Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Publication year range
1.
Mater Sci Eng C Mater Biol Appl ; 118: 111427, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33255024

ABSTRACT

Streptokinase, a clot-dissolving agent, is widely used in treatment of cardiovascular diseases such as blood clots and deep thrombosis. Streptokinase is a cost-effective drug with a short biological half-life (i.e. 15 to 30 min). In addition, due to its prokaryotic source, the immune response quickly reacts to the drug. Despite these limitations, streptokinase is still the first choice for diseases associated with thrombosis. In this work, streptokinase was encapsulated in mPEG-PLGA nanoparticles to improve its pharmacokinetic properties. The nanoparticles containing the enzyme were prepared by coaxial electrospray and their physicochemical properties, blood compatibility, circulation time and cell toxicity were evaluated. The results showed that the use of mPEG-PLGA nanoparticles to encapsulate the enzyme resulted in prolonged circulation time (up to 120 min) with a slight decrease in its activity. In vivo studies also showed that the nanoparticles containing streptokinase did not have adverse effect on blood biochemistry parameters as well as liver and kidney tissues. As a result, the mPEG-PLGA nanoparticles showed the potential for increasing the biological activity of streptokinase with no important adverse effect.


Subject(s)
Nanoparticles , Streptokinase , Drug Carriers , Particle Size , Polyesters , Polyethylene Glycols
2.
Int J Nanomedicine ; 13: 6517-6530, 2018.
Article in English | MEDLINE | ID: mdl-30410336

ABSTRACT

PURPOSE: Nitric oxide (NO) can be clinically applied at low concentrations to regulate angiogenesis. However, studies using small molecule NO donors (N-diazeniumdiolate, S-nitrosothiol, etc) have yet to meet clinical requirements due to the short half-life and initial burst-release profile of NO donors. In this study, we report the feasibility of methoxy poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) (mPEG-PLGA) nanoparticles (NPs) as NO-releasing polymers (NO-NPs) for inducing angiogenesis. MATERIALS AND METHODS: The mPEG-PLGA copolymers were synthesized by typical ring-opening polymerization of lactide, glycolide and mPEG as macroinitiators. Double emulsion methods were used to prepare mPEG-PLGA NPs incorporating hydrophilic NONOate (dieth-ylenetriamine NONOate). RESULTS: This liposomal NP encapsulates hydrophilic diethylenetriamine NONOate (70%±4%) more effectively than other previously reported materials. The application of NO-NPs at different ratios resulted in varying NO-release profiles with no significant cytotoxicity in various cell types: normal cells (fibroblasts, human umbilical vein endothelial cells and epithelial cells) and cancer cells (C6, A549 and MCF-7). The angiogenic potential of NO-NPs was confirmed in vitro by tube formation and ex vivo through an aorta ring assay. Tubular formation increased 189.8% in NO-NP-treated groups compared with that in the control group. Rat aorta exhibited robust sprouting angiogenesis in response to NO-NPs, indicating that NO was produced by polymeric NPs in a sustained manner. CONCLUSION: These findings provide initial results for an angiogenesis-related drug development platform by a straightforward method with biocompatible polymers.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Biocompatible Materials/chemistry , Nanoparticles/chemistry , Nitric Oxide/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , A549 Cells , Animals , Cell Death/drug effects , Delayed-Action Preparations/pharmacology , Drug Carriers , Emulsions/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Nanoparticles/ultrastructure , Particle Size , Rats, Sprague-Dawley , Static Electricity
3.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-707030

ABSTRACT

Objective To investigate the uptake mechanism of HepG2.2.15 cells to the nanoparticles co-loaded with syringopicroside and hydroxytyrosol (SH-NPs). Methods The nanoparticles were prepared by using a nanoprecipitation method with mPEG-PLGA as nano-carrier co-loaded with syringopicroside and hydroxytyrosol. The uptake mechanism of HepG2.2.15 cells to SH-NPs was studied by fluorescence microscopy and flow cytometry using fluoresceineisothiocyanate (FITC) as a fluorescent marker. Results With colchicine as the inhibitor, the incubation time ranged from 0.5 to 24 h, the percentage of positive cells increased from 1.9% to 56.4%; When the drug concentration was 125, 250 μg/mL and 500 μg/mL, the positive cell percentages were 4.9%, 3.4% and 3.9%. With chloroquine as the inhibitor; the incubation time ranged from 0.5 to 24 h, the percentage of positive cells increased from 7.4% to 55.4%; When the drug concentration was 125, 250 and 500 μg/mL, the percentage of positive cells was 19.5%, 22.5% and 27.6%. Conclusion Colchicine and chloroquine have an inhibitory effect on HepG2.2.15 cells uptake, and the uptake of SH-NPs in HepG2.2.15 cells was positively correlated with drug concentration and incubation time. It can be concluded that the uptake mechanism of HepG2.2.15 cells to SH-NPs was nonspecific adsorption endocytosis.

4.
Int J Nanomedicine ; 12: 5255-5269, 2017.
Article in English | MEDLINE | ID: mdl-28769562

ABSTRACT

Previous studies have shown that mithramycin A (MIT) is a promising candidate for the treatment of pancreatic carcinoma through inhibiting transcription factor Sp1. However, systemic toxicities may limit its clinical application. Here, we report a rationally designed formulation of MIT-loaded nanoparticles (MIT-NPs) with a small size and sustained release for improved passive targeting and enhanced therapeutic efficacy. Nearly spherical MIT-NPs with a mean particle size of 25.0±4.6 nm were prepared by encapsulating MIT into methoxy poly(ethylene glycol)-block-poly(d,l-lactic-co-glycolic acid) (mPEG-PLGA) nanoparticles (NPs) with drug loading of 2.11%±0.51%. The in vitro release of the MIT-NPs lasted for >48 h with a sustained-release pattern. The cytotoxicity of MIT-NPs to human pancreatic cancer BxPC-3 and MIA Paca-2 cells was comparable to that of free MIT. Determined by flow cytometry and confocal microscopy, the NPs internalized into the cells quickly and efficiently, reaching the peak level at 1-2 h. In vivo fluorescence imaging showed that the prepared NPs were gradually accumulated in BxPC-3 and MIA Paca-2 xenografts and retained for 168 h. The fluorescence intensity in both BxPC-3 and MIA Paca-2 tumors was much stronger than that of various tested organs. Therapeutic efficacy was evaluated with the poorly permeable BxPC-3 pancreatic carcinoma xenograft model. At a well-tolerated dose of 2 mg/kg, MIT-NPs suppressed BxPC-3 tumor growth by 96%. Compared at an equivalent dose, MIT-NPs exerted significantly higher therapeutic effect than free MIT (86% versus 51%, P<0.01). Moreover, the treatment of MIT and MIT-NPs reduced the expression level of oncogene c-Myc regulated by Sp1, and notably, both of them decreased the protein level of CD47. In summary, the novel formulation of MIT-NPs shows highly therapeutic efficacy against pancreatic carcinoma xenograft. In addition, MIT-NPs can downregulate CD47 expression, implying that it might play a positive role in cancer immunotherapy.


Subject(s)
Nanoparticles/administration & dosage , Pancreatic Neoplasms/drug therapy , Plicamycin/administration & dosage , Polyesters/chemistry , Polyethylene Glycols/chemistry , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Microscopy, Confocal , Nanoparticles/chemistry , Particle Size , Plicamycin/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...