Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 429, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594496

ABSTRACT

The study aims to explore the effect of PPARγ signaling on ferroptosis and preeclampsia (PE) development. Serum and placental tissue are collected from healthy subjects and PE patients. The PPARγ and Nrf2 decreases in the PE. Rosiglitazone intervention reverses hypoxia-induced trophoblast ferroptosis and decreases lipid synthesis by regulating Nfr2 and SREBP1. Compared to the Hypoxia group, the migratory and invasive abilities enhance after rosiglitazone and ferr1 treatment. Rosiglitazone reduces the effect of hypoxia and erastin. The si-Nrf2 treatment attenuats the effects of rosiglitazone on proliferation, migration, and invasion. The si-Nrf2 does not affect SREBP1 expression. PPARγ agonists alleviates ferroptosis in the placenta of the PE rats. The study confirms that PPARγ signaling and ferroptosis-related indicators were dysregulated in PE. PPARγ/Nrf2 signaling affects ferroptosis by regulating lipid oxidation rather than SREBP1-mediated lipid synthesis. In conclusion, our study find that PPARγ can alleviate PE development by regulating lipid oxidation and ferroptosis.


Subject(s)
Ferroptosis , Pre-Eclampsia , Humans , Female , Pregnancy , Rats , Animals , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , PPAR gamma/metabolism , Lipid Metabolism , Placenta/metabolism , Pre-Eclampsia/drug therapy , Pre-Eclampsia/prevention & control , Pre-Eclampsia/metabolism , NF-E2-Related Factor 2/metabolism , Hypoxia/metabolism , Lipids
2.
Planta Med ; 90(5): 388-396, 2024 May.
Article in English | MEDLINE | ID: mdl-38490239

ABSTRACT

Diabetes mellitus, linked with insulin resistance and hyperglycaemia, is a leading cause of mortality. Glucose uptake through glucose transporter type 4, especially in skeletal muscle, is crucial for maintaining euglycaemia and is a key pathway targeted by antidiabetic medication. Abrus precatorius is a medicinal plant with demonstrated antihyperglycaemic activity in animal models, but its mechanisms are unclear.This study evaluated the effect of a 50% ethanolic (v/v) A. precatorius leaf extract on (1) insulin-stimulated glucose uptake and (2) related gene expression in differentiated C2C12 myotubes using rosiglitazone as a positive control, and (3) generated a comprehensive phytochemical profile of A. precatorius leaf extract using liquid chromatography-high resolution mass spectrometry to elucidate its antidiabetic compounds. A. precatorius leaf extract significantly increased insulin-stimulated glucose uptake, and insulin receptor substrate 1 and Akt substrate of 160 kDa gene expression; however, it had no effect on glucose transporter type 4 gene expression. At 250 µg/mL A. precatorius leaf extract, the increase in glucose uptake was significantly higher than 1 µM rosiglitazone. Fifty-five phytochemicals (primarily polyphenols, triterpenoids, saponins, and alkaloids) were putatively identified, including 24 that have not previously been reported from A. precatorius leaves. Abrusin, precatorin I, glycyrrhizin, hemiphloin, isohemiphloin, hispidulin 4'-O-ß-D-glucopyranoside, homoplantaginin, and cirsimaritin were putatively identified as known major compounds previously reported from A. precatorius leaf extract. A. precatorius leaves contain antidiabetic phytochemicals and enhance insulin-stimulated glucose uptake in myotubes via the protein kinase B/phosphoinositide 3-kinase pathway by regulating insulin receptor substrate 1 and Akt substrate of 160 kDa gene expression. Therefore, A. precatorius leaves may improve skeletal muscle insulin sensitivity and hyperglycaemia. Additionally, it is a valuable source of bioactive phytochemicals with potential therapeutic use for diabetes.


Subject(s)
Abrus , Diabetes Mellitus , Hyperglycemia , Insulin Resistance , Animals , Insulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Abrus/chemistry , Insulin Receptor Substrate Proteins/metabolism , Rosiglitazone/metabolism , Rosiglitazone/pharmacology , Glucose Transporter Type 4 , Phosphatidylinositol 3-Kinases , Muscle, Skeletal/metabolism , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Plant Extracts/chemistry , Glucose/pharmacology
3.
Environ Pollut ; 347: 123761, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38467365

ABSTRACT

Adipose tissue compromises one of the principal depots where brominated flame retardants (BFR) accumulate in vivo, yet whether BFR disturb thermogenic brown/beige adipocytes is still not referred to date. Herein, effects of BDE-99, a major congener of polybrominated diphenyl ethers (PBDEs) detected in humans, on brown/beige adipocytes were explored for the first time, aiming to provide new knowledge evaluating the obesogenic and metabolic disrupting effects of BFR. Our results firstly demonstrated that exposure to BDE-99 during the lineage commitment period significantly promoted C3H10T1/2 MSCs differentiating into brown/beige adipocytes, evidenced by the increase of brown/beige adipocyte marker UCP1, Cidea as well as mitochondrial membrane potential and basal respiration rate, which was similar to pharmacological PPARγ agonist rosiglitazone. Unexpectedly, the mitochondrial maximal respiration rate of BDE-99 stimulated brown/beige adipocytes was not synchronously enhanced and resulted in a significant reduction of mitochondrial spare respiration capacity (SRC) compared to control or rosiglitazone stimulated adipocytes, indicating a deficient energy-dissipating capacity of BDE-99 stimulated thermogenic adipocytes. Consistently with compromised mitochondrial SRC, lipidomic analysis further revealed that the lipids profile of mitochondria derived from BDE-99 stimulated brown/beige adipocytes were quite different from control or rosiglitazone stimulated cells. In detail, BDE-99 group contains more free fatty acid (FFA) and lyso-PE in mitochondria. In addition to energy metabolism, our results also demonstrated that BDE-99 stimulated brown/beige adipocytes were deficient in endocrine, which secreted more adverse adipokine named resistin, coinciding with comparable beneficial adipokine adiponectin compared with that of rosiglitazone. Taken together, our results showed for the first time that BDE-99 stimulated brown/beige adipocytes were aberrant in energy metabolism and endocrine, which strongly suggests that BDE-99 accumulated in human adipose tissue could interfere with brown/beige adipocytes to contribute to the occurrence of obesity and relevant metabolic disorders.


Subject(s)
Adipocytes, Beige , Humans , Adipocytes, Beige/metabolism , Halogenated Diphenyl Ethers/metabolism , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Adipocytes, Brown/metabolism , Adipokines
4.
J Complement Integr Med ; 21(1): 123-130, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38253264

ABSTRACT

OBJECTIVES: This study aimed to investigate the antioxidant effect of rosiglitazone (ROG) and pioglitazone (POG) on oxidative damage and dysfunction of hepatic tissue in hypothyroid rats. METHODS: The male rats were classified into six groups: (1) Control; (2) Hypothyroid, (3) Hypothyroid-POG 10, (4) Hypothyroid-POG 20, (5) Hypothyroid-ROG 2, and (6) Hypothyroid-ROG 4. To induction hypothyroidism in rats, propylthiouracil (PTU) (0.05 %w/v) was added to drinking water. In groups 2-6, besides PTU, the rats were also intraperitoneal administrated with 10 or 20 mg/kg POG or 2 or 4 mg/kg ROG for six weeks. Finally, after deep anesthesia, the blood was collected to measure the serum biochemical markers and hepatic tissue was separated for biochemical oxidative stress markers. RESULTS: Administration of PTU significantly reduced serum thyroxin concentration, total thiol levels, activity of superoxide dismutase (SOD) and catalase (CAT) enzymes, and increased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (Alk-P) and malondialdehyde (MDA) in the liver. Additionally, our results showed that prescription of POG or ROG for six weeks to hypothyroid rats resulted in an improvement in liver dysfunction (decrease in serum levels of AST, ALT, and ALK-P) through reducing oxidative damage in hepatic tissue (increase in CAT, SOD, or total thiols and decrease in MDA levels). CONCLUSIONS: The findings of the present study presented that the IP administration of POG and ROG for six weeks improves liver dysfunction induced by hypothyroidism in juvenile rats by reducing oxidative damage.


Subject(s)
Hypothyroidism , Liver Diseases , Rats , Animals , Male , Pioglitazone/adverse effects , Pioglitazone/metabolism , Rosiglitazone/adverse effects , Rosiglitazone/metabolism , Rats, Wistar , Hypothyroidism/drug therapy , Antioxidants/pharmacology , Antioxidants/therapeutic use , Oxidative Stress , Propylthiouracil/adverse effects , Propylthiouracil/metabolism , Superoxide Dismutase/metabolism , Liver , Receptor Protein-Tyrosine Kinases/adverse effects , Receptor Protein-Tyrosine Kinases/metabolism
5.
J Ethnopharmacol ; 321: 117550, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38065350

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Vascular endothelial cell senescence is associated with cardiovascular complications in diabetes. Essential oil from Fructus Alpiniae zerumbet (Pers.) B.L.Burtt & R.M.Sm. (EOFAZ) has potentially beneficial and promising diabetes-related vascular endothelial cell senescence-mitigating effects; however, the underlying molecular mechanisms remain unclear. AIM OF THE STUDY: To investigate the molecular effects of EOFAZ on vascular endothelial cell senescence in diabetes. MATERIALS AND METHODS: A diabetes mouse model was developed using a high-fat and high-glucose diet (HFD) combined with intraperitoneal injection of low-dose streptozotocin (STZ, 30 mg/kg) and oral treatment with EOFAZ. 4D label-free quantitative proteomics, network pharmacology, and molecular docking techniques were employed to explore the molecular mechanisms via which EOFAZ alleviates diabetes-related vascular endothelial cell senescence. A human aortic endothelial cells (HAECs) senescence model was developed using high palmitic acid and high glucose (PA/HG) concentrations in vitro. Western blotting, immunofluorescence, SA-ß-galactosidase staining, cell cycle, reactive oxygen species (ROS), cell migration, and enzyme linked immunosorbent assays were performed to determine the protective role of EOFAZ against vascular endothelial cell senescence in diabetes. Moreover, the PPAR-γ agonist rosiglitazone, inhibitor GW9662, and siRNA were used to verify the underlying mechanism by which EOFAZ combats vascular endothelial cell senescence in diabetes. RESULTS: EOFAZ treatment ameliorated abnormal lipid metabolism, vascular histopathological damage, and vascular endothelial aging in diabetic mice. Proteomics and network pharmacology analysis revealed that the differentially expressed proteins (DEPs) and drug-disease targets were associated with the peroxisome proliferator-activated receptor gamma (PPAR-γ) signalling pathway, a key player in vascular endothelial cell senescence. Molecular docking indicated that the small-molecule compounds in EOFAZ had a high affinity for the PPAR-γ protein. Western blotting and immunofluorescence analyses confirmed the significance of DEPs and the involvement of the PPAR-γ signalling pathway. In vitro, EOFAZ and rosiglitazone treatment reversed the effects of PA/HG on the number of senescent endothelial cells, expression of senescence-related proteins, the proportion of cells in the G0/G1 phase, ROS levels, cell migration rate, and expression of pro-inflammatory factors. The protective effects of EOFAZ against vascular endothelial cell senescence in diabetes were aborted following treatment with GW9662 or PPAR-γ siRNA. CONCLUSIONS: EOFAZ ameliorates vascular endothelial cell senescence in diabetes by activating PPAR-γ signalling. The results of the present study highlight the potential beneficial and promising therapeutic effects of EOFAZ and provide a basis for its clinical application in diabetes-related vascular endothelial cell senescence.


Subject(s)
Diabetes Mellitus, Experimental , Oils, Volatile , Humans , Mice , Animals , Endothelial Cells , PPAR gamma/metabolism , Rosiglitazone/metabolism , Rosiglitazone/pharmacology , Reactive Oxygen Species/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Oils, Volatile/pharmacology , Molecular Docking Simulation , Network Pharmacology , Proteomics , RNA, Small Interfering , Glucose/metabolism
6.
Obesity (Silver Spring) ; 32(1): 70-79, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37929774

ABSTRACT

OBJECTIVE: This study investigated remodeling of cellular metabolism and structures during browning of primary human adipocytes derived from both visceral and subcutaneous adipose tissues. Effects of glucocorticoids on the browning were also assessed. METHODS: Differentiated omental and subcutaneous human adipocytes were treated with rosiglitazone, with or without dexamethasone, and expression levels of brite adipocyte markers, lipolysis, and lipid droplet and mitochondrial structures were examined. RESULTS: Both omental and subcutaneous adipocytes acquired brite phenotypes upon peroxisome proliferator-activated receptor-γ agonist treatment, and dexamethasone tended to enhance the remodeling. Although rosiglitazone increased lipolysis during treatment, brite adipocytes exhibited lower basal lipolytic rates and enhanced responses to ß-adrenergic agonists or atrial natriuretic peptide. Transcriptome analysis identified induction of both breakdown and biosynthesis of lipids in brite adipocytes. After 60+ days in culture, lipid droplet size increased to ~50 microns, becoming almost unilocular in control adipocytes, and after browning, they acquired paucilocular morphology, clusters of small lipid droplets (1-2 micron) surrounded by mitochondria appearing on the periphery of the central large one. CONCLUSIONS: Metabolic and structural remodeling during browning of primary human adipocytes is similar to previous findings in human adipocytes in vivo, supporting their uses for mechanical studies investigating browning with translational relevance.


Subject(s)
Adipocytes , Subcutaneous Fat , Humans , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Adipocytes/metabolism , Subcutaneous Fat/metabolism , Lipolysis , Dexamethasone
7.
Clin Epigenetics ; 15(1): 178, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37932845

ABSTRACT

The role of PRKAG2 in the maintenance of heart function is well established, but little is known about how PRKAG2 is regulated in cardiomyocytes. In this study, we investigated the role of the lncRNA PRKAG2-AS, which is present at the PRKAG2 promoter, in the regulation of PRKAG2 expression. PRKAG2-AS expression was predominantly nuclear, as determined by RNA nucleoplasmic separation and fluorescence in situ hybridization. Knockdown of PRKAG2-AS in the nucleus, but not the cytoplasm, significantly decreased the expression of PRKAG2b and PRKAG2d. Interestingly, we found that PRKAG2-AS and its target genes, PRKAG2b and PRKAG2d, were reduced in the hearts of patients with ischemic cardiomyopathy, suggesting a potential role for PRKAG2-AS in myocardial ischemia. Indeed, knockdown of PRKAG2-AS in the nucleus resulted in apoptosis of cardiomyocytes. We further elucidated the mechanism by which PRKAG2-AS regulates PRKAG2 transcription by identifying 58 PRKAG2-AS interacting proteins. Among them, PPARG was selected for further investigation based on its correlation and potential interaction with PRKAG2-AS in regulating transcription. Overexpression of PPARG, or its activation with rosiglitazone, led to a significant increase in the expression of PRKAG2b and PRKAG2d in cardiomyocytes, which could be attenuated by PRKAG2-AS knockdown. This finding suggests that PRKAG2-AS mediates, at least partially, the protective effects of rosiglitazone on hypoxia-induced apoptosis. However, given the risk of rosiglitazone in heart failure, we also examined the involvement of PRKAG2-AS in this condition and found that PRKAG2-AS, as well as PRKAG2b and PRKAG2d, was elevated in hearts with dilated cardiomyopathy (DCM) and that overexpression of PRKAG2-AS led to a significant increase in PRKAG2b and PRKAG2d expression, indicating that up-regulation of PRKAG2-AS may contribute to the mechanism of heart failure by promoting transcription of PRKAG2. Consequently, proper expression of PRKAG2-AS is essential for maintaining cardiomyocyte function, and aberrant PRKAG2-AS expression induced by hypoxia or other stimuli may cause cardiac dysfunction.


Subject(s)
AMP-Activated Protein Kinases , Heart Failure , Myocardial Ischemia , PPAR gamma , RNA, Long Noncoding , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Apoptosis , DNA Methylation , Heart Failure/genetics , Hypoxia , In Situ Hybridization, Fluorescence , Myocytes, Cardiac/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Rosiglitazone/metabolism , RNA, Long Noncoding/genetics
8.
BMC Urol ; 23(1): 169, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37875848

ABSTRACT

BACKGROUND: Dysregulation of the terminal differentiation of bladder urothelium is associated with the pathogenesis of urinary tract disorders. Fibroblast growth factor (Fgf)7 and Fgf10 stimulate urothelial proliferation; however, their roles in cellular differentiation remain unclear. In this study, we used an organoid system to investigate the roles of these Fgfs in regulating bladder urothelium differentiation and identify their distribution patterns in the mouse bladder. METHODS: Adult bladder epithelia (AdBE) isolated from adult mouse bladder tissues (AdBTs) were used to culture adult bladder organoids (AdBOs) in the presence of Fgf7 and Fgf10. The differentiation status of the cells in AdBTs, AdBEs, AdBOs, and neonatal bladder tissues (NeoBTs) was analyzed via quantitative real-time-PCR for the presence of undifferentiated cell markers (Krt5, Trp63, and Krt14) and differentiated cell markers (Krt20, Upk1a, Upk2, and Upk3a). Organoid cell proliferation was assessed by counting cell numbers using the trypan blue method. The effects of Fgf7 and Fgf10 on organoid differentiation were assessed using different doses of Fgfs, and the involvement of peroxisome proliferator-activated receptor γ (PPARγ) signaling in these processes was tested by introducing a PPARγ agonist (Rosiglitazone) and antagonist (T0070907) to the culture. The expression patterns of Fgf7 and Fgf10 were examined via in situ hybridization of AdBTs. RESULTS: AdBOs showed higher expression of undifferentiated cell markers and lower expression of differentiated cell markers than AdBTs, NeoBTs, and AdBEs, indicating the relatively immature state of AdBOs. Differentiation of AdBOs was enhanced by Rosiglitazone and Fgf7, suggesting an interplay of intracellular signals between Fgf7 and PPARγ. Co-addition of T0070907 suppressed Fgf7-mediated differentiation, demonstrating that PPARγ is activated downstream of Fgf7 to promote cellular differentiation into umbrella cells. Furthermore, we found that Fgf7 is predominantly expressed in the umbrella cells of the urothelium, whereas Fgf10 is predominantly expressed in the urothelium and stroma of AdBTs. CONCLUSIONS: We demonstrated that unlike Fgf10, Fgf7 induces cellular differentiation via PPARγ activity and has a unique tissue distribution pattern in the adult bladder. Further studies on the Fgf7-PPARγ signaling axis would provide insights into the differentiation mechanisms toward functional umbrella cells and the pathogenesis of several urinary tract diseases.


Subject(s)
PPAR gamma , Urinary Bladder , Mice , Animals , PPAR gamma/metabolism , Rosiglitazone/metabolism , Urothelium/metabolism , Cell Differentiation , Organoids , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 7/metabolism , Uroplakin III/metabolism
9.
Brain Behav ; 13(12): e3275, 2023 12.
Article in English | MEDLINE | ID: mdl-37837628

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) seriously threatens the health of people. In addition, microglia M1 polarization was confirmed to be involved in the progression of ICH. Rosiglitazone was able to be used as an antidiabetic agent, which could activate PPAR-γ, and PPAR-γ was reported to inhibit inflammation in microglia. However, the detailed function of Rosiglitazone in ICH remains unclear. METHODS: In vivo and in vitro experiments were used to test the function of Rosiglitazone in ICH. In addition, RT-qPCR and western blot were performed to evaluate the mRNA and protein level of PPAR-γ, respectively. Immunofluorescence staining was performed to detect the levels of CD206 and CD86, and ELISA was used to measure the levels of pro-inflammatory cytokines. RESULTS: PPAR-γ was downregulated in ICH mice, whereas p-JNK and p-STAT3 were upregulated. Thrombin notably downregulated the level of PPAR-γ in BV2 cells, whereas Rosiglitazone partially reversed this phenomenon. In addition, Rosiglitazone markedly reversed thrombin-induced microglia M1 polarization. Consistently, thrombin-induced inflammatory response in BV2 cells was abolished in the presence of Rosiglitazone. SP600125 (JNK/STAT3 inhibitor) greatly reversed thrombin-induced M1 polarization in microglia, and GW9662 abolished the effect of SP600125. Meanwhile, Rosiglitazone could inactivate JNK/STAT3 pathway through the upregulation of PPAR-γ. Furthermore, Rosiglitazone notably alleviated the symptom of ICH in vivo through inhibiting the apoptosis and mediating PPAR-γ/JNK/STAT3 axis. CONCLUSION: Rosiglitazone could attenuate the inflammation in ICH through inhibiting microglia M1 polarization. Thus, our research would shed now lights on exploring new therapeutic strategies against ICH.


Subject(s)
Microglia , Thrombin , Humans , Mice , Animals , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Rosiglitazone/therapeutic use , Thrombin/metabolism , Thrombin/pharmacology , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Signal Transduction , Inflammation/drug therapy , Inflammation/metabolism , STAT3 Transcription Factor/metabolism
10.
Nutrients ; 15(7)2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37049636

ABSTRACT

Shikonin, a natural ingredient produced by Lithospermum erythrorhizon, has anti-inflammatory, anti-cancer, and anti-obesity effects. It also inhibits adipocyte differentiation; however, the underlying molecular and epigenetic mechanisms remain unclear. We performed RNA-sequencing of shikonin-treated 3T3-L1 cells. Gene ontology and gene set enrichment analysis showed that shikonin is significantly associated with genes related to adipogenesis, histone modification, and PPARγ. Shikonin treatment downregulated the mRNA expression of PPARγ-responsive genes and rosiglitazone-induced transcriptional activity of PPARγ. Microscale thermophoresis assays showed a KD value 1.4 ± 0.13 µM for binding between shikonin and PPARγ. Glutathione S-transferase pull-down assays exhibited that shikonin blocked the rosiglitazone-dependent association of PPARγ with its coactivator CBP. In addition, shikonin decreased the enrichment of the active histone code H3K4me3 and increased the repressive code H3K27me3 of PPARγ target promoters. Shikonin is a PPARγ antagonist that suppresses adipogenesis by regulating the enrichment of histone codes during adipogenesis. Therefore, it may be used to treat obesity-related disorders via epigenetic changes.


Subject(s)
Histones , PPAR gamma , Mice , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Histones/metabolism , Rosiglitazone/metabolism , Rosiglitazone/pharmacology , Methylation , Adipocytes , Adipogenesis , Cell Differentiation , 3T3-L1 Cells
11.
Immunol Cell Biol ; 101(5): 428-443, 2023 05.
Article in English | MEDLINE | ID: mdl-36918728

ABSTRACT

Macrophages exhibit a range of functional pro- and anti-inflammatory states that induce changes in their cellular metabolism. We aimed to elucidate whether these changes affect the molecular properties of their circadian clock focusing on their anti-inflammatory phenotype. Primary cell cultures of bone marrow-derived macrophages (BMDMs; nonpolarized M0 BMDM) from PER2::LUC (fusion protein of PERIOD2 and LUCIFERASE) mice were polarized into the M1 (proinflammatory) or M2 (anti-inflammatory) phenotype, and PER2-driven bioluminescence was recorded in real-time at the cell-population and single-cell levels. Viability, clock gene expression profiles, polarization plasticity and peroxisome proliferator-activated receptor γ (PPARγ) protein levels were analyzed. The effects of pharmacological activation/inhibition of PPARγ (rosiglitazone/GW9662) and inhibition of fatty acid oxidation (FAO) by etomoxir in M2 BMDM cell cultures were examined. The parameters of PER2-driven bioluminescence rhythms differed between M0, M1 and M2 BMDM cultures at cell-population and single-cell levels. Compared with M0, polarization to M2 did not change the period but increased amplitude, mean bioluminescence level and rhythm persistence. Polarization to M1 shortened the period but had no effect on the amplitude of the rhythm. The same period changes were observed after a bidirectional switch between M1- and M2-polarized states in the same culture. Both PPARγ activation/inhibition and FAO inhibition modulated the clock in M2 BMDMs, suggesting metabolic regulation of the M2 clock. Our results indicate that bidirectional changes in the properties of BMDM circadian clocks in response to their actual polarization are mediated via changes in their metabolic state. They provide new information on the interrelationship between the BMDM polarization, their circadian clock and cellular metabolism.


Subject(s)
Circadian Clocks , Mice , Animals , PPAR gamma/metabolism , Macrophages/metabolism , Rosiglitazone/metabolism , Anti-Inflammatory Agents/metabolism
12.
Article in English | MEDLINE | ID: mdl-36972780

ABSTRACT

Methamphetamine (METH) is a highly addictive psychostimulant. The adipocyte-derived hormone adiponectin has a broad spectrum of functions in the brain. However, limited research has been conducted on the effect of adiponectin signaling on METH-induced conditioned place preference (CPP) and knowledge of the underlying neural mechanisms is scarce. The METH induced adult male C57/BL6J mice model were used for testing the therapeutic activities of intraperitoneal injection of AdipoR agonist AdipoRon and peroxisome proliferator-activated receptor gamma (PPARγ)-selective agonist rosiglitazone, adiponectin receptor 1 (AdipoR1) overexpression in hippocampal dentate gyrus (DG), and chemogenetic inhibiting the neural activity of DG, and the changes of neurotrophic factors, synaptic molecules, glutamate receptors, and inflammatory cytokines were also measured. We found that adiponectin expression was significantly reduced in METH addicted patients and mice. Our findings also showed that injection of AdipoRon or rosiglitazone alleviated the METH-induced CPP behavior. Moreover, the expression of AdipoR1 in the hippocampus was also reduced, and AdipoR1 overexpression blocked the development of METH-induced CPP behavior through regulatory effects on neurotrophic factors, synaptic molecules, and glutamate receptors. The observed inhibitory neural activity of the hippocampal dentate gyrus (DG) induced via a chemogenetic approach produced a therapeutic effect on the METH-induced CPP behavior. Finally, we identified an abnormal expression of some key inflammatory cytokines through the PPARγ/Adiponectin/AdipoR1 axis. This study demonstrates that adiponectin signaling is a promising diagnostic and therapeutic target for METH addiction.


Subject(s)
Central Nervous System Stimulants , Methamphetamine , Male , Mice , Animals , Methamphetamine/pharmacology , PPAR gamma/metabolism , Adiponectin , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/metabolism , Hippocampus/metabolism , Signal Transduction , Cytokines/metabolism
13.
Sleep Breath ; 27(3): 1155-1164, 2023 06.
Article in English | MEDLINE | ID: mdl-35943692

ABSTRACT

PURPOSE: Patients with obstructive sleep apnoea (OSA) have a high incidence of vascular endothelial injury. The most important pathophysiological feature of OSA is chronic intermittent hypoxia (CIH). This study aimed to investigate the mechanisms of CIH-related vascular endothelial injury. METHODS: IH exposure was applied to human umbilical vein endothelial cells (HUVECs). After modeling, cell viability, the expression levels of peroxisome proliferator activated receptor γ (PPARγ), apoptosis-associated proteins and mitochondrial division fusion proteins, and the levels of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were assessed via Cell Counting Kit-8 (CCK-8), western blotting, fluorescent microscope, and flow cytometry, respectively. Rosiglitazone (PPARγ agonist), tempo (the mitochondrial-specific antioxidant), and tempo combined with PPARγ interfering RNA were used to treat HUVECs, respectively. RESULTS: After IH exposure, cell viability and levels of MMP decreased, cell apoptosis and ROS levels increased, and the expression levels of PPARγ decreased. Both tempo and rosiglitazone pretreatment ameliorated cell apoptosis and improved cell viability. In addition, mitochondrial function became better after tempo pretreatment. PPARγ interference reversed the protective effects of tempo on IH-related mitochondrial function injury and cell injury. CONCLUSIONS: PPARγ regulated the apoptosis and cell viability of IH-treated HUVECs by altering mitochondrial function. This finding clarifies the mechanism of CIH-related vascular endothelial injury.


Subject(s)
PPAR gamma , Sleep Apnea, Obstructive , Humans , Human Umbilical Vein Endothelial Cells/metabolism , PPAR gamma/genetics , Reactive Oxygen Species/metabolism , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Hypoxia/metabolism , Sleep Apnea, Obstructive/metabolism , Apoptosis
14.
Acta Pharmacol Sin ; 44(2): 454-464, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35918412

ABSTRACT

Rheumatoid arthritis (RA) is characterized by synovial inflammation, synoviocyte expansion and damage to cartilage and bone. We recently reported that peroxisome proliferator-activated receptor (PPAR)-γ inhibited the proliferation and activation of fibroblast-like synoviocytes (FLS), and was downregulated in RA synovial. In this study we investigated the role of PPAR-γ in RA and the underlying mechanisms. Adjuvant-induced arthritis (AIA) was induced in rats; from D15, AIA rats were orally administered pioglitazone (30 mg·kg-1·d-1) or rosiglitazone (4 mg·kg-1·d-1) for 14 days. Collagen-induced arthritis (CIA) was induced in wild-type and Ppar-γ+/- mice. We showed that the expression of PPAR-γ was significantly reduced, whereas that of TNF-α was markedly increased in human RA FLS. In CIA mice, knockdown of PPAR-γ expression (Ppar-γ+/-) aggravated the ankle inflammation. Similarly, T0070907 (a PPAR-γ antagonist) or si-PPAR-γ promoted the activation and inflammation of TNF-α-induced FLS in vitro. On the contrary, administration of PPAR-γ agonist pioglitazone or rosiglitazone, or injection of ad-Ppar-γ into the ankle of AIA rat in vivo induced overexpression of PPAR-γ, reduced the paw swelling and inflammation, and downregulated activation and inflammation of FLS in RA. Interesting, injection of ad-Ppar-γ into the ankle also reversed the ankle inflammation in Ppar-γ+/- CIA mice. We conducted RNA-sequencing and KEGG pathway analysis, and revealed that PPAR-γ overexpression was closely related to p53 signaling pathway in TNF-α-induced FLS. Co-IP study confirmed that p53 protein was bound to PPAR-γ in RA FLS. Taken together, PPAR-γ alleviates the inflammatory response of TNF-α-induced FLS by binding p53 in RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Synoviocytes , Rats , Mice , Humans , Animals , Synoviocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , PPAR gamma/metabolism , Rosiglitazone/pharmacology , Rosiglitazone/therapeutic use , Rosiglitazone/metabolism , Pioglitazone/pharmacology , Pioglitazone/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Proliferation , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Fibroblasts/metabolism , Cells, Cultured , Synovial Membrane/metabolism
15.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(12): 1111-1117, 2022 Dec.
Article in Chinese | MEDLINE | ID: mdl-36585234

ABSTRACT

Objective To investigate the effect of Achyranthes bidentata polysaccharides (ABPS) on adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and its mechanism. Methods Five SD rats were sacrificed, and the BMSCs were dissected and isolated. The BMSCs were adherently cultured to logarithmic growth phase after identification, and treated with different doses of ABPS for 48 hours. The cell survival rates were detected by MTT assay. The highest dose of ABPS without toxicity to BMSCs was selected for subsequent experiments. Cells were randomly divided into control group, ABPS group, rosiglitazone group and ABPS combined with rosiglitazone group. Cell survival rates were detected by MTT assay. Triglyceride (TG) levels in BMSCs were detected by spectrophotometry. Lipid droplet formation in BMSCs was observed by oil red O staining. The mRNA and protein expression of peroxisome proliferater-activated receptor γ (PPARγ), transient receptor potential vanilloid 4 (TRPV4) and CCAAT/enhancer binding protein α (C/EBPα) were detected by real time quantitative PCR and Western blot analysis. Results The dose of ABPS≤200 mg/L had no obvious toxic effect on the growth of BMSCs after 48 hours, and the cell survival rate of 400 mg/L ABPS group was lower. Compared with the control group, the ABPS group showed decreased levels in TG, decreased relative expression of PPARγ, TRPV4 and C/EBPα mRNA and protein, and the decreased number of cytoplasmic lipid droplets. In the rosiglitazone group, observation reported the decreased cell survival rate, increased TG level, increased relative expression levels of PPARγ, TRPV4 and C/EBPα mRNA and protein, along with the increased number of cytoplasmic lipid droplets. Compared with the ABPS group, the cell survival rate was decreased, TG level was increased, the relative expression levels of PPARγ, TRPV4 and C/EBPα mRNA and protein increased, and the number of cytoplasmic lipid droplets increased in the ABPS combined with rosiglitazone group. Compared with rosiglitazone group, the survival rate was increased, TG level was decreased, the relative expression levels of PPARγ, TRPV4 and C/EBPα mRNA and protein were decreased, and the number of cytoplasmic lipid droplets was decreased in the ABPS combined with rosiglitazone group. Conclusion ABPS can inhibit adipogenic differentiation of BMSCs, and the mechanism may be related to the regulation of PPARγ/TRPV4 pathway.


Subject(s)
Achyranthes , Antineoplastic Agents , Mesenchymal Stem Cells , Rats , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Achyranthes/genetics , Achyranthes/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/pharmacology , Rosiglitazone/pharmacology , Rosiglitazone/metabolism , Peroxisomes/metabolism , Rats, Sprague-Dawley , Cell Differentiation , Adipogenesis , CCAAT-Enhancer-Binding Protein-alpha/genetics , Antineoplastic Agents/pharmacology , Polysaccharides/pharmacology , RNA, Messenger/metabolism , Bone Marrow Cells , Cells, Cultured
16.
Molecules ; 27(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35011234

ABSTRACT

The endocannabinoidome (expanded endocannabinoid system, eCBome)-gut microbiome (mBIome) axis plays a fundamental role in the control of energy intake and processing. The liver-expressed antimicrobial peptide 2 (LEAP2) is a recently identified molecule acting as an antagonist of the ghrelin receptor and hence a potential effector of energy metabolism, also at the level of the gastrointestinal system. Here we investigated the role of the eCBome-gut mBIome axis in the control of the expression of LEAP2 in the liver and, particularly, the intestine. We confirm that the small intestine is a strong contributor to the circulating levels of LEAP2 in mice, and show that: (1) intestinal Leap2 expression is profoundly altered in the liver and small intestine of 13 week-old germ-free (GF) male mice, which also exhibit strong alterations in eCBome signaling; fecal microbiota transfer (FMT) from conventionally raised to GF mice completely restored normal Leap2 expression after 7 days from this procedure; in 13 week-old female GF mice no significant change was observed; (2) Leap2 expression in organoids prepared from the mouse duodenum is elevated by the endocannabinoid noladin ether, whereas in human Caco-2/15 epithelial intestinal cells it is elevated by PPARγ activation by rosiglitazone; (3) Leap2 expression is elevated in the ileum of mice with either high-fat diet-or genetic leptin signaling deficiency-(i.e., ob/ob and db/db mice) induced obesity. Based on these results, we propose that LEAP2 originating from the small intestine may represent a player in eCBome- and/or gut mBIome-mediated effects on food intake and energy metabolism.


Subject(s)
Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Endocannabinoids/genetics , Gastrointestinal Microbiome/genetics , Receptors, Ghrelin/antagonists & inhibitors , Animals , Caco-2 Cells , Diet, High-Fat , Female , Glycerides/metabolism , Humans , Intestines , Liver , Male , Mice , Mice, Inbred C57BL , Models, Animal , Obesity , RNA, Messenger/genetics , Rosiglitazone/metabolism , Signal Transduction , Tandem Mass Spectrometry
17.
PLoS One ; 15(11): e0242427, 2020.
Article in English | MEDLINE | ID: mdl-33186383

ABSTRACT

BACKGROUND: Gulf War (GW) Illness (GWI) is a debilitating condition with a complex constellation of immune, endocrine and neurological symptoms, including cognitive impairment, anxiety and depression. We studied a novel model of GWI based on 3 known common GW exposures (GWE): (i) intranasal lipopolysaccharide, to which personnel were exposed during desert sand storms; (ii) pyridostigmine bromide, used as prophylaxis against chemical warfare; and (iii) chronic unpredictable stress, an inescapable element of war. We used this model to evaluate prophylactic treatment with the PPARγ agonist, rosiglitazone (ROSI). METHODS: Rats were subjected to the three GWE for 33 days. In series 1 and 2, male and female GWE-rats were compared to naïve rats. In series 3, male rats with GWE were randomly assigned to prophylactic treatment with ROSI (GWE-ROSI) or vehicle. After the 33-day exposures, three neurofunctional domains were evaluated: cognition (novel object recognition), anxiety-like behaviors (elevated plus maze, open field) and depression-like behaviors (coat state, sucrose preference, splash test, tail suspension and forced swim). Brains were analyzed for astrocytic and microglial activation and neuroinflammation (GFAP, Iba1, tumor necrosis factor and translocator protein). Neurofunctional data from rats with similar exposures were pooled into 3 groups: naïve, GWE and GWE-ROSI. RESULTS: Compared to naïve rats, GWE-rats showed significant abnormalities in the three neurofunctional domains, along with significant neuroinflammation in amygdala and hippocampus. There were no differences between males and females with GWE. GWE-ROSI rats showed significant attenuation of neuroinflammation and of some of the neurofunctional abnormalities. CONCLUSION: This novel GWI model recapitulates critical neurofunctional abnormalities reported by Veterans with GWI. Concurrent prophylactic treatment with ROSI was beneficial in this model.


Subject(s)
Persian Gulf Syndrome/drug therapy , Persian Gulf Syndrome/metabolism , Rosiglitazone/pharmacology , Animals , Anxiety/metabolism , Astrocytes/metabolism , Brain/metabolism , Cognition/physiology , Disease Models, Animal , Female , Hippocampus/metabolism , Lipopolysaccharides/pharmacology , Male , PPAR gamma/agonists , PPAR gamma/metabolism , Persian Gulf Syndrome/physiopathology , Pyridostigmine Bromide/adverse effects , Rats , Rats, Wistar , Rosiglitazone/metabolism , Stress, Psychological/physiopathology
18.
Spectrochim Acta A Mol Biomol Spectrosc ; 234: 118268, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32203688

ABSTRACT

The binding of an anti-diabetic drug rosiglitazone (RG) with calf-thymus DNA (CT-DNA) in physiological buffer (pH 7.4) has been investigated using various spectral techniques such as UV-Vis, fluorescence, 1H NMR and circular dichroism (CD) coupled with viscosity measurement and molecular docking studies. The binding of RG with CT-DNA results in small hypochromism without any change in absorption maximum and fluorescence quenching with hardly any shifts in emission maximum suggesting groove binding mode of interaction. The binding constant is found to be 4.2 × 102 M-1 at 298 K. Thermodynamic analysis reveal that the binding is spontaneous and H-bonding and van der Waals forces play predominant role in the binding of RG with CT-DNA. Competitive interaction between RG and ethidium bromide with CT-DNA, viscosity measurements, KI quenching, 1H NMR and CD studies substantiate the prosed mode of binding. Voltammetric investigations suggest that the electro-reduction of RG is an adsorption controlled process and shift of reduction peak to more negative potential, with a binding constant of 3.4 × 103 M-1, validates the groove binding mode of interaction between RG and CT-DNA. Molecular docking reveals that RG binds in the minor groove of DNA and the dominating interaction forces are H-bonding and hydrophobic interactions.


Subject(s)
DNA/chemistry , DNA/metabolism , Electrochemical Techniques , Hypoglycemic Agents/chemistry , Molecular Docking Simulation , Rosiglitazone/chemistry , Rosiglitazone/metabolism , Binding, Competitive , Circular Dichroism , Ethidium/chemistry , Kinetics , Potassium Iodide/chemistry , Proton Magnetic Resonance Spectroscopy , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Thermodynamics , Viscosity
19.
Biochem Biophys Res Commun ; 524(1): 70-76, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31980182

ABSTRACT

Given the highly heterogeneity of diffuse large B cell lymphoma (DLBCL) and the diverse demands for proper treatment, many patients would relapse or show resistance to current therapeutic regimens, new treatment options are urgent to be explored. Curcumin harbored anti-tumor potential in various cancers, here, we investigated the possible effects and mechanism of curcumin on human DLBCL in vitro and in vivo, we found that curcumin inhibited cell viability in a concentration and time dependent manner, promoted cell apoptosis and arrested cell cycle at G2 phase, and these effects were mediated by PPARγ promotion and Akt/mTOR pathway inactivation. Furthermore, effects of curcumin on human DLBCL cells could be partly rescued by PPARγ antagonist GW9662, and enhanced by PPARγ agonist rosiglitazone. Taken together, our results demonstrated that curcumin inhibited the proliferation of DLBCL cells by up-regulating the expression of PPARγ, and our results might provide novel therapeutic approaches and a potential target to DLBCL treatment.


Subject(s)
Antineoplastic Agents/metabolism , Curcumin/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , PPAR gamma/metabolism , Anilides/metabolism , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, SCID , Neoplasms, Experimental , Proto-Oncogene Proteins c-akt/metabolism , Rosiglitazone/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
20.
Drug Chem Toxicol ; 43(6): 656-662, 2020 Nov.
Article in English | MEDLINE | ID: mdl-30880486

ABSTRACT

Metabolically induced drug-toxicity is a major cause of drug failure late in drug optimization phases. Accordingly, in vitro metabolic profiling of compounds is being introduced at earlier stages of the drug discovery pipeline. An increasingly common method to obtain these profiles is through overexpression of key CYP450 metabolic enzymes in immortalized liver cells, to generate competent hepatocyte surrogates. Enhanced cytotoxicity is presumed to be due to toxic metabolite production via the overexpressed enzyme. However, metabolically induced toxicity is a complex multi-parameter phenomenon and the potential background contribution to metabolism arising from the use of liver cells which endogenously express CYP450 isoforms is consistently overlooked. In this study, we sought to reduce the potential background interference by applying this methodology in kidney-derived HEK293 cells which lack endogenous CYP450 expression. Overexpression of CYP3A4 resulted in increased HEK293 proliferation, while exposure to four compounds with reported metabolically induced cytotoxicity in liver-derived cells overexpressing CYP3A4 resulted in no increase in cytotoxicity. Our results indicate that overexpression of a single CYP450 isoform in hepatic cell lines may not be a reliable method to discriminate which enzymes are responsible for metabolic induced cytotoxicity.


Subject(s)
Chlorpromazine/toxicity , Cytochrome P-450 CYP3A/metabolism , Epithelial Cells/drug effects , Labetalol/toxicity , Propranolol/toxicity , Rosiglitazone/toxicity , Activation, Metabolic , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorpromazine/metabolism , Cytochrome P-450 CYP3A/genetics , Epithelial Cells/enzymology , Epithelial Cells/pathology , HEK293 Cells , Humans , Labetalol/metabolism , Propranolol/metabolism , Risk Assessment , Rosiglitazone/metabolism , Substrate Specificity , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...