Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
In Silico Pharmacol ; 12(1): 23, 2024.
Article in English | MEDLINE | ID: mdl-38584776

ABSTRACT

The dynamic behavior of Protein Disulfide Isomerase (PDI) in an aqueous solution environment under physiologically active pH has been experimentally verified in this study using Small Angle X-ray Scattering (SAXS) technique. The structural mechanism of dimerization for full-length PDI molecules and co-complex with two renowned substrates has been comprehensively discussed. The structure models obtained from the SAXS data of PDI purified from bovine liver display behavior duality between unaccompanied-enzyme and after engaged with substrates. The analysis of SAXS data revealed that PDI exists as a homo-dimer in the solution environment, and substrate induction provoked its segregation into monomer to enable the enzyme to interact systematically with incoming clients. Supplementary Information: The online version contains supplementary material available at 10.1007/s40203-024-00198-0.

2.
Biofactors ; 44(3): 289-298, 2018 May.
Article in English | MEDLINE | ID: mdl-29672963

ABSTRACT

One of the worldwide metabolic health dilemma is nonalcoholic fatty liver diseases (NAFLD). Researchers are searching effective drug to manage NAFLD patients. One of the best way to manage the metabolic imperfection is through natural principal isolated from different sources. Butein, a natural compound known to have numerous pharmacological application. In the current study we assessed the therapeutic effect of butein administration on liver function tests, oxidative stress, antioxidants, lipid abnormalities, serum inflammatory cytokines, and mitochondrial reactive oxygen species levels, in rats with methionine-choline deficient (MCD) diet induced NAFLD. Male Wistar rats were treated with MCD diet with/without butein (200 mg/kg body wt. orally) for 6 weeks. The protective effect of butein, were evident from decreased transaminase activities, restoration of albumin, globulin, albumin/globulin ratio, and oxidants in serum (P < 0.01), further it improved liver antioxidant status (P < 0.01). Butein significantly lowered lipid profile parameters (P < 0.01), suppressed inflammatory cytokines (P < 0.01), and improved liver histology. Further to understand the possible mechanism behind the hepatoprotective and lipid lowering effect of butein, the activities of heme oxygenase (HO1), myeloperoxidase (MPO), and mitochondrial reactive oxygen species (ROS) were measured. We found that butein supplementation significantly decreased the activity of HO1 (P < 0.001), and increased the activity of MPO (P < 0.001). Furthermore butein attenuated mitochondrial ROS produced in NAFLD condition. Present study shows that butein supplementation restore liver function by altering liver oxidative stress, inflammatory markers, vital defensive enzyme activities, and mitochondrial ROS. In summary, butein has remarkable potential to develop effective hepato-protective drug. © 2018 BioFactors, 44(3):289-298, 2018.


Subject(s)
Chalcones/pharmacology , Choline Deficiency/drug therapy , Diet/adverse effects , Hypolipidemic Agents/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Administration, Oral , Albumins/metabolism , Animals , Choline/metabolism , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Globulins/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Liver/metabolism , Liver/pathology , Male , Methionine/deficiency , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Transaminases/antagonists & inhibitors , Transaminases/metabolism
3.
J Pharm Biomed Anal ; 154: 294-301, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29567572

ABSTRACT

Drug repurposing has been an emerging therapeutic strategy, which involves exploration of a new therapeutic approach for the use of an existing drug. Glibenclamide (Gli) is an anti-diabetic sulfonylurea drug extensively used for the treatment of type-2 diabetes, it has also been shown to possess anti-proliferative effect against several types of tumors. The present study was executed to understand the mechanisms underlying the interaction of Gli with DNA under physiological conditions. The binding mechanism of Gli with DNA was scrutinized by UV-vis absorption spectroscopy and fluorescence emission spectroscopy. The conformational changes and electrochemical properties were analyzed by circular dichroism spectroscopy and cyclic voltammetry. Isothermal titration calorimetry was employed to examine the thermodynamic changes and molecular docking technique used to analyze the interaction mode of Gli with DNA. The spectroscopic studies revealed that Gli interacts with DNA through groove binding mode. Further, isothermal titration calorimetry depicted a stronger mode of interaction favorably groove-binding. Recently, systemic combination therapy has shown significant promise in inhibiting multiple targets simultaneously yielding high therapeutic competence with lesser side effects. With this concern, we intended to study the combined cytotoxicity of Gli with doxorubicin (Dox). The results of MTT assay and acridine orange (AO)/ethidium bromide (EtBr) staining showed synergistic cytotoxicity of Gli + Dox combination on HepG2 & A549 cells. The present study documents the intricate mechanism of Gli-DNA interaction and delivers a multifaceted access for chemotherapy by Gli + Dox combination.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Doxorubicin/pharmacology , Glyburide/pharmacology , A549 Cells , Calorimetry/methods , Cell Line, Tumor , Circular Dichroism/methods , DNA/drug effects , Drug Synergism , Fluorescence , Hep G2 Cells , Humans , Molecular Docking Simulation/methods , Spectrometry, Fluorescence/methods , Thermodynamics
4.
Nanomedicine ; 14(2): 415-428, 2018 02.
Article in English | MEDLINE | ID: mdl-29166623

ABSTRACT

Drawbacks and limitations of recently available therapies to hepatocellular cancer (HCC) devoted the scientist to focus on emerging new strategies. ZnO nanoparticles (ZnONPs) based chemotherapeutics has been emanating as a promising approach to maximize therapeutic synergy facilitating the discovery of novel multitargeted combinations. In the present study we conjugated ZnONPs with ferulic acid (ZnONPs-FAC) characterized by computational, spectroscopic and microscopic techniques. In vitro anticancer potential has been evaluated by assessing cell viability, morphology, ROS generation, mitochondrial membrane permeability, comet assay, immunofluorescent staining of 8-OHdG, Ki67 and γ-H2AX, cell cycle analysis and western blot analysis and in vivo anticancer potential against DEN induced HCC was analyzed by histopathological and immunohistochemical methods. The results revealed that ZnONPs-FAC induces cell death through apoptosis and can suppress the DEN-induced HCC. Our study documents therapeutic potential of nanoparticle conjugated with phytochemicals, suggesting a new platform for combinatorial chemotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Coumaric Acids/chemistry , Diethylnitrosamine/toxicity , Liver Neoplasms, Experimental/drug therapy , Nanoparticles/administration & dosage , Zinc Oxide/chemistry , Alkylating Agents/toxicity , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Nanoparticles/chemistry , Rats , Rats, Wistar , Tumor Cells, Cultured
5.
Mol Biol Rep ; 44(6): 463-474, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29185131

ABSTRACT

Nimbolide is a bioactive compound found in Azadirachta indica. This work was devised to investigate the potential effects of nimbolide on intracellular lipid deposition and its associated redox modulation in primary hepatocytes (Heps). Lipid accumulation was induced in Heps by supplementing 1 mM oleic acid for 24 h which was marked by significant accumulation of lipids. The results demonstrated that nimbolide can decrease intracellular cholesterol, free fatty acids and triglycerides. Nimbolide may also improve hepatocytes function through its antioxidant effects by inhibiting oxidative DNA damage and lipid peroxidation by curtailing the reactive oxygen species levels. Further it also restore the mitochondrial potential, improving the endogenous antioxidant levels such as GSH and antioxidant enzyme activities. Nimbolide increased (P < 0.05) liver X receptor-α (LXRα), peroxisome proliferator-activated receptor-γ (PPARγ) and sterol regulatory element-binding protein-1c (SREBP1c) gene expression in Heps. The biological significance of nimbolide may involve hypolipidemic effect, lipid peroxidation inhibition, DNA damage inhibition, ROS inhibition, restoring mitochondrial function, increases in GSH and SOD & CAT activities, and direct regulation of LXRα, PPARγ and SREBP1c gene expression. Nimbolide may be used as effective lipid lowering compound and lipid deposition-induced Heps changes.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Limonins/pharmacology , Lipid Metabolism/drug effects , Animals , Antioxidants/pharmacology , Cells, Cultured , Humans , Hypolipidemic Agents/pharmacology , Lipid Peroxidation/drug effects , Liver X Receptors/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , PPAR gamma/metabolism , Reactive Oxygen Species/metabolism , Triglycerides/metabolism
6.
Inflamm Res ; 66(9): 753-763, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28560522

ABSTRACT

BACKGROUND AND AIM: Liver inflammation stimulates various inflammatory cytokines and initiates injury through oxidative stress. The aim of this study was to curtaile the liver injury through natural principles such as 2-hydroxy-4-methoxy benzoic acid (HMBA). METHODS: The current study examines the hepatoprotective and lipid lowering effect of HMBA against carbon tetra chloride (CCl4)-mediated liver toxicity in male Wistar rats. RESULTS: The hepatoprotective effects of HMBA against CCl4-induced liver damage, were evident from low serum transaminases activities, reduced hepatic lipid peroxidation and collagen content, restoration of total glutathione, and recouping of the inflammatory cytokines, such as TNF-α, IL-1ß, IL-10, and IL-6 levels. Further it was found that the treatment of HMBA, significantly lowered (P<0.01) the levels of total cholesterol, triglycerides, free fatty acids and phospholipids in serum and liver. To investigate the mechanism behind the hepatoprotective and lipid lowering effect, the activities of heme oxygenase (HO1), and myeloperoxidase (MPO) were measured and expression levels were quantified through western blot following HMBA administration. The results showed that HMBA administration significantly decreased the activity of HO1 (P<0.001), and increased the activity of MPO (P<0.001); further similar finding was observed in western analysis. The hepatoprotective, lipid lowering and shifting key defensive enzyme activities are similar to that of standard drug such as N-acetylcysteine. CONCLUSION: HMBA is competent of shielding liver from CCl4-induced hepatotoxicity, and this is associated with the lipid lowering, inflammatory cytokine restoration and induction of defensive enzyme activities.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Benzoates/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Benzoates/pharmacology , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Collagen/metabolism , Cytokines/blood , Glutathione/metabolism , Heme Oxygenase-1/metabolism , Hydroxyproline/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Peroxidase/metabolism , Rats, Wistar
7.
Mol Biol Rep ; 40(12): 6533-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24197690

ABSTRACT

The present study brings out the preventive role of (-)-epigallocatechin-gallate (EGCG) on cardiac mitochondrial metabolism and apoptosis in cigarette smoke (CS)-exposed rats. The CS-exposed rats showed significantly decreased activities of TCA cycle enzymes and mitochondrial enzymatic antioxidants, on the other hand, mitochondrial lipid peroxidation was increased and GSH level was decreased. Further, CS exposure was found to induce cardiac apoptosis through release of cytochrome c into the cytosol, cleavage of pro-caspase-3 to active caspase-3, up-regulation of pro-apoptotic (Bax) and down-regulation of antiapoptotic (Bcl-2) molecules. The CS-induced apoptosis was further confirmed by mitochondrial and nuclear ultra structural apoptotic features as evaluated by electron microscopic studies. EGCG supplementation shelters the activities of TCA cycle enzymes and antioxidant enzymes, with concomitant decrease in lipid peroxidation and increase in GSH level. EGCG administration inhibited apoptosis through the inhibition of cytochrome c release into cytosol, activation of pro-caspase-3, down regulation of Bax and significant up regulation of Bcl-2. EGCG reversed the ultra structural apoptotic alterations of mitochondria and nucleus. The present study has provided experimental evidences that the EGCG treatment enduring to cardio protection at mitochondrial level.


Subject(s)
Apoptosis/drug effects , Catechin/analogs & derivatives , Heart/physiopathology , Mitochondria, Heart/enzymology , Smoking/adverse effects , Animals , Antioxidants/metabolism , Body Weight/drug effects , Catechin/pharmacology , Citric Acid Cycle/drug effects , Electrophoresis, Agar Gel , Enzyme Stability/drug effects , Glutathione/metabolism , Heart/drug effects , Lipid Peroxidation/drug effects , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocardium/enzymology , Myocardium/pathology , Myocardium/ultrastructure , Organ Size/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...