Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
J Control Release ; 341: 646-660, 2022 01.
Article in English | MEDLINE | ID: mdl-34921973

ABSTRACT

We report copper(II) arsenite (CuAS)-integrated polymer micelles (CuAS-PMs) as a new class of Fenton-like catalytic nanosystem that can display reactive oxygen species (ROS)-manipulating anticancer therapeutic activity. CuAS-PMs were fabricated through metal-catechol chelation-based formation of the CuAS complex on the core domain of poly (ethylene glycol)-b-poly(3,4-dihydroxy-L-phenylalanine) (PEG-PDOPA) copolymer micelles. CuAS-PMs maintained structural robustness under serum conditions. The insoluble state of the CuAS complex was effectively retained at physiological pH, whereas, at endosomal pH, the CuAS complex was ionized to release arsenite and cuprous Fenton catalysts (Cu+ ions). Upon endocytosis, CuAS-PMs simultaneously released hydrogen peroxide (H2O2)-generating arsenite and Fenton-like reaction-catalyzing Cu+ ions in cancer cells, which synergistically elevated the level of highly cytotoxic hydroxyl radicals (•OH), thereby preferentially killing cancer cells. Animal experiments demonstrated that CuAS-PMs could effectively suppress the growth of solid tumors without systemic in vivo toxicity. The design rationale of CuAS-PMs may provide a promising strategy to develop diverse oxidative stress-amplifying agents with great potential in cancer-specific therapy.


Subject(s)
Antineoplastic Agents , Arsenites , Nanoparticles , Animals , Antineoplastic Agents/chemistry , Arsenites/pharmacology , Copper , Hydrogen Peroxide/chemistry , Nanoparticles/chemistry , Oxidative Stress
2.
Pharmaceutics ; 12(7)2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32635231

ABSTRACT

Theranostic nanoparticles can deliver therapeutic agents as well as diverse imaging agents to tumors. The enhanced permeation and retention (EPR) effect is regarded as a crucial mechanism for the tumor-targeted delivery of nanoparticles. Although a large number of studies of the EPR effect of theranostic nanoparticles have been performed, the effect of the change in the body size of the host on the EPR effect is not fully understood. In this regard, comparative research is needed on the behavior of nanoparticles in large animals for developing the nanoparticles to the clinical stage. In this study, we prepared fluorophore (indocyanine green (ICG) or cyanine 5.5 (Cy5.5))-conjugated glycol chitosan nanoparticles (CNPs) for comparing the tumor-targeting efficacy in VX2 tumor-bearing mouse and rabbit models. As expected, the CNPs formed nano-sized spherical nanoparticles and were stable for 8 days under aqueous conditions. The CNPs also exhibited dose-dependent cellular uptake into VX2 tumor cells without cytotoxicity. The half-life of the near-infrared fluorescence (NIRF) signals in the blood were 3.25 h and 4.73 h when the CNPs were injected into mice and rabbits, respectively. Importantly, the CNPs showed excellent tumor accumulation and prolonged biodistribution profiles in both the VX2 tumor-bearing mouse and rabbit models, wherein the tumor accumulation was maximized at 48 h and 72 h, respectively. Based on the excellent tumor accumulation of the CNPs, finally, the CNPs were used in the image-guided surgery of the rabbit orthotopic VX2 lung tumor model. The lung tumor tissue was successfully removed based on the NIRF signal from the CNPs in the tumor tissue. This study shows that CNPs can be potentially used for tumor theragnosis in small animals and large animals.

3.
Sci Rep ; 7(1): 16635, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29192289

ABSTRACT

Apoptosis is one of the most important intracellular events in living cell, which is a programmed cell death interrelated with caspase enzyme activity for maintaining homeostasis in multicellular organisms. Therefore, direct apoptosis imaging of living cells can provide enormous advantages for diagnosis, drug discovery, and therapeutic monitoring in various diseases. However, a method of direct apoptosis imaging has not been fully validated, especially for live cells in in vitro and in vivo. Herein, we developed a new apoptosis imaging technology via a direct visualization of active caspase-3/-7 activity in living cells. For this, we synthesized a caspase-3/-7-specific cleavable peptide (KGDEVD) conjugated triacetylated N-azidoacetyl-D-mannosamine (Apo-S-Ac3ManNAz), wherein the Apo-S-Ac3ManNAz can be cleaved by the active caspase-3/-7 in live apoptotic cells and the cleaved Ac3ManNAz molecules can further generate targetable azido groups (N3) on the living cell surface. Importantly, the azido groups on the apoptotic tumor cells could be visualized with Cy5.5-conjugated dibenzylcyclooctyne (DBCO-Cy5.5) via bioorthogonal click chemistry in vitro cell culture condition and in vivo tumor-bearing mice. Therefore, our Apo-S-Ac3ManNAz can be utilized for the further applications in tumor therapy as a monitoring tool for anticancer efficacy and optimization of anticancer new drugs in cell culture system and in tumor-bearing mice.


Subject(s)
Apoptosis , Caspase 3/metabolism , Caspase 7/metabolism , Molecular Imaging , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor/methods , Enzyme Activation , Fluorescent Dyes , Humans , Mice , Molecular Imaging/methods , Staining and Labeling , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
4.
Eur J Pharmacol ; 812: 243-249, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28734931

ABSTRACT

Cancer is a life-threatening disease, and the occurrence of metastasis, which increases the lethality of primary tumors, is increasing. The epithelial-to-mesenchymal transition (EMT) is a biological process by which epithelial cells lose cell-cell adhesion properties and acquire mesenchymal properties, including motility and invasiveness. EMT is considered an early stage of metastasis; therefore, inhibiting EMT may be an effective anticancer therapy. In the present study, the antimetastatic effect of piperlongumine (PL) was assessed in human cancer cells. PL is a single component isolated from long pepper (Piper longum) and it has been studied for its antibacterial, antiangiogenic, and antidiabetic activities. Migration assays (wound healing assay) and transwell invasion assays showed that PL inhibited the migration and invasion of cancer cells. Western blotting and immunofluorescence imaging showed that TGF-ß upregulated the transcription factors Snail1 and Twist1 and downregulated E-cadherin, a marker of epithelial cells, inducing EMT. PL might inhibit TGF-ß-induced EMT by downregulating Snail1 and Twist1 and upregulating E-cadherin in cancer cells. In summary, PL might inhibit TGF-ß-induced EMT, suggesting that it is a promising anticancer agent.


Subject(s)
Cadherins/metabolism , Dioxolanes/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Twist-Related Protein 1/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , MCF-7 Cells , Neoplasm Invasiveness
5.
Chem Pharm Bull (Tokyo) ; 64(8): 1108-17, 2016.
Article in English | MEDLINE | ID: mdl-27477648

ABSTRACT

The purposes of the present study were to develop a self-microemulsifying drug delivery system (SMEDDS) containing bortezomib, a proteasome inhibitor. The solubility of the drug was evaluated in 15 pharmaceutical excipients. Combinations of oils, surfactants and cosurfactants were screened by drawing pseudo-ternary phase diagrams. The system exhibiting the largest region of microemulsion was considered optimal. Bortezomib SMEDDS spontaneously formed a microemulsion when diluted with an aqueous medium with a median droplet size of approximately 20-30 nm. In vitro release studies showed that the SMEDDS had higher initial release rates for the drug when compared with the raw drug material alone. Measurement of the viscosity, size, and ion conductivity indicated that a phase inversion from water in an oil system to oil in a water system occurred when the weight ratio of the water exceeded 30% of the entire microemulsion system. In a pharmacokinetics study using rats, the bortezomib microemulsion failed to improve the bioavailability of the drug. The reason was assumed to be degradation of the drug in the microemulsion in the gastrointestinal tract. However, bortezomib in Labrasol(®) solution (an aqueous solution containing 0.025% Labrasol(®)) showed significantly increased area under the curve from 0-24 h (AUC0-24 h) and maximum plasma concentration (Cmax) values compared to the drug suspension. The findings of this study imply that oral delivery of a bortezomib and colloidal system containing Labrasol(®) could be an effective strategy for the delivery of bortezomib.


Subject(s)
Bortezomib/administration & dosage , Bortezomib/pharmacokinetics , Drug Compounding , Drug Delivery Systems/methods , Animals , Biological Availability , Emulsions , Gastrointestinal Tract/metabolism , Glycerides/chemistry , Male , Particle Size , Rats , Rats, Sprague-Dawley , Solubility , Surface Properties , Viscosity
6.
J Mater Chem B ; 4(7): 1219-1227, 2016 Feb 21.
Article in English | MEDLINE | ID: mdl-32262977

ABSTRACT

In this work, we have developed photosensitizer-loaded bubble-generating calcium carbonate (CaCO3)-mineralized nanoparticles that have potential for ultrasound imaging (US)-guided photodynamic therapy (PDT) of tumors. A photosensitizer, chlorin e6 (Ce6)-loaded CaCO3-mineralized nanoparticles (Ce6-BMNs), was prepared using an anionic block copolymer-templated in situ mineralization method. Ce6-BMNs were composed of the Ce6-loaded CaCO3 core and the hydrated poly(ethylene glycol) (PEG) shell. Ce6-BMNs exhibited excellent stability under serum conditions. Ce6-BMNs showed enhanced echogenic US signals at tumoral acid pH by generating carbon dioxide (CO2) bubbles. Ce6-BMNs effectively inhibited Ce6 release at physiological pH (7.4). At a tumoral acidic pH (6.4), Ce6 release was accelerated with CO2 bubble generation due to the dissolution of the CaCO3 mineral core. Upon irradiation of Ce6-BMN-treated MCF-7 breast cancer cells, the cell viability dramatically decreased with increasing Ce6 concentration. The phototoxicity of the Ce6-BMNs was much higher than that of free Ce6. On the basis of tumoral pH-responsive CO2 bubble-generation and simultaneous Ce6 release at the target tumor site, these CaCO3 mineralized nanoparticles can be considered as promising theranostic nanoparticles for US imaging-guided PDT in the field of tumor therapy.

7.
Int J Mol Med ; 36(6): 1601-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26498588

ABSTRACT

Shikonin (SK), a natural naphthoquinone isolated from the Chinese medicinal herb, has been known to suppress the proliferation of several cancer cells. However, its role in the epithelial mesenchymal transition (EMT) has yet to be demonstrated. The aim of the present study was to examine the effects of SK on EMT. Lipopolysaccharide (LPS) induced EMT-like phenotypic changes, enhancing cell migration and invasion. SK markedly reduced the expression of the LPS-induced EMT markers, including N-cadherin in MDA-MB­231 cells, and increased the expression of E-cadherin in MCF-7 cells. SK also inhibited cell migration and invasion in vitro. The effects of SK on the LPS-induced EMT were mediated by the inactivation of the NF-κB-Snail signaling pathway. The results provided new evidence that SK suppresses breast cancer cell invasion and migration by inhibiting the EMT. Therefore, SK is a potentially effective anticancer agent for breast tumors, by inhibiting metastasis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lipopolysaccharides/pharmacology , Naphthoquinones/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Microscopy, Fluorescence , Snail Family Transcription Factors , Transcription Factor RelA/metabolism , Transcription Factors/metabolism
8.
Biochem Biophys Res Commun ; 465(1): 71-6, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26235879

ABSTRACT

Shikonin, a natural naphthoquinone isolated from the Chinese traditional medicine Zi Cao (purple gromwell), is known to suppress the growth of several cancer cell types. In this study, we evaluated the pro-apoptotic effects of shikonin on MCF-7 and HeLa cells, and investigated the underlying mechanism. Shikonin-induced apoptosis was associated with activation of caspase-3, poly(ADP-ribose) polymerase (PARP) cleavage, up-regulation of p73, and down-regulation of BCL-2. Shikonin also induced up-regulation of the tumor suppressor gene, p16(INK4A). Increasing transcriptional activity of p16(INK4A) by shikonin treatment, we observed in luciferase promoter assay, reflects reduced promoter binding by down-regulation of ICBP90 (inverted CCAAT box binding protein, 90 kDa), which are involved in down-regulation of its partner, DNMT1 (DNA methyltransferase 1). On the basis of these results, we conclude that shikonin causes apoptosis via a p73-related, caspase-3-dependent pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/agonists , Gene Expression Regulation, Neoplastic , Naphthoquinones/pharmacology , Nuclear Proteins/agonists , Tumor Suppressor Proteins/agonists , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cyclin-Dependent Kinase Inhibitor p16/agonists , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, Reporter , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , MCF-7 Cells , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases
9.
J Pharm Sci ; 104(8): 2611-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26053132

ABSTRACT

A pH-responsive nanofibrous mesh was prepared for the controlled release of antibiotics in response to pH changes. Eudragit EPO (EPO) and Eudragit L100 (L100) were injected through inner and outer needle and simultaneously electrospun through coaxial nozzles composed of inner and outer needles. Various amounts of EPO and L100 were coejected with tetracycline through the needle and simultaneously electrospun to the fibrous meshes. The mass erosion rates of the meshes at pH 6.0 gradually decreased as the amounts of EPO increased, whereas those at pH 2.0 showed negligible differences; these differences were confirmed by scanning electron microscopy and monitoring the dry weight changes. At pH 6.0, the fibrous structures of the meshes rapidly disappeared compared to those under acidic conditions because Eudragit L100 is localized to the shell of the nanofiber during the electrospinning process. Both the pH changes and the blend ratio of the two polymers significantly affected the tetracycline release; tetracycline was rapidly released from the meshes at pH 6.0, whereas the release rates were attenuated at pH 2.0. Tetracycline was released faster from the mesh at higher blend ratios of EPO for both pH values. The electrostatic interaction between EPO and L100 is expected to yield different release profiles of tetracycline. Consequently, higher amounts of encapsulated drugs were released from the mesh at neutral pH and successfully inhibited bacterial growth.


Subject(s)
Anti-Bacterial Agents/chemistry , Biocompatible Materials/chemistry , Drug Delivery Systems , Nanofibers/chemistry , Polymethacrylic Acids/chemistry , Tetracycline/chemistry , Administration, Topical , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Drug Compounding , Escherichia coli/drug effects , Escherichia coli/growth & development , Hydrogen-Ion Concentration , Kinetics , Microscopy, Electron, Scanning , Nanofibers/ultrastructure , Solubility , Static Electricity , Tetracycline/administration & dosage , Tetracycline/pharmacology , Transdermal Patch
10.
ACS Nano ; 9(1): 134-45, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25559896

ABSTRACT

We report a theranostic nanoparticle that can express ultrasound (US) imaging and simultaneous therapeutic functions for cancer treatment. We developed doxorubicin-loaded calcium carbonate (CaCO3) hybrid nanoparticles (DOX-CaCO3-MNPs) through a block copolymer templated in situ mineralization approach. The nanoparticles exhibited strong echogenic signals at tumoral acid pH by producing carbon dioxide (CO2) bubbles and showed excellent echo persistence. In vivo results demonstrated that the DOX-CaCO3-MNPs generated CO2 bubbles at tumor tissues sufficient for echogenic reflectivity under a US field. In contrast, the DOX-CaCO3-MNPs located in the liver or tumor-free subcutaneous area did not generate the CO2 bubbles necessary for US contrast. The DOX-CaCO3-MNPs could also trigger the DOX release simultaneously with CO2 bubble generation at the acidic tumoral environment. The DOX-CaCO3-MNPs displayed effective antitumor therapeutic activity in tumor-bearing mice. The concept described in this work may serve as a useful guide for development of various theranostic nanoparticles for US imaging and therapy of various cancers.


Subject(s)
Calcium Carbonate/chemistry , Carbon Dioxide/chemistry , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/drug therapy , Drug Carriers/chemistry , Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Transport , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers/metabolism , Drug Liberation , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Male , Mice , Models, Molecular , Molecular Conformation , Ultrasonography
11.
J Control Release ; 198: 1-9, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25481438

ABSTRACT

P-glycoprotein (Pgp) mediated multi-drug resistance (MDR) is a major cause of failure in chemotherapy. In this study, small interfering RNA (siRNA) for Pgp down-regulation was delivered to tumors to overcome MDR in cancer. To achieve an efficient siRNA delivery in vivo, self-polymerized 5'-end thiol-modified siRNA (poly-siRNA) was incorporated in tumor targeting glycol chitosan nanoparticles. Pgp-targeted poly-siRNA (psi-Pgp) and thiolated glycol chitosan polymers (tGC) formed stable nanoparticles (psi-Pgp-tGC NPs), and the resulting nanoparticles protected siRNA molecules from enzymatic degradation. The psi-Pgp-tGC NPs could release functional siRNA molecules after cellular delivery, and they were able to facilitate siRNA delivery to Adriamycin-resistant breast cancer cells (MCF-7/ADR). After intravenous administration, the psi-Pgp-tGC NPs accumulated in MCF-7/ADR tumors and down-regulated P-gp expression to sensitize cancer cells. Consequently, chemo-siRNA combination therapy significantly inhibited tumor growth without systemic toxicity. These psi-Pgp-tGC NPs showed great potential as a supplementary therapeutic agent for drug-resistant cancer.


Subject(s)
Chitosan/administration & dosage , Drug Resistance, Neoplasm , Nanoparticles/administration & dosage , Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Gene Silencing , Humans , MCF-7 Cells , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Oncol Rep ; 31(6): 2827-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24789371

ABSTRACT

Shikonin, a natural naphthoquinone isolated from a traditional Chinese medicinal herb, has been reported to promote tumor cell death. However, there are few reports concerning its effect on metastasis-related cell invasion and migration behavior. In the present study, we investigated the effect of shikonin on human breast cancer invasion and migration. We found that shikonin inhibited phorbol 12-myristate 13-acetate (PMA)-induced cell migration and invasion in MCF-7 breast cancer cells, which was correlated with modulation of matrix metalloproteinase-9 (MMP-9) through suppression of both expression and proteolytic and promoter activity. We also found that shikonin inhibited both MMP-9 expression and promoter activity in MDA-MB­231 cells with high metastatic potential. These results indicated that shikonin induces the suppression of migration and invasion through modulation of MMP-9 in human breast cancer cells. Therefore, shikonin may be a potential anticancer drug for human breast cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors/administration & dosage , Naphthoquinones/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/administration & dosage , Tetradecanoylphorbol Acetate/analogs & derivatives
13.
ACS Nano ; 8(5): 4257-67, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24754537

ABSTRACT

In general, traditional gene carriers contain strong cationic charges to efficiently load anionic genes, but this cationic character also leads to destabilization of plasma membranes and causes severe cytotoxicity. Here, we developed a PCR-based nanofactory as a safe gene delivery system. A few template plasmid DNA can be amplified by PCR inside liposomes about 200 nm in diameter, and the quantity of loaded genes highly increased by more than 8.8-fold. The liposome membrane was composed of neutral lipids free from cationic charges. Consequently, this system is nontoxic, unlike other traditional cationic gene carriers. Intense red fluorescent protein (RFP) expression in CHO-K1 cells showed that the amplified genes could be successfully transfected to cells. Animal experiments with the luciferase gene also showed in vivo gene expression by our system without toxicity. We think that this PCR-based nanofactory system can overcome the toxicity problem that is the critical limitation of current gene delivery to clinical application.


Subject(s)
DNA/chemistry , Gene Transfer Techniques , Liposomes/chemistry , Nanotechnology/methods , Animals , Anions , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival , Cricetinae , Cricetulus , Genetic Vectors/chemistry , Humans , Interleukin-12/metabolism , Lipids/chemistry , Luminescent Proteins/chemistry , Microscopy, Fluorescence , Nanoparticles/chemistry , Neoplasms/metabolism , Polymerase Chain Reaction , Transfection , Red Fluorescent Protein
14.
Theranostics ; 4(4): 420-31, 2014.
Article in English | MEDLINE | ID: mdl-24578725

ABSTRACT

Cell labeling and tracking are important processes in understanding biologic mechanisms and the therapeutic effect of inoculated cells in vivo. Numerous attempts have been made to label and track inoculated cells in vivo; however, these methods have limitations as a result of their biological effects, including secondary phagocytosis of macrophages and genetic modification. Here, we investigated a new cell labeling and tracking strategy based on metabolic glycoengineering and bioorthogonal click chemistry. We first treated cells with tetra-acetylated N-azidoacetyl-D-mannosamine to generate unnatural sialic acids with azide groups on the surface of the target cells. The azide-labeled cells were then transplanted to mouse liver, and dibenzyl cyclooctyne-conjugated Cy5 (DBCO-Cy5) was intravenously injected into mice to chemically bind with the azide groups on the surface of the target cells in vivo for target cell visualization. Unnatural sialic acids with azide groups could be artificially induced on the surface of target cells by glycoengineering. We then tracked the azide groups on the surface of the cells by DBCO-Cy5 in vivo using bioorthogonal click chemistry. Importantly, labeling efficacy was enhanced and false signals by phagocytosis of macrophages were reduced. This strategy will be highly useful for cell labeling and tracking.


Subject(s)
Cell Tracking/methods , Click Chemistry , Macrophages, Peritoneal/cytology , Phagocytosis , Staining and Labeling/methods , Animals , Azides/chemistry , Cell Line, Tumor , Cell Transplantation , Flow Cytometry/methods , Hexosamines/chemistry , Humans , Liver/cytology , Male , Mice , Mice, Nude , Molecular Imaging/methods , Sialic Acids/chemistry
15.
Chem Commun (Camb) ; 50(33): 4351-3, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24643304

ABSTRACT

We report on pH-responsive core-shell polymer micelles with catechol-Fe(3+) coordinated core cross-links, which provide robustness to drug-loaded polymer micelles and allow the facilitated intracellular release of loaded anticancer drugs in response to an endosomal acidic pH.


Subject(s)
Ferric Compounds/chemistry , Micelles , Polymers/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catechols/chemistry , Cell Proliferation/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , Ligands , MCF-7 Cells
16.
Int J Oncol ; 44(5): 1455-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24573652

ABSTRACT

Shikonin, a natural naphthoquinone isolated from the traditional Chinese medicine Zi Cao (gromwell), has been shown to possess tumor cell killing activity. The human androgen receptor (AR) is a nuclear transcription factor that serves as a major therapeutic target for prostate cancer. However, AR regulation by shikonin has not been reported. We investigated the effects of shikonin on the growth of prostate cancer cells. We observed that shikonin decreased the expression of AR at both the mRNA and the protein levels in LNCaP and 22RV1 human prostate cancer cells. The results from a luciferase assay showed that shikonin decreased the transcriptional activity of AR. Moreover, shikonin treatment inhibited AR target gene expression, PSA and growth inhibition of prostate cancer cells. In conclusion, the present study shows for the first time that shikonin treatment causes transcriptional repression of AR and inhibition of its nuclear localization in human prostate cancer cells. We propose that shikonin, an anticancer drug extracted from natural sources, induces inhibition of cell growth through modulation of AR in androgen-responsive prostate cancer cells and is a candidate for use in cancer chemotherapy for human prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Naphthoquinones/pharmacology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostate-Specific Antigen/metabolism , Receptors, Androgen/genetics , Signal Transduction/drug effects
17.
ACS Nano ; 8(3): 2048-63, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24499346

ABSTRACT

Tumor-targeting strategies for nanoparticles have been predominantly based on optimization of physical properties or conjugation with biological ligands. However, their tumor-targeting abilities remain limited and insufficient. Furthermore, traditional biological binding molecules have intrinsic limitations originating from the limited amount of cellular receptors and the heterogeneity of tumor cells. Our two-step in vivo tumor-targeting strategy for nanoparticles is based on metabolic glycoengineering and click chemistry. First, an intravenous injection of precursor-loaded glycol chitosan nanoparticles generates azide groups on tumor tissue specifically by the enhanced permeation and retention (EPR) effect followed by metabolic glycoengineering. These 'receptor-like' chemical groups then enhance the tumor-targeting ability of drug-containing nanoparticles by copper-free click chemistry in vivo during a second intravenous injection. The advantage of this protocol over traditional binding molecules is that there are significantly more binding molecules on the surface of most tumor cells regardless of cell type. The subsequent enhanced tumor-targeting ability can significantly enhance the cancer therapeutic efficacy in animal studies.


Subject(s)
Click Chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Targeted Therapy/methods , Nanoparticles/chemistry , Nanoparticles/metabolism , Animals , Azides/chemistry , Cell Line, Tumor , Chitosan/chemistry , Hexosamines/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Lung Neoplasms/pathology , Male , Mice , Nanoparticles/therapeutic use
18.
Pharm Res ; 31(6): 1418-25, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23934255

ABSTRACT

PURPOSE: The application of gold nanoparticles (AuNPs) in biomedical field was limited due to the low stability in the biological condition. Herein, to enhance stability and tumor targeting ability of AuNPs, their surface was modified with biocompatible glycol chitosan (GC) and the in vivo biodistribution of GC coated AuNPs (GC-AuNPs) were studied through computed tomography (CT). METHODS: Polymer-coated gold nanoparticles were produced using GC as a reducing agent and a stabilizer. Their feasibility in biomedical application was explored through CT in tumor-bearing mice. RESULTS: Stability of gold nanoparticles increased in the physiological condition due to the GC coating layer on the surface. Tomographic images of tumor were successfully obtained in the tumor-xenografted animal model when the GC-AuNPs were used as a CT contrast agent. The tumor targeting property of the gold nanoparticles was due to the properties of GC because GC-AuNPs were accumulated in the tumor, while most of heparin-coated nanoparticles were found in the liver and spleen. CONCLUSIONS: The polymer properties on the surface played an important role in the behavior of gold nanoparticles in the biological condition and the enhanced stability and tumor targeting property of nanoparticles were inherited from GC on the surface.


Subject(s)
Biocompatible Materials , Chitosan/chemistry , Contrast Media/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Neoplasms/diagnosis , Animals , Cell Survival/drug effects , Chitosan/toxicity , Contrast Media/pharmacokinetics , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Mice , Neoplasms/pathology , Radiography , Tissue Distribution
19.
J Nanosci Nanotechnol ; 14(10): 7606-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25942834

ABSTRACT

Cancer cells overexpressing folate receptors have been targeted using a folate decorated carriers for anti-cancer drugs in aims to overcome the tissue non-specificity of anti-cancer agents. We here prepared magnetic nanoparticles and surface-decorated them with different amounts of folate to optimize the number of the immobilized folate on the carriers for superior targeting effects. Magnetic nanoparticles were prepared by oxidizing ferric or ferrous chloride solution to iron oxide in the presence of poly(vinyl alcohol). The magnetic nanoparticles were functionalized with primary amines for subsequent reactions with the different feed ratios of the activated folate. The magnetization degree of the folate magnetic magnetization were slightly decreased when the folate on the particles were increased. Intracellular uptakes of the nanoparticles were shown to be increased and become saturated dependent on the amounts of the surface-immobilized folate. The folate-decorated magnetic nanoparticles showed negligible cytotoxicity against KB cells from 5 µg to 35 µg of the nanoparticle weights.


Subject(s)
Drug Carriers/chemistry , Folic Acid/chemistry , Magnetite Nanoparticles/chemistry , Biological Transport , Cell Survival/drug effects , Drug Carriers/metabolism , Drug Carriers/toxicity , Folic Acid/metabolism , Folic Acid/toxicity , Humans , Intracellular Space/metabolism , KB Cells , Structure-Activity Relationship , Surface Properties
20.
Biomaterials ; 35(7): 2302-11, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24360720

ABSTRACT

An increasing number of treatments of metastases rely on diagnostics and imaging these days. The facts that the activity of cathepsin B (CB) is markedly linked to the metastatic process and that CB is found highly expressed in the pericellular regions in this process make CB an attractive target for diagnosing metastases. We have developed a CB-sensitive nanoprobe (CB-CNP) consisting of self-quenched CB-sensitive fluorogenic peptide probes conjugated onto the surface of tumor-targeting glycol chitosan nanoparticles (CNPs). The freshly prepared CB-CNP formed a spherical nanoparticle structure (280 nm in diameter) and the fluorescence intensity of CB-CNP was strongly quenched in physiological condition. However, self-quenched CB-CNP boosted strong fluorescence signals in the presence of CB, not of cathepsin l or cathepsin d, due to the CB-specific cleavage of self-quenched peptide probes. Importantly, the intravenously injected CB-CNP demonstrated the potential to discriminate metastases in vivo in three metastatic mouse models, including 4T1-luc2 liver metastases, RFP-B16F10 lung metastases and HT1080 peritoneal metastases. Indeed, Western blot analysis confirmed that the CB expression of metastases had increased compared to normal organ in these metastatic mouse models. CB-CNPs may be useful for depicting metastases through non-invasive CB molecular imaging.


Subject(s)
Cathepsin B/metabolism , Fluorescent Dyes/chemistry , Models, Biological , Nanostructures , Neoplasm Metastasis , Optical Imaging/methods , Cell Line, Tumor , Chitosan/chemistry , Colonic Neoplasms/pathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...