210
Views
0
CrossRef citations to date
0
Altmetric
Review

Nanotherapeutic Strategies for the Treatment of Alzheimer's Disease

, &
Pages 177-195 | Published online: 18 Feb 2015

References

  • Addressing global dementia (Editorial) . Lancet383 (9936), 2185 (2014).
  • Hardy J Selkoe DJ . The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science297 (5580), 353–356 (2002).
  • Mondragon-Rodriguez S Perry G Zhu X et al. Phosphorylation of tau protein as the link between oxidative stress, mitochondrial dysfunction, and connectivity failure: implications for Alzheimer's disease. Oxid. Med. Cell. Longev.940603 (2013).
  • Manafirad A Farzadfar F Habibi L et al. Is Amyloid-β an innocent bystander and marker in Alzheimer's disease? Is the liability of multivalent cation homeostasis and its influence on amyloid-β function the real mechanism? J. Alzheimers Dis. 42 (1), 69–85 (2014).
  • Huang Y Mucke L . Alzheimer mechanisms and therapeutic strategies. Cell148 (6), 1204–1222 (2012).
  • Spuch C Saida O Navarro C . Advances in the treatment of neurodegenerative disorders employing nanoparticles. Recent Pat. Drug Deliv. Formul.6 (1), 2–18 (2012).
  • Foley P . Lipids in Alzheimer's disease: A century-old story. Biochim. Biophys. Acta1801 (8), 750–753 (2010).
  • Haass C Kaether C Thinakaran G et al. Trafficking and proteolytic processing of APP. Cold Spring Harb. Perspect. Med.2 (5), a6270 (2012).
  • Wang Y Xia Z Xu JR et al. Alpha-mangostin, a polyphenolic xanthone derivative from mangosteen, attenuates β-amyloid oligomers-induced neurotoxicity by inhibiting amyloid aggregation. Neuropharmacology62 (2), 871–881 (2012).
  • Gilbert BJ . The role of amyloid-β in the pathogenesis of Alzheimer's disease. J. Clin. Pathol.66 (5), 362–366 (2013).
  • Blennow K de Leon MJ Zetterberg H . Alzheimer's disease. Lancet368 (9533), 387–403 (2006).
  • Spillantini MG Goedert M . Tau pathology and neurodegeneration. Lancet Neurol.12 (6), 609–622 (2013).
  • Bartus RT . On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp. Neurol.163 (2), 495–529 (2000).
  • Camps P Munoz-Torrero D . Cholinergic drugs in pharmacotherapy of Alzheimer's disease. Mini Rev. Med. Chem.2 (1), 11–25 (2002).
  • Padurariu M Ciobica A Lefter R et al. The oxidative stress hypothesis in Alzheimer's disease. Psychiatr. Danub.25 (4), 401–409 (2013).
  • Massaad CA . Neuronal and vascular oxidative stress in Alzheimer's disease. Curr. Neuropharmacol.9 (4), 662–673 (2011).
  • Tiiman A Palumaa P Tougu V . The missing link in the amyloid cascade of Alzheimer's disease–metal ions. Neurochem. Int.62 (4), 367–378 (2013).
  • Barcia E Salama A Fernandez-Carballido A et al. Protective effects of clioquinol on human neuronal-like cells: a new formulation of clioquinol-loaded PLGA microspheres for Alzheimer's disease. J. Drug Target19 (8), 637–646 (2011).
  • de Boer AG Gaillard PJ . Strategies to improve drug delivery across the blood–brain barrier. Clin. Pharmacokinet.46 (7), 553–576 (2007).
  • Ronaldson PT Davis TP . Blood–brain barrier integrity and glial support: mechanisms that can be targeted for novel therapeutic approaches in stroke. Curr. Pharm. Des.18 (25), 3624–3644 (2012).
  • Reese TS Karnovsky MJ . Fine structural localization of a blood–brain barrier to exogenous peroxidase. J. Cell Biol.34 (1), 207–217 (1967).
  • Lai CH Kuo KH . The critical component to establish in vitro BBB model: pericyte. Brain Res. Brain Res. Rev.50 (2), 258–265 (2005).
  • Abbott NJ Ronnback L Hansson E . Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci.7 (1), 41–53 (2006).
  • Mahringer A Ott M Reimold I et al. The ABC of the blood–brain barrier-regulation of drug efflux pumps. Curr. Pharm. Des.17 (26), 2762–2770 (2011).
  • Bendayan R Ronaldson PT Gingras D et al. In situ localization of P-glycoprotein (ABCB1) in human and rat brain. J. Histochem. Cytochem.54 (10), 1159–1167 (2006).
  • Sanchez-Covarrubias L Slosky LM Thompson BJ et al. Transporters at CNS barrier sites: obstacles or opportunities for drug delivery? Curr. Pharm. Des. 20 (10), 1422–1449 (2014).
  • Wong HL Wu XY Bendayan R . Nanotechnological advances for the delivery of CNS therapeutics. Adv. Drug Deliv. Rev.64 (7), 686–700 (2012).
  • Ronaldson PT Finch JD Demarco KM et al. Inflammatory pain signals an increase in functional expression of organic anion transporting polypeptide 1a4 at the blood–brain barrier. J. Pharmacol. Exp. Ther.336 (3), 827–839 (2011).
  • Pardridge WM . Drug targeting to the brain. Pharm. Res.24 (9), 1733–1744 (2007).
  • Alafuzoff I Adolfsson R Bucht G et al. Albumin and immunoglobulin in plasma and cerebrospinal fluid, and blood-cerebrospinal fluid barrier function in patients with dementia of Alzheimer type and multi-infarct dementia. J. Neurol. Sci.60 (3), 465–472 (1983).
  • Banks WA Kumar VB Farr SA et al. Impairments in brain-to-blood transport of amyloid-β and reabsorption of cerebrospinal fluid in an animal model of Alzheimer's disease are reversed by antisense directed against amyloid-β protein precursor. J. Alzheimers Dis.23 (4), 599–605 (2011).
  • Erickson MA Banks WA . Blood–brain barrier dysfunction as a cause and consequence of Alzheimer's disease. J. Cereb. Blood Flow Metab.33 (10), 1500–1513 (2013).
  • Lacalle-Aurioles M Mateos-Perez JM Guzman-De-Villoria JA et al. Cerebral blood flow is an earlier indicator of perfusion abnormalities than cerebral blood volume in Alzheimer's disease. J. Cereb. Blood Flow Metab.34 (4), 654–659 (2014).
  • Miyakawa T Shimoji A Kuramoto R et al. The relationship between senile plaques and cerebral blood vessels in Alzheimer's disease and senile dementia. Morphological mechanism of senile plaque production. Virchows Arch. B, Cell Pathol.40 (2), 121–129 (1982).
  • Jancso G Domoki F Santha P et al. β-amyloid (1–42) peptide impairs blood-brain barrier function after intracarotid infusion in rats. Neurosci. Lett.253 (2), 139–141 (1998).
  • Kook SY Hong HS Moon M et al. Aβ1–42-RAGE interaction disrupts tight junctions of the blood–brain barrier via Ca2+-calcineurin signaling. J. Neurosci.32 (26), 8845–8854 (2012).
  • Do TM Alata W Dodacki A et al. Altered cerebral vascular volumes and solute transport at the blood–brain barriers of two transgenic mouse models of Alzheimer's disease. Neuropharmacology81, 311–317 (2014).
  • Burgmans S van de Haar HJ Verhey FR et al. Amyloid-β interacts with blood–brain barrier function in dementia: a systematic review. J. Alzheimers Dis.35 (4), 859–873 (2013).
  • Hartz AM Bauer B Soldner EL et al. Amyloid-β contributes to blood–brain barrier leakage in transgenic human amyloid precursor protein mice and in humans with cerebral amyloid angiopathy. Stroke43 (2), 514–523 (2012).
  • Donahue JE Flaherty SL Johanson CE et al. RAGE, LRP-1, and amyloid-β protein in Alzheimer's disease. Acta Neuropathol.112 (4), 405–415 (2006).
  • Deane R Du Yan S Submamaryan RK et al. RAGE mediates amyloid-β peptide transport across the blood–brain barrier and accumulation in brain. Nat. Med.9 (7), 907–913 (2003).
  • Deane R Zlokovic BV . Role of the blood–brain barrier in the pathogenesis of Alzheimer's disease. Curr. Alzheimer Res.4 (2), 191–197 (2007).
  • Hartz AM Miller DS Bauer B . Restoring blood–brain barrier P-glycoprotein reduces brain amyloid-β in a mouse model of Alzheimer's disease. Mol. Pharmacol.77 (5), 715–723 (2010).
  • Miller DS Bauer B Hartz AM . Modulation of P-glycoprotein at the blood–brain barrier: opportunities to improve central nervous system pharmacotherapy. Pharmacol. Rev.60 (2), 196–209 (2008).
  • Cirrito JR Deane R Fagan AM et al. P-glycoprotein deficiency at the blood–brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model. J. Clin. Invest.115 (11), 3285–3290 (2005).
  • Bauer B Hartz AM Miller DS . Tumor necrosis factor alpha and endothelin-1 increase P-glycoprotein expression and transport activity at the blood–brain barrier. Mol. Pharmacol.71 (3), 667–675 (2007).
  • McDade E Kim A James J et al. Cerebral perfusion alterations and cerebral amyloid in autosomal dominant Alzheimer disease. Neurology83 (8), 710–717 (2014).
  • Banks WA . Drug delivery to the brain in Alzheimer's disease: consideration of the blood–brain barrier. Adv. Drug Deliv. Rev.64 (7), 629–639 (2012).
  • Pardridge WM . The blood–brain barrier and neurotherapeutics. NeuroRx2 (1), 1–2 (2005).
  • Gabathuler R . Approaches to transport therapeutic drugs across the blood–brain barrier to treat brain diseases. Neurobiol. Dis.37 (1), 48–57 (2010).
  • Fung LK Shin M Tyler B et al. Chemotherapeutic drugs released from polymers: distribution of 1,3-bis(2-chloroethyl)-1-nitrosourea in the rat brain. Pharm. Res.13 (5), 671–682 (1996).
  • Barua NU Gill SS Love S . Convection-enhanced drug delivery to the brain: therapeutic potential and neuropathological considerations. Brain Pathol.24 (2), 117–127 (2013).
  • Barua NU Miners JS Bienemann AS et al. Convection-enhanced delivery of neprilysin: a novel amyloid-β-degrading therapeutic strategy. J. Alzheimers Dis.32 (1), 43–56 (2012).
  • Peng KA Masliah E . Lentivirus-expressed siRNA vectors against Alzheimer disease. Methods Mol. Biol.614, 215–224 (2010).
  • Wahlberg LU Lind G Almqvist PM et al. Targeted delivery of nerve growth factor via encapsulated cell biodelivery in Alzheimer disease: a technology platform for restorative neurosurgery. J. Neurosurg.117 (2), 340–347 (2012).
  • Perlstein B Ram Z Daniels D et al. Convection-enhanced delivery of maghemite nanoparticles: increased efficacy and MRI monitoring. Neuro. Oncol.10 (2), 153–161 (2008).
  • MacKay JA Deen DF Szoka FJ . Distribution in brain of liposomes after convection enhanced delivery; modulation by particle charge, particle diameter, and presence of steric coating. Brain Res.1035 (2), 139–153 (2005).
  • Fortin D Gendron C Boudrias M et al. Enhanced chemotherapy delivery by intraarterial infusion and blood–brain barrier disruption in the treatment of cerebral metastasis. Cancer109 (4), 751–760 (2007).
  • Hynynen K McDannold N Vykhodtseva N et al. Focal disruption of the blood-brain barrier due to 260-kHz ultrasound bursts: a method for molecular imaging and targeted drug delivery. J. Neurosurg.105 (3), 445–454 (2006).
  • Yao L Song Q Bai W et al. Facilitated brain delivery of poly (ethylene glycol)-poly (lactic acid) nanoparticles by microbubble-enhanced unfocused ultrasound. Biomaterials35 (10), 3384–3395 (2014).
  • Pardridge WM . Blood–brain barrier drug targeting: the future of brain drug development. Mol. Interv.3 (2), 90–105 (2003).
  • Patel MM Goyal BR Bhadada SV et al. Getting into the brain: approaches to enhance brain drug delivery. CNS Drugs.23 (1), 35–58 (2009).
  • Mittapalli RK Manda VK Adkins CE et al. Exploiting nutrient transporters at the blood–brain barrier to improve brain distribution of small molecules. Ther. Deliv.1 (6), 775–784 (2010).
  • Cornford EM Young D Paxton JW et al. Melphalan penetration of the blood–brain barrier via the neutral amino acid transporter in tumor-bearing brain. Cancer Res.52 (1), 138–143 (1992).
  • Mena I Cotzias GC . Protein intake and treatment of Parkinson's disease with levodopa. N. Engl. J. Med.292 (4), 181–184 (1975).
  • Uchino H Kanai Y Kim DK et al. Transport of amino acid-related compounds mediated by L-type amino acid transporter 1 (LAT1): insights into the mechanisms of substrate recognition. Mol. Pharmacol.61 (4), 729–737 (2002).
  • Schroeder U Sommerfeld P Ulrich S et al. Nanoparticle technology for delivery of drugs across the blood–brain barrier. J. Pharm. Sci.87 (11), 1305–1307 (1998).
  • Herve F Ghinea N Scherrmann JM . CNS delivery via adsorptive transcytosis. AAPS J.10 (3), 455–472 (2008).
  • Lu W . Adsorptive-mediated brain delivery systems. Curr. Pharm. Biotechnol.13 (12), 2340–2348 (2012).
  • Lockman PR Koziara JM Mumper RJ et al. Nanoparticle surface charges alter blood–brain barrier integrity and permeability. J. Drug Target12 (9–10), 635–641 (2004).
  • Dhuria SV Hanson LR Frey WN . Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J. Pharm. Sci.99 (4), 1654–1673 (2010).
  • Chen Y Zhao Y Dai CL et al. Intranasal insulin restores insulin signaling, increases synaptic proteins, and reduces Aβ level and microglia activation in the brains of 3xTg-AD mice. Exp. Neurol.261C, 610–619 (2014).
  • Yarchoan M Arnold SE . Repurposing diabetes drugs for brain insulin resistance in Alzheimer disease. Diabetes63 (7), 2253–2261 (2014).
  • Aguzzi A O'Connor T . Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat. Rev. Drug Discov.9 (3), 237–248 (2010).
  • Wong GT Manfra D Poulet FM et al. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits β-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J. Biol. Chem.279 (13), 12876–12882 (2004).
  • Luo Y Bolon B Kahn S et al. Mice deficient in BACE1, the Alzheimer's β-secretase, have normal phenotype and abolished β-amyloid generation. Nat. Neurosci.4 (3), 231–232 (2001).
  • Alvarez-Erviti L Seow Y Yin H et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol.29 (4), 341–345 (2011).
  • Heffern MC Velasco PT Matosziuk LM et al. Modulation of amyloid-β aggregation by histidine-coordinating cobalt(III) schiff base complexes. Chembiochem.15 (11), 1584–1589 (2014).
  • Churches QI Caine J Cavanagh K et al. Naturally occurring polyphenolic inhibitors of amyloid-β aggregation. Bioorg. Med. Chem. Lett.24 (14), 3108–3112 (2014).
  • Takahashi T Mihara H . Peptide and protein mimetics inhibiting amyloid β-peptide aggregation. Acc. Chem. Res.41 (10), 1309–1318 (2008).
  • Pan H Soman NR Schlesinger PH et al. Cytolytic peptide nanoparticles (‘NanoBees’) for cancer therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol.3 (3), 318–327 (2011).
  • Liu L Xu K Wang H et al. Self-assembled cationic peptide nanoparticles as an efficient antimicrobial agent. Nat. Nanotechnol.4 (7), 457–463 (2009).
  • Li M Xu C Wu L et al. Self-assembled peptide-polyoxometalate hybrid nanospheres: two in one enhances targeted inhibition of amyloid β-peptide aggregation associated with Alzheimer's disease. Small9 (20), 3455–3461 (2013).
  • Shen Y Yu LC . Potential protection of curcumin against hypoxia-induced decreases in β-III tubulin content in rat prefrontal cortical neurons. Neurochem. Res.33 (10), 2112–2117 (2008).
  • Reinke AA Gestwicki JE . Structure-activity relationships of amyloid β-aggregation inhibitors based on curcumin: influence of linker length and flexibility. Chem. Biol. Drug Des.70 (3), 206–215 (2007).
  • Lim GP Chu T Yang F et al. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci.21 (21), 8370–8377 (2001).
  • Sharma RA Gescher AJ Steward WP . Curcumin: the story so far. Eur. J. Cancer41 (13), 1955–1968 (2005).
  • Cheng KK Yeung CF Ho SW et al. Highly stabilized curcumin nanoparticles tested in an in vitro blood–brain barrier model and in Alzheimer's disease Tg2576 mice. AAPS J.15 (2), 324–336 (2013).
  • Mathew A Fukuda T Nagaoka Y et al. Curcumin loaded-PLGA nanoparticles conjugated with Tet-1 peptide for potential use in Alzheimer's disease. PLoS ONE7 (3), e32616 (2012).
  • Kogan MJ Bastus NG Amigo R et al. Nanoparticle-mediated local and remote manipulation of protein aggregation. Nano. Lett.6 (1), 110–115 (2006).
  • Prades R Guerrero S Araya E et al. Delivery of gold nanoparticles to the brain by conjugation with a peptide that recognizes the transferrin receptor. Biomaterials33 (29), 7194–7205 (2012).
  • Lee JH Engler JA Collawn JF et al. Receptor mediated uptake of peptides that bind the human transferrin receptor. Eur. J. Biochem.268 (7), 2004–2012 (2001).
  • Mawuenyega KG Sigurdson W Ovod V et al. Decreased clearance of CNS β-amyloid in Alzheimer's disease. Science330 (6012), 1774 (2010).
  • Cramer PE Cirrito JR Wesson DW et al. ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. Science335 (6075), 1503–1506 (2012).
  • Prada CM Garcia-Alloza M Betensky RA et al. Antibody-mediated clearance of amyloid-β peptide from cerebral amyloid angiopathy revealed by quantitative in vivo imaging. J. Neurosci.27 (8), 1973–1980 (2007).
  • Sagare A Deane R Bell RD et al. Clearance of amyloid-β by circulating lipoprotein receptors. Nat. Med.13 (9), 1029–1031 (2007).
  • Song Q Huang M Yao L et al. Lipoprotein-based nanoparticles rescue the memory loss of mice with Alzheimer's disease by accelerating the clearance of amyloid-β. ACS Nano8 (3), 2345–2359 (2014).
  • Lee CY Tse W Smith JD et al. Apolipoprotein E promotes β-amyloid trafficking and degradation by modulating microglial cholesterol levels. J. Biol. Chem.287 (3), 2032–2044 (2012).
  • Canovi M Markoutsa E Lazar AN et al. The binding affinity of anti-Aβ1–42 MAb-decorated nanoliposomes to Aβ1–42 peptides in vitro and to amyloid deposits in post-mortem tissue. Biomaterials32 (23), 5489–5497 (2011).
  • Wilcock DM Rojiani A Rosenthal A et al. Passive amyloid immunotherapy clears amyloid and transiently activates microglia in a transgenic mouse model of amyloid deposition. J. Neurosci.24 (27), 6144–6151 (2004).
  • DeMattos RB Bales KR Cummins DJ et al. Peripheral anti-Aβ antibody alters CNS and plasma Aβ clearance and decreases brain Aβ burden in a mouse model of Alzheimer's disease. Proc. Natl Acad. Sci. USA98 (15), 8850–8855 (2001).
  • Zhang C Wan X Zheng X et al. Dual-functional nanoparticles targeting amyloid plaques in the brains of Alzheimer's disease mice. Biomaterials35 (1), 456–465 (2014).
  • Bartnik D Funke SA Andrei-Selmer LC et al. Differently selected D-enantiomeric peptides act on different Aβ species. Rejuvenation Res.13 (2–3), 202–205 (2010).
  • Li J Feng L Fan L et al. Targeting the brain with PEG-PLGA nanoparticles modified with phage-displayed peptides. Biomaterials32 (21), 4943–4950 (2011).
  • Dong J Atwood CS Anderson VE et al. Metal binding and oxidation of amyloid-β within isolated senile plaque cores: Raman microscopic evidence. Biochemistry42 (10), 2768–2773 (2003).
  • Ehmann WD Markesbery WR Alauddin M et al. Brain trace elements in Alzheimer's disease. Neurotoxicology7 (1), 195–206 (1986).
  • Adlard PA Bush AI . Metals and Alzheimer's disease. J. Alzheimers Dis.10 (2–3), 145–163 (2006).
  • Lovell MA Robertson JD Teesdale WJ et al. Copper, iron and zinc in Alzheimer's disease senile plaques. J. Neurol. Sci.158 (1), 47–52 (1998).
  • Bush AI . The metallobiology of Alzheimer's disease. Trends Neurosci.26 (4), 207–214 (2003).
  • Bolognin S Messori L Drago D et al. Aluminum, copper, iron and zinc differentially alter amyloid-Aβ(1–42) aggregation and toxicity. Int. J. Biochem. Cell Biol.43 (6), 877–885 (2011).
  • Jomova K Vondrakova D Lawson M et al. Metals, oxidative stress and neurodegenerative disorders. Mol. Cell Biochem.345 (1–2), 91–104 (2010).
  • Cherny RA Barnham KJ Lynch T et al. Chelation and intercalation: complementary properties in a compound for the treatment of Alzheimer's disease. J. Struct. Biol.130 (2–3), 209–216 (2000).
  • Liu G Garrett MR Men P et al. Nanoparticle and other metal chelation therapeutics in Alzheimer disease. Biochim. Biophys. Acta1741 (3), 246–252 (2005).
  • Liu G Men P Kudo W et al. Nanoparticle-chelator conjugates as inhibitors of amyloid-β aggregation and neurotoxicity: a novel therapeutic approach for Alzheimer disease. Neurosci. Lett.455 (3), 187–190 (2009).
  • Francis PT Palmer AM Snape M et al. The cholinergic hypothesis of Alzheimer's disease: a review of progress. J. Neurol. Neurosurg. Psychiatr.66 (2), 137–147 (1999).
  • Standridge JB . Pharmacotherapeutic approaches to the treatment of Alzheimer's disease. Clin. Ther.26 (5), 615–630 (2004).
  • Raschetti R Albanese E Vanacore N et al. Cholinesterase inhibitors in mild cognitive impairment: a systematic review of randomised trials. PLoS Med.4 (11), e338 (2007).
  • Tse FL Laplanche R . Absorption, metabolism, and disposition of [14C]SDZ ENA 713, an acetylcholinesterase inhibitor, in minipigs following oral, intravenous, and dermal administration. Pharm. Res.15 (10), 1614–1620 (1998).
  • Mutlu NB Degim Z Yilmaz S et al. New perspective for the treatment of Alzheimer diseases: liposomal rivastigmine formulations. Drug Dev. Ind. Pharm.37 (7), 775–789 (2011).
  • Garberg P Ball M Borg N et al. In vitro models for the blood-brain barrier. Toxicol. In vitro.19 (3), 299–334 (2005).
  • Ismail MF Elmeshad AN Salem NA . Potential therapeutic effect of nanobased formulation of rivastigmine on rat model of Alzheimer's disease. Int. J. Nanomedicine8, 393–406 (2013).
  • Fujimura M Usuki F Kawamura M et al. Inhibition of the Rho/ROCK pathway prevents neuronal degeneration in vitro and in vivo following methylmercury exposure. Toxicol. Appl. Pharmacol.250 (1), 1–9 (2011).
  • Herskowitz JH Seyfried NT Gearing M et al. Rho kinase II phosphorylation of the lipoprotein receptor LR11/SORLA alters amyloid-β production. J. Biol. Chem.286 (8), 6117–6127 (2011).
  • Yang P Wen HZ Zhang JH . Expression of a dominant-negative Rho-kinase promotes neurite outgrowth in a microenvironment mimicking injured central nervous system. Acta Pharmacol. Sin.31 (5), 531–539 (2010).
  • Liu Y Liu Z Wang Y et al. Investigation of the performance of PEG-PEI/ROCK-II-siRNA complexes for Alzheimer's disease in vitro. Brain Res.1490, 43–51 (2013).
  • Craig MC Murphy DG . Alzheimer's disease in women. Best Pract. Res. Clin. Obstet. Gynaecol.23 (1), 53–61 (2009).
  • Sherwin BB . Can estrogen keep you smart? Evidence from clinical studies. J. Psychiatry Neurosci.24 (4), 315–321 (1999).
  • Ohkura T Isse K Akazawa K et al. Long-term estrogen replacement therapy in female patients with dementia of the Alzheimer type: 7 case reports. Dementia6 (2), 99–107 (1995).
  • Simpkins JW Singh M . More than a decade of estrogen neuroprotection. Alzheimers Dement.4 (1 Suppl 1), S131–S136 (2008).
  • Vasudevan N Pfaff DW . Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol.29 (2), 238–257 (2008).
  • Mittal G Carswell H Brett R et al. Development and evaluation of polymer nanoparticles for oral delivery of estradiol to rat brain in a model of Alzheimer's pathology. J. Control. Release150 (2), 220–228 (2011).
  • Krol S Macrez R Docagne F et al. Therapeutic benefits from nanoparticles: the potential significance of nanoscience in diseases with compromise to the blood–brain barrier. Chem. Rev.113 (3), 1877–1903 (2013).
  • Wu X Tan Y Mao H et al. Toxic effects of iron oxide nanoparticles on human umbilical vein endothelial cells. Int. J. Nanomedicine5, 385–399 (2010).
  • Onoue S Yamada S Chan HK . Nanodrugs: pharmacokinetics and safety. Int. J. Nanomedicine9, 1025–1037 (2014).
  • Frick R Muller-Edenborn B Schlicker A et al. Comparison of manganese oxide nanoparticles and manganese sulfate with regard to oxidative stress, uptake and apoptosis in alveolar epithelial cells. Toxicol. Lett.205 (2), 163–172 (2011).
  • Ginzburg VV Balijepalli S . Modeling the thermodynamics of the interaction of nanoparticles with cell membranes. Nano Lett.7 (12), 3716–3722 (2007).
  • Hutter E Boridy S Labrecque S et al. Microglial response to gold nanoparticles. ACS Nano4 (5), 2595–2606 (2010).

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.