Mesoporous silica nanoparticles for treating spinal cord injury

An estimated 12,000 new cases of spinal cord injury (SCI) occur every year in the United States. A small oxidative molecule responsible for secondary injury, acrolein, is an important target in SCI. Acrolein attacks essential proteins and lipids, creating a feed-forward loop of oxidative stress in both the primary injury area and the surrounding areas. A small molecule used and FDA-approved for hypertension, hydralazine, has been found to "scavenge" acrolein after injury, but its delivery and short half-life, as well as its hypertension effects, hinder its application for SCI. Nanomedical systems broaden the range of therapeutic availability and efficacy over conventional medicine. They allow for targeted delivery of therapeutic molecules to tissues of interest, reducing side effects of untargeted therapies in unwanted areas. Nanoparticles made from silica form porous networks that can carry therapeutic molecules throughout the body. To attenuate the acrolein cascade and improve therapeutic availability, we have used a one-step, modified Stober method to synthesize two types of silica nanoparticles. Both particles are “stealth-coated” with poly(ethylene) glycol (PEG) (to minimize interactions with the immune system and to increase circulation time), which is also a therapeutic agent for SCI by facilitating membrane repair. One nanoparticle type contains an amine-terminal PEG (SiNP-mPEG-Am) and the other possesses a terminal hydrazide group (SiNP-mPEG-Hz). The former allows for exploration of hydralazine delivery, loading, and controlled release. The latter group has the ability to react with acrolein, allowing the nanoparticle to scavenge directly. The nanoparticles have been characterized and are being explored using neuronal PC-12 cells in vitro, demonstrating the potential of novel silica nanoparticles for use in attenuating secondary injury after SCI.

[1]  P. Couvreur,et al.  Stealth PEGylated polycyanoacrylate nanoparticles for intravenous administration and splenic targeting. , 1999, Journal of controlled release : official journal of the Controlled Release Society.

[2]  Wolfhard Semmler,et al.  Silica- and alkoxysilane-coated ultrasmall superparamagnetic iron oxide particles: a promising tool to label cells for magnetic resonance imaging. , 2007, Langmuir : the ACS journal of surfaces and colloids.

[3]  Riyi Shi,et al.  Chitosan produces potent neuroprotection and physiological recovery following traumatic spinal cord injury , 2010, Journal of Experimental Biology.

[4]  Takuro Niidome,et al.  PEG-modified gold nanorods with a stealth character for in vivo applications. , 2006, Journal of controlled release : official journal of the Controlled Release Society.

[5]  R. Shi,et al.  Acrolein induces axolemmal disruption, oxidative stress, and mitochondrial impairment in spinal cord tissue , 2004, Neurochemistry International.

[6]  A. Karydas,et al.  PLGA-mPEG nanoparticles of cisplatin: in vitro nanoparticle degradation, in vitro drug release and in vivo drug residence in blood properties. , 2002, Journal of controlled release : official journal of the Controlled Release Society.

[7]  Jianlin Shi,et al.  The effect of PEGylation of mesoporous silica nanoparticles on nonspecific binding of serum proteins and cellular responses. , 2010, Biomaterials.

[8]  R. Shi,et al.  Acrolein scavenging: a potential novel mechanism of attenuating oxidative stress following spinal cord injury , 2009, Journal of neurochemistry.

[9]  Hiroaki Sai,et al.  Ultrasmall sub-10 nm near-infrared fluorescent mesoporous silica nanoparticles. , 2012, Journal of the American Chemical Society.

[10]  R. Shi,et al.  Acrolein inflicts axonal membrane disruption and conduction loss in isolated guinea-pig spinal cord , 2002, Neuroscience.

[11]  Yaping Li,et al.  In vivo biodistribution and urinary excretion of mesoporous silica nanoparticles: effects of particle size and PEGylation. , 2011, Small.

[12]  Laetitia Gonzalez,et al.  Size-dependent cytotoxicity of monodisperse silica nanoparticles in human endothelial cells. , 2009, Small.

[13]  D. Burke,et al.  Incidence rates and populations at risk for spinal cord injury: A regional study , 2001, Spinal Cord.

[14]  R. Shi,et al.  Accumulation of Acrolein–Protein Adducts after Traumatic Spinal Cord Injury , 2005, Neurochemical Research.

[15]  James F. Leary,et al.  Biosensor-controlled gene therapy/drug delivery with nanoparticles for nanomedicine , 2005, SPIE BiOS.

[16]  E. Niki,et al.  Acrolein Is a Product of Lipid Peroxidation Reaction , 1998, The Journal of Biological Chemistry.

[17]  R. Shi,et al.  Neuroprotection from secondary injury by polyethylene glycol requires its internalization , 2007, Journal of Experimental Biology.

[18]  R. Shi,et al.  Hydralazine rescues PC12 cells from acrolein‐mediated death , 2006, Journal of neuroscience research.

[19]  Raoul Kopelman,et al.  Room-temperature preparation and characterization of poly (ethylene glycol)-coated silica nanoparticles for biomedical applications. , 2003, Journal of biomedical materials research. Part A.

[20]  Zhenkun Zhang,et al.  Synthesis of poly(ethylene glycol) (PEG)-grafted colloidal silica particles with improved stability in aqueous solvents. , 2007, Journal of colloid and interface science.

[21]  B. Duerstock,et al.  Hydralazine inhibits compression and acrolein‐mediated injuries in ex vivo spinal cord , 2007, Journal of neurochemistry.

[22]  M. Radomski,et al.  Amorphous silica nanoparticles trigger nitric oxide/peroxynitrite imbalance in human endothelial cells: inflammatory and cytotoxic effects , 2011, International journal of nanomedicine.

[23]  Weihong Tan,et al.  Optimization of dye-doped silica nanoparticles prepared using a reverse microemulsion method. , 2004, Langmuir : the ACS journal of surfaces and colloids.

[24]  Libang Feng,et al.  Preparation of poly(ethylene glycol)-grafted silica nanoparticles using a facile esterification condensation method , 2009 .

[25]  S. Aiba,et al.  Synthesis of a chitosan-dendrimer hybrid and its biodegradation. , 2003, Biomacromolecules.

[26]  R. Shi,et al.  Anti-acrolein treatment improves behavioral outcome and alleviates myelin damage in experimental autoimmune enchephalomyelitis mouse , 2011, Neuroscience.

[27]  Qiang Cai,et al.  Dilute solution routes to various controllable morphologies of MCM-41 silica with a basic medium , 2001 .

[28]  Michael J Sailor,et al.  Biodegradable luminescent porous silicon nanoparticles for in vivo applications. , 2009, Nature materials.

[29]  R. Shi,et al.  Critical role of acrolein in secondary injury following ex vivo spinal cord trauma , 2008, Journal of neurochemistry.

[30]  Weihong Tan,et al.  Surface modification of silica nanoparticles to reduce aggregation and nonspecific binding. , 2006, Langmuir : the ACS journal of surfaces and colloids.

[31]  T. Heinonen,et al.  Decrease of reduced glutathione in isolated rat hepatocytes caused by acrolein, acrylonitrile, and the thermal degradation products of styrene copolymers. , 1980, Toxicology.

[32]  Riyi Shi,et al.  Functionalized mesoporous silica nanoparticle-based drug delivery system to rescue acrolein-mediated cell death. , 2008, Nanomedicine.

[33]  R. Shi,et al.  Acrolein‐mediated mechanisms of neuronal death , 2006, Journal of neuroscience research.