Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.426
Filter
1.
Int J Mol Sci ; 25(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39337503

ABSTRACT

The role of astroglial and microglial cells in the pathogenesis of epilepsy is currently under active investigation. It has been proposed that the activity of these cells may be regulated by the agonists of peroxisome proliferator-activated nuclear receptors (PPARs). This study investigated the effects of a seven-day treatment with the PPAR ß/δ agonist GW0742 (Fitorine, 5 mg/kg/day) on the behavior and gene expression of the astroglial and microglial proteins involved in the regulation of epileptogenesis in the rat brain within a lithium-pilocarpine model of temporal lobe epilepsy (TLE). TLE resulted in decreased social and increased locomotor activity in the rats, increased expression of astro- and microglial activation marker genes (Gfap, Aif1), pro- and anti-inflammatory cytokine genes (Tnfa, Il1b, Il1rn), and altered expression of other microglial (Nlrp3, Arg1) and astroglial (Lcn2, S100a10) genes in the dorsal hippocampus and cerebral cortex. GW0742 attenuated, but did not completely block, some of these impairments. Specifically, the treatment affected Gfap gene expression in the dorsal hippocampus and Aif1 gene expression in the cortex. The GW0742 injections attenuated the TLE-specific enhancement of Nlrp3 and Il1rn gene expression in the cortex. These results suggest that GW0742 may affect the expression of some genes involved in the regulation of epileptogenesis.


Subject(s)
Astrocytes , Epilepsy, Temporal Lobe , Microglia , PPAR delta , PPAR-beta , Thiazoles , Animals , Male , Rats , Astrocytes/drug effects , Astrocytes/metabolism , Cytokines/metabolism , Cytokines/genetics , Disease Models, Animal , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Microglia/drug effects , Microglia/metabolism , Phenols , Pilocarpine/pharmacology , PPAR delta/agonists , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/agonists , PPAR-beta/genetics , PPAR-beta/metabolism , Sulfhydryl Compounds , Thiazoles/pharmacology , Thiazoles/therapeutic use
2.
Respir Res ; 25(1): 345, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39313791

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung epithelial phenotypes, fibroblast activation, and increased extracellular matrix deposition. Transforming growth factor-beta (TGF-ß)1-induced Smad signaling and downregulation of peroxisomal genes are involved in the pathogenesis and can be inhibited by peroxisome proliferator-activated receptor (PPAR)-α activation. However, the three PPARs, that is PPAR-α, PPAR-ß/δ, and PPAR-γ, are known to interact in a complex crosstalk. METHODS: To mimic the pathogenesis of lung fibrosis, primary lung fibroblasts from control and IPF patients with comparable levels of all three PPARs were treated with TGF-ß1 for 24 h, followed by the addition of PPAR ligands either alone or in combination for another 24 h. Fibrosis markers (intra- and extracellular collagen levels, expression and activity of matrix metalloproteinases) and peroxisomal biogenesis and metabolism (gene expression of peroxisomal biogenesis and matrix proteins, protein levels of PEX13 and catalase, targeted and untargeted lipidomic profiles) were analyzed after TGF-ß1 treatment and the effects of the PPAR ligands were investigated. RESULTS: TGF-ß1 induced the expected phenotype; e.g. it increased the intra- and extracellular collagen levels and decreased peroxisomal biogenesis and metabolism. Agonists of different PPARs reversed TGF-ß1-induced fibrosis even when given 24 h after TGF-ß1. The effects included the reversals of (1) the increase in collagen production by repressing COL1A2 promoter activity (through PPAR-ß/δ activation); (2) the reduced activity of matrix metalloproteinases (through PPAR-ß/δ activation); (3) the decrease in peroxisomal biogenesis and lipid metabolism (through PPAR-γ activation); and (4) the decrease in catalase protein levels in control (through PPAR-γ activation) and IPF (through a combined activation of PPAR-ß/δ and PPAR-γ) fibroblasts. Further experiments to explore the role of catalase showed that an overexpression of catalase protein reduced collagen production. Additionally, the beneficial effect of PPAR-γ but not of PPAR-ß/δ activation on collagen synthesis depended on catalase activity and was thus redox-sensitive. CONCLUSION: Our data provide evidence that IPF patients may benefit from a combined activation of PPAR-ß/δ and PPAR-γ.


Subject(s)
Idiopathic Pulmonary Fibrosis , PPAR delta , PPAR gamma , PPAR-beta , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , PPAR-beta/metabolism , PPAR-beta/genetics , PPAR-beta/agonists , Cells, Cultured , PPAR delta/metabolism , PPAR delta/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/drug effects , Peroxisomes/metabolism , Peroxisomes/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Male , Transforming Growth Factor beta1/metabolism , Female
3.
Shock ; 62(4): 574-581, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39227395

ABSTRACT

ABSTRACT: Background: The kidney is the most commonly affected organ in sepsis patients, and Krüppel-like transcription factor 15 (KLF15) has a kidney-protective effect and is highly enriched in the kidneys. This study aims to explore the role of KLF15 in sepsis-related acute kidney injury. Methods: A septic injury model in HK2 cells was established through the administration of lipopolysaccharide (LPS), followed by the transfection of an overexpression plasmid for KLF15. Cell viability was assessed using Cell Counting Kit-8 assay, and apoptosis was measured via flow cytometry. The levels of inflammatory cytokines were detected using ELISA, and western blot assay was employed to assess the expression of KLF15, PPARδ, as well as inflammatory and apoptosis-related proteins. The interaction between KLF15 and PPARδ was confirmed through the utilization of online databases and immunoprecipitation experiments. The mechanism was further validated using PPARδ agonists and small interfering RNA. Results: LPS-induced HK2 cells showed downregulated expression of KLF15 and PPARδ, along with decreased viability, accompanied by increased levels of apoptosis, TNFα, IL-1ß, and IL-6. Additionally, LPS upregulated the expression of Bax, cytoplasmic cytochrome C [Cytc (cyt)], Cox-2, and p-NF-κB-p65 in HK2 cells, while simultaneously downregulating the expression of Bcl2 and mitochondrial cytochrome c [Cytc (mit)]. immunoprecipitation experiment revealed a possible interaction between KLF15 and PPARδ in HK2 cells. Ov-KLF15, Ov-PPARδ, or administration of PPARδ agonists effectively alleviated the aforementioned alterations induced by LPS. However, interference with PPARδ significantly attenuated the protective effect of Ov-KLF15 on HK2 cells. Conclusion: KLF15 attenuates LPS-induced apoptosis and inflammatory responses in HK2 cells via PPARδ.


Subject(s)
Apoptosis , Epithelial Cells , Inflammation , Kidney Tubules , Kruppel-Like Transcription Factors , Lipopolysaccharides , PPAR delta , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Lipopolysaccharides/toxicity , Lipopolysaccharides/pharmacology , Apoptosis/drug effects , Humans , PPAR delta/metabolism , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Inflammation/metabolism , Cell Line , Transcription Factors/metabolism , Transcription Factors/genetics , Acute Kidney Injury/metabolism
4.
Chem Res Toxicol ; 37(9): 1574-1587, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39235066

ABSTRACT

ZLY06 is a dual agonist of peroxisome proliferator-activated receptor (PPAR) δ/γ, showing potential therapeutic effects on metabolic syndrome. However, our research has revealed that ZLY06 exhibits hepatotoxicity in normal C57BL/6J mice, though the precise mechanism remains unclear. This study aims to investigate the manifestations and mechanisms of ZLY06-induced hepatotoxicity. We administered ZLY06 via oral gavage to C57BL/6J mice (once daily for six weeks) and monitored various indicators to preliminarily explore its hepatotoxicity. Additionally, we further investigate the specific mechanisms of ZLY06-induced hepatotoxicity using PPAR inhibitors (GW9662 and GSK0660) and the Protein kinase B (AKT) activator (SC79). Results showed that ZLY06 led to increased serum ALP, ALT and AST, as well as elevated liver index and hepatic lipid levels. There was upregulation in the gene and protein expression of lipid metabolism-related molecules Acc, Scd1, Cd36, Fabp1 and Fabp2 in hepatocytes, with Cd36 showing the most significant change. Furthermore, cotreatment with SC79 significantly reduced ZLY06-induced hepatotoxicity in AML12 cells, evidenced by decreased intracellular TG levels and downregulation of CD36 expression. Specific knockdown of CD36 also mitigated ZLY06-induced hepatotoxicity. The study found that ZLY06 may bind to AKT1, inhibiting its phosphorylation activation, with the downregulation of p-AKT1 preceding the upregulation of CD36. In summary, ZLY06 mediates the upregulation of CD36 by potentially binding to and inhibiting the phosphorylation of AKT1, leading to hepatic lipid metabolism disorder and inducing liver toxicity.


Subject(s)
CD36 Antigens , Lipid Metabolism , Liver , Mice, Inbred C57BL , PPAR gamma , Proto-Oncogene Proteins c-akt , Up-Regulation , Animals , CD36 Antigens/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation/drug effects , Mice , Up-Regulation/drug effects , Liver/drug effects , Liver/metabolism , Male , PPAR gamma/agonists , PPAR gamma/metabolism , Lipid Metabolism/drug effects , PPAR delta/metabolism , PPAR delta/agonists , PPAR delta/antagonists & inhibitors
5.
Biomed Pharmacother ; 179: 117303, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153437

ABSTRACT

The role of peroxisome proliferator-activated receptor (PPAR)ß/δ in hepatic fibrosis remains a subject of debate. Here, we examined the effects of a PPARß/δ agonist on the pathogenesis of liver fibrosis and the activation of hepatic stellate cells (HSCs), the main effector cells in liver fibrosis, in response to the pro-fibrotic stimulus transforming growth factor-ß (TGF-ß). The PPARß/δ agonist GW501516 completely prevented glucose intolerance and peripheral insulin resistance, blocked the accumulation of collagen in the liver, and attenuated the expression of inflammatory and fibrogenic genes in mice fed a choline-deficient high-fat diet (CD-HFD). The antifibrogenic effect of GW501516 observed in the livers CD-HFD-fed mice could occur through an action on HSCs since primary HSCs isolated from Ppard-/- mice showed increased mRNA levels of the profibrotic gene Col1a1. Moreover, PPARß/δ activation abrogated TGF-ß1-mediated cell migration (an indicator of cell activation) in LX-2 cells (immortalized activated human HSCs). Likewise, GW501516 attenuated the phosphorylation of the main downstream intracellular protein target of TGF-ß1, suppressor of mothers against decapentaplegic (SMAD)3, as well as the levels of the SMAD3 co-activator p300 via the activation of AMP-activated protein kinase (AMPK) and the subsequent inhibition of extracellular signal-regulated kinase-1/2 (ERK1/2) in LX-2 cells. Overall, these findings uncover a new mechanism by which the activation of AMPK by a PPARß/δ agonist reduces TGF-ß1-mediated activation of HSCs and fibrosis via the reduction of both SMAD3 phosphorylation and p300 levels.


Subject(s)
AMP-Activated Protein Kinases , E1A-Associated p300 Protein , Hepatic Stellate Cells , Liver Cirrhosis , Mice, Inbred C57BL , PPAR delta , PPAR-beta , Smad3 Protein , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Animals , Phosphorylation/drug effects , PPAR-beta/agonists , PPAR-beta/metabolism , PPAR-beta/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , PPAR delta/metabolism , PPAR delta/agonists , PPAR delta/genetics , Smad3 Protein/metabolism , AMP-Activated Protein Kinases/metabolism , E1A-Associated p300 Protein/metabolism , Male , Mice , Humans , Thiazoles/pharmacology , Diet, High-Fat/adverse effects , Mice, Knockout , Insulin Resistance , Cell Line , Transforming Growth Factor beta1/metabolism
6.
World J Gastroenterol ; 30(28): 3428-3446, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39091710

ABSTRACT

BACKGROUND: Alcohol-associated liver disease (ALD) is a leading cause of liver-related morbidity and mortality, but there are no therapeutic targets and modalities to prevent ALD-related liver fibrosis. Peroxisome proliferator activated receptor (PPAR) α and δ play a key role in lipid metabolism and intestinal barrier homeostasis, which are major contributors to the pathological progression of ALD. Meanwhile, elafibranor (EFN), which is a dual PPARα and PPARδ agonist, has reached a phase III clinical trial for the treatment of metabolic dysfunction-associated steatotic liver disease and primary biliary cholangitis. However, the benefits of EFN for ALD treatment is unknown. AIM: To evaluate the inhibitory effects of EFN on liver fibrosis and gut-intestinal barrier dysfunction in an ALD mouse model. METHODS: ALD-related liver fibrosis was induced in female C57BL/6J mice by feeding a 2.5% ethanol (EtOH)-containing Lieber-DeCarli liquid diet and intraperitoneally injecting carbon tetrachloride thrice weekly (1 mL/kg) for 8 weeks. EFN (3 and 10 mg/kg/day) was orally administered during the experimental period. Histological and molecular analyses were performed to assess the effect of EFN on steatohepatitis, fibrosis, and intestinal barrier integrity. The EFN effects on HepG2 lipotoxicity and Caco-2 barrier function were evaluated by cell-based assays. RESULTS: The hepatic steatosis, apoptosis, and fibrosis in the ALD mice model were significantly attenuated by EFN treatment. EFN promoted lipolysis and ß-oxidation and enhanced autophagic and antioxidant capacities in EtOH-stimulated HepG2 cells, primarily through PPARα activation. Moreover, EFN inhibited the Kupffer cell-mediated inflammatory response, with blunted hepatic exposure to lipopolysaccharide (LPS) and toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling. EFN improved intestinal hyperpermeability by restoring tight junction proteins and autophagy and by inhibiting apoptosis and proinflammatory responses. The protective effect on intestinal barrier function in the EtOH-stimulated Caco-2 cells was predominantly mediated by PPARδ activation. CONCLUSION: EFN reduced ALD-related fibrosis by inhibiting lipid accumulation and apoptosis, enhancing hepatocyte autophagic and antioxidant capacities, and suppressing LPS/TLR4/NF-κB-mediated inflammatory responses by restoring intestinal barrier function.


Subject(s)
Chalcones , Disease Models, Animal , Intestinal Mucosa , Liver Cirrhosis , Liver Diseases, Alcoholic , Mice, Inbred C57BL , PPAR alpha , Animals , Mice , Humans , Female , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/drug therapy , PPAR alpha/metabolism , PPAR alpha/agonists , Chalcones/pharmacology , Liver Cirrhosis/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Caco-2 Cells , Liver/pathology , Liver/drug effects , Liver/metabolism , Ethanol/toxicity , Apoptosis/drug effects , Lipid Metabolism/drug effects , PPAR delta/agonists , PPAR delta/metabolism , Signal Transduction/drug effects , Oxidative Stress/drug effects , Propionates
7.
Sci Immunol ; 9(98): eadn2717, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178275

ABSTRACT

The formation of memory T cells is a fundamental feature of adaptative immunity, allowing the establishment of long-term protection against pathogens. Although emerging evidence suggests that metabolic reprogramming is crucial for memory T cell differentiation and survival, the underlying mechanisms that drive metabolic rewiring in memory T cells remain unclear. Here, we found that up-regulation of the nuclear receptor peroxisome proliferator-activated receptor ß/δ (PPARß/δ) instructs the metabolic reprogramming that occurs during the establishment of central memory CD8+ T cells. PPARß/δ-regulated changes included suppression of aerobic glycolysis and enhancement of oxidative metabolism and fatty acid oxidation. Mechanistically, exposure to interleukin-15 and expression of T cell factor 1 facilitated activation of the PPARß/δ pathway, counteracting apoptosis induced by antigen clearance and metabolic stress. Together, our findings indicate that PPARß/δ is a master metabolic regulator orchestrating a metabolic switch that may be favorable for T cell longevity.


Subject(s)
CD8-Positive T-Lymphocytes , Mice, Inbred C57BL , PPAR delta , PPAR-beta , Animals , PPAR-beta/metabolism , PPAR-beta/immunology , CD8-Positive T-Lymphocytes/immunology , PPAR delta/immunology , PPAR delta/metabolism , Mice , Immunologic Memory/immunology , Memory T Cells/immunology , Mice, Knockout , Interleukin-15/immunology , Interleukin-15/metabolism , Mice, Transgenic , Metabolic Reprogramming , Receptors, Cytoplasmic and Nuclear
8.
Expert Opin Pharmacother ; 25(11): 1517-1523, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107982

ABSTRACT

INTRODUCTION: Seladelpar (MBX-8025) is a once-daily administered highly specific PPAR-δ agonist in Phase 3 and extension trials for use in patients with primary biliary cholangitis (PBC). AREAS COVERED: This review provides background on current treatment options for PBC, and summarizes clinical trial data regarding the safety and effectiveness of seladelpar within the context of these treatments. EXPERT OPINION: Clinical trials results demonstrate the safety and tolerability of seladelpar use for PBC, including in patients with cirrhosis. The primary composite endpoint (ALP <1.67 times ULN, decrease ≥ 15% from baseline, and TB ≤ULN) was met in 61.7% of the patients treated with seladelpar and in 20% receiving placebo (p < 0.001). Moreover, pruritus - a cardinal and often intractable symptom of PBC - was improved with seladelpar treatment, as were overall quality of life measurements. Improvements in markers of inflammation were likewise observed. These biochemical and clinical findings therefore represent landmark developments in PBC treatment and offer a therapeutic option for PBC.


Subject(s)
Liver Cirrhosis, Biliary , Quality of Life , Humans , Liver Cirrhosis, Biliary/drug therapy , Adult , PPAR delta/agonists , Pruritus/drug therapy , Pruritus/etiology , Propionates , Chalcones
9.
Immunity ; 57(8): 1864-1877.e9, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39111315

ABSTRACT

Tumor-infiltrating lymphocyte (TIL) hypofunction contributes to the progression of advanced cancers and is a frequent target of immunotherapy. Emerging evidence indicates that metabolic insufficiency drives T cell hypofunction during tonic stimulation, but the signals that initiate metabolic reprogramming in this context are largely unknown. Here, we found that Meteorin-like (METRNL), a metabolically active cytokine secreted by immune cells in the tumor microenvironment (TME), induced bioenergetic failure of CD8+ T cells. METRNL was secreted by CD8+ T cells during repeated stimulation and acted via both autocrine and paracrine signaling. Mechanistically, METRNL increased E2F-peroxisome proliferator-activated receptor delta (PPARδ) activity, causing mitochondrial depolarization and decreased oxidative phosphorylation, which triggered a compensatory bioenergetic shift to glycolysis. Metrnl ablation or downregulation improved the metabolic fitness of CD8+ T cells and enhanced tumor control in several tumor models, demonstrating the translational potential of targeting the METRNL-E2F-PPARδ pathway to support bioenergetic fitness of CD8+ TILs.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Mitochondria , Tumor Microenvironment , CD8-Positive T-Lymphocytes/immunology , Animals , Mitochondria/metabolism , Mitochondria/immunology , Mice , Tumor Microenvironment/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Humans , Mice, Inbred C57BL , Cytokines/metabolism , Signal Transduction , Energy Metabolism , PPAR delta/metabolism , Cell Line, Tumor , Neoplasms/immunology , Glycolysis , Mice, Knockout , Oxidative Phosphorylation
10.
Biomolecules ; 14(8)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39199415

ABSTRACT

Leptin, acting centrally or peripherally, has complex effects on cardiac remodeling and heart function. We previously reported that central leptin exerts an anti-hypertrophic effect in the heart via cardiac PPARß/δ activation. Here, we assessed the impact of central leptin administration and PPARß/δ inhibition on cardiac function. Various cardiac properties, including QRS duration, R wave amplitude, heart rate (HR), ejection fraction (EF), end-diastolic left ventricular mass (EDLVM), end-diastolic volume (EDV), and cardiac output (CO) were analyzed. Central leptin infusion increased cardiac PPARß/δ protein content and decreased HR, QRS duration, and R wave amplitude. These changes induced by central leptin suggested a decrease in the ventricular wall growth, which was confirmed by MRI. In fact, the EDLVM was reduced by central leptin while increased in rats co-treated with leptin and GSK0660, a selective antagonist of PPARß/δ activity. In summary, central leptin plays a dual role in cardiac health, potentially leading to ventricular atrophy and improving heart function when PPARß/δ signaling is intact. The protective effects of leptin are lost by PPARß/δ inhibition, underscoring the importance of this pathway. These findings highlight the therapeutic potential of targeting leptin and PPARß/δ pathways to combat cardiac alterations and heart failure, particularly in the context of obesity.


Subject(s)
Leptin , PPAR delta , PPAR-beta , Animals , Leptin/pharmacology , Leptin/metabolism , PPAR-beta/metabolism , PPAR-beta/agonists , PPAR delta/metabolism , PPAR delta/agonists , Rats , Male , Heart/drug effects , Rats, Wistar , Atrophy , Heart Rate/drug effects , Myocardium/metabolism , Myocardium/pathology , Sulfones , Thiophenes
11.
J Pineal Res ; 76(5): e12988, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38982751

ABSTRACT

Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.


Subject(s)
Kruppel-Like Factor 4 , Melatonin , Myocytes, Smooth Muscle , PPAR delta , Plaque, Atherosclerotic , Animals , Melatonin/pharmacology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Kruppel-Like Factor 4/metabolism , Humans , PPAR delta/metabolism , PPAR delta/genetics , Mice, Knockout , Male , Mice, Knockout, ApoE , Phenotype , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apolipoproteins E/deficiency , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/drug effects , Mice, Inbred C57BL
12.
Biomolecules ; 14(6)2024 May 21.
Article in English | MEDLINE | ID: mdl-38927010

ABSTRACT

Nuclear hormone receptors exist in dynamic equilibrium between transcriptionally active and inactive complexes dependent on interactions with ligands, proteins, and chromatin. The present studies examined the hypothesis that endogenous ligands activate peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in keratinocytes. The phorbol ester treatment or HRAS infection of primary keratinocytes increased fatty acids that were associated with enhanced PPARß/δ activity. Fatty acids caused PPARß/δ-dependent increases in chromatin occupancy and the expression of angiopoietin-like protein 4 (Angptl4) mRNA. Analyses demonstrated that stearoyl Co-A desaturase 1 (Scd1) mediates an increase in intracellular monounsaturated fatty acids in keratinocytes that act as PPARß/δ ligands. The activation of PPARß/δ with palmitoleic or oleic acid causes arrest at the G2/M phase of the cell cycle of HRAS-expressing keratinocytes that is not found in similarly treated HRAS-expressing Pparb/d-null keratinocytes. HRAS-expressing Scd1-null mouse keratinocytes exhibit enhanced cell proliferation, an effect that is mitigated by treatment with palmitoleic or oleic acid. Consistent with these findings, the ligand activation of PPARß/δ with GW0742 or oleic acid prevented UVB-induced non-melanoma skin carcinogenesis, an effect that required PPARß/δ. The results from these studies demonstrate that PPARß/δ has endogenous roles in keratinocytes and can be activated by lipids found in diet and cellular components.


Subject(s)
Keratinocytes , PPAR delta , PPAR-beta , Stearoyl-CoA Desaturase , Keratinocytes/metabolism , Keratinocytes/drug effects , PPAR-beta/metabolism , PPAR-beta/genetics , Animals , Mice , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , PPAR delta/metabolism , PPAR delta/genetics , Fatty Acids/metabolism , Angiopoietin-Like Protein 4/metabolism , Angiopoietin-Like Protein 4/genetics , Humans , Oleic Acid/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Monounsaturated/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology
13.
Theriogenology ; 226: 130-140, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38878465

ABSTRACT

Inflammation in the reproductive tract has become a serious threat to animal fertility. Recently, the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the context of reproduction and the inflammatory response has been highlighted, but the role of PPARß/δ has not been fully elucidated. The aim of the present study was to investigate the in vitro effect of PPARß/δ ligands (agonist: L-165,041 and antagonist: GSK 3787) on the transcriptome profile of porcine endometrium during LPS-induced inflammation in the mid-luteal and follicular phases of the oestrous cycle (days 10-12 and 18-20, respectively) using the RNA-Seq method. During the mid-luteal phase of the oestrous cycle, the current study identified 145 and 143 differentially expressed genes (DEGs) after treatment with an agonist or antagonist, respectively. During the follicular phase of the oestrous cycle, 55 and 207 DEGs were detected after treatment with an agonist or antagonist, respectively. The detected DEGs are engaged in the regulation of various processes, such as the complement and coagulation cascade, NF-κB signalling pathway, or the pathway of 15-eicosatetraenoic acid derivatives synthesis. The results of the current study indicate that PPARß/δ ligands are involved in the control of the endometrial inflammatory response.


Subject(s)
Endometrium , Inflammation , Lipopolysaccharides , PPAR delta , PPAR-beta , Animals , Female , Swine , Endometrium/drug effects , Endometrium/metabolism , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/metabolism , PPAR-beta/genetics , Inflammation/chemically induced , Phenoxyacetates/pharmacology , Gene Expression Regulation/drug effects , Transcriptome
14.
Int J Mol Sci ; 25(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38791255

ABSTRACT

A robust predictive model was developed using 136 novel peroxisome proliferator-activated receptor delta (PPARδ) agonists, a distinct subtype of lipid-activated transcription factors of the nuclear receptor superfamily that regulate target genes by binding to characteristic sequences of DNA bases. The model employs various structural descriptors and docking calculations and provides predictions of the biological activity of PPARδ agonists, following the criteria of the Organization for Economic Co-operation and Development (OECD) for the development and validation of quantitative structure-activity relationship (QSAR) models. Specifically focused on small molecules, the model facilitates the identification of highly potent and selective PPARδ agonists and offers a read-across concept by providing the chemical neighbours of the compound under study. The model development process was conducted on Isalos Analytics Software (v. 0.1.17) which provides an intuitive environment for machine-learning applications. The final model was released as a user-friendly web tool and can be accessed through the Enalos Cloud platform's graphical user interface (GUI).


Subject(s)
PPAR delta , Quantitative Structure-Activity Relationship , Software , PPAR delta/agonists , PPAR delta/chemistry , PPAR delta/metabolism , Molecular Docking Simulation , Humans , Machine Learning
15.
Med ; 5(5): 377-379, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38733969

ABSTRACT

The study by Hirschfield et al.1 demonstrated safety profile and clinically significant effectiveness of the peroxisome proliferator-activated receptor delta (PPARδ) agonist seladelpar in patients with primary biliary cholangitis, highlighting its plausible use as a second-line treatment to reduce disease activity and pruritus.


Subject(s)
Liver Cirrhosis, Biliary , Pruritus , Humans , Pruritus/drug therapy , Liver Cirrhosis, Biliary/drug therapy , PPAR delta/agonists , PPAR delta/metabolism
16.
Cancer Lett ; 592: 216937, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38704134

ABSTRACT

Dysfunctional bone marrow (BM) endothelial progenitor cells (EPCs) with high levels of reactive oxygen species (ROS) are responsible for defective hematopoiesis in poor graft function (PGF) patients with acute leukemia or myelodysplastic neoplasms post-allotransplant. However, the underlying mechanism by which BM EPCs regulate their intracellular ROS levels and the capacity to support hematopoiesis have not been well clarified. Herein, we demonstrated decreased levels of peroxisome proliferator-activated receptor delta (PPARδ), a lipid-activated nuclear receptor, in BM EPCs of PGF patients compared with those with good graft function (GGF). In vitro assays further identified that PPARδ knockdown contributed to reduced and dysfunctional BM EPCs, characterized by the impaired ability to support hematopoiesis, which were restored by PPARδ overexpression. Moreover, GW501516, an agonist of PPARδ, repaired the damaged BM EPCs triggered by 5-fluorouracil (5FU) in vitro and in vivo. Clinically, activation of PPARδ by GW501516 benefited the damaged BM EPCs from PGF patients or acute leukemia patients in complete remission (CR) post-chemotherapy. Mechanistically, we found that increased expression of NADPH oxidases (NOXs), the main ROS-generating enzymes, may lead to elevated ROS level in BM EPCs, and insufficient PPARδ may trigger BM EPC damage via ROS/p53 pathway. Collectively, we found that defective PPARδ contributes to BM EPC dysfunction, whereas activation of PPARδ in BM EPCs improves their hematopoiesis-supporting ability after myelosuppressive therapy, which may provide a potential therapeutic target not only for patients with leukemia but also for those with other cancers.


Subject(s)
Endothelial Progenitor Cells , Hematopoiesis , PPAR delta , Reactive Oxygen Species , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Young Adult , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/drug effects , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Mice, Inbred C57BL , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/drug therapy , NADPH Oxidases/metabolism , PPAR delta/metabolism , PPAR delta/genetics , Reactive Oxygen Species/metabolism , Thiazoles/pharmacology , Tumor Suppressor Protein p53/metabolism
17.
Phytomedicine ; 129: 155695, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728922

ABSTRACT

BACKGROUND: Exercise is an effective strategy to prevent sarcopenia, but high physical inactivity in the elderly requires alternative therapeutic approaches. Exercise mimetics are therapeutic compounds that simulate the beneficial effects of exercise on skeletal muscles. However, the toxicity and adverse effects of exercise mimetics raise serious concerns. PURPOSE: We aimed to search novel plant-based alternatives to activate exercise induced-signaling. METHODS: We used open databases and luciferase assays to identify plant-derived alternatives to activate exercise-induced signaling and compared its efficacy to mild intensity continuous training (MICT) in aged C57BL/6 mice. The nineteen-month-old mice were either fed an experimental diet supplemented with the isolated alternative or subjected to MICT for up to 21 mo of age. RESULTS: Our analysis revealed that Chrysanthemum zawadskii Herbich var latillobum (Maxim.) Kitamura (CZH), a medicinal plant rich in linarin, is a novel activator of peroxisome proliferator-activated receptor δ (PPARδ) and estrogen-related receptor γ (ERRγ), key regulators of exercise-induced positive effects on muscles. CZH supplementation ameliorated the loss of muscle function and mass, and increased PPARδ and ERRγ expression in mouse muscles. CZH also improved mitochondrial functions and proteostasis in aged mice, similar to MICT. Furthermore, CZH and linarin induced the activation of Sestrin 1, a key mediator of exercise benefits, in muscle. Silencing Sestrin 1 negated the increase in myogenesis and mitochondrial respiration by CZH and linarin in primary myoblasts from old mice. CONCLUSION: Our findings suggest the potential of CZH as a novel plant-derived alternative to activate exercise-induced signaling for preventing sarcopenia in sedentary older adults. This could offer a safer therapeutic option for sarcopenia treatment.


Subject(s)
Chrysanthemum , Mice, Inbred C57BL , Sarcopenia , Signal Transduction , Animals , Chrysanthemum/chemistry , Signal Transduction/drug effects , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Male , PPAR delta/metabolism , Plant Extracts/pharmacology , Receptors, Estrogen/metabolism , Humans , Aging/drug effects , Glycosides
18.
Biochem Biophys Res Commun ; 722: 150158, 2024 08 30.
Article in English | MEDLINE | ID: mdl-38795455

ABSTRACT

The cytokine interleukin-38 (IL-38), a recently discovered member of the IL-1 family, has been shown to regulate inflammation and improve hepatic endoplasmic reticulum stress and lipid metabolism in individuals with obesity. However, its impact on insulin signaling in skeletal muscle cells and the underlying mechanisms remain unclear. In vitro obesity models were established using palmitate treatment, and Western blot analysis was performed to assess target proteins. Commercial kits were used to measure glucose uptake in cultured myocytes. Our study showed that IL-38 treatment alleviated the impairment of insulin signaling, including IRS-1 and Akt phosphorylation, and increased glucose uptake in palmitate-treated C2C12 myocytes. Increased levels of STAT3-mediated signaling and oxidative stress were observed in these cells following palmitate treatment, and these effects were reversed by IL-38 treatment. In addition, IL-38 treatment upregulated the expression of PPARδ, SIRT1 and antioxidants. Knockdown of PPARδ or SIRT1 using appropriate siRNAs abrogated the effects of IL-38 on insulin signaling, oxidative stress, and the STAT3-dependent pathway. These results suggest that IL-38 alleviates insulin resistance by inhibiting STAT3-mediated signaling and oxidative stress in skeletal muscle cells through PPARδ/SIRT1. This study provides fundamental evidence to support the potential use of IL-38 as a safe therapeutic agent for the treatment of insulin resistance and type 2 diabetes.


Subject(s)
Hyperlipidemias , Insulin Resistance , Oxidative Stress , STAT3 Transcription Factor , Signal Transduction , Sirtuin 1 , Animals , Oxidative Stress/drug effects , Sirtuin 1/metabolism , Sirtuin 1/genetics , STAT3 Transcription Factor/metabolism , Mice , Signal Transduction/drug effects , Cell Line , Hyperlipidemias/metabolism , Hyperlipidemias/drug therapy , PPAR delta/metabolism , PPAR delta/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Interleukins/metabolism , Interleukins/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects , Interleukin-1/metabolism , Interleukin-1/genetics
19.
Metabolism ; 156: 155934, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38762141

ABSTRACT

BACKGROUND AND AIM: Clinically, septic males tend to have higher mortality rates, but it is unclear if this is due to sex differences in cardiac dysfunction, possibly influenced by hormonal variations. Cardiac dysfunction significantly contributes to sepsis-related mortality, primarily influenced by metabolic imbalances. Peroxisome proliferator-activated receptor delta (PPARδ) is a key player in cardiac metabolism and its activation has been demonstrated to favor sepsis outcomes. While estradiol (E2) is abundant and beneficial in females, its impact on PPARδ-mediated metabolism in the heart with regards to sex during sepsis remains unknown. METHODS AND RESULTS: Here, we unveil that while sepsis diminishes PPARδ nuclear translocation and induces metabolic dysregulation, oxidative stress, apoptosis and dysfunction in the heart thereby enhancing mortality, these effects are notably more pronounced in males than females. Mechanistic experiments employing ovariectomized(OVX) mice, E2 administration, and G protein-coupled estrogen receptor 1(GPER-1) knockout (KO) mice revealed that under lipopolysaccharide (LPS)-induced sepsis, E2 acting via GPER-1 enhances cardiac electrical activity and function, promotes PPARδ nuclear translocation, and subsequently ameliorates cardiac metabolism while mitigating oxidative stress and apoptosis in females. Furthermore, PPARδ specific activation using GW501516 in female GPER-1-/- mice reduced oxidative stress, ultimately decreasing NLRP3 expression in the heart. Remarkably, targeted GPER-1 activation using G1 in males mirrors these benefits, improving cardiac electrical activity and function, and ultimately enhancing survival rates during LPS challenge. By employing NLRP3 KO mice, we demonstrated that the targeted GPER-1 activation mitigated injury, enhanced metabolism, and reduced apoptosis in the heart of male mice via the downregulation of NLRP3. CONCLUSION: Our findings collectively illuminate the sex-specific cardiac mechanisms influencing sepsis mortality, offering insights into physiological and pathological dimensions. From a pharmacological standpoint, this study introduces specific GPER-1 activation as a promising therapeutic intervention for males under septic conditions. These discoveries advance our understanding of the sex differences in sepsis-induced cardiac dysfunction and also present a novel avenue for targeted interventions with potential translational impact.


Subject(s)
Estradiol , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis , Signal Transduction , Animals , Female , Male , Mice , Estradiol/pharmacology , Estradiol/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , PPAR delta/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Sepsis/metabolism , Sepsis/complications , Sex Characteristics
20.
Bioorg Chem ; 147: 107364, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636434

ABSTRACT

Osteoporosis is particularly prevalent among postmenopausal women and the elderly. In the present study, we investigated the effect of the novel small molecule E0924G (N-(4-methoxy-pyridine-2-yl)-5-methylfuran-2-formamide) on osteoporosis. E0924G significantly increased the protein expression levels of osteoprotegerin (OPG) and runt-related transcription factor 2 (RUNX2), and thus significantly promoted osteogenesis in MC3T3-E1 cells. E0924G also significantly decreased osteoclast differentiation and inhibited bone resorption and F-actin ring formation in receptor activator of NF-κB ligand (RANKL)-induced osteoclasts from RAW264.7 macrophages. Importantly, oral administration of E0924G in both ovariectomized (OVX) rats and SAMP6 senile mice significantly increased bone mineral density and decreased bone loss compared to OVX controls or SAMR1 mice. Further mechanistic studies showed that E0924G could bind to and then activate peroxisome proliferator-activated receptor delta (PPARδ), and the pro-osteoblast effect and the inhibition of osteoclast differentiation induced by E0924G were significantly abolished when PPARδ was knocked down or inhibited. In conclusion, these data strongly suggest that E0924G has the potential to prevent OVX-induced and age-related osteoporosis by dual regulation of bone formation and bone resorption through activation of the PPARδ signaling pathway.


Subject(s)
Bone Resorption , Osteogenesis , Ovariectomy , PPAR delta , Signal Transduction , Animals , Mice , Bone Resorption/drug therapy , Bone Resorption/prevention & control , Bone Resorption/metabolism , Rats , PPAR delta/metabolism , Female , Osteogenesis/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Molecular Structure , RAW 264.7 Cells , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Osteoporosis/metabolism , Dose-Response Relationship, Drug , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Rats, Sprague-Dawley , Osteoclasts/drug effects , Osteoclasts/metabolism , Cell Differentiation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL