Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Control Release ; 268: 198-211, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29061511

ABSTRACT

Malignant proliferation and metastasis in non-small cell lung carcinoma (NSCLC) are great challenges for effective clinical treatment through conventional chemotherapy. The combinational therapy strategy of RNA interfering (RNAi) technology and chemotherapeutic agents have been reported to be promising for effective cancer therapy. In this study, based on multifunctional nanoparticles (NPs), the simultaneous delivery of etoposide (ETP) and anti-Enhancer of Zeste Homologue 2 (EZH2) siRNA for the effective treatment of orthotopic lung tumor was achieved. The NPs exhibited pH/redox dual sensitivity verified by particle size changes, morphological changes, and in vitro release of drugs. Confocal microscopy analysis confirmed that the NPs exhibited endosomal escape property and on-demand intracellular drug release behavior, which can protect siRNA from degradation and facilitate the chemotherapeutic effect respectively. In vitro tumor cell motility study demonstrated that EZH2 siRNA loaded in NPs can decrease the migration and invasion capabilities of tumor cells by downregulating the expression of EZH2 mRNA and protein. In particular, an antiproliferation study revealed that the co-delivery of siRNA and ETP in the multifunctional NPs can induce a synergistic therapeutic effect on NSCLC. In vivo targeting evaluation showed that cRGDyC-PEG modification on NPs exhibited a low distribution in normal organs and an obvious accumulation in orthotopic lung tumor. Furthermore, targeted NPs co-delivering siRNA and ETP showed superior inhibition on tumor growth and metastasis and produced minimal systemic toxicity. These findings indicated that multifunctional NPs can be utilized as a co-delivery system, and that the combination of EZH2 siRNA and ETP can effectively treat NSCLC.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Enhancer of Zeste Homolog 2 Protein/genetics , Etoposide/administration & dosage , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Combined Modality Therapy , Drug Liberation , Etoposide/chemistry , Female , Humans , Mice, Nude , Nanoparticles/chemistry , Polymers/administration & dosage , Polymers/chemistry , RNA, Small Interfering/chemistry
2.
Int J Nanomedicine ; 12: 4241-4256, 2017.
Article in English | MEDLINE | ID: mdl-28652730

ABSTRACT

Physicochemical properties, including particle size, zeta potential, and drug release behavior, affect targeting efficiency, cellular uptake, and antitumor effect of nanocarriers in a formulated drug-delivery system. In this study, a novel stepwise pH-responsive nanodrug delivery system was developed to efficiently deliver and significantly promote the therapeutic effect of doxorubicin (DOX). The system comprised dimethylmaleic acid-chitosan-urocanic acid and elicited stepwise responses to extracellular and intracellular pH. The nanoparticles (NPs), which possessed negative surface charge under physiological conditions and an appropriate nanosize, exhibited advantageous stability during blood circulation and enhanced accumulation in tumor sites via enhanced permeability and retention effect. The tumor cellular uptake of DOX-loaded NPs was significantly promoted by the first-step pH response, wherein surface charge reversion of NPs from negative to positive was triggered by the slightly acidic tumor extracellular environment. After internalization into tumor cells, the second-step pH response in endo/lysosome acidic environment elicited the on-demand intracellular release of DOX from NPs, thereby increasing cytotoxicity against tumor cells. Furthermore, stepwise pH-responsive NPs showed enhanced antiproliferation effect and reduced systemic side effect in vivo. Hence, the stepwise pH-responsive NPs provide a promising strategy for efficient delivery of antitumor agents.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Chitosan/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Evaluation, Preclinical/methods , Drug Liberation , Female , Hydrogen-Ion Concentration , Malonates/chemistry , Mice, Inbred BALB C , Nanoparticles/chemistry , Particle Size , Tissue Distribution , Urocanic Acid/chemistry
3.
J Biomed Mater Res B Appl Biomater ; 105(7): 2093-2106, 2017 10.
Article in English | MEDLINE | ID: mdl-27405391

ABSTRACT

P-glycoprotein (P-gp) plays an importantrole in multidrug resistance (MDR), proved to be one of the major obstacles in cancer chemotherapy. Cationic polymers could specifically deliver siRNA to tumor cells and thus reverse MDR by the downregulation of P-gp. In this study, a triblock copolymer micelle was prepared based on the polymer of N-succinyl-chitosan-poly-l-lysine-palmitic acid (NSC-PLL-PA) to deliver siRNA-P-gp (siRNA-micelle) or doxorubicin (Dox-micelle). The resulting micelle exhibited an efficient binding ability for siRNA and high encapsulation efficiency for Dox, with an average particle size of ∼170 nm. siRNA-micelle and Dox-micellewere instable at low pH, thereby enhancing tumor accumulation and intracellular release of the encapsulated siRNA and Dox. siRNA-micelle micelles could enhance the knockdown efficacy of siRNA by improving the transfection efficiency, downregulating P-gp expression, and passing the drug efflux transporters, thereby improving the therapeutic effects of Dox-micelle. However, P-gp could transfer from HepG2/ADM to HepG2 cells independent of the expression of mdr1, and the acquired resistance could permit tumor cells to survive and develop intrinsic P-gp-mediated resistance, thereby limiting the desired efficiency of chemotherapeutics. This study demonstrated the effectiveness of siRNA-micelle for tumor-targeted delivery, MDR reversal, and provided an effective strategy for the treatment of cancers that develop MDR. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2093-2106, 2017.


Subject(s)
Doxorubicin/pharmacology , Drug Delivery Systems/methods , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Micelles , Neoplasm Proteins , Neoplasms , RNA, Small Interfering , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Hep G2 Cells , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
4.
J Biomed Mater Res B Appl Biomater ; 105(5): 1114-1125, 2017 07.
Article in English | MEDLINE | ID: mdl-27008163

ABSTRACT

The development of effective and stable carriers of small interfering RNA (siRNA) is important for treating cancer with multidrug resistance (MDR). We developed a new gene and drug co-delivery system and checked its characteristics. Low-density lipoprotein (LDL) was coupled with N-succinyl chitosan (NSC) Lipoic acid (LA) micelles and co-delivered MDR1 siRNA and paclitaxel (PTX-siRNA/LDL-NSC-LA) to enhance antitumor effects by silencing the MDR gene of tumors (Li et al., Adv Mater 2014;26:8217-8224). In our study, we developed a new type of containing paclitaxel-loaded micelles and siRNA-loaded LDL nanoparticle. This "binary polymer" is pH and reduction dual-sensitive core-crosslinked micelles. PTX-siRNA/LDL-NSC-LA had an average particle size of (171.6 ± 6.42) nm, entrapment efficiency of (93.92 ± 1.06) %, and drug-loading amount of (12.35% ± 0.87) %. In vitro, MCF-7 cells, high expressed LDL receptor, were more sensitive to this delivery system than to taxol® and cell activity was inhibited significantly. Fluorescence microscopy showed that PTX-siRNA/LDL-NSC-LA was uptaken very conveniently and played a key role in antitumor activity. PTX-siRNA/LDL-NSC-LA protected the siRNA from degradation by macrophage phagocytosis and evidently down-regulated the level of mdr1 mRNA as well as the expression of P-gp. We tested the target ability of PTX-siRNA/LDL-NSC-LA in vivo in tumor-bearing nude mice. Results showed that this system could directly deliver siRNA and PTX to cancer cells. Thus, new co-delivering siRNA and antitumor drugs should be explored for solving MDR in cancer. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1114-1125, 2017.


Subject(s)
Chitosan , Gene Transfer Techniques , Lipoproteins, LDL , Micelles , Neoplasm Proteins , Neoplasms, Experimental , Paclitaxel/pharmacology , RNA, Small Interfering , Thioctic Acid , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Chitosan/chemistry , Chitosan/pharmacology , Female , Humans , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/pharmacology , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Thioctic Acid/chemistry , Thioctic Acid/pharmacology , Xenograft Model Antitumor Assays
5.
ACS Appl Mater Interfaces ; 8(47): 32146-32158, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27933846

ABSTRACT

The efficient delivery of antitumor agents to tumor sites faces numerous obstacles, such as poor cellular uptake and slow intracellular drug release. In this regard, smart nanoparticles (NPs) that respond to the unique microenvironment of tumor tissues have been widely used for drug delivery. In this study, novel charge-reversal and reduction-responsive histidine-grafted chitosan-lipoic acid NPs (HCSL-NPs) were selected for efficient therapy of breast cancer by enhancing cell internalization and intracellular pH- and reduction-triggered doxorubicin (DOX) release. The surface charge of HCSL-NPs presented as negative at physiological pH and reversed to positive at the extracellular and intracellular pH of the tumor. In vitro release investigation revealed that DOX/HCSL-NPs demonstrated a sustained drug release under the physiological condition, whereas rapid DOX release was triggered by both endolysosome pH and high-concentration reducing glutathione (GSH). These NPs exhibited enhanced internalization at extracellular pH, rapid intracellular drug release, and improved cytotoxicity against 4T1 cells in vitro. Excellent tumor penetrating efficacy was also found in 4T1 tumor spheroids and solid tumor slices. In vivo experiments demonstrated that HCSL-NPs exhibited excellent tumor-targeting ability in tumor tissues as well as excellent antitumor efficacy and low systemic toxicity in breast tumor-bearing BALB/c mice. These results indicated that the novel charge-reversal and reduction-responsive HCSL-NPs have great potential for targeted and efficient delivery of chemotherapeutic drugs in cancer treatments.


Subject(s)
Nanoparticles , Animals , Doxorubicin , Drug Carriers , Drug Delivery Systems , Drug Liberation , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C
6.
Int J Pharm ; 511(2): 728-40, 2016 Sep 25.
Article in English | MEDLINE | ID: mdl-27484835

ABSTRACT

Internal stimuli, such as intracellular lysosomal pH, enzyme, redox and reduction, can be applied to improve biological specificity of chemotherapeutic drugs for cancer therapy. Thus, functionalized copolymers based on their response to specific microenvironment of tumor regions have been designed as smart drug vesicles for enhanced anti-cancer efficiency and reduced side effects. Herein, we reported dually pH/reduction-responsive novel micelles based on self-assembly of carboxymethyl chitosan-cysteamine-N-acetyl histidine (CMCH-SS-NA) and doxorubicin (DOX). The tailor-made dually responsive micelles demonstrated favorable stability in normal physiological environment and triggered rapid drug release in acidic and/or reduction environment. Additionally, the nanocarriers responded to the intracellular environment in an ultra-fast manner within several minutes, which led to the pinpointed release of DOX in tumor cells effectively and ensured higher DOX concentrations within tumor areas with the aid of targeted delivery, thereby leading to enhanced tumor ablation. Thus, this approach with sharp drug release behavior represented a versatile strategy to provide a promising paradigm for cancer therapy.


Subject(s)
Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Micelles , Tumor Microenvironment/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Chitosan/analogs & derivatives , Chitosan/chemistry , Cysteamine/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Histidine/chemistry , Hydrogen-Ion Concentration , Liver/metabolism , Mice , Oxidation-Reduction , Rats , Tissue Distribution , Xenograft Model Antitumor Assays
7.
Sci Rep ; 6: 23859, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27030638

ABSTRACT

Co-delivery of chemotherapeutics and siRNA with different mechanisms in a single system is a promising strategy for effective cancer therapy with synergistic effects. In this study, a triblock copolymer micelle was prepared based on the polymer of N-succinyl chitosan-poly-L-lysine-palmitic acid (NSC-PLL-PA) to co-deliver doxorubicin (Dox) and siRNA-P-glycoprotein (P-gp) (Dox-siRNA-micelle). Dox-siRNA-micelle was unstable in pH 5.3 medium than in pH 7.4 medium, which corresponded with the in vitro rapid release of Dox and siRNA in acidic environments. The antitumor efficacy of Dox-siRNA-micelle in vitro significantly increased, especially in HepG2/ADM cells, which was due to the downregulation of P-gp. Moreover, almost all the Dox-siRNA-micelles accumulated in the tumor region beyond 24 h post-injection, and the co-delivery system significantly inhibited tumor growth with synergistic effects in vivo. This study demonstrated the effectiveness of Dox-siRNA-micelles in tumor-targeting and MDR reversal, and provided a promising strategy to develop a co-delivery system with synergistic effects for combined cancer therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antibiotics, Antineoplastic/pharmacology , Combined Modality Therapy/methods , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Liver Neoplasms/therapy , RNA, Small Interfering/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cell Survival/drug effects , Drug Liberation , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Micelles , RNA, Small Interfering/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Int J Nanomedicine ; 11: 325-36, 2016.
Article in English | MEDLINE | ID: mdl-26855571

ABSTRACT

In this study, harmine liposomes (HM-lip) were prepared through the thin-film hydration-pH-gradient method and then coated with N-trimethyl chitosan (TMC). Particle size, zeta potential, entrapment efficiency, and in vitro release of HM-lip and TMC-coated harmine liposomes (TMC-HM-lip) were also determined. Sprague Dawley rats were further used to investigate the pharmacokinetics in vivo. Retention behavior in mouse gastrointestinal tract (GIT) was studied through high-performance liquid chromatography and near-infrared imaging. Degradation was further evaluated through incubation with Caco-2 cell homogenates, and a Caco-2 monolayer cell model was used to investigate the uptake and transport of drugs. HM-lip and TMC-HM-lip with particle size of 150-170 nm, an entrapment efficiency of about 81%, and a zeta potential of negative and positive, respectively, were prepared. The release of HM from HM-lip and TMC-HM-lip was slower than that from HM solution and was sensitive to pH. TMC-HM-lip exhibited higher oral bioavailability and had prolonged retention time in GIT. HM-lip and TMC-HM-lip could also protect HM against degradation in Caco-2 cell homogenates. The uptake amount of TMC-HM-lip was higher than that of HM and HM-lip. TMC-HM-lip further demonstrated higher apparent permeability coefficient (P(app)) from the apical to the basolateral side than HM and HM-lip because of its higher uptake and capability to open tight junctions in the cell monolayers. TMC-HM-lip can prolong the retention time in the GIT, protect HM against enzyme degradation, and improve transport across Caco-2 cell monolayers, thus enhancing the oral bioavailability of HM.


Subject(s)
Chitosan/chemistry , Gastrointestinal Tract/drug effects , Harmine/metabolism , Liposomes/chemistry , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Cell Membrane Permeability , Cell Proliferation/drug effects , Cells, Cultured , Drug Carriers/chemistry , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Humans , In Vitro Techniques , Liposomes/administration & dosage , Mice , Particle Size , Rats , Rats, Sprague-Dawley
9.
Yao Xue Xue Bao ; 51(4): 642-9, 2016 04.
Article in Chinese | MEDLINE | ID: mdl-29860751

ABSTRACT

cRGD-carboxymethyl chitosan-palmitic acid (cRGD-CMCh-PA) was synthesized and a pH- sensitive paclitaxel-loaded cRGD-CMCh-PA micelles(PTX-cRGD-CMCh-PA) was prepared with the film dispersion method; related substances were characterized by FT-IR and (1)H NMR. PTX-cRGD-CMCh-PA micelles were studied with the particle size distribution, zeta potential, morphology and release behavior in vitro was investigated by the method of equilibrium dialysis. In vitro cytotoxicity of different formulations on A549 cells was tested by MTT assay. The uptake process of micelles was explored using confocal microscopy and a live cell station was used to observe the dynamic phagocytosis. The subcutaneous and orthotropic tumor models were built to study the distribution of Di R-labeled micelles by near-infrared fluorescence(NIR) imaging system. The FT-IR spectra and (1)H NMR spectra confirmed the successful conjugation of cRGD-CMCh-PA polymer and the degree of carboxymethyl and the palmitic acid grafted on chitosan were 45.0% and 15.0%. PTX-cRGD-CMCh-PA micelles were prepared with particle size of(162.9 ± 1.5) nm, zeta potential of +26.3 m V and encapsulation efficiency and the drug loading of 99.67% and 28.5%, respectively. The micelles released slowly in pH 7.4 whose release curves were accorded with the Higuchi equation; they had an initial burst effect in second hours and showed a pH sensitive release behavior in pH 5.3. The IC(50) of PXT-CMCh-PA and PTX-cRGD-CMCh-PA were 2.077 µg·mL(-1) and 0.876 µg·mL(-1), respectively. The cells uptake process of micelles in A549 cells revealed that the micelles were mainly co-located with lysosome and PTX-cRGD-CMCh- PA showed much better targeting effect. The NIR fluorescence imaging results showed that the micelles had a good targeting effect on both subcutaneous and orthotropic tumors. In this study, a novel copolymer cRGD- CMCh-PA was synthesized with a sustained and pH-dependent drug release activity which would potentially become a new carrier for hydrophobic drugs.


Subject(s)
Chitosan/analogs & derivatives , Drug Carriers/chemistry , Oligopeptides/chemistry , Paclitaxel/administration & dosage , Palmitic Acid/chemistry , A549 Cells , Antineoplastic Agents, Phytogenic/administration & dosage , Chitosan/chemistry , Drug Liberation , Humans , Hydrophobic and Hydrophilic Interactions , Micelles , Particle Size , Polymers , Spectroscopy, Fourier Transform Infrared
10.
Sci Rep ; 5: 17904, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26639052

ABSTRACT

An antibody that specifically interacts with an antigen could be applied to an active targeting delivery system. In this study, CD147 antibody was coupled with α-hed chitosan nanoparticles (α-Hed-CS-NPs). α-Hed-CS-CD147-NPs were round and spherical in shape, with an average particle size of 148.23 ± 1.75 nm. The half-maximum inhibiting concentration (IC50) of α-Hed-CS-CD147-NPs in human liver cancer cell lines HepG2 and SMMC-7721 was lower than that of free α-Hed and α-Hed-CS-NPs. α-Hed-induced cell death was mainly triggered by apoptosis. The increase in intracellular accumulation of α-Hed-CS-CD147-NPs was also related to CD147-mediated internalization through the Caveolae-dependent pathway and lysosomal escape. The higher targeting antitumor efficacy of α-Hed-CS-CD147-NPs than that α-Hed-CS-NPs was attributed to its stronger fluorescence intensity in the tumor site in nude mice.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Basigin/immunology , Chitosan/chemistry , Endocytosis/drug effects , Liver Neoplasms/pathology , Nanoparticles/chemistry , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Flow Cytometry , Fluorescence , Hep G2 Cells , Humans , Imaging, Three-Dimensional , Intracellular Space/metabolism , Mice, Nude , Microscopy, Confocal , Nanoparticles/ultrastructure , Oleanolic Acid/chemical synthesis , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Particle Size , Propidium/metabolism , Saponins/chemical synthesis , Saponins/chemistry , Spectroscopy, Fourier Transform Infrared , Spectroscopy, Near-Infrared , Subcellular Fractions/metabolism
11.
Int J Pharm ; 492(1-2): 141-51, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26188316

ABSTRACT

This study aimed to prepare efficient cRGDyK peptide-decorated micelles for the targeted therapy of non-small-cell lung cancer (NSCLC). An amphiphilic copolymer N-succinyl-palmitoyl-chitosan (SPCS) was synthesized and characterized. cRGDyK peptide is a ligand that can target tumors via specific binding integrin receptor overexpressed on tumor neovascularization and cells. cRGDyK-functionalized SPCS micelles loaded with paclitaxel (PTX/cRGDyK-SPCS) were prepared by film dispersion method and then characterized according to morphology, size, and zeta potential. PTX/cRGDyK-SPCS micelles presented pH-triggered drug release behavior under acidic conditions. The accumulation of micelles detected by laser confocal fluorescence microscopy and flow cytometry showed that cRGDyK-SPCS micelles were easily taken up by A549 cells marked with the luciferase gene (luc-A549). Meanwhile, co-localization of the micelles and lysosomes was recorded dynamically using a live cell station. MTT assays and cell apoptosis studies revealed that cell viability was significantly inhibited by PTX/cRGDyK-SPCS micelles. More importantly, in vivo animal studies showed that cRGDyK-SPCS micelles mainly accumulated in the orthotopic tumor site. PTX/cRGDyK-SPCS micelles exhibited better anti-tumor activity in subcutaneous and orthotopic lung tumors compared with PTX/SPCS micelles and Taxol(®). These results suggested that PTX/cRGDyK-SPCS micelles had better cancer targeting capacity and superior anti-tumor efficacy. Thus, these micelles have great potential as novel carriers in delivering anti-tumor drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Chitosan/analogs & derivatives , Chitosan/administration & dosage , Paclitaxel/administration & dosage , Peptides, Cyclic/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Chitosan/chemistry , Chitosan/therapeutic use , Drug Delivery Systems , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice, Nude , Micelles , Microtubules/drug effects , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Tumor Burden/drug effects
12.
CNS Neurosci Ther ; 19(12): 917-25, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24267641

ABSTRACT

AIM: Remote ischemic preconditioning protects against ischemic organ damage by giving short periods of subcritical ischemia to a remote organ. We tested the hypothesis that remote ischemic conditioning can attenuate cerebral stroke in a rat middle cerebral artery occlusion (MCAO) model by microparticles (MPs). METHODS AND RESULTS: MPs were extracted from healthy rats that underwent hindlimb ischemia-reperfusion preconditioning (RIPC), and were transfused into rats that had undergone MCAO without RIPC. The transfusion resulted in an increase in platelet-derived MPs in blood and reduction in infarction area, confirmed by both 2-3-5-triphenyltetrazolium chloride staining and magnetic resonance imaging, albeit to a lesser degree than RIPC itself. Behavioral tests (modified Neurological Severity Score [mNSS]) were calculated to judge the behavioral change. However, no significant difference was observed after MP transfusion in 24 h or the following consecutive 9 days. CONCLUSIONS: RIPC induces an increase in MPs, and platelet-derived MPs may confer at least part of the remote protective effect against cerebral ischemic-reperfusion injury.


Subject(s)
Cell-Derived Microparticles/pathology , Cerebral Infarction/etiology , Cerebral Infarction/prevention & control , Infarction, Middle Cerebral Artery/complications , Ischemic Preconditioning , Animals , Cell-Derived Microparticles/ultrastructure , Disease Models, Animal , Endothelial Cells/pathology , Flow Cytometry , Male , Microscopy, Electron, Transmission , Nervous System Diseases/diagnosis , Nervous System Diseases/etiology , Platelet Membrane Glycoprotein IIb/metabolism , Rats , Rats, Sprague-Dawley , Upper Extremity/physiopathology
13.
Article in Chinese | MEDLINE | ID: mdl-16188098

ABSTRACT

OBJECTIVE: To explore the relationship between the polymorphism of HLA-DRB1*, DQB* genes and the susceptibility of pneumoconiosis. METHODS: 1:1 case-control study was adopted. one hundred and thirteen cases of I grade pneumoconiosis were investigated. The control group were workers exposed to dust, who were the same sex, nationality, work place, time of beginning exposure and the cumulative exposure ages not over 2 years. PCR-SSP was used to detect 9 alleles in HLA-DRB1*, DQB1*. Information on related factors of pneumoconiosis was collected using a questionnaire. Univariate and multivariate logistic regression analysis were carried out with 1:1 case-control methodology. RESULTS: The frequency of HLA-DRB1*08 allele in case group was significantly higher than that of the controls (OR: 6.000; 95% CI: 1.9060 - 18.9414). The frequencies of HLA-DRB1*09, HLA-DQB1*06 in case group were significantly lower than those of the controls (OR: 0.259, 0.300; 95% CI: 0.1436 - 0.6268, 0.1149 - 0.5837 respectively). There were significant relationship between HLA-DRB1*08, HLA-DRB1*09, HLA-DQB1*06 alleles and pneumoconiosis after adjusting age, smoking, beginning age of exposure and cumulative length of exposure with multivariate logistic regression analysis (OR: 7.804, 0.225, and 0.269; 95% CI: 2.077 - 29.307, 0.083 - 0.609 and 0.117 - 0.613 respectively. Survival analysis showed that HLA-DQB1*06 allele was a protective factor and HLA-DRB1*08 allele was a risk factor for affecting pneumoconiosis latent period. CONCLUSION: HLA-DRB1*08 allele may be the susceptible risk gene for pneumoconiosis. HLA-DQB1*06 may be the protective gene against developing pneumoconiosis.


Subject(s)
HLA-DQ beta-Chains/genetics , HLA-DRB1 Chains/genetics , Pneumoconiosis/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Case-Control Studies , Gene Frequency , Genetic Predisposition to Disease , Humans , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...