Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.503
Filter
Add more filters

Publication year range
1.
Cell ; 179(6): 1289-1305.e21, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31761534

ABSTRACT

Adult mesenchymal stem cells, including preadipocytes, possess a cellular sensory organelle called the primary cilium. Ciliated preadipocytes abundantly populate perivascular compartments in fat and are activated by a high-fat diet. Here, we sought to understand whether preadipocytes use their cilia to sense and respond to external cues to remodel white adipose tissue. Abolishing preadipocyte cilia in mice severely impairs white adipose tissue expansion. We discover that TULP3-dependent ciliary localization of the omega-3 fatty acid receptor FFAR4/GPR120 promotes adipogenesis. FFAR4 agonists and ω-3 fatty acids, but not saturated fatty acids, trigger mitosis and adipogenesis by rapidly activating cAMP production inside cilia. Ciliary cAMP activates EPAC signaling, CTCF-dependent chromatin remodeling, and transcriptional activation of PPARγ and CEBPα to initiate adipogenesis. We propose that dietary ω-3 fatty acids selectively drive expansion of adipocyte numbers to produce new fat cells and store saturated fatty acids, enabling homeostasis of healthy fat tissue.


Subject(s)
Adipogenesis , Cilia/metabolism , Fatty Acids, Omega-3/pharmacology , Receptors, G-Protein-Coupled/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipose Tissue, White/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Cilia/drug effects , Cyclic AMP/metabolism , Docosahexaenoic Acids/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism
2.
Mol Cell ; 73(3): 446-457.e6, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30612880

ABSTRACT

Multisite phosphorylation of kinases can induce on-off or graded regulation of catalytic activity; however, its influence on substrate specificity remains unclear. Here, we show that multisite phosphorylation of ribosomal protein S6 kinase 1 (S6K1) alters target selection. Agonist-inducible phosphorylation of glutamyl-prolyl tRNA synthetase (EPRS) by S6K1 in monocytes and adipocytes requires not only canonical phosphorylation at Thr389 by mTORC1 but also phosphorylation at Ser424 and Ser429 in the C terminus by cyclin-dependent kinase 5 (Cdk5). S6K1 phosphorylation at these additional sites induces a conformational switch and is essential for high-affinity binding and phosphorylation of EPRS, but not canonical S6K1 targets, e.g., ribosomal protein S6. Unbiased proteomic analysis identified additional targets phosphorylated by multisite phosphorylated S6K1 in insulin-stimulated adipocytes-namely, coenzyme A synthase, lipocalin 2, and cortactin. Thus, embedded within S6K1 is a target-selective kinase phospho-code that integrates signals from mTORC1 and Cdk5 to direct an insulin-stimulated, post-translational metabolon determining adipocyte lipid metabolism.


Subject(s)
Adipocytes/enzymology , Lipid Metabolism , Myeloid Cells/enzymology , Protein Processing, Post-Translational , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cyclin-Dependent Kinase 5/metabolism , Enzyme Activation , HEK293 Cells , Hep G2 Cells , Humans , Insulin/pharmacology , Interferon-gamma/pharmacology , Lipid Metabolism/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Myeloid Cells/drug effects , Phosphorylation , Proteomics/methods , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Substrate Specificity , U937 Cells
3.
J Biol Chem ; 300(9): 107721, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39214307

ABSTRACT

Obesity has emerged as a major health risk on a global scale. Hinokiflavone (HF), a natural small molecule, extracted from plants like cypress, exhibits diverse chemical structures and low synthesis costs. Using high-fat diet-induced obese mice models, we found that HF suppresses obesity by inducing apoptosis in adipose tissue. Adipocyte apoptosis helps maintain tissue health by removing aging, damaged, or excess cells in adipose tissue, which is crucial in preventing obesity and metabolic diseases. We found that HF can specifically bind to insulin-like growth factor 2 mRNA binding protein 2 to promote the stability of N6-methyladenosine-modified Bim, inducing mitochondrial outer membrane permeabilization. Mitochondrial outer membrane permeabilization leads to Caspase9/3-mediated adipocyte mitochondrial apoptosis, alleviating obesity induced by a high-fat diet. The proapoptotic effect of HF offers a controlled means for weight loss. This study reveals the potential of small molecule HF in developing new therapeutic approaches in drug development and biomedical research.


Subject(s)
Apoptosis , Bcl-2-Like Protein 11 , Diet, High-Fat , Obesity , Animals , Obesity/metabolism , Obesity/pathology , Obesity/drug therapy , Obesity/etiology , Apoptosis/drug effects , Diet, High-Fat/adverse effects , Mice , Bcl-2-Like Protein 11/metabolism , Bcl-2-Like Protein 11/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Male , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Mice, Inbred C57BL , Adipocytes/metabolism , Adipocytes/drug effects , Adipocytes/pathology , Humans
4.
FASEB J ; 38(14): e23733, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38995329

ABSTRACT

High-quality fat (HQF) improves the survival rate of fat and volumetric filling compared to traditional Coleman fat. However, this HQF strategy inevitably leads to a significant amount of unused fat being wasted. "CEFFE" (cell-free fat extract) is an acellular aqueous-phase liquid, rich in bioactive proteins. The remaining fat from preparing HQF can be further processed into CEFFE to promote the survival of HQF. HQF was obtained and the remaining fat was processed into CEFFE, then HQF was transplanted subcutaneously in nude mice. Animal studies showed that CEFFE significantly improved the survival rate of HQF. Histological analysis revealed that CEFFE improved the survival rate of HQF, by enhancing cell proliferation activity, reducing apoptosis, increasing angiogenesis, and improving the inflammatory state. Under simulated anaerobic conditions, CEFFE also improved the viability of HQF. In vitro, studies demonstrated that CEFFE enhanced the survival rate of HQF through multiple mechanisms. Transcriptomic analysis and qPCR showed that CEFFE increased the expression of angiogenesis-related genes in ADSCs while enhancing their proliferation-related gene expression and suppressing the expression of three differentiation-related genes. Moreover, functional experiments demonstrated that CEFFE-induced ADSCs exhibited stronger proliferation and adipogenic differentiation abilities. Tube formation and migration assays revealed that CEFFE promoted tube formation and migration of HUVECs, indicating its inherent pro-angiogenic properties. CEFFE facilitated the development of M0 to M2 macrophages, suggesting its role in improving the inflammatory state. This innovative clinical strategy optimizes HQF transplantation strategy, minimizing fat wastage and enhancing the efficiency of fat utilization.


Subject(s)
Cell Proliferation , Mice, Nude , Animals , Mice , Cell Proliferation/drug effects , Adipose Tissue/metabolism , Adipose Tissue/cytology , Cell Survival/drug effects , Cell Differentiation/drug effects , Humans , Male , Apoptosis/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Adipocytes/cytology
5.
Mol Cell ; 65(2): 260-271, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28107648

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation) is a post-translational modification of proteins mediated by PARP family members, such as PARP-1. Although PARylation has been studied extensively, few examples of definitive biological roles for site-specific PARylation have been reported. Here we show that C/EBPß, a key pro-adipogenic transcription factor, is PARylated by PARP-1 on three amino acids in a conserved regulatory domain. PARylation at these sites inhibits C/EBPß's DNA binding and transcriptional activities and attenuates adipogenesis in various genetic and cell-based models. Interestingly, PARP-1 catalytic activity drops precipitously during the first 48 hr of differentiation, corresponding to a release of C/EBPß from PARylation-mediated inhibition. This promotes the binding of C/EBPß at enhancers controlling the expression of adipogenic target genes and continued differentiation. Depletion or chemical inhibition of PARP-1, or mutation of the PARylation sites on C/EBPß, enhances these early adipogenic events. Collectively, our results provide a clear example of how site-specific PARylation drives biological outcomes.


Subject(s)
Adipocytes/enzymology , Adipogenesis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Embryonic Stem Cells/enzymology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/genetics , DNA/genetics , DNA/metabolism , Embryonic Stem Cells/drug effects , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NIH 3T3 Cells , Phenotype , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Binding , Protein Domains , RNA Interference , Signal Transduction , Time Factors , Transcription, Genetic/drug effects , Transcriptional Activation , Transfection
6.
J Biol Chem ; 299(2): 102804, 2023 02.
Article in English | MEDLINE | ID: mdl-36529290

ABSTRACT

Fibroblast growth factor (FGF) is a multifunctional protein that exhibits a wide range of biological effects. Most commonly, it acts as a mitogen, but it also has regulatory, morphological, and endocrine effects. The four receptor subtypes of FGF are activated by more than 20 different FGF ligands. FGF2, one of the FGF ligands, is an essential factor for cell culture in stem cells for regenerative medicine; however, recombinant FGF2 is extremely unstable. Here, we successfully generated homobivalent agonistic single-domain antibodies (variable domain of heavy chain of heavy chain antibodies referred to as VHHs) that bind to domain III and induce activation of the FGF receptor 1 and thus transduce intracellular signaling. This agonistic VHH has similar biological activity (EC50) as the natural FGF2 ligand. Furthermore, we determined that the agonistic VHH could support the proliferation of human-induced pluripotent stem cells (PSCs) and human mesenchymal stem cells, which are PSCs for regenerative medicine. In addition, the agonistic VHH could maintain the ability of mesenchymal stem cells to differentiate into adipocytes or osteocytes, indicating that it could maintain the properties of PSCs. These results suggest that the VHH agonist may function as an FGF2 mimetic in cell preparation of stem cells for regenerative medicine with better cost effectiveness.


Subject(s)
Fibroblast Growth Factor 2 , Protein Domains , Receptor, Fibroblast Growth Factor, Type 1 , Single-Domain Antibodies , Humans , Adipocytes/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Ligands , Mesoderm/cytology , Mesoderm/drug effects , Osteocytes/drug effects , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regenerative Medicine , Signal Transduction/drug effects , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology
7.
Immunology ; 173(1): 76-92, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38720202

ABSTRACT

Our newly developed menthyl esters of valine and isoleucine exhibit anti-inflammatory properties beyond those of the well-known menthol in macrophages stimulated by lipopolysaccharide (LPS) and in a mouse model of colitis induced by sodium dextran sulfate. Unlike menthol, which acts primarily through the cold-sensitive TRPM8 channel, these menthyl esters displayed unique mechanisms that operate independently of this receptor. They readily penetrated target cells and efficiently suppressed LPS-stimulated tumour necrosis factor-alpha (Tnf) expression mediated by liver X receptor (LXR), a key nuclear receptor that regulates intracellular cholesterol and lipid balance. The menthyl esters showed affinity for LXR and enhanced the transcriptional activity through their non-competitive and potentially synergistic agonistic effect. This effect can be attributed to the crucial involvement of SCD1, an enzyme regulated by LXR, which is central to lipid metabolism and plays a key role in the anti-inflammatory response. In addition, we discovered that the menthyl esters showed remarkable efficacy in suppressing adipogenesis in 3T3-L1 adipocytes at the mitotic clonal expansion stage in an LXR-independent manner as well as in mice subjected to diet-induced obesity. These multiple capabilities of our compounds establish them as formidable allies in the fight against inflammation and obesity, paving the way for a range of potential therapeutic applications.


Subject(s)
Anti-Inflammatory Agents , Anti-Obesity Agents , Liver X Receptors , Obesity , Animals , Mice , Obesity/drug therapy , Obesity/metabolism , Liver X Receptors/metabolism , Liver X Receptors/agonists , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Adipogenesis/drug effects , Esters/chemistry , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Humans , Menthol/pharmacology , Mice, Inbred C57BL , Lipopolysaccharides , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Dextran Sulfate , Adipocytes/metabolism , Adipocytes/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , TRPM Cation Channels/metabolism
8.
Biochem Biophys Res Commun ; 734: 150459, 2024 Nov 19.
Article in English | MEDLINE | ID: mdl-39083977

ABSTRACT

Abnormal adipose tissue formation is associated with metabolic disorders such as obesity, diabetes, and liver and cardiovascular diseases. Thus, identifying the novel factors that control adipogenesis is crucial for understanding these conditions and developing targeted treatments. In this study, we identified the melanosome-related factor MLPH as a novel adipogenic factor. MLPH was induced during the adipogenesis of 3T3-L1 cells and human mesenchymal stem cells. Although MLPH did not affect lipid metabolism, such as lipogenesis or lipolysis, adipogenesis was severely impaired by MLPH depletion. We observed that MLPH prevented excess reactive oxygen species (ROS) accumulation and lipid peroxidation during adipogenesis and in mature adipocytes. In addition, increased MLPH expression was observed under cirrhotic conditions in liver cancer cells and its overexpression also reduced ROS and lipid peroxidation. Our findings demonstrate that MLPH is a novel adipogenic factor that maintains redox homeostasis by preventing lipid peroxidation and ROS accumulation, which could lead to metabolic diseases.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Homeostasis , Lipid Peroxidation , Oxidation-Reduction , Reactive Oxygen Species , Lipid Peroxidation/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Humans , Animals , Mice , Reactive Oxygen Species/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology
9.
Biochem Biophys Res Commun ; 721: 150129, 2024 08 20.
Article in English | MEDLINE | ID: mdl-38762933

ABSTRACT

Melanoma, the deadliest skin cancer, originates from epidermal melanocytes. The influence of preadipocytes on melanoma is less understood. We co-cultured mouse melanoma B16 cells with 3T3L1 preadipocytes to form mixed spheroids and observed increased melanoma proliferation and growth compared to B16-only spheroids. Metastasis-related proteins YAP, TAZ, and PD-L1 levels were also higher in mixed spheroids. Treatment with exosome inhibitor GW4869 halted melanoma growth and reduced expression of these proteins, suggesting exosomal crosstalk between B16 and 3T3L1 cells. MiR-155 expression was significantly higher in mixed spheroids, and GW4869 reduced its levels. Additionally, co-culturing with Raw264.7 macrophage cells increased M2 markers IL-4 and CD206 in Raw264.7 cells, effects that were diminished by GW4869. These results indicate that preadipocytes may enhance melanoma progression and metastasis via exosomal interactions.


Subject(s)
Adipocytes , Exosomes , Macrophages , Melanoma, Experimental , Tumor Microenvironment , Animals , Mice , Macrophages/metabolism , Macrophages/pathology , Macrophages/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipocytes/drug effects , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , RAW 264.7 Cells , Exosomes/metabolism , Coculture Techniques , Disease Progression , 3T3-L1 Cells , Benzylidene Compounds/pharmacology , Aniline Compounds/pharmacology , Cell Proliferation/drug effects , Melanoma/pathology , Melanoma/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Cell Line, Tumor , MicroRNAs/metabolism , MicroRNAs/genetics
10.
Biochem Biophys Res Commun ; 731: 150279, 2024 10 30.
Article in English | MEDLINE | ID: mdl-39018972

ABSTRACT

This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARß, PPARγ, C/EBPα, C/EBPß, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.


Subject(s)
3T3-L1 Cells , AMP-Activated Protein Kinases , Gastrointestinal Microbiome , Lacticaseibacillus paracasei , Obesity , Probiotics , Animals , Obesity/metabolism , Obesity/microbiology , Obesity/prevention & control , Gastrointestinal Microbiome/drug effects , Mice , Lacticaseibacillus paracasei/metabolism , Male , Rats , AMP-Activated Protein Kinases/metabolism , Probiotics/administration & dosage , Probiotics/pharmacology , Rats, Sprague-Dawley , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Lipid Metabolism/drug effects , Liver/metabolism , Anti-Obesity Agents/pharmacology
11.
Int J Obes (Lond) ; 48(7): 923-933, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38388800

ABSTRACT

BACKGROUND/OBJECTIVES: Obesity and its associated metabolic diseases are increasing globally. Sedentary lifestyle, high caloric diet, and genetic predisposition are known to contribute to the onset of obesity. It is increasingly recognized that exposure to environmental chemicals such as Bisphenol A (BPA) may also play a significant role. BPA has been correlated with an array of adverse health effects, including obesity and metabolic disorders. Due to public concern, manufacturers are replacing BPA with structural analogues for which there is limited toxicological data. The objective of this study was to assess the effects of these BPA analogues on adipogenesis. METHODS: The adipogenic effects of Tetra Methyl Bisphenol F (TMBPF), Bisphenol F (BPF), Bisphenol AP (BPAP), and fluorine-9-bisphenol (BHPF) were evaluated in murine 3T3-L1 cells. The cells were treated with BPA and its analogues at concentrations from 0.01 µM to 20 µM, throughout differentiation, in the absence of Dexamethasone (Dex). Lipid accumulation, mRNA and protein levels of adipogenic markers was assessed. RESULTS: We found that TMBPF, BPF and BPA increased 3T3-L1 lipid accumulation and the expression levels of adipogenic markers lipoprotein lipase (Lpl), fatty acid binding protein 4 (Fabp4) and perilipin (Plin) (1-20 µM; p < 0.05), whereas BHPF and BPAP had no effect in this model. Further, TMBPF induced adipogenesis to a greater extent than all the other chemicals including BPA (1-20 µM; p < 0.05). The effect mediated by TMBPF on expression levels of Fabp4, but not Plin, is likely mediated via peroxisome proliferator-activated receptor (PPAR) γ activation. CONCLUSIONS: Of the BPA analogues tested, BPF was most similar to BPA in its effects, while TMBPF was most adipogenic. In addition, TMBPF is likely a PPARγ agonist, it is likely an obesogenic chemical and may be a metabolic disruptor.


Subject(s)
3T3-L1 Cells , Adipogenesis , Benzhydryl Compounds , Obesity , Phenols , Animals , Mice , Phenols/pharmacology , Adipogenesis/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Cell Differentiation/drug effects , Fatty Acid-Binding Proteins/metabolism , PPAR gamma/metabolism , Endocrine Disruptors/pharmacology
12.
Toxicol Appl Pharmacol ; 486: 116937, 2024 May.
Article in English | MEDLINE | ID: mdl-38643950

ABSTRACT

Selective Serotonin Reuptake Inhibitors (SSRIs) are widely used medications for the treatment of major depressive disorder. However, long-term SSRI use has been associated with weight gain and altered lipid profiles. These findings suggest that SSRIs may have negative effects on metabolism. Exposure to certain chemicals called 'obesogens' is known to promote lipid accumulation and obesity by modulating adipogenesis. Here, we investigated whether citalopram (CIT) and sertraline (SER) interfere with the process of adipogenesis, using human mesenchymal stem cells (MSCs) in a 2D and a 3D model. Assessment of intracellular lipid accumulation by fluorescence staining was used as a measure for enhanced adipogenesis. To explore possible mechanisms behind SSRIs' effects, receptor mediated activity was studied using responsive cell lines for various nuclear receptors. Furthermore, RNA sequencing was performed in the 3D model, followed by differential gene expression and pathway analysis. A dose dependent increase in lipid accumulation was observed in both models with CIT and SER. For the 3D model, the effect was seen in a range close to reported steady-state plasma concentrations (0.065-0.65 µM for SER and 0.12-0.92 µM for CIT). Pathway analysis revealed unexpected results of downregulation in adipogenesis-related pathways and upregulation in phospholipids and lysosomal pathways. This was confirmed by an observed increase in lysosomes in the 2D model. Our findings suggest lysosomal dysfunction and disrupted lipid metabolism in mature adipocytes, leading to excessive phospholipid synthesis. Moreover, important adipogenic processes are inhibited, potentially leading to dysfunctional adipocytes, which might have implications in the maintenance of a healthy metabolic balance.


Subject(s)
Adipogenesis , Antidepressive Agents , Citalopram , Lipid Metabolism , Mesenchymal Stem Cells , Selective Serotonin Reuptake Inhibitors , Sertraline , Adipogenesis/drug effects , Sertraline/pharmacology , Sertraline/toxicity , Humans , Citalopram/pharmacology , Lipid Metabolism/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/toxicity , Antidepressive Agents/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Cells, Cultured , Dose-Response Relationship, Drug
13.
Diabet Med ; 41(7): e15319, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38711201

ABSTRACT

INTRODUCTION: Berberine (BBR) is an alkaloid found in plants. It has neuroprotective, anti-inflammatory and lipid-lowering activity. However, the efficacy of treatment with BBR and the mechanisms through which it acts need further study. AIMS: This study investigated the therapeutic effects and the mechanism of action of BBR on obesity-induced insulin resistance in peripheral tissues. METHODS: High-fat-fed C57BL/6J mice and low-fat-fed C57BL/6J mice with miR-27a overexpression were given BBR intervention (100 mg/kg, po), and the oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed. Palmitic acid-stimulated hypertrophic adipocyte models were treated with BBR (10 µM). Related indicators and protein expression levels were examined. RESULTS: The AUCs of the OGTT and the ITT in the BBR intervention group were reduced significantly (p < 0.01) (p < 0.05), and the serum biochemical parameters, including FBG, TC, TG and LDL-C were significantly reduced after BBR intervention. In the in vitro experiments, the triglyceride level and volume of lipid droplets decreased significantly after BBR intervention (p < 0.01) (p < 0.05). Likewise, BBR ameliorates skeletal muscle and pancreas insulin signalling pathways in vivo and in vitro. DISCUSSION: The results showed that BBR significantly ameliorated insulin resistance, reduced body weight and percent body fat and improved serum biochemical parameters in mice. Likewise, BBR reduced triglyceride level and lipid droplet volume in hypertrophic adipocytes, BBR improved obesity effectively. Meanwhile, BBR ameliorated the histomorphology of the pancreas, and skeletal muscle and pancreas insulin related signalling pathways of islets in in vitro and in vivo experiments. The results further demonstrated that BBR inhibited miR-27a levels in serum from obese mice and supernatant of hypertrophic adipocytes. miR-27a overexpression in low-fat fed mice indicated that miR-27a caused insulin resistance, and BBR intervention significantly improved the miR-27a induced insulin resistance status. CONCLUSION: This study demonstrates the important role of BBR in obesity-induced peripheral insulin resistance and suggest that the mechanism of its effect may be inhibition of miR-27a secretion.


Subject(s)
Berberine , Insulin Resistance , Mice, Inbred C57BL , MicroRNAs , Obesity , Berberine/pharmacology , Berberine/therapeutic use , Animals , Obesity/metabolism , Obesity/drug therapy , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Male , Diet, High-Fat , Adipocytes/metabolism , Adipocytes/drug effects , Glucose Tolerance Test
14.
Cell Commun Signal ; 22(1): 443, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285292

ABSTRACT

BACKGROUND: Epithelial ovarian cancer (EOC) is the deadliest gynaecological cancer with high mortality rates driven by the common development of resistance to chemotherapy. EOC frequently invades the omentum, an adipocyte-rich organ of the peritoneum and omental adipocytes have been implicated in promoting disease progression, metastasis and chemoresistance. The signalling mechanisms underpinning EOC omentum tropism have yet to be elucidated. METHODS: Three-dimensional co-culture models were used to explore adipocyte-EOC interactions. The impact of adipocytes on EOC proliferation, response to therapy and invasive capacity was assessed. Primary adipocytes and omental tissue were isolated from patients with ovarian malignancies and benign ovarian neoplasms. Exosomes were isolated from omentum tissue conditioned media and the effect of omentum-derived exosomes on EOC evaluated. Exosomal microRNA (miRNA) sequencing was used to identify miRNAs abundant in omental exosomes and EOC cells were transfected with highly abundant miRNAs miR-21, let-7b, miR-16 and miR-92a. RESULTS: We demonstrate the capacity of adipocytes to induce an invasive phenotype in EOC populations through driving epithelial-to-mesenchymal transition (EMT). Exosomes secreted by omental tissue of ovarian cancer patients, as well as patients without malignancies, induced proliferation, upregulated EMT markers and reduced response to paclitaxel therapy in EOC cell lines and HGSOC patient samples. Analysis of the omentum-derived exosomes from cancer patients revealed highly abundant miRNAs that included miR-21, let-7b, miR-16 and miR-92a that promoted cancer cell proliferation and protection from chemotherapy when transfected in ovarian cancer cells. CONCLUSIONS: These observations highlight the capacity of omental adipocytes to generate a pro-tumorigenic and chemoprotective microenvironment in ovarian cancer and other adipose-related malignancies.


Subject(s)
Adipocytes , Exosomes , MicroRNAs , Neoplasm Invasiveness , Ovarian Neoplasms , Paclitaxel , Female , Exosomes/metabolism , Humans , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Adipocytes/metabolism , Adipocytes/drug effects , Adipocytes/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Omentum/pathology , Omentum/metabolism , Cell Proliferation/drug effects , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/drug effects
15.
Nitric Oxide ; 149: 75-84, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38879114

ABSTRACT

Obesity is commonly linked with white adipose tissue (WAT) dysfunction, setting off inflammation and oxidative stress, both key contributors to the cardiometabolic complications associated with obesity. To improve metabolic and cardiovascular health, countering these inflammatory and oxidative signaling processes is crucial. Offering potential in this context, the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) by nitro-fatty acids (NO2-FA) promote diverse anti-inflammatory signaling and counteract oxidative stress. Additionally, we previously highlighted that nitro-oleic acid (NO2-OA) preferentially accumulates in WAT and provides protection against already established high fat diet (HFD)-mediated impaired glucose tolerance. The precise mechanism accounting for these protective effects remained largely unexplored until now. Herein, we reveal that protective effects of improved glucose tolerance by NO2-OA is absent when Nrf2 is specifically ablated in adipocytes (ANKO mice). NO2-OA treatment did not alter body weight between ANKO and littermate controls (Nrf2fl/fl) mice on both the HFD and low-fat diet (LFD). As expected, at day 76 (before NO2-OA treatment) and notably at day 125 (daily treatment of 15 mg/kg NO2-OA for 48 days), both HFD-fed Nrf2fl/fl and ANKO mice exhibited increased fat mass and reduced lean mass compared to LFD controls. However, throughout the NO2-OA treatment, no distinction was observed between Nrf2fl/fl and ANKO in the HFD-fed mice as well as in the Nrf2fl/fl mice fed a LFD. Glucose tolerance tests revealed impaired glucose tolerance in HFD-fed Nrf2fl/fl and ANKO compared to LFD-fed Nrf2fl/fl mice. Notably, NO2-OA treatment improved glucose tolerance in HFD-fed Nrf2fl/fl but did not yield the same improvement in ANKO mice at days 15, 30, and 55 of treatment. Unraveling the pathways linked to NO2-OA's protective effects in obesity-mediated impairment in glucose tolerance is pivotal within the realm of precision medicine, crucially propelling future applications and refining novel drug-based strategies.


Subject(s)
Adipocytes , Diet, High-Fat , NF-E2-Related Factor 2 , Obesity , Animals , NF-E2-Related Factor 2/metabolism , Obesity/metabolism , Obesity/drug therapy , Diet, High-Fat/adverse effects , Mice , Adipocytes/metabolism , Adipocytes/drug effects , Male , Mice, Inbred C57BL , Glucose Intolerance/metabolism , Oleic Acids/pharmacology , Mice, Knockout
16.
Prostaglandins Other Lipid Mediat ; 174: 106874, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002709

ABSTRACT

Dahuang Huanglian Xiexin Decoction (DHXD) is the representative clinical formula for treating epigastric oppression. In this study, we aim to explore the effect of DHXD on obesity and attempt to investigate its potential mechanism. 3T3-L1 preadipocytes were differentiated and high-fat diet-induced obese rat model was established. DHXD was used for treatment and tunicamycin, the activator of endoplasmic reticulum (ER) stress, was adopted to investigate the related regulatory mechanism. Cell viability was evaluated using CCK-8 assay. Oil-Red O staining was performed to determine lipid accumulation. Glycerol production and Triglyceride content were measured using their commercial kits. Western blot was conducted to examine the expression of critical proteins. Results indicated that DHXD could greatly reduce intracellular lipid droplets and triglyceride in differentiated 3T3-L1 cells. Moreover, the elevated expression of mature adipocytes markers, PPARγ, aP2, during adipogenesis was decreased by DHXD treatment. In addition, DHXD aggravated the lipolysis in differentiated 3T3-L1 cells, as evidenced by the upregulated ATGL expression and the downregulated HSL expression. Besides, DHXD inhibited endoplasmic reticulum (ER) stress in 3T3-L1 cells. Further experiments indicated that the impacts of DHXD on adipocyte differentiation and lipid degradation were partly abolished by tunicamycin. Finally, DHXD alleviated lipid accumulation and ER stress in obese rats. In conclusion, DHXD ameliorates obesity via modulating adipocyte differentiation and lipid degradation through inhibiting ER stress.


Subject(s)
3T3-L1 Cells , Adipocytes , Cell Differentiation , Drugs, Chinese Herbal , Endoplasmic Reticulum Stress , Obesity , Animals , Endoplasmic Reticulum Stress/drug effects , Mice , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Adipocytes/drug effects , Adipocytes/metabolism , Drugs, Chinese Herbal/pharmacology , Rats , Male , Cell Differentiation/drug effects , Lipid Metabolism/drug effects , Rats, Sprague-Dawley , Diet, High-Fat , Adipogenesis/drug effects
17.
Mol Biol Rep ; 51(1): 562, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38644407

ABSTRACT

BACKGROUND: Obesity is associated with a wide variety of metabolic disorders that impose significant burdens on patients and society. The "browning" phenomenon in white adipose tissue (WAT) has emerged as a promising therapeutic strategy to combat metabolic disturbances. However, though the anti-diabetic drug dapagliflozin (DAPA) is thought to promote "browning," the specific mechanism of this was previously unclear. METHODS: In this study, C57BL/6 J male mice were used to establish an obesity model by high-fat diet feeding, and 3T3-L1 cells were used to induce mature adipocytes and to explore the role and mechanism of DAPA in "browning" through a combination of in vitro and in vivo experiments. RESULTS: The results show that DAPA promotes WAT "browning" and improves metabolic disorders. Furthermore, we discovered that DAPA activated "browning" through the fibroblast growth factor receptors 1-liver kinase B1-adenosine monophosphate-activated protein kinase signaling pathway. CONCLUSION: These findings provide a rational basis for the use of DAPA in treating obesity by promoting the browning of white adipose tissue.


Subject(s)
Adipose Tissue, White , Benzhydryl Compounds , Glucosides , Protein Serine-Threonine Kinases , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Animals , Male , Mice , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , AMP-Activated Protein Kinases/metabolism , Benzhydryl Compounds/pharmacology , Diet, High-Fat , Glucosides/pharmacology , Mice, Inbred C57BL , Obesity/metabolism , Obesity/drug therapy , Protein Serine-Threonine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects
18.
Mol Biol Rep ; 51(1): 785, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951450

ABSTRACT

BACKGROUND: Kaempferia parviflora Wall. ex. Baker (KP) has been reported to exhibit anti-obesity effects. However, the detailed mechanism of the anti-obesity effect of KP extract (KPE) is yet to be clarified. Here, we investigated the effect of KPE and its component polymethoxyflavones (PMFs) on the adipogenic differentiation of human mesenchymal stem cells (MSCs). METHODS AND RESULTS: KPE and PMFs fraction (2.5 µg/mL) significantly inhibited lipid and triacylglyceride accumulation in MSCs; lipid accumulation in MSCs was suppressed during the early stages of differentiation (days 0-3) but not during the mid (days 3-7) or late (days 7-14) stages. Treatment with KPE and PMFs fractions significantly suppressed peroxisome proliferator-activated receptor-γ (PPARγ), CCAAT/enhancer binding protein α (C/EBPα), and various adipogenic metabolic factors. Treatment with KPE and PMFs fraction induced the activation of AMP-activated protein kinase (AMPK) signaling, and pretreatment with an AMPK signaling inhibitor significantly attenuated KPE- and PMFs fraction-induced suppression of lipid formation. CONCLUSIONS: Our findings demonstrate that KPE and PMFs fraction inhibit lipid formation by inhibiting the differentiation of undifferentiated MSCs into adipocyte lineages via AMPK signaling, and this may be the mechanism underlying the anti-obesity effects of KPE and PMFs. Our study lays the foundation for the elucidation of the anti-obesity mechanism of KPE and PMFs.


Subject(s)
AMP-Activated Protein Kinases , Adipogenesis , Cell Differentiation , Flavones , Mesenchymal Stem Cells , Plant Extracts , Signal Transduction , Zingiberaceae , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Adipogenesis/drug effects , Plant Extracts/pharmacology , Zingiberaceae/chemistry , AMP-Activated Protein Kinases/metabolism , Flavones/pharmacology , Cell Differentiation/drug effects , Signal Transduction/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cells, Cultured
19.
J Nat Prod ; 87(5): 1441-1453, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38722764

ABSTRACT

Herein, we report an extensive phytochemical study on the whole plant of Drymaria cordata, which led to the isolation of ten new orbitides, named drymariamides A-J (1-10). Compounds 2, 3, and 5 incorporate rare residues of noncanonical amino acids of kynurenine (Kyn) or 3a-hydroxypyrroloindoline (HPI). Their structures with absolute configurations were elucidated by a combination of spectroscopic analysis, advanced Marfey's method, X-ray diffraction, and electronic circular dichroism analysis. Compounds 1-10 exhibited antiadipogenic effects in 3T3-L1 adipocytes, and the most potent compound 7 showed an EC50 value of 1.17 ± 0.19 µM.


Subject(s)
3T3-L1 Cells , Amino Acids , Peptides, Cyclic , Animals , Mice , Amino Acids/chemistry , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Adipogenesis/drug effects , Adipocytes/drug effects , Adipocytes/metabolism
20.
J Nat Prod ; 87(7): 1673-1681, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38597733

ABSTRACT

Enhanced glucose uptake in insulin-sensitive tissues is one of the therapeutic strategies to ameliorate hyperglycemia and maintain glucose homeostasis in type 2 diabetes. This study disclosed the role of fungal depsidones in glucose uptake and the underlying mechanism in 3T3-L1 adipocytes. Depsidones, including nidulin, nornidulin, and unguinol, isolated from Aspergillus unguis, stimulate glucose uptake in adipocytes. Compared to the others, nidulin exhibited an upward trend in glucose uptake. The effect of nidulin was found to be dose- and time-dependent. Nidulin also enhanced insulin- and metformin-stimulated glucose uptake. Upregulation of GLUT4 expression and AKT and AMPK phosphorylation were observed with nidulin treatment. Blockage of AKT, but not AMPK, phosphorylation was largely accompanied by diminished glucose uptake. In agreement, nidulin triggered the translocation of GLUT4 to the plasma membrane. Importantly, nidulin elevated glucose uptake associated with increased AKT phosphorylation in insulin-resistant adipocytes. Taken together, nidulin could stimulate glucose uptake mainly through AKT-dependent GLUT4 translocation, serving as a seed compound in drug discovery for type 2 diabetes.


Subject(s)
3T3-L1 Cells , Adipocytes , Glucose Transporter Type 4 , Glucose , Proto-Oncogene Proteins c-akt , Animals , Mice , Adipocytes/metabolism , Adipocytes/drug effects , Glucose/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glucose Transporter Type 4/metabolism , Phosphorylation , Lactones/pharmacology , Lactones/chemistry , Molecular Structure , Insulin/metabolism , Depsides/pharmacology , Metformin/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL