Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 544
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.
Eur Rev Med Pharmacol Sci ; 26(8): 2794-2801, 2022 04.
Article in English | MEDLINE | ID: mdl-35503624

ABSTRACT

OBJECTIVE: The nitric oxide (NO) metabolite nitrite has been shown to attenuate hyperglycemia via its increase in insulin sensitivity and glucose uptake. However, the oral use of nitrite is limited due to its potential formation of the carcinogenic N-nitrosamines via reaction of acidic nitrite and the secondary amines. We investigated the anti-diabetic effect of sodium nitrite (SN) combined with glutathione (GSH) in streptozotocin (STZ)-induced diabetic mice for potential use of GSH as a protective agent in future nitrite therapy. MATERIALS AND METHODS: STZ-induced diabetic mice were orally treated for 5 weeks with vehicle, SN, GSH or SN + GSH. Oral glucose tolerance test and the measurement of fasting blood glucose (FBG) and glycosylated hemoglobin (HbA1c) levels were carried out to evaluate anti-diabetic effects of SN and SN + GSH. Plasma levels of total NO metabolites (NOx) were measured to confirm nitrite absorption. RESULTS: SN and SN + GSH significantly improved the glucose tolerance (p < 0.05), but GSH alone did not. The efficacy of combination treatment with SN and GSH in improving the glucose tolerance was higher than that of SN alone. Oral treatment with SN or SN + GSH significant reduced FBG and HbA1c levels (p < 0.05). Interestingly, daily oral administration of SN + GSH was more effective in reducing FBG and HbA1c levels than that of SN alone. Administration of SN or SN + GSH significantly increased plasma NOx levels (p < 0.05), and combination treatment with SN + GSH was more effective in increasing plasma NOx levels than that with SN alone. CONCLUSIONS: Combination treatment with SN and GSH is more effective in controlling hyperglycemia and increasing the plasma NOx levels in an experimental mouse model of diabetes. Since oral administration of GSH has been shown to be non-toxic in humans, the combination of SN and GSH may be important in potential future nitrite therapy.


Subject(s)
Diabetes Mellitus, Experimental , Glutathione , Hyperglycemia , Sodium Nitrite , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Glutathione/administration & dosage , Glycated Hemoglobin/metabolism , Hyperglycemia/drug therapy , Mice , Nitric Oxide/metabolism , Sodium Nitrite/administration & dosage , Streptozocin
3.
Biomed Pharmacother ; 145: 112435, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34798469

ABSTRACT

INTRODUCTION: Cold ischemia-reperfusion injury (IRI) is an inevitable event that increases post-transplant complications. We have previously demonstrated that supplementation of University of Wisconsin (UW) solution with non-FDA-approved hydrogen sulfide (H2S) donor molecules minimizes cold IRI and improves renal graft function after transplantation. The present study investigates whether an FDA-approved H2S donor molecule, sodium thiosulfate (STS), will have the same or superior effect in a clinically relevant rat model of syngeneic orthotopic kidney transplantation. METHOD: Thirty Lewis rats underwent bilateral nephrectomy followed by syngeneic orthotopic transplantation of the left kidney after 24-hour preservation in either UW or UW+STS solution at 4 °C. Rats were monitored to post-transplant day 14 and sacrificed to assess renal function (urine output, serum creatinine and blood urea nitrogen). Kidney sections were stained with H&E, TUNEL, CD68, and myeloperoxidase (MPO) to detect acute tubular necrosis (ATN), apoptosis, macrophage infiltration, and neutrophil infiltration. RESULT: UW+STS grafts showed significantly improved graft function immediately after transplantation, with improved recipient survival compared to UW grafts (p < 0.05). Histopathological examination revealed significantly reduced ATN, apoptosis, macrophage and neutrophil infiltration and downregulation of pro-inflammatory and pro-apoptotic genes in UW+STS grafts compared to UW grafts (p < 0.05). CONCLUSION: We show for the first time that preservation of renal grafts in STS-supplemented UW solution protects against prolonged cold IRI by suppressing apoptotic and inflammatory pathways, and thereby improving graft function and prolonging recipient survival. This could represent a novel clinically applicable therapeutic strategy to minimize the detrimental clinical outcome of prolonged cold IRI in kidney transplantation.


Subject(s)
Kidney Transplantation/methods , Organ Preservation Solutions/pharmacology , Reperfusion Injury/prevention & control , Thiosulfates/pharmacology , Adenosine/administration & dosage , Adenosine/pharmacology , Allopurinol/administration & dosage , Allopurinol/pharmacology , Animals , Apoptosis/physiology , Blood Urea Nitrogen , Cold Ischemia/adverse effects , Creatinine/blood , Glutathione/administration & dosage , Glutathione/pharmacology , Insulin/administration & dosage , Insulin/pharmacology , Kidney Function Tests , Male , Organ Preservation Solutions/administration & dosage , Raffinose/administration & dosage , Raffinose/pharmacology , Rats , Rats, Inbred Lew , Survival Rate , Thiosulfates/administration & dosage
4.
Molecules ; 26(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34946570

ABSTRACT

The study aimed to develop a new glutathione (GSH) oral formulation to enhance the delivery of GSH and counter the nephrotoxicity of the anticancer drug, cyclophosphamide (CP). A nanostructured lipid carrier glutathione formulation (GSH-NLCs) composed of glutathione (500 mg), stearic and oleic acid (300 mg, each), and Tween® 80 (2%, w/v) was prepared through the emulsification-solvent-evaporation technique, which exhibited a 452.4 ± 33.19 nm spheroidal-sized particulate material with narrow particle size distributions, -38.5 ± 1.4 mV zeta potential, and an entrapment efficiency of 79.8 ± 1.9%. The GSH formulation was orally delivered, and biologically tested to ameliorate the CP-induced renal toxicity in a rat model. Detailed renal morphology, before and after the GSH-NLCs administration, including the histopathological examinations, confirmed the ameliorating effects of the prepared glutathione formulation together with its safe oral delivery. CP-induced oxidative stress, superoxide dismutase depletion, elevation of malondialdehyde levels, depletion of Bcl-2 concentration levels, and upregulated NF-KB levels were observed and were controlled within the recommended and near normal/control levels. Additionally, the inflammatory mediator marker, IL-1ß, serum levels were marginally normalized by delivery of the GHS-NLCs formulation. Oral administration of the pure glutathione did not exhibit any ameliorating effects on the renal tissues, which suggested that the pure glutathione is reactive and is chemically transformed during the oral delivery, which affected its pharmacological action at the renal site. The protective effects of the GSH-NLCs formulation through its antioxidant and anti-inflammatory effects suggested its prominent role in containing CP-induced renal toxicity and renal tissue damage, together with the possibility of administrating higher doses of the anticancer drug, cyclophosphamide, to achieve higher and effective anticancer action in combination with the GSH-NLCs formulation.


Subject(s)
Glutathione/pharmacology , Kidney Diseases/drug therapy , Lipids/chemistry , Nanoparticles/chemistry , Protective Agents/pharmacology , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Cyclophosphamide , Drug Carriers/chemistry , Drug Compounding , Glutathione/administration & dosage , Glutathione/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Male , Micelles , Oxidative Stress/drug effects , Particle Size , Protective Agents/administration & dosage , Protective Agents/metabolism , Rats , Rats, Sprague-Dawley
5.
Sci Rep ; 11(1): 24114, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916571

ABSTRACT

Delivering bioactive compounds into skin tissue has long been a challenge. Using ex vivo porcine and rat skins, here we demonstrate that a detachable dissolvable microneedle (DDMN) array, a special dissolvable microneedle that allows needle detachment from the base within 2 min post administration, can effectively embed a model compound into epidermis and dermis. Diffusion of the compound from the needle embedding sites to the nearby skin tissue is demonstrated at various post administration periods. The relationship between the time that a conventional dissolvable microneedle array is left on skin without needle detachment from the base and the degree of skin surface abrasion at each microneedle penetration spot is also demonstrated on skin of human volunteers. Co-loading glutathione with vitamin C (vitC) can stabilize vitC in the DDMN. DDMN loaded with vitC and glutathione can help erasing post-acne-hyperpigmentation spots.


Subject(s)
Ascorbic Acid/administration & dosage , Drug Delivery Systems/methods , Glutathione/administration & dosage , Hyperpigmentation/drug therapy , Microinjections/methods , Needles , Animals , Ascorbic Acid/metabolism , Diffusion , Drug Stability , Epidermis/metabolism , Glutathione/metabolism , Humans , Injections, Intradermal , Rats , Skin Physiological Phenomena , Swine
6.
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
7.
Int J Mol Sci ; 22(16)2021 08 17.
Article in English | MEDLINE | ID: mdl-34445563

ABSTRACT

Choroid plexus (CP) sequesters cadmium and other metals, protecting the brain from these neurotoxins. These metals can induce cellular stress and modulate homeostatic functions of CP, such as solute transport. We previously showed in primary cultured neonatal rat CP epithelial cells (CPECs) that cadmium induced cellular stress and stimulated choline uptake at the apical membrane, which interfaces with cerebrospinal fluid in situ. Here, in CPECs, we characterized the roles of glutathione (GSH) and Zinc supplementation in the adaptive stress response to cadmium. Cadmium increased GSH and decreased the reduced GSH-to-oxidized GSH (GSSG) ratio. Heat shock protein-70 (Hsp70), heme oxygenase (HO-1), and metallothionein (Mt-1) were induced along with the catalytic and modifier subunits of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH synthesis. Inhibition of GCL by l-buthionine sulfoximine (BSO) enhanced stress protein induction and stimulation of choline uptake by cadmium. Zinc alone did not induce Hsp70, HO-1, or GCL subunits, or modulate choline uptake. Zinc supplementation during cadmium exposure attenuated stress protein induction and stimulation of choline uptake; this effect persisted despite inhibition of GSH synthesis. These data indicated up-regulation of GSH synthesis promotes adaptation to cadmium-induced cellular stress in CP, but Zinc may confer cytoprotection independent of GSH.


Subject(s)
Cadmium/toxicity , Choline/metabolism , Choroid Plexus/drug effects , Epithelium/drug effects , Glutathione/administration & dosage , Oxidative Stress/drug effects , Zinc/administration & dosage , Animals , Animals, Newborn , Choroid Plexus/metabolism , Choroid Plexus/pathology , Dietary Supplements , Epithelium/metabolism , Epithelium/pathology , Rats , Rats, Sprague-Dawley
8.
Biomol Concepts ; 12(1): 16-26, 2021 May 09.
Article in English | MEDLINE | ID: mdl-33966361

ABSTRACT

Tuberculosis (TB) caused by Mycobacterium tuberculosis (M. tb) still remains a devastating infectious disease in the world. There has been a daunting increase in the incidence of Type 2 Diabetes Mellitus (T2DM) worldwide. T2DM patients are three times more vulnerable to M. tb infection compared to healthy individuals. TB-T2DM coincidence is a challenge for global health control. Despite some progress in the research, M. tb still has unexplored characteristics in successfully evading host defenses. The lengthy duration of treatment, the emergence of multi-drug-resistant strains and extensive-drug-resistant strains of M. tb have made TB treatment very challenging. Previously, we have tested the antimycobacterial effects of everolimus within in vitro granulomas generated from immune cells derived from peripheral blood of healthy subjects. However, the effectiveness of everolimus treatment against mycobacterial infection in individuals with T2DM is unknown. Furthermore, the effectiveness of the combination of in vivo glutathione (GSH) supplementation in individuals with T2DM along with in vitro treatment of isolated immune cells with everolimus against mycobacterial infection has never been tested. Therefore, we postulated that liposomal glutathione (L-GSH) and everolimus would offer great hope for developing adjunctive therapy for mycobacterial infection. L-GSH or placebo was administered to T2DM individuals orally for three months. Study subjects' blood was drawn pre- and post-L-GSH/or placebo supplementation, where Peripheral Blood Mononuclear Cells (PBMCs) were isolated from whole blood to conduct in vitro studies with everolimus. We found that in vitro treatment with everolimus, an mTOR (membrane target of rapamycin) inhibitor, significantly reduced intracellular M. bovis BCG infection alone and in conjunction with L-GSH supplementation. Furthermore, we found L-GSH supplementation coupled with in vitro everolimus treatment produced a greater effect in inhibiting the growth of intracellular Mycobacterium bovis BCG, than with the everolimus treatment alone. We also demonstrated the functions of L-GSH along with in vitro everolimus treatment in modulating the levels of cytokines such as IFN-γ, TNF-α, and IL-2 and IL-6, in favor of improving control of the mycobacterial infection. In summary, in vitro everolimus-treatment alone and in combination with oral L-GSH supplementation for three months in individuals with T2DM, was able to increase the levels of T-helper type 1 (Th1) cytokines IFN-γ, TNF-α, and IL-2 as well as enhance the abilities of granulomas from individuals with T2DM to improve control of a mycobacterial infection.


Subject(s)
BCG Vaccine/administration & dosage , Diabetes Mellitus, Type 2/immunology , Everolimus/pharmacology , Glutathione/administration & dosage , Leukocytes, Mononuclear/immunology , Mycobacterium bovis/immunology , Tuberculosis/immunology , Administration, Oral , Adolescent , Adult , Aged , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Dietary Supplements , Double-Blind Method , Female , Granuloma/immunology , Humans , Immunity , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/microbiology , Male , Middle Aged , Mycobacterium bovis/drug effects , Mycobacterium bovis/metabolism , Tuberculosis/drug therapy , Tuberculosis/metabolism , Tuberculosis/microbiology , Young Adult
9.
Appl Physiol Nutr Metab ; 46(9): 1133-1142, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33740389

ABSTRACT

The effect of oral glutathione (GSH) supplementation was studied in obese subjects with and without type 2 diabetes (T2DM) on measures of glucose homeostasis and markers of oxidative stress. Twenty subjects (10 patients with T2DM and 10 obese subjects) were recruited for the study, and randomized in a double-blinded placebo-controlled manner to consume either 1000 mg GSH per day or placebo for 3 weeks. Before and after the 3 weeks insulin sensitivity was measured with the hyperinsulinemic-euglycemic clamp and a muscle biopsy was obtained to measure GSH and skeletal muscle mitochondrial hydrogen peroxide (H2O2) emission rate. Whole body insulin sensitivity increased significantly in the GSH group. Skeletal muscle GSH was numerically increased (∼19%) in the GSH group; no change was seen in GSH to glutathione disulfide ratio. Skeletal muscle mitochondrial H2O2 emission rate did not change in response to the intervention and neither did the urinary excretion of the RNA oxidation product 8-oxo-7,8-dihydroguanosine or the DNA oxidation product 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), although 8-oxodG decreased as a main effect of time. Oral GSH supplementation improves insulin sensitivity in obese subjects with and without T2DM, although it does not alter markers of oxidative stress. The study has been registered in clinicaltrials.gov (NCT02948673). Novelty: Reduced glutathione supplementation increases insulin sensitivity in obese subjects with and without T2DM. H2O2 emission rate from skeletal muscle mitochondria was not affected by GSH supplementation.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Dietary Supplements , Glutathione/administration & dosage , Insulin Resistance/physiology , Obesity/physiopathology , Administration, Oral , Biomarkers/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Dietary Supplements/adverse effects , Glucose Tolerance Test , Glutathione/adverse effects , Glutathione/blood , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Hydrogen Peroxide/metabolism , Middle Aged , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Oxidative Stress , Oxygen Consumption
10.
J Mater Chem B ; 9(12): 2866-2876, 2021 03 28.
Article in English | MEDLINE | ID: mdl-33720270

ABSTRACT

The human sodium iodide symporter (hNIS) can be linked to the downstream of radiation-sensitive early growth response protein1 (Egr1) promoter, and activated by the Egr1 following 131I treatment. However, the rapid outflow of 131I restricted the radiotherapy effect. To overcome this barrier, ultrasmall gold nanoclusters (usAuNCs) were used to enhance the radiotherapy efficacy of Egr1-hNIS for its radiation sensitization. In this work, we prepared "cell bomb" BMSCs carrying both GSH@AuNCs and Egr1-hNIS. We found that the "cell bomb" can target TNBC tumor and reach a maximum 131I concentration 9 h following 131I injection. Colony formation assay revealed that 131I, 131I combined with GSH@AuNCs could independently inhibit 39.5% and 66.4% of cell growth, respectively. Moreover, in vivo131I therapy further demonstrated that the growth of triple negative breast cancer (TNBC) was controlled by BMSC-Egr1-hNIS + AuNCs group, with relative volume inhibition percentages of 56.16% (compared with the control group) and 36.20% (compared with the BMSC-Egr1-hNIS group), respectively. To summarize, we successfully prepared BMSC-Egr1-hNIS carrying GSH@AuNCs to target TNBC which could synergistically improve the efficacy of hNIS gene therapy.


Subject(s)
Biocompatible Materials/metabolism , Breast Neoplasms/radiotherapy , Gold/metabolism , Mesenchymal Stem Cells/metabolism , Metal Nanoparticles/chemistry , Symporters/metabolism , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Glutathione/administration & dosage , Glutathione/chemistry , Glutathione/metabolism , Gold/administration & dosage , Gold/chemistry , Injections, Intravenous , Iodine Radioisotopes , Metal Nanoparticles/administration & dosage , Mice , Particle Size , Promoter Regions, Genetic/genetics , Symporters/genetics
12.
Curr Eye Res ; 46(4): 470-481, 2021 04.
Article in English | MEDLINE | ID: mdl-32838548

ABSTRACT

PURPOSE: Tissues in the eye are particularly susceptible to oxidative damage due to light exposure. While vitamin C (ascorbic acid) has been noted as a vital antioxidant in the vitreous humor, its physiological concentration (1-2 mM) has been shown to be toxic to retinal and lens epithelial cells in in vitro cell culture. We have explored adding vitamin C to hydrogel vitreous substitutes as a potential therapeutic to prevent oxidative damage to intraocular tissues after vitrectomy. However, vitamin C degrades rapidly even when loaded at high concentrations, limiting its long-term effectiveness. Glutathione, another antioxidant found abundantly in the lens at concentrations of 2-10 mM, was proposed to be used in conjunction with vitamin C. METHODS: Cell viability and reactive oxygen species activity of human retinal and lens epithelial cells treated with various combinations of vitamin C, glutathione, hydrogen peroxide, and a hydrogel vitreous substitute were determined using CellTiter-Glo luminescent cell viability assay and dichlorofluorescein assay, respectively. The vitamin C remaining in hydrogel vitreous substitute or glutathione-vitamin C solutions was determined using a microplate reader at 265 nm wavelength, compared against standard solutions with known concentrations. RESULTS: Glutathione protected the lens and retinal cells from the negative effect of vitamin C on cell viability and prolonged the antioxidant effect of vitamin C in vitro. While the detected reading of pure vitamin C solution decreased rapidly from 100% to 10% by 3 days, glutathione provided a significant extension to vitamin C stability, with 70% remaining after 14 days when the glutathione was used at physiological concentrations found in the lens (2-10 mM). CONCLUSIONS: These results indicate glutathione might be an effective addition to vitamin C in intraocular implants, including potential vitreous substitutes, and warrants additional studies on the effectiveness of the vitamin C - glutathione combination in preventing oxidative stress post-vitrectomy.


Subject(s)
Antioxidants/administration & dosage , Ascorbic Acid/administration & dosage , Epithelial Cells/drug effects , Glutathione/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Lens, Crystalline/cytology , Retinal Pigment Epithelium/drug effects , Biomimetic Materials/chemistry , Cell Survival , Cells, Cultured , Drug Carriers , Drug Combinations , Epithelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Vitreous Body
13.
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
14.
J Dev Orig Health Dis ; 12(3): 484-495, 2021 06.
Article in English | MEDLINE | ID: mdl-32741420

ABSTRACT

Extremely premature birth is associated with a permanent disruption of energy metabolism. The underlying mechanisms are poorly understood. The oxidative stress induced by parenteral nutrition (PN) during the first week of life is suspected to reprogram energy metabolism in the liver. Full-term male Hartley guinea pigs (to isolate PN from prematurity) receiving PN enriched or not with glutathione (to isolate PN effects from PN-induced oxidative stress effects) or an Oral Nutrition (ON) during the first week of life were used. At 1 week (neonatal) and 16 weeks (adult), measurements of liver glutathione (GSH and GSSG) and activities of three key enzymes of energy metabolism (glucokinase (GCK), phosphofructokinase (PFK), and acetyl-CoA carboxylase (ACC)) were performed. Differences between groups were reported if p ≤ 0.05 (Analysis of Variance). At 1 week, compared to ON, PN induced higher GSSG (oxidative stress), higher GCK activity, and lower PFK and ACC activity, the glutathione supplement prevented all PN effects. At 16 weeks, early PN induced lower GSSG (reductive stress) and lower GCK activity, which was prevented by added glutathione, and higher ACC activity independent of glutathione supplement. ACC was negatively associated (r2 = 0.33) with GSSG. Increased nicotinamide adenine dinucleotide phosphate levels confirmed the glucose-6-phosphate accumulation at 1 week, whereas our protocol failed to document lipid accumulation at 16 weeks. In adult male guinea pigs, neonatal exposure to PN affected glutathione metabolism leading to reductive stress (lower GSSG) and an altered metabolic flow of glucose. Partial prevention with glutathione supplementation suggests that, in addition to peroxides, other factors of PN are involved.


Subject(s)
Energy Metabolism , Liver/enzymology , Oxidative Stress , Parenteral Nutrition/adverse effects , Premature Birth , Animals , Animals, Newborn , Glucose/metabolism , Glutathione/administration & dosage , Guinea Pigs , Male
15.
J Mater Chem B ; 9(1): 187-194, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33237120

ABSTRACT

In this study, an enhanced anticancer strategy combining the chemotherapy from antineoplastics with the oxidative damage from a sulfur dioxide (SO2) prodrug is presented. Based on the characteristics of a high glutathione (GSH) level in the tumor microenvironment, a novel GSH-responsive SO2 polymeric prodrug mPEG-b-P(PA-alt-GDNs) was designed and synthesized via a ring-opening alternating copolymerization and "click" reaction. The GSH-sensitive mechanism of the polymer was investigated in detail. Furthermore, Irinotecan was loaded into the polymeric prodrug nanoparticles by a self-assembly method with a drug loading content of 12.3 wt% and a loading efficiency of 42.2%. The drug-loaded nanoparticles showed a sensitive response to high concentrations of GSH in the tumor cells and rapidly released both Irinotecan and SO2. The depletion of GSH and the release of SO2 were supposed to increase the level of reactive oxygen species in the tumor cell, which, in combination with the released Irinotecan, exerted an enhanced anti-proliferative effect against HepG2 cells. Finally, Irinotecan-loaded nanoparticles exhibited a stronger antitumor effect than free antineoplastics in HepG2 cells. Thus, these results indicated that our polymeric prodrug SO2 is a promising candidate for chemotherapeutic drug delivery and would be a new weapon in anticancer treatment.


Subject(s)
Drug Delivery Systems/methods , Glutathione/chemical synthesis , Irinotecan/chemical synthesis , Polyethylene Glycols/chemical synthesis , Prodrugs/chemical synthesis , Sulfur Dioxide/chemical synthesis , Dose-Response Relationship, Drug , Glutathione/administration & dosage , Glutathione/metabolism , Hep G2 Cells , Humans , Irinotecan/administration & dosage , Irinotecan/metabolism , Nuclear Magnetic Resonance, Biomolecular/methods , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/metabolism , Polymers/administration & dosage , Polymers/chemical synthesis , Polymers/metabolism , Prodrugs/administration & dosage , Prodrugs/metabolism , Sulfur Dioxide/administration & dosage , Sulfur Dioxide/metabolism
16.
J Pediatr Gastroenterol Nutr ; 71(6): 771-777, 2020 12.
Article in English | MEDLINE | ID: mdl-32960827

ABSTRACT

OBJECTIVES: The nutritional status of children with cystic fibrosis (CF) is associated with mortality and morbidity. Intestinal inflammation may contribute to impaired digestion, absorption, and nutrient utilization in patients with CF and oral glutathione may reduce inflammation, promoting improved nutritional status in patients with CF. METHODS: The GROW study was a prospective, multicenter, randomized, placebo-controlled, double-blind, phase II clinical trial in pancreatic insufficient patients with CF between the ages of 2 and 10 years. Patients received reduced glutathione or placebo orally daily for 24 weeks. The primary endpoint was the difference in change in weight-for-age z-scores from baseline through week 24 between treatment groups. Secondary endpoints included other anthropometrics, serum, and fecal inflammatory markers in addition to other clinical outcomes. RESULTS: Fifty-eight participants completed the study. No significant differences were seen between glutathione (n = 30) and placebo (n = 28) groups in the 6-month change in weight-for-age z-score (-0.08; 95% CI: -0.22 to 0.06; P = 0.25); absolute change in weight (kg) (-0.18; 95% CI: -0.55 to 0.20; P = 0.35); or absolute change in BMI kg/m (-0.06; 95% CI: -0.37 to 0.25; P = 0.69). There were no significant differences in other secondary endpoints. Overall, glutathione was safe and well tolerated. CONCLUSIONS: Oral glutathione supplementation did not impact growth or change serum or fecal inflammatory markers in pancreatic insufficient children with CF when compared with placebo.


Subject(s)
Cystic Fibrosis , Exocrine Pancreatic Insufficiency , Glutathione , Child , Child, Preschool , Cystic Fibrosis/drug therapy , Double-Blind Method , Exocrine Pancreatic Insufficiency/drug therapy , Exocrine Pancreatic Insufficiency/etiology , Glutathione/administration & dosage , Humans , Prospective Studies
17.
Arq. bras. med. vet. zootec. (Online) ; 72(4): 1321-1328, July-Aug. 2020. ilus
Article in English | LILACS, VETINDEX | ID: biblio-1131480

ABSTRACT

Fifteen New Zealand adult rabbits were randomly allocated into three groups: Sham-operated (group A), Ischemia and Reperfusion (group B) and Carolina Rinse Solution (CRS) (group C). Groups B and C were subjected to one hour of ischemia and two hours of reperfusion. In group C, ten minutes before reperfusion, the bowel lumen was filled with CRS, and the segment immersed in CRS. Necrosis and loss of integrity of the villi were visible in groups B and C. Edema of the submucosa and circular muscle was observed in all groups. Hemorrhage was observed in different layers for groups B and C, but group C showed more severe hemorrhage in different layers during reperfusion. All groups showed polymorphonuclear leukocyte infiltration on the base of the mucosa, submucosa, and longitudinal muscle, in addition to polymorphonuclear leukocytes margination in the mucosal and submucosal vessels. Necrosis of enterocytes, muscles, crypts of Lieberkühn and myenteric plexus was observed in groups B and C during reperfusion. Topical and intraluminal Carolina Rinse Solution did not attenuate the effects of ischemia and reperfusion in the small intestine of rabbits.(AU)


Quinze coelhos da raça Nova Zelândia foram alocados em três grupos: instrumentado (grupo A), isquemia e reperfusão (grupo B) e solução de Carolina rinse (CRS) (grupo C). Os grupos B e C foram submetidos a uma hora de isquemia e a duas horas de reperfusão. No grupo C, 10 minutos antes da reperfusão, o segmento isolado foi imerso e teve seu lúmen preenchido com CRS. Os grupos B e C apresentaram necrose e perda progressiva da integridade das vilosidades. Foi observado edema na submucosa e na camada muscular circular em todos os grupos. Nos grupos B e C, foi observada hemorragia em diferentes camadas, mas, no grupo C, a hemorragia foi mais intensa durante a reperfusão. Todos os grupos apresentaram infiltrado de PMN na base da mucosa, na submucosa e na camada muscular longitudinal e marginação de PMN nos vasos da mucosa e da submucosa. Durante a reperfusão, foi observada necrose dos enterócitos, das camadas musculares, das criptas de Lieberkühn e do plexo mioentérico nos grupos B e C. O uso tópico e intraluminal de CRS não atenuou os efeitos da isquemia e da reperfusão no intestino delgado de coelhos.(AU)


Subject(s)
Animals , Rabbits , Reperfusion/veterinary , Allopurinol/administration & dosage , Deferoxamine/administration & dosage , Glutathione/administration & dosage , Ischemia/veterinary , Jejunum/surgery
18.
Nutrients ; 12(7)2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32635181

ABSTRACT

Vitamin B-6 and glutathione (GSH) are antioxidant nutrients, and inadequate vitamin B-6 may indirectly limit glutathione synthesis and further affect the antioxidant capacities. Since liver cirrhosis is often associated with increased oxidative stress and decreased antioxidant capacities, we conducted a double-blind randomized controlled trial to assess the antioxidative effect of vitamin B-6, GSH, or vitamin B-6/GSH combined supplementation in cirrhotic patients. We followed patients after the end of supplementation to evaluate the association of vitamin B-6 and GSH with disease severity. In total, 61 liver cirrhosis patients were randomly assigned to placebo, vitamin B-6 (50 mg pyridoxine/d), GSH (500 mg/d), or B-6 + GSH groups for 12 weeks. After the end of supplementation, the condition of patient's disease severity was followed until the end of the study. Neither vitamin B-6 nor GSH supplementation had significant effects on indicators of oxidative stress and antioxidant capacities. The median follow-up time was 984 d, and 21 patients were lost to follow-up. High levels of GSH, a high GSH/oxidized GSH ratio, and high GSH-St activity at baseline (Week 0) had a significant effect on low Child-Turcotte-Pugh scores at Week 0, the end of supplementation (Week 12), and the end of follow-up in all patients after adjusting for potential confounders. Although the decreased GSH and its related enzyme activity were associated with the severity of liver cirrhosis, vitamin B-6 and GSH supplementation had no significant effect on reducing oxidative stress and increasing antioxidant capacities.


Subject(s)
Antioxidants/administration & dosage , Dietary Supplements , Glutathione/administration & dosage , Liver Cirrhosis/therapy , Vitamin B 6/administration & dosage , Double-Blind Method , Female , Follow-Up Studies , Humans , Liver Cirrhosis/physiopathology , Male , Middle Aged , Oxidative Stress/drug effects , Severity of Illness Index , Treatment Outcome
19.
Chem Biol Interact ; 328: 109195, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32707044

ABSTRACT

A previous study demonstrated that glutathione (GSH) produces specific antidepressant-like effect in the forced swimming test (FST), a predictive test of antidepressant activity. The present study investigated the involvement of multiple cellular targets implicated in the antidepressant-like effect of GSH in the FST. The antidepressant-like effect of GSH (300 nmol/site, icv) lasted up to 3 h when mice were submitted to FST. The central administration of oxidized GSH (GSSG, 3-300 nmol/site) did not alter the behavior of mice submitted to the FST. Furthermore, the combined treatment of sub-effective doses of GSH (100 nmol/site, icv) with a sub-effective dose of classical antidepressants (fluoxetine 10 mg/kg, and imipramine 5 mg/kg, ip) presented synergistic effect by decreasing the immobility time in the FST. The antidepressant-like effect of GSH was abolished by prazosin (1 mg/kg, ip, α1-adrenoceptor antagonist), baclofen (1 mg/kg, ip, GABAB receptor agonist), bicuculline (1 mg/kg, ip, GABAA receptor antagonist), l-arginine (750 mg/kg, ip, NO precursor), SNAP (25 µg/site, icv, NO donor), but not by yohimbine (1 mg/kg, ip, α2-adrenoceptor antagonist). The NMDA receptor antagonists, MK-801(0.001 mg/kg, ip) or GMP (0.5 mg/kg, ip), potentiated the effect of a sub-effective dose of GSH in the FST. These results suggest that the antidepressant-like effect induced by GSH is connected to the activation of α1 adrenergic and GABAA receptors, as well as the inhibition of GABAB and NMDA receptors and NO biosyntesis. We speculate that redox-mediated signaling on the extracelular portion of cell membrane receptors would be a common mechanism of action of GSH.


Subject(s)
Antidepressive Agents/pharmacology , Glutathione/pharmacology , Molecular Targeted Therapy , Adrenergic Antagonists/pharmacology , Animals , Arginine/pharmacology , Drug Synergism , Female , Glutathione/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Immobilization , Male , Mice , Receptors, Adrenergic/metabolism , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...