Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.897
Filter
1.
Sci Rep ; 14(1): 18252, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107469

ABSTRACT

Brown fat is a therapeutic target for the treatment of obesity-associated metabolic diseases. However, nutritional intervention strategies for increasing the mass and activity of human brown adipocytes have not yet been established. To identify vitamins required for brown adipogenesis and adipocyte browning, chemical compound-induced brown adipocytes (ciBAs) were converted from human dermal fibroblasts under serum-free and vitamin-free conditions. Choline was found to be essential for adipogenesis. Additional treatment with pantothenic acid (PA) provided choline-induced immature adipocytes with browning properties and metabolic maturation, including uncoupling protein 1 (UCP1) expression, lipolysis, and mitochondrial respiration. However, treatment with high PA concentrations attenuated these effects along with decreased glycolysis. Transcriptome analysis showed that a low PA concentration activated metabolic genes, including the futile creatine cycle-related thermogenic genes, which was reversed by a high PA concentration. Riboflavin treatment suppressed thermogenic gene expression and increased lipolysis, implying a metabolic pathway different from that of PA. Thiamine treatment slightly activated thermogenic genes along with decreased glycolysis. In summary, our results suggest that specific B vitamins and choline are uniquely involved in the regulation of adipocyte browning via cellular energy metabolism in a concentration-dependent manner.


Subject(s)
Adipocytes, Brown , Choline , Pantothenic Acid , Riboflavin , Thiamine , Humans , Riboflavin/pharmacology , Pantothenic Acid/pharmacology , Pantothenic Acid/metabolism , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Thiamine/pharmacology , Thiamine/metabolism , Choline/metabolism , Choline/pharmacology , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Lipolysis/drug effects , Energy Metabolism/drug effects , Thermogenesis/drug effects , Adipogenesis/drug effects , Glycolysis/drug effects , Cells, Cultured , Mitochondria/metabolism , Mitochondria/drug effects
2.
PLoS One ; 19(8): e0306903, 2024.
Article in English | MEDLINE | ID: mdl-39116155

ABSTRACT

Malva parviflora has shown anti-inflammatory, antihypertensive, antihyperlipidemic, and hypoglycemic effects. This study is aimed to evaluate the anti-adipogenic effect of M. parviflora on 3T3-L1 adipocytes. Fibroblast differentiation was induced either in the absence or presence of M. parviflora fractions (F3, F4, F7, F12, F13, F17, F18 and F19) for 4 days; F17 and 18 were the most effective fractions in reducing intracellular lipid accumulation (by 25.6% and 23.1%, respectively). EC50 of F17 and F18 (14 µg/mL and 17 µg/mL, respectively) were used to evaluate their anti adipogenic effect. After 10 days of inducing differentiation in the absence or presence of the extracts at the EC50 of F17 and F18, lipid accumulation, the concentration of interleukin 6 (IL-6) were measured in the culture medium; the presence of PPAR-γ, AKT, and p-AKT was also determined. In differentiated adipocytes (C2), F17 maintained intracellular lipid concentration at levels comparable to metformin, while decreasing PPAR-γ and increasing p-AKT presence; it also prevented IL-6 expression. F17 consists of alanine, valine, phenylalanine, and proline. On the other hand, F18 reduced intracellular lipid concentrations, prevented the increase of PPAR-γ and p-AKT, and maintained IL-6 expression at similar levels as metformin. F18 is mainly constituted by alanine, valine, proline, and sucrose. In conclusion, M. parviflora fractions (F17 and F18) control the process of adipogenesis, lipogenesis, and cellular dysfunction.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , PPAR gamma , Plant Extracts , Animals , Mice , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Adipogenesis/drug effects , Plant Extracts/pharmacology , PPAR gamma/metabolism , Interleukin-6/metabolism , Cell Differentiation/drug effects , Lipid Metabolism/drug effects , Proto-Oncogene Proteins c-akt/metabolism
3.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201308

ABSTRACT

Polybrominated diphenyl ethers (PBDEs), commonly used as synthetic flame retardants, are present in a variety of consumer products, including electronics, polyurethane foams, textiles, and building materials. Initial evidence from epidemiological and experimental studies suggests that maternal PBDE exposure may be associated with a higher BMI in children, with disturbance of energy metabolism and an increased risk of Type 2 diabetes. However, the causality between early exposure to real-life PBDE concentrations and increased weight as well as mechanisms underlying impaired metabolic pathways in the offspring remain elusive. Here, using a mouse model we examined the effect of maternal exposure to 2,2',4,4'-tetrabrominated diphenyl ether (BDE-47), the most abundant congener in human samples, on offspring weight gain and energy homeostasis using a mouse model. Maternal exposure to BDE-47 at low dose resulted in weight gain in female offspring together with an impaired glucose and insulin tolerance in both female and male mice. In vitro and in vivo data suggest increased adipogenesis induced by BDE-47, possibly mediated by DNA hypermethylation. Furthermore, mRNA data suggest that neuronal dysregulation of energy homeostasis, driven via a disturbed leptin signaling may contribute to the observed weight gain as well as impaired insulin and glucose tolerance.


Subject(s)
Halogenated Diphenyl Ethers , Insulin Resistance , Maternal Exposure , Prenatal Exposure Delayed Effects , Weight Gain , Animals , Halogenated Diphenyl Ethers/toxicity , Female , Mice , Maternal Exposure/adverse effects , Weight Gain/drug effects , Pregnancy , Male , Prenatal Exposure Delayed Effects/metabolism , DNA Methylation/drug effects , Adipogenesis/drug effects , Leptin/metabolism , Flame Retardants/toxicity , Flame Retardants/adverse effects , Energy Metabolism/drug effects
4.
Mar Drugs ; 22(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39195446

ABSTRACT

This study explores the anti-obesity effects of the ethyl acetate extract of Ecklonia cava (EC-ETAC) on 3T3-L1 preadipocytes, focusing on its impact on adipogenesis, lipolysis, and adipose browning via the HO-1/Nrf2 pathway. Western blot analysis revealed that EC-ETAC significantly inhibited adipogenic transcription factors (PPARγ, C/EBPα, SREBP-1) and lipogenesis-related proteins (FAS, LPL). Concurrently, EC-ETAC enhanced lipolytic markers (p-AMPK, p-HSL) and adipose browning-related proteins (UCP-1, PGC-1α), indicating its role in promoting lipolysis and adipose browning. The inhibition of HO-1 by zinc protoporphyrin (ZnPP) significantly reversed these effects, underscoring the critical role of HO-1 in mediating the anti-obesity properties of EC-ETAC. Additionally, fluorescence measurements and Oil Red O staining confirmed the reduction of lipid accumulation and oxidative stress upon EC-ETAC treatment. These findings suggest that EC-ETAC exerts its anti-obesity effects by modulating the HO-1/Nrf2 pathway, which is crucial for regulating adipogenesis, lipolysis, and adipose browning. This study highlights the potential of EC-ETAC as a natural therapeutic agent for obesity management and supports further research into its clinical applications. By targeting the HO-1/Nrf2 pathway, EC-ETAC could offer a novel approach to enhancing energy expenditure and reducing fat mass, thereby improving metabolic health.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Anti-Obesity Agents , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Phaeophyceae , Signal Transduction , Animals , Mice , Adipogenesis/drug effects , NF-E2-Related Factor 2/metabolism , Phaeophyceae/chemistry , Signal Transduction/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Heme Oxygenase-1/metabolism , Anti-Obesity Agents/pharmacology , Lipolysis/drug effects , Plant Extracts/pharmacology , Oxidative Stress/drug effects , Obesity/drug therapy , Obesity/metabolism , Membrane Proteins
5.
Stem Cell Res Ther ; 15(1): 252, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135105

ABSTRACT

BACKGROUND: Obesity is characterized by excessive fat accumulation, which is related with abnormal pluripotency of mesenchymal stem cells (MSCs). Recently, there is growing evidence that the disorder of maternal vitamin D (VD) intake is a well-known risk factor for long-term adverse health outcomes to their offspring. Otherwise, less is known of its repercussion and underlying mechanisms on the different differentiation potential of MSCs. METHODS: Four-week-old female C57BL/6J mice were fed with different VD reproductive diets throughout the whole pregnancy and lactation. The characteristics of BMSCs from their seven-day male offspring, VDR knockdown establishment of HuMSCs and HuMSCs under the different VD interventions in vitro were confirmed by flow cytometry, RT-PCR, and immunofluorescence. The roles of VD on their mitochondrial dysfunction and differentiation potential were also investigated. Then their remaining weaned male pups were induced by administrating high-fat-diet (HFD) for 16 weeks and normal fat diet was simultaneously as controls. Their lipid accumulation and adipocytes hypertrophy were determined by histological staining and related gene expressions. RESULTS: Herein, it was proved that imbalance of early-life VD intake could significantly aggravate the occurrence of obesity by inducing the adipogenesis through affecting the VD metabolism and related metabolites (P < 0.05). Moreover, abnormally maternal VD intake might be involved on the disorders of differentiation potential to inhibit the maintenance of MSCs stemness through increasing the productions of ROS, which was accompanied by impairing the expression of related genes on the adipo-osteogenic differentiation (P < 0.05). Moreover, it was along with increasing potential of adipogenic differentiation of MSCs as higher ROS in the state of VD deficiency, while excessive maternal VD status could conversely enhance the osteogenic differentiation with slightly lower ROS (P < 0.05). Furthermore, the underlying mechanisms might be involved on the mitochondria dysfunctional, especially the mitophagy, by activating the LC3b, P62 and etc. using in vivo and in vitro studies (P < 0.05). CONCLUSION: These findings demonstrated that imbalance of early-life VD intake could target ROS-mediated crosstalk between mitochondrial dysfunction and differentiation potential of MSCs, which was significantly associated with the later obesity. Obviously, our results could open up an attractive modality for the benefits of suitable VD intake during the pregnancy and lactation.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Mitochondria , Obesity , Reactive Oxygen Species , Vitamin D , Animals , Mesenchymal Stem Cells/metabolism , Mice , Obesity/metabolism , Obesity/pathology , Cell Differentiation/drug effects , Female , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Vitamin D/pharmacology , Vitamin D/metabolism , Male , Pregnancy , Diet, High-Fat/adverse effects , Adipogenesis/drug effects
6.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201266

ABSTRACT

Obesity is a complex health condition characterized by excessive adipose tissue accumulation, leading to significant metabolic disturbances such as insulin resistance and cardiovascular diseases. Fatty acid synthase (FAS), a key enzyme in lipogenesis, has been identified as a potential therapeutic target for obesity due to its role in adipocyte differentiation and lipid accumulation. This study employed a multidisciplinary approach involving in silico and in vitro analyses to investigate the anti-adipogenic properties of maclurin, a natural phenolic compound derived from Morus alba. Using SwissDock software (ChEMBL version 23), we predicted protein interactions and demonstrated a high probability (95.6%) of maclurin targeting FAS, surpassing the interaction rates of established inhibitors like cerulenin. Docking simulations revealed maclurin's superior binding affinity to FAS, with a binding score of -7.3 kcal/mol compared to -6.7 kcal/mol for cerulenin. Subsequent in vitro assays confirmed these findings, with maclurin effectively inhibiting FAS activity in a concentration-dependent manner in 3T3-L1 adipocytes, without compromising cell viability. Furthermore, maclurin treatment resulted in significant reductions in lipid accumulation and the downregulated expression of critical adipogenic genes such as PPARγ, C/EBPα, and FAS, indicating the suppression of adipocyte differentiation. Maclurin shows potential as a novel FAS inhibitor with significant anti-adipogenic effects, offering a promising therapeutic avenue for the treatment and prevention of obesity.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Molecular Docking Simulation , Mice , Animals , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cell Differentiation/drug effects , Adipogenesis/drug effects , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/antagonists & inhibitors , PPAR gamma/metabolism , PPAR gamma/genetics , Lipid Metabolism/drug effects , Cell Survival/drug effects , 4-Butyrolactone/analogs & derivatives
7.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201373

ABSTRACT

Low or insufficient testosterone levels caused by caponization promote fat deposition in animals. However, the molecular mechanism of fat deposition in caponized animals remains unclear. This study aimed to investigate the metabolomics and transcriptomic profiles of adipose tissues and study the effect of testosterone and leptin on the proliferation of adipocytes. We observed a significant enlargement in the areas of adipocytes in the abdominal fat tissues in capon, as well as increased luciferase activity of the serum leptin and a sharp decrease in the serum testosterone in caponized gander. Metabolomics and transcriptomic results revealed differentially expressed genes and differentially expressed metabolites with enhanced PARR signal pathway. The mRNA levels of peroxisome proliferators-activated receptor γ, fatty acid synthase, and suppressor of cytokine signaling 3 in goose primary pre-adipocytes were significantly upregulated with high leptin treatment and decreased significantly with increasing testosterone dose. Hence, reduced testosterone and increased leptin levels after caponization possibly promoted adipocytes proliferation and abdominal fat deposition by altering the expression of PPAR pathway related genes in caponized ganders. This study provides a new direction for the mechanism through which testosterone regulates the biological function of leptin and fat deposition in male animals.


Subject(s)
Adipogenesis , Leptin , Signal Transduction , Testosterone , Animals , Leptin/metabolism , Testosterone/pharmacology , Testosterone/metabolism , Adipogenesis/drug effects , Male , Signal Transduction/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Geese/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Cell Proliferation/drug effects , Adipose Tissue/metabolism , Adipose Tissue/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Orchiectomy
8.
Mol Med Rep ; 30(4)2024 10.
Article in English | MEDLINE | ID: mdl-39129299

ABSTRACT

Tanshinone IIA (Tan IIA) may have therapeutic effects on avascular necrosis of the femoral head (ANFH) by targeting bone marrow mesenchymal stem cells (BMSCs). The effect and underlying mechanism of Tan IIA on adipogenesis and osteogenesis ability of BMSCs remain to be elucidated. In the present study BMSCs were treated with osteogenic or adipogenic differentiation medium with or without Tan IIA under hypoxic environment. Osteogenic differentiation potential was evaluated by alkaline phosphatase (ALP) measurement, alizarin red staining and reverse transcription­quantitative (RT­q) PCR of osteogenic marker genes. Adipogenic differentiation potential was evaluated with oil red staining and RT­qPCR of adipogenic marker genes. Detailed mechanism was explored by RNA­seq and small molecular treatment during osteogenesis and adipogenesis of BMSCs. ALP level, mineralized nodules and expression level of osteogenic marker genes significantly increased following Tan IIA treatment during osteogenic differentiation of BMSCs. Lipid droplet and expression levels of adipogenic marker genes significantly decreased following Tan IIA treatment during adipogenic differentiation of BMSCs. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses of RNA­seq data indicated increased Akt and TGFß signaling following Tan IIA treatment. Further western blot assay confirmed that Tan IIA significantly activated Akt/cAMP response element­binding protein signaling and TGFß/Smad3 signaling. Application of Akti1/2 (an Akt inhibitor) significantly decreased the promotion effect of osteogenesis induced by Tan IIA, while the addition of SB431542 significantly reduced inhibition effect of adipogenesis caused by Tan IIA. Tan IIA could promote osteogenic differentiation potential of BMSCs by activating AKT signaling and suppress adipogenic differentiation potential of BMSCs by activating TGFß signaling.


Subject(s)
Abietanes , Adipogenesis , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Proto-Oncogene Proteins c-akt , Signal Transduction , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Abietanes/pharmacology , Adipogenesis/drug effects , Cell Differentiation/drug effects , Signal Transduction/drug effects , Animals , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Cells, Cultured , Smad3 Protein/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/cytology
9.
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
10.
Gene ; 931: 148873, 2024 Dec 30.
Article in English | MEDLINE | ID: mdl-39159793

ABSTRACT

Sarcopenia has become a prominent health problem among the elderly because of its adverse consequence, including physical disabilities and death. Fibro-adipogenic progenitors (FAPs) exhibit adipogenic and fibrogenic potencies and regulate skeletal muscle development, which plays important role in sarcopenia. Mairin, as an ingredient of Astragalus membranaceus, has the effect of anti-fibrosis. Therefore, we predicted that mairin targeted the fibrosis of FAPs and then affected sarcopenia. To verify our ideas, mairin (30 mg/kg/day or 60 mg/kg/day) was given to senescence accelerated mouse-prone 8 (SAMP8) mice by oral administration. Aging led to loss of weight, skeletal muscle mass, strength, and function, and an increase in muscle atrophy and fibrosis, while mairin administration inhibited physiological decline caused by aging. Similarly, mairin (20 µM or 40 µM) treatment enhanced FAP proliferation but blocked the differentiation into fibroblasts. Mechanically, mairin played an anti-fibrotic role via AMP-activated protein kinase-transforming growth factor beta-drosophila mothers against decapentaplegic protein (AMPK-TGF-ß-SMAD) axis, as evidenced by increased phosphorylation of AMPKα and decreased TGF-ß and phosphorylated-SMAD2/3. In addition, the potential target genes of mairin were explored by mRNA sequencing in our study. In conclusion, mairin may interfere with the AMPK/TGF-ß/SMAD pathway to repress the fibrosis of FAPs and eventually ameliorate sarcopenia.


Subject(s)
AMP-Activated Protein Kinases , Fibrosis , Sarcopenia , Signal Transduction , Transforming Growth Factor beta , Animals , Sarcopenia/metabolism , Sarcopenia/drug therapy , Sarcopenia/pathology , Mice , Transforming Growth Factor beta/metabolism , AMP-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Aging/metabolism , Adipogenesis/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Cell Differentiation/drug effects
11.
Environ Sci Pollut Res Int ; 31(39): 51593-51603, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39115733

ABSTRACT

Accumulating epidemiological evidence underscores the association between pervasive environmental factors and an increased risk of metabolic diseases. Environmental chemicals, recognized disruptors of endocrine and metabolic processes, may contribute to the global prevalence of metabolic disorders, including obesity. Acetyl tributyl citrate (ATHC), categorized as a citric acid ester plasticizer, serves as a substitute for di-(2-ethylhexyl) phthalate (DEHP) in various everyday products. Despite its widespread use and the increasing risk of exposure in humans and animals due to its high leakage rates, information regarding the safety of exposure to environmentally relevant doses of ATHC remains limited. This study aimed to investigate the potential impact of ATHC exposure on metabolic homeostasis. Both in vivo and in vitro exposure models were used to characterize the effects induced by ATHC exposure. C57BL/6 J male mice were subjected to a diet containing ATHC for 12 weeks, and metabolism-related parameters were monitored and analyzed throughout and after the exposure period. Results indicated that sub-chronic dietary exposure to ATHC induced an increase in body fat percentage, elevated serum lipid levels, and increased lipid content in the liver tissue of mice. Furthermore, the effect of ATHC exposure on murine hepatocytes were examined and results indicated that ATHC significantly augmented lipid levels in AML12 hepatocytes, disrupting energy homeostasis and altering the expression of genes associated with fatty acid synthesis, uptake, oxidation, and secretion pathways. Conclusively, both in vivo and in vitro results suggest that exposure to low levels of ATHC may be linked to an elevated risk of obesity and fatty liver in mice. The potential implications of ATHC on human health warrant comprehensive evaluation in future studies.


Subject(s)
Adipogenesis , Hepatocytes , Mice, Inbred C57BL , Plasticizers , Animals , Mice , Plasticizers/toxicity , Hepatocytes/drug effects , Male , Adipogenesis/drug effects , Citrates
12.
Medicine (Baltimore) ; 103(33): e39350, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151516

ABSTRACT

BACKGROUND: Previous researches have demonstrated that the traditional Chinese medicine could therapeutically treat inflammatory and hypoxic diseases by enhancing the functionality of mesenchymal stem cells. However, its mechanism was not yet clear. This research aimed to investigate the impact of the traditional Chinese medicine Sijunzi decoction and its herb monomer ginsenoside Rg1 on the proliferation and differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs) and explore the underlying mechanisms. METHODS: Different concentrations of Sijunzi decoction and Rg1 were applied to differentiating induced hUC-MSCs. The CCK-8 test was utilized to evaluate cell proliferation activity and identify suitable drug concentrations. Alizarin Red staining was employed to detect the formation of calcium nodules, and Oil Red O staining was used to assess the formation of lipid droplets. PCR was utilized to examine gene expression related to osteogenic differentiation, adipogenic differentiation, and the HIF-1α signaling pathway in hUC-MSCs. Western blot analysis was conducted to evaluate protein expression in osteogenic differentiation and HIF-1α. ELISA was performed to measure HIF-1α signaling factors and inflammatory cytokine expression. Biochemical assays were used to assess changes in oxidative stress indicators. RESULTS: The Sijunzi decoction and Rg1 both demonstrated a dose-dependent promotion of hUC-MSC proliferation. The Sijunzi decoction significantly increased the expression of genes and proteins relevant to osteogenesis, such as osterix, osteocalcin, RUNX2, and osteopontin, and activated the HIF-1α pathway in hUC-MSCs. (P < .05). Similar effects were observed at the gene level after treatment with Rg1. Simultaneously, Sijunzi decoction significantly reduced the secretion of pro-inflammatory cytokines TNF-α, IL-6, and IL-1ß, while increasing the secretion of the anti-inflammatory cytokine IL-10 during osteogenic differentiation (P < .05). Moreover, Sijunzi decoction lowered oxidative stress levels and enhanced the antioxidant capacity of hUC-MSCs during osteogenic differentiation (P < .05). However, the impact of Sijunzi decoction on hUC-MSCs toward adipogenic differentiation was not significant (P > .05). CONCLUSION: Sijunzi decoction promotes the proliferation and osteogenic differentiation of hUC-MSCs, potentially through the activation of the HIF-1α signaling pathway and by modulating the microenvironment via reducing inflammation and oxidative stress levels. Rg1 might be involved in this process.


Subject(s)
Cell Differentiation , Cell Proliferation , Drugs, Chinese Herbal , Ginsenosides , Hypoxia-Inducible Factor 1, alpha Subunit , Mesenchymal Stem Cells , Osteogenesis , Umbilical Cord , Humans , Mesenchymal Stem Cells/drug effects , Drugs, Chinese Herbal/pharmacology , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Umbilical Cord/cytology , Osteogenesis/drug effects , Ginsenosides/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Adipogenesis/drug effects , Signal Transduction/drug effects , Cells, Cultured
13.
Front Endocrinol (Lausanne) ; 15: 1401120, 2024.
Article in English | MEDLINE | ID: mdl-39040675

ABSTRACT

Whilst western diet and sedentary lifestyles heavily contribute to the global obesity epidemic, it is likely that chemical exposure may also contribute. A substantial body of literature implicates a variety of suspected environmental chemicals in metabolic disruption and obesogenic mechanisms. Chemically induced obesogenic metabolic disruption is not yet considered in regulatory testing paradigms or regulations, but this is an internationally recognised human health regulatory development need. An early step in the development of relevant regulatory test methods is to derive appropriate minimum chemical selection lists for the target endpoint and its key mechanisms, such that the test method can be suitably optimised and validated. Independently collated and reviewed reference and proficiency chemicals relevant for the regulatory chemical universe that they are intended to serve, assist regulatory test method development and validation, particularly in relation to the OECD Test Guidelines Programme. To address obesogenic mechanisms and modes of action for chemical hazard assessment, key initiating mechanisms include molecular-level Peroxisome Proliferator-Activated Receptor (PPAR) α and γ agonism and the tissue/organ-level key event of perturbation of the adipogenesis process that may lead to excess white adipose tissue. Here we present a critical literature review, analysis and evaluation of chemicals suitable for the development, optimisation and validation of human PPARα and PPARγ agonism and human white adipose tissue adipogenesis test methods. The chemical lists have been derived with consideration of essential criteria needed for understanding the strengths and limitations of the test methods. With a weight of evidence approach, this has been combined with practical and applied aspects required for the integration and combination of relevant candidate test methods into test batteries, as part of an Integrated Approach to Testing and Assessment for metabolic disruption. The proposed proficiency and reference chemical list includes a long list of negatives and positives (20 chemicals for PPARα, 21 for PPARγ, and 11 for adipogenesis) from which a (pre-)validation proficiency chemicals list has been derived.


Subject(s)
Adipogenesis , Obesity , PPAR alpha , PPAR gamma , Humans , PPAR alpha/metabolism , PPAR alpha/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Adipogenesis/drug effects , Obesity/metabolism , Obesity/chemically induced , Transcriptional Activation/drug effects
14.
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
15.
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
16.
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
17.
Eur J Pharmacol ; 978: 176793, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38960061

ABSTRACT

In recent years, a common-used antidiabetic drug, liraglutide, was identified with extra effects on lipid metabolism. Its effects against excessive lipid deposition in bone marrow were gained much attention but not well established. Our aim in the present study is to explore the interaction of miRNAs-mRNAs altered by liraglutide administration during bone marrow adipogenesis in diabetes. To establish the diabetic animal model, rats were treated with high fat diet (HFD) and STZ injection. We then identified the lowering effect of liraglutide on lipids metabolism in the diabetes. During this process, high-throughput sequencing and bioinformatics analyses on miRNAs extracted from bone marrow mesenchymal stem cells (BMSCs) were conducted after liraglutide administration. We then identified five differentially expressed miRNAs (miRNA-150-5p, miRNA-129-5p, miRNA-201-3p, miRNA-201-5p, and miRNA-214-5p). The expressions of the DE miRNAs were verified as temporal specific expression patterns in Day 3 and in Day 7. Among them, miRNA-150-5p expression was more stable and consistent with the sequencing data. Of interest, miR-150-5p overexpression facilitated adipogenesis of BMSCs. But this promotion was alleviated by liraglutide. The predicted target gene of miR-150-5p, GDF11, was validated to be involved in liraglutide alleviated BMSCs' lipid accumulation in diabetes. In vitro, liraglutide increased the GDF11 expression, rescued its down-expression by siGDF11 and inhibit the adipogenesis of BMSCs cultured in high glucose medium. In vivo, liraglutide reversed the HFD-STZ induced excessive lipid droplets by up-regulation of GDF11 expression, which was discounted by agomiR-150-5p injection. Above all, liraglutide might alleviate bone marrow fat accumulation via inactivating miR-150-5p/GDF11 axis in diabetes.


Subject(s)
Adipogenesis , Diabetes Mellitus, Experimental , Liraglutide , Mesenchymal Stem Cells , MicroRNAs , Rats, Sprague-Dawley , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Rats , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism
18.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000199

ABSTRACT

Adiponectin is a circulating hormone secreted by adipose tissue that exerts, unlike other adipokines such as leptin, anti-inflammatory, anti-atherosclerotic and other protective effects on health. Adiponectin receptor agonists are being tested in clinical trials and are expected to show benefits in many diseases. In a recent article, LW Chen's group used monocyte chemoattractant protein-1 (MCP-1/CCL2) to improve plasma levels of adiponectin, suggesting the involvement of dipeptidyl peptidase 4 (DPP4/CD26) in the mechanism. Here, we discuss the significance of the role of DPP4, favoring the increase in DPP4-positive interstitial progenitor cells, a finding that fits with the greater stemness and persistence of other DPP4/CD26-positive cells.


Subject(s)
Adipogenesis , Adipose Tissue , Dipeptidyl Peptidase 4 , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl Peptidase 4/genetics , Adipogenesis/genetics , Adipogenesis/drug effects , Humans , Adipose Tissue/metabolism , Animals , Adiponectin/metabolism , Adiponectin/genetics , Gene Expression Regulation/drug effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Stromal Cells/metabolism , Adipocytes/metabolism , Adipocytes/drug effects
19.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062809

ABSTRACT

The study evaluated the effects of Arthrospira maxima phycobiliproteins (PBPs), rosiglitazone (RSG), and 17ß-estradiol (E) on the differentiation process of 3T3-L1 cells and on their regulation of lipogenic and inflammatory gene expression at different stages of the process. The results showed that phycobiliproteins promoted cell proliferation after 24 h of treatment. Furthermore, for all three treatments, the regulation of the highest number of markers occurred on days 6 and 12 of differentiation, regardless of when the treatment was applied. Phycobiliproteins reduced lipid droplet accumulation on days 3, 6, 10, and 13 of the adipogenic process, while rosiglitazone showed no differences compared to the control. On day 6, both phycobiliproteins and rosiglitazone positively regulated Acc1 mRNA. Meanwhile, all three treatments negatively regulated Pparγ and C/ebpα. Phycobiliproteins and estradiol also negatively regulated Ucp1 and Glut4 mRNAs. Rosiglitazone and estradiol, on the other hand, negatively regulated Ppara and Il-6 mRNAs. By day 12, phycobiliproteins and rosiglitazone upregulated Pparγ mRNA and negatively regulated Tnfα and Il-1ß. Additionally, phycobiliproteins and estradiol positively regulated Il-6 and negatively regulated Ppara, Ucp2, Acc1, and Glut4. Rosiglitazone and estradiol upregulate C/ebpα and Ucp1 mRNAs. The regulation exerted by phycobiliproteins on the mRNA expression of the studied markers was dependent on the phase of cell differentiation. The results of this study highlight that phycobiliproteins have an anti-adipogenic and anti-inflammatory effect by reducing the expression of adipogenic, lipogenic, and inflammatory genes in 3T3-L1 cells at different stages of the differentiation process.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Estradiol , Phycobiliproteins , Rosiglitazone , Animals , Mice , Estradiol/pharmacology , Rosiglitazone/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cell Differentiation/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Phycobiliproteins/pharmacology , Phycobiliproteins/metabolism , Phycobiliproteins/genetics , Gene Expression Regulation/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Proliferation/drug effects , Inflammation/metabolism , Inflammation/genetics , Spirulina
20.
Phytomedicine ; 132: 155854, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032276

ABSTRACT

BACKGROUND: Recent investigations underscore the capacity of photodynamic therapy (PDT) to induce adipocyte apoptosis, thereby mitigating obesity. Nonetheless, extant synthetic photosensitizers manifest limitations that hinder their clinical viability. PURPOSE: In the current study, we used Hypericum perforatum-derived exosomes-like nanovesicles (HPExos) as a novel photosensitizer, and investigated its PDT effects in adipose tissue during obesity. METHOD: HPExos-were administered to high fat diet mice via intraperitoneal injection, followed by targeted irradiation with specialized LED lights. Mass spectrometric analysis was analyzed in adipose tissues. CCK8 assay and Oil Red O staining were used to investigate lipid accumulation in 3T3-L1 cells to clarify adipocyte differentiation. The expression levels of related markers associated with adipogenesis and lipogenesis were assessed by RT-PCR. Apoptosis analysis was performed by TUNEL staining of and western blotting. RESULTS: HPExos combined with PDT accumulated in visceral white adipose tissues results in a reduced body weight and improved insulin sensitivity. HPExos combined with PDT induced apoptosis by driving high levels of ROS. In addition, HPExos combined with PDT significantly downregulated the expression of transcription factors, PPARγ, C/EBPα, and SREBP and lipogenesis protein FABP4 both in vitro and in vivo, associated with a decreased FFA levels. CONCLUSION: These findings suggest that HPExos could act as an effective photosensitizer in regulating glucose hemostasis by inhibiting adipocyte differentiation and lipogenesis, offering a promising approach for obesity treatment.


Subject(s)
3T3-L1 Cells , Apoptosis , Exosomes , Hypericum , Obesity , Photochemotherapy , Hypericum/chemistry , Animals , Mice , Exosomes/metabolism , Photochemotherapy/methods , Male , Apoptosis/drug effects , Obesity/drug therapy , Diet, High-Fat , Mice, Inbred C57BL , Adipose Tissue/drug effects , Photosensitizing Agents/pharmacology , PPAR gamma/metabolism , Adipocytes/drug effects , Adipogenesis/drug effects , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Lipogenesis/drug effects , Reactive Oxygen Species/metabolism , Plant Extracts/pharmacology , Insulin Resistance , CCAAT-Enhancer-Binding Proteins , Fatty Acid-Binding Proteins , Sterol Regulatory Element Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL