Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 226
1.
J Mater Chem B ; 9(42): 8832-8841, 2021 11 03.
Article En | MEDLINE | ID: mdl-34636390

Tumor-targeting gold nanorods (AuNRs) assembled through Au-S bonds have been widely used for photothermal therapy (PTT) via intravenous injection. However, with extended in vivo circulation times, biothiols can replace some S-modified targeting ligands on the surface of the AuNRs, which lowers their targeting efficacy towards cancer cells, resulting in a non-ideal PTT effect. To address this problem, herein, we utilized Se-modified AuNRs to establish a dual functional nanoprobe (Casp-RGD-Se-AuNRs) for improving the therapeutic effect and real-time monitoring of Caspase-9 levels to indicate the degree of cell apoptosis. The experiments demonstrated that the Casp-RGD-Se-AuNRs are better at avoiding interference from biothiols than the S-modified nanoprobe (Casp-RGD-S-AuNRs) for extended blood-circulation times after intravenous injection, significantly improving the PTT efficacy via more effectively targeting cancer cells. Simultaneously, the change of Caspase-9 levels visually shows the degree of apoptosis. Moreover, an in vivo study showed that, compared with the S-modified nanoprobe, the Se-modified nanoprobe exhibits a higher delivery efficiency to the tumor region after intravenous injection (accumulation in the tumor increased by 87%) and a better anticancer efficacy under NIR light irradiation (the tumor inhibition rate increased 6-fold). This work provides a valuable strategy to overcome the off-target problem, and new ideas for avoiding interference by biomolecules during blood circulation.


Antineoplastic Agents/pharmacology , Gold/pharmacology , Nanotubes/chemistry , Photosensitizing Agents/pharmacology , Photothermal Therapy , Selenium/pharmacology , Sulfhydryl Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gold/blood , Gold/chemistry , Humans , Infrared Rays , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Selenium/blood , Selenium/chemistry , Sulfhydryl Compounds/blood , Sulfhydryl Compounds/chemistry
2.
Anal Chem ; 92(22): 14872-14877, 2020 11 17.
Article En | MEDLINE | ID: mdl-32972134

Gold nanoparticles (AuNPs) are increasingly being used as diagnostic and therapeutic agents owing to their excellent properties; however, there is not much data available on their dynamics in vivo on a single particle basis in a single mouse. Here, we developed a method for the direct analysis of nanoparticles in trace blood samples based on single particle inductively coupled plasma-mass spectrometry (spICP-MS). A flexible, highly configurable, and precisely controlled sample introduction system was designed by assembling an ultralow-volume autosampler (flow rate in the range of 5-5000 µL/min) and a customized cyclonic spray chamber (transfer efficiency up to 99%). Upon systematic optimization, the detection limit of the nanoparticle size (LODsize) of AuNPs in ultrapure water was 19 nm, and the detection limit of the nanoparticle number concentration (LODNP) was 8 × 104 particle/L. Using a retro-orbital blood sampling method and subsequent dilution, the system was successfully applied to track the dynamic changes in size and concentration for AuNPs in the blood of a single mouse, and the recovery for the blood sample was 111.74%. Furthermore, the concentration of AuNPs in mouse blood reached a peak in a short period of time and, then, gradually decreased. This study provides a promising technique for analyzing and monitoring the size and concentration of nanoparticles in ultralow-volume blood samples with low concentrations, making it a powerful tool for analyzing and understanding the fate of nanoparticles in vivo.


Analytic Sample Preparation Methods/methods , Blood Chemical Analysis/methods , Gold/blood , Gold/chemistry , Mass Spectrometry/methods , Metal Nanoparticles , Animals , Male , Mice, Inbred C57BL
3.
PLoS One ; 15(7): e0234916, 2020.
Article En | MEDLINE | ID: mdl-32614882

A great deal of attention has been focused on nanoparticles for cancer therapy, with the promise of tumor-selective delivery. However, despite intense work in the field over many years, the biggest obstacle to this vision remains extremely low delivery efficiency of nanoparticles into tumors. Due to the cost, time, and impact on the animals for in vivo studies, the nanoparticle field predominantly uses cellular uptake assays as a proxy to predict in vivo outcomes. Extensive research has focused on decreasing macrophage uptake in vitro as a proxy to delay nanoparticle accumulation in the mononuclear phagocytic system (MPS), mainly the liver and spleen, and thereby increase tumor accumulation. We have recently reported novel synthetic methods employing small molecule crosslinkers for the controlled assembly of small nanoparticles into larger aggregates and found that these nanoaggregates had remarkably high surface coverage and low cell uptake, even in macrophages. We further found that this extremely low cellular uptake could be recapitulated on solid gold nanoparticles by densely coating their surface with small molecules. Here we report our studies on the biodistribution and clearance of these materials in comparison to more conventional PEGylated gold nanoparticles. It was expected that the remarkably low macrophage uptake in vitro would translate to extended blood circulation time in vivo, but instead we found no correlation between either surface coverage or in vitro macrophage cell uptake and in vivo blood circulation. Gold nanoaggregates accumulate more rapidly and to a higher level in the liver compared to control gold nanoparticles. The lack of correlation between in vitro macrophage uptake and in vivo blood circulation suggests that the field must find other in vitro assays to use as a primary proxy for in vivo outcomes or use direct in vivo experimentation as a primary assay.


Coated Materials, Biocompatible/pharmacokinetics , Gold/pharmacokinetics , Metal Nanoparticles , Polyethylene Glycols , Animals , Endocytosis , Fasting/metabolism , Female , Gold/administration & dosage , Gold/blood , Half-Life , Kidney/metabolism , Liver/metabolism , Macrophages/physiology , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/classification , Mice , Organ Specificity , Pilot Projects , RAW 264.7 Cells , Specific Pathogen-Free Organisms , Spleen/metabolism , Tissue Distribution
4.
PLoS One ; 15(5): e0233357, 2020.
Article En | MEDLINE | ID: mdl-32433650

Trace elements and minerals are compounds that are essential for the support of a variety of biological functions and play an important role in the formation of and the defense against oxidative stress. Here we describe a technique, allowing sequential detection of the trace elements (K, Zn, Se, Cu, Mn, Fe, Mg) in serum and whole blood by an ICP-MS method using single work-up, which is a simple, quick and robust method for the sequential measurement and quantification of the trace elements Sodium (Na), Potassium (K), Calcium (Ca), Zinc (Zn), Selenium (Se), Copper (Cu), Iron (Fe), Manganese (Mn) and Magnesium (Mg) in whole blood as well as Copper (Cu), Selenium (Se), Zinc (Zn), Iron (Fe), Magnesium (Mg), Manganese (Mn), Chromium (Cr), Nickel (Ni), Gold (Au) and Lithium (Li) in human serum. For analysis, only 100 µl of serum or whole blood is sufficient, which make this method suitable for detecting trace element deficiency or excess in newborns and infants. All samples were processed and analyzed by ICP-MS (Agilent Technologies). The accuracy, precision, linearity and the limit of quantification (LOQ), Limit of Blank (LOB) and the limit of detection (LOD) of the method were assessed. Recovery rates were between 80-130% for most of the analyzed elements; repeatabilities (Cv %) calculated were below 15% for most of the measured elements. The validity of the proposed methodology was assessed by analyzing a certified human serum and whole blood material with known concentrations for all elements; the method described is ready for routine use in biomonitoring studies.


Spectrophotometry, Atomic/methods , Tandem Mass Spectrometry/methods , Trace Elements/blood , Calcium/blood , Chromium/blood , Copper/blood , Gold/blood , Humans , Iron/blood , Limit of Detection , Lithium/blood , Magnesium/blood , Manganese/blood , Nickel/blood , Potassium/blood , Selenium/blood , Sodium/blood , Zinc/blood
5.
Article En | MEDLINE | ID: mdl-32305711

The bimetallic metal complex Titanocref exhibits relevant anticancer activity, but it is unknown if it is stable to reach target tissues intact. To gain insight, a pharmacologically relevant dose was added to human blood plasma and the mixture was incubated at 37 °C. The obtained mixture was analyzed 5 and 60 min later by size-exclusion chromatography hyphenated to an inductively coupled plasma atomic emission spectrometer (SEC-ICP-AES). We simultaneously detected several titanium (Ti), gold (Au) and sulfur (S)-peaks, which corresponded to a Ti degradation product that eluted partially, and a Au degradation product that eluted entirely bound to plasma proteins (both time points). Although ~70% of the intact Titanocref was retained on the column after 60 min, our results allowed us to establish - for the first time - its likely degradation pathway in human plasma at near physiological conditions. These results suggest that ~70% of Titanocref remain in plasma after 60 min, which supports results from a recent in vivo study in which mice were treated with Titanocref and revealed Ti:Au molar ratios in tumors and organs close to 1:1. Thus, our stability studies suggest that the intact drug is able to reach target tissue. Overall, our results exemplify that SEC-ICP-AES enables the execution of intermediate in vitro studies with human plasma in the context of advancing bimetallic metal-based drugs to more costly clinical studies.


Antineoplastic Agents/blood , Gold/blood , Plasma/chemistry , Sulfur/blood , Titanium/blood , Antineoplastic Agents/isolation & purification , Blood Proteins/chemistry , Blood Proteins/isolation & purification , Chromatography, Gel , Gold/isolation & purification , Humans , Male , Protein Binding , Spectrophotometry, Atomic , Titanium/isolation & purification
6.
J Nanobiotechnology ; 18(1): 45, 2020 Mar 14.
Article En | MEDLINE | ID: mdl-32169073

BACKGROUND: To effectively applied nanomaterials (NMs) in medicine, one of the top priorities is to address a better understanding of the possible sub-organ transfer, clearance routes, and potential toxicity of the NMs in the liver and kidney. RESULTS: Here we explored how the surface chemistry of polyethylene glycol (PEG), chitosan (CS), and polyethylenimine (PEI) capped gold nanoparticles (GNPs) governs their sub-organ biodistribution, transfer, and clearance profiles in the liver and kidney after intravenous injection in mice. The PEG-GNPs maintained dispersion properties in vivo, facilitating passage through the liver sinusoidal endothelium and Disse space, and were captured by hepatocytes and eliminated via the hepatobiliary route. While, the agglomeration/aggregation of CS-GNPs and PEI-GNPs in hepatic Kupffer and endothelial cells led to their long-term accumulation, impeding their elimination. The gene microarray analysis shows that the accumulation of CS-GNPs and PEI-GNPs in the liver induced obvious down-regulation of Cyp4a or Cyp2b related genes, suggesting CS-GNP and PEI-GNP treatment impacted metabolic processes, while the PEI-GNP treatment is related with immune responses. CONCLUSIONS: This study demonstrates that manipulation of nanoparticle surface chemistry can help NPs selectively access distinct cell types and elimination pathways, which help to clinical potential of non-biodegradable NPs.


Gold/metabolism , Gold/toxicity , Kidney/metabolism , Liver/metabolism , Metal Nanoparticles/toxicity , Animals , Chitosan/metabolism , Cytosol , Disease Models, Animal , Gene Expression/drug effects , Gold/blood , Kidney/pathology , Kinetics , Liver/pathology , Male , Metal Nanoparticles/chemistry , Mice , Mice, Inbred ICR , Particle Size , Polyethylene Glycols/metabolism , Polyethyleneimine/metabolism , Rats , Rats, Wistar , Tissue Distribution , Transcriptome
7.
ACS Nano ; 13(5): 5002-5014, 2019 05 28.
Article En | MEDLINE | ID: mdl-30916928

Antibiotics that are most used to cure bacterial infections in the clinic result in the imbalance of intestinal microflora, destroy the intestinal barrier, and induce bacterial resistance. There is an urgent need for antibacterial agent therapy for bacterial infections that does not destroy intestinal microflora. Herein, we applied 4,6-diamino-2-pyrimidinethiol (DAPT)-coated Au nanoparticles (D-Au NPs) for therapy of bacterial infection induced by Escherichia coli ( E. coli) in the gut. We cultured D-Au NPs and E. coli in an anaerobic atmosphere to evaluate their bactericidal effect. We studied the microflora, distribution of Au, and biomarkers in mice after a 28-day oral administration to analyze the effect of Au NPs on mice. D-Au NPs cured bacterial infections more effectively than levofloxacin without harming intestinal microflora. D-Au NPs showed great potential as alternatives to oral antibiotics.


Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Gastrointestinal Microbiome , Gold/therapeutic use , Metal Nanoparticles/therapeutic use , Administration, Oral , Animals , Bacterial Infections/blood , Biocompatible Materials/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/ultrastructure , Feces/chemistry , Gastrointestinal Microbiome/drug effects , Gold/administration & dosage , Gold/blood , Gold/pharmacology , Intestine, Small/ultrastructure , Male , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C
8.
Colloids Surf B Biointerfaces ; 177: 141-148, 2019 May 01.
Article En | MEDLINE | ID: mdl-30721790

Adsorption of biomolecules onto nanoparticles surface in biological samples led to the formation of a bio-corona, it could modified the "identity" of nanoparticles, contributing to the determination of their toxicity and biocompatibility. Gel electrophoresis in combination with liquid chromatography-tandem mass spectrometry (LC-MS/MS) was employed to qualitatively analyze and identify the human serum proteins adsorbed on the surface of three different nanomaterials stabilized with citrate: 10.02 ± 0.91 nm gold nanoparticles (AuNPs), 9.73 ± 1.70 nm silver nanoparticles (AgNPs) and, 2.40 ± 0.30 nm platinum nanoparticles (PtNPs). An exhaustive analysis and classification of all identified proteins related with their function were also developed.


Gold/blood , Metal Nanoparticles/chemistry , Platinum/blood , Protein Corona/analysis , Proteomics , Silver/blood , Gold/chemistry , Humans , Platinum/chemistry , Silver/chemistry , Surface Properties
9.
ACS Infect Dis ; 5(2): 228-238, 2019 02 08.
Article En | MEDLINE | ID: mdl-30521752

A well-developed, functional immune system is paramount to combat harmful attacks from pathogenic organisms and prevent infectious diseases. Newborn animals and humans have only limited immunity upon birth, but their immune functions are expected to develop within weeks to months and eventually to reach a maturity that will provide full protection. Despite the importance of immune activity in animal and human health management, there is no convenient test available that allows for rapid assessment of the state of immune function in nonlaboratory settings. Here we report an extremely simple and rapid blood test that may be used in point-of-care clinics or field settings to evaluate the humoral immune status of animals. The test detects a cooperative interaction between a gold nanoparticle and arguably the three most important proteins involved in the immune system: immunoglobulin M (IgM), immunoglobulin G (IgG), and at least one complement protein, C3, in the blood serum. Such interactions cause the gold nanoparticles to form clusters and aggregates. The average particle size of the gold nanoparticle-serum mixture, measured by dynamic light scattering, corresponds positively to the immune status and activity of the subject. Our study demonstrates that the test may be used not only for monitoring the immune function development from neonates to adults, but also for detecting active immune responses during infection. Although data reported here are largely based on murine and bovine models, it is likely that this test will be applicable to humans as well.


Complement C3/immunology , Gold/blood , Immunity, Humoral , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Metal Nanoparticles/chemistry , Animals , Animals, Newborn/immunology , Biological Assay , Cattle , Dynamic Light Scattering , Hematologic Tests , Mice , Virus Diseases/immunology
10.
Toxicol Sci ; 158(1): 23-35, 2017 07 01.
Article En | MEDLINE | ID: mdl-28402537

Many physiologically based pharmacokinetic (PBPK) models for environmental chemicals, drugs, and nanomaterials have been developed to aid risk and safety assessments using acslX. However, acslX has been rendered sunset since November 2015. Alternative modeling tools and tutorials are needed for future PBPK applications. This forum article aimed to: (1) demonstrate the performance of 4 PBPK modeling software packages (acslX, Berkeley Madonna, MATLAB, and R language) tested using 2 existing models (oxytetracycline and gold nanoparticles); (2) provide a tutorial of PBPK model code conversion from acslX to Berkeley Madonna, MATLAB, and R language; (3) discuss the advantages and disadvantages of each software package in the implementation of PBPK models in toxicology, and (4) share our perspective about future direction in this field. Simulation results of plasma/tissue concentrations/amounts of oxytetracycline and gold from different models were compared visually and statistically with linear regression analyses. Simulation results from the original models were correlated well with results from the recoded models, with time-concentration/amount curves nearly superimposable and determination coefficients of 0.86-1.00. Step-by-step explanations of the recoding of the models in different software programs are provided in the Supplementary Data. In summary, this article presents a tutorial of PBPK model code conversion for a small molecule and a nanoparticle among 4 software packages, and a performance comparison of these software packages in PBPK model implementation. This tutorial helps beginners learn PBPK modeling, provides suggestions for selecting a suitable tool for future projects, and may lead to the transition from acslX to alternative modeling tools.


Gold/pharmacokinetics , Metal Nanoparticles/chemistry , Models, Biological , Oxytetracycline/pharmacokinetics , Animals , Dogs , Gold/blood , Gold/chemistry , Oxytetracycline/blood , Swine , Tissue Distribution
11.
Nanomedicine ; 13(4): 1531-1542, 2017 05.
Article En | MEDLINE | ID: mdl-28238752

In this study, we investigated gold nanoparticle (AuNP) interactions in blood using thromboelastography as a rapid screening tool to monitor their influence on blood coagulation. 1.2 nM colloidal AuNPs ranging from 12 to 85 nm have no effect in the blood, however, 5 nM AuNPs demonstrate pro-thrombogenic concentration dependent effects with a reduction in clot formation. Size effects exhibit a non-linear trend with 45 and 85 nm particles resulting in a faster pro-thrombotic response. Clot strength decreased with AuNP size with the greatest reduction with 28 nm particles. We assessed AuNP interactions in the blood focusing on their biological activity. AuNP-RGD possessed pro-coagulant activities, while PEG-thiol, human fibrinogen and clopidogrel prevented blood clot formation and influenced platelet activity, and were more efficient when bound to nanocarriers than unbound ligands. Such tests could fill the knowledge gaps in thrombogenicity of NPs between in vitro test methods and predict in vivo haemocompatibility.


Blood Platelets/drug effects , Coagulants/chemistry , Gold/blood , Metal Nanoparticles/chemistry , Anticoagulants/chemistry , Humans , Ligands , Particle Size , Thrombelastography
12.
ACS Nano ; 11(3): 2428-2443, 2017 03 28.
Article En | MEDLINE | ID: mdl-28040885

A significant challenge to delivering therapeutic doses of nanoparticles to targeted disease sites is the fact that most nanoparticles become trapped in the liver. Liver-resident macrophages, or Kupffer cells, are key cells in the hepatic sequestration of nanoparticles. However, the precise role that the macrophage phenotype plays in nanoparticle uptake is unknown. Here, we show that the human macrophage phenotype modulates hard nanoparticle uptake. Using gold nanoparticles, we examined uptake by human monocyte-derived macrophages that had been driven to a "regulatory" M2 phenotype or an "inflammatory" M1 phenotype and found that M2-type macrophages preferentially take up nanoparticles, with a clear hierarchy among the subtypes (M2c > M2 > M2a > M2b > M1). We also found that stimuli such as LPS/IFN-γ rather than with more "regulatory" stimuli such as TGF-ß/IL-10 reduce per cell macrophage nanoparticle uptake by an average of 40%. Primary human Kupffer cells were found to display heterogeneous expression of M1 and M2 markers, and Kupffer cells expressing higher levels of M2 markers (CD163) take up significantly more nanoparticles than Kupffer cells expressing lower levels of surface CD163. Our results demonstrate that hepatic inflammatory microenvironments should be considered when studying liver sequestration of nanoparticles, and that modifying the hepatic microenvironment might offer a tool for enhancing or decreasing this sequestration. Our findings also suggest that models examining the nanoparticle/macrophage interaction should include studies with primary tissue macrophages.


Gold/metabolism , Liver/metabolism , Macrophages/metabolism , Metal Nanoparticles/chemistry , Gold/blood , Gold/chemistry , Humans , Liver/cytology , Macrophages/chemistry , Monocytes/chemistry , Monocytes/metabolism , Phenotype
13.
Article En | MEDLINE | ID: mdl-27821451

Under an NIH priority to identify new drugs to treat class B parasitic agents, we performed high-throughput screens, which identified the activity of auranofin (Ridaura) against Entamoeba histolytica and Giardia intestinalis, major causes of water- and foodborne outbreaks. Auranofin, an orally administered, gold (Au)-containing compound that was approved by the FDA in 1985 for treatment of rheumatoid arthritis, was effective in vitro and in vivo against E. histolytica and both metronidazole-sensitive and -resistant strains of Giardia We now report the results of an NIH-sponsored phase I trial to characterize the pharmacokinetics (PK) and safety of auranofin in healthy volunteers using modern techniques to measure gold levels. Subjects received orally 6 mg (p.o.) of auranofin daily, the recommended dose for rheumatoid arthritis, for 7 days and were followed for 126 days. Treatment-associated adverse events were reported by 47% of the subjects, but all were mild and resolved without treatment. The mean gold maximum concentration in plasma (Cmax) at day 7 was 0.312 µg/ml and the half-life (t1/2) 35 days, so steady-state blood levels would not be reached in short-term therapy. The highest concentration of gold, 13 µM (auranofin equivalent), or more than 25× the 50% inhibitory concentration (IC50) for E. histolytica and 4× that for Giardia, was in feces at 7 days. Modeling of higher doses (9 and 21 mg/day) was performed for systemic parasitic infections, and plasma gold levels of 0.4 to 1.0 µg/ml were reached after 14 days of treatment at 21 mg/day. This phase I trial supports the idea of the safety of auranofin and provides important PK data to support its potential use as a broad-spectrum antiparasitic drug. (This study has been registered at ClinicalTrials.gov under identifier NCT02089048.).


Antiparasitic Agents/pharmacokinetics , Antirheumatic Agents/pharmacokinetics , Auranofin/pharmacokinetics , Entamoeba histolytica/drug effects , Giardia lamblia/drug effects , Models, Statistical , Administration, Oral , Adult , Antiparasitic Agents/blood , Antirheumatic Agents/blood , Auranofin/blood , Computer Simulation , Drug Administration Schedule , Drug Dosage Calculations , Drug Repositioning , Entamoeba histolytica/growth & development , Female , Giardia lamblia/growth & development , Gold/blood , Half-Life , Healthy Volunteers , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Male , Metronidazole/pharmacology , Tissue Distribution
14.
Nano Lett ; 16(6): 3540-4, 2016 06 08.
Article En | MEDLINE | ID: mdl-27172130

We use individual gold nanorods as pointlike detectors for the intrinsic dynamics of an oscillating biological system. We chose the pattern forming MinDE protein system from Escherichia coli (E. coli), a prominent example for self-organized chemical oscillations of membrane-associated proteins that are involved in the bacterial cell division process. Similar to surface plasmon resonance (SPR), the gold nanorods report changes in their protein surface coverage without the need for fluorescence labeling, a technique we refer to as NanoSPR. Comparing the dynamics for fluorescence labeled and unlabeled proteins, we find a reduction of the oscillation period by about 20%. The absence of photobleaching allows us to investigate Min proteins attaching and detaching from lipid coated gold nanorods with an unprecedented bandwidth of 100 ms time resolution and 1 h observation time. The long observation reveals small changes of the oscillation period over time. Averaging many cycles yields the precise wave profile that exhibits the four phases suggested in previous reports. Unexpected from previous fluorescence-based studies, we found an immobile static protein layer not dissociating during the oscillation cycle. Hence, NanoSPR is an attractive label-free real-time technique for the local investigation of molecular dynamics with high observation bandwidth. It gives access to systems, which cannot be fluorescently labeled, and resolves local dynamics that would average out over the sensor area used in conventional SPR.


Adenosine Triphosphatases/chemistry , Cell Cycle Proteins/chemistry , Escherichia coli Proteins/chemistry , Gold/chemistry , Lipid Bilayers/chemistry , Nanotubes/chemistry , Surface Plasmon Resonance/instrumentation , Biosensing Techniques/methods , Escherichia coli , Fluorescent Dyes/chemistry , Gold/blood , Surface Plasmon Resonance/methods
15.
Ther Deliv ; 6(7): 777-83, 2015 Jul.
Article En | MEDLINE | ID: mdl-26228771

AIM: Gold nanoparticles are employed for imaging and treatment of surgically inaccessible tumors owing to their inherent optical absorption and ability to extravasate through intravenous distribution. These nanoparticles are cleared from the blood by the reticuloendothelial system (RES) as expected given their size. MATERIALS & METHODS: This study demonstrates the effects of RES blockade through the intravenous administration of λ-carrageenan, resulting in a decrease in the median clearance rate from 18.9 (95% CrI: 15.9-22.6) to 11.2 (95% CrI: 8.8-13.9) µl/min and an increase in nanoparticle circulation half-life t(½)( = 264 ± 73 vs 160 ± 22 min; p < 0.01). RESULTS: This 59.3% decrease in clearance is greater than the 15% previously reported for liposomes [ 1 ]. CONCLUSION: The primary benefit of nontoxic RES blockade is to increase the circulation time, where traditional particle modification is ineffective or impractical.


Antineoplastic Agents/pharmacokinetics , Carrageenan/administration & dosage , Gold/pharmacokinetics , Metal Nanoparticles , Mononuclear Phagocyte System/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Chemistry, Pharmaceutical , Female , Gold/administration & dosage , Gold/blood , Half-Life , Injections, Intravenous , Metabolic Clearance Rate , Mice, Inbred BALB C , Models, Biological , Mononuclear Phagocyte System/metabolism , Nanotechnology , Particle Size , Photoplethysmography , Technology, Pharmaceutical/methods
16.
Metallomics ; 7(9): 1364-70, 2015 Sep.
Article En | MEDLINE | ID: mdl-26095799

The development and optimization of a versatile analytical system for the speciation analysis of metal-containing nanoscale materials in blood serum is reported herein. Based on capillary electrophoresis (CE) interfaced with inductively coupled plasma mass spectrometry (ICP-MS), the method was shown to be feasible to investigate the interactions between serum proteins and gold nanoparticles of potential medicinal use, which are their first and foremost occurrence upon their entry into the circulatory system. To improve the separation resolution between the intact nanoparticles and different protein conjugates, the CE system was optimized with an emphasis on compatibility with physiological conditions, avoiding aggregation effects, and analyte recovery. Optimization allowed also for acquiring the acceptable figures of merit such as migration time and peak area precision of 1.0-6.4% and 2.4-6.9%, respectively, detection limits in the range of 0.8-1.0 µg L(-1) Au, and capillary recoveries on the order of 86-97%, depending on the nanoparticle size and conjugate type. We sytematically investigated the role of size in mediating protein adsorption to gold nanoparticles in a real-serum environment. At the initial stage of surface coating, the speciation of smaller particles (5 and 10 nm) was found to be dominated by albumin, transferrin (both in apo- and holo-form) playing the secondary role in developing the protein corona. For 20 and 50 nm nanoparticles, the contribution of transferrin is initially comparable; however, with time it becomes replaced by albumin. The time of attaining equilibrium adsorption is also a function of particle size but for the whole size range investigated, albumin is the only equilibrium binding partner. These principal findings prove that for metal-based nanomaterials in general, serum protein conjugates could be variable in composition depending on the protein abundance and binding affinity, as well as the residence time in the bloodstream.


Electrophoresis, Capillary/methods , Gold/blood , Mass Spectrometry/methods , Metal Nanoparticles/analysis , Blood Proteins/metabolism , Gold/chemistry , Gold/metabolism , Humans , Limit of Detection , Male , Metal Nanoparticles/chemistry , Particle Size , Reproducibility of Results
17.
Sci Rep ; 5: 8669, 2015 Mar 02.
Article En | MEDLINE | ID: mdl-25727895

Radiotherapy is often the most straightforward first line cancer treatment for solid tumors. While it is highly effective against tumors, there is also collateral damage to healthy proximal tissues especially with high doses. The use of radiosensitizers is an effective way to boost the killing efficacy of radiotherapy against the tumor while drastically limiting the received dose and reducing the possible damage to normal tissues. Here, we report the design and application of a good radiosensitizer by using ultrasmall Au(29-43)(SG)(27-37) nanoclusters (<2 nm) with a naturally-occurring peptide (e.g., glutathione or GSH) as the protecting shell. The GSH-coated Au(29-43)(SG)(27-37) nanoclusters can escape the RES absorption, leading to a good tumor uptake (~8.1% ID/g at 24 h post injection). As a result, the as-designed Au nanoclusters led to a strong enhancement for radiotherapy, as well as a negligible damage to normal tissues. After the treatment, the ultrasmall Au(29-43)(SG)(27-37) nanoclusters can be efficiently cleared by the kidney, thereby avoiding potential long-term side-effects caused by the accumulation of gold atoms in the body. Our data suggest that the ultrasmall peptide-protected Au nanoclusters are a promising radiosensitizer for cancer radiotherapy.


Gold/therapeutic use , Kidney/metabolism , Metal Nanoparticles/therapeutic use , Neoplasms/metabolism , Radiation-Sensitizing Agents/therapeutic use , Animals , Glutathione/blood , Gold/blood , Hydrodynamics , Imaging, Three-Dimensional , Kidney/drug effects , Male , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C , Neoplasms/diagnostic imaging , Neoplasms/pathology , Organ Specificity/drug effects , Radiation-Sensitizing Agents/pharmacology , Radionuclide Imaging , Spectrophotometry, Ultraviolet , Time Factors , Tomography, X-Ray Computed , Tumor Burden/drug effects
18.
Analyst ; 140(9): 3090-7, 2015 May 07.
Article En | MEDLINE | ID: mdl-25802895

As biospectroscopy techniques continue to be developed for screening or diagnosis within a point-of-care setting, an important development for this field will be high-throughput optimization. For many of these techniques, it is therefore necessary to adapt and develop parameters to generate a robust yet simple approach delivering high-quality spectra from biological samples. Specifically, this is important for surface-enhanced Raman spectroscopy (SERS) wherein there are multiple variables that can be optimised to achieve an enhancement of the Raman signal from a sample. One hypothesis is that "large" diameter (>100 nm) gold nanoparticles provide a greater enhancement at near-infrared (NIR) and infrared (IR) wavelengths than those <100 nm in diameter. Herein, we examine this notion using examples in which SERS spectra were acquired from MCF-7 breast cancer cells incubated with 150 nm gold nanoparticles. It was found that 150 nm gold nanoparticles are an excellent material for NIR/IR SERS. Larger gold nanoparticles may better satisfy the theoretical restraints for SERS enhancement at NIR/IR wavelengths compared to smaller nanoparticles. Also, larger nanoparticles or their aggregates are more readily observed via optical microscopy (and especially electron microscopy) compared to smaller ones. This allows rapid and straightforward identification of target areas containing a high concentration of nanoparticles and facilitating SERS spectral acquisition. To some extent, these observations appear to extend to biofluids such as blood plasma or (especially) serum; SERS spectra of such biological samples often exhibit a low signal-to-noise ratio in the absence of nanoparticles. With protein-rich biofluids such as serum, a dramatic SERS effect can be observed; although this might facilitate improved spectral biomarker identification in the future, it may not always improve classification between control vs. cancer. Thus, use of "large" gold nanoparticles are a good starting point in order to derive informative NIR/IR SERS analysis of biological samples.


Breast Neoplasms/pathology , Breast/pathology , Gold/analysis , Metal Nanoparticles/analysis , Spectrum Analysis, Raman/methods , Breast/chemistry , Breast Neoplasms/chemistry , Female , Gold/blood , Humans , MCF-7 Cells , Metal Nanoparticles/ultrastructure , Serum/chemistry
19.
Metallomics ; 7(3): 516-24, 2015 Mar.
Article En | MEDLINE | ID: mdl-25671498

Engineered gold nanoparticles (AuNPs) have recently drawn an increased interest in disease diagnostics and therapies. However, reports on detailed studies of AuNPs regarding their pharmacodynamics, pharmacokinetics, biodistribution, metabolism and potential toxicity are limited. It is common knowledge that the in vivo behavior and fate of various AuNPs are influenced by their surface and size. However, a comprehensive description and understanding of all variables is crucial for their further development toward potential clinical use. In this article, we describe the pharmacokinetics and biodistribution of mesoporous silica-coated gold nanorods functionalized with polyethylene glycol or bovine serum albumin (AuNR@SiO2-PEG and AuNR@SiO2-BSA, respectively) in tumor-bearing balb/c mice. To gain further insight into the pharmacokinetics, biodistribution and tumor uptake, we also compare the results with BSA functionalized gold nanorods (AuNR-BSA) and gold clusters (AuNC-BSA). The results reveal that AuNR@SiO2-PEG have the longest blood half-life and the maximum percentage content in the tumor at 24 h and 3 days compared to other AuNPs. AuNR@SiO2-PEG, AuNR@SiO2-BSA and AuNR-BSA had primarily accumulated in the liver and spleen without apparent metabolism after 3 days, while the content of AuNC-BSA in the liver, spleen and kidneys showed an obvious decrease, indicating a size-dependent metabolism process. Our results demonstrate how to manipulate the size and surface chemistry of AuNPs to prolong their blood circulation time, improve delivery into target organs and achieve a safer design of nanomedicines.


Gold/chemistry , Gold/pharmacokinetics , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Nanotechnology , Neoplasms/metabolism , Particle Size , Administration, Intravenous , Animals , Cattle , Gold/blood , Gold/toxicity , Hydrodynamics , Male , Metal Nanoparticles/toxicity , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C , Neoplasms/blood , Neoplasms/pathology , Serum Albumin, Bovine/metabolism , Spectrophotometry, Atomic , Static Electricity , Tissue Distribution , Toxicity Tests
20.
Analyst ; 139(22): 5977-82, 2014 Nov 21.
Article En | MEDLINE | ID: mdl-25267979

We report a rapid and simple assay for colorimetric visualization of thrombin at nanomolar levels using functional gold nanoparticles (FAuNPs) coupled with microporous membranes. We used a 29-mer thiolated-thrombin-binding-aptamer (TBA29) to prepare TBA29 functionalized AuNPs (TBA29-AuNPs) for the selective detection of human thrombin. The sensing mechanism is based on the principle of TBA29-AuNPs flowing down through the nitrocellulose membrane (NCM) pores at different flow rates after binding to thrombin. Compared with free TBA29-AuNPs, when thrombin-TBA29-AuNPs were dropped on the NCM, the particles flowed down more easily through the NCM pores along with the buffer solution due to the increase in the gravity of particles. Therefore, color intensities of TBA29-AuNPs on the NCM depended on the concentration of thrombin; the color intensity was lighter when the concentration of thrombin was higher. Thrombin can be detected at the nanomolar level with the naked eye using this colorimetric probe. A protein G modified AuNP based probe (PG-AuNPs/NCM) was employed to detect human immunoglobulin G (hIgG) in plasma samples to demonstrate the practicality of our sensing system. Also, fibrinogen modified Au NPs were analyzed to demonstrate that this concept of detection could be extended to other proteins or systems, by functionalizing with suitable molecules.


Colorimetry/methods , Gold/chemistry , Membranes, Artificial , Metal Nanoparticles , Proteins/analysis , Gold/blood
...