Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37.909
Filter
1.
Food Res Int ; 188: 114433, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823827

ABSTRACT

Whey derived peptides have shown potential activity improving brain function in pathological condition. However, there is little information about their mechanism of action on glial cells, which have important immune functions in brain. Astrocytes and microglia are essential in inflammatory and oxidative defense that take place in neurodegenerative disease. In this work we evaluate antioxidant and anti-inflammatory potential bioactivity of whey peptide in glial cells. Peptides were formed during simulated gastrointestinal digestion (Infogest protocol), and low molecular weight (<5kDA) peptides (WPHf) attenuated reactive oxygen species (ROS) production induced by hydrogen peroxide stimulus in both cells in dose-dependent manner. WPHf induced an increase in the antioxidant glutathione (GSH) content and prevented GSH reduction induced by lipopolysaccharides (LPS) stimulus in astrocytes cells in a cell specific form. An increase in cytokine mRNA expression (TNFα and IL6) and nitric oxide secretion induced by LPS was attenuated by WPHf pre-treatment in both cells. The inflammatory pathway was dependent on NFκB activation. Bioactive peptide ranking analysis showed positive correlation with hydrophobicity and negative correlation with high molecular weights. The sequence identification revealed 19 peptides cross-referred with bioactive database. Whey peptides were rich in leucine, valine and tyrosine in the C-terminal region and lysine in the N-terminal region. The anti-inflammatory and antioxidant potential of whey peptides were assessed in glia cells and its mechanisms of action were related, such as modulation of antioxidant enzymes and anti-inflammatory pathways. Features of the peptide structure, such as molecular size, hydrophobicity and types of amino acids present in the terminal region are associated to bioactivity.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Neuroglia , Whey Proteins , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Whey Proteins/pharmacology , Whey Proteins/chemistry , Whey Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Animals , Reactive Oxygen Species/metabolism , Lipopolysaccharides/pharmacology , Glutathione/metabolism , Peptides/pharmacology , Nitric Oxide/metabolism , Astrocytes/drug effects , Astrocytes/metabolism
2.
Proc Natl Acad Sci U S A ; 121(24): e2404668121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833473

ABSTRACT

Developing anticancer drugs with low side effects is an ongoing challenge. Immunogenic cell death (ICD) has received extensive attention as a potential synergistic modality for cancer immunotherapy. However, only a limited set of drugs or treatment modalities can trigger an ICD response and none of them have cytotoxic selectivity. This provides an incentive to explore strategies that might provide more effective ICD inducers free of adverse side effects. Here, we report a metal-based complex (Cu-1) that disrupts cellular redox homeostasis and effectively stimulates an antitumor immune response with high cytotoxic specificity. Upon entering tumor cells, this Cu(II) complex enhances the production of intracellular radical oxidative species while concurrently depleting glutathione (GSH). As the result of heightening cellular oxidative stress, Cu-1 gives rise to a relatively high cytotoxicity to cancer cells, whereas normal cells with low levels of GSH are relatively unaffected. The present Cu(II) complex initiates a potent ferroptosis-dependent ICD response and effectively inhibits in vivo tumor growth in an animal model (c57BL/6 mice challenged with colorectal cancer). This study presents a strategy to develop metal-based drugs that could synergistically potentiate cytotoxic selectivity and promote apoptosis-independent ICD responses through perturbations in redox homeostasis.


Subject(s)
Copper , Glutathione , Homeostasis , Oxidation-Reduction , Animals , Mice , Humans , Glutathione/metabolism , Mice, Inbred C57BL , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Oxidative Stress/drug effects , Drug Synergism , Immunogenic Cell Death/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism
3.
Nat Commun ; 15(1): 4751, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834573

ABSTRACT

Intracellular potassium (K+) homeostasis is fundamental to cell viability. In addition to channels, K+ levels are maintained by various ion transporters. One major family is the proton-driven K+ efflux transporters, which in gram-negative bacteria is important for detoxification and in plants is critical for efficient photosynthesis and growth. Despite their importance, the structure and molecular basis for K+-selectivity is poorly understood. Here, we report ~3.1 Å resolution cryo-EM structures of the Escherichia coli glutathione (GSH)-gated K+ efflux transporter KefC in complex with AMP, AMP/GSH and an ion-binding variant. KefC forms a homodimer similar to the inward-facing conformation of Na+/H+ antiporter NapA. By structural assignment of a coordinated K+ ion, MD simulations, and SSM-based electrophysiology, we demonstrate how ion-binding in KefC is adapted for binding a dehydrated K+ ion. KefC harbors C-terminal regulator of K+ conductance (RCK) domains, as present in some bacterial K+-ion channels. The domain-swapped helices in the RCK domains bind AMP and GSH and they inhibit transport by directly interacting with the ion-transporter module. Taken together, we propose that KefC is activated by detachment of the RCK domains and that ion selectivity exploits the biophysical properties likewise adapted by K+-ion-channels.


Subject(s)
Cryoelectron Microscopy , Escherichia coli Proteins , Escherichia coli , Potassium , Potassium/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Potassium-Hydrogen Antiporters/metabolism , Potassium-Hydrogen Antiporters/chemistry , Potassium-Hydrogen Antiporters/genetics , Molecular Dynamics Simulation , Glutathione/metabolism , Protein Domains
4.
PeerJ ; 12: e17541, 2024.
Article in English | MEDLINE | ID: mdl-38832034

ABSTRACT

Introduction: Oxidative and antioxidant pathways play essential roles in the development of alcohol-induced brain injury. The Nrf2 pathway is an endogenous antioxidant response pathway, but there has been little research on the role of Nrf2 in alcohol-related diseases. Thus, we examined the effects of alcohol and an Nrf2 agonist (TBHQ) on astrocyte function, mRNA expression, and metabolite content to further explore the protective mechanisms of Nrf2 agonists in astrocytes following alcohol exposure. Methods: CTX TNA2 astrocytes were cultured with alcohol and TBHQ and then subjected to transcriptome sequencing, LC-MS/MS analysis, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and malondialdehyde (MDA) and superoxide dismutase (SOD) activity assays. Results: Alcohol exposure significantly increased malondialdehyde (MDA) levels while decreasing superoxide dismutase (SOD) levels in astrocytes. Treatment with TBHQ effectively reversed these effects, demonstrating its protective role against oxidative stress induced by alcohol. Transcriptome sequencing and qRT-PCR analysis revealed that TBHQ specifically upregulates genes involved in glutathione metabolism, including a notable increase in the expression of the glutathione S-transferase A5 (GSTA5) gene, which was suppressed by alcohol exposure. Additionally, metabolomic analysis showed that TBHQ regulates key components of ether lipid metabolism in alcohol-exposed astrocytes, with significant reductions in the levels of lysophosphatidylcholine (18:0) (LysoPC (18:0)) and 2-acetyl-1-alkyl-sn-glycero-3-phosphocholine, both of which are critical markers in the ether lipid metabolic pathway. Discussion: The findings underscore the role of TBHQ as an Nrf2 agonist in mitigating alcohol-induced oxidative damage in astrocytes by modulating glutathione metabolism and ether lipid metabolism. The regulation of GSTA5 gene expression emerges as a key mechanism through which Nrf2 agonists confer neuroprotection against oxidative stress and lipid oxidation. These insights pave the way for potential therapeutic strategies targeting the Nrf2 pathway to protect astrocytes from alcohol-induced damage.


Subject(s)
Astrocytes , Ethanol , Glutathione , NF-E2-Related Factor 2 , Oxidative Stress , Astrocytes/drug effects , Astrocytes/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Animals , Ethanol/pharmacology , Oxidative Stress/drug effects , Glutathione/metabolism , Hydroquinones/pharmacology , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Malondialdehyde/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Cells, Cultured
5.
Sci Rep ; 14(1): 10835, 2024 05 12.
Article in English | MEDLINE | ID: mdl-38736022

ABSTRACT

Research on the relationships between oligoelements (OE) and the development of cancer or its prevention is a field that is gaining increasing relevance. The aim was to evaluate OE and their interactions with oncology treatments (cytarabine or etoposide) to determine the effects of this combination on biogenic amines and oxidative stress biomarkers in the brain regions of young Wistar rats. Dopamine (DA), 5-Hydroxyindoleacetic acid (5-Hiaa), Glutathione (Gsh), Tiobarbituric acid reactive substances (TBARS) and Ca+2, Mg+2 ATPase enzyme activity were measured in brain regions tissues using spectrophometric and fluorometric methods previously validated. The combination of oligoelements and cytarabine increased dopamine in the striatum but decreased it in cerebellum/medulla-oblongata, whereas the combination of oligoelements and etoposide reduced lipid peroxidation. These results suggest that supplementation with oligoelements modifies the effects of cytarabine and etoposide by redox pathways, and may become promising therapeutic targets in patients with cancer.


Subject(s)
Brain , Cytarabine , Dopamine , Etoposide , Oxidative Stress , Rats, Wistar , Animals , Etoposide/pharmacology , Oxidative Stress/drug effects , Cytarabine/pharmacology , Dopamine/metabolism , Rats , Brain/metabolism , Brain/drug effects , Male , Lipid Peroxidation/drug effects , Dietary Supplements , Glutathione/metabolism
6.
ACS Appl Mater Interfaces ; 16(20): 25788-25798, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38716694

ABSTRACT

Phototherapy, represented by photodynamic therapy (PDT) and photothermal therapy (PTT), has great potential in tumor treatment. However, the presence of antioxidant glutathione (GSH) and the heat shock proteins (HSPs) expression caused by high temperature can weaken the effects of PDT and PTT. Here, a multifunctional nanocomplex BT&GA@CL is constructed to realize enhanced synergistic PDT/PTT. Cinnamaldehyde liposomes (CLs) formed by cinnamaldehyde dimer self-assembly were loaded with in gambogic acid (GA) and an aggregation-induced emission molecule BT to obtain BT&GA@CL. As a drug carrier, CL can consume glutathione (GSH) and release drugs responsively. The released BT aggregates can simultaneously act as both a photothermal agent and photosensitizer to achieve PDT and PTT under 660 nm laser irradiation. Specifically, GA as an HSP90 inhibitor can attenuate PTT-induced HSP90 protein expression, thereby weakening the tolerance of tumor cells to high temperatures and enhancing PTT. Such a multifunctional nanocomplex simultaneously modulates the content of GSH and HSP90 in tumor cells, thus enhancing both PDT and PTT, ultimately achieving the goal of efficient combined tumor suppression.


Subject(s)
Glutathione , Liposomes , Photochemotherapy , Photosensitizing Agents , Xanthones , Liposomes/chemistry , Glutathione/metabolism , Glutathione/chemistry , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Xanthones/chemistry , Xanthones/pharmacology , Animals , Mice , Photothermal Therapy , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
7.
PLoS One ; 19(5): e0304290, 2024.
Article in English | MEDLINE | ID: mdl-38787841

ABSTRACT

The aim of the study was to assess the impact of solarium light therapy on selected biological and biochemical parameters of peripheral blood in recreational horses. The study involved 10 horses divided into two groups of young (aged 5 to 7 years) and old (aged 14 to 19 years) individuals. All animals participated in light therapy sessions every other day. Blood was sampled three times during the study: before the treatment, after five light sessions, and after ten light sessions. Morphological parameters, the activity of antioxidant enzymes, TAS values, and the levels of glutathione (GSH), vitamin D3, vitamin C, and malondialdehyde (MDA) were measured in the whole blood. Light therapy contributed to an increase in MCV, HDW, MCVr, CHr and MPV indices, and simultaneously a decrease in the basophil counts, MCHC, RDW and CHCMr indices in both groups of horses (p ≤ 0.05). At the same time reticulocytes fell in older whereas white blood cells and monocytes counts expanded in younger individuals. The treatment also increased the activity of glutathione reductase (GR) and glutathione peroxidase (GPx) in young but decreased the activity of mentioned enzymes in blood plasma of old horses. The total antioxidant status (TAS) of the blood plasma rose progressively, whereas GSH levels declined in all individuals. Moreover, vitamin D3 levels did not change, whereas vitamin C levels gradually decreased during the experiment. The therapy also helped to reduce levels of MDA in the blood plasma, especially of older horses (p ≤ 0.05). In turn, GPx and GR activities as well as MDA levels significantly declined, whereas GSH levels notably elevated in erythrocytes (p ≤ 0.05). Solarium light therapy appears to have a beneficial impact on the morphological parameters and antioxidant status of blood in recreational horses in the winter season. However, the observed results could in part be attributed to the natural physiological adaptation of each individual organism to the treatment.


Subject(s)
Antioxidants , Animals , Horses/blood , Antioxidants/metabolism , Glutathione/blood , Glutathione/metabolism , Phototherapy/methods , Malondialdehyde/blood , Ascorbic Acid/blood , Male , Female , Glutathione Reductase/blood , Glutathione Reductase/metabolism , Glutathione Peroxidase/blood , Glutathione Peroxidase/metabolism , Cholecalciferol/blood , Aging/blood
8.
Spectrochim Acta A Mol Biomol Spectrosc ; 317: 124446, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38759396

ABSTRACT

Developing the efficient nanozymes for reactive oxygen species (ROS)-mediated highly potent tumor catalytic therapy has become a great challenge. In this study, we prepared the DNA-Fe, -FeAg, and -FeCuAg nanocluster (NCs) using the G-/C-rich single-stranded DNA (ssDNA) templates. The steady-state kinetic and the catalytic performances and mechanisms of DNA-metal NCs were first systematically investigated. The results indicated that c-kit-TBA-Fe, c-kit-TBA-FeAg, and c-kit-TBA-FeCuAg NCs exhibited the high peroxidase-like activity. All of three types of NCs presented the higher affinity to the substrate TMB and better storage stability at 4 °C than horseradish peroxidase (HRP). Moreover, c-kit-TBA-FeAg and c-kit-TBA-FeCuAg NCs presented the 6.7- and 4.7-fold stronger affinity to TMB than c-kit-TBA-Fe, respectively. However, the maximum reaction rate (Vmax) of c-kit-TBA-FeCuAg NCs with H2O2 was the largest, which promoted the generation of much more •OH in the reaction system. More importantly, c-kit-TBA-FeCuAg NCs were able to deplete largely the intracellular GSH and thus generate lots of endogenous ROS in HeLa cells, thereby exhibiting the significant and specific in vitro cancer cells toxicity. Therefore, c-kit-TBA-FeCuAg NCs, with peroxidase-like activity and glutathione (GSH) consumption ability, hold the ROS-based promising therapeutic effects for cancer.


Subject(s)
Glutathione , Humans , Glutathione/metabolism , Silver/chemistry , Silver/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , DNA/metabolism , DNA/chemistry , Gold/chemistry , Iron/chemistry , Iron/metabolism , Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Kinetics , HeLa Cells , Hydrogen Peroxide/pharmacology , Cell Line, Tumor
9.
Biomed Mater ; 19(4)2024 May 30.
Article in English | MEDLINE | ID: mdl-38772383

ABSTRACT

The traditional chemotherapeutic agents' disadvantages such as high toxicity, untargeting and poor water solubility lead to disappointing chemotherapy effects, which restricts its clinical application. In this work, novel size-appropriate and glutathione (GSH)-responsive nano-hydrogels were successfully prepared via the active ester method between chitosan (containing -NH2) and cross-linker (containing NHS). Especially, the cross-linker was elaborately designed to possess a disulfide linkage (SS) as well as two terminal NHS groups, namely NHS-SS-NHS. These functionalities endowed chitosan-based cross-linked scaffolds with capabilities for drug loading and delivery, as well as a GSH-responsive mechanism for drug release. The prepared nano-hydrogels demonstrated excellent performance applicable morphology, excellent drug loading efficiency (∼22.5%), suitable size (∼100 nm) and long-term stability. The prepared nano-hydrogels released over 80% doxorubicin (DOX) after incubation in 10 mM GSH while a minimal DOX release less than 25% was tested in normal physiological buffer (pH = 7.4). The unloaded nano-hydrogels did not show any apparent cytotoxicity to A 549 cells. In contrast, DOX-loaded nano-hydrogels exhibited marked anti-tumor activity against A 549 cells, especially in high GSH environment. Finally, through fluorescent imaging and flow cytometry analysis, fluorescein isothiocyanate-labeled nano-hydrogels show obvious specific binding to the GSH high-expressing A549 cells and nonspecific binding to the GSH low-expressing A549 cells. Therefore, with this cross-linking approach, our present finding suggests that cross-linked chitosan nano-hydrogel drug carrier improves the anti-tumor effect of the A 549 cells and may serve as a potential injectable delivery carrier.


Subject(s)
Antineoplastic Agents , Chitosan , Cross-Linking Reagents , Doxorubicin , Glutathione , Hydrogels , Chitosan/chemistry , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Glutathione/chemistry , Glutathione/metabolism , Hydrogels/chemistry , Cross-Linking Reagents/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Drug Liberation , Cell Line, Tumor , A549 Cells , Drug Carriers/chemistry , Drug Delivery Systems , Disulfides/chemistry , Delayed-Action Preparations/chemistry
10.
Med Sci Monit ; 30: e944335, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38783538

ABSTRACT

BACKGROUND Either a reduction in antioxidant levels or an accumulation of reactive oxygen species can heighten susceptibility to oxidative damage in disc cells. To date, no research has investigated the levels of lipid peroxidation products (thiobarbituric acid reactive substances [TBARs]), reduced glutathione (GSH), and glutathione peroxidase (GPx) in excised human lumbar disc tissues affected by degenerative disease. Therefore, this study aimed to evaluate lipid peroxidation products in excised disc tissues from patients with degenerative disc disease. MATERIAL AND METHODS Forty-two patients were enrolled. Patients were divided into lumbar disc degeneration (LDD) and nonlumbar disc degeneration (nonLDD) groups according to Pfirrmann classification. Intervertebral discs were obtained from all patients during the operation and were homogenized for analysis. TBARs levels were measured using fluorometry. GSH levels and GPx activity were quantified spectrophotometrically using a kinetic method. RESULTS TBARs levels in excised discs from LDD patients (5.18±4.14) were significantly higher than those from nonLDD patients (2.56±1.23, P=0.008). The levels of TBARs tended to increase with the severity of degeneration according to the Pfirrmann classification. However, these 2 groups showed no significant differences in reduced glutathione levels or glutathione peroxidase activity (P>0.05). Patients with LDD exhibited a worse health-related quality of life, reflected in lower utility and EQ-VAS scores and higher Oswestry disability index scores. CONCLUSIONS There was a notable increase in lipid peroxidation products in the excised intervertebral discs of patients with LDD. This finding suggests that oxidative stress may contribute to the development of disc degeneration.


Subject(s)
Glutathione Peroxidase , Glutathione , Intervertebral Disc Degeneration , Intervertebral Disc , Lipid Peroxidation , Lumbar Vertebrae , Oxidative Stress , Thiobarbituric Acid Reactive Substances , Humans , Intervertebral Disc Degeneration/metabolism , Lipid Peroxidation/physiology , Glutathione Peroxidase/metabolism , Male , Female , Adult , Middle Aged , Glutathione/metabolism , Lumbar Vertebrae/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Oxidative Stress/physiology
11.
Drug Dev Res ; 85(4): e22199, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38812443

ABSTRACT

It has been reported that lipophilic statins such as atorvastatin can more readily penetrate into ß-cells and reach the mitochondria, resulting in mitochondrial dysfunction, oxidative stress, decrease in insulin release. Many studies have shown that natural products can protect mitochondrial dysfunction induced by drug in different tissue. We aimed to explore mitochondrial protection potency of hesperidin, vanillic acid, and sinapic acid as natural compounds against mitochondrial dysfunction induced by atorvastatin in pancreas isolated mitochondria. Mitochondria were isolated form rat pancreas and directly treated with toxic concentration of atorvastatin (500 µM) in presence of various concentrations hesperidin, vanillic acid, and sinapic acid (1, 10, and 100 µM) separately. Mitochondrial toxicity parameters such as the reactive oxygen species (ROS) formation, succinate dehydrogenases (SDH) activity, mitochondrial swelling, depletion of glutathione (GSH), mitochondrial membrane potential (MMP) collapse, and malondialdehyde (MDA) production were measured. Our findings demonstrated that atorvastatin directly induced mitochondrial toxicity at concentration of 500 µM and higher in pancreatic mitochondria. Except MDA, atorvastatin caused significantly reduction in SDH activity, mitochondrial swelling, ROS formation, depletion of GSH, and collapse of MMP. While, our data showed that all three protective compounds at low concentrations ameliorated atorvastatin-induced mitochondrial dysfunction with the increase of SDH activity, improvement of mitochondrial swelling, MMP collapse and mitochondrial GSH, and reduction of ROS formation. We can conclude that hesperidin, vanillic acid, and sinapic acid can directly reverse the toxic of atorvastatin in rat pancreas isolated mitochondria, which may be beneficial for protection against diabetogenic-induced mitochondrial dysfunction in pancreatic ß-cells.


Subject(s)
Atorvastatin , Coumaric Acids , Hesperidin , Membrane Potential, Mitochondrial , Mitochondria , Mitochondrial Swelling , Pancreas , Reactive Oxygen Species , Vanillic Acid , Animals , Atorvastatin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Coumaric Acids/pharmacology , Rats , Reactive Oxygen Species/metabolism , Male , Mitochondrial Swelling/drug effects , Membrane Potential, Mitochondrial/drug effects , Vanillic Acid/pharmacology , Hesperidin/pharmacology , Glutathione/metabolism , Rats, Wistar , Succinate Dehydrogenase/metabolism , Malondialdehyde/metabolism
12.
J Nanobiotechnology ; 22(1): 299, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38812031

ABSTRACT

BACKGROUND: Discrepancies in the utilization of reactive oxygen species (ROS) between cancer cells and their normal counterparts constitute a pivotal juncture for the precise treatment of cancer, delineating a noteworthy trajectory in the field of targeted therapies. This phenomenon is particularly conspicuous in the domain of nano-drug precision treatment. Despite substantial strides in employing nanoparticles to disrupt ROS for cancer therapy, current strategies continue to grapple with challenges pertaining to efficacy and specificity. One of the primary hurdles lies in the elevated levels of intracellular glutathione (GSH). Presently, predominant methods to mitigate intracellular GSH involve inhibiting its synthesis or promoting GSH efflux. However, a conspicuous gap remains in the absence of a strategy capable of directly and efficiently clearing GSH. METHODS: We initially elucidated the chemical mechanism underpinning oridonin, a diminutive pharmacological agent demonstrated to perturb reactive oxygen species, through its covalent interaction with glutathione. Subsequently, we employed the incorporation of maleimide-liposomes, renowned for their capacity to disrupt the ROS delivery system, to ameliorate the drug's water solubility and pharmacokinetics, thereby enhancing its ROS-disruptive efficacy. In a pursuit to further refine the targeting for acute myeloid leukemia (AML), we harnessed the maleic imide and thiol reaction mechanism, facilitating the coupling of Toll-like receptor 2 (TLR2) peptides to the liposomes' surface via maleic imide. This strategic approach offers a novel method for the precise removal of GSH, and its enhancement endeavors are directed towards fortifying the precision and efficacy of the drug's impact on AML targets. RESULTS: We demonstrated that this peptide-liposome-small molecule machinery targets AML and consequently induces cell apoptosis both in vitro and in vivo through three disparate mechanisms: (I) Oridonin, as a Michael acceptor molecule, inhibits GSH function through covalent bonding, triggering an initial imbalance of oxidative stress. (II) Maleimide further induces GSH exhaustion, aggravating redox imbalance as a complementary augment with oridonin. (III) Peptide targets TLR2, enhances the directivity and enrichment of oridonin within AML cells. CONCLUSION: The rationally designed nanocomplex provides a ROS drug enhancement and targeted delivery platform, representing a potential solution by disrupting redox balance for AML therapy.


Subject(s)
Diterpenes, Kaurane , Glutathione , Leukemia, Myeloid, Acute , Liposomes , Reactive Oxygen Species , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/pharmacology , Glutathione/metabolism , Glutathione/chemistry , Liposomes/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Humans , Reactive Oxygen Species/metabolism , Animals , Mice , Cell Line, Tumor , Toll-Like Receptor 2/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects
13.
J Hazard Mater ; 472: 134453, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38723481

ABSTRACT

Crop plants face complex tropospheric ozone (O3) stress, emphasizing the need for a food security-focused management strategy. While research extensively explores O3's harmful effects, this study delves into the combined impacts of O3 and CO2. This study investigates the contrasting responses of O3-sensitive (PBW-550) and O3-resistant (HUW-55) wheat cultivars, towards elevated ozone (eO3) and elevated carbon dioxide (eCO2), both individually and in combination. The output of the present study confirms the positive effect of eCO2 on wheat cultivars exposed to eO3 stress, with more prominent effects on O3-sensitive cultivar PBW-550, as compared to the O3-resistant HUW-55. The differential response of the two wheat cultivars can be attributed to the mechanistic variations in the enzyme activities of the Halliwell-Asada pathway (AsA-GSH cycle) and the ascorbate and glutathione pool. The results indicate that eCO2 was unable to uplift the regeneration of the glutathione pool in HUW-55, however, PBW-550 responded well, under similar eO3 conditions. The study's findings, highlighting mechanistic variations in antioxidants, show a more positive yield response in PBW-550 compared to HUW-55 under ECO treatment. This insight can inform agricultural strategies, emphasizing the use of O3-sensitive cultivars for sustained productivity in future conditions with high O3 and CO2 concentrations.


Subject(s)
Ascorbic Acid , Carbon Dioxide , Glutathione , Ozone , Triticum , Ozone/toxicity , Ozone/pharmacology , Triticum/drug effects , Triticum/metabolism , Carbon Dioxide/metabolism , Glutathione/metabolism , Ascorbic Acid/metabolism , Air Pollutants/toxicity
14.
Int J Biol Macromol ; 270(Pt 1): 132093, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710247

ABSTRACT

Long-term and excessive alcohol consumption can lead to the development of alcoholic liver disease (ALD), characterized by oxidative damage, intestinal barrier injury, and disruption of intestinal microbiota. In this study, we extracted fucoidan (Aj-FUC) from Apostichopus japonicus using enzymatic methods and characterized its structure. The ALD model was established in male Balb/c mice using 56° Baijiu, with silymarin as a positive control. Mice were orally administered 100 mg/kg·bw and 300 mg/kg·bw of Aj-FUC for 28 days to evaluate its effects on liver injury in ALD mice and explore its potential role in modulating the gut-liver axis. The results showed significant improvements in histopathological changes and liver disease in the Aj-FUC group. Aj-FUC treatment significantly increased the levels of glutathione (GSH) and glutathione peroxidase (GSH-Px) while weakly reduced the elevation of malondialdehyde (MDA) induced by ALD. It also regulated the Nrf2/HO-1 signaling pathway, collectively alleviating hepatic oxidative stress. Aj-FUC intervention upregulated the expression of ZO-1 and Occludin, thus contributing to repair the intestinal barrier. Additionally, Aj-FUC increased the content of short-chain fatty acids (SCFAs) and regulated the imbalance in gut microbiota. These results suggested that Aj-FUC alleviates ALD by modulating the gut-liver axis homeostasis. It may prove to be a useful dietary supplement in the treatment of alcoholic liver damage.


Subject(s)
Homeostasis , Liver Diseases, Alcoholic , Liver , Oxidative Stress , Polysaccharides , Stichopus , Animals , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Polysaccharides/pharmacology , Polysaccharides/chemistry , Mice , Male , Liver/drug effects , Liver/metabolism , Liver/pathology , Homeostasis/drug effects , Oxidative Stress/drug effects , Stichopus/chemistry , Mice, Inbred BALB C , Malondialdehyde/metabolism , Gastrointestinal Microbiome/drug effects , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Disease Models, Animal , Glutathione Peroxidase/metabolism
15.
Nat Commun ; 15(1): 3684, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693181

ABSTRACT

The metal-nucleic acid nanocomposites, first termed metal-nucleic acid frameworks (MNFs) in this work, show extraordinary potential as functional nanomaterials. However, thus far, realized MNFs face limitations including harsh synthesis conditions, instability, and non-targeting. Herein, we discover that longer oligonucleotides can enhance the synthesis efficiency and stability of MNFs by increasing oligonucleotide folding and entanglement probabilities during the reaction. Besides, longer oligonucleotides provide upgraded metal ions binding conditions, facilitating MNFs to load macromolecular protein drugs at room temperature. Furthermore, longer oligonucleotides facilitate functional expansion of nucleotide sequences, enabling disease-targeted MNFs. As a proof-of-concept, we build an interferon regulatory factor-1(IRF-1) loaded Ca2+/(aptamer-deoxyribozyme) MNF to target regulate glucose transporter (GLUT-1) expression in human epidermal growth factor receptor-2 (HER-2) positive gastric cancer cells. This MNF nanodevice disrupts GSH/ROS homeostasis, suppresses DNA repair, and augments ROS-mediated DNA damage therapy, with tumor inhibition rate up to 90%. Our work signifies a significant advancement towards an era of universal MNF application.


Subject(s)
Aptamers, Nucleotide , DNA, Catalytic , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Humans , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Cell Line, Tumor , DNA, Catalytic/metabolism , DNA, Catalytic/chemistry , Animals , Receptor, ErbB-2/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Reactive Oxygen Species/metabolism , Mice , DNA Repair , DNA Damage , Glutathione/metabolism , Glutathione/chemistry , Nucleic Acids/metabolism , Nucleic Acids/chemistry
16.
Biomed Khim ; 70(2): 73-82, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38711406

ABSTRACT

Thiram is a dithiocarbamate derivative, which is used as a fungicide for seed dressing and spraying during the vegetation period of plants, and also as an active vulcanization accelerator in the production of rubber-based rubber products. In this study the content of reactive oxygen species (ROS) and the state of the glutathione system have been investigated in the oral fluid and gum tissues of adult male Wistar rats treated with thiram for 28 days during its administration with food at a dose of 1/50 LD50. Thiram induced formation of ROS in the oral cavity; this was accompanied by an imbalance in the ratio of reduced and oxidized forms of glutathione due to a decrease in glutathione and an increase in its oxidized form as compared to the control. Thiram administration caused an increase in the activity of glutathione-dependent enzymes (glutathione peroxidase, glutathione transferase, and glutathione reductase). However, the time-course of enzyme activation in the gum tissues and oral fluid varied in dependence on the time of exposure to thiram. In the oral fluid of thiram-treated rats changes in the antioxidant glutathione system appeared earlier. The standard diet did not allow the glutathione pool to be fully restored to physiological levels after cessation of thiram intake. The use of exogenous antioxidants resviratrol and an Echinacea purpurea extract led to the restoration of redox homeostasis in the oral cavity.


Subject(s)
Antioxidants , Fungicides, Industrial , Glutathione , Rats, Wistar , Reactive Oxygen Species , Thiram , Animals , Male , Rats , Glutathione/metabolism , Reactive Oxygen Species/metabolism , Fungicides, Industrial/toxicity , Thiram/toxicity , Antioxidants/pharmacology , Mouth/metabolism , Mouth/drug effects , Glutathione Reductase/metabolism , Glutathione Transferase/metabolism , Glutathione Peroxidase/metabolism
17.
Reprod Domest Anim ; 59(5): e14596, 2024 May.
Article in English | MEDLINE | ID: mdl-38757656

ABSTRACT

Chlorogenic acid (CGA) is an effective phenolic antioxidant that can scavenge hydroxyl radicals and superoxide anions. Herein, the protective effects and mechanisms leading to CGA-induced porcine parthenogenetic activation (PA) in early-stage embryos were investigated. Our results showed that 50 µM CGA treatment during the in vitro culture (IVC) period significantly increased the cleavage and blastocyst formation rates and improved the blastocyst quality of porcine early-stage embryos derived from PAs. Then, genes related to zygotic genome activation (ZGA) were identified and investigated, revealing that CGA can promote ZGA in porcine PA early-stage embryos. Further analysis revealed that CGA treatment during the IVC period decreased the abundance of reactive oxygen species (ROS), increased the abundance of glutathione and enhanced the activity of catalase and superoxide dismutase in porcine PA early-stage embryos. Mitochondrial function analysis revealed that CGA increased mitochondrial membrane potential and ATP levels and upregulated the mitochondrial homeostasis-related gene NRF-1 in porcine PA early-stage embryos. In summary, our results suggest that CGA treatment during the IVC period helps porcine PA early-stage embryos by regulating oxidative stress and improving mitochondrial function.


Subject(s)
Chlorogenic Acid , Embryo Culture Techniques , Embryonic Development , Mitochondria , Oxidative Stress , Parthenogenesis , Reactive Oxygen Species , Animals , Oxidative Stress/drug effects , Parthenogenesis/drug effects , Mitochondria/drug effects , Embryo Culture Techniques/veterinary , Chlorogenic Acid/pharmacology , Embryonic Development/drug effects , Reactive Oxygen Species/metabolism , Blastocyst/drug effects , Swine , Membrane Potential, Mitochondrial/drug effects , Antioxidants/pharmacology , Female , Glutathione/metabolism
18.
J Nanobiotechnology ; 22(1): 253, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755600

ABSTRACT

Improving cancer therapy by targeting the adverse tumor microenvironment (TME) rather than the cancer cells presents a novel and potentially effective strategy. In this study, we introduced FexMoyS nanoparticles (NPs), which act as sequential bioreactors to manipulate the TME. FexMoyS NPs were synthesized using thermal decomposition and modified with polyethylene glycol (PEG). Their morphology, chemical composition, and photothermal properties were characterized. The capability to produce ROS and deplete GSH was evaluated. Effects on CRC cells, including cell viability, apoptosis, and glycolysis, were tested through various in vitro assays. In vivo efficacy was determined using CRC-bearing mouse models and patient-derived xenograft (PDX) models. The impact on the MAPK signaling pathway and tumor metabolism was also examined. The FexMoyS NPs showed efficient catalytic activity, leading to increased ROS production and GSH depletion, inducing ferroptosis, and suppressing glycolysis in CRC cells. In vivo, the NPs significantly inhibited tumor growth, particularly when combined with NIR light therapy, indicating a synergistic effect of photothermal therapy and chemodynamic therapy. Biosafety assessments revealed no significant toxicity in treated mice. RNA sequencing suggested that the NPs impact metabolism and potentially immune processes within CRC cells. FexMoyS NPs present a promising multifaceted approach for CRC treatment, effectively targeting tumor cells while maintaining biosafety. The nanoparticles exhibit potential for clinical translation, offering a new avenue for cancer therapy.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Glycolysis , Polyethylene Glycols , Reactive Oxygen Species , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Humans , Mice , Polyethylene Glycols/chemistry , Ferroptosis/drug effects , Glycolysis/drug effects , Cell Line, Tumor , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , Mice, Nude , Apoptosis/drug effects , Cell Survival/drug effects , Female , Glutathione/metabolism
19.
Neuroimage ; 293: 120632, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701994

ABSTRACT

During aging, the brain is subject to greater oxidative stress (OS), which is thought to play a critical role in cognitive impairment. Glutathione (GSH), as a major antioxidant in the brain, can be used to combat OS. However, how brain GSH levels vary with age and their associations with cognitive function is unclear. In this study, we combined point-resolved spectroscopy and edited spectroscopy sequences to investigate extended and closed forms GSH levels in the anterior cingulate cortex (ACC), posterior cingulate cortex (PCC), and occipital cortex (OC) of 276 healthy participants (extended form, 166 females, age range 20-70 years) and 15 healthy participants (closed form, 7 females, age range 26-56 years), and examined their relationships with age and cognitive function. The results revealed decreased extended form GSH levels with age in the PCC among 276 participants. Notably, the timecourse of extended form GSH level changes in the PCC and ACC differed between males and females. Additionally, positive correlations were observed between extended form GSH levels in the PCC and OC and visuospatial memory. Additionally, a decreased trend of closed form GSH levels with age was also observed in the PCC among 15 participants. Taken together, these findings enhance our understanding of the brain both closed and extended form GSH time course during normal aging and associations with sex and memory, which is an essential first step for understanding the neurochemical underpinnings of healthy aging.


Subject(s)
Aging , Glutathione , Humans , Female , Middle Aged , Male , Adult , Aged , Glutathione/metabolism , Aging/metabolism , Aging/physiology , Young Adult , Spatial Memory/physiology , Occipital Lobe/metabolism , Gyrus Cinguli/metabolism , Brain/metabolism
20.
Bull Exp Biol Med ; 176(5): 617-619, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38730108

ABSTRACT

We studied the effect of the HSP27 inhibitor, 5-(5-ethyl-2-hydroxy-4-methoxyphenyl)-4-(4-methoxyphenyl)-isoxazole, at a final concentration of 0.1 µM and/or the apoptosis inducer dexamethasone at a final concentration of 10 µM on the content of hydroxyl radical, reduced and oxidized glutathione, HSP27, activity of glutathione reductase, glutathione peroxidase, caspase-3, and the number of Annexin+ Jurkat tumor cells. The involvement of HSP27 in apoptosis of Jurkat tumor cells was demonstrated. Simultaneous exposure to the HSP27 inhibitor and dexamethasone resulted in an increase in the level of HSP27 against the background of developing oxidative stress (increase in the concentration of hydroxyl radicals and changes in the state of the glutathione system).


Subject(s)
Apoptosis , Caspase 3 , Dexamethasone , Glutathione , HSP27 Heat-Shock Proteins , Oxidative Stress , Humans , Dexamethasone/pharmacology , Jurkat Cells , Apoptosis/drug effects , HSP27 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/genetics , Glutathione/metabolism , Caspase 3/metabolism , Caspase 3/genetics , Oxidative Stress/drug effects , Glutathione Reductase/metabolism , Glutathione Peroxidase/metabolism , Hydroxyl Radical/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL