Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.325
Filter
1.
Chem Biol Drug Des ; 104(3): e14625, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289148

ABSTRACT

Arctigenin (Ar) is a promising therapeutic candidate for postmenopausal osteoporosis (PMOP). This study explores its mechanism by examining its effects on adipogenesis and osteogenesis in ovariectomized (OVX) rats. In vitro, Ar effectively suppressed the adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) from OVX rats, reducing lipid droplet formation and downregulating proteins associated with lipid synthesis. In vivo, Ar treatment significantly reduced bone loss, inhibited adipocyte development, improved lipid metabolism, and promoted bone formation in OVX rats. Mechanistically, Ar inhibited the phosphorylation of Mitogen-Activated Protein Kinase 1 (MEK1), downregulated Peroxisome Proliferator-Activated Receptor gamma (PPARγ), promoted the accumulation of ß-catenin in the nucleus, and prevented the direct binding of PPARγ to ß-catenin in BMSCs. This regulation of the PPARγ/Wnt signaling axis underlies its dual role in inhibiting adipogenesis and promoting osteogenesis. Notably, co-treatment with rosiglitazone (RGZ) reversed the effects of Ar on adipogenesis and osteogenesis without affecting MEK1 inhibition. These findings offer valuable insights into arctigenin's potential as a therapeutic strategy for PMOP by modulating MEK1 signaling and regulating the PPARγ/Wnt axis.


Subject(s)
Adipogenesis , Furans , Lignans , MAP Kinase Kinase 1 , Mesenchymal Stem Cells , Osteogenesis , Ovariectomy , PPAR gamma , Rats, Sprague-Dawley , Wnt Signaling Pathway , beta Catenin , Animals , PPAR gamma/metabolism , Osteogenesis/drug effects , Female , Adipogenesis/drug effects , Lignans/pharmacology , Lignans/chemistry , Rats , Wnt Signaling Pathway/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Furans/pharmacology , Furans/chemistry , MAP Kinase Kinase 1/metabolism , beta Catenin/metabolism , Bone Marrow/metabolism , Bone Marrow/drug effects , Humans
2.
Cells ; 13(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273057

ABSTRACT

Cardiovascular diseases remain the leading cause of death in the world, and that is why finding an effective and multi-functional treatment alternative to combat these diseases has become more important. Fibrates and thiazolidinediones, peroxisome proliferator-activated receptors alpha and gamma are the pharmacological therapies used to treat dyslipidemia and type 2 diabetes, respectively. New mechanisms of action of these drugs have been found, demonstrating their pleiotropic effects, which contribute to preserving the heart by reducing or even preventing myocardial damage. Here, we review the mechanisms underlying the cardioprotective effects of PPAR agonists and regulating morphological and physiological heart alterations (metabolic flexibility, mitochondrial damage, apoptosis, structural remodeling, and inflammation). Moreover, clinical evidence regarding the cardioprotective effect of PPAR agonists is also addressed.


Subject(s)
Myocardium , PPAR alpha , PPAR gamma , Humans , PPAR gamma/agonists , PPAR gamma/metabolism , PPAR alpha/agonists , PPAR alpha/metabolism , Myocardium/pathology , Myocardium/metabolism , Animals , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use
3.
Cells ; 13(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273065

ABSTRACT

Prostate apoptosis response-4 (Par-4, also known as PAWR) is a ubiquitously expressed tumor suppressor protein that induces apoptosis selectively in cancer cells, while leaving normal cells unaffected. Our previous studies indicated that genetic loss of Par-4 promoted hepatic steatosis, adiposity, and insulin-resistance in chow-fed mice. Moreover, low plasma levels of Par-4 are associated with obesity in human subjects. The mechanisms underlying obesity in rodents and humans are multi-faceted, and those associated with adipogenesis can be functionally resolved in cell cultures. We therefore used pluripotent mouse embryonic fibroblasts (MEFs) or preadipocyte cell lines responsive to adipocyte differentiation cues to determine the potential role of Par-4 in adipocytes. We report that pluripotent MEFs from Par-4-/- mice underwent rapid differentiation to mature adipocytes with an increase in lipid droplet accumulation relative to MEFs from Par-4+/+ mice. Knockdown of Par-4 in 3T3-L1 pre-adipocyte cultures by RNA-interference induced rapid differentiation to mature adipocytes. Interestingly, basal expression of PPARγ, a master regulator of de novo lipid synthesis and adipogenesis, was induced during adipogenesis in the cell lines, and PPARγ induction and adipogenesis caused by Par-4 loss was reversed by replenishment of Par-4. Mechanistically, Par-4 downregulates PPARγ expression by directly binding to its upstream promoter, as judged by chromatin immunoprecipitation and luciferase-reporter studies. Thus, Par-4 transcriptionally suppresses the PPARγ promoter to regulate adipogenesis.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Apoptosis Regulatory Proteins , PPAR gamma , Animals , PPAR gamma/metabolism , PPAR gamma/genetics , Adipogenesis/genetics , Mice , Adipocytes/metabolism , Adipocytes/cytology , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Differentiation , Humans , Transcription, Genetic , Promoter Regions, Genetic/genetics , Fibroblasts/metabolism
4.
Cells ; 13(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39273076

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays a major role in breast cancer progression and the development of drug resistance. We have previously demonstrated a trans-differentiation therapeutic approach targeting invasive dedifferentiated cancer cells. Using a combination of PPARγ agonists and MEK inhibitors, we forced the differentiation of disseminating breast cancer cells into post-mitotic adipocytes. Utilizing murine breast cancer cells, we demonstrated a broad class effect of PPARγ agonists and MEK inhibitors in inducing cancer cell trans-differentiation into adipocytes. Both Rosiglitazone and Pioglitazone effectively induced adipogenesis in cancer cells, marked by PPARγ and C/EBPα upregulation, cytoskeleton rearrangement, and lipid droplet accumulation. All tested MEK inhibitors promoted adipogenesis in the presence of TGFß, with Cobimetinib showing the most prominent effects. A metastasis ex vivo culture from a patient diagnosed with triple-negative breast cancer demonstrated a synergistic upregulation of PPARγ with the combination of Pioglitazone and Cobimetinib. Our results highlight the potential for new therapeutic strategies targeting cancer cell plasticity and the dedifferentiation phenotype in aggressive breast cancer subtypes. Combining differentiation treatments with standard therapeutic approaches may offer a strategy to overcome drug resistance.


Subject(s)
Cell Differentiation , PPAR gamma , Pioglitazone , PPAR gamma/metabolism , PPAR gamma/agonists , Humans , Animals , Mice , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Pioglitazone/pharmacology , Protein Kinase Inhibitors/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Rosiglitazone/pharmacology , Azetidines/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Piperidines/pharmacology
5.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273102

ABSTRACT

Embryonic stem cells are crucial for studying developmental biology due to their self-renewal and pluripotency capabilities. This research investigates the differentiation of mouse ESCs into adipocytes, offering insights into obesity and metabolic disorders. Using a monolayer differentiation approach over 30 days, lipid accumulation and adipogenic markers, such as Cebpb, Pparg, and Fabp4, confirmed successful differentiation. RNA sequencing revealed extensive transcriptional changes, with over 15,000 differentially expressed genes linked to transcription regulation, cell cycle, and DNA repair. This study utilized Robust Rank Aggregation to identify critical regulatory genes like PPARG, CEBPA, and EP300. Network analysis further highlighted Atf5, Ccnd1, and Nr4a1 as potential key players in adipogenesis and its mature state, validated through RT-PCR. While key adipogenic factors showed plateaued expression levels, suggesting early differentiation events, this study underscores the value of ESCs in modeling adipogenesis. These findings contribute to our understanding of adipocyte differentiation and have significant implications for therapeutic strategies targeting metabolic diseases.


Subject(s)
Adipocytes , Adipogenesis , Cell Differentiation , Mouse Embryonic Stem Cells , Animals , Adipogenesis/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Cell Differentiation/genetics , Adipocytes/metabolism , Adipocytes/cytology , PPAR gamma/metabolism , PPAR gamma/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Gene Expression Profiling , Gene Regulatory Networks , Transcription, Genetic , Gene Expression Regulation
6.
Int J Mol Sci ; 25(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39273161

ABSTRACT

The Target-Based Virtual Screening approach is widely employed in drug development, with docking or molecular dynamics techniques commonly utilized for this purpose. This systematic review (SR) aimed to identify in silico therapeutic targets for treating Diabetes mellitus (DM) and answer the question: What therapeutic targets have been used in in silico analyses for the treatment of DM? The SR was developed following the guidelines of the Preferred Reporting Items Checklist for Systematic Review and Meta-Analysis, in accordance with the protocol registered in PROSPERO (CRD42022353808). Studies that met the PECo strategy (Problem, Exposure, Context) were included using the following databases: Medline (PubMed), Web of Science, Scopus, Embase, ScienceDirect, and Virtual Health Library. A total of 20 articles were included, which not only identified therapeutic targets in silico but also conducted in vivo analyses to validate the obtained results. The therapeutic targets most frequently indicated in in silico studies were GLUT4, DPP-IV, and PPARγ. In conclusion, a diversity of targets for the treatment of DM was verified through both in silico and in vivo reassessment. This contributes to the discovery of potential new allies for the treatment of DM.


Subject(s)
Computer Simulation , Diabetes Mellitus , Dietary Supplements , Hypoglycemic Agents , Humans , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Glucose Transporter Type 4/metabolism , Animals , Drug Development/methods , PPAR gamma/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy/methods
7.
Int J Mol Sci ; 25(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39273687

ABSTRACT

The global obesity epidemic, exacerbated by the sedentary lifestyle fostered by the COVID-19 pandemic, presents a growing socioeconomic burden due to decreased physical activity and increased morbidity. Current obesity treatments show promise, but they often come with expensive medications, frequent injections, and potential side effects, with limited success in improving obesity through increased energy expenditure. This study explores the potential of a refined sulfated polysaccharide (SPSL), derived from the brown seaweed Scytosiphon lomentaria (SL), as a safe and effective anti-obesity treatment by promoting energy expenditure. Chemical characterization revealed that SPSL, rich in sulfate and L-fucose content, comprises nine distinct sulfated glycan structures. In vitro analysis demonstrated potent anti-lipogenic properties in adipocytes, mediated by the downregulation of key adipogenic modulators, including 5' adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ (PPARγ) pathways. Inhibiting AMPK attenuated the anti-adipogenic effects of SPSL, confirming its involvement in the mechanism of action. Furthermore, in vivo studies using zebrafish models showed that SPSL increased energy expenditure and reduced lipid accumulation. These findings collectively highlight the therapeutic potential of SPSL as a functional food ingredient for mitigating obesity-related metabolic dysregulation by promoting energy expenditure. Further mechanistic and preclinical investigations are warranted to fully elucidate its mode of action and evaluate its efficacy in obesity management, potentially offering a novel, natural therapeutic avenue for this global health concern.


Subject(s)
Adipogenesis , Energy Metabolism , Fucose , Functional Food , Obesity , Polysaccharides , Seaweed , Zebrafish , Animals , Energy Metabolism/drug effects , Obesity/drug therapy , Obesity/metabolism , Polysaccharides/chemistry , Polysaccharides/pharmacology , Seaweed/chemistry , Fucose/metabolism , Adipogenesis/drug effects , Mice , Adipocytes/metabolism , Adipocytes/drug effects , Humans , Sulfates/chemistry , Sulfates/metabolism , PPAR gamma/metabolism , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism
8.
Nat Commun ; 15(1): 7730, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231983

ABSTRACT

Mutations in mitochondrial energy-producing genes lead to a heterogeneous group of untreatable disorders known as primary mitochondrial diseases (MD). Leigh syndrome (LS) is the most common pediatric MD and is characterized by progressive neuromuscular affectation and premature death. Here, we show that daily cannabidiol (CBD) administration significantly extends lifespan and ameliorates pathology in two LS mouse models, and improves cellular function in fibroblasts from LS patients. CBD delays motor decline and neurodegenerative signs, improves social deficits and breathing abnormalities, decreases thermally induced seizures, and improves neuropathology in affected brain regions. Mechanistically, we identify peroxisome proliferator-activated receptor gamma (PPARγ) as a key nuclear receptor mediating CBD's beneficial effects, while also providing proof of dysregulated PPARγ expression and activity as a common feature in both mouse neurons and fibroblasts from LS patients. Taken together, our results provide the first evidence for CBD as a potential treatment for LS.


Subject(s)
Cannabidiol , Mitochondrial Diseases , PPAR gamma , Animals , Female , Humans , Male , Mice , Brain/metabolism , Brain/drug effects , Brain/pathology , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Leigh Disease/drug therapy , Leigh Disease/metabolism , Leigh Disease/genetics , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Neurons/drug effects , Neurons/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics
9.
Mol Med Rep ; 30(5)2024 11.
Article in English | MEDLINE | ID: mdl-39301631

ABSTRACT

Fibrosis is the basis of structural remodeling in atrial fibrillation (AF), during which inflammation is crucial. Programmed cell death factor 4 (PDCD4) is a newly identified inflammatory gene, with unknown mechanisms of action in AF. The present study aimed to elucidate the effects of PDCD4 on the inflammation and structural remodeling of atrial myocytes. For this purpose, a PDCD4 overexpression plasmid (oePDCD4) and PDCD4 small interfering (si)RNA (siPDCD4) were used to modulate PDCD4 expression in mouse atrial myocytes (HL­1 cells). The expression of PDCD4 was detected using reverse transcription­quantitative PCR and western blot analysis. The optimal drug concentrations of peroxisome proliferator­activated receptor γ (PPARγ) agonist (pioglitazone hydrochloride), NF­κB inhibitor (CBL0137), PPARγ inhibitor (GW9962) and NF­κB agonist (betulinic acid) were screened using a Cell Counting Kit­8 assay. The levels of inflammatory factors were detected using enzyme­linked immunosorbent assays, the expression levels of fibrosis­related proteins and NF­κB subunits were detected using western blot analysis, and the expression of phosphorylated (p­)p65/p65 was detected using immunofluorescence staining. The results revealed that PDCD4 overexpression increased the levels of fibrotic factors (collagen I, collagen III, fibronectin, α­smooth muscle actin and matrix metalloproteinase 2), pro­inflammatory cytokines (IFN­Î³, IL­6, IL­17A and TNF­α) and p­p65, whereas it reduced the levels of anti­inflammatory cytokines (IL­4) in HL­1 cells. Additionally, treatment with the PPARγ agonist and NF­κB inhibitor reversed the levels of fibrotic­, pro­inflammatory and anti­inflammatory factors in oePDCD4­HL­1 cells. By contrast, PDCD4 silencing exerted the opposite effects on fibrotic factors, pro­inflammatory cytokines, anti­inflammatory cytokines and p­p65. In addition, treatment with the PPARγ inhibitor and NF­κB agonist reversed the levels of fibrotic­, pro­inflammatory and anti­inflammatory factors in siPDCD4­HL­1 cells. In conclusion, the present study demonstrated that PDCD4 may induce inflammation and fibrosis by activating the PPARγ/NF­κB signaling pathway, thereby promoting the structural remodeling of atrial myocytes in AF.


Subject(s)
Apoptosis Regulatory Proteins , Fibrosis , Inflammation , Myocytes, Cardiac , NF-kappa B , PPAR gamma , RNA-Binding Proteins , Signal Transduction , Animals , PPAR gamma/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , NF-kappa B/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Heart Atria/metabolism , Heart Atria/pathology , Cell Line
10.
Allergol Immunopathol (Madr) ; 52(5): 73-79, 2024.
Article in English | MEDLINE | ID: mdl-39278854

ABSTRACT

This study examines the therapeutic effects of Shengmai Powder (SMP) on both in vitro and in vivo models of chronic obstructive pulmonary disease (COPD) and the underlying mechanisms. Cigarette smoke and cigarette extracts were used to create in vitro and in vivo models of COPD. ELISA was used to measure the levels of pro-inflammatory factors (IL-6, TNF-α, and IL-1ß) in mouse lung tissue and alveolar macrophages. Flow cytometry assessed the phagocytic capacity of alveolar macrophage. Western blotting was used to analyze the expression of RhoA, PPARγ, IκBα, p-IκBα, P65, and p-P65 in alveolar. The results show that SMP reversed the increased levels of pro-inflammatory factors (IL-6, TNF-α, and IL-1ß) in mouse lung tissue and alveolar macrophages induced by cigarette smoke and cigarette extract. SMP also restored the decreased fluorescence intensity and RhoA levels in alveolar macrophages caused by cigarette extract. Additionally, SMP increased PPARγ expression and decreased IκBα and P65 phosphorylation in alveolar macrophages exposed to cigarette extract. Also, the effects of SMP were reversed by PPARγ inhibitors. The study concluded that SMP regulates alveolar macrophage phagocytic function through the PPAR-γ/NF-κB pathway, thereby improving the chronic inflammatory state of COPD.


Subject(s)
Drug Combinations , Drugs, Chinese Herbal , Macrophages, Alveolar , PPAR gamma , Pulmonary Disease, Chronic Obstructive , Animals , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , PPAR gamma/metabolism , Drugs, Chinese Herbal/pharmacology , Mice , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Disease Models, Animal , Phagocytosis/drug effects , Humans , Male , Cytokines/metabolism , Powders , Signal Transduction/drug effects , NF-kappa B/metabolism
11.
Int J Oncol ; 65(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39301639

ABSTRACT

Macrophages have crucial roles in immune responses and tumor progression, exhibiting diverse phenotypes based on environmental cues. In the present study, the impact of cinobufagin (CB) on macrophage polarization and the consequences on tumor­associated behaviors were investigated. Morphological transformations of THP­1 cells into M0, M1 and M2 macrophages were observed, including distinct changes in the size, shape and adherence properties of these cells. CB treatment inhibited the viability of A549 and LLC cells in a concentration­dependent manner, with an IC50 of 28.8 and 30.12 ng/ml, respectively. CB at concentrations of <30 ng/ml had no impact on the viability of M0 macrophages and lung epithelial (BEAS­2B) cells. CB influenced the expression of macrophage surface markers, reducing CD206 positivity in M2 macrophages without affecting CD86 expression in M1 macrophages. CB also altered certain expression profiles at the mRNA level, notably downregulating macrophage receptor with collagenous structure (MARCO) expression in M2 macrophages and upregulating tumor necrosis factor­α and interleukin­1ß in both M0 and M1 macrophages. Furthermore, ELISA analyses revealed that CB increased the levels of pro­inflammatory cytokines in M1 macrophages and reduced the levels of anti­inflammatory factors in M2 macrophages. CB treatment also attenuated the migration and invasion capacities of A549 and LLC cells stimulated by M2 macrophage­conditioned medium. Additionally, CB modulated peroxisome proliferator­activated receptor γ (PPARγ) and MARCO expression in M2 macrophages and epithelial­mesenchymal transition in A549 cells, which was partially reversed by rosiglitazone, a PPARγ agonist. Finally, CB and cisplatin treatments hindered tumor growth in vivo, with distinct impacts on animal body weight and macrophage marker expression in tumor tissues. In conclusion, the results of the present study demonstrated that CB exerted complex regulatory effects on macrophage polarization and tumor progression, suggesting its potential as a modulator of the tumor microenvironment and a therapeutic for cancer treatment.


Subject(s)
Bufanolides , Cell Movement , Lung Neoplasms , Neoplasm Invasiveness , Tumor-Associated Macrophages , Bufanolides/pharmacology , Bufanolides/therapeutic use , Humans , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/immunology , Animals , Mice , Cell Movement/drug effects , A549 Cells , Xenograft Model Antitumor Assays , THP-1 Cells , PPAR gamma/metabolism , Macrophage Activation/drug effects , Cell Line, Tumor
12.
Mol Med ; 30(1): 151, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39278948

ABSTRACT

Erythropoietin (EPO), expressed in red blood progenitor cells, primarily regulates erythropoiesis by binding to its receptor. Besides anemia, recent studies have identified new therapeutic indications for EPO that are not connected to red blood cell formation. Elevated EPO levels harm bone homeostasis in adult organisms and are associated with increased osteoclast; however, the underlying molecular mechanisms remain unclear. This study demonstrated that EPO enhanced osteoclast differentiation and bone resorption in vitro. We showed that EPO promoted osteoclast formation by up-regulating PPARγ expression through activating the Jak2/ERK signaling pathway. Consistently, PPARγ antagonists rescued the hyperactivation of osteoclasts due to EPO, while PPARγ agonists reversed the EMP9-mediated decrease in osteoclast differentiation. Further, exposing female mice to EPO for two months led to a decrease in bone mass and increased osteoclast numbers. The present results suggested that EPO promotes osteoclastogenesis by regulating the Jak2/ERK/ PPARγ signaling pathway. From a clinical perspective, the risk of compromised bone health should be considered when using EPO to treat anemia in post-operative patients with intertrochanteric fractures of the femur, as it could significantly impact the patient's recovery and quality of life.


Subject(s)
Cell Differentiation , Erythropoietin , Osteoclasts , PPAR gamma , Erythropoietin/pharmacology , Erythropoietin/metabolism , Animals , PPAR gamma/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Mice , Female , Cell Differentiation/drug effects , Osteogenesis/drug effects , Janus Kinase 2/metabolism , MAP Kinase Signaling System/drug effects , Signal Transduction/drug effects , Humans , Up-Regulation/drug effects , Gene Expression Regulation/drug effects , Bone Resorption/metabolism , Mice, Inbred C57BL
13.
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
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1518-1528, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276047

ABSTRACT

OBJECTIVE: To elucidate the therapeutic mechanism of Qingxin Jieyu Granule (QXJYG) against atherosclerosis (AS) based on network pharmacology. METHODS: The major targets and pathways of QXJYG against AS were analyzed using network pharmacology. Rat models of AS established by high-fat feeding combined with intraperitoneal vitamin D3 injection were treated daily with normal saline, atorvastatin (13.15 mg/kg), or QXJYG at 0.99, 1.98, and 3.96 g/kg for 8 weeks (n=6). Ultrasound and HE staining were used to assess the function and pathologies of the abdominal aorta. Blood lipids and serum levels of Ang Ⅱ, ET-1, TXA2, PGI2, and ox-LDL of the rats were detected using an automatic biochemical analyzer or ELISA. The expressions of LOX-1, PPARγ, RXRα, p-P65, VCAM-1 and ICAM-1 in the abdominal aorta were detected with immunohistochemistry. RESULTS: The rat models of AS showed obvious abdominal aorta wall thickening, increased pulse wave velocity and pulse index, decreased inner diameter of the abdominal aorta, elevated levels of TC, LDL-C, Ang Ⅱ, ET-1 and TXA2, and lowered levels of HDL-C and PGI2. QXJYG and atorvastatin treatment of the rat models significantly alleviated histopathological changes of the abdominal aorta, decreased serum levels of TC, LDL-C, Ang Ⅱ, ET-1 and TXA2, and increased the levels of HDL-C and PGI2. Network pharmacology study suggested the therapeutic effect of QXJYG against AS was mediated by regulating lipid metabolism, PPAR and NF-κB pathways. Consistently, treatments with QXJYG were found to significantly decrease ox-LDL level and LOX-1, P-P65, VCAM-1 and ICAM-1 protein expressions while increasing PPARγ and RXRα expressions in the aorta of AS rats. CONCLUSION: QXJYG alleviates lipid metabolism disorder and improves histopathological changes of the abdominal aorta of AS rats possibly by lowering ox-LDL level, reducing LOX-1 expression, activating PPARγ and RXRα, and inhibiting P65 phosphorylation to reduce VCAM-1 and ICAM-1 expression in the aorta.


Subject(s)
Atherosclerosis , Drugs, Chinese Herbal , Lipid Metabolism , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Rats , Lipid Metabolism/drug effects , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Aorta, Abdominal/metabolism , Aorta, Abdominal/drug effects , Network Pharmacology , Lipoproteins, LDL/metabolism , Intercellular Adhesion Molecule-1/metabolism , PPAR gamma/metabolism , Male , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Vascular Cell Adhesion Molecule-1/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Lipids/blood , Thromboxane A2/metabolism , Epoprostenol/analogs & derivatives , Scavenger Receptors, Class E
15.
Clin Exp Pharmacol Physiol ; 51(11): e13917, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39285148

ABSTRACT

Ischaemic stroke is a common condition that can lead to cerebral ischaemia-reperfusion injury. Phillygenin (PHI), a natural bioactive compound derived from Forsythia suspensa, has been shown to play a crucial role in regulating inflammation across various diseases. However, its specific regulatory effects in ischaemic stroke progression remain unclear. In this study, we established a middle cerebral artery occlusion (MCAO) rat model. Treatment with PHI (50 or 100 mg/kg) significantly reduced cerebral infarction in MCAO rats. PHI treatment also mitigated the increased inflammatory response observed in these rats. Additionally, PHI suppressed microglial activation by reducing iNOS expression, a marker of M1-type polarization of microglia, and attenuated increased brain tissue apoptosis in MCAO rats. Furthermore, PHI's anti-inflammatory effects in MCAO rats were abrogated upon co-administration with GW9662, a peroxisome proliferator-activated receptor γ (PPARγ) inhibitor. In summary, PHI attenuated microglial activation and apoptosis in cerebral ischaemia-reperfusion injury through PPARγ activation, suggesting its potential as a therapeutic agent for mitigating cerebral ischaemia-reperfusion injury.


Subject(s)
Apoptosis , Infarction, Middle Cerebral Artery , Microglia , PPAR gamma , Rats, Sprague-Dawley , Reperfusion Injury , Animals , PPAR gamma/metabolism , Apoptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Male , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Lignans
16.
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
17.
Int J Biol Sci ; 20(11): 4297-4313, 2024.
Article in English | MEDLINE | ID: mdl-39247816

ABSTRACT

The heart relies on various defense mechanisms, including metabolic plasticity, to maintain its normal structure and function under high-altitude hypoxia. Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ), sensitizes insulin, which in turn regulates blood glucose levels. However, its preventive effects against hypoxia-induced cardiac dysfunction at high altitudes have not been reported. In this study, pioglitazone effectively prevented cardiac dysfunction in hypoxic mice for 4 weeks, independent of its effects on insulin sensitivity. In vitro experiments demonstrated that pioglitazone enhanced the contractility of primary cardiomyocytes and reduced the risk of QT interval prolongation under hypoxic conditions. Additionally, pioglitazone promoted cardiac glucose metabolic reprogramming by increasing glycolytic capacity; enhancing glucose oxidation, electron transfer, and oxidative phosphorylation processes; and reducing mitochondrial reactive ROS production, which ultimately maintained mitochondrial membrane potential and ATP production in cardiomyocytes under hypoxic conditions. Notably, as a PPARγ agonist, pioglitazone promoted hypoxia-inducible factor 1α (HIF-1α) expression in hypoxic myocardium. Moreover, KC7F2, a HIF-1α inhibitor, disrupted the reprogramming of cardiac glucose metabolism and reduced cardiac function in pioglitazone-treated mice under hypoxic conditions. In conclusion, pioglitazone effectively prevented high-altitude hypoxia-induced cardiac dysfunction by reprogramming cardiac glucose metabolism.


Subject(s)
Glucose , Hypoxia , Myocytes, Cardiac , PPAR gamma , Pioglitazone , Pioglitazone/pharmacology , Pioglitazone/therapeutic use , Animals , PPAR gamma/metabolism , PPAR gamma/agonists , Mice , Glucose/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Hypoxia/complications , Hypoxia/metabolism , Male , Mice, Inbred C57BL , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Reactive Oxygen Species/metabolism
18.
Int Immunopharmacol ; 140: 112822, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39096877

ABSTRACT

Sepsis is a systemic inflammatory response syndrome caused by a dysregulated host response to infection. CD4+T cell reduction is crucial to sepsis-induced immunosuppression. Pyroptosis, a programmed necrosis, is concerned with lymphocytopenia. Peroxisome proliferator-activated receptor gamma (PPARγ) regulated by upstream mTOR, exerts anti-pyroptosis effects. To investigate the potential effects of mTOR-PPARγ on sepsis-induced CD4+T cell depletion and the underlying mechanisms, we observed mTOR activation and pyroptosis with PPARγ-Nrf suppression through cecal ligation and puncture (CLP) sepsis mouse model. Further mechanism research used genetically modified mice with T cell-specific knockout mTOR or Tuberous Sclerosis Complex1 (TSC1). It revealed that mTOR mediated CD4 + T cell pyroptosis in septic mice by negatively regulating the PPARγ-Nrf2 signaling pathway. Taken together, mTOR-PPARγ-Nrf2 signaling mediated the CD4+ T cell pyroptosis in sepsis, contributing to CD4+T cell depletion and immunosuppression.


Subject(s)
CD4-Positive T-Lymphocytes , NF-E2-Related Factor 2 , PPAR gamma , Pyroptosis , Sepsis , Signal Transduction , TOR Serine-Threonine Kinases , Animals , PPAR gamma/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Sepsis/immunology , Sepsis/metabolism , CD4-Positive T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Male , Disease Models, Animal , Humans
19.
Int Immunopharmacol ; 140: 112729, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39098229

ABSTRACT

ADORA3 is mainly expressed in intestinal tract, and has the potential to promote the expression of mucin 2 (MUC2), the function-related factor of goblet cells, under asthma conditions. This study aims to confirm the induction and mechanisms of ADORA3 activation on goblet cells in ulcerative colitis (UC). A significant decrease in ADORA3 expression was found in mucosal biopsies from UC patients and in the colons of colitis mice. This reduction correlated negatively with disease severity and positively with goblet cell number. ADORA3 activation mitigated dextran sulfate sodium (DSS)-induced colitis and facilitated ATOH1-mediated goblet cell differentiation in both in vivo and in vitro. Metabolomics analysis unveiled that ADORA3 activation bolstered ketogenesis, leading to elevated levels of the metabolite BHB. Subsequently, BHB heightened the activity of HDAC1/2, augmenting histone acetylation at the H3K9ac site within the promoter region of the ATOH1 gene. Furthermore, the reason for ADORA3 activation to enhance ketogenesis was attributed to controlling the competitive binding among ß-arrestin2, SHP1 and PPARγ. This results in the non-ligand-dependent activation of PPARγ, thereby promoting the transcription of HMGCS2. The exact mechanisms by which ADORA3 promoted goblet cell differentiation and alleviated UC were elucidated using MRS1191 and shHMGCS2 plasmid. Collectively, ADORA3 activation promoted goblet cell differentiation and alleviated UC by enhancing ketogenesis via the "BHB-HDAC1/2-H3K9ac" pathway.


Subject(s)
Cell Differentiation , Colitis, Ulcerative , Goblet Cells , Hydroxymethylglutaryl-CoA Synthase , Adult , Animals , Female , Humans , Male , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Butyric Acid/pharmacology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Colon/pathology , Colon/metabolism , Dextran Sulfate , Goblet Cells/pathology , Goblet Cells/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Mice, Inbred C57BL , PPAR gamma/metabolism , PPAR gamma/genetics
20.
J Microbiol Biotechnol ; 34(8): 1688-1697, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39086228

ABSTRACT

The current study aimed to determine whether Strongylocentrotus intermedius (S. intermedius) extract (SIE) exerts anti-obesity potentials employing 3T3-L1 cells as in vitro model. Herein we reported that treatment of SIE for 6 days reduced lipid accretion and triglyceride content whereas it increased the release of free glycerol. The inhibited lipid accumulation and induced lipolysis were evidenced by the downregulation of lipogenesis proteins, such as fatty acid synthase and lipoprotein lipase, and the upregulation of hormone-sensitive lipase expression. Furthermore, the downregulation of adipogenic transcription factors, including peroxisome proliferator-activated receptor gamma, CCAAT/enhancer-binding protein α, and sterol regulatory element-binding protein 1, highlights that reduced lipid accumulation is supported by lowering adipocyte differentiation. Additionally, treatment activates brown adipocyte phenotype in 3T3-L1 cells by inducing expression of brown adipose tissue-specific proteins, such as uncoupling protein 1 and peroxisome proliferator-activated receptor-γ coactivator 1α. Moreover, SIE induced the phosphorylation of AMP-activated protein kinase (AMPK). The pharmacological approach using AMPK inhibitor revealed that the restraining effect of SIE on adipogenesis and promotion of adipocyte browning were blocked. In GC-MS analysis, SIE was mainly composed of cholest-5-en-3-ol (36.71%) along with saturated and unsaturated fatty acids which have favorable anti-obesity potentials. These results reveal that SIE has the possibility as a lipid-lowering agent for the intervention of obesity.


Subject(s)
3T3-L1 Cells , AMP-Activated Protein Kinases , Adipogenesis , Animals , Adipogenesis/drug effects , Mice , AMP-Activated Protein Kinases/metabolism , Lipolysis/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Adiposity/drug effects , Anti-Obesity Agents/pharmacology , Lipid Metabolism/drug effects , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Triglycerides/metabolism , Cell Differentiation/drug effects , Lipogenesis/drug effects , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL