Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Nanobiotechnology ; 21(1): 82, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36894943

ABSTRACT

BACKGROUND: Glycol chitosan nanoparticles (CNPs) have emerged as an effective drug delivery system for cancer diagnosis and treatment. Although they have great biocompatibility owing to biodegradable chemical structure and low immunogenicity, sufficient information on in vivo toxicity to understand the potential risks depending on the repeated high-dose have not been adequately studied. Herein, we report the results of in vivo toxicity evaluation for CNPs focused on the number and dose of administration in healthy mice to provide a toxicological guideline for a better clinical application of CNPs. RESULTS: The CNPs were prepared by conjugating hydrophilic glycol chitosan with hydrophobic 5ß-cholanic acid and the amphiphilic glycol chitosan-5ß-cholanic acid formed self-assembled nanoparticles with its concentration-dependent homogeneous size distributions (265.36-288.3 nm) in aqueous condition. In cell cultured system, they showed significantly high cellular uptake in breast cancer cells (4T1) and cardiomyocytes (H9C2) than in fibroblasts (L929) and macrophages (Raw264.7) in a dose- and time-dependent manners, resulting in severe necrotic cell death in H9C2 at a clinically relevant highly concentrated condition. In particular, when the high-dose (90 mg/kg) of CNPs were intravenously injected into the healthy mice, considerable amount was non-specifically accumulated in major organs (liver, lung, spleen, kidney and heart) after 6 h of injection and sustainably retained for 72 h. Finally, repeated high-dose of CNPs (90 mg/kg, three times) induced severe cardiotoxicity accompanying inflammatory responses, tissue damages, fibrotic changes and organ dysfunction. CONCLUSIONS: This study demonstrates that repeated high-dose CNPs induce severe cardiotoxicity in vivo. Through the series of toxicological assessments in the healthy mice, this study provides a toxicological guideline that may expedite the application of CNPs in the clinical settings.


Subject(s)
Chitosan , Nanoparticles , Neoplasms , Mice , Animals , Cardiotoxicity/etiology , Drug Delivery Systems , Chitosan/toxicity , Chitosan/chemistry , Nanoparticles/chemistry
2.
J Control Release ; 351: 37-49, 2022 11.
Article in English | MEDLINE | ID: mdl-36089170

ABSTRACT

Many preclinically tested nanoparticles in existing animal models fail to be directly translated into clinical applications because of their poor resemblance to human cancer. Herein, the enhanced permeation and retention (EPR) effect of glycol chitosan nanoparticles (CNPs) in different tumor microenvironments (TMEs) was compared using different pancreatic tumor models, including pancreatic cancer cell line (BxPC3), patient-derived cancer cell (PDC), and patient-derived xenograft (PDX) models. CNPs were intravenously injected into different tumor models, and their accumulation efficiency was evaluated using non-invasive near-infrared fluorescence (NIRF) imaging. In particular, differences in angiogenic vessel density, collagen matrix, and hyaluronic acid content in tumor tissues of the BxPC3, PDC, and PDX models greatly affected the tumor-targeting efficiency of CNPs. In addition, different PDX models were established using different tumor tissues of patients to predict the clinical EPR effect of CNPs in inter-patient TMEs, wherein the gene expression levels of PECAM1, COL4A1, and HAS1 in human tumor tissues were observed to be closely related to the EPR effect of CNPs in PDX models. The results suggested that the PDX models could mimic inter-patient TMEs with different blood vessel structures and extracellular matrix (ECM) content that critically affect the tumor-targeting ability of CNPs in different pancreatic PDX models. This study provides a better understanding of the heterogeneity and complexity of inter-patient TMEs that can predict the response of various nanoparticles in individual tumors for personalized cancer therapy.


Subject(s)
Nanoparticles , Neoplasms , Animals , Humans , Heterografts , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/metabolism , Tumor Microenvironment , Extracellular Matrix/metabolism , Disease Models, Animal , Xenograft Model Antitumor Assays
3.
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.

4.
J Control Release ; 267: 223-231, 2017 Dec 10.
Article in English | MEDLINE | ID: mdl-28917532

ABSTRACT

In cancer theranostics, the main strategy of nanoparticle-based targeted delivery system has been understood by enhanced permeability and retention (EPR) effect of macromolecules. Studies on diverse nanoparticles provide a better understanding of different EPR effects depending on their structure, physicochemical properties, and chemical modifications. Recently the tumor microenvironment has been considered as another important factor for determining tumor-targeted delivery of nanoparticles, but the correlation between EPR effects and tumor microenvironment has not yet been fully elucidated. Herein, ectopic subcutaneous tumor models presenting different tumor microenvironments were established by inoculation of SCC7, U87, HT29, PC3, and A549 cancer cell lines into athymic nude mice, respectively. In the five different types of tumor-bearing mice, tumor-targeted delivery of self-assembled glycol chitosan nanoparticles (CNPs) were comparatively evaluated to identify the correlation between the tumor microenvironments and targeted delivery of CNPs. As a result, neovascularization and extents of intratumoral extracellular matrix (ECM) were both important in determining the tumor targeted delivery of CNPs. The EPR effect was maximized in the tumors which include large extent of angiogenic blood vessels and low intratumoral ECM content. This comprehensive study provides substantial evidence that the EPR effects based tumor-targeted delivery of nanoparticles can be different depending on the tumor microenvironment in individual tumors. To overcome current limitations in clinical nanomedicine, the tumor microenvironment of the patients and EPR effects in clinical tumors should also be carefully studied.


Subject(s)
Chitosan/administration & dosage , Drug Delivery Systems , Nanoparticles/administration & dosage , Neoplasms/metabolism , Tumor Microenvironment , Animals , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacokinetics , Extracellular Matrix , Female , Humans , Mice, Nude , Microvessels/drug effects , Nanoparticles/chemistry , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Tissue Distribution , Tumor Burden/drug effects
5.
Int J Nanomedicine ; 12: 6089-6105, 2017.
Article in English | MEDLINE | ID: mdl-28860768

ABSTRACT

Recent progress in nanomedicine has shown a strong possibility of targeted therapy for obstinate chronic lung diseases including idiopathic pulmonary fibrosis (IPF). IPF is a fatal lung disease characterized by persistent fibrotic fibroblasts in response to type I collagen-rich extracellular matrix. As a pathological microenvironment is important in understanding the biological behavior of nanoparticles, in vitro cellular uptake of glycol chitosan nanoparticles (CNPs) in human lung fibroblasts was comparatively studied in the presence or absence of type I collagen matrix. Primary human lung fibroblasts from non-IPF and IPF patients (n=6/group) showed significantly increased cellular uptake of CNPs (>33.6-78.1 times) when they were cultured on collagen matrix. To elucidate the underlying mechanism of enhanced cellular delivery of CNPs in lung fibroblasts on collagen, cells were pretreated with chlorpromazine, genistein, and amiloride to inhibit clathrin-mediated endocytosis, caveolae-mediated endocytosis, and macropinocytosis, respectively. Amiloride pretreatment remarkably reduced the cellular uptake of CNPs, suggesting that lung fibroblasts mainly utilize the macropinocytosis-dependent mechanism when interacted with collagen. In addition, the internalization of CNPs was predominantly suppressed by a phosphoinositide 3-kinase (PI3K) inhibitor in IPF fibroblasts, indicating that enhanced PI3K activity associated with late-stage macropinocytosis can be particularly important for the enhanced cellular delivery of CNPs in IPF fibroblasts. Our study strongly supports the concept that a pathological microenvironment which surrounds lung fibroblasts has a significant impact on the intracellular delivery of nanoparticles. Based on the property of enhanced intracellular delivery of CNPs when fibroblasts are made to interact with a collagen-rich matrix, we suggest that CNPs may have great potential as a drug-carrier system for targeting fibrotic lung fibroblasts.


Subject(s)
Chitosan/administration & dosage , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Nanoparticles/administration & dosage , Amiloride/pharmacology , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacokinetics , Chlorpromazine/pharmacology , Collagen Type I/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/methods , Endocytosis/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Genistein/pharmacology , Humans , Lung/metabolism , Nanoparticles/chemistry , Phosphatidylinositol 3-Kinases/metabolism
6.
Biochem Biophys Res Commun ; 489(1): 35-41, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28533089

ABSTRACT

A variety of VEGF inhibitors have been reported to treat cancers by suppressing tumor angiogenesis. Bevacizumab, a monoclonal VEGF antibody, was the first FDA approved anti-angiogenic agent for cancer treatments. However, bevacizumab shows modest therapeutic efficiency and often cause resistant problem in significant populations of cancer patients. To solve these problem, we investigated the therapeutic efficacy of siRNA drugs targeting VEGF and combination of the RNAi drug with bevacizumab for cancer treatments. For efficient VEGF siRNA delivery, chemically polymerized siRNAs were complexed with thiolated-glycol chitosan (psi(VEGF)/tGC). The poly-VEGF siRNA and thiolated-glycol chitosan formed stable nanoparticles via electrostatic interaction and chemical crosslinking, and showed high accumulation in tumor tissues resulting in efficient gene silencing. Both VEGF siRNA nanoparticles and bevacizumab had efficient therapeutic effects in tumor xenograft mouse models. Interestingly, most pronounced therapeutic efficacy was observed when the two distinct VEGF inhibitors were treated in combination revealing synergistic effects. The results showed that the psi(VEGF)/tGC nanoparticle mediated knockdown of VEGF exerts anti-tumor effects and the combination treatments with bevacizumab can extend the treatments options to conventional bevacizumab treatments for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bevacizumab/pharmacology , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bevacizumab/administration & dosage , Bevacizumab/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gene Silencing/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polymerization/drug effects , RNA, Small Interfering/chemistry , Tumor Cells, Cultured , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
7.
Top Curr Chem (Cham) ; 375(2): 31, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28251564

ABSTRACT

Nanoparticles based on nanotechnology and biotechnology have emerged as efficient carriers for various biopharmaceutical agents including proteins and genes. In particular, polysaccharides have attracted interest of many researchers in the drug delivery field due to their advantages such as biocompatibility, biodegradability, low toxicity, and ease of modification. A number of polysaccharides including chitosan, hyaluronic acid, and dextran, and their derivatives have been widely used as polymeric backbones for the formation of nanoparticles, which can be provided as valuable gene delivery carriers. In this review, we introduce the chemical and physical natures of different polysaccharides particularly used in biomedical applications, and then discuss recent progress in the development of polysaccharide-based nanoparticles for gene delivery.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Nanoparticles/chemistry , Polysaccharides/chemistry , Biomedical Technology , Genetic Vectors/chemistry , Humans
8.
Theranostics ; 7(1): 9-22, 2017.
Article in English | MEDLINE | ID: mdl-28042312

ABSTRACT

Nucleic acid-directed self-assembly provides an attractive method to fabricate prerequisite nanoscale structures for a wide range of technological applications due to the remarkable programmability of DNA/RNA molecules. In this study, exquisite RNAi-AuNP nanoconstructs with various geometries were developed by utilizing anti-VEGF siRNA molecules as RNAi-based therapeutics in addition to their role as building blocks for programmed self-assembly. In particular, the anti-VEGF siRNA-functionalized AuNP nanoconstructs can take additional advantage of gold-nanoclusters for photothermal cancer therapeutic agent. A noticeable technical aspect of self-assembled RNAi-AuNP nanoconstructs in this study is the precise conjugation and separation of designated numbers of therapeutic siRNA onto AuNP to develop highly sophisticated RNA-based building blocks capable of creating various geometries of RNAi-AuNP nano-assemblies. The therapeutic potential of RNAi-AuNP nanoconstructs was validated in vivo as well as in vitro by combining heat generation capability of AuNP and anti-angiogenesis mechanism of siRNA. This strategy of combining anti-VEGF mechanism for depleting angiogenesis process at initial tumor progression and complete ablation of residual tumors with photothermal activity of AuNP at later tumor stage showed effective tumor growth inhibition and tumor ablation with PC-3 tumor bearing mice.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Therapy/methods , Gold/therapeutic use , Hyperthermia, Induced/methods , Nanostructures/administration & dosage , RNA, Small Interfering/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gold/metabolism , Heterografts , Male , Mice , Neovascularization, Pathologic , Prostatic Neoplasms/therapy , RNA, Small Interfering/metabolism , Treatment Outcome
9.
ACS Biomater Sci Eng ; 3(6): 990-999, 2017.
Article in English | MEDLINE | ID: mdl-29457127

ABSTRACT

We report unique bioactivity of a polycation-polyanion complex with potential utility for cancer therapy. A complex of disulfide-crosslinked polyethyleneimine (CLPEI), a polycation used for gene complexation, and dermatan sulfate (DS), an anionic polysaccharide to shield excessive cationic charge of the former, has toxicity to a specific group of cancer cell lines, including B16-F10 murine melanoma, A375SM human melanoma, and PC-3 human prostate cancer cells. These CLPEI-DS-sensitive cells express CD146, which binds to the complex via interaction with DS. There is a positive correlation between toxicity and intracellular level of CLPEI, indicating that the CLPEI-DS-sensitivity is attributable to the increased cellular uptake of CLPEI mediated by the DS-CD146 interactions. In vitro studies show that CLPEI-DS complex causes G0/G1 phase arrest and apoptotic cell death. In syngeneic and allograft models of B16-F10 melanoma, CLPEI-DS complex administered with a sub-toxic level of doxorubicin potentiates the chemotherapeutic effect of the drug by loosening tumor tissues. Given the unique toxicity, CLPEI-DS complex may be a useful carrier of gene or chemotherapeutics for the therapy of CD146-positive cancers.

10.
Physiol Rep ; 4(21)2016 11.
Article in English | MEDLINE | ID: mdl-27905295

ABSTRACT

Alternaria alternata is an allergenic fungus and known to cause an upper respiratory tract infection and asthma in humans with compromised immunity. Although A. alternata's effect on airway epithelial cells has previously been examined, the potential role of A. alternata on lung fibroblast viability is not understood. Since lung fibroblasts derived from patients with idiopathic pulmonary fibrosis (IPF) display a distinct phenotype that is resistant to stress and cell death inducing conditions, the investigation of the role of Alternaria on pathological IPF fibroblasts provides a better understanding of the fibrotic process induced by an allergenic fungus. Therefore, we examined cell viability of control and IPF fibroblasts (n = 8 each) in response to A. alternata extract. Control fibroblast cell death was increased while IPF fibroblasts were resistant when exposed to 50-100 µg/mL of A. alternata extract. However, there was no significant difference in kinetics or magnitude of Ca2+ responses from control lung and IPF fibroblasts. In contrast, unlike control fibroblasts, intracellular reactive oxygen species (ROS) levels remained low when IPF cells were treated with A. alternata extracts as a function of time. Caspase 3/7 and TUNEL assay revealed that enhanced cell death caused by A. alternata extract was likely due to necrosis, and 7-AAD assay and the use of sodium pyruvate for ATP generation further supported our findings that IPF fibroblasts become resistant to A. alternata extract-induced necrotic cell death. Our results suggest that exposure to A. alternata potentially worsens the fibrotic process by promoting normal lung fibroblast cell death in patients with IPF.


Subject(s)
Alternaria/enzymology , Cell Death/drug effects , Cell Survival/drug effects , Desensitization, Immunologic , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Necrosis , Sphingosine/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Survival/physiology , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Reactive Oxygen Species/metabolism , Sphingosine/administration & dosage , Sphingosine/adverse effects
11.
Biochem Biophys Res Commun ; 479(4): 779-786, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27693784

ABSTRACT

Labeling of stem cells aims to distinguish transplanted cells from host cells, understand in vivo fate of transplanted cells, particularly important in stem cell therapy. Adipose-derived mesenchymal stem cells (ASCs) are considered as an emerging therapeutic option for tissue regeneration, but much remains to be understood regarding the in vivo evidence. In this study, a simple and efficient cell labeling method for labeling and tracking of stem cells was developed based on bio-orthogonal copper-free click chemistry, and it was applied in a mouse hindlimb ischemia model. The human ASCs were treated with tetra-acetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz) to generate glycoprotein with unnatural azide groups on the cell surface, and the generated azide groups were fluorescently labeled by specific binding of dibenzylcyclooctyne-conjugated Cy5 (DBCO-Cy5). The safe and long-term labeling of the hASCs by this method was first investigated in vitro. Then the DBCO-Cy5-hASCs were transplanted into the hindlimb ischemia mice model, and we could monitor and track in vivo fate of the cells using optical imaging system. We could clearly observe the migration potent of the hASCs toward the ischemic lesion. This approach to design and tailor new method for labeling of stem cells may be useful to provide better understanding on the therapeutic effects of transplanted stem cells into the target diseases.


Subject(s)
Cell Tracking/methods , Ischemia/therapy , Mesenchymal Stem Cells/cytology , Adipose Tissue/cytology , Animals , Azides/chemistry , Click Chemistry/methods , Disease Models, Animal , Fluorescent Dyes/chemistry , Hindlimb , Humans , Imaging, Three-Dimensional , Ischemia/pathology , Mesenchymal Stem Cell Transplantation , Mice
12.
J Clin Med ; 5(9)2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27657144

ABSTRACT

Chronic lung diseases include a variety of obstinate and fatal diseases, including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), and lung cancers. Pharmacotherapy is important for the treatment of chronic lung diseases, and current progress in nanoparticles offers great potential as an advanced strategy for drug delivery. Based on their biophysical properties, nanoparticles have shown improved pharmacokinetics of therapeutics and controlled drug delivery, gaining great attention. Herein, we will review the nanoparticle-based drug delivery system for the treatment of chronic lung diseases. Various types of nanoparticles will be introduced, and recent innovative efforts to utilize the nanoparticles as novel drug carriers for the effective treatment of chronic lung diseases will also be discussed.

13.
Biomaterials ; 108: 57-70, 2016 11.
Article in English | MEDLINE | ID: mdl-27619240

ABSTRACT

Although there is great versatility of ultrasound (US) technologies in the real clinical field, one main technical challenge is the compromising of high quality of echo properties and size engineering of ultrasound contrast agents (UCAs); a high echo property is offset by reducing particle size. Herein, a new strategy for overcoming the dilemma by devising chemical gas (CO2) generating carbonate copolymer nanoparticles (Gas-NPs), which are clearly distinguished from the conventional gas-encapsulated micro-sized UCAs. More importantly, Gas-NPs could be readily engineered to strengthen the desirable in vivo physicochemical properties for nano-sized drug carriers with higher tumor targeting ability, as well as the high quality of echo properties for tumor-targeted US imaging. In tumor-bearing mice, anticancer drug-loaded Gas-NPs showed the desirable theranostic functions for US-triggered drug delivery, even after i.v. injection. In this regard, and as demonstrated in the aforementioned study, our technology could serve a highly effective platform in building theranostic UCAs with great sophistication and therapeutic applicability in tumor-targeted US imaging and US-triggered drug delivery.


Subject(s)
Delayed-Action Preparations/administration & dosage , Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Theranostic Nanomedicine/methods , Ultrasonography/methods , Animals , Carbon Dioxide/chemical synthesis , Carbon Dioxide/radiation effects , Contrast Media/chemical synthesis , Delayed-Action Preparations/radiation effects , Doxorubicin/chemistry , Drug Delivery Systems/methods , High-Energy Shock Waves , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Nanoparticles/radiation effects
14.
Macromol Biosci ; 16(11): 1583-1597, 2016 11.
Article in English | MEDLINE | ID: mdl-27503770

ABSTRACT

The aim of this study is to establish the safe and effective ocular delivery system of therapeutic small interfering RNA (siRNA) in corneal neovascularization therapy. The major hurdle present in siRNA-based corneal neovascularization (CNV) therapy is severe cytotoxicity caused by repetitive drug treatment. A reducible branched polyethylenimine (rBPEI)-based nanoparticle (NP) system is utilized as a new siRNA carrier as a hope for CNV therapy. The thiolated BPEI is readily self-crosslinked in mild conditions to make high molecular weight rBPEI thus allowing the creation of stable siRNA/rBPEI nanoparticles (siRNA-rBPEI-NPs). In the therapeutic region, the rBPEI polymeric matrix is effectively degraded into nontoxic LMW BPEI inside the reductive cytosol causing the rapid release of the encapsulated siRNA into the cytosol to carry out its function. The fluorescent-labeled siRNA-rBPEI-NPs can release siRNA into the entire corneal region after subconjuctival injection into the eye of Sprague Dawley rats thus confirming the proof of concept of this system.


Subject(s)
Corneal Neovascularization/drug therapy , Drug Delivery Systems/methods , Nanoparticles , Polyethyleneimine , RNA, Small Interfering , Animals , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Human Umbilical Vein Endothelial Cells , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
15.
J Control Release ; 244(Pt B): 205-213, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27435681

ABSTRACT

Nanoparticles have resulted in great progress in biomedical imaging and targeted drug delivery in cancer theranostics. To develop nanoparticles as an effective carrier system for therapeutics, chemical structures and physicochemical properties of nanoparticle may provide a reliable means to predict the in vitro characteristics of nanoparticles. However, in vivo fates of nanoparticles, such as pharmacokinetics and tumor targeting efficiency of nanoparticles, have been difficult to predict beforehand. To predict the in vivo fates of nanoparticles in tumor-bearing mice, differences in physicochemical properties and in vitro cancer cell/macrophage uptake of 5 different nanoparticles with mean diameter of 200-250nm were comparatively analyzed, along with their circulation in adult zebrafish. The nanoparticles which showed favorable cellular uptake by macrophages indicated high unintended liver accumulation in vivo, which is attributed to the clearance by the reticuloendothelial system (RES). In addition, blood circulation of nanoparticles was closely correlated in adult zebrafish and in mice that the zebrafish experiment may elucidate the in vivo behavior of nanoparticles in advance of the in vivo experiment using mammal animal models. This comparative study on various nanoparticles was conducted to provide the basic information on predicting the in vivo fates of nanoparticles prior to the in vivo experiments.


Subject(s)
Macrophages/metabolism , Nanoparticles/administration & dosage , Neoplasms/metabolism , Animals , Biological Transport , Blood Circulation , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/analogs & derivatives , Dextrans/administration & dosage , Female , Humans , Hyaluronic Acid/administration & dosage , Mice , Mice, Inbred C3H , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Polystyrenes/administration & dosage , RAW 264.7 Cells , Titanium/administration & dosage , Zebrafish
16.
Bioconjug Chem ; 27(4): 927-36, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26930274

ABSTRACT

Establishment of an appropriate cell labeling and tracking method is essential for the development of cell-based therapeutic strategies. Here, we are introducing a new method for cell labeling and tracking by combining metabolic gylcoengineering and bioorthogonal copper-free Click chemistry. First, chondrocytes were treated with tetraacetylated N-azidoacetyl-D-mannosamine (Ac4ManNAz) to generate unnatural azide groups (-N3) on the surface of the cells. Subsequently, the unnatural azide groups on the cell surface were specifically conjugated with near-infrared fluorescent (NIRF) dye-tagged dibenzyl cyclooctyne (DBCO-650) through bioorthogonal copper-free Click chemistry. Importantly, DBCO-650-labeled chondrocytes presented strong NIRF signals with relatively low cytotoxicity and the amounts of azide groups and DBCO-650 could be easily controlled by feeding different amounts of Ac4ManNAz and DBCO-650 to the cell culture system. For the in vivo cell tracking, DBCO-650-labeled chondrocytes (1 × 10(6) cells) seeded on the 3D scaffold were subcutaneously implanted into mice and the transplanted DBCO-650-labeled chondrocytes could be effectively tracked in the prolonged time period of 4 weeks using NIRF imaging technology. Furthermore, this new cell labeling and tracking technology had minimal effect on cartilage formation in vivo.


Subject(s)
Cartilage/cytology , Chondrocytes/cytology , Click Chemistry , Copper/chemistry , Animals , Flow Cytometry , Mice , Tissue Engineering
17.
J Control Release ; 220(Pt B): 631-41, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26307351

ABSTRACT

Cancer is a multifactorial disease which involves complex genetic mutation and dysregulation. Combinatorial RNAi technology and concurrent multiple gene silencing are expected to provide advanced strategies for effective cancer therapy, but a safe and effective carrier system is a prerequisite to successful siRNA delivery in vivo. We previously developed an effective tumor-targeting siRNA delivery system for in vivo application. In response to the success of this development, herein we present a dual-gene targeted siRNA and its delivery system, to achieve synergistic effects in cancer therapy. Two different sequences of siRNA were chemically modified to be randomly copolymerized in a single backbone of siRNA polymer (Dual-poly-siRNA), and the resulting Dual-poly-siRNA was incorporated into tumor-homing glycol chitosan nanoparticles. Based on the stability in serum and delivery in a tumor-targeted manner, intravenously administered Dual-poly-siRNA carrying glycol chitosan nanoparticles (Dual-NP) demonstrated successful dual-gene silencing in tumors. Notably, co-delivery of VEGF and Bcl-2 targeting siRNA led to more effective cancer therapy for convenient application.


Subject(s)
Chitosan/chemistry , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , RNA, Small Interfering/genetics , RNAi Therapeutics/methods , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Injections, Intravenous , Male , Mice, Nude , Nanoparticles , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Tumor Burden , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
18.
Macromol Biosci ; 15(9): 1224-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26175259

ABSTRACT

Cathepsin B (CB)-specific molecular imaging probe is applied to monitor the changes of CB expression in 3D cultured chondrocytes during the chondrogenesis. The probe is synthesized with a CB-cleavable peptide linked to a near infrared fluorescence (NIRF) dye and a dark quencher, which can recover the NIRF signal in the presence of CB enzyme. The CB activities in two different sets of chondrocytes are comparatively analyzed. The chondrocytes with higher CB activity show more extensive area of chondrogenesis that CB molecular imaging directly reflects the chondrogenic potency of the cells. The CB probe is expected to provide a reliable prediction for the quality of engineered cartilage by visualizing the activity of the relevant enzyme in chondrocytes.


Subject(s)
Cartilage, Articular/physiology , Cathepsin B/analysis , Chondrogenesis , Optical Imaging , Tissue Engineering/standards , Animals , Cartilage, Articular/enzymology , Cathepsin B/genetics , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/physiology , Gene Expression , Molecular Imaging , Quality Control , Rabbits
19.
Curr Pharm Des ; 21(22): 3158-66, 2015.
Article in English | MEDLINE | ID: mdl-26027570

ABSTRACT

In recent years, combination of different types of therapies using nanoparticles has emerged as an advanced strategy for cancer treatment. Most of all, combination of chemotherapeutic drug and siRNA in nanoformulation has shown a great potential, because siRNA-mediated specific gene silencing can compensate for the incomplete anti-cancer actions of chemotherapy. In this article, nanoparticle-based combination therapy for cancer treatment is introduced to be focused on the therapeutic chemical and siRNA combination. It is classified into 3 groups: 1) general chemotherapy combined with siRNA carrying nanoparticle, 2) co-delivery of chemical and siRNA therapeutics within a single nanoparticle, and 3) Use of multiple nanoparticles for chemical and siRNA therapeutics. The purpose of the combination and the mechanisms of anti-cancer action was described according to the categories. Examples of some recent developments of nanotechnology-based chemo- and siRNA- therapeutics combination therapy are summarized for better understanding of its practical application.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Carriers/administration & dosage , Nanomedicine , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Drug Carriers/chemistry , Humans , Nanoparticles/chemistry , Neoplasms/pathology , RNA, Small Interfering/therapeutic use
20.
J Biomed Mater Res A ; 103(11): 3474-82, 2015 11.
Article in English | MEDLINE | ID: mdl-25904025

ABSTRACT

Methylprednisolone (MP) is a glucocorticoid that is used as an anti-inflammatory agent to the treat spinal cord injury (SCI). A low molecular weight chitosan was used to synthesize chitosan-MP conjugate, which was used to evaluate the gene therapy, anti-inflammatory and anti-apoptotic effects of MP. The cytotoxicity of chitosan-MP nanoparticles and the transfection efficiency of plasmid DNA were evaluated by MTT and luciferase assays. A chitosan-MP/pDNA complexes was injected into injured spinal cord to evaluate the anti-inflammatory and anti-apoptotic effects of these complexes using terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL) and ED1 staining, respectively. In addition, to evaluate the distribution of chitosan-MP/pDNA complexes, pß-gal encapsulated chitosan-MP was injected into the injected site. Cell survival was similar in cells treated with chitosan-MP conjugate and untreated cells. Luciferase expression was higher in cells treated with the chitosan-MP/pDNA than cells treated with the chitosan/pDNA. The chitosan-MP/pDNA complexes also reduced apoptosis and inflammation at the injury site. These results suggest that chitosan-MP conjugation is an effective gene delivery system to treat SCI.


Subject(s)
Gene Transfer Techniques , Nanoparticles/chemistry , Spinal Cord Injuries/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Chitosan/pharmacology , Luciferases/metabolism , Male , Methylprednisolone/pharmacology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Proton Magnetic Resonance Spectroscopy , Rats, Sprague-Dawley , Transfection , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...