Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Adv Mater ; 35(10): e2209603, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36524741

ABSTRACT

Glutathione (GSH)-activatable probes hold great promise for in vivo cancer imaging, but are restricted by their dependence on non-selective intracellular GSH enrichment and uncontrollable background noise. Here, a holographically activatable nanoprobe caging manganese tetraoxide is shown for tumor-selective contrast enhancement in magnetic resonance imaging (MRI) through cooperative GSH/albumin-mediated cascade signal amplification in tumors and rapid elimination in normal tissues. Once targeting tumors, the endocytosed nanoprobe effectively senses the lysosomal microenvironment to undergo instantaneous decomposition into Mn2+ with threshold GSH concentration of ≈ 0.12 mm for brightening MRI signals, thus achieving high contrast tumor imaging and flexible monitoring of GSH-relevant cisplatin resistance during chemotherapy. Upon efficient up-regulation of extracellular GSH in tumor via exogenous injection, the relaxivity-silent interstitial nanoprobe remarkably evolves into Mn2+ that are further captured/retained and re-activated into ultrahigh-relaxivity-capable complex by stromal albumin in the tumor, and simultaneously allows the renal clearance of off-targeted nanoprobe in the form of Mn2+ via lymphatic vessels for suppressing background noise to distinguish tiny liver metastasis. These findings demonstrate the concept of holographic tumor activation via both tumor GSH/albumin-mediated cascade signal amplification and simultaneous background suppression for precise tumor malignancy detection, surveillance, and surgical guidance.


Subject(s)
Albumins , Glutathione , Magnetic Resonance Imaging , Metal Nanoparticles , Molecular Probes , Neoplasms , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Glutathione/pharmacology , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Molecular Probes/pharmacology , Albumins/administration & dosage , Albumins/pharmacokinetics , Albumins/pharmacology , Magnetic Resonance Imaging/methods , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Image Enhancement/methods , Holography/methods , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Metal Nanoparticles/administration & dosage , Transferrin/administration & dosage , Transferrin/pharmacokinetics , Transferrin/pharmacology , Tissue Distribution , A549 Cells , Humans , Animals , Mice , Mice, Inbred BALB C , Mice, Nude , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Cisplatin/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology
2.
J Mater Chem B ; 9(45): 9370-9382, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34726686

ABSTRACT

Injectable cell-based hydrogels allow surgical operation in a minimally invasive way for articular cartilage lesions but the chondrocytes in the injectable hydrogels are difficultly arrayed and fixed at the site of interest to repair the cartilage tissue. In this study, an injectable hyaluronic acid-polyacrylic acid (HA-pAA) hydrogel was first synthesized using hyaluronic acid-cyclodextrin (HA-CD) and polyacrylic acid-ferrocene (pAA-Fc) to provide cell-delivery and self-healing. To promote the cell fixation and alignment, porous poly(lactic-co-glycolic acid) (PLGA) magnetic microcapsules (PPMMs) with glutathione (GSH) loaded and iron oxide nanoparticles (IO) located in the shell were designed. The GSH-loaded PPMMs with layer-by-layer (LbL) assembly of hyaluronic acid (HA) and GSH (LbL-PPMMs) can provide a two-stage rapid and slow release of GSH to modulate the self-healing of the HA-pAA hydrogel at the injured site. Furthermore, the chondrocytes embedded in the HA-pAA hydrogel could be delivered through CD44 receptors on the HA polymer chains of LbL-PPMMs toward the surface of the damaged site by an internal magnetic force. The composite hydrogel system of chondrocytes/LbL-PPMMs/HA-pAA can provide the damaged cartilage with a more even and smooth surface than other groups in a rabbit model after 8 weeks of implantation. In addition, the chondrocytes in the deep zone tissue exhibit a columnar array, similar to the cell arrangement in normal cartilage tissue. Together with the cell navigation behavior and GSH release from the LbL-PPMM/HA-pAA hydrogel, a full closure of lesions on the cartilage tissue can be achieved. Our results demonstrate the highly promising potential of the injectable LbL-PPMM/HA-pAA system in cartilage tissue repair.


Subject(s)
Cartilage/injuries , Chondrocytes/drug effects , Glutathione/chemistry , Glutathione/pharmacokinetics , Hydrogels/chemistry , Magnetic Phenomena , Animals , Cell Survival , Delayed-Action Preparations , Glutathione/administration & dosage , Hyaluronic Acid , Magnetic Iron Oxide Nanoparticles , Male , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits
3.
Jpn J Clin Oncol ; 51(2): 218-227, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33051668

ABSTRACT

OBJECTIVE: Two phase I studies of darinaparsin including Japanese and Korean patients with relapsed/refractory peripheral T-cell lymphoma were performed to evaluate its safety (primary purpose), efficacy and pharmacokinetic profile (ClinicalTrials.gov: NCT01435863 and NCT01689220). METHODS: Patients received intravenous darinaparsin for 5 consecutive days at 200 mg/m2/day in 4-week cycles, 300 mg/m2/day in 4-week cycles or 300 mg/m2/day in 3-week cycles. RESULTS: Seventeen Japanese and 6 Korean patients were enrolled and treated. Drug-related adverse events developed in 18 patients (78%). Dose-limiting toxicity, grade 3 hepatic dysfunction, was reported on Day 15 of cycle 1 in 1 Japanese patient who received 300 mg/m2/day. The most common drug-related, grade ≥ 3 adverse events were lymphopenia (9%), neutropenia (9%) and thrombocytopenia (9%). No deaths occurred. In 14 evaluable patients, 1 and 3 patients had complete response and partial response, respectively. The plasma concentration-time profiles of arsenic, a surrogate marker for darinaparsin, were similar between Japanese and Korean patients. No significant difference was found in its pharmacokinetic profile. CONCLUSIONS: These data indicate the good tolerability and potential efficacy of darinaparsin in patients with relapsed/refractory peripheral T-cell lymphoma. Darinaparsin 300 mg/m2/day for 5 consecutive days in 3-week cycles is the recommended regimen for phase II study.


Subject(s)
Arsenicals/therapeutic use , Glutathione/analogs & derivatives , Lymphoma, T-Cell, Peripheral/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Aged, 80 and over , Arsenicals/administration & dosage , Arsenicals/adverse effects , Arsenicals/pharmacokinetics , Female , Glutathione/administration & dosage , Glutathione/adverse effects , Glutathione/pharmacokinetics , Glutathione/therapeutic use , Humans , Japan , Male , Middle Aged , Republic of Korea , Time Factors , Treatment Outcome , Young Adult
4.
Arch Biochem Biophys ; 695: 108643, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33122162

ABSTRACT

Artificial nanostructures using polymers to produce polymeric vesicles are inspired by the many intricate structures found in living organisms. Polymersomes are a class of self-assembled vesicles known for their great stability and application in drug delivery. They can be tuned according to their intended use by changing their components and introducing activable block copolymers that transform these polymersomes into smart nanocarriers. In this study, we propose the synthesis of a poly (ethylene oxide)-poly (ε-caprolactone)-based polymersome (PEO-PCL) loaded with GSH as a pH-responsive drug delivery molecule for cancer and protein alteration inhibition. Initially, the nanocarrier was synthesized and characterized by DLS, TEM/SEM microscopy as well as gel permeation chromatography (GPC) and 1H NMR. Their CMC formation, encapsulation efficiency, and pH responsiveness were analyzed. In addition, empty and GSH-loaded PEO-PCL polymersomes were tested for their toxicity and therapeutic effect on normal and cancer cells via an MTT test. Subsequently, protein alteration models (aggregation, glycation, and oxidation) were performed in vitro where the polymersomes were tested. Results showed that other than being non-toxic and able to highly encapsulate and release the GSH in response to acidic conditions, the nanocomposites do not hinder its content's ameliorative effects on cancer cells and protein alterations. This infers that polymeric nanocarriers can be a base for future smart biomedicine applications and theranostics.


Subject(s)
Drug Carriers , Glutathione , Neoplasm Proteins/metabolism , Neoplasms , Polyesters , Animals , Chlorocebus aethiops , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Glutathione/chemistry , Glutathione/pharmacokinetics , Glutathione/pharmacology , Glycosylation/drug effects , HeLa Cells , Humans , Hydrogen-Ion Concentration , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Vero Cells
5.
Bioorg Chem ; 100: 103910, 2020 07.
Article in English | MEDLINE | ID: mdl-32388424

ABSTRACT

Selenium nanoparticles (SeNPs) have become one of the most prospective and promising tools in the course of cancer diagnosis and therapy. Here we describe the synthesis of a novel radioactive platform for tumor imaging using selenium nanoparticles. SeNPs were synthetized using dithionite and glutathione as reducing and capping agent respectively with 5 mmol/L sodium selenite as a precursor and then SeNPs radiolabeled with technetium-99 m, the most common and famous radioactive isotope used for imaging purposes. A characteristic profile for the synthesized SeNPs was performed including size analysis, zeta potential, antioxidant activity, radiochemical yield and in-vivo biodistribution in normal and solid tumor bearing mice. Size analysis showed amorphous SeNP cores of a mean diameter of 21 ± 5 nm with a hydrodynamic size of 43 ± 8 nm and -28 mV zeta potential. The particles also showed a superior antioxidant activity of radical scavenging activity 55.6% according to DPPH assay, in addition, satisfying radiochemical yield up to 97 ± 1.5 was achieved. In vivo studies were applied on male Swiss albino mice that demonstrated a good biodistribution pattern in normal mice with a moderate accumulation in liver at 30 min post injection. Excellent T/NT ratios were obtained in solid tumor bearing mice throughout the experimental time points. The as-synthetized selenium nanoparticles demonstrated surprising and satisfying features which make them promising enough for tumor theranosis.


Subject(s)
Glutathione/chemistry , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Selenium/chemistry , Technetium/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Glutathione/pharmacokinetics , Male , Mice , Nanoparticles/analysis , Radionuclide Imaging , Selenium/pharmacokinetics , Technetium/pharmacokinetics , Tissue Distribution
6.
Drug Metab Dispos ; 48(1): 52-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31704713

ABSTRACT

Glutathione (GSH) has been reported to be closely related to various diseases of the central nervous system, yet its authentic active ingredients and action sites remain unclear. In the present study, oral exogenous GSH significantly alleviated ischemic brain injury, but this result was inconsistent with its low bioavailability and blood-brain barrier (BBB) permeability. To ascertain the exposure of GSH-derived ingredients, including GSH, cysteine (CYS), glutamate (Glu), glycine (GLY), CYS-GLY, and γ-glutamylcysteine (γ-GC) were systematically studied both in vitro and in vivo. The outcomes demonstrated that oral GSH not only increases the GSH and CYS levels in rat striatum and cortex, but it also can decrease the rise of intracerebral Glu concentration caused by ischemia/reperfusion surgery. Then the influence of GSH on the BBB was investigated via measuring IgG leakage, intracerebral endotoxin, and tight-junction proteins. All indicators showed that GSH dosing can repair the destroyed BBB. Oral GSH greatly enhances the exposure of GSH, CYS, CYS-GLY, and γ-GC in rat duodenum, jejunum, ileum, and colon. Accumulating evidence reveals a close link between brain injury and gastrointestinal dysfunction. Our findings further suggest that oral GSH significantly improves intestinal inflammatory damage and barrier disruptions. In conclusion, oral GSH can have a direct therapeutic role in brain injury by stabilizing intracerebral levels of GSH, CYS, and Glu. It can also play an indirect therapeutic role by enhancing the intestinal exposure of GSH, CYS, CYS-GLY, and γ-GC and improving intestinal barrier disruptions. SIGNIFICANCE STATEMENT: The authentic active ingredients and action sites of exogenous glutathione (GSH) in the treatment of ischemic brain injury are unclear. We have shown that oral exogenous GSH not only stabilizes intracerebral levels of GSH, cysteine (CYS), and glutamate (Glu) to act directly on brain injury, but it can also exert an indirect therapeutic role by improving intestinal barrier disruptions. These findings have great significance for revealing the therapeutic effect of GSH on ischemic brain injury and for promoting its further development and clinical application.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Glutathione/pharmacokinetics , Glutathione/therapeutic use , Models, Biological , Reperfusion Injury/drug therapy , Administration, Oral , Animals , Binding Sites , Biotransformation , Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Glutathione/administration & dosage , Humans , Ileum/drug effects , Ileum/metabolism , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
7.
Hell J Nucl Med ; 22(2): 131-134, 2019.
Article in English | MEDLINE | ID: mdl-31273355

ABSTRACT

OBJECTIVE: Glutathione (GSH) plays an important role in a horde of cellular events that include cell proliferation and apoptosis.The present study describes the radio-synthesis and characterization of gallium-68 (68Ga)-labelled glutathione for its application in radionuclide imaging of cancer. SUBJECT AND METHODS: The radio-synthesis of radio-complex 68Ga-GSH was performed by the direct labeling method. The developed radio-complex was subjected to quality control tests. Colon tumors were developed in healthy male Sprague Dawley (S.D) rats by giving subcutaneous injections of dimethylhydrazine (DMH) in order to monitor the uptake of 68Ga-GSH radio-complex. RESULTS: Gallium-68-labelled glutathione was synthesized with a labeling efficiency of 73.5%±1%. Percentage plasma protein binding and log Po/w values for the radio-complex were found to be 20%-30% and -0.223±0.12, respectively. A significantly higher percentage specific uptake value of newly developed 68Ga-GSH complex was observed in colon tumor in comparison to soft tissue at 90 minutes post administration thereby exhibiting specificity for cancerous cells, which was also witnessed significantly increased overtime from the ratio of colon tumor uptake to normal colon uptake (P≤0.05). CONCLUSION: Therefore, 68Ga-labelled glutathione can further be exploited for radionuclide imaging and assessment of tumor drug resistance in patients.


Subject(s)
Colonic Neoplasms/metabolism , Gallium Radioisotopes , Glutathione/chemistry , Glutathione/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Animals , Glutathione/pharmacokinetics , Isotope Labeling , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
8.
J Cosmet Dermatol ; 18(3): 728-737, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30895708

ABSTRACT

BACKGROUND: Glutathione is one of agents which is commonly used to lighten skin color in Asia as a dietary supplement. Previous studies suggest its potential effect of glutathione on skin color. However, the clinical efficacy of glutathione in oral form is still questionable due to its limited absorption and bioavailability. AIM: To determine the clinical effects of glutathione on skin color and related skin conditions. PATIENTS/METHODS: A systematic review was conducted using PubMed, CINAHL, Scopus, EMBASE and Cochrane library were searched from inceptions to October 2017. All clinical studies evaluating the effect of glutathione on any skin effects in healthy volunteer were included. RESULTS: A total of four studies were included. Three studies were RCTs with placebo control, while one was a single-arm trial. One study used topical form, while others used oral form of glutathione with 250 to 500 mg/day. We found that both oral glutathione with the dosage of 500 mg/day and topical 2.0% oxidized glutathione could brighten skin color in sun-exposed area measured by skin melanin index. No significant differences in the reduction in skin melanin index were observed in sun-protected area for any products. In addition, glutathione also has a trend to improve skin wrinkle, skin elasticity, and UV spots. Some adverse events but nonserious were reported. CONCLUSIONS: Current evidence of the skin whitening effect of glutathione is still inconclusive due to the quality of included studies and inconsistent findings. However, there is a trend that glutathione might brighten skin color at skin-exposed area.


Subject(s)
Dietary Supplements , Glutathione/administration & dosage , Skin Lightening Preparations/administration & dosage , Skin Pigmentation/drug effects , Administration, Cutaneous , Administration, Oral , Biological Availability , Glutathione/pharmacokinetics , Humans , Melanins/analysis , Melanins/metabolism , Randomized Controlled Trials as Topic , Skin/chemistry , Skin/metabolism , Skin/radiation effects , Skin Absorption , Skin Lightening Preparations/pharmacokinetics , Skin Pigmentation/radiation effects , Sunlight/adverse effects , Treatment Outcome
9.
Drug Deliv ; 26(1): 216-225, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30843439

ABSTRACT

To develop proliposome formulations to improve the oral bioavailability of l-glutathione (GSH), GSH-loaded proliposomes were prepared using the granule method. Mannitol was selected as an effective excipient to achieve the desired particle size, entrapment efficiency (EE), and solubility for oral delivery of the final formulation. To evaluate the effect of surface charge of proliposomes on the oral bioavailability of GSH, negative (F1-F4) and positive proliposomes (F5-F9) were prepared. Particle size of F1 and F5 was 167.8 ± 0.9 and 175.9 ± 2.0 nm, and zeta potential of F1 and F5 was -8.1 ± 0.7 and 21.1 ± 2.0 mV, respectively. Encapsulation efficiency of F1 and F5 was 58.6% and 54.7%, respectively. Considering their particle size, zeta potential, and EE, the proliposomes F1 and F5 were adopted as the optimal formulations for further experiments. Solid state characterization of the proliposomes confirmed lipid coating on the surface of mannitol. The release of GSH from F1 and F5 formulations was prolonged until 24 h and pH independent. The total antioxidant capacity of GSH was concentration-dependent and maintained after formulation of GSH proliposomes. Circular dichroism spectroscopy confirmed that the molecular structure of GSH was maintained in the proliposome formulations. GSH proliposomes exhibited no significant changes in particle size and zeta potential for 4 weeks. An oral bioavailability study in rats revealed that F5 exhibited 1.05-, 1.08-, and 1.11-fold higher bioavailability than F1, commercial capsule formulation, and pure GSH, respectively. In conclusion, the prepared GSH proliposomes enhanced the poor bioavailability of GSH and prolonged its duration of action.


Subject(s)
Antioxidants/chemistry , Glutathione/chemistry , Liposomes/chemistry , Peptides/chemistry , Administration, Oral , Animals , Antioxidants/pharmacokinetics , Biological Availability , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Drug Carriers/chemistry , Drug Delivery Systems/methods , Excipients/chemistry , Glutathione/pharmacokinetics , Humans , KB Cells , Lipids/chemistry , Male , Particle Size , Peptides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility
10.
Acta Pharmacol Sin ; 40(5): 699-709, 2019 May.
Article in English | MEDLINE | ID: mdl-30218071

ABSTRACT

The combinational administration of antioxidants and chemotherapeutic agents during conventional cancer treatment is among one of the most controversial areas in oncology. Although the data on the combinational usage of doxorubicin (DOX) and glutathione (GSH) agents have been explored for over 20 years, the duration, administration route, and authentic rationality have not yet been fully understood yet. In the current study, we systematically investigated the pharmacokinetics (PK) and pharmacodynamics (PD) with both in vivo and in vitro models to elucidate the influence of GSH on the toxicity and efficacy of DOX. We first studied the cardioprotective and hepatoprotective effects of GSH in Balb/c mice, H9c2, and HL7702 cells. We showed that coadministration of exogenous GSH (5, 50, and 500 mg/kg per day, intragastric) significantly attenuated DOX-induced cardiotoxicity and hepatotoxicity by increasing intracellular GSH levels, whereas the elevated GSH concentrations did not affect the exposure of DOX in mouse heart and liver. From PK and PD perspectives, then the influences of GSH on the chemotherapeutic efficacy of DOX were investigated in xenografted nude mice and cancer cell models, including MCF-7, HepG2, and Caco-2 cells, which revealed that administration of exogenous GSH dose-dependently attenuated the anticancer efficacy of DOX in vivo and in vitro, although the elevated GSH levels neither influenced the concentration of DOX in tumors in vivo, nor the uptake of DOX in MCF-7 tumor cells in vitro. Based on the results we suggest that the combined administration of GSH and DOX should be contraindicated during chemotherapy unless DOX has caused serious hepatotoxicity and cardiotoxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Cardiotonic Agents/therapeutic use , Cardiotoxicity/prevention & control , Chemical and Drug Induced Liver Injury/prevention & control , Doxorubicin/therapeutic use , Glutathione/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacokinetics , Cell Line, Tumor , Contraindications, Drug , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/toxicity , Drug Therapy, Combination , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Heterografts , Humans , Liver/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Myocardium/metabolism , Rats , Tissue Distribution
11.
J Mater Sci Mater Med ; 30(1): 5, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30569308

ABSTRACT

Glutathione-modified superparamagnetic iron oxide nanoparticles (GSH-SPIONs) were prepared by conjugating glutathione (GSH) on the surface of the PEG (Polyethylene glycol)/PEI (polyethyleneimine)-SPIONs which were synthesized by thermal decomposition method. Thermogravimetric analysis showed that the mass fraction of GSH on the surface of SPIONs was 30.64 wt%. GSH-SPIONs in PBS were injected into the substantia nigra of rat brains. The subcellular distributions of the nanoparticles in the brains was examined by the transmission electron microscope (TEM). A remarkable amount of GSH-SPIONs were found in vesicles inside cell bodies and axons, and in mitochondria. TEM pictures show that GSH-SPIONs enter the neuronal cells by endocytosis and travel through axoplasmic transport. GSH-SPIONs have great potential as drug delivery agents in the brain to treat diseases or study brain function via mitochondria-targeting way or axoplasmic transport way.


Subject(s)
Glutathione/chemistry , Glutathione/pharmacokinetics , Magnetite Nanoparticles/chemistry , Substantia Nigra/metabolism , Animals , Cell Survival , PC12 Cells , Rats , Substantia Nigra/chemistry , Tissue Distribution
12.
Int J Pharm ; 552(1-2): 16-26, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30253209

ABSTRACT

In the present study, we have developed the robust nanoparticles (MGC-GNP/PTX), which are TAMs and tumor cells-dual recognizable, for targeting cancer therapy. Of great importance, the developed nano-platforms are glutathione (GSH)-activable, which means it remains structure intact under normal physiological condition and can be disrupted when exposed to certain concentration of GSH. As demonstrated by the drug release assay in vitro, MGC-GNP/PTX exhibited an excellent structure stability under the normal condition with only 10% of cumulative drug release at 72 h. However, after increasing the concentration of GSH to 1 mM or 10 mM, the release of PTX from the nanoparticles was significantly accelerated and approximately 35% or 95% of drugs was released. Cellular experiments and in vivo tumor targeting assay displayed that the developed nanoparticles have a super capacity of tumor cells and TAMs-dual targeting drug delivery, which resulted in much stronger cytotoxicity when compared to the unmodified ones. Finally, the pharmacodynamic evaluation indicated that the mice treated with MGC-GNP/PTX displayed the strongest tumor suppression ability versus other groups. More importantly, the treatment of MGC-GNP/PTX did not significantly influence the body weight and pathological of the mice, indicated that the prepared nanoparticle system had a satisfactory bio-safety for targeting tumor drug delivery.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Glutathione/administration & dosage , Lung Neoplasms/drug therapy , Nanoparticles/administration & dosage , Paclitaxel/administration & dosage , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Line , Drug Liberation , Glutathione/chemistry , Glutathione/pharmacokinetics , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics
13.
Int J Mol Sci ; 19(4)2018 Mar 29.
Article in English | MEDLINE | ID: mdl-29596317

ABSTRACT

Feline injection site sarcomas (FISS) are malignant skin tumors with high recurrence rates despite the primary treatment of radical surgical resections. Adjunctive radiotherapy or chemotherapy with doxorubicin is mostly ineffective. Cellular and molecular causes of multidrug resistance, specific physio-chemical properties of solid tumors impairing drug transport, and the tumor microenvironment have been indicated for causing standard chemotherapy failure. Gold nanoparticles are promising imaging tools, nanotherapeutics, and drug delivery systems (DDS) for chemotherapeutics, improving drug transport within solid tumors. This study was conducted to assess the distribution of 4-nm glutathione-stabilized gold nanoparticles in FISS and their influence on kidney and liver parameters in nude mice. The role of gold nanoparticles as a doxorubicin DDS in FISS was examined to determine the potential reasons for failure to translate results from in vitro to in vivo studies. Grade III tumors characterized by a large area of necrosis at their core displayed positive immuneexpression of tumor-associated macrophages (TAM) at both the periphery and within the tumor core near the area of necrosis. Gold nanoparticles did not cause necrosis at the injection site and had no negative effect on liver and kidney parameters in nude mice. Gold nanoparticles accumulated in the tumor core and at the periphery and co-internalized with TAM-an important observation and potential therapeutic target warranting further investigation. The large area of necrosis and high immunoexpression of TAM, indicating "pro-tumor macrophages", may be responsible for FISS tumor progression and therapeutic failure. However, further studies are required to test this hypothesis.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Drug Delivery Systems/methods , Glutathione , Gold , Metal Nanoparticles , Sarcoma , Skin Neoplasms , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Glutathione/chemistry , Glutathione/pharmacokinetics , Glutathione/pharmacology , Gold/chemistry , Gold/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Nude , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Adv Healthc Mater ; 6(16)2017 Aug.
Article in English | MEDLINE | ID: mdl-28504415

ABSTRACT

Gold nanoclusters are emerging as new materials for biomedical applications because of promises offered by their ultrasmall size and excellent biocompatibility. Here, the synthesis and optical and biological characterizations of a highly luminescent folate-functionalized Au22 cluster (Au22 -FA) are reported. The Au22 -FA clusters are synthesized by functionalizing the surface of Au22 (SG)18 clusters, where SG is glutathione, with benzyl chloroformate and folate. The functionalized clusters are highly water-soluble and exhibit remarkably bright luminescence with a quantum yield of 42%, significantly higher than any other water-soluble gold clusters protected with thiolate ligands. The folate groups conjugated to the gold cluster give rise to additional luminescence enhancement by energy transfer sensitization. The brightness of Au22 -FA is found to be 4.77 mM-1 cm-1 , nearly 8-fold brighter than that of Au22 (SG)18 . Further biological characterizations have revealed that the Au22 -FA clusters are well-suited for bioimaging. The Au22 -FA clusters exhibit excellent photostability and low toxicity; nearly 80% cell viability at 1000 ppm of the cluster. Additionally, the Au22 -FA clusters show target specificity to folate-receptor positive cells. Finally, the time-course in vivo luminescence images of intravenous-injected mice show that the Au22 -FA clusters are renal-clearable, leaving only 8% of them remained in the body after 24 h post-injection.


Subject(s)
Folic Acid/chemistry , Gold/chemistry , Luminescent Agents/chemistry , Metal Nanoparticles/chemistry , Molecular Imaging/methods , Animals , Cell Line, Tumor , Cell Survival/drug effects , Folic Acid/pharmacokinetics , Glutathione/chemistry , Glutathione/pharmacokinetics , Glutathione/toxicity , Gold/pharmacokinetics , Humans , Luminescent Agents/pharmacokinetics , Luminescent Agents/toxicity , Metal Nanoparticles/toxicity , Mice , Nanomedicine
15.
J Neurochem ; 141(5): 738-749, 2017 06.
Article in English | MEDLINE | ID: mdl-28294337

ABSTRACT

Copper is an essential metal for the function of many proteins related to important cellular reactions and also involved in the synaptic transmission. Although there are several mechanisms involved in copper homeostasis, a dysregulation in this process can result in serious neurological consequences, including degeneration of dopaminergic neurons. 6-Hydroxydopamine is a dopaminergic neurotoxin mainly used in experimental models of Parkinson's disease, whose neurotoxicity has been related to its ability to generate free radicals. In this study, we examined the effects induced by copper on 6-OHDA autoxidation. Our data show that both Cu+ and Cu2+ caused an increase in • OH production by 6-OHDA autoxidation, which was accompanied by an increase in the rate of both p-quinone formation and H2 O2 accumulation. The presence of ascorbate greatly enhanced this process by establishing a redox cycle which regenerates 6-OHDA from its p-quinone. However, the presence of glutathione did not change significantly the copper-induced effects. We observed that copper is able to potentiate the ability of 6-OHDA to cause both lipid peroxidation and protein oxidation, with the latter including a reduction in free-thiol content and an increase in carbonyl content. Ascorbate also increases the lipid peroxidation induced by the action of copper and 6-OHDA. Glutathione protects against the copper-induced lipid peroxidation, but does not reduce its potential to oxidize free thiols. These results clearly demonstrate the potential of copper to increase the capacity of 6-OHDA to generate oxidative stress and the ability of ascorbate to enhance this potential, which may contribute to the destruction of dopaminergic neurons.


Subject(s)
Adrenergic Agents/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Copper/pharmacology , Glutathione/pharmacokinetics , Oxidative Stress/drug effects , Oxidopamine/pharmacology , Animals , Brain/ultrastructure , Drug Synergism , Hydrogen Peroxide/metabolism , Lipid Peroxidation/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors
16.
J Cyst Fibros ; 16(3): 342-345, 2017 May.
Article in English | MEDLINE | ID: mdl-27988297

ABSTRACT

Glutathione (GSH) is decreased in cystic fibrosis (CF) airways, thus its resupply by inhalation has been employed to restore antioxidant defense. CF airways present however increased activity of gamma-glutamyltransferase (GGT), the enzyme specifically capable of degrading GSH, and thus inhaled GSH might be promptly catabolized. In addition, prooxidant reactions are known to originate during GGT-mediated GSH catabolism. We determined levels of GGT in the sputum samples obtained from a previously published trial of GSH inhalation treatment, and analyzed their correlations with inflammatory markers and FEV1% values. Results indicate that differentiating patients with increasing vs. decreasing GGT activity - as measured in sputum before and after the six months duration of the study - may discriminate subjects more likely profiting from inhaled GSH, as opposed to those with increasing GGT in which these treatments might even produce aggravation of the damage.


Subject(s)
Cystic Fibrosis , Glutathione , Sputum , gamma-Glutamyltransferase , Administration, Inhalation , Adolescent , Adult , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Biological Availability , Cystic Fibrosis/diagnosis , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Drug Monitoring/methods , Female , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Humans , Male , Prognosis , Sputum/drug effects , Sputum/metabolism , Treatment Outcome , gamma-Glutamyltransferase/analysis , gamma-Glutamyltransferase/metabolism
17.
Biochem Pharmacol ; 126: 79-86, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27889539

ABSTRACT

Inorganic arsenicals are well-known carcinogens, whereas arsenite (iAsIII) compounds are now recognized as potent therapeutic agents for several leukemias, and arsenic trioxide has been used for the treatment of recurrent acute promyelocytic leukemia (APL). However, recent clinical trials revealed that arsenite is not always effective for non-APL malignancies. Another arsenical, S-dimethylarsino-glutathione ([DMAIII(GS)]), which is a putative metabolic intermediate in the hepatic metabolism of iAsIII, shows promise for treating several types of lymphoma. However, the metabolism of [DMAIII(GS)] has not been well investigated, probably because [DMAIII(GS)] is not stable in biological fluids where the concentration of glutathione is low. In the present study, we injected [DMAIII(GS)] intravenously into mice and compared the tissue distribution and metabolic dynamics of [DMAIII(GS)] with those of sodium arsenite (NaAsO2). We found a unique organ preference for the distribution of [DMAIII(GS)] to the lung and brain in comparison to NaAsO2. Furthermore, [DMAIII(GS)] appeared to bind to serum albumin by exchanging its glutathione moiety quickly after administration, providing novel insights into the longer retention of [DMAIII(GS)] in plasma.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Arsenicals/pharmacokinetics , Arsenites/pharmacokinetics , Glutathione/analogs & derivatives , Sodium Compounds/pharmacokinetics , Animals , Antineoplastic Agents/blood , Arsenicals/blood , Glutathione/blood , Glutathione/pharmacokinetics , Injections, Intravenous , Male , Mice, Inbred C57BL , Protein Binding , Serum Albumin/metabolism , Tissue Distribution
18.
Drug Discov Today Technol ; 20: 59-69, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27986226

ABSTRACT

The blood-brain barrier (BBB) represents a major obstacle for the delivery and development of drugs curing brain pathologies. However, this biological barrier presents numerous endogenous specialized transport systems that can be exploited by engineered nanoparticles to enable drug delivery to the brain. In particular, conjugation of glutathione (GSH) onto PEGylated liposomes (G-Technology®) showed to safely enhance delivery of encapsulated drugs to the brain. Yet, understanding of the mechanism of action remains limited and full mechanistic understanding will aid in the further optimization of the technology. In order to elucidate the mechanism of brain targeting by GSH-PEG liposomes, we here demonstrate that the in vivo delivery of liposomal ribavirin is increased in brain extracellular fluid according to the extent of GSH conjugation onto the liposomes. In vitro, using the hCMEC/D3 human cerebral microvascular endothelial (CMEC) cell line, as well as primary bovine and porcine CMEC (and in contrast to non-brain derived endothelial and epithelial cells), we show that liposomal uptake occurs through the process of endocytosis and that the brain-specific uptake is also glutathione conjugation-dependent. Interestingly, the uptake mechanism is an active process that is temperature-, time- and dose-dependent. Finally, early endocytosis events rely on cytoskeleton remodeling, as well as dynamin- and clathrin-dependent endocytosis pathways. Overall, our data demonstrate that the glutathione-dependent uptake mechanism of the G-Technology involves a specific endocytosis pathway indicative of a receptor-mediated mechanism, and supports the benefit of this drug delivery technology for the treatment of devastating brain diseases.


Subject(s)
Antiviral Agents/administration & dosage , Brain/metabolism , Glutathione/administration & dosage , Polyethylene Glycols/administration & dosage , Ribavirin/administration & dosage , Animals , Antiviral Agents/pharmacokinetics , Biological Transport , Cattle , Cell Line , Cells, Cultured , Endothelial Cells/metabolism , Glutathione/chemistry , Glutathione/pharmacokinetics , HEK293 Cells , Humans , Liposomes , Male , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Rats, Wistar , Ribavirin/pharmacokinetics , Swine
19.
J Control Release ; 241: 1-14, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27586188

ABSTRACT

The targeted drug delivery to the central nervous system represents one of the major challenges in pharmaceutical formulations since it is strictly limited through the highly selective blood-brain barrier (BBB). l-Glutathione (GSH), a tripeptide and well-known antioxidant, has been studied in the last years as potential candidate to facilitate the receptor-mediated transcytosis of nanocarriers. We thus tested whether GSH decoration of a positively charged polymer, poly(ethylene imine), with this vector enables the transport of genetic material and, simultaneously, the passage through the BBB. In this study, we report the synthesis of GSH conjugated cationic poly(ethylene imine)s via ecologically desirable thiol-ene photo-addition. The copolymers, containing 80% primary or secondary amine groups, respectively, were investigated concerning their bio- and hemocompatibility as well as their ability to cross a hCMEC/D3 endothelial cell layer mimicking the BBB within microfluidically perfused biochips. We demonstrate that BBB passage depends on the used amino-groups and on the GSH ratio. Thereby the copolymer containing secondary amines showed an enhanced performance. We thus conclude that GSH-coupling represents a feasible and promising approach for the functionalization of nanocarriers intended to cross the BBB for the delivery of drugs to the central nervous system.


Subject(s)
Biocompatible Materials/chemical synthesis , Blood-Brain Barrier/metabolism , Drug Carriers/chemical synthesis , Gene Transfer Techniques , Glutathione/analogs & derivatives , Polyethyleneimine/analogs & derivatives , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Endothelial Cells/metabolism , Glutathione/chemical synthesis , Glutathione/chemistry , Glutathione/pharmacokinetics , HEK293 Cells , Humans , Mice , Microscopy, Fluorescence , Polyethyleneimine/chemical synthesis , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacokinetics
20.
Chemistry ; 22(44): 15937-15944, 2016 Oct 24.
Article in English | MEDLINE | ID: mdl-27649691

ABSTRACT

The kinetics and mechanisms of the copper(II)-catalyzed GSH (glutathione) oxidation are examined in the light of its biological importance and in the use of blood and/or saliva samples for GSH monitoring. The rates of the free thiol consumption were measured spectrophotometrically by reaction with DTNB (5,5'-dithiobis-(2-nitrobenzoic acid)), showing that GSH is not auto-oxidized by oxygen in the absence of a catalyst. In the presence of Cu2+ , reactions with two timescales were observed. The first step (short timescale) involves the fast formation of a copper-glutathione complex by the cysteine thiol. The second step (longer timescale) is the overall oxidation of GSH to GSSG (glutathione disulfide) catalyzed by copper(II). When the initial concentrations of GSH are at least threefold in excess of Cu2+ , the rate law is deduced to be -d[thiol]/dt=k[copper-glutathione complex][O2 ]0.5 [H2 O2 ]-0.5 . The 0.5th reaction order with respect to O2 reveals a pre-equilibrium prior to the rate-determining step of the GSSG formation. In contrast to [Cu2+ ] and [O2 ], the rate of the reactions decreases with increasing concentrations of GSH. This inverse relationship is proposed to be a result of the competing formation of an inactive form of the copper-glutathione complex (binding to glutamic and/or glycine moieties).


Subject(s)
Copper/chemistry , Glutathione/pharmacokinetics , Organometallic Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry , Catalysis , Glutathione/chemistry , Organometallic Compounds/chemistry , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...