Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 270
1.
Acta Med Okayama ; 77(3): 243-254, 2023 Jun.
Article En | MEDLINE | ID: mdl-37357625

Brown adipose tissue (BAT) plays a critical role in metabolic homeostasis. BAT dysfunction is associated with the development of obesity through an imbalance between energy expenditure and energy intake. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is the master regulator of adipogenesis. However, the roles of PPARγ and thiazolidinediones (TZDs) in the regulation of BAT metabolism remain unclear. TZDs, which are selective PPARγ activators, improve systemic insulin resistance in animals and humans. In the present study, we generated brown adipocyte-specific PPARγ-deficient mice (BATγKO) to examine the in vivo roles of PPARγ and TZDs in BAT metabolism. In electron microscopic examinations, brown adipocyte-specific PPARγ deletion promoted severe whitening of brown fat and morphological alteration of mitochondria. Brown adipocyte-specific PPARγ deletion also reduced mRNA expression of BATselective genes. Although there was no difference in energy expenditure between control and BATγKO mice in calorimetry, norepinephrine-induced thermogenesis was impaired in BATγKO mice. Moreover, pioglitazone treatment improved diet-induced insulin resistance in the control mice but not in the BATγKO mice. These findings suggest that BAT PPARγ is necessary for the maintenance of brown adipocyte function and for the insulin-sensitizing action of TZDs.


Insulin Resistance , Insulins , Thiazolidinediones , Humans , Mice , Animals , Thiazolidinediones/pharmacology , Thiazolidinediones/metabolism , Adipose Tissue, Brown/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Insulin Resistance/physiology , Insulins/metabolism , Adipose Tissue
2.
Eur J Cell Biol ; 102(2): 151320, 2023 Jun.
Article En | MEDLINE | ID: mdl-37130450

Adipose tissue (AT) dysfunctions are associated with the onset of insulin resistance (IR) and type 2 diabetes mellitus (T2DM). Targeting glucose-dependent insulinotropic peptide receptor (GIPR) is a valid option to increase the efficacy of glucagon-like peptide 1 (GLP-1) receptor agonists in T2DM treatment. Nevertheless, the therapeutic potential of targeting the GIP/GIPR axis and its effect on the AT are controversial. In this work, we explored the expression and regulation of GIPR in precursor cells and mature adipocytes, investigating if and how obesogenic stimuli and thiazolidinediones perturb GIPR expression. Using publicly available gene expression datasets, we assessed that, among white adipose tissue (WAT) cells, adipocytes express lower levels of GIPR compared to cells of mesothelial origin, pericytes, dendritic and NK/T cells. However, we report that GIPR levels markedly increase during the in vitro differentiation of both murine and human adipocytes, from 3T3-L1 and human mesenchymal precursor cells (MSCs), respectively. Notably, we demonstrated that thiazolidinediones - ie. synthetic PPARγ agonists widely used as anti-diabetic drugs and contained in the adipogenic mix - markedly induce GIPR expression. Moreover, using multiple in vitro systems, we assessed that thiazolidinediones induce GIPR in a PPARγ-independent manner. Our results support the hypothesis that PPARγ synthetic agonists may be used to increase GIPR levels in AT, potentially affecting in turn the targeting of GIP system in patients with metabolic dysfunctions. Furthermore, we demonstrate in vitro and in vivo that proinflammatory stimuli, and especially the TNFα, represses GIPR both in human and murine adipocytes, even though discordant results were obtained between human and murine cellular systems for other cytokines. Finally, we demonstrated that GIPR is negatively affected also by the excessive lipid engulfment. Overall, we report that obesogenic stimuli - ie. pro-inflammatory cytokines and the increased lipid accumulation - and PPARγ synthetic ligands oppositely modulate GIPR expression, possibly influencing the effectiveness of GIP agonists.


Diabetes Mellitus, Type 2 , Thiazolidinediones , Humans , Mice , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/metabolism , Adipocytes/metabolism , Lipids/pharmacology
3.
Article En | MEDLINE | ID: mdl-37156296

Podocytes are specialized epithelial cells that maintain the glomerular filtration barrier. These cells are susceptible to lipotoxicity in the obese state and irreversibly lost during kidney disease leading to proteinuria and renal injury. PPARγ is a nuclear receptor whose activation can be renoprotective. This study examined the role of PPARγ in the lipotoxic podocyte using a PPARγ knockout (PPARγKO) cell line and since the activation of PPARγ by Thiazolidinediones (TZD) is limited by their side effects, it explored other alternative therapies to prevent podocyte lipotoxic damage. Wild-type and PPARγKO podocytes were exposed to the fatty acid palmitic acid (PA) and treated with the TZD (Pioglitazone) and/or the Retinoid X receptor (RXR) agonist Bexarotene (BX). It revealed that podocyte PPARγ is essential for podocyte function. PPARγ deletion reduced key podocyte proteins including podocin and nephrin while increasing basal levels of oxidative and ER stress causing apoptosis and cell death. A combination therapy of low-dose TZD and BX activated both the PPARγ and RXR receptors reducing PA-induced podocyte damage. This study confirms the crucial role of PPARγ in podocyte biology and that their activation in combination therapy of TZD and BX may be beneficial in the treatment of obesity-related kidney disease.


Kidney Diseases , Podocytes , Thiazolidinediones , Humans , PPAR gamma/metabolism , Pioglitazone/pharmacology , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Kidney Diseases/drug therapy , Bexarotene/pharmacology
4.
J Endocrinol ; 257(1)2023 04 01.
Article En | MEDLINE | ID: mdl-36688873

Peroxisome proliferator-activated receptor γ (PPARγ) belongs to a family of nuclear receptors that could serve as lipid sensors. PPARγ is the target of a group of insulin sensitizers called thiazolidinediones (TZDs) which regulate the expression of genes involved in glucose and lipid metabolism as well as adipokines that regulate metabolic function in other tissues. Non-alcoholic fatty liver disease (NAFLD) has a high prevalence worldwide and is even higher in patients with obesity and insulin resistance. TZD-mediated activation of PPARγ could serve as a good treatment for NAFLD because TZDs have shown anti-fibrogenic and anti-inflammatory effectsin vitro and increase insulin sensitivity in peripheral tissues which improves liver pathology. However, mechanistic studies in mouse models suggest that the activation of PPARγ in hepatocytes might reduce or limit the therapeutic potential of TZD against NAFLD. In this review, we briefly describe the short history of PPAR isoforms, the relevance of their expression in different tissues, as well as the pathogenesis and potential therapeutics for NAFLD. We also discuss some evidence derived from mouse models that could be useful for endocrinologists to assess tissue-specific roles of PPARs, complement reverse endocrinology approaches, and understand the direct role that PPARγ has in hepatocytes and non-parenchymal cells.


Insulin Resistance , Non-alcoholic Fatty Liver Disease , Thiazolidinediones , Animals , Mice , Hepatocytes/metabolism , Insulin Resistance/physiology , Non-alcoholic Fatty Liver Disease/metabolism , PPAR gamma/metabolism , Thiazolidinediones/metabolism , Thiazolidinediones/therapeutic use
5.
Ren Fail ; 44(1): 2056-2065, 2022 Dec.
Article En | MEDLINE | ID: mdl-36420656

BACKGROUND: In recent years, peroxisome proliferator-activated receptor γ (PPARγ) has been found to be closely associated with hypoxia renal disease. The aim of this study was to investigate the relationship between rosiglitazone and mitochondrial apoptosis in renal tissue and its associated mechanisms. METHODS: Twenty-four male Sprague-Dawley rats were randomly divided into three groups (n = 8 in each): normal control group, hypoxia injury group (equal volume of 0.9% saline), and PPARγ agonist group (Rosiglitazone, 10 mg/kg · d, intraperitoneally). The hypoxia injury group and PPARγ agonist group were placed in a hypoxia chamber and the simulated altitude was set at 7,000 m for 7 days. Blood and kidney samples were collected after 7 days. The quantitative real-time polymerase chain reaction and Western blot methods were used to determine the expression of PPARγ, nuclear factor kappa-B (NF-κB), B-cell lymphoma-2 (Bcl-2), and Bax. RESULTS: The results showed that compared with the normal control group, the renal tissue of rats after hypoxia was severely damaged, as shown by massive renal tubular epithelial cell degeneration and detachment, and renal tubular dilation. The NF-κB protein expression significantly increased, the Bcl-2 protein and mRNA expression significantly decreased, and Bax protein and mRNA expression significantly increased (p < .05 for all). Renal injury was much less severe in the PPARγ agonist group compared to the hypoxia injury group. CONCLUSIONS: Rosiglitazone can alleviate hypoxia renal injury, with the possible mechanism involving attenuation of apoptosis by inhibiting the activation of the NF-κB signaling pathway in a PPARγ-dependent manner and increasing Bcl-2 and decreasing Bax expression.


PPAR gamma , Thiazolidinediones , Male , Rats , Animals , Rosiglitazone/pharmacology , PPAR gamma/metabolism , NF-kappa B/metabolism , bcl-2-Associated X Protein/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/metabolism , Rats, Sprague-Dawley , Signal Transduction , Apoptosis , Epithelial Cells/metabolism , Hypoxia/complications , Hypoxia/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Hypoglycemic Agents , Kidney/metabolism , RNA, Messenger/metabolism
6.
Life Sci ; 299: 120537, 2022 Jun 15.
Article En | MEDLINE | ID: mdl-35398016

AIMS: To accommodate surplus energy, adipose tissue expands by increasing both adipose cell size (hypertrophy) and cell number (hyperplasia). Enlarged, hypertrophic adipocytes are known to have reduced insulin response and impaired glucose transport, which negatively influence whole-body glucose homeostasis. Rosiglitazone is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, known to stimulate hyperplasia and to efficiently improve insulin sensitivity. Still, a limited amount of research has investigated the effects of rosiglitazone in mature, hypertrophic adipocytes. Therefore, the objective of this study was to examine rosiglitazone's effect on insulin-stimulated glucose uptake in hypertrophic adipocytes. MAIN METHODS: C57BL/6J male mice were subjected to 2 weeks of high-fat diet (HFD) followed by 1 week of HFD combined with daily administration of rosiglitazone (10 mg/kg). Adipose cell-size distribution and gene expression were analysed in intact adipose tissue, and glucose uptake, insulin response, and protein expression were examined using primary adipocytes isolated from epididymal and inguinal adipose tissue. KEY FINDINGS: HFD-feeding induced an accumulation of hypertrophic adipocytes, which was not affected by rosiglitazone-treatment. Still, rosiglitazone efficiently improved insulin-stimulated glucose transport without restoring insulin signaling or GLUT4 expression in similar-sized adipocytes. This improvement occurred concurrently with extracellular matrix remodelling and restored intracellular levels of targets involved in actin turnover. SIGNIFICANCE: These results demonstrate that rosiglitazone improves glucose transport in hypertrophic adipocytes, and highlights the importance of the cytoskeleton and extracellular matrix as potential therapeutic targets.


Actins , Thiazolidinediones , Actins/metabolism , Adipocytes/metabolism , Animals , Glucose/metabolism , Hyperplasia/metabolism , Hypertrophy , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , Rosiglitazone/pharmacology , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
7.
Ecotoxicol Environ Saf ; 233: 113308, 2022 Mar 15.
Article En | MEDLINE | ID: mdl-35176672

Thiazolidinedione (TZD) is an oral anti-diabetic drug that exhibits some side effects on the male reproductive system by interfering with the steroidogenesis and androgenic activity and also shows anti-proliferative effect on several cell types. This study investigated the effect of TZD on immature chicken Sertoli cell (SC) proliferation and the potential mechanism by which 17ß-estradiol regulated this process. Chicken SC viability was investigated under different treatment concentration and time of TZD. 17ß-estradiol (0.001 µM, 24 h) was added to analyze its effects on TZD-mediated cell viability, cell metabolic activity, cell growth, cell cycle progression, reactive oxygen species (ROS) level, antioxidant enzyme activity, mitochondria activity, oxygen consumption rate, adenosine triphosphate (ATP) level, and mitochondrial respiratory chain enzyme activity, adiponectin expression and several cell proliferation-related genes mRNA and protein levels. We performed the microRNA (miRNA) array to find TZD-induced differentially expressed miRNAs and validated whether miR-1577 can target on adiponectin via the dual luciferase reporter assay, as well as verified the effect of adiponectin addition with different concentrations on the SC viability. Further, SCs were transfected with miR-1577 agomir (a double-stranded synthetic miRNA mimic) in the presence or absence of TZD and antagomir (a single-stranded synthetic miRNA inhibitor) in the presence or absence of 17ß-estradiol to analyze whether miR-1577 was involved in TZD-mediated SC proliferation and whether 17ß-estradiol regulated this process. Results showed that TZD significantly inhibited SC viability, cell metabolic activity, cell growth, and cell cycle progression, while increased adiponectin level and ROS generation. TZD-treated SCs presented decreases of antioxidant enzyme activity, mitochondria activity, basal and maximal respiration, ATP production and level, mitochondrial respiratory chain enzyme activity, and mRNA and protein expressions of several cell proliferation-related genes, as well as the significant alteration of miRNA expressions (a total number of 55 miRNAs were up-regulated whereas 53 miRNAs down-regulated). Whereas, 17ß-estradiol played a positive role in chicken SC proliferation and rescued the damage of TZD on SC proliferation by up-regulating miR-1577 expression whose target gene was validated to be the adiponectin. In addition, exogenous adiponectin (more than 1 µg/ml) treatment exhibited a significant inhibition on the SC viability. Transfection of miR-1577 agomir promoted the SC proliferation via down-expressed adiponectin, and increased the mitochondrial function and cell proliferation-related gene expression, while TZD weakened the positive effect of miR-1577 agomir on SCs. On the other hand, transfection of miR-1577 antagomir inhibited SC proliferation by producing the opposite effects on above parameters, while 17ß-estradiol attenuated the negative effect of miR-1577 antagomir on SCs. These findings suggest down-expressed miR-1577 is involved in the regulation of TZD-inhibited SC proliferation through increasing adiponectin level, and this damage of TZD on the immature chicken SC proliferation can be ameliorated by appropriate dose of exogenous 17ß-estradiol treatment. This study provides an insight into the cytoprotective effect of 17ß-estradiol on TZD-damaged SC proliferation and may suggest a potential strategy for reducing the risk of SC dysfunction caused by the abuse of TZD.


Chickens , Thiazolidinediones , Adiponectin/genetics , Animals , Cell Proliferation , Chickens/metabolism , Estradiol/metabolism , Male , Sertoli Cells/metabolism , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
8.
Proteins ; 90(1): 142-154, 2022 01.
Article En | MEDLINE | ID: mdl-34331342

Derivatives of 2,4-thiazolidinedione have been reported to inhibit the aggregation of tau protein, in which compound 30 (C30) not only inhibit 80% of paired helical filament 6 (PHF6) aggregation, but also inhibit K18 and full-length tau aggregation. However, its inhibitory mechanism is unclear. In this study, to investigate the effect of C30 on tau protein, all-atom molecular dynamics simulation was performed on the PHF6 oligomer with and without C30. The results show that C30 can cause significant conformational changes in the PHF6 oligomer. The nematic order parameter P2 and secondary structure analyses show that C30 destroys the ordered structure of PHF6 oligomer, reduces the content of ß-sheet structure, and transforms ß-sheet into random coil structure. By clustering analysis, it was found that C30 has four possible binding sites on the PFH6 oligomer, and the binding ability order is S1 > S2 > S4 > S3. Following a more in-depth analyses of each site, it was determined that the S1 site is the most possible binding site mainly located between layers of L1 and L3. The hydrophobic interaction is the driving force for the binding of C30 to PHF6 oligomer. In addition, L1P4_Y310, L1P5_Y310, L3P1_V309, and L3P2_V309 are key residues for C30 binding to oligomer. Moreover, π-π interaction formed by L1P4_Y310 and L1P5_Y310 with C30 and the hydrogen bonding interaction formed by C30 with L3P3_Q307 are beneficial to the combination of C30 and oligomer. The fully understanding disrupt the mechanism of 2,4-thiazolidinedione derivative on PHF6 oligomer and the identification of binding sites will help design and discover new AD inhibitors in the future.


Oligopeptides , Thiazolidinediones , tau Proteins , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Oligopeptides/chemistry , Oligopeptides/metabolism , Poisson Distribution , Thermodynamics , Thiazolidinediones/chemistry , Thiazolidinediones/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
9.
Bioorg Chem ; 119: 105533, 2022 02.
Article En | MEDLINE | ID: mdl-34902647

A novel ANAP (Aspergillus niger from alkaline protease) catalyzed one pot three component approach in the synthesis of new thiazolidinedione festooned quinoline analogues via Knoevenagel condensation and N-alkylation have been reported. The catalytic effect of enzyme was monitored and optimized by adjusting various parameters including catalyst concentration, choice of solvent and temperature. The isolated alkaline protease exhibits favorable features for the reaction response such as the shorter reaction time, simple work-up procedure, clean reaction profiles and excellent product yields through reusability of the catalyst upto five cycles. In silico molecular docking simulations were carried out to find out the effective binding affinity of the synthesized quinoline analogues 4(a-i) towards PPARγ protein (Id-2XKW). In vitro α-amylase and α-glucosidase assays were performed for hypoglycemic activity evaluation. In vivo hypoglycemic studies carried out on streptozotocin (SZT) induced diabetic male albino rats have shown that compounds 4e and 4f significantly reduced blood glucose levels with percentage reduction of 43.7 ± 0.91 and 45.6 ± 0.28 at a concentration of 50 mg/kg body wt. The results obtained from molecular docking simulations and in vitro enzyme assays are in consistent with in-vivo studies which clearly demonstrated that out of the synthesized quinoline analogues, compounds 4e and 4f possess promising hypoglycemic activity which was on par to that of standards pioglitazone and rosiglitazone respectively.


Bacterial Proteins/metabolism , Diabetes Mellitus, Experimental/drug therapy , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Quinolines/pharmacology , Thiazolidinediones/pharmacology , Animals , Aspergillus niger/enzymology , Biocatalysis , Diabetes Mellitus, Experimental/chemically induced , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Male , Models, Molecular , Molecular Structure , Quinolines/chemistry , Quinolines/metabolism , Rats , Streptozocin , Structure-Activity Relationship , Thiazolidinediones/chemistry , Thiazolidinediones/metabolism , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
10.
Bioorg Chem ; 115: 105162, 2021 10.
Article En | MEDLINE | ID: mdl-34314919

A newly designed series of imidazolyl-methyl- l-2,4-thiazolidinediones 9 (a-m) were synthesized and In Silico studies were carried out to rationalize their anti-diabetic activity. Generally, all newly synthesized thiazolidinediones had anti-hyperglycemic activity compared with a diabetic-control group, without toxicity in 3T3 cells (viability ≥ 90%). These studies revealed that the compounds 9e and 9b (11∗10-6mol/kg) lowered blood glucose more effectively when compared to pioglitazone at the same dose. Following the administration of compound 9e, no weight gains or any serious side effects on liver and pancreas were observed. Moreover, the glucose consumption assay results showed a significant glucose-lowering effect (p < 0.001) in HepG2 cells, which were exposed to 11 mM of glucose at concentrations of 1.25-10 mM of compound 9e. Also, the PPAR-γ gene expression study revealed that pioglitazone and 9e showed similar behavior relative to the control group.


Drug Design , Hypoglycemic Agents/chemical synthesis , Thiazolidinediones/chemistry , 3T3 Cells , Animals , Binding Sites , Catalytic Domain , Cell Survival/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Glucose/metabolism , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Mice , Molecular Docking Simulation , PPAR gamma/agonists , PPAR gamma/metabolism , Pancreas/drug effects , Pancreas/metabolism , Pioglitazone/pharmacology , Rats , Structure-Activity Relationship , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
11.
Eur J Med Chem ; 223: 113630, 2021 Nov 05.
Article En | MEDLINE | ID: mdl-34175538

In search of dually active PPAR-modulators/aldose reductase (ALR2) inhibitors, 16 benzylidene thiazolidinedione derivatives, previously reported as partial PPARγ agonists, together with additional 18 structural congeners, were studied for aldose reductase inhibitory activity. While no compounds had dual property, our efforts led to the identification of promising inhibitors of ALR2. Eight compounds (11, 15-16, 20-24, 30) from the library of 33 compounds were identified as potent and selective inhibitors of ALR2. Compound 21 was the most effective and selective inhibitor with an IC50 value of 0.95 ± 0.11 and 13.52 ± 0.81 µM against ALR2 and aldehyde reductase (ALR1) enzymes, respectively. Molecular docking and dynamics studies were performed to understand inhibitor-enzyme interactions at the molecular level that determine the potency and selectivity. Compound 21 was further subjected to in silico and in vitro studies to evaluate the pharmacokinetic profile. Being less acidic (pKa = 9.8), the compound might have a superior plasma membrane permeability and reach the cytosolic ALR2. This fact together with excellent drug-likeness criteria points to improved bioavailability compared to the clinically used compound Epalrestat. The designed compounds represent a novel group of non-carboxylate inhibitors of aldose reductase with an improved physicochemical profile.


Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Thiazolidinediones/pharmacology , Aldehyde Reductase/chemistry , Aldehyde Reductase/metabolism , Animals , Caco-2 Cells , Catalytic Domain , Dogs , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Madin Darby Canine Kidney Cells , Male , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Rats, Wistar , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacokinetics
12.
Bioorg Chem ; 114: 104970, 2021 09.
Article En | MEDLINE | ID: mdl-34120026

Thiazolidinediones (TZD), benzopyrans are the proven scaffolds for inhibiting Aldose reductase (ALR2) activity and their structural confluence with the retention of necessary fragments helped in designing a series of hybrid compounds 2-(5-cycloalkylidene-2,4-dioxothiazolidin-3-yl)-N-(2-oxo-2H-chromen-3-yl)acetamide (10a-n) for better ALR2 inhibition. The compounds were synthesized by treating substituted 3-(N-bromoacetyl amino)coumarins (9a-d) with potassium salt of 5-cyclo alkylidene-1,3-thiazolidine-2,4-diones (4a-d). The inhibition activity against ALR2 with IC50 values range from 0.012 ± 0.001 to 0.056 ± 0.007 µM. N-[(6-Bromo-3-coumarinyl)-2-(5-cyclopentylidene-2,4-dioxothiazolidin-3-yl)] acetamide (10c) with cyclopentylidene group on one end and the 6-bromo group on the other end showed better inhibitory property (IC50 = 0.012 µM) and selectivity index (324.166) against the ALR2, a forty fold superiority over sorbinil, a better molecule over epalrestat and rest of the analogues exhibited a far superior response over sorbinil and slightly better as compared with epalrestat. It was further confirmed by the insilico studies that compound 10c showed best inhibition activity among the synthesized compounds with a high selectivity index against the ALR2. In invivo experiments, supplementation of compound 10c to STZ induced rats delayed the progression of cataract in a dose-dependent manner warranting its further development as a potential agent to treat thediabetic secondary complications especially cataract.


Aldehyde Reductase/antagonists & inhibitors , Coumarins/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Enzyme Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Thiazolidinediones/therapeutic use , Aldehyde Reductase/metabolism , Animals , Cataract/prevention & control , Coumarins/chemical synthesis , Coumarins/metabolism , Coumarins/pharmacokinetics , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Male , Molecular Docking Simulation , Molecular Structure , Protein Binding , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacokinetics
13.
J Med Chem ; 64(10): 6949-6971, 2021 05 27.
Article En | MEDLINE | ID: mdl-34006099

Monotargeting anticancer agents suffer from resistance and target nonspecificity concerns, which can be tackled with a multitargeting approach. The combined treatment with HDAC inhibitors and PPARγ agonists has displayed potential antitumor effects. Based on these observations, this work involves design and synthesis of molecules that can simultaneously target PPARγ and HDAC. Several out of 25 compounds inhibited HDAC4, and six compounds acted as dual-targeting agents. Compound 7i was the most potent, with activity toward PPARγ EC50 = 0.245 µM and HDAC4 IC50 = 1.1 µM. Additionally, compounds 7c and 7i were cytotoxic to CCRF-CEM cells (CC50 = 2.8 and 9.6 µM, respectively), induced apoptosis, and caused DNA fragmentation. Furthermore, compound 7c modulated the expression of c-Myc, cleaved caspase-3, and caused in vivo tumor regression in CCRF-CEM tumor xenografts. Thus, this study provides a basis for the rational design of dual/multitargeting agents that could be developed further as anticancer therapeutics.


Drug Design , Histone Deacetylases/metabolism , PPAR gamma/metabolism , Repressor Proteins/metabolism , Thiazolidinediones/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Mice , Mice, SCID , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/pathology , PPAR gamma/chemistry , PPAR gamma/genetics , Repressor Proteins/antagonists & inhibitors , Structure-Activity Relationship , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Transcriptional Activation/drug effects , Transplantation, Heterologous
14.
PLoS One ; 16(3): e0247619, 2021.
Article En | MEDLINE | ID: mdl-33661961

Thiazolidinediones are very important and used as a drug for the treatment of type 2 diabetes. Here, we report a convenient approach to synthesis 3-m-tolyl-5-arylidene-2,4-thiazolidinediones (TZDs) derivatives 7a-e in two steps with moderate to good yield using morpholine as a catalyst. All the structures were confirmed by their spectral IR, 1H NMR and 13C NMR data. The anti-diabatic activity of all synthesized molecules is evaluated by docking with peroxisome proliferator-activated receptor-γ (PPARγ). Preliminary flexible docking studies reveals that our compounds 7a, 7d and 7e showed better binding affinity with the protein and could be a potential candidate for the treatment of type 2 diabetes in near future.


Hypoglycemic Agents/chemistry , Morpholines/chemistry , Thiazolidinediones/chemistry , Thiazolidines/chemistry , Carbon-13 Magnetic Resonance Spectroscopy/methods , Catalysis , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Models, Chemical , Molecular Docking Simulation , Molecular Structure , PPAR gamma/chemistry , PPAR gamma/metabolism , Protein Binding , Proton Magnetic Resonance Spectroscopy/methods , Spectrophotometry, Infrared/methods , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/metabolism
15.
Life Sci ; 271: 119182, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33577851

Glitazones are synthetic derivatives of thiazolidinedione, and are designated as oral anti-diabetic agents, primarily acting on peroxisome proliferator-activated receptor-gamma (PPAR-γ) receptors and driving some crucial metabolic pathways linked to glucose and lipid metabolism at transcriptional level. Despite presenting adverse effects, including weight gain, fluid retention, prostate hyperplasia, hyperinsulinemia, and myocardial infarction, they are still preferred in clinical settings due to their utmost efficacy and selectivity. However, these complications kept glitazones restrained for long-term usage. The present review briefly highlights some important synthetic derivatives of thiazolidine2,4-dione and emphasizes the influence of various structural manipulations on their bio-efficacy.


Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Thiazolidinediones/chemistry , Thiazolidinediones/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/antagonists & inhibitors , Glucose/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , PPAR gamma/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Thiazolidinediones/therapeutic use
16.
Eur J Pharm Biopharm ; 156: 155-164, 2020 Nov.
Article En | MEDLINE | ID: mdl-32927077

During the last few decades, extensive efforts has been made to design nanocarriers to transport drugs into the central nervous system (CNS). However, its efficacy is limited due to the presence of the Blood-Brain Barrier (BBB) which greatly reduces drug penetration making Drug Delivery Systems (DDS) necessary. Polymeric nanoparticles (NPs) have been reported to be appropriate for this purpose and in particular, poly(lactic-co-glycolic acid) (PLGA) has been used for its ability to entrap small molecule drugs with great efficiency and the ease with which it functionalizes NPs. Despite the fact that their synthetic identity has been studied in depth, the biological identity of such manufactured polymers still remains unknown as does their biodistribution and in vivo fate. This biological identity is a result of their interaction with blood proteins, the so-called "protein corona" which tends to alter the behavior of polymeric nanoparticles in the body. The aim of the present research is to identify the proteins bounded to polymeric nanoparticles designed to selectively interact with the BBB. For this purpose, four different PLGA NPs were prepared and analyzed: (i) "PLGA@Drug," in which a model drug was encapsulated in its core; (ii) "8D3-PLGA" NPs where the PLGA surface was functionalized with a monoclonal anti-transferrin receptor antibody (8D3 mAb) in order to specifically target the BBB; (iii) "8D3-PLGA@Drug" in which the PLGA@Drug surface was functionalized using the same antibody described above and (iv) bare PLGA NPs which were used as a control. Once the anticipated protein corona NPs were obtained, proteins decorating both bare and functionalized PLGA NPs were isolated and analyzed. Apart from the indistinct interaction with PLGA NPs with the most abundant serum proteins, specific proteins could also be identified in the case of functionalized PLGA NPs. These findings may provide valuable insight into designing novel vehicles based on PLGA NPs for crossing the BBB.


Blood-Brain Barrier/metabolism , Nanoparticles/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/metabolism , Proteins/metabolism , Thiazolidinediones/metabolism , Blood-Brain Barrier/drug effects , Emulsions/chemical synthesis , Emulsions/metabolism , Humans , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Protein Transport/drug effects , Protein Transport/physiology , Proteins/chemical synthesis , Thiazolidinediones/chemical synthesis
17.
Bioorg Chem ; 100: 103927, 2020 07.
Article En | MEDLINE | ID: mdl-32422389

Protein tyrosine phosphatase 1B (PTP1B) is considered a potential therapeutic target for the treatment of type 2 diabetes mellitus (T2DM), since this enzyme plays a significant role to down-regulate insulin and leptin signalling and its over expression has been implicated in the development of insulin resistance, T2DM and obesity. Some thiazolidinediones (TZD) derivatives have been reported as promising PTP1B inhibitors with anti hyperglycemic effects. Recently, lobeglitazone, a new TZD, was described as an antidiabetic drug that targets the PPAR-γ (peroxisome γ proliferator-activated receptor) pathway, but no information on its effects on PTP1B have been reported to date. We investigated the effects of lobeglitazone on PTP1B activity in vitro. Surprisingly, lobeglitazone led to moderate inhibition on PTP1B (IC50 42.8 ± 3.8 µM) activity and to a non-competitive reversible mechanism of action. As lobeglitazone inhibits PTP1B activity in vitro, we speculate that it could also target PTP1B signalling pathway in vivo and thus contribute to potentiate its antidiabetic effects.


Hypoglycemic Agents/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Pyrimidines/chemistry , Thiazolidinediones/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Inhibitory Concentration 50 , Kinetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Pyrimidines/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
18.
Bioorg Chem ; 100: 103934, 2020 07.
Article En | MEDLINE | ID: mdl-32446120

Epigenetics plays a fundamental role in cancer progression, and developing agents that regulate epigenetics is crucial for cancer management. Among Class I and Class II HDACs, HDAC8 is one of the essential epigenetic players in cancer progression. Therefore, we designed, synthesized, purified, and structurally characterized novel compounds containing N-substituted TZD (P1-P25). Cell viability assay of all compounds on leukemic cell lines (CEM, K562, and KCL22) showed the cytotoxic potential of P8, P9, P10, P12, P19, and P25. In-vitro screening of different HDACs isoforms revealed that P19 was the most potent and selective inhibitor for HDAC8 (IC50 - 9.3 µM). Thermal shift analysis (TSA) confirmed the binding of P19 to HDAC8. In-vitro screening of all compounds on the transport activity of GLUT1, GLUT4, and GLUT5 indicated that P19 inhibited GLUT1 (IC50 - 28.2 µM). P10 and P19 induced apoptotic cell death in CEM cells (55.19% and 60.97% respectively) and P19 was less cytotoxic on normal WBCs (CC50 - 104.2 µM) and human fibroblasts (HS27) (CC50 - 105.0 µM). Thus, among this novel series of TZD derivatives, compound P19 was most promising HDAC8 inhibitor and cytotoxic on leukemic cells. Thus, P19 could serve as a lead for further development of optimized molecules with enhanced selectivity and potency.


Histone Deacetylase Inhibitors/metabolism , Repressor Proteins/antagonists & inhibitors , Thiazolidinediones/chemistry , Apoptosis/drug effects , Binding Sites , Cell Line , Cell Survival/drug effects , Drug Evaluation, Preclinical , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Molecular Docking Simulation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Structure-Activity Relationship , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
19.
Bioorg Chem ; 91: 103128, 2019 10.
Article En | MEDLINE | ID: mdl-31369977

α-Glucosidase is considered as a therapeutic target for the treatment of type 2 diabetes mellitus (DM2). In current study, we synthesized pyrrolidine-2,5-dione (succinimide) and thiazolidine-2,4-dione derivatives and evaluated for their ability to inhibit α-Glucosidase. Pyrrolidine-2,5-dione derivatives (11a-o) showed moderate to poor α-glucosidase inhibition. Compound 11o with the IC50 value of 28.3 ±â€¯0.28 µM emerged as a good inhibitor of α-glucosidase. Thiazolidine-2,4-dione and dihydropyrimidine (TZD-DHPM) hybrids (22a-c) showed excellent inhibitory activities. The most active compound 22a displayed IC50 value of 0.98 ±â€¯0.008 µM. Other two compounds of this series also showed activity in low micromolar range. The in-vivo antidiabetic study of three compounds 11n, 11o and 22a were also determined using alloxan induced diabetes mice model. Compounds 11o and 22a showed significant hypoglycemic effect compared to the reference drug. In-vivo acute toxicity study showed the safety of these selected compounds. In-silico docking studies were carried out to rationalize the in-vitro results. The binding modes and bioassay results of TZD-DHPM hybrids showed that interactions with important residues appeared significant for high potency.


Antioxidants/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Glycoside Hydrolase Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Succinimides/therapeutic use , Thiazolidinediones/therapeutic use , Alloxan , Animals , Antioxidants/chemical synthesis , Antioxidants/metabolism , Catalytic Domain , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/metabolism , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Mice , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Succinimides/chemical synthesis , Succinimides/metabolism , Thiazolidinediones/chemical synthesis , Thiazolidinediones/metabolism , alpha-Glucosidases/chemistry , alpha-Glucosidases/metabolism
20.
Bioorg Chem ; 89: 102993, 2019 08.
Article En | MEDLINE | ID: mdl-31129500

A series of thirteen novel 2,4-thiazolidinedione derivatives were synthesized through three step reaction procedure. The title compounds were synthesized by Knoevenagel condensation at the 5th position of the 2,4-thiazolidinedione ring. Various physicochemical and spectral studies were conducted to characterize the synthesized derivatives including- IR, Mass, 1H NMR, 13C NMR and elemental analysis. The derivatives were screened for in vivo anti diabetic, in vivo anti-inflammatory and in vitro free radical scavenging activities by carrageenan induced rat paw edema method, alloxan induced diabetes in wistar rats method and FRAP (ferric reducing antioxidant power) method respectively. Some of the derivatives emerged out as potent antidiabetic, anti inflammatory and free radical scavenging agents. Molecular docking was carried out to investigate some possible structural insights into the potential binding patterns of the most potent anti-diabetic molecules NB7,NB12 and NB13 with the active sites of target PPARγ (PDB ID: 2PRG) using MOE software. Dichloro derivative compound NB-7 has shown great potential in the present study as it not only has maximum antidiabetic activity but also possess excellent anti-inflammatory and antioxidant potential.


Thiazolidinediones/chemistry , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Antioxidants/chemistry , Binding Sites , Blood Glucose/analysis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Edema/chemically induced , Edema/drug therapy , Edema/pathology , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Molecular Docking Simulation , PPAR gamma/chemistry , PPAR gamma/metabolism , Protein Structure, Tertiary , Rats , Rats, Wistar , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/metabolism
...