Construction, gene delivery, and expression of DNA tethered nanoparticles.

PURPOSE Layered nanoparticles have the potential to deliver any number of substances to cells both in vitro and in vivo. The purpose of this study was to develop and test a relatively simple alternative to custom synthesized nanoparticles for use in multiple biological systems, with special focus on the eye. METHODS The biotin-labeled transcriptionally active PCR products (TAP) were conjugated to gold, semiconductor nanocrystals, and magnetic nanoparticles (MNP) coated with streptavidin. The process of nanoparticle construction was monitored with gel electrophoresis. Fluorescence microscopy followed by image analysis was used to examine gene expression levels from DNA alone and tethered MNP in human hepatoma derived Huh-7 cells. Adult retinal endothelial cells from both dog (ADREC) and human (HREC) sources were transfected with nanoparticles and reporter gene expression evaluated with confocal and fluorescent microscopy. Transmission electron microscopy was used to quantify the concentration of nanoparticles in a stock solution. Nanoparticles were evaluated for transfection efficiency, determined by fluorescence microscopy cell counts. Cells treated with MNP were evaluated for increased reactive oxygen species (ROS) and necrosis with flow cytometry. RESULTS Both 5' and 3' biotin-labeled TAP bound equally to MNP and there were no differences in functionality between the two tethering orientations. Free DNA was easily removed by the use of magnetic columns. These particles were also able to deliver genes to a human hepatoma cell line, Huh-7, but transfection efficiency was greater than TAP. The semiconductor nanocrystals and MNP had the highest transfection efficiencies. The MNP did not induce ROS formation or necrosis after 48 h of incubation. CONCLUSIONS Once transfected, the MNP had reporter gene expression levels equivalent to TAP. The nanoparticles, however, had better transfection efficiencies than TAP. The magnetic nanoparticles were the most easily purified of all the nanoparticles tested. This strategy for bioconjugating TAP to nanoparticles is valuable because nanoparticle composition can be changed and the system optimized quickly. Since endothelial cells take up MNP, this strategy could be used to target neovascularization as occurs in proliferative retinopathies. Multiple cell types were used to test this technology and in each the nanoparticles were capable of transfection. In adult endothelial cells the MNP appeared innocuous, even at the highest doses tested with respect to ROS and necrosis. This technology has the potential to be used as more than just a vector for gene transfer, because each layer has the potential to perform its own unique function and then degrade to expose the next functional layer.

[1]  D. S. Mcleod,et al.  Nanoparticle tethered antioxidant response element as a biosensor for oxygen induced toxicity in retinal endothelial cells. , 2006, Molecular vision.

[2]  Jayanth Panyam,et al.  Biodegradable nanoparticles for drug and gene delivery to cells and tissue. , 2003, Advanced drug delivery reviews.

[3]  D. S. Mcleod,et al.  Adenosine stimulates canine retinal microvascular endothelial cell migration and tube formation. , 1998, Current eye research.

[4]  V. Ciccarone,et al.  Improving the transfection efficiency of post-mitotic neurons , 2001, Journal of Neuroscience Methods.

[5]  J. C. Mullikin Discrete and Continuous Methods for Three-Dimensional Image Analysis , 1993 .

[6]  J. Benns,et al.  Tailoring New Gene Delivery Designs for Specific Targets , 2000, Journal of drug targeting.

[7]  Kerry K. Karukstis,et al.  Targeted Antiproliferative Drug Delivery to Vascular Smooth Muscle Cells With a Magnetic Resonance Imaging Nanoparticle Contrast Agent: Implications for Rational Therapy of Restenosis , 2002, Circulation.

[8]  C. Plank,et al.  Magnetofection: Enhancing and Targeting Gene Delivery with Superparamagnetic Nanoparticles and Magnetic Fields , 2003, Journal of liposome research.

[9]  Shelton D Caruthers,et al.  Molecular imaging of angiogenesis in nascent Vx-2 rabbit tumors using a novel alpha(nu)beta3-targeted nanoparticle and 1.5 tesla magnetic resonance imaging. , 2003, Cancer research.

[10]  Viola Mr Potential of microparticles for diagnostic tracer imaging. , 1990 .

[11]  N. Kotov,et al.  Nanorainbows: graded semiconductor films from quantum dots. , 2001, Journal of the American Chemical Society.

[12]  B. Bonnemain,et al.  Superparamagnetic agents in magnetic resonance imaging: physicochemical characteristics and clinical applications. A review. , 1998, Journal of drug targeting.

[13]  C. Plank,et al.  Magnetofection Potentiates Gene Delivery to Cultured Endothelial Cells , 2003, Journal of Vascular Research.

[14]  R. Weissleder,et al.  Liver. II: Iron oxide-based reticuloendothelial contrast agents for MR imaging. Clinical review. , 1996, Magnetic resonance imaging clinics of North America.

[15]  J. Rosenecker,et al.  The Magnetofection Method: Using Magnetic Force to Enhance Gene Delivery , 2003, Biological chemistry.

[16]  S. Davis,et al.  Nanoparticles in drug delivery. , 1987, Critical reviews in therapeutic drug carrier systems.

[17]  Joseph Rosenecker,et al.  Enhancing and targeting nucleic acid delivery by magnetic force , 2003, Expert opinion on biological therapy.

[18]  Vasilis Ntziachristos,et al.  High throughput magnetic resonance imaging for evaluating targeted nanoparticle probes. , 2002, Bioconjugate chemistry.

[19]  T. Thomas,et al.  DNA nanoparticles and development of DNA delivery vehicles for gene therapy. , 2002, Biochemistry.

[20]  D. Mitchell MR imaging contrast agents — what's in a name? , 1997, Journal of magnetic resonance imaging : JMRI.

[21]  S. Zahler,et al.  Magnetofection--a highly efficient tool for antisense oligonucleotide delivery in vitro and in vivo. , 2003, Molecular therapy : the journal of the American Society of Gene Therapy.

[22]  S. Kaufman,et al.  Peer Reviewed: Nanoparticle Detection Technology for Chemical Analysis. , 1999 .

[23]  M. Violante Potential of microparticles for diagnostic tracer imaging. , 1990, Acta radiologica. Supplementum.

[24]  J. Kreuter,et al.  Distribution and elimination of poly(methyl-2-14C-methacrylate) nanoparticle radioactivity after injection in rats and mice. , 1979, Journal of pharmaceutical sciences.

[25]  S. Kaufman,et al.  Nanoparticle detection technology for chemical analysis. , 1999, Analytical chemistry.