Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nanomedicine ; 14(4): 1279-1287, 2018 06.
Article in English | MEDLINE | ID: mdl-29597048

ABSTRACT

Canine lower urinary tract neoplasia is a clinically important disease process that has high mortality due to late stage diagnosis and poorly durable response to treatment. Non-invasive diagnostic techniques (e.g. dipstick test, urine cytology) currently have poor diagnostic value, while more invasive tests (e.g. cystoscopy and biopsy) are costly and often require general anesthesia. We have developed and herein describe a quantitative cytological analysis method based on the use of surface-enhanced Raman spectroscopy (SERS), for identifying cancerous transitional cells in urine using SERS biotags (SBTs) carrying the peptide PLZ4 (amino acid sequence cQDGRMGFc) that targets malignant transitional cells. By analyzing the ratio of the PLZ4-SBTs to an on board control we were able to show that transitional cells had significantly higher ratios (P < 0.05) in patients diagnosed with transitional cell carcinoma (TCC) than in healthy samples.


Subject(s)
Carcinoma, Transitional Cell/diagnosis , Spectrum Analysis, Raman/methods , Animals , Biomarkers, Tumor/urine , Biopsy/methods , Carcinoma, Transitional Cell/urine , Cystoscopy/methods , Dogs , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/urine
2.
Nano Lett ; 16(9): 5495-502, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27490923

ABSTRACT

Glutamatergic cytotoxicity mediated by overactivation of N-methyl-d-aspartate receptors (NMDARs) is implicated in numerous neurological disorders. To be therapeutically viable, NMDAR antagonists must preserve physiological role of synaptic NMDARs (sNMDARs) in synaptic transmission and block only excessive pathological activation of NMDARs. Here we present a novel NMDAR antagonist that satisfies this two-fold requirement by exploiting spatial differences in NMDAR subcellular locations. Specifically, we designed a hybrid nanodrug (AuM) to be larger than the synaptic cleft by attaching memantine, NMDAR antagonist, via polymer linkers to a gold nanoparticle. We show that AuM efficiently and selectively inhibited extrasynaptic NMDARs (eNMDARs), while having no effect on sNMDARs and synaptic transmission. AuM exhibited neuroprotective properties both in vitro and ex vivo during such neurotoxic insults as NMDAR-mediated cytotoxicity in cerebrocortical cell culture and oxygen-glucose deprivation in acute hippocampal slices. Furthermore, AuM prevented dendritic spine loss triggered by Aß oligomers in organotypic hippocampal slices and was more effective than free memantine. Using a novel rational design strategy, we demonstrate a proof of concept for a new class of neuroprotective drugs that might be beneficial for treatment of several neurological disorders.


Subject(s)
Metal Nanoparticles , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Transmission , Animals , Cells, Cultured , Cerebral Cortex/cytology , Gold , Memantine/pharmacology , Neuroprotective Agents/pharmacology , Rats, Sprague-Dawley , Synapses
3.
Adv Funct Mater ; 26(2): 267-276, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-27441036

ABSTRACT

The rapid development of fluorescence imaging technologies requires concurrent improvements in the performance of fluorescent probes. Quantum dots have been extensively used as an imaging probe in various research areas because of their inherent advantages based on unique optical and electronic properties. However, their clinical translation has been limited by the potential toxicity especially from cadmium. Here, a versatile bioimaging probe is developed by using highly luminescent cadmium-free CuInSe2/ZnS core/shell quantum dots conjugated with CGKRK (Cys-Gly-Lys-Arg-Lys) tumor-targeting peptides. This probe exhibits excellent photostability, reasonably long circulation time, minimal toxicity, and strong tumor-specific homing property. The most important feature of this probe is that it shows distinctive versatility in tumor-targeted multimodal imaging including near-infrared, time-gated, and two-photon imaging in different tumor models. In a glioblastoma mouse model, the targeted probe clearly denotes tumor boundaries and positively labels a population of diffusely infiltrating tumor cells, suggesting its utility in precise tumor detection during surgery. This work lays a foundation for potential clinical translation of the probe.

4.
Chembiochem ; 17(7): 570-5, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895508

ABSTRACT

Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.


Subject(s)
Aminopyridines/chemistry , Breast Neoplasms/drug therapy , Drug Delivery Systems , Ethylenediamines/chemistry , Mitochondrial Proteins/administration & dosage , Peptides, Cyclic/administration & dosage , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Carrier Proteins , Cell Line, Tumor , Ethylenediamines/pharmacology , Female , Humans , Ligands , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Nanoparticles/chemistry
5.
Nano Lett ; 15(10): 6745-50, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26317146

ABSTRACT

Surface enhanced Raman spectroscopy (SERS) is a powerful analytical technique that has been proposed as a substitute for fluorescence for biological imaging and detection but is not yet commercially utilized. The reason lies primarily in the lower intensity and poor reproducibility of most metal nanoparticle-based tags as compared to their fluorescence-based counterparts. Here, using a technique that scrupulously preserves the same number of dye molecules in both the SERS and fluorescence measurements, we show that SERS-based biotags (SBTs) with highly reproducible optical properties can be nanoengineered such that their brightness is at least equal to that of fluorescence-based tags.

6.
Nat Mater ; 13(9): 904-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907927

ABSTRACT

There is considerable interest in using nanoparticles as labels or to deliver drugs and other bioactive compounds to cells in vitro and in vivo. Fluorescent imaging, commonly used to study internalization and subcellular localization of nanoparticles, does not allow unequivocal distinction between cell surface-bound and internalized particles, as there is no methodology to turn particles 'off'. We have developed a simple technique to rapidly remove silver nanoparticles outside living cells, leaving only the internalized pool for imaging or quantification. The silver nanoparticle (AgNP) etching is based on the sensitivity of Ag to a hexacyanoferrate-thiosulphate redox-based destain solution. In demonstration of the technique we present a class of multicoloured plasmonic nanoprobes comprising dye-labelled AgNPs that are exceptionally bright and photostable, carry peptides as model targeting ligands, can be etched rapidly and with minimal toxicity in mice, and that show tumour uptake in vivo.


Subject(s)
Cells/metabolism , Metal Nanoparticles , Molecular Imaging/methods , Molecular Probes/chemistry , Molecular Probes/metabolism , Silver/chemistry , Silver/metabolism , Animals , Avidin/chemistry , Biological Transport , Cell Line, Tumor , Female , Humans , Mice , Molecular Probes/analysis , Molecular Probes/toxicity , Polyethylene Glycols/chemistry , Silver/toxicity
7.
Mol Pharm ; 12(2): 600-9, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25490248

ABSTRACT

While a host of methods exist to deliver genetic materials or small molecules to cells, very few are available for protein delivery to the cytosol. We describe a modular, light-activated nanocarrier that transports proteins into cells by receptor-mediated endocytosis and delivers the cargo to the cytosol by light triggered endosomal escape. The platform is based on hollow gold nanoshells (HGN) with polyhistidine tagged proteins attached through an avidity-enhanced, nickel chelation linking layer; here, we used green fluorescent protein (GFP) as a model deliverable cargo. Endosomal uptake of the GFP loaded nanocarrier was mediated by a C-end Rule (CendR) internalizing peptide fused to the GFP. Focused femtosecond pulsed-laser excitation triggered protein release from the nanocarrier and endosome disruption, and the released protein was capable of targeting the nucleoli, a model intracellular organelle. We further demonstrate the generality of the approach by loading and releasing Sox2 and p53. This method for targeting of individual cells, with resolution similar to microinjection, provides spatial and temporal control over protein delivery.


Subject(s)
Drug Delivery Systems/methods , Proteins/administration & dosage , Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival , Endocytosis , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Confocal
8.
Nano Lett ; 14(4): 2046-51, 2014.
Article in English | MEDLINE | ID: mdl-24597503

ABSTRACT

We have combined a versatile and powerful route to deliver nucleic acids with peptide-based cell-specific targeting. siRNA targeting the polo-like kinase gene is in clinical trials for cancer treatment, and here we deliver this RNA selectively to cancer cells displaying the neuropilin-1 epitope using gold nanoshells. Release of the siRNA from the nanoparticles results from irradiation with a pulsed near-infrared laser, which also provides efficient endosomal escape within the cell. As a result, our approach requires 10-fold less material than standard nucleic acid transduction materials and is significantly more efficient than other particle-based methods. We also describe a particle-nucleic acid design that does not rely on modified RNA, thereby making the preparation of these materials more efficient and much less expensive. These improvements, when combined with control over when and where the siRNA is released, could provide the basis for diverse cell biological studies.


Subject(s)
Delayed-Action Preparations/chemistry , Gene Transfer Techniques , Gold/chemistry , Nanocapsules/chemistry , Peptides/chemistry , Prostatic Neoplasms/genetics , RNA, Small Interfering/administration & dosage , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Delayed-Action Preparations/metabolism , Drug Delivery Systems , Endosomes/metabolism , Humans , Lasers , Male , Neuropilin-1/metabolism , Peptides/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
9.
Angew Chem Int Ed Engl ; 54(13): 4018-22, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25651530

ABSTRACT

We present a silica nanoparticle (SNP) functionalized with polyphosphate (polyP) that accelerates the natural clotting process of the body. SNPs initiate the contact pathway of the blood-clotting system; short-chain polyP accelerates the common pathway by the rapid formation of thrombin, which enhances the overall blood-clotting system, both by accelerating fibrin generation and by facilitating the regulatory anticoagulation mechanisms essential for hemostasis. Analysis of the clotting properties of bare SNPs, bare polyP, and polyP-functionalized SNPs in plasma demonstrated that the attachment of polyP to SNPs to form polyP-SNPs creates a substantially enhanced synergistic effect that lowers clotting time and increases thrombin production at low concentrations. PolyP-SNP even retains its clotting function at ambient temperature. The polyP-SNP system has the potential to significantly improve trauma-treatment protocols and outcomes in hospital and prehospital settings.


Subject(s)
Blood Coagulation/drug effects , Nanoparticles , Polyphosphates/chemistry , Silicon Dioxide/pharmacology , Fibrin/chemistry , Hemorrhage/drug therapy , Hemostasis , Magnetic Resonance Spectroscopy , Particle Size , Spectrophotometry, Atomic , Temperature , Thrombin/chemistry , Whole Blood Coagulation Time , Zirconium/chemistry
10.
Proc Natl Acad Sci U S A ; 108(40): 16559-64, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21930955

ABSTRACT

A multiplexed, ratiometric method is described that can confidently distinguish between cancerous and noncancerous epithelial prostate cells in vitro. The technique is based on bright surface-enhanced resonance Raman scattering (SERRS) biotags (SBTs) infused with unique Raman reporter molecules, and carrying cell-specific peptides. Two sets of SBTs were used. One targets the neuropilin-1 (NRP-1) receptors of cancer cells through the RPARPAR peptide. The other functions as a positive control (PC) and binds to both noncancerous and cancer cells through the HIV-derived TAT peptide. Point-by-point 2D Raman maps of the spatial distribution of the two tags were constructed with subcellular resolution from cells simultaneously incubated with the two sets of SBTs. Averaging the SERRS signal over a given cell yielded an NRP/PC ratio from which a robust quantitative measure of the overexpression of the NRP-1 by the cancer cell line was extracted. The use of a local, on-cell reference produces quantitative, statistically robust measures of overexpression independent of such sources of uncertainty as variations in the location of the focal plane, the local cell concentration, and turbidity.


Subject(s)
Neuropilin-1/metabolism , Prostatic Neoplasms/diagnosis , Cell Count/methods , Cell Line, Tumor , Humans , In Vitro Techniques , Male , Prostatic Neoplasms/metabolism , Silver , Spectrum Analysis, Raman/methods
11.
Nano Lett ; 12(6): 2912-7, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22551121

ABSTRACT

We characterize the distribution of surface-enhanced Raman spectroscopy (SERS) enhancement factors observed in individual hot spots of single Ag "nanocapsules", encapsulated Ag nanoparticle dimers formed via controlled nanoparticle linking, polymer encapsulation, and small molecule infusion. The enhancement factors are calculated for over 1000 individual nanocapsules by comparing Raman scattering intensities of 4-mercaptobenzoic acid (MBA) measured from single SERS hot spots to intensities measured from high-concentration solutions of MBA. Correlation spectroscopy measurements of the rotational diffusion identify nanocapsules with signals dominated by single hot spots via their strong polarization response. Averaging over the entire surface of the nanocapsules, the distribution of enhancement factors is found to range from 10(6) to 10(8), with a mean of 6 × 10(6). Averaging only over nanoparticle junctions (where most SERS signals are expected) increases this average value to 10(8), with a range from 2 × 10(7) to 2 × 10(9). This significant statistical sampling shows that very high SERS enhancement factors can be obtained on a consistent basis using nanoparticle linking.


Subject(s)
Models, Statistical , Nanostructures/chemistry , Nanostructures/ultrastructure , Spectrum Analysis/methods , Surface Plasmon Resonance/methods , Computer Simulation , Materials Testing , Molecular Conformation , Rotation , Statistics as Topic , Surface Properties
12.
Nano Lett ; 12(1): 61-7, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22133237

ABSTRACT

Nanorattles consisting of hydrophilic, rare-earth-doped NaYF(4) shells each containing a loose magnetic nanoparticle were fabricated through an ion-exchange process. The inner magnetic Fe(3)O(4) nanoparticles are coated with a SiO(2) layer to avoid iron leaching in acidic biological environments. This multifunctional mesoporous nanostructure with both upconversion luminescent and magnetic properties has excellent water dispersibility and a high drug-loading capacity. The material emits visible luminescence upon NIR excitation and can be directed by an external magnetic field to a specific target, making it an attractive system for a variety of biological applications. Measurements on cells incubated with the nanorattles show them to have low cytotoxicity and excellent cell imaging properties. In vivo experiments yield highly encouraging tumor shrinkage with the antitumor drug doxorubicin (DOX) and significantly enhanced tumor targeting in the presence of an applied magnetic field.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Doxorubicin/administration & dosage , Luminescent Measurements/methods , Magnetite Nanoparticles/therapeutic use , Nanocapsules/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Magnetite Nanoparticles/chemistry , Mice , Nanocapsules/chemistry , Porosity
13.
Nano Lett ; 10(5): 1780-6, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20415427

ABSTRACT

The surface-enhanced Raman spectroscopy (SERS) activity and the optical reflectance of a subwavelength gold nanograting fabricated entirely using top down technologies on silicon wafers are presented. The grating consists of 120 nm gold cladding on top of parallel silica nanowires constituting the grating's lines, with gaps between nanowires <10 nm wide at their narrowest point. The grating produces inordinately intense SERS and shows very strong polarization dependence. Reflectance measurements for the optimized grating indicate that (when p-polarization is used and at least one of the incident electric field components lies across the grating lines) the reflectance drops to <1% at resonance, indicating that essentially all of the radiant energy falling on the surface is coupled into the grating. The SERS intensity and the reflectance at resonance anticorrelate predicatively, suggesting that reflectance measurements can provide a nondestructive, wafer-level test of SERS efficacy. The SERS performance of the gratings is very uniform and reproducible. Extensive measurements on samples cut from both the same wafer and from different wafers, produce a SERS intensity distribution function that is similar to that obtained for ordinary Raman measurements carried out at multiple locations on a polished (100) silicon wafer.


Subject(s)
Crystallization/methods , Gold/chemistry , Nanostructures/chemistry , Nanostructures/ultrastructure , Nanotechnology/instrumentation , Refractometry/instrumentation , Spectrum Analysis, Raman/instrumentation , Equipment Design , Equipment Failure Analysis , Macromolecular Substances/chemistry , Materials Testing , Molecular Conformation , Particle Size , Surface Properties
14.
Nat Commun ; 12(1): 1541, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750829

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by marked desmoplasia and drug resistance due, in part, to poor drug delivery to extravascular tumor tissue. Here, we report that carcinoma-associated fibroblasts (CAFs) induce ß5 integrin expression in tumor cells in a TGF-ß dependent manner, making them an efficient drug delivery target for the tumor-penetrating peptide iRGD. The capacity of iRGD to deliver conjugated and co-injected payloads is markedly suppressed when ß5 integrins are knocked out in the tumor cells. Of note, ß5 integrin knock-out in tumor cells leads to reduced disease burden and prolonged survival of the mice, demonstrating its contribution to PDAC progression. iRGD significantly potentiates co-injected chemotherapy in KPC mice with high ß5 integrin expression and may be a powerful strategy to target an aggressive PDAC subpopulation.


Subject(s)
Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Progression , Drug Delivery Systems , Drug Therapy , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Oligopeptides , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
15.
J Am Chem Soc ; 132(9): 2850-1, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20158187

ABSTRACT

We demonstrated that the nanostructures comprising silver cores and dense layers of Y(2)O(3):Er separated by a silica shell are an excellent model system to study the interaction between upconversion materials and metals in nanoscale. This architecture allows for versatile control of the Y(2)O(3):Er-metal interaction through control of the silica dielectric spacer thickness and the metal-core size. Finally, the nanoparticles are potentially interesting as fluorescent labels in, for instance (single particle), imaging experiments or bioassays which require low background or tissue penetrating wavelengths.


Subject(s)
Europium/chemistry , Fluorescent Dyes/pharmacokinetics , Molecular Imaging/methods , Nanostructures/chemistry , Silicon Dioxide/chemistry , Silver/chemistry , Yttrium/chemistry , Cell Line, Tumor , Fluorescence , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Luminescent Measurements , Male , Materials Testing , Particle Size , Prostatic Neoplasms/diagnosis , Surface Properties , Tissue Distribution
16.
J Am Chem Soc ; 131(40): 14237-9, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-19754205

ABSTRACT

We present two strategies for attaching self-assembled DNA arrays to the surfaces of cells. Our first approach uses biotin-streptavidin interactions to bind DNA architectures to biotinylated cells. The second approach takes advantage of specific antibody-cell surface interactions, conjugated arrays and the subsequent binding to native epidermal growth factor receptors expressed on cancer cells. DNA array-cell surface interactions were visualized by fluorescence, confocal microscopy, and scanning electron microscopy. This novel application of DNA nanoarrays provides strategies to specifically label cell surfaces with micrometer-sized patches, bind cells onto a designed functionalized DNA scaffold, engineer cell/cell networks into microtissues, and deliver materials to cell surfaces.


Subject(s)
DNA, Single-Stranded/chemistry , Nanostructures/chemistry , Oligonucleotide Array Sequence Analysis/methods , Antibodies/immunology , Antibodies/metabolism , Biotin/chemistry , DNA, Single-Stranded/chemical synthesis , DNA, Single-Stranded/metabolism , ErbB Receptors/immunology , ErbB Receptors/metabolism , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Jurkat Cells , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Models, Molecular , Streptavidin/chemistry
17.
Sci Adv ; 4(5): eaat0351, 2018 05.
Article in English | MEDLINE | ID: mdl-29795786

ABSTRACT

Noninvasive stimulation of cells is crucial for the accurate examination and control of their function both at the cellular and the system levels. To address this need, we present a pioneering optical stimulation platform that does not require genetic modification of cells but instead capitalizes on unique optoelectronic properties of graphene, including its ability to efficiently convert light into electricity. We report the first studies of optical stimulation of cardiomyocytes via graphene-based biointerfaces (G-biointerfaces) in substrate-based and dispersible configurations. The efficiency of stimulation via G-biointerfaces is independent of light wavelength but can be tuned by changing the light intensity. We demonstrate that an all-optical evaluation of use-dependent drug effects in vitro can be enabled using substrate-based G-biointerfaces. Furthermore, using dispersible G-biointerfaces in vivo, we perform optical modulation of the heart activity in zebrafish embryos. Our discovery is expected to empower numerous fundamental and translational biomedical studies.


Subject(s)
Graphite/chemistry , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Nanostructures , Photic Stimulation , Animals , Biophysical Phenomena , Cells, Cultured , Hydrogen-Ion Concentration , Light , Rats , Temperature , Zebrafish
18.
Nat Commun ; 9(1): 1070, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523838

ABSTRACT

The original version of the Supplementary Information associated with this Article inadvertently omitted Supplementary Table 1. The HTML has now been updated to include a corrected version of the Supplementary Information.

19.
Nat Biomed Eng ; 2(2): 95-103, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29955439

ABSTRACT

Bacterial resistance to antibiotics has made it necessary to resort to antibiotics that have considerable toxicities. Here, we show that the cyclic 9-amino acid peptide CARGGLKSC (CARG), identified via phage display on Staphylococcus aureus (S. aureus) bacteria and through in vivo screening in mice with S. aureus-induced lung infections, increases the antibacterial activity of CARG-conjugated vancomycin-loaded nanoparticles in S. aureus-infected tissues and reduces the needed overall systemic dose, minimizing side effects. CARG binds specifically to S. aureus bacteria but not Pseudomonas bacteria in vitro, selectively accumulates in S. aureus-infected lungs and skin of mice but not in non-infected tissue and Pseudomonas-infected tissue, and significantly enhances the accumulation of intravenously injected vancomycin-loaded porous silicon nanoparticles bearing the peptide in S. aureus-infected mouse lung tissue. The targeted nanoparticles more effectively suppress staphylococcal infections in vivo relative to equivalent doses of untargeted vancomycin nanoparticles or of free vancomycin. The therapeutic delivery of antibiotic-carrying nanoparticles bearing peptides targeting infected tissue may help combat difficult-to-treat infections.

20.
Nat Commun ; 8(1): 343, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839238

ABSTRACT

In vivo tumor imaging with nanoprobes suffers from poor tumor specificity. Here, we introduce a nanosystem, which allows selective background quenching to gain exceptionally tumor-specific signals. The system uses near-infrared quantum dots and a membrane-impermeable etchant, which serves as a cation donor. The etchant rapidly quenches the quantum dots through cation exchange (ionic etching), and facilitates renal clearance of metal ions released from the quantum dots. The quantum dots are intravenously delivered into orthotopic breast and pancreas tumors in mice by using the tumor-penetrating iRGD peptide. Subsequent etching quenches excess quantum dots, leaving a highly tumor-specific signal provided by the intact quantum dots remaining in the extravascular tumor cells and fibroblasts. No toxicity is noted. The system also facilitates the detection of peritoneal tumors with high specificity upon intraperitoneal tumor targeting and selective etching of excess untargeted quantum dots. In vivo cation exchange may be a promising strategy to enhance specificity of tumor imaging.The imaging of tumors in vivo using nanoprobes has been challenging due to the lack of sufficient tumor specificity. Here, the authors develop a tumor-specific quantum dot system that permits in vivo cation exchange to achieve selective background quenching and high tumor-specific imaging.


Subject(s)
Cations/chemistry , Diagnostic Imaging/methods , Neoplasms/diagnostic imaging , Quantum Dots , Animals , Cell Line, Tumor , Humans , Mice , Nanomedicine/methods , Neoplasms/pathology , Reproducibility of Results , Sensitivity and Specificity , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL