Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomed Opt ; 17(2): 026006, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22463038

ABSTRACT

We determined the effect of aggregation and coating thickness of gold on the luminescence of nanoparticles engulfed by macrophages and in gelatin phantoms. Thin gold-coated iron oxide nanoclusters (nanoroses) have been developed to target macrophages to provide contrast enhancement for near-infrared optical imaging applications. We compare the brightness of nanoroses luminescent emissions in response to 635 nm laser excitation to other nanoparticles including nanoshells, nanorods, and Cy5 conjugated iron oxide nanoparticles. Luminescent properties of all these nanoparticles were investigated in monomeric and aggregated form in gelatin phantoms and primary macrophage cell cultures using confocal microscopy. Aggregation of the gold nanoparticles increased luminescence emission and correlated with increased surface mass of gold per nanoparticle (nanoshells 37 ± 14.30 × 10(-3) brightness with 1.23 × 10(-4) wt of gold (g)/nanoparticle versus original nanorose 1.45 ± 0.37 × 10(-3) with 2.10 × 10(-16) wt of gold/nanoparticle, p<0.05). Nanoshells showed greater luminescent intensity than original nanoroses or Cy5 conjugated iron oxide nanoparticles when compared as nanoparticles per macrophage (38 ± 10 versus 11 ± 2.8 versus 17 ± 6.5, p<0.05, respectively, ANOVA), but showed relatively poor macrophage uptake (1025 ± 128 versus 7549 ± 236 versus 96,000 nanoparticles/cell, p<0.05, student t-test nanoshells versus nanoroses). Enhancement of gold fluorescent emissions by nanoparticles can be achieved by reducing the thickness of the gold coating, by clustering the gold on the surface of the nanoparticles (nanoshells), and by clustering the gold nanoparticles themselves.


Subject(s)
Cell Tracking/methods , Gold , Macrophages/cytology , Microscopy, Confocal/methods , Nanoparticles , Animals , Cells, Cultured , Infrared Rays , Mice , Mice, Inbred C57BL , Reproducibility of Results , Sensitivity and Specificity
2.
Langmuir ; 27(12): 7681-90, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21591638

ABSTRACT

The ability of smaller than 100 nm antibody (Ab) nanoparticle conjugates to target and modulate the biology of specific cell types may enable major advancements in cellular imaging and therapy in cancer. A key challenge is to load a high degree of targeting, imaging, and therapeutic functionality into small, yet stable particles. A versatile method called thin autocatalytic growth on substrate (TAGs) has been developed in our previous study to form ultrathin and asymmetric gold coatings on iron oxide nanocluster cores producing exceptional near-infrared (NIR) absorbance. AlexaFluor 488 labeled Abs were used to correlate the number of Abs conjugated to iron oxide/gold nanoclusters (nanoroses) with the hydrodynamic size. A transition from submonolayer to multilayer aggregates of Abs on the nanorose surface was observed for 54 Abs and an overall particle diameter of ∼60-65 nm. The hydrodynamic diameter indicated coverage of a monolayer of 54 Abs, in agreement with the prediction of a geometric model, by assuming a circular footprint of 16.9 nm diameter per Ab molecule. The targeting efficacy of nanoclusters conjugated with monoclonal Abs specific for epidermal growth factor receptor (EGFR) was evaluated in A431 cancer cells using dark field microscopy and atomic absorbance spectrometry (AAS) analysis. Intense NIR scattering was achieved from both high uptake of nanoclusters in cells and high intrinsic NIR absorbance of individual nanoclusters. Dual mode imaging with dark field reflectance microscopy and fluorescence microscopy indicates the Abs remained attached to the Au surfaces upon the uptake by the cancer cells. The ability to load intense multifunctionality, specifically strong NIR absorbance, conjugation of an Ab monolayer in addition to a strong r2 MRI contrast that was previously demonstrated in a total particle size of only 63 nm, is an important step forward in development of theranostic agents for combined molecular specific imaging and therapy.


Subject(s)
Antibodies/immunology , ErbB Receptors/immunology , Nanostructures , Cell Line, Tumor , Humans , Microscopy, Fluorescence , Spectrophotometry, Ultraviolet
3.
ACS Nano ; 4(4): 2178-84, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20373747

ABSTRACT

Metal nanoparticles with surface plasmon resonance (SPR) in the near-infrared region (NIR) are of great interest for imaging and therapy. Presently, gold nanoparticles with NIR absorbance are typically larger than 50 nm, above the threshold size of approximately 5 nm required for efficient renal clearance. As these nanoparticles are not biodegradable, concerns about long-term toxicity have restricted their translation into the clinic. Here, we address this problem by developing a flexible platform for the kinetically controlled assembly of sub-5 nm ligand-coated gold particles to produce metal/polymer biodegradable nanoclusters smaller than 100 nm with strong NIR absorbance for multimodal application. A key novel feature of the proposed synthesis is the use of weakly adsorbing biodegradable polymers that allows tight control of nanocluster size and, in addition, results in nanoclusters with unprecedented metal loadings and thus optical functionality. Over time, the biodegradable polymer stabilizer degrades under physiological conditions that leads to disassembly of the nanoclusters into sub-5 nm primary gold particles which are favorable for efficient body clearance. This synthesis of polymer/inorganic nanoclusters combines the imaging contrast and therapeutic capabilities afforded by the NIR-active nanoparticle assembly with the biodegradability of a polymer stabilizer.


Subject(s)
Infrared Rays , Molecular Imaging/methods , Nanocomposites/chemistry , Polymers/metabolism , Polymers/therapeutic use , Surface Plasmon Resonance , Adsorption , Animals , Cell Line , Gold/chemistry , Kinetics , Lactic Acid/chemistry , Metal Nanoparticles/chemistry , Mice , Polyesters , Polyethylene Glycols/chemistry , Polymers/chemistry , Polymers/toxicity
4.
Biomed Opt Express ; 1(1): 2-16, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-21258441

ABSTRACT

Application of photothermal Optical Coherence Tomography (OCT) to detect macrophages in ex vivo rabbit arteries which have engulfed nanoclusters of gold coated iron oxide (nanorose) is reported. Nanorose engulfed by macrophages associated with atherosclerotic lesions in rabbit arteries absorb incident laser (800nm) energy and cause optical pathlength (OP) variation which is measured using photothermal OCT. OP variation in polydimethyl siloxane tissue phantoms containing varying concentrations of nanorose match values predicted from nanoparticle and material properties. Measurement of OP variation in rabbit arteries in response to laser excitation provides an estimate of nanorose concentration in atherosclerotic lesions of 2.5x10(9) particles/ml. OP variation in atherosclerotic lesions containing macrophages taking up nanorose has a different magnitude and profile from that observed in control thoracic aorta without macrophages and is consistent with macrophage presence as identified with RAM-11 histology staining. Our results suggest that tissue regions with macrophages taking up nanorose can be detected using photothermal OCT.

5.
Article in English | MEDLINE | ID: mdl-19964846

ABSTRACT

Ultrasound is a widely used modality with excellent spatial resolution, low cost, portability, reliability and safety. In clinical practice and in the biomedical field, molecular ultrasound-based imaging techniques are desired to visualize tissue pathologies, such as cancer. In this paper, we present an advanced imaging technique - combined photoacoustic and magneto-acoustic imaging - capable of visualizing the anatomical, functional and biomechanical properties of tissues or organs. The experiments to test the combined imaging technique were performed using dual, nanoparticle-based contrast agents that exhibit the desired optical and magnetic properties. The results of our study demonstrate the feasibility of the combined photoacoustic and magneto-acoustic imaging that takes the advantages of each imaging techniques and provides high sensitivity, reliable contrast and good penetrating depth. Therefore, the developed imaging technique can be used in wide range of biomedical and clinical application.


Subject(s)
Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Magnetics , Microscopy, Acoustic/instrumentation , Microscopy, Acoustic/methods , Optics and Photonics/instrumentation , Optics and Photonics/methods , Ferric Compounds/chemistry , Nanoparticles/chemistry
6.
ACS Nano ; 3(9): 2686-96, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19711944

ABSTRACT

The ability of 20-50 nm nanoparticles to target and modulate the biology of specific types of cells will enable major advancements in cellular imaging and therapy in cancer and atherosclerosis. A key challenge is to load an extremely high degree of targeting, imaging, and therapeutic functionality into small, yet stable particles. Herein we report approximately 30 nm stable uniformly sized near-infrared (NIR) active, superparamagnetic nanoclusters formed by kinetically controlled self-assembly of gold-coated iron oxide nanoparticles. The controlled assembly of nanocomposite particles into clusters with small primary particle spacings produces collective responses of the electrons that shift the absorbance into the NIR region. The nanoclusters of approximately 70 iron oxide primary particles with thin gold coatings display intense NIR (700-850 nm) absorbance with a cross section of approximately 10(-14) m(2). Because of the thin gold shells with an average thickness of only 2 nm, the r(2) spin-spin magnetic relaxivity is 219 mM(-1) s(-1), an order of magnitude larger than observed for typical iron oxide particles with thicker gold shells. Despite only 12% by weight polymeric stabilizer, the particle size and NIR absorbance change very little in deionized water over 8 months. High uptake of the nanoclusters by macrophages is facilitated by the dextran coating, producing intense NIR contrast in dark field and hyperspectral microscopy, both in cell culture and an in vivo rabbit model of atherosclerosis. Small nanoclusters with optical, magnetic, and therapeutic functionality, designed by assembly of nanoparticle building blocks, offer broad opportunities for targeted cellular imaging, therapy, and combined imaging and therapy.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/therapy , Ferric Compounds/chemistry , Ferric Compounds/metabolism , Gold/chemistry , Macrophages/metabolism , Nanoparticles/chemistry , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Biological Transport , Cell Line , Humans , Magnetics , Particle Size , Rabbits , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...