Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.842
Filter
1.
Nutrients ; 16(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38999918

ABSTRACT

This study aimed to investigate the therapeutic potential of Citrullus mucosospermus extract (CME) in counteracting adipogenesis and its associated metabolic disturbances in murine models. In vitro experiments utilizing 3T3-L1 preadipocytes revealed that CME potently inhibited adipocyte differentiation, as evidenced by a dose-dependent reduction in lipid droplet formation. Remarkably, CME also attenuated glucose uptake and intracellular triglyceride accumulation in fully differentiated adipocytes, suggesting its ability to modulate metabolic pathways in mature adipose cells. Translating these findings to an in vivo setting, we evaluated the effects of CME in C57BL/6N mice fed a high-fat diet (HFD) for 10 weeks. CME administration, concomitantly with the HFD, resulted in a significant attenuation of body weight gain compared to the HFD control group. Furthermore, CME treatment led to substantial reductions in liver weight, total fat mass, and deposits of visceral and retroperitoneal adipose tissue, underscoring its targeted impact on adipose expansion. Histological analyses revealed the remarkable effects of CME on hepatic steatosis. While the HFD group exhibited severe lipid accumulation within liver lobules, CME dose-dependently mitigated this pathology, with the highest dose virtually abolishing hepatic fat deposition. An examination of adipose tissue revealed a progressive reduction in adipocyte hypertrophy upon CME treatment, culminating in a near-normalization of adipocyte morphology at the highest dose. Notably, CME exhibited potent anti-inflammatory properties, significantly attenuating the upregulation of pro-inflammatory cytokines' mRNA levels (TNF-α, IL-1ß and IL-6) in the livers of HFD-fed mice. This suggests a potential mechanism through which CME may exert protective effects against inflammation associated with obesity and fatty liver disease.


Subject(s)
3T3-L1 Cells , Adipogenesis , Diet, High-Fat , Mice, Inbred C57BL , Plant Extracts , Weight Gain , Animals , Diet, High-Fat/adverse effects , Plant Extracts/pharmacology , Mice , Weight Gain/drug effects , Male , Adipogenesis/drug effects , Adipocytes/drug effects , Obesity , Liver/drug effects , Liver/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism
2.
Adipocyte ; 13(1): 2381262, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39039652

ABSTRACT

Obesity is a low-grade chronic inflammation induced by the pathological expansion of adipocytes which allows the development of obesity-associated metabolic diseases like type 2 diabetes mellitus (T2D) and non-alcoholic fatty liver disease (NAFLD). However, mechanisms regulating adipocyte inflammation remain poorly understood. Here, we observed that TRIM8 was upregulated in adipocyte inflammation and insulin resistance while DUSP14 was downregulated. TRIM8 deficiency and DUSP14 over-expression decreased the level of inflammatory cytokines, increased glucose uptake content, and improved insulin signalling transduction compared to LPS treatment alone. Conversely, silencing DUSP14 increased the expression of inflammatory cytokines. It decreased the glucose uptake content and the phosphorylation level of proteins involved in insulin signalling, further impairing insulin signalling and aggravating insulin resistance. Furthermore, The decreased level of inflammatory cytokines, increased glucose uptake, and improved insulin signalling transduction caused by TRIM8 deficiency were reversed by down-regulated DUSP14. Collectively, our findings revealed that TRIM8 can regulate adipocyte inflammation and insulin resistance by regulating the MAPKs pathway which is dependent on DUSP14.


Subject(s)
Adipocytes , Dual-Specificity Phosphatases , Inflammation , Insulin Resistance , Animals , Adipocytes/metabolism , Mice , Inflammation/metabolism , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , MAP Kinase Signaling System , 3T3-L1 Cells , Signal Transduction , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mice, Inbred C57BL
3.
Adipocyte ; 13(1): 2374062, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38953241

ABSTRACT

Obesity is associated with a low-grade chronic inflammatory process characterized by higher circulating TNFα levels, thus contributing to insulin resistance. This study evaluated the effect of silybin, the main bioactive component of silymarin, which has anti-inflammatory properties, on TNFα levels and its impact on glucose uptake in the adipocyte cell line 3T3-L1 challenged with two different inflammatory stimuli, TNFα or lipopolysaccharide (LPS). Silybin's pre-treatment effect was evaluated in adipocytes pre-incubated with silybin (30 or 80 µM) before challenging with the inflammatory stimuli (TNFα or LPS). For the post-treatment effect, the adipocytes were first challenged with the inflammatory stimuli and then post-treated with silybin. After treatments, TNFα production, glucose uptake, and GLUT4 protein expression were determined. Both inflammatory stimuli increased TNFα secretion, diminished GLUT4 expression, and significantly decreased glucose uptake. Silybin 30 µM only reduced TNFα secretion after the LPS challenge. Silybin 80 µM as post-treatment or pre-treatment decreased TNFα levels, improving glucose uptake. However, glucose uptake enhancement induced by silybin did not depend on GLUT4 protein expression. These results show that silybin importantly reduced TNFα levels and upregulates glucose uptake, independently of GLUT4 protein expression.


Subject(s)
3T3-L1 Cells , Adipocytes , Glucose , Lipopolysaccharides , Silybin , Tumor Necrosis Factor-alpha , Animals , Silybin/pharmacology , Mice , Tumor Necrosis Factor-alpha/metabolism , Glucose/metabolism , Adipocytes/metabolism , Adipocytes/drug effects , Lipopolysaccharides/pharmacology , Glucose Transporter Type 4/metabolism , Silymarin/pharmacology
4.
Clin Epigenetics ; 16(1): 96, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033117

ABSTRACT

BACKGROUND: Obesity is a major health burden. Preadipocytes proliferate and differentiate in mature adipocytes in the adipogenic process, which could be a potential therapeutic approach for obesity. Deficiency of SIRT6, a stress-responsive protein deacetylase and mono-ADP ribosyltransferase enzyme, blocks adipogenesis. Mutants of SIRT6 (N308K/A313S) were recently linked to the in the long lifespan Ashkenazi Jews. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect adipogenesis at the transcriptional and epigenetic level. METHODS: We analyzed the role of SIRT6 wild-type (WT) or SIRT6 centenarian-associated mutant (N308K/A313S) overexpression in adipogenesis, by creating stably transduced preadipocyte cell lines using lentivirus on the 3T3-L1 model. Histone post-translational modifications (PTM: acetylation, methylation) and transcriptomic changes were analyzed by mass spectrometry (LC-MS/MS) and RNA-Seq, respectively, in 3T3-L1 adipocytes. In addition, the adipogenic process and related signaling pathways were investigated by bioinformatics and biochemical approaches. RESULTS: Overexpression of centenarian-associated SIRT6 mutant increased adipogenic differentiation to a similar extent compared to the WT form. However, it triggered distinct histone PTM profiles in mature adipocytes, with significantly higher acetylation levels, and activated divergent transcriptional programs, including those dependent on signaling related to the sympathetic innervation and to PI3K pathway. 3T3-L1 mature adipocytes overexpressing SIRT6 N308K/A313S displayed increased insulin sensitivity in a neuropeptide Y (NPY)-dependent manner. CONCLUSIONS: SIRT6 N308K/A313S overexpression in mature adipocytes ameliorated glucose sensitivity and impacted sympathetic innervation signaling. These findings highlight the importance of targeting SIRT6 enzymatic activities to regulate the co-morbidities associated with obesity.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Epigenesis, Genetic , Sirtuins , Sirtuins/genetics , Sirtuins/metabolism , Mice , Adipocytes/metabolism , Animals , Epigenesis, Genetic/genetics , Adipogenesis/genetics , Humans , Mutation , Obesity/genetics , Obesity/metabolism , Protein Processing, Post-Translational/genetics , Histones/metabolism , Histones/genetics
5.
Sci Rep ; 14(1): 15064, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956395

ABSTRACT

Sargassum horneri (S. horneri), a brown seaweed excessively proliferating along Asian coastlines, are damaging marine ecosystems. Thus, this study aimed to enhance nutritional value of S. horneri through lactic acid bacteria fermentation to increase S. horneri utilization as a functional food supplement, and consequently resolve coastal S. horneri accumulation. S. horneri supplemented fermentation was most effective with Lactiplantibacillus pentosus SH803, thus this product (F-SHWE) was used for further in vitro studies. F-SHWE normalized expressions of oxidative stress related genes NF-κB, p53, BAX, cytochrome C, caspase 9, and caspase 3, while non-fermented S. horneri (SHWE) did not, in a H2O2-induced HT-29 cell model. Moreover, in an LPS-induced HT-29 cell model, F-SHWE repaired expressions of inflammation marker genes ZO1, IL1ß, IFNγ more effectively than SHWE. For further functional assessment, F-SHWE was also treated in 3T3-L1 adipocytes. As a result, F-SHWE decreased lipid accumulation, along with gene expression of adipogenesis markers PPARγ, C/EBPα, C/EBPß, aP2, and Lpl; lipogenesis markers Lep, Akt, SREBP1, Acc, Fas; inflammation markers IFN-γ and NF-κB. Notably, gene expression of C/EBPß, IFN-γ and NF-κB were suppressed only by F-SHWE, suggesting the enhancing effect of fermentation on obesity-related properties. Compositional analysis attributed the protective effects of F-SHWE to acetate, an organic acid significantly higher in F-SHWE than SHWE. Therefore, F-SHWE is a novel potential anti-obesity agent, providing a strategy to reduce excess S. horneri populations along marine ecosystems.


Subject(s)
3T3-L1 Cells , Adipocytes , Fermentation , Inflammation , Oxidative Stress , Sargassum , Sargassum/chemistry , Mice , Animals , Adipocytes/metabolism , Adipocytes/drug effects , Oxidative Stress/drug effects , Humans , Inflammation/metabolism , Lactobacillus pentosus/metabolism , HT29 Cells , Adipogenesis/drug effects
6.
Int J Nanomedicine ; 19: 5441-5458, 2024.
Article in English | MEDLINE | ID: mdl-38868593

ABSTRACT

Introduction: Quercetin (QUER), a flavonoid abundant in fruits and vegetables, is emerging as a promising alternative therapeutic agent for obesity treatment due to its antioxidant and anti-adipogenic properties. However, the clinical application of QUER is limited by its poor solubility, low bioavailability, and potential toxicity at high doses. To address these challenges, this study aims to develop an advanced drug delivery system using fluorescent mesoporous silica nanoparticles (FMSNs) coated with polydopamine (PDA) for the efficient and sustained delivery of QUER to inhibit adipogenesis. Methods: The research included the synthesis of PDA-coated FMSNs for encapsulation of QUER, characterization of their mesoporous structures, and systematic investigation of the release behavior of QUER. The DPPH assay was used to evaluate the sustained radical scavenging potential. Concentration-dependent effects on 3T3-L1 cell proliferation, cellular uptake and adipogenesis inhibition were investigated. Results: PDA-coated FMSNs exhibited well-aligned mesoporous structures. The DPPH assay confirmed the sustained radical scavenging potential, with FMSNs-QUER@PDA showing 53.92 ± 3.48% inhibition at 72 h, which was higher than FMSNs-QUER (44.66 ± 0.57%) and free QUER (43.37 ± 5.04%). Concentration-dependent effects on 3T3-L1 cells highlighted the enhanced efficacy of PDA-coated FMSNs for cellular uptake, with a 1.5-fold increase compared to uncoated FMSNs. Adipogenesis inhibition was also improved, with relative lipid accumulation of 44.6 ± 4.6%, 37.3 ± 4.6%, and 36.5 ± 7.3% at 2.5, 5, and 10 µM QUER concentrations, respectively. Conclusion: The study successfully developed a tailored drug delivery system, emphasizing sustained QUER release and enhanced therapeutic effects. FMSNs, especially when coated with PDA, exhibit promising properties for efficient QUER delivery, providing a comprehensive approach that integrates advanced drug delivery technology and therapeutic efficacy.


Subject(s)
3T3-L1 Cells , Adipogenesis , Delayed-Action Preparations , Drug Carriers , Indoles , Nanoparticles , Polymers , Quercetin , Silicon Dioxide , Quercetin/chemistry , Quercetin/pharmacology , Quercetin/pharmacokinetics , Quercetin/administration & dosage , Animals , Mice , Adipogenesis/drug effects , Silicon Dioxide/chemistry , Indoles/chemistry , Indoles/pharmacology , Indoles/pharmacokinetics , Indoles/administration & dosage , Nanoparticles/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/chemistry , Polymers/chemistry , Porosity , Drug Liberation , Cell Proliferation/drug effects , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/pharmacokinetics , Antioxidants/administration & dosage
7.
Int J Biol Macromol ; 272(Pt 1): 132738, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825269

ABSTRACT

Piperine (PIP) has been known for its pharmacological activities with low water solubility and poor dissolution, which limits its nutritional application. The purpose of this research was to enhance PIP stability, dispersibility and biological activity by preparing PIP nanoparticles using the wet-media milling approach combined with nanosuspension solidification methods of spray/freeze drying. Octenyl succinic anhydride (OSA)-modified waxy maize starch was applied as the stabilizer to suppress aggregation of PIP nanoparticles. The particle size, redispersibility, storage stability and in vitro release behavior of PIP nanoparticles were measured. The regulating effect on adipocyte differentiation was evaluated using 3T3-L1 cell model. Results showed that PIP nanoparticles had a reduced particle size of 60 ± 1 nm, increased release rate in the simulated gastric (SGF) and intestinal fluids (SIF) and enhanced inhibition effect on adipogenesis in 3T3-L1 cells compared with free PIP, indicating that PIP-loaded nanoparticles with improved stability and anti-adipogenic property were developed successfully by combining wet-media milling and drying methods.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Alkaloids , Benzodioxoles , Nanoparticles , Piperidines , Polyunsaturated Alkamides , Starch , Animals , Mice , Nanoparticles/chemistry , Polyunsaturated Alkamides/chemistry , Polyunsaturated Alkamides/pharmacology , Benzodioxoles/pharmacology , Benzodioxoles/chemistry , Piperidines/pharmacology , Piperidines/chemistry , Adipogenesis/drug effects , Alkaloids/chemistry , Alkaloids/pharmacology , Adipocytes/drug effects , Starch/chemistry , Starch/analogs & derivatives , Particle Size , Drug Liberation , Cell Differentiation/drug effects
8.
Nutrients ; 16(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38931278

ABSTRACT

Corn peptide (CP) is a short, naturally occurring, and physiologically active peptide generated from corn-protease-catalyzed hydrolysis. CP plays a role in preventing obesity-related disorders, but its impact on reducing inflammation is unknown. Hence, this study examined the possible protective effects of corn peptide powder (CPP) against the harmful effects of lipopolysaccharide (LPS), with a particular emphasis on reducing oxidative damage and inflammation in adipocytes. Hence, mature 3T3-L1 adipocytes underwent exposure to 10 ng/mL LPS, with or without CPP (10 and 20 µg/mL). LPS stimulation increased reactive oxygen species and superoxide anion generation. However, this effect was reduced in a dose-dependent manner by pretreatment with CPP. CPP treatment elevated the mRNA expressions of the antioxidant enzymes manganese superoxide dismutase (mnSOD) and glutathione peroxidase 1 (Gpx1) while reducing the mRNA expressions of the cytosolic reactive oxygen species indicators p40 and p67 (NADPH oxidase 2). In addition, CPP inhibited the monocyte chemoattractant protein-1, tumor necrosis factor-alpha, Toll-like receptor 4, and nuclear factor kappa B mRNA expressions induced by LPS. These findings demonstrate that CPP may ameliorate adipocyte dysfunction by suppressing oxidative damage and inflammatory responses through a new mechanism known as Toll-like receptor 4/nuclear factor kappa B-mediated signaling.


Subject(s)
3T3-L1 Cells , Adipocytes , Inflammation , Lipopolysaccharides , Oxidative Stress , Reactive Oxygen Species , Superoxide Dismutase , Toll-Like Receptor 4 , Zea mays , Animals , Mice , Adipocytes/drug effects , Adipocytes/metabolism , Zea mays/chemistry , Reactive Oxygen Species/metabolism , Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Powders , Peptides/pharmacology , Glutathione Peroxidase/metabolism , NF-kappa B/metabolism , Antioxidants/pharmacology , Glutathione Peroxidase GPX1 , Signal Transduction/drug effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Tumor Necrosis Factor-alpha/metabolism , Anti-Inflammatory Agents/pharmacology
9.
Nutrients ; 16(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38931313

ABSTRACT

Obesity is increasingly prevalent worldwide and is linked to metabolic diseases, such as insulin resistance (IR) and type 2 diabetes mellitus (T2DM), due to excessive free fatty acids (FFAs). Although lifestyle changes are effective, they often prove to be insufficient as initial treatments for obesity. Additionally, while surgical and pharmacological interventions are available, they are not entirely safe or effective. Recently, interest has grown in utilizing food waste and plant-derived phenolic compounds for their health benefits, presenting a promising avenue for managing obesity and its related disorders. Indeed, many studies have examined the potential inhibitory effects of the natural extract on adipocyte differentiation and lipid accumulation. This study focused on the evaluation of the effects of standardized extracts obtained from red oranges and olive leaf waste on 3T3-L1 murine pre-adipocyte and adipocyte functionality. Red orange extract (ROE) and olive leaf extract (OLE), alone and in combination, were tested to assess their anti-obesity and anti-inflammatory effects, as well as their potential therapeutic benefits. Three in vitro models were established to investigate the effects of the extracts on (I) adipocyte differentiation; (II) mature and hypertrophic adipocytes challenged with palmitic acid (PA) and erastin (ER), respectively; and (III) erastin-induced cytotoxicity on pre-adipocytes.


Subject(s)
3T3-L1 Cells , Adipocytes , Olea , Plant Extracts , Plant Leaves , Animals , Olea/chemistry , Adipocytes/drug effects , Plant Extracts/pharmacology , Mice , Plant Leaves/chemistry , Cell Differentiation/drug effects , Anti-Obesity Agents/pharmacology , Adipogenesis/drug effects , Obesity/drug therapy
10.
J Ethnopharmacol ; 333: 118423, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38878841

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Murraya koenigii commonly known as curry leaf, is traditionally used in India to manage various ailments including diabetes mellitus. Curry leaves are well documented in Indian Ayurvedic system of medicine for beneficial effects in skin eruptions, dysentery, emesis, poisonous bites and bruises. The anti-hyperglycemic and anti-hyperlipidemic effects of curry leaf extracts have been demonstrated through several in vitro and in vivo experiments previously. AIM OF THE STUDY: To prepare an alkaloid enriched fraction (AEF) from M. koenigii and its evaluation on i) in vitro adipogenesis process and ii) in vivo high fat diet-induced obesity in C57BL/6J mice. MATERIALS AND METHODS: MKME and AEF were prepared from M. koenigii leaves. The four carbazole alkaloids (bioactive markers) isolated from AEF were quantitatively determined in the leaves by RP-HPLC method. MKME and AEF were studied for anti-obesogenic activity in adipocytes in vitro and in HFD-induced C57BL/6J obese mice in vivo. At the termination of the in vivo study, lipid profile, hepatic and renal injury and glucose levels were analyzed in the blood samples. Animal tissues were examined histopathologically to determine any signs of damage. Repeated dose oral toxicity study for 28 days on Sprague-Dawley rats was also performed to determine the safety profile of AEF. RESULTS: Both MKME and AEF displayed anti-obesogenic activity at 25 µg/ml concentration in vitro and showed 54.06 ± 3.86% and 37.46 ± 3.17% lipid accumulation, respectively compared to control. Further, supplementation of AEF and MKME in HFD-fed C57BL/6J mice helped in controlling weight gain, improved dyslipidemia and glucose intolerance significantly. AEF showed better anti-obesity activity than MKME both in vitro and in vivo study. Repeated administration of AEF up to 1 g/kg dose for 28 days showed no pathological tissue damage. Both MKME and AEF were standardized using a simple and validated RP-HPLC method. CONCLUSION: Present study was aimed at preparation of a novel alkaloid-enriched fraction from methanolic extract of M. koenigii leaf and its evaluation for anti-diabesity effect. Our results demonstrated AEF to be a promising plant-based therapy for ameliorating obesity and related metabolic complications in HFD-fed C57BL/6J mice.


Subject(s)
3T3-L1 Cells , Alkaloids , Diet, High-Fat , Mice, Inbred C57BL , Murraya , Obesity , Plant Extracts , Plant Leaves , Animals , Murraya/chemistry , Alkaloids/pharmacology , Plant Leaves/chemistry , Obesity/drug therapy , Diet, High-Fat/adverse effects , Plant Extracts/pharmacology , Mice , Male , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/isolation & purification , Adipogenesis/drug effects , Adipocytes/drug effects
11.
Lipids Health Dis ; 23(1): 192, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909257

ABSTRACT

BACKGROUND: Overweight, often known as obesity, is the abnormal and excessive accumulation of fat that exposes the health of a person at risk by increasing the likelihood that they may experience many chronic conditions. Consequently, obesity has become a global health threat, presenting serious health issues, and attracting a lot of attention in the healthcare profession and the scientific community. METHOD: This study aims to explore the anti-adipogenic properties of 7-MEGA™ in an attempt to address obesity, using both in vitro and in vivo research. The effects of 7MEGA™ at three distinct concentrations were investigated in obese mice who were given a high-fat diet (HFD) and 3T3-L1 adipocytes. RESULTS: 7MEGA™ decreased the total fat mass, overall body weight, and the perirenal and subcutaneous white adipose tissue (PWAT and SWAT) contents in HFD mice. Additionally, 7MEGA™ showed promise in improving the metabolic health of individuals with obesity and regulate the levels of insulin hormone, pro-inflammatory cytokines and adipokines. Furthermore, Peroxisome proliferator-activated receptors (PPAR) α and γ, Uncoupling Protein 1 (UCP-1), Sterol Regulatory Element-Binding Protein 1 (SREBP-1), Fatty Acid-Binding Protein 4 (FABP4), Fatty Acid Synthase (FAS), Acetyl-CoA Carboxylase (ACC), Stearoyl-CoA Desaturase-1 (SCD-1) and CCAAT/Enhancer-Binding Protein (C/EBPα) were among the adipogenic regulators that 7MEGA™ could regulate. CONCLUSION: In summary, this study uncovered that 7MEGA™ demonstrates anti-adipogenic and anti-obesity effects, suggesting its potential in combating obesity.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Animals , Diet, High-Fat/adverse effects , Adipogenesis/drug effects , Obesity/metabolism , Mice , Adipocytes/drug effects , Adipocytes/metabolism , Male , PPAR gamma/metabolism , PPAR gamma/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Mice, Obese , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Adipokines/metabolism , Anti-Obesity Agents/pharmacology , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , CCAAT-Enhancer-Binding Proteins
12.
Molecules ; 29(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38893341

ABSTRACT

Perilla frutescens var. acuta (Lamiaceae) is widely used not only as an oil or a spice, but also as a traditional medicine to treat colds, coughs, fever, and indigestion. As an ongoing effort, luteolin-7-O-diglucuronide (1), apigenin-7-O-diglucuronide (2), and rosmarinic acid (3) isolated from P. frutescens var. acuta were investigated for their anti-adipogenic and thermogenic activities in 3T3-L1 cells. Compound 1 exhibited a strong inhibition against adipocyte differentiation by suppressing the expression of Pparg and Cebpa over 52.0% and 45.0%, respectively. Moreover, 2 inhibited the expression of those genes in a dose-dependent manner [Pparg: 41.7% (5 µM), 62.0% (10 µM), and 81.6% (50 µM); Cebpa: 13.8% (5 µM), 18.4% (10 µM), and 37.2% (50 µM)]. On the other hand, the P. frutescens var. acuta water extract showed moderate thermogenic activities. Compounds 1 and 3 also induced thermogenesis in a dose-dependent manner by stimulating the mRNA expressions of Ucp1, Pgc1a, and Prdm16. Moreover, an LC-MS/MS chromatogram of the extract was acquired using UHPLC-MS2 and it was analyzed by feature-based molecular networking (FBMN) and the Progenesis QI software (version 3.0). The chemical profiling of the extract demonstrated that flavonoids and their glycoside derivatives, including those isolated earlier as well as rosmarinic acid, are present in P. frutescens var. acuta.


Subject(s)
3T3-L1 Cells , Anti-Obesity Agents , Cinnamates , Depsides , Perilla frutescens , Plant Extracts , Rosmarinic Acid , Mice , Perilla frutescens/chemistry , Animals , Plant Extracts/pharmacology , Plant Extracts/chemistry , Depsides/pharmacology , Depsides/chemistry , Depsides/isolation & purification , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/isolation & purification , Cinnamates/pharmacology , Cinnamates/chemistry , Cinnamates/isolation & purification , Adipogenesis/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Cell Differentiation/drug effects , Obesity/drug therapy , Obesity/metabolism , Thermogenesis/drug effects
13.
Mol Cell Biol ; 44(7): 261-272, 2024.
Article in English | MEDLINE | ID: mdl-38828991

ABSTRACT

The protein tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) plays an important role in modulating glucose and lipid homeostasis. We previously suggested a potential role of SHP-1 in the regulation of peroxisome proliferator-activated receptor γ2 (PPARγ2) expression and activity but the mechanisms were unexplored. PPARγ2 is the master regulator of adipogenesis, but how its activity is regulated by tyrosine phosphorylation is largely unknown. Here, we found that SHP-1 binds to PPARγ2 primarily via its N-terminal SH2-domain. We confirmed the phosphorylation of PPARγ2 on tyrosine-residue 78 (Y78), which was reduced by SHP-1 in vitro resulting in decreased PPARγ2 stability. Loss of SHP-1 led to elevated, agonist-induced expression of the classical PPARγ2 targets FABP4 and CD36, concomitant with increased lipid content in cells expressing PPARγ2, an effect blunted by abrogation of PPARγ2 phosphorylation. Collectively, we discovered that SHP-1 affects the stability of PPARγ2 through dephosphorylation thereby influencing adipogenesis.


Subject(s)
Adipogenesis , PPAR gamma , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , PPAR gamma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Phosphorylation , Humans , Animals , Mice , CD36 Antigens/metabolism , CD36 Antigens/genetics , HEK293 Cells , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Protein Stability , 3T3-L1 Cells , src Homology Domains , Protein Binding
14.
STAR Protoc ; 5(2): 102977, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38875117

ABSTRACT

By differentiating into mature adipocytes, 3T3-L1 cells can be utilized as a model cell line to investigate (pre)adipocyte function in vitro. Here, we present a protocol for combining qualitative and quantitative analysis of lipid droplets in mature 3T3-L1 adipocytes using oil red O. We describe steps to differentiate 3T3-L1 preadipocytes to adipocytes and give detailed procedures to determine total lipid amount as well as lipid droplet size and number using microscopic devices and an ImageJ macro. For complete details on the use and execution of this protocol, please refer to Kaczmarek et al.1.


Subject(s)
3T3-L1 Cells , Adipocytes , Azo Compounds , Lipid Droplets , Animals , Mice , Adipocytes/metabolism , Adipocytes/cytology , Lipid Droplets/metabolism , Azo Compounds/chemistry , Cell Differentiation , Staining and Labeling/methods , Lipid Metabolism
15.
Eur J Pharmacol ; 977: 176705, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38830457

ABSTRACT

Obesity is a major health issue that contributes significantly to increased mortality and morbidity worldwide. Obesity is caused by uncontrolled adipogenesis and lipogenesis, leading to several metabolism-associated problems. Pancreatic lipase, an enzyme that breaks down dietary lipids, is a prominent target for obesity. Orlistat, a known inhibitor of pancreatic lipase, is commonly employed for the management of obesity. However, its side effects, such as diarrhoea, nausea and bladder pain, urge to look out for safer alternatives. Morin is a pentahydroxyflavone, exerts a broad spectrum of pharmacological effects including antioxidant, anti-inflammatory, lipid lowering, anti-diabetic, anti-fibrotic, anti-cancer, etc. This study investigated the effect of morin on pancreatic lipase activity, in vitro and in vivo adipogenesis. Molecular docking and simulation studies showed morin to have a higher binding affinity towards pancreatic lipase compared with orlistat, which also inhibited its activity in vitro. Morin also reduced lipid droplet accretion and downregulated the expression of adipogenic and lipogenic genes. The acute oral toxicity of morin was determined in C57BL/6 mice, where morin did not show toxicity up to 2000 mg/kg body weight dose. Oral administration of morin to high fat diet fed mice reduced body weight, glucose and insulin levels. Also, the histopathological examination revealed reduction in adipocyte size and decreased mRNA expression of adipogenesis markers in white adipose tissue of morin administered group compared to high fat diet group. Overall, the results suggested morin inhibited pancreatic lipase activity, adipogenesis and further studies are warranted to explore its therapeutic potential for obesity.


Subject(s)
Adipogenesis , Flavonoids , Lipase , Mice, Inbred C57BL , Molecular Docking Simulation , Animals , Adipogenesis/drug effects , Flavonoids/pharmacology , Mice , Lipase/antagonists & inhibitors , Lipase/metabolism , Male , 3T3-L1 Cells , Diet, High-Fat/adverse effects , Pancreas/drug effects , Pancreas/pathology , Anti-Obesity Agents/pharmacology , Obesity/drug therapy , Obesity/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Humans , Orlistat/pharmacology , Flavones
16.
Biochem Pharmacol ; 226: 116381, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909786

ABSTRACT

The escalating prevalence of obesity presents formidable challenges, necessitating the development of effective therapeutic strategies. In this study, we aimed to elucidate the preventive effects on obesity of tetrahydroberberrubine (THBru), a derivative of berberine (BBR) and to unravel its underlying mechanism. Using an obese mouse model induced by a high-fat diet (HFD), THBru was found to markedly ameliorate obesity, as evidenced by reduced body weight, decreased Lee's index, diminished fat mass in epididymal white adipose tissue (WAT) and brown adipose tissue (BAT), alongside improved dyslipidemia. Notably, at the same dose, THBru exhibited superior efficacy compared to BBR. RNA-sequencing and gene set enrichment analysis indicated THBru activated thermogenesis, which was further confirmed in WAT, BAT, and 3T3-L1 cells. Bioinformatics analysis of RNA-sequencing data revealed the candidate gene Pgc1α, a key regulator involved in thermogenesis. Moreover, THBru was demonstrated to elevate the expression of PGC1α by stabilizing its mRNA in WAT, BAT and 3T3-L1 cells. Furthermore, PGC1α knockdown blocked the pro-thermogenic and anti-obesity action of THBru both in vivo and in vitro. This study unravels the preventive effects of THBru on obesity through the activation of PGC1α-mediated thermogenesis, thereby delineating its potential therapeutic implications for obesity and associated disorders.


Subject(s)
3T3-L1 Cells , Adipose Tissue, Brown , Adipose Tissue, White , Berberine , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Thermogenesis , Animals , Thermogenesis/drug effects , Mice , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Male , Berberine/pharmacology , Berberine/analogs & derivatives , Berberine/therapeutic use , Obesity/prevention & control , Obesity/metabolism , Obesity/drug therapy , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Diet, High-Fat/adverse effects , Anti-Obesity Agents/pharmacology
17.
Bioorg Chem ; 150: 107536, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38878751

ABSTRACT

Carboxylesterase 1 (CES1), a member of the serine hydrolase superfamily, is involved in a wide range of xenobiotic and endogenous substances metabolic reactions in mammals. The inhibition of CES1 could not only alter the metabolism and disposition of related drugs, but also be benefit for treatment of metabolic disorders, such as obesity and fatty liver disease. In the present study, we aim to develop potential inhibitors of CES1 and reveal the preferred inhibitor structure from a series of synthetic pyrazolones (compounds 1-27). By in vitro high-throughput screening method, we found compounds 25 and 27 had non-competitive inhibition on CES1-mediated N-alkylated d-luciferin methyl ester (NLMe) hydrolysis, while compound 26 competitively inhibited CES1-mediated NLMe hydrolysis. Additionally, Compounds 25, 26 and 27 can inhibit CES1-mediated fluorescent probe hydrolysis in live HepG2 cells with effect. Besides, compounds 25, 26 and 27 could effectively inhibit the accumulation of lipid droplets in mouse adipocytes cells. These data not only provided study basis for the design of newly CES1 inhibitors. The present study not only provided the basis for the development of lead compounds for novel CES1 inhibitors with better performance, but also offered a new direction for the explore of candidate compounds for the treatment of hyperlipidemia and related diseases.


Subject(s)
Adipocytes , Carboxylic Ester Hydrolases , Enzyme Inhibitors , Pyrazolones , Humans , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/antagonists & inhibitors , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Animals , Mice , Pyrazolones/pharmacology , Pyrazolones/chemistry , Pyrazolones/chemical synthesis , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Molecular Structure , Hep G2 Cells , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , 3T3-L1 Cells
18.
Biomed Pharmacother ; 176: 116851, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838506

ABSTRACT

Glinus oppositifolius L., a member of the Molluginaceae family, has a long-standing history of utilization as both a vegetable and a medicinal agent across numerous countries. This plant possesses a diverse range of pharmacological activities and attracts scientific interest in studying its chemical profile. The present phytochemical investigation of the plant resulted in the isolation of eleven new triterpenoid saponins, accompanied by three known compounds. Their structures were elucidated by intensive spectroscopic analysis, DFT calculations, and comparison with previously reported data. The isolates were evaluated for their anti-adipogenic effect and cytotoxicity against human cancer cell lines, namely, colorectal carcinoma HCT116, hepatoblastoma cell HepG2, breast cancer cell MDA-MB-231, and human lung adenocarcinoma cell A549. Compounds 5, 7, and 13 exhibited a potent inhibitory effect against the differentiation of preadipocyte 3T3-L1. In addition, compound 13 displayed inhibitory effects against the growth of A549 cancer cells.


Subject(s)
3T3-L1 Cells , Plant Components, Aerial , Saponins , Triterpenes , Saponins/pharmacology , Saponins/isolation & purification , Saponins/chemistry , Humans , Triterpenes/pharmacology , Triterpenes/isolation & purification , Triterpenes/chemistry , Animals , Mice , Plant Components, Aerial/chemistry , Adipogenesis/drug effects , A549 Cells , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/chemistry , Hep G2 Cells , Cell Line, Tumor , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Cell Differentiation/drug effects , HCT116 Cells
20.
Molecules ; 29(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38893431

ABSTRACT

BACKGROUND: With the changes in lifestyle and diet structure, the incidence of obesity has increased year by year, and obesity is one of the inducements of many chronic metabolic diseases. Epigallocatechin gallate (EGCG), which is the most abundant component of tea polyphenols, has been used for many years to improve obesity and its complications. Though it has been reported that EGCG can improve obesity through many molecular mechanisms, EGCG may have many mechanisms yet to be explored. In this study, we explored other possible mechanisms through molecular docking and in vitro experiments. METHODS: AutoDock Vina was selected for conducting the molecular docking analysis to elucidate the interaction between EGCG and Notch1, while molecular dynamics simulations were employed to validate this interaction. Then, the new regulation mechanism of EGCG on obesity was verified with in vitro experiments, including a Western blot experiment, immunofluorescence experiment, oil red O staining, and other experiments in 3T3-L1 adipocytes. RESULTS: The molecular docking results showed that EGCG could bind to Notch1 protein through hydrogen bonding. In vitro cell experiments demonstrated that EGCG can significantly reduce the sizes of lipid droplets of 3T3-L1 adipocytes and promote UCP-1 expression by inhibiting the expression of Notch1 in 3T3-L1 adipocytes, thus promoting mitochondrial biogenesis. CONCLUSIONS: In this study, molecular docking and in vitro cell experiments were used to explore the possible mechanism of EGCG to improve obesity by inhibiting Notch1.


Subject(s)
Adipogenesis , Catechin , Molecular Docking Simulation , Receptor, Notch1 , Animals , Mice , 3T3-L1 Cells , Adipogenesis/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/chemistry , Gene Expression Regulation/drug effects , Molecular Dynamics Simulation , Obesity/drug therapy , Obesity/metabolism , Receptor, Notch1/metabolism , Uncoupling Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL