Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharm Res ; 38(8): 1397-1404, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34282500

ABSTRACT

PURPOSES: Liquid protein-based biopharmaceutical formulations have been reported to form aggregation and protein sub-visible particles (SbVPs) during dropping (Randolph et al., J Pharm Sci 2015, 104, 602). However, effects of secondary package on liquid biopharmaceutical formulation stability during dropping are overlooked and have not been reported so far. This study reports the first real-world evaluation on effects of secondary package on liquid biopharmaceutical formulation stability during dropping, using two monoclonal antibodies (mAb-1 and mAb-2) and one fusion protein (FP-1) as model biopharmaceuticals. METHODS: The potential protective effects of secondary package and formulation composition on liquid biopharmaceutical formulations during dropping were evaluated with micro-flow imaging (MFI) and dynamic light scattering (DLS). RESULTS: The dropping-induced degradation could be detected with the two sensitive particle analyzing techniques MFI and DLS. Formulation compositions have dramatic impact on biopharmaceutical stability during dropping. Surprisingly, unlike the primary packages that have been reported to impact liquid biopharmaceutical stability, the secondary packaging system as described in our current preliminary design has little or no protective effect during dropping. CONCLUSIONS: Our study is the first real-world data showing that the secondary package system has little to no effect on the liquid biopharmaceutical formulation quality during dropping. On the contrary, the stability of liquid biopharmaceutical formulations during dropping is more relevant to formulation compositions and primary packages.


Subject(s)
Biological Products/chemistry , Drug Packaging , Antibodies, Monoclonal/chemistry , Drug Compounding , Drug Stability , Dynamic Light Scattering
2.
Pharm Dev Technol ; 26(1): 21-29, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33070673

ABSTRACT

Multidrug resistance (MDR) is a serious challenge in chemotherapy and also a major threat to breast cancer treatment. As an intracellular energy factory, mitochondria provide energy for drug efflux and are deeply involved in multidrug resistance. Mitochondrial targeted delivery of doxorubicin can overcome multidrug resistance by disrupting mitochondrial function. By incorporating a reactive oxygen species (ROS)-responsive hydrophobic group into the backbone structure of hyaluronic acid - a natural ligand for the highly expressed CD44 receptor on tumor surfaces, a novel ROS-responsive and CD44-targeting nano-carriers was constructed. In this study, mitochondria-targeted triphenylphosphine modified-doxorubicin (TPP-DOX) and amphipathic ROS-responsive hyaluronic acid derivatives (HA-PBPE) were synthesized and confirmed by 1H NMR. The nanocarriers TPP-DOX @ HA-PBPE was prepared in a regular shape and particle size of approximately 200 nm. Compared to free DOX, its antitumor activity in vitro and tumor passive targeting in vivo has been enhanced. The ROS-responsive TPP-DOX@HA-PBPE nanocarriers system provide a promising strategy for the reverse of MDR and efficient delivery of doxorubicin derivatives into drug-resistant cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Breast Neoplasms/metabolism , Doxorubicin/metabolism , Drug Resistance, Multiple/drug effects , Nanoparticles/metabolism , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems/methods , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Reactive Oxygen Species/chemistry
3.
Pharm Res ; 37(11): 228, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33098017

ABSTRACT

PURPOSES: The main purposes of this article are to describe an unprecedented phenomenon in which significant amount of a shoulder peak impurity was observed during normal non-reducing capillary electrophoresis-sodium dodecyl sulfate (CE-SDS) analysis of a recombinant fusion protein X, and to evaluate the root cause for this phenomenon. METHODS: A series of experiments were conducted to study the nature of this degradation. Effects of iodoacetamide (IAM), heating temperature, duration, and SDS on the formation of this specific impurity were evaluated using a variety of characterization techniques. RESULTS: The formation of the impurity as observed in CE-SDS was actually due to alkylation of lysine and serine residues with IAM, as confirmed by peptide mapping and LC-MS/MS, which increased the molecular weight and therefore decreased the electrophoretic mobility. The amount of impurity was also strongly dependent on sample preparation conditions including the presence or absence of SDS. CONCLUSIONS: Our study clearly suggested that even though IAM has been used extensively as an alkylation reagent in the traditional non-reducing CE-SDS analysis of monoclonal antibodies and other proteins, alkylation with IAM could potentially lead to additional impurity peak, and therefore complicating analysis. Therefore, before performing CE-SDS and other analyses, the effects of sample preparation procedures on analytical results must be evaluated. For protein X, IAM should be excluded for CE-SDS analysis.


Subject(s)
Recombinant Proteins/chemistry , Sodium Dodecyl Sulfate/chemistry , Antibodies, Monoclonal/chemistry , Chromatography, Liquid/methods , Electrophoresis, Capillary/methods , Electrophoresis, Polyacrylamide Gel/methods , Tandem Mass Spectrometry/methods
4.
Biol Pharm Bull ; 43(12): 1924-1930, 2020.
Article in English | MEDLINE | ID: mdl-33268710

ABSTRACT

Nanoparticles are used in many fields and in everyday products. Silver nanoparticles are the most frequently used nanoparticles; for example, in food-related products, owing to their antibacterial activity. However, it has been pointed out that they might have unexpected biological effects, and evaluation of their effects is underway. Although there is a growing body of evidence that nanoparticles can also induce epigenetic changes, there is still little information on the underlying mechanisms. Here, we evaluated changes in DNA methylation induced by silver nanoparticles and attempted to elucidate the induction mechanism. Immunofluorescence staining analysis revealed that silver nanoparticles with a diameter of 10, 50, or 100 nm (nAg10, nAg50, and nAg100, respectively) decreased the content of methylated DNA in A549 alveolar epithelial cells. The level of DNA methyltransferase 1 (Dnmt1) protein, which is involved in maintaining methylation during DNA replication, was significantly decreased, whereas that of Dnmt3b, which is responsible for de novo DNA methylation, was significantly increased by nAg10 treatment. Co-treatment with nAg10 and cycloheximide, which inhibits translation by inhibiting the translocation step of protein synthesis, decreased the level of Dnmt1 in comparison with nAg10-treated A549 cells, indicating a post-translational effect of nAg10. Furthermore, pretreatment with the proteasome inhibitor lactacystin restored the levels of Dnmt1 protein and DNA methylation in nAg10-treated cells. Collectively, these results suggest that nAg10 induced DNA hypomethylation through a proteasome-mediated degradation of Dnmt1.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA Methylation/drug effects , Metal Nanoparticles/administration & dosage , Proteolysis/drug effects , Silver/pharmacology , A549 Cells , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/physiology , Dose-Response Relationship, Drug , Humans , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism
5.
Mol Pharm ; 16(7): 2966-2979, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31095914

ABSTRACT

Doxorubicin (DOX) is a first-line chemo drug for cancer therapy, yet it fails to treat multi-drug-resistant tumors. Hypoxia is a major causative factor leading to chemotherapy failure. Particularly, hypoxia up-regulates its responsive transcription factor-hypoxia-inducible factors (HIF)-to induce the overexpression of drug resistant genes. Metformin (MET) is recently found to cooperate with DOX against multiple tumors. As a mitochondrial inhibitor, MET could suppress tumor oxygen consumption, and thereby modulate the hypoxic tumor microenvironment. In this study, we used cationic liposomes to codeliver both DOX and MET for treating multi-drug-resistant breast cancer cells-MCF7/ADR. Faster release of MET enhanced the cytotoxicity of DOX through attenuating hypoxic stress both in vivo and in vitro. MET diminished the cellular oxygen consumption and inhibited HIF1α and P-glycoprotein (Pgp) expression in vitro. In addition, the dual-drug-loaded liposomes increased tumor targeting and intratumoral blood oxygen saturation, which suggested that the tumor reoxygenation effect of MET facilitated the exertion of its synergistic activity with DOX against MCF7/ADR xenografts. In general, our study represents a feasible strategy to boost the therapeutic effect in treating multi-drug-resistant cancer by improving the hypoxic tumor microenvironment.


Subject(s)
Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Cell Hypoxia/drug effects , Doxorubicin/therapeutic use , Drug Delivery Systems/methods , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Metformin/therapeutic use , Tumor Microenvironment/drug effects , Animals , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/metabolism , Drug Compounding/methods , Drug Liberation , Drug Synergism , Female , Humans , Liposomes/chemistry , MCF-7 Cells , Metformin/administration & dosage , Metformin/metabolism , Mice , Mice, Nude , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Pharm Res ; 36(8): 118, 2019 Jun 03.
Article in English | MEDLINE | ID: mdl-31161359

ABSTRACT

PURPOSE: The main purposes of this manuscript are to report a surprising and interesting degradation reaction of glucagon from a specific vendor in which glucagon underwent cleavage among several peptide bonds quickly under near neutral to basic conditions, and to propose the root cause of mechanism for the degradation reaction. METHODS: The degradation reaction was monitored by HPLC and the fragment structures were confirmed by LC-MS. Possible impurities responsible for the degradation were either confirmed or excluded by a variety of techniques such as addition of chelator EDTA and transitional metal ions or separation by ultrafiltration. RESULTS: This type of degradation was rarely reported in literature, especially considering its extreme cleavage efficiency. Contamination by a thermostable high molecular impurity (such as a peptidase with molecular weight between 10 and 30 KDa) during the manufacturing process was the main reason for this interesting phenomenon. CONCLUSIONS: The degradation phenomenon described here could be used as an excellent example showing that products ordered from vendors meeting the rudimentary quality standards might contain impurities which could cause significant degradation. We suggest that a simple solution, i.e. additional tests of stability under real or accelerated conditions by manufacturers and inclusion of the "accelerated stability criteria" in the Certificate of Analysis (CoAs), especially for sensitive biological reagents prone to faster degradation, would be very helpful for avoiding losses for both vendors and users.


Subject(s)
Glucagon/chemistry , Chelating Agents/chemistry , Chromatography, High Pressure Liquid , Drug Contamination , Drug Stability , Edetic Acid/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Peptide Hydrolases/chemistry , Protein Stability , Tandem Mass Spectrometry , Transition Elements/chemistry , Ultrafiltration
7.
Acta Pharmacol Sin ; 40(4): 546-555, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29930276

ABSTRACT

Despite more effective chemotherapy combined with limb-salvage surgery for the osteosarcoma treatment, survival rates for osteosarcoma patients have stagnated over the past three decades due to the poor prognosis. Osteosarcoma cancer stem cells (OSCs) are responsible for the growth and metastasis of osteosarcoma. The existence of OSCs offers a theoretical explanation for therapeutic failures including tumor recurrence, metastasis, and drug resistance. Understanding the pathways that regulate properties of OSCs may shed light on mechanisms that lead to osteosarcoma and suggest better modes of treatment. In this study, we showed that the expression level of Kruppel-like factor 4 (KLF4) is highly associated with human osteosarcoma cancer stemness. KLF4-overexpressed osteosarcoma cells displayed characteristics of OSCs: increased sphere-forming potential, enhanced levels of stemness-associated genes, great chemoresistance to adriamycin and CDDP, as well as more metastasis potential. Inversely, KLF4 knockdown could reduce colony formation in vitro and inhibit tumorigenesis in vivo, supporting an oncogenic role for KLF4 in osteosarcoma pathogenesis. Furthermore, KLF4 was shown to activate the p38 MAPK signaling pathway to promote cancer stemness. Altogether, our studies uncover an essential role for KLF4 in regulation of OSCs and identify KLF4-p38 MAPK axis as a potential therapeutic target for osteosarcoma treatment.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Neoplastic Stem Cells/metabolism , Osteosarcoma/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Phenotype , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Drug Dev Ind Pharm ; 45(6): 1009-1016, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30950303

ABSTRACT

Pearl powder has been used to treat many diseases like palpitations, insomnia, and epilepsy for thousands of years in Chinese medicine. It has demonstrated antioxidant, antiaging, antiradiative, and tonic activities. Pearl powder contains multiple active proteins, which are nutritious for skin cells and might be advantageous for wound repair and regeneration. However, its healing effect in vivo was not reported yet. This study aims to investigate the effects and the underlying mechanism of the pearl powders with different particle sizes in wound treatment. Briefly, the pearl powder with different sizes was characterized for their particle sizes and morphology. The protein release profiles of these powders were also studied. The influence of the different size of pearl powder in the proliferation, migration of skin cells was evaluated. Then, with the rat skin excision model, the effect of pearl powder on wound repair and regeneration was investigated. It was demonstrated that, all the micro and nanosized pearl powders could both increase the proliferation and migration of skin cells and accelerate the wound closure, as well as significantly enhanced the biomechanic strength of the healed skins. Moreover, the pearl powder treatment could improve the formation and regular deposition of collagen, and enhance the skin angiogenesis. Among all these in vitro and in vivo investigations, nanoscale pearl powder expressed the highest efficiency for healing. The mechanism might be contributed to the increased release of active proteins, enhanced tissue attachment, and the increased cellular uptake for the nano powder at the topical site.


Subject(s)
Nacre/administration & dosage , Nanoparticles/administration & dosage , Pinctada/chemistry , Skin Physiological Phenomena/drug effects , Wound Healing/drug effects , Administration, Cutaneous , Animals , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Fibroblasts , Humans , Nacre/chemistry , Nanoparticles/chemistry , Particle Size , Powders , Rabbits , Rats , Rats, Sprague-Dawley , Skin/drug effects , Skin/injuries
9.
Drug Dev Ind Pharm ; 45(9): 1556-1564, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31271317

ABSTRACT

Objective: This study was aimed to develop DOX-TPP loaded acetal-PEG-PCCL micelles to improve the clinical efficacy of drug resistance tumor. Significance: Chemotherapy is one of the main treatments for breast cancer but is plagued by multidrug resistance (MDR). DOX-TPP-loaded micelles can enhance the specific concentration of drugs in the tumor and improve the efficacy and overcome MDR. Methods: In this study, DOX-TPP-loaded micelles based on acetal-PEG-PCCL were prepared and their physicochemical properties were characterized. The cellular uptake and ability to induce apoptosis of the micelles was confirmed by flow cytometry in MCF-7/ADR cells. In addition, cytotoxicity of the micelles was studied in MCF-7 cells and MCF-7/ADR cells. Confocal is used to study the subcellular distribution of DOX. Free DOX-TPP or DOX-TPP-loaded acetal-PEG-PCCL micelles were administered via intravenous injection in the tail vain for the biodistribution study in vivo. Results: The diameter of micelles was about 102.4 nm and their drug-loading efficiency is 61.8%. The structural characterization was confirmed by 1H NMR. The micelles exhibited better antitumor efficacy compared to free doxorubicin in MCF-7/ADR cells by MTT assay. The apoptotic rate and the cellular uptake of micelles were significantly higher than free DOX and DOX-TPP. Micelles can efficiently deliver mitochondria-targeting DOX-TPP to tumor cells. The result of bio-distribution showed that the micelles had stronger tumor infiltration ability than free drugs. Conclusions: In this study, mitochondriotropic DOX-TPP was conjugated to the nanocarrier acetal-PEG-PCCL via ionic interaction to form a polymer, which spontaneously formed spherical micelles. The cytotoxicity and cellular uptake of the micelles are superior to free DOX and exhibit mitochondrial targeting and passive tumor targeting, indicating that they have potential prospects.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Nanoconjugates/chemistry , Organophosphorus Compounds/administration & dosage , Acetals/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Drug Compounding , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Micelles , Mitochondria/drug effects , Mitochondria/pathology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Distribution
10.
Neurol Neurochir Pol ; 53(4): 265-270, 2019.
Article in English | MEDLINE | ID: mdl-31343070

ABSTRACT

AIM: The purpose of this study was to analyse the diagnostic value of gated myocardial perfusion imaging (G-MPI) in the evaluation of myocardial injury in sarcoglycanopathy. MATERIALS AND METHODS: Twenty-eight patients diagnosed with sarcoglycanopathy were evaluated using 99m- -methoxyisobutylisonitrile(99Tcm-MIBI) G-MPI. The data was processed into tomographic images, and the left ventricular function was analysed using quantitative gated SPECT (QGS) to assess the degree of impairment in myocardial and cardiac function. RESULTS: The images of 23 of the patients (82.1%) were positive. Two hundred and twenty-nine sub-segments with abnormal lesions were detected out of 391 cardiac sub-segments of these 23 positive cases. According to the segmental abnormalities, the cases were divided into two cases (8.7%) with single abnormal wall segment, six cases (26.1%) with two abnormal wall segments, and 15 cases (65.2%) with three or more abnormal wall segments or scattered lesions. CONCLUSIONS: 99Tcm-MIBI G-MPI can objectively show impaired myocardium in patients with sarcoglycanopathy. Therefore, this method is helpful for early diagnosis and follow-up of myocardial damage.


Subject(s)
Myocardial Perfusion Imaging , Sarcoglycanopathies , Humans , Radiopharmaceuticals , Sarcoglycanopathies/diagnosis , Technetium Tc 99m Sestamibi , Tomography, Emission-Computed, Single-Photon
11.
Mol Pharm ; 15(3): 882-891, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29357260

ABSTRACT

Multidrug resistance (MDR) is the major obstacle for chemotherapy. In a previous study, we have successfully synthesized a novel doxorubicin (DOX) derivative modified by triphenylphosphonium (TPP) to realize mitochondrial delivery of DOX and showed the potential of this compound to overcome DOX resistance in MDA-MB-435/DOX cells. (1) To introduce specificity for DOX-TPP to cancer cells, here we report on the conjugation of DOX-TPP to hyaluronic acid (HA) by hydrazone bond with adipic acid dihydrazide (ADH) as the acid-responsive linker, producing HA- hydra-DOX-TPP nanoparticles. Hyaluronic acid (HA) is a natural water-soluble linear glycosaminoglycan, which was hypothesized to increase the accumulation of nanoparticles containing DOX-TPP in the mitochondria of tumor cells upon systemic administration, overcoming DOX resistance, in vivo. Our results showed HA- hydra-DOX-TPP to self-assemble to core/shell nanoparticles of good dispersibility and effective release of DOX-TPP from the HA- hydra-DOX-TPP conjugate in cancer cells, which was followed by enhanced DOX mitochondria accumulation. The HA- hydra-DOX-TPP nanoparticles also showed improved anticancer effects, better tumor cell apoptosis, and better safety profile compared to free DOX in MCF-7/ADR bearing mice.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Mitochondria/metabolism , Nanoconjugates/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/chemistry , Drug Liberation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Lysosomes/drug effects , Lysosomes/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Acta Pharmacol Sin ; 39(10): 1681-1692, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29849132

ABSTRACT

Breast cancer is the leading cause of cancer-related death for women, and multidrug resistance (MDR) is the major obstacle faced by chemotherapy for breast cancer. We have previously synthesized a doxorubicin (DOX) derivative by conjugating DOX with triphenylphosphonium (TPP) to achieve mitochondrial delivery, which induced higher cytotoxicity in drug-resistant breast cancer cells than DOX itself. Due to its amphiphilicity, TPP-DOX is difficult to physically entrap in nanocarriers. Thus, we linked it to hyaluronic acid (HA) by a novel ionic bond utilizing the specific bromide ion of TPP to form supra-molecular self-assembled structures (HA-ionic-TPP-DOX). The product was analyzed uisng 1H-NMR, 13C-NMR and mass spectrometry. The HA nanocarriers (HA-ionic-TPP-DOX) were shown to self-assemble into spherical nanoparticles, and sensitive to acidic pH in terms of morphology and drug release. Compared with free DOX, HA-ionic-TPP-DOX produced much greater intracellular DOX accumulation and mitochondrial localization, leading to increased ROS production, slightly decreased mitochondrial membrane potential, increased cytotoxicity in MCF-7/ADR cells and enhanced tumor targeting in vivo. In xenotransplant zebrafish model with the MCF-7/ADR cell line, both TPP-DOX and HA-ionic-TPP-DOX inhibited tumor cell proliferation without inducing significant side effects compared with free DOX. In addition, we observed a better anti-tumor effect of HA-ionic-TPP-DOX on MCF-7/ADR cells in zebrafish than that of TPP-DOX treatment. Furthermore, HA-ionic-DOX-TPP exhibited favorable biocompatibility and anti-tumor effects in MCF-7/ADR tumor-bearing nude mice in comparison with the effects of TPP-DOX and DOX, suggesting the potential of HA-ionic-TPP-DOX for the targeted delivery and controlled release of TPP-DOX, which can lead to the sensitization of resistant breast tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Mitochondria/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Doxorubicin/chemistry , Drug Liberation , Drug Resistance, Neoplasm/drug effects , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Mice, Nude , Onium Compounds/chemistry , Organophosphorus Compounds/chemistry , Zebrafish
13.
Biochem Biophys Res Commun ; 484(3): 543-549, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28130106

ABSTRACT

When considering the safety of ingested nanomaterials, it is important to quantitate their transfer across intestinal cells; however, little information exists about the effects of nanomaterial size or exposure side (apical versus basolateral epithelial surface) on nanomaterial transfer. Here, we examined cellular internalization and transcellular transport, and the effects of nanomaterials on Caco-2 monolayers after apical or basolateral exposure to Ag or Au nanoparticles with various sizes. After apical treatment, both internalization and transfer to the basolateral side of the monolayers were greater for smaller Ag nanoparticles than for larger Ag nanoparticles. In contrast, after basolateral treatment, larger Ag nanoparticles were more internalized than smaller Ag nanoparticles, but the transfer to the apical side was greater for smaller Ag nanoparticles. Au nanoparticles showed different rules of internalization and transcellular transport compared with Ag nanoparticles. Furthermore, the paracellular permeability of the Caco-2 monolayers was temporarily increased by Ag nanoparticles (5 µg/mL; diameters, ≤10 nm) following basolateral but not apical exposure. We conclude that the internalization, transfer, and effects of nanomaterials in epithelial cell monolayers depend on the size and composition of nanomaterials, and the exposure side.


Subject(s)
Cell Membrane Permeability/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Gold/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry , Subcellular Fractions/chemistry , Caco-2 Cells , Diffusion , Gold/administration & dosage , Humans , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/ultrastructure , Particle Size , Respiratory Tract Absorption , Silver/administration & dosage , Subcellular Fractions/drug effects
14.
Biol Pharm Bull ; 40(5): 726-728, 2017.
Article in English | MEDLINE | ID: mdl-28458361

ABSTRACT

We previously reported that unmodified silica nanoparticles with diameters of 70 nm (nSP70) induced liver damage in mice, whereas nSP70 modified with carboxyl or amino groups did not. In addition, we have found that both unmodified and modified nSP70s localize in both Kupffer cells and parenchymal hepatocytes. We therefore evaluated the contributions of nSP70 uptake by these cell populations to liver damage. To this end, we pretreated mice with gadolinium (III) chloride hydrate (GdCl3) to prevent nSP70 uptake by Kupffer cells, subsequently injected the mice with either type of nSP70, and then assessed plasma levels of alanine aminotransferase (ALT). In mice given GdCl3, unmodified nSP70 increased ALT levels. From these data, we hypothesized that in GdCl3-treated mice, the unmodified nSP70 that was prevented from entering Kupffer cells was shunted to parenchymal hepatocytes, where it induced cytotoxicity and increased liver damage. In contrast, GdCl3 pretreatment had no effect on ALT levels in mice injected with surface-modified nSP70s, suggesting that modified nSP70s spared parenchymal hepatocytes and thus induced negligible liver damage. In cytotoxicity analyses, the viability of a parenchymal hepatocyte line was greater when exposed to surface-modified nSP70s than to unmodified nSP70s. These findings imply that the decreased liver damage associated with surface-modified compared with unmodified nSP70 is attributable to decreased cytotoxicity to parenchymal hepatocytes.


Subject(s)
Amines/chemistry , Carboxylic Acids/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Alanine Transaminase/analysis , Animals , Cell Line , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Female , Gadolinium/chemistry , Hepatocytes/drug effects , Kupffer Cells/drug effects , Liver Function Tests , Mice , Mice, Inbred BALB C , Nanoparticles/toxicity , Particle Size , Silicon Dioxide/toxicity , Surface Properties
15.
Drug Dev Ind Pharm ; 43(1): 55-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27498809

ABSTRACT

This study aims to investigate the solid lipid nanoparticle (SLN) as a novel vehicle for the sustained release and transdermal delivery of piroxicam, as well as to determine the anti-inflammation effect of piroxicam-loaded SLN. SLN formulation was optimized and the particle size, polydispersity index, zeta potential (ZP), encapsulation efficiency, drug release, and morphological properties were characterized. The transdermal efficiency and mechanism of the piroxicam-loaded SLNs were investigated in vitro. With the inflammation induced edema model in rat, the anti-inflammatory efficiency of piroxicam-enriched SLNs (Pir-SLNs) was evaluated. The SLN formulation was optimized as: lecithin 100 mg, glycerin monostearate 200 mg, and Tween (1%, w/w). The particle size is around 102 ± 5.2 nm with a PDI of 0.262. The ZP is 30.21 ± 2.05 mV. The prepared SLNs showed high entrapment efficiency of 87.5% for piroxicam. There is no interaction between piroxicam and the vehicle components. The presence of polymorphic form of lipid with higher drug content in the optimized Pir-SLNs enables the Pir-SLNs to release the drug with a sustained manner. Pir-SLNs with oleic acid as enhancer can radically diffuse into both the stratum corneum and dermal layer, as well as penetrate through the hair follicles and sebaceous glands with significantly higher density than the other control groups. Pir-SLNs promptly inhibited the inflammation since the 3rd hour after the treatment by decreasing the PGE2 level. SLN was demonstrated to be a promising carrier for encapsulation and sustained release of piroxicam. Pir-SLN is a novel topical preparation with great potential for anti-inflammation application.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Drug Carriers/pharmacokinetics , Drug Liberation , Nanoparticles/metabolism , Piroxicam/pharmacokinetics , Skin Absorption/drug effects , Administration, Cutaneous , Animals , Anti-Inflammatory Agents/administration & dosage , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/administration & dosage , Edema/drug therapy , Edema/metabolism , Edema/pathology , Nanoparticles/administration & dosage , Organ Culture Techniques , Piroxicam/administration & dosage , Rats , Rats, Sprague-Dawley , Skin Absorption/physiology
16.
J Nanosci Nanotechnol ; 16(6): 5498-505, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27427589

ABSTRACT

Development of novel strategy stimulating the healing with skin appendages regeneration is the critical goal for wound therapy. In this study, influence of the transplantation of bone marrow derived mesenchymal stem cells (MSCs) and epidermal stem cells (ESCs) with the nanofiberous scaffold prepared from silk fibroin protein in wound re-epithelization, collagen synthesis, as well as the skin appendages regeneration were investigated. It was shown that both the transplantation of MSCs and ESCs could significantly accelerate the skin re-epithelization, stimulate the collagen synthesis. Furthermore, the regenerative features of MSCs and ESCs in activating the blood vessels and hair follicles formation, respectively were suggested. These results demonstrated that the electrospinning nanofiberous scaffold is an advantageous carrier for the cells transplantation, but also provided the experimental proofs for the application of MSCs and ESCs as promising therapeutics in skin tissue engineering.


Subject(s)
Adult Stem Cells/cytology , Electricity , Fibroins/chemistry , Fibroins/pharmacology , Nanofibers/chemistry , Regeneration/drug effects , Wound Healing/drug effects , Adult , Adult Stem Cells/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Humans , Materials Testing , Skin/cytology , Skin/drug effects , Tissue Scaffolds/chemistry
17.
Nanomedicine ; 12(2): 421-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26711963

ABSTRACT

α-Mangostin (α-M) is a polyphenolic xanthone that protects and improves the survival of cerebral cortical neurons against Aß oligomer-induced toxicity in rats. α-M is a potential candidate as a treatment for Alzheimer's disease (AD). However, the efficacy was limited by the poor penetration of the drug through the blood-brain barrier (BBB). In this study, we modified the α-M liposome with transferrin (Tf) and investigated the intracellular distribution of liposomes in bEnd3 cells. In addition, the transport of α-M across the BBB in the Tf(α-M) liposome group was examined. In vitro studies demonstrated that the Tf(α-M) liposome could cross the BBB in the form of an integrated liposome. Results of the in vivo studies on the α-M distribution in the brain demonstrated that the Tf(α-M) liposome improved the brain delivery of α-M. These results indicated that the Tf liposome is a potential carrier of α-M against AD. FROM THE CLINICAL EDITOR: The use of α-Mangostin (α-M) as a potential agent to treat Alzheimer's disease (AD) has been reported. However, its use is limited by the poor penetration through the blood brain barrier. The delivery of this agent by transferrin-modified liposomes was investigated by the authors in this study. The positive results could point to a better drug delivery system for brain targeting.


Subject(s)
Blood-Brain Barrier/metabolism , Liposomes/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Transferrin/metabolism , Xanthones/administration & dosage , Xanthones/pharmacokinetics , Alzheimer Disease/drug therapy , Animals , Brain/metabolism , Cell Line , Drug Delivery Systems , Garcinia mangostana/chemistry , Mice , Neuroprotective Agents/chemistry , Rats, Sprague-Dawley , Xanthones/chemistry
18.
Yao Xue Xue Bao ; 51(2): 257-63, 2016 02.
Article in Zh | MEDLINE | ID: mdl-29856579

ABSTRACT

Mitochondrion is one of the most vital organelles in cells of human body, and it is involved in many metabolic processes. Mitochondrion dysfunction is closely related to many diseases such as cancers, neurodegenerative diseases, obesity and ischemia reperfusion injury. As a result, mitochondrial drug delivery has gained more and more attention in the drug discovery against these diseases. This review gives a brief introduction to the relationship between mitochondria and human diseases(e.g., cancer), and summarizes the latest trend of mitochondrial targeting drug delivery system(MTDDS).


Subject(s)
Drug Delivery Systems , Mitochondria/drug effects , Humans , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Obesity/drug therapy , Reperfusion Injury/drug therapy
19.
Biochem Biophys Res Commun ; 447(3): 383-7, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24727452

ABSTRACT

Mesenchymal stem cells (MSCs) are non-hematopoietic cells with multi-lineage potential, which makes them attractive targets for regenerative medicine applications. Efficient gene transfer into MSCs is essential for basic research in developmental biology and for therapeutic applications involving gene-modification in regenerative medicine. Adenovirus vectors (Advs) can efficiently and transiently introduce an exogenous gene into many cell types via their primary receptors, the coxsackievirus and adenovirus receptors (CARs), but not into MSCs, which lack CAR expression. To overcome this problem, an Adv coated with cationic polymer polyethyleneimine (PEI) was developed. In this study, we demonstrated that PEI coating with an optimal ratio can enhance adenoviral transduction of MSCs without cytotoxicity. We also investigated the physicochemical properties and internalization mechanisms of the PEI-coated Adv. These results could help to evaluate the potentiality of the PEI-coated Adv as a prototype vector for efficient and safe transduction into MSCs.


Subject(s)
Adenoviridae/chemistry , Genetic Vectors/chemistry , Mesenchymal Stem Cells , Polyethyleneimine/chemistry , Transduction, Genetic/methods , Adenoviridae/physiology , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/physiology , Endocytosis , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Virion/chemistry , Virion/physiology , Virus Internalization
20.
Mol Pharm ; 11(10): 3322-9, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-24735422

ABSTRACT

The incidence of hepatic diseases continuously increases worldwide and causes significant mortality because of inefficient pharmacotherapy. Gene therapy is a new strategy in the treatment of hepatic diseases, but the disadvantages of insufficient retention in the liver and undesirable side effects hinder its application. In this study, we developed a novel nonviral vehicle targeted to liver. Mannan was cationized with spermine at varying grafted ratios to deliver the gene and was optimized in cytotoxicity and transfection in vitro. A spermine-mannan (SM)-based delivery system was proven to be hepatic targeted and was capable of prolonging the gene retention period in the liver. Moreover, SM at N/P of 20 was confirmed to be less interfered with by the serum. Optimized SM vehicle has relatively high hepatic transfection with almost no toxicity induction in the liver, which highlighted its potential in the treatment of hepatic diseases.


Subject(s)
Cations/chemistry , Genetic Vectors/chemistry , Genetic Vectors/chemical synthesis , Liver/metabolism , Mannans/chemistry , Spermine/chemistry , Transfection/methods , Animals , Gene Transfer Techniques , Hep G2 Cells , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL