid Interface Sci 2014;207:32–42
  • Croy SR, Kwon GS. Polymeric micelles for drug delivery. Curr Pharm Des 2006;12:4669–84
  • Bae YH, Yin H. Stability issues of polymeric micelles. J Control Release 2008;131:2–4
  • Gillies ER, Fréchet JM. Dendrimers and dendritic polymers in drug delivery. Drug Discov Today 2005;10:35–43
  • Cheng Y, Tongwen X. Dendrimers as potential drug carriers. Part I: solubilization of non-steroidal anti-inflammatory drugs in the presence of polyamidoamine dendrimers. Eur J Med Chem 2005;40:1188–92
  • He H, Li Y, Jia XR, et al. PEGylated poly(amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials 2011;23:478–87
  • Wong HL, Li Y, Bendayan R, et al. Solid-lipid nanoparticles for antitumor drug delivery. In: Amiji MM, ed. Nanotechnology for cancer therapeutics. Boca Raton, FL: CRC Press; 2006:714–76
  • Alam MI, Baboota S, Ahuja A, et al. Pharmacoscintigraphic evaluation of potential of lipid nanocarriers for nose-to-brain delivery of antidepressant drug. Int J Pharm 2014;470:99–106
  • Alam MI, Baboota S, Ahuja A, et al. Intranasal administration of nanostructured lipid carriers containing CNS acting drug: pharmacodynamic studies and estimation in blood and brain. J Psychiatr Res 2012;46:1133–8
  • Wong HL, Bendayan R, Rauth AM, et al. Chemotherapy with anticancer drugs encapsulated in solid lipid nanoparticles. Adv Drug Deliv Rev 2007;59:491–504
  • Kathe N, Henriksen B, Chauhan H. Physicochemical characterization techniques for solid lipid nanoparticles: principles and limitations. Drug Dev Ind Pharm 2014;40:1565–75
  • Iqbal MA, Md S, Sahni JK, et al. Nanostructured lipid carriers system: recent advances in drug delivery. J Drug Target 2012;20:813–30
  • Rempe R, Cramer S, Huwel S, Galla HJ. Transport of poly (n-butylcyano-acrylate) nanoparticles across the blood–brain barrier in vitro and their influence on barrier integrity. Biochem Biophys Res Commun 2011;406:64–9
  • Tian XH, Lin XN, Wei F, et al. Enhanced brain targeting of temozolomide in polysorbate-80 coated poly butyl cyanoacrylate nanoparticles. Int J Nanomed 2011;6:445–52
  • Li T, Shu YJ, Cheng JY, et al. Pharmacokinetics and efficiency of brain targeting of ginsenosides Rg1 and Rb1 given as Nao-Qing microemulsion. Drug Dev Ind Pharm 2015;41:224–31
  • Kreuter J. Nanoparticles. In: Kreuter J, ed. Colloidal drug delivery systems. New York: Marcel Dekker; 1994:219–342
  • Kreuter J. Nanoparticulate systems for brain delivery of drugs. Adv Drug Deliv Rev 2001;47:65–81
  • Hoet PHM, Brüske-Hohlfeld I, Salata OV. Nanoparticles – known and unknown health risks. J Nanobiotechnol 2004;2:12–27
  • Borm PJ, Muller-Schulte D. Nanoparticles in drug delivery and environmental exposure: same size, same risks? Nanomedicine 2006;1:235–49
  • Muller RH, Gohla S, Keck CM. State of the art of nanocrystals – special features, production nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm 2011;78:1–9
  • Furumoto K, Nagayama S, Ogawara K, et al. Hepatic uptake of negatively charged particles in rats: possible involvement of serum proteins in recognition by scavenger receptor. J Control Release 2004;97:133–41
  • McNeeley KM, Karathanasis E, Annapragada AV, Bellamkonda RV. Masking and triggered unmasking of targeting ligands on nanocarriers to improve drug delivery to brain tumors. Biomaterials 2009;30:3986–95
  • Olivier JC, Huerta R, Lee HJ, et al. Synthesis of PEGylated immune nanoparticles. Pharm Res 2002;19:117–43
  • Aktaş Y, Yemisci M, Andrieux K, et al. Development and brain delivery of chitosan–PEG nanoparticles functionalized with the monoclonal antibody OX26. Bioconjug Chem 2005;16:1503–11
  • Costantino L, Gandolfi F, Tosi G, et al. Peptide-derivatized biodegradable nanoparticles able to cross the blood brain barrier. J Control Release 2005;108:84–96
  • Lockman PR, Oyewumi MO, Koziara JM, et al. Brain uptake of thiamine-coated nanoparticles. J Control Release 2003;93:271–82
  • Goppert TM, Muller RH. Polysorbate-stabilized solid lipid nanoparticles as colloidal carriers for intravenous targeting of drugs to the brain: comparison of plasma protein adsorption patterns. J Drug Target 2005;13:179–87
  • Koziara JM, Lockman PR, Allen DD, Mumper RJ. In situ blood-brain barrier transport of nanoparticles. Pharm Res 2003;20:1772–8
  • Novakovic D, Feligioni M, Scaccianoce S, et al. Profile of gantenerumab and its potential in the treatment of Alzheimer's disease. Drug Des Devel Ther 2013;7:1359–64
  • Kar S, Slowikowski SP, Westaway D, Mount HT. Interactions between beta-amyloid and central cholinergic neurons: implications for Alzheimer's disease. J Psychiatry Neurosci 2004;29:427–41
  • Lahiri DK, Greig NH. Lethal weapon: amyloid beta-peptide, role in the oxidative stress and neurodegeneration of Alzheimer's disease. Neurobiol Aging 2004;25:581–7
  • Steffensmeier AM, Tare M, Puli OR, et al. Novel neuroprotective function of apical-basal polarity gene crumbs in amyloid beta 42 (aβ42) mediated neurodegeneration. PLoS One 2013;8:78717
  • Fazil M, Md S, Haque S, et al. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur J Pharm Sci 2012;47:6–15
  • Bhavna, Md S, Ali M, et al. Donepezil nanosuspension intended for nose to brain targeting: in vitro and in vivo safety evaluation. Int J Biol Macromol 2014;67:418–25
  • Wang X, Chi N, Tang X. Preparation of estradiol chitosan nanoparticles for improving nasal absorption and brain targeting. Eur J Pharm Biopharm 2008;70:735–40
  • Tahara K, Sakai T, Yamamoto H, et al. Improved cellular uptake of chitosan-modified PLGA nanospheres by A549 cells. Int J Pharm 2009;382:198–204
  • Liu Z, Jiang M, Kang T, et al. Lactoferrin-modified PEG-co-PCL nanoparticles for enhanced brain delivery of NAP peptide following intranasal administration. Biomaterials 2013;34:3870–81
  • Lang AE, Lozano AM. Parkinson's disease. First of two parts. New Engl J Med 1998;339:1044–53
  • Gibb WR. Neuropathology of Parkinson's disease and related syndromes. Neurol Clin 1992;10:361–76
  • Kanwar JR, Sun X, Punj V, et al. Nanoparticles in the treatment and diagnosis of neurological disorders: untamed dragon with fire power to heal. Nanomedicine 2012;8:399–414
  • Le W. Role of iron in UPS impairment model of Parkinson's disease. Park Relat Disord 2014;20:158–61
  • Doorn JA, Florang VR, Schamp JH, Vanle BC. Aldehyde dehydrogenase inhibition generates a reactive dopamine metabolite autotoxic to dopamine neurons. Parkinsonism Relat Disord 2014;20S:S73–5
  • Sampath U, Janardhanam VA. Asiaticoside, a trisaccaridetriterpene induces biochemical and molecular variations in brain of mice with parkinsonism. Transl Neurodegener 2013;2:23. doi: 10.1186/2047-9158-2-23
  • Jafarieh O, Md S, Ali M, et al. Design, characterization, and evaluation of intranasal delivery of ropinirole-loaded mucoadhesive nanoparticles for brain targeting. Drug Dev Ind Pharm 2014. [Epub ahead of print]. doi: 10.3109/03639045.2014.991400
  • Sharma S, Lohan S, Murthy RSR. Formulation and characterization of intranasal mucoadhesive nanoparticulates and thermo-reversible gel of levodopa for brain delivery. Drug Dev Ind Pharm 2014;40:869–78
  • Abuirmeileh A, Harkavyi A, Kingsbury A, et al. The CRF-like peptide urocortin produces a long-lasting recovery in rats made hemiparkinsonian by 6-hydroxydopamine or lipopolysaccharide. J Neurol Sci 2008;271:131–6
  • Brasnjevic I, Steinbusch HW, Schmitz C, Martinez-Martinez P. Delivery of peptide and protein drugs over the blood–brain barrier. Prog Neurobiol 2009;87:212–51
  • Wen Z, Yan Z, Hu K, et al. Odorranalectin-conjugated nanoparticles: preparation, brain delivery and pharmacodynamic study on Parkinson's disease following intranasal administration. J Control Release 2011;151:131–8
  • Pardeshi CV, Rajput PV, Belgamwar VS, et al. Novel surface modified solid lipid nanoparticles as intranasal carriers for ropinirole hydrochloride: application of factorial design approach. Drug Deliv 2013;20:47–56
  • Pardeshi CV, Belgamwar VS, Tekade AR, Surana SJ. Novel surface modified polymer-lipid hybrid nanoparticles as intranasal carriers for ropinirole hydrochloride: in vitro, ex vivo and in vivo pharmacodynamic evaluation. J Mater Sci Mater Med 2013;24:2101–15
  • Howells DW, Porritt MJ, Wong JY, et al. Reduced BDNF mRNA expression in the Parkinson's disease substantia nigra. Exp Neurol 2000;166:127–35
  • Rangasamy SB, Soderstrom K, Bakay RAE, Kordower JH. Chapter 13 – neurotrophic factor therapy for Parkinson's disease. In: Anders B, Cenci MA, eds. Progress in brain research. Amsterdam: Elsevier; 2010:237–64
  • Zhao YZ, Li X, Lu CT, et al. Gelatin nanostructured lipid carriers-mediated intranasal delivery of basic fibroblast growth factor enhances functional recovery in hemiparkinsonian rats. Nanomed Nanotech Biol Med 2014;10:755–64
  • Pan W, Banks WA, Fasold MB, et al. Transport of brain derived neurotrophic factor across the blood–brain barrier. Neuropharmacology 1998;37:1553–61
  • Shinoyama M, Ideguchi M, Kida H, et al. Cortical region-specific engraftment of embryonic stem cell-derived neural progenitor cells restores axonal sprouting to a subcortical target and achieves motor functional recovery in a mouse model of neonatal hypoxic-ischemic brain injury. Front Cell Neurosci 2013;7:128
  • Brodie MJ, Elder AT, Kwan P. Epilepsy in later life. Lancet Neurol 2009;8:1019–30
  • Thurman DJ, Beghi E, Begley CE, et al. Standards for epidemiologic studies and surveillance of epilepsy. Epilepsia 2011;52:2–26
  • Vyas TK, Babbar AK, Sharma RK, et al. Intranasal mucoadhesive microemulsions of clonazepam: preliminary studies on brain targeting. J Pharm Sci 2006;95:570–80
  • Vyas TK, Shahiwala A, Marathe S, Misra A. Intranasal drug delivery for brain targeting. Curr Drug Deliv 2005;2:165–75
  • Gaskin PL, Alexander SP, Fone KC. Neonatal phencyclidine administration and post-weaning social isolation as a dual-hit model of ‘schizophrenia-like’ behaviour in the rat. Psychopharmacology (Berlin) 2014;231:2533–45
  • Stachowiak MK, Kucinski A, Curl R, et al. Schizophrenia: a neurodevelopment disorder-integrative genomic hypothesis and therapeutic implications from a transgenic mouse model. Schizophr Res 2013;143:367–76
  • Sa YK, Yang H, Jung HK, et al. Olanzapine-induced diabetic ketoacidosis and neuroleptic malignant syndrome with rhabdomyolysis: a case report. Endocrinol Metab 2013;28:70–5
  • Seju U, Kumar A, Sawant KK. Development and evaluation of olanzapine-loaded PLGA nanoparticles for nose-to-brain delivery: in vitro and in vivo studies. Acta Biomater 2011;7:4169–76
  • Chalikwar SS, Mena BS, Pardeshi CV, et al. Self-assembled, chitosan grafted PLGA nanoparticles for intranasal delivery: design, development and ex vivo characterization. Polym Plastic Technol Eng 2013;52:368–80
  • Kumar M, Pathak K, Misra A. Formulation and characterization of nanoemulsion-based drug delivery system of risperidone. Drug Dev Ind Pharm 2009;35:387–95
  • Patel S, Chavhan S, Soni H, et al. Brain targeting of risperidone-loaded solid lipid nanoparticles by intranasal route. J Drug Target 2011;19:468–74
  • Piazza J, Hoare T, Molinaro L, et al. Haloperidol-loaded intranasally administered lectin functionalized poly(ethylene glycol)–block-poly(D,L)-lactic-co-glycolic acid (PEG–PLGA) nanoparticles for the treatment of schizophrenia. Eur J Pharm Biopharm 2014;87:30–9
  • Chen J, Zhang C, Liu Q, et al. Solanum tuberosum lectin-conjugated PLGA nanoparticles for nose-to-brain delivery: in vivo and in vitro evaluations. J Drug Target 2011;2:1–11
  • Ferrari AJ, Charlson FJ, Norman R, et al. The epidemiological modelling of major depressive disorder: application for the Global Burden of Disease Study 2010. PLoS One 2013;8:69637
  • Charlson FJ, Ferrari AJ, Somerville AJ, et al. The epidemiological modelling of dysthymia: application for the Global Burden of Disease 2010 Study. J Affect Disord 2013;151:111–20
  • Rosenzweig-Lipson S, Beyer CE, Hughes ZA, et al. Differentiating antidepressants of the future: efficacy and safety. Pharmacol Ther 2007;113:134–53
  • Al-Ghananeem AM, Smith M, Coronel ML, Tran H. Advances in brain targeting and drug delivery of anti-HIV therapeutic agents. Exp Opin Drug Deliv 2013;10:973–85
  • Borm PJ, Robbins D, Haubold S, et al. The potential risks of nanomaterials: a review carried out for ECETOC. Part Fibre Toxicol 2006;3:11
  • Medina C, Santos-Martinez MJ, Radomski A, et al. Nanoparticles: pharmacological and toxicological significance. Br J Pharmacol 2007;150:552–8
  • Oberdorster E. Manufactured nanomaterials (fullerenes, C60) induce oxidative stress in the brain of juvenile largemouth bass. Environ Health Perspect 2004;112:1058–62
  • Sharma HS. Nanoneuroscience: emerging concepts on nanoneurotoxicity and nanoneuroprotection. Nanomedicine 2007;2:753–8
  • Sharma HS, Sharma A. Nanoparticles aggravate heat stress induced cognitive deficits, blood–brain barrier disruption, edema formation and brain pathology. Prog Brain Res 2007;162:245–73
  • Greene LA, Tischler AS. Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA 1976;73:2424–8
  • Long TC, Tajuba J, Sama P, et al. Nanosize titanium dioxide stimulates reactive oxygen species in brain microglia and damages neurons in vitro. Environ Health Perspect 2007;115:1631–7
  • Trapani A, De Giglio E, Cafagna D, et al. Characterization and evaluation of chitosan nanoparticles for dopamine brain delivery. Int J Pharm 2011;419:296–307
  • Hu YL, Gao JQ. Potential neurotoxicity of nanoparticles. Int J Pharm 2010;394:115–21
  • Rahman MF, Wang J, Patterson TA, et al. Expression of genes related to oxidative stress in the mouse brain after exposure to silver-25 nanoparticles. Toxicol Lett 2009;187:15–21
  • Ma L, Liu J, Li N, et al. Oxidative stress in the brain of mice caused by translocated nanoparticulate TiO2 delivered to the abdominal cavity. Biomaterials 2009;31:99–105
  • Kircher MF, Mahmood U, King RS, et al. A multimodal nanoparticle for preoperative magnetic resonance imaging and intraoperative optical brain tumor delineation. Cancer Res 2003;63:8122–5
  • Tin TW, Yamamoto S, Ahmed S, et al. Brain cytokine and chemokine mRNA expression in mice induced by intranasal instillation with ultrafine carbon black. Toxicol Lett 2006;163:153–60
  • Maurer-Jones MA, Bantz KC, Love SA, et al. Toxicity of therapeutic nanoparticles. Nanomedicine 2009;4:219–41
  • Xia T, Kovochich M, Liong M, et al. Cationic polystyrene nanosphere toxicity depends on cell-specific endocytic and mitochondrial injury pathways. ACS Nano 2008;2:85–96
  • Yi X, Manickam DS, Brynskikh A, Kabanov AV. Agile delivery of protein therapeutics to CNS. J Control Release 2014;190:637–63
  • Graff CL, Pollack GM. P-Glycoprotein attenuates brain uptake of substrates after nasalinstillation. Pharm Res 2003;20:1225–30
  • Reynoso-Camacho R, Gonzalez DME, Loarca-Pina G. Purification and acute toxicity of a lectin extracted from tepary bean (Phaseolusacutifolius). Food Chem Toxicol 2003;41:21–7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.