Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Phytomedicine ; 126: 155447, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38394732

ABSTRACT

BACKGROUD: High comorbidity rates have been reported in patients with atherosclerosis and osteoporosis, posing a serious risk to the health and well-being of elderly patients. To improve and update clinical practice regarding the joint treatment of these two diseases, the common mechanisms of atherosclerosis and osteoporosis need to be clarified. MicroRNAs (miRNAs), are importance molecules in the pathogenesis of human diseases, including in cardiovascular and orthopedic fields. They have garnered interest as potential targets for novel therapeutic strategies. However, the key miRNAs involved in atherosclerosis and osteoporosis and their precise regulation mechanisms remain unknown. Paeonol (Pae), an active ingredient in Cortex Moutan, has shown promising results in improving both lipid and bone metabolic abnormalities. However, it is uncertain whether this agent can exert a cotherapeutic effect on atherosclerosis and osteoporosis. OBJECTIVE: This study aimed to screen important shared miRNAs in atherosclerotic and osteoporotic complications, and explore the mechanism of the protective effects of Pae against atherosclerosis and osteoporosis in high-fat diet (HFD)-fed ApoE-/- mice. METHODS: An experimental atherosclerosis and osteoporosis model was established in 40-week-old HFD ApoE-/- mice. Various techniques such as Oil Red O staining, HE staining and micro-CT were used to confirm the co-occurrence of these two diseases and efficacy of Pae in addition to the associated biochemical changes. Bioinformatics was used to screen key miRNAs in the atherosclerosis and osteoporosis model, and gene involvement was assessed through serum analyses, qRT-PCR, and western blot. To investigate the effect of Pae on the modulation of the miR let-7g/HMGA2/CEBPß pathway, Raw 264.7 cells were cocultured with bone marrow mesenchymal stem cells (BMSCs) and treated with an miR let-7g mimic/inhibitor. RESULTS: miR let-7g identified using bioinformatics was assessed to evaluate its participation in atherosclerosis-osteoporosis. Experimental analysis showed reduced miR let-7g levels in the atherosclerosis-osteoporosis mice model. Moreover, miR let-7g was required for BMSC - Raw 264.7 cell crosstalk, thereby promoting foam cell formation and adipocyte differentiation. Treatment with Pae significantly reduced plaque accumulation and foam cell number in the aorta while increasing bone density and improving trabecular bone microarchitecture in HFD ApoE-/- mice. Pae also increased the level of miR let-7g in the bloodstream of model mice. In vitro studies, Pae enhanced miR let-7g expression in BMSCs, thereby suppressing the HMGA2/CEBPß pathway to prevent the formation of foam cells and differentiation of adipocytes induced by oxidized low-density lipoprotein (ox-LDL). CONCLUSION: The study results suggested that miR let-7g participates in atherosclerosis -osteoporosis regulation and that Pae acts as a potential therapeutic agent for preventing atherosclerosis-osteoporosis through regulatory effects on the miR let-7g/HMGA2/CEBPß pathway to hinder foam cell formation and adipocyte differentiation.


Subject(s)
Acetophenones , Adipogenesis , Atherosclerosis , Foam Cells , MicroRNAs , Osteoporosis , Animals , Mice , Acetophenones/pharmacology , Acetophenones/therapeutic use , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Differentiation , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Adipogenesis/genetics
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(3): 159424, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37956708

ABSTRACT

LGALS12, also known as galectin12, belongs to the galectin family with ß-galactoside-binding activity. We previously reported that LGALS12 is an important regulator of adipogenesis in porcine adipocytes in vitro, but its value in pig breeding needed to be explored in vivo. In this study, we used CRISPR/Cas9 to construct porcine fetal fibroblasts (PFFs) with a 43 bp deletion in LGALS12 exon 2. Using these PFFs as donor cells, a LGALS12 knockout pig model was generated via somatic cell nuclear transfer. Primary cultures of porcine intramuscular (IM) and subcutaneous (SC) adipocytes were established using cells from LGALS12 knockout pigs and wild-type pigs. A comparison of these cells proved that LGALS12 deficiency suppresses cell proliferation via the RAS-p38MAPK pathway and promotes lipolysis via the PKA pathway in both IM and SC adipocytes. In addition, we observed AKT activation only in IM adipocytes and suppression of the Wnt/ß-catenin only in SC adipocytes. Our findings suggest that LGALS12 deficiency affects the adipogenesis of IM and SC adipocytes through different mechanisms.


Subject(s)
Adipocytes , CRISPR-Cas Systems , Swine , Animals , Gene Knockout Techniques , Adipocytes/metabolism , Adipogenesis/genetics , Cell Proliferation
3.
BMC Res Notes ; 16(1): 338, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37974253

ABSTRACT

OBJECTIVE: Placental extract, which contains various bioactive compounds, has been used as traditional medicine. Many studies have demonstrated additional applications of placental extract and provided a scientific basis for the broad spectrum of its effects. We have previously reported that porcine placental extract (PPE) strongly suppresses adipogenesis in a 3T3-L1 preadipocyte cell line, inhibiting differentiation. This study aimed to examine the effect of PPE on the accumulation of lipid droplets (LD) in adipose-derived mesenchymal stromal/stem cells (ASC). RESULTS: The study findings revealed that PPE decreased the size of LD during the differentiation of ASC into mature adipocytes. RT-qPCR analysis revealed that PPE increased the gene expression of lysosomal acid lipase A (Lipa), a lipolysis-related gene, in ASC-differentiated adipocytes. However, no differences were noted in the adipocyte differentiation markers (Pparg, Cebpa, and Adipoq), or the adipogenesis-related genes (Dgat1, Dgat2, Fasn, Soat1, and Soat2). In addition, PPE promoted autophagosome formation, which was partially co-localized with the LD, indicating that PPE accelerated the degradation of LD by inducing autophagy (termed lipophagy) during the differentiation of ASC into mature adipocytes. These results suggest that the use of PPE may be a potential novel treatment for regulating adipogenesis for the treatment of obesity.


Subject(s)
Placental Extracts , Pregnancy , Female , Animals , Swine , Placental Extracts/metabolism , Placental Extracts/pharmacology , Lipid Droplets/metabolism , Placenta/metabolism , Cell Differentiation , Adipocytes/metabolism , Adipogenesis/genetics , Lipolysis , Autophagy , Stem Cells
4.
Sci Rep ; 13(1): 19251, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37935755

ABSTRACT

Recent studies have shown that the health benefits of probiotics are not limited to those offered by living bacteria. It was reported that both live and killed cells of Propionibacterium freudenreichii MJ2 (MJ2) isolated from raw milk showed antiobesity activity in 3T3-L1 cells and high-fat diet-induced obese mice. This study was aimed at identifying the active component(s) responsible for the antiadipogenic activity of MJ2. Cell wall, surface protein, and cytoplasmic fractions of MJ2 were investigated for their inhibitory effects on adipogenesis in 3T3-L1 cells. Adipocytes treated with the surface protein fraction showed significantly lower lipid accumulation. Using the MASCOT algorithm following LC-MS/MS analysis, 131 surface proteins were identified and they were principally classified into three categories (network clusters related to ribosomes, carbon metabolism, and chaperones). Among them, chaperonin 60 (Cpn60) was selected as a potential candidate protein. Cpn60 inhibited lipid accumulation and adipogenesis during the early period of differentiation (days 0-2) and decreased expression of genes related to adipogenesis (Pparg and Cebpa) and lipogenesis (Fas and Scd1). The expression of Gata2/3, which suppresses adipogenesis, significantly increased in Cpn60-treated cells. Moreover, the nuclear translocation of C/EBPß was inhibited by Cpn60 treatment. In conclusion, Cpn60, a surface protein in MJ2, shows antiadipogenic activity by reducing the expression of C/EBPß through the upregulation of Gata2/3 expression followed by downregulation of Pparg and Cebpa expression.


Subject(s)
Adipogenesis , Propionibacterium freudenreichii , Mice , Animals , Adipogenesis/genetics , PPAR gamma/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Chaperonin 60/pharmacology , Obesity/metabolism , Chromatography, Liquid , Plant Extracts/pharmacology , Tandem Mass Spectrometry , Cell Differentiation , CCAAT-Enhancer-Binding Protein-beta , Triglycerides/pharmacology , Membrane Proteins/pharmacology , 3T3-L1 Cells
5.
Mitochondrion ; 72: 22-32, 2023 09.
Article in English | MEDLINE | ID: mdl-37451354

ABSTRACT

Mitochondrial functions play a crucial role in determining the metabolic and thermogenic status of brown adipocytes. Increasing evidence reveals that the mitochondrial oxidative phosphorylation (OXPHOS) system plays an important role in brown adipogenesis, but the mechanistic insights are limited. Herein, we explored the potential metabolic mechanisms leading to OXPHOS regulation of brown adipogenesis in pharmacological and genetic models of mitochondrial respiratory complex I deficiency. OXPHOS deficiency inhibits brown adipogenesis through disruption of the brown adipogenic transcription circuit without affecting ATP levels. Neither blockage of calcium signaling nor antioxidant treatment can rescue the suppressed brown adipogenesis. Metabolomics analysis revealed a decrease in levels of tricarboxylic acid cycle intermediates and heme. Heme supplementation specifically enhances respiratory complex I activity without affecting complex II and partially reverses the inhibited brown adipogenesis by OXPHOS deficiency. Moreover, the regulation of brown adipogenesis by the OXPHOS-heme axis may be due to the suppressed histone methylation status by increasing histone demethylation. In summary, our findings identified a heme-sensing retrograde signaling pathway that connects mitochondrial OXPHOS to the regulation of brown adipocyte differentiation and metabolic functions.


Subject(s)
Adipogenesis , Histones , Adipogenesis/genetics , Histones/metabolism , Electron Transport Complex I/metabolism , Demethylation , Cell Differentiation
6.
Int J Mol Sci ; 23(19)2022 Oct 09.
Article in English | MEDLINE | ID: mdl-36233307

ABSTRACT

Salix pseudolasiogyne (Salicaceae) is a willow tree and has been used as a medicinal herb in Korea to treat pain and fever. As a part of an ongoing study to identify bioactive natural products, potential anti-adipogenic compounds were investigated using the ethanol (EtOH) extract of S. pseudolasiogyne twigs. Phytochemical investigation of the EtOH extracts using liquid chromatography-mass spectrometry (LC/MS) led to the separation of two compounds, oregonin (1) and 2'-O-acetylsalicortin (2). The structures of the isolates were identified using nuclear magnetic resonance spectroscopy and LC/MS analysis. To the best of our knowledge, it is the first report identifying oregonin (1) in twigs of S. pseudolasiogyne. Here, we found that the isolated compounds, oregonin (1) and 2'-O-acetylsalicortin (2), showed anti-adipogenic effects during 3T3-L1 cell differentiation. Notably, 2'-O-acetylsalicortin (2), at a concentration of 50 µM, significantly suppressed lipid accumulation. Moreover, the mRNA and protein levels of lipogenic and adipogenic transcription factors were reduced in 2'-O-acetylsalicortin (2)-treated 3T3-L1 cells. Taken together, these results indicate that 2'-O-acetylsalicortin (2), isolated from S. pseudolasiogyne twigs, has the potential to be applied as a therapeutic agent to effectively control adipocyte differentiation, a critical stage in the progression of obesity.


Subject(s)
Salix , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Cell Differentiation , Diarylheptanoids , Ethanol/pharmacology , Lipids/pharmacology , Mice , PPAR gamma/metabolism , Phytochemicals/metabolism , Phytochemicals/pharmacology , Plant Extracts/metabolism , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Salix/genetics , Transcription Factors/metabolism
7.
Gene ; 838: 146705, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-35772657

ABSTRACT

Although the precise pathogenesis of steroid-induced osteonecrosis of femoral head (SONFH) is not yet fully understood, evidence shows miRNAs-mediated posttranscription control directs the adipogenesis of bone marrow mesenchymal stem cells (BMSCs) and plays a pivotal role in the SONFH processes. Huogu injection formulated according to traditional Chinese medicine (TCM) theory has been used to treat SONFH by intra-articular injection. In this study, we asked whether the therapeutic effects of Huogu injection might depend on the inhibition of adipogenic differentiation of BMSCs, and if so, the pathway might be a therapeutic target to promote bone repair. Consequently, miR-34c-5p was upregulated in the dexamethasone (DEX)-treated BMSCs and might participate in the adipogenesis of BMSCs. TargetScan database and the luciferase reporter assay showed miR-34c-5p targeted on the MDM4 and negatively regulated its expression. Huogu injection in vitro inhibited the adipogenesis in the DEX-treated BMSCs by inhibiting the expression levels of PPARγ and C/EBPα, as well as reducing miR-34c-5p to prevent the degradation of MDM4. Moreover, miR-34c-5p mimic or MDM4 knockdown using shRNA neutralized the anti-adipogenesis of Huogu injection in BMSCs. In vivo, the results of X-ray imaging confirmed that Huogu injection alleviated the bone loss in rat SONFH. Consistent with results in vitro, Huogu injection reduced the lipid accumulation, removed the suppression of MDM4 by downregulating the expression of miR-34c-5p, and inhibited the expression of C/EBPα and PPARγ in bone tissues. When the lentivirus encoding miR-34c-5p was conducted by intra-articular injection, the overexpression of miR-34c-5p antagonized the therapeutic effects of Huogu injection. Our results underline the critical importance of the miR-34c-5p/MDM4 pathway in regulating the adipogenic outcome of BMSCs, suggesting the miR-34c-5p as a potentially effective therapeutic target in SONFH. These results further reinforce the potential of Huogu injection as an alternative approach in SONFH.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Animals , Rats , Adipogenesis/genetics , Cell Differentiation , Cells, Cultured , Femur Head/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteogenesis , PPAR gamma/metabolism
8.
PeerJ ; 10: e13417, 2022.
Article in English | MEDLINE | ID: mdl-35529487

ABSTRACT

After adipogenic differentiation, key regulators of adipogenesis are stimulated and cells begin to accumulate lipids. To identify specific changes in lipid composition and gene expression patterns during 3T3-L1 cell adipogenesis, we carried out lipidomics and RNA sequencing analysis of undifferentiated and differentiated 3T3-L1 cells. The analysis revealed significant changes in lipid content and gene expression patterns during adipogenesis. Slc2a4 was up-regulated, which may enhance glucose transport; Gpat3, Agpat2, Lipin1 and Dgat were also up-regulated, potentially to enrich intracellular triacylglycerol (TG). Increased expression levels of Pnpla2, Lipe, Acsl1 and Lpl likely increase intracellular free fatty acids, which can then be used for subsequent synthesis of other lipids, such as sphingomyelin (SM) and ceramide (Cer). Enriched intracellular diacylglycerol (DG) can also provide more raw materials for the synthesis of phosphatidylinositol (PI), phosphatidylcholine (PC), phosphatidylethanolamine (PE), ether-PE, and ether-PC, whereas high expression of Pla3 may enhance the formation of lysophophatidylcholine (LPC) and lysophosphatidylethanolamine (LPE). Therefore, in the process of adipogenesis of 3T3-L1 cells, a series of genes are activated, resulting in large changes in the contents of various lipid metabolites in the cells, especially TG, DG, SM, Cer, PI, PC, PE, etherPE, etherPC, LPC and LPE. These findings provide a theoretical basis for our understanding the pathophysiology of obesity.


Subject(s)
Adipogenesis , Lipidomics , Mice , Animals , Adipogenesis/genetics , 3T3-L1 Cells , Lecithins , Ceramides , Sequence Analysis, RNA , 1-Acylglycerol-3-Phosphate O-Acyltransferase
9.
Article in English | MEDLINE | ID: mdl-35248800

ABSTRACT

Inhibition of adipogenesis is an important strategy for obesity treatment. Rocaglamide-A (Roc-A) is a natural herbal medicine isolated from the genus Aglaia (family Meliaceae), which has a cyclopenta[b]benzofuran core structure. Roc-A exhibits various pharmacological effects against diverse human cancer cells. However, the exact role of Roc-A during adipogenesis in adipocytes has not been studied at all. In this study, we demonstrate that Roc-A is crucial for reducing adipogenesis via downregulating PPARγ transcriptional activity. Consistently, Western-blot and RT-PCR analyses clearly showed that Roc-A inhibits the expression of PPARγ target genes and lipogenic markers in a dose-dependent manner along with suppression of lipid accumulation, in both 3T3-L1 cells and mouse adipose-derived stem cells. Mechanistically, Roc-A significantly decreased STAT3 phosphorylation in a dose-dependent manner in 3T3-L1 adipocytes. In particular, we confirmed that Roc-A effectively suppressed the expression of genes involved in cell-cycle regulation, such as cyclin A, B, D1, and E1, early during mitotic clonal expansion in 3T3-L1 adipocytes, and this effect was abolished by the JAK2/STAT3 activator FGF2. Taken together, our results demonstrated that Roc-A reduces adipogenesis by inhibiting PPARγ transactivation and STAT3 phosphorylation and thus may serve as a therapeutic target in obesity.


Subject(s)
Adipogenesis , Benzofurans , Adipogenesis/genetics , Animals , Benzofurans/pharmacology , Mice , Obesity , PPAR gamma/genetics
10.
Autophagy ; 18(3): 518-539, 2022 03.
Article in English | MEDLINE | ID: mdl-34101546

ABSTRACT

The overexpansion of adipose tissues leads to obesity and eventually results in metabolic disorders. Garcinia cambogia (G. cambogia) has been used as an antiobesity supplement. However, the molecular mechanisms underlying the effects of G. cambogia on cellular processes have yet to be fully understood. Here, we discovered that G. cambogia attenuated the expression of CEBPB (CCAAT/enhancer binding protein (C/EBP), beta), an important adipogenic factor, suppressing its transcription in differentiated cells. In addition, G. cambogia inhibited macroautophagic/autophagic flux by decreasing autophagy-related gene expression and autophagosome formation. Notably, G. cambogia markedly elevated the expression of KLF3 (Kruppel-like factor 3 (basic)), a negative regulator of adipogenesis, by reducing SQSTM1/p62-mediated selective autophagic degradation. Furthermore, increased KLF3 induced by G. cambogia interacted with CTBP2 (C-terminal binding protein 2) to form a transcriptional repressor complex and inhibited Cebpa and Pparg transcription. Importantly, we found that RPS6KA1 and STAT3 were involved in the G. cambogia-mediated regulation of CEBPB and autophagic flux. In an obese animal model, G. cambogia reduced high-fat diet (HFD)-induced obesity by suppressing epididymal and inguinal subcutaneous white adipose tissue mass and adipocyte size, which were attributed to the regulation of targets that had been consistently identified in vitro. These findings provide new insight into the mechanism of G. cambogia-mediated regulation of adipogenesis and suggest molecular links to therapeutic targets for the treatment of obesity.Abbreviations: 3-MA: 3-methyladenine; ACTB: actin beta; ATG: autophagy-related; Baf: bafilomycin A1; BECN1: beclin 1; CEBP: CCAAT/enhancer binding protein (C/EBP); CHX: cycloheximide; CREB: cAMP response element binding protein; CTBP: C-terminal binding protein; EGCG: (-)-epigallocatechin gallate; eWAT: epididymal white; G. cambogia: Garcinia cambogia; GFP: green fluorescent protein; H&E: hematoxylin and eosin; HFD: high-fat diet; iWAT: inguinal subcutaneous white; KLF: Kruppel-like factor; LAP: liver-enriched transcriptional activating proteins; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; ND: normal diet; PPARG: peroxisome proliferator activated receptor gamma; qPCR: quantitative real-time PCR; RFP: red fluorescent protein; RPS6KA1: ribosomal protein S6 kinase A1; siRNA: small-interfering RNA; SQSTM1/p62: sequestosome 1; STAT: signal transducer and activator of transcription; TEM: transmission electron microscopy.


Subject(s)
Adipogenesis , Garcinia cambogia , Adipogenesis/genetics , Animals , Autophagy/physiology , Garcinia cambogia/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Obesity , PPAR gamma/metabolism , Protein Serine-Threonine Kinases , Sequestosome-1 Protein/metabolism
11.
Biomed Res Int ; 2021: 2521273, 2021.
Article in English | MEDLINE | ID: mdl-34812408

ABSTRACT

Obesity is a public health problem characterized by increased body weight due to abnormal adipose tissue expansion. Bioactive compound consumption from the diet or intake of dietary supplements is one of the possible ways to control obesity. Natural products with adipogenesis-regulating potential act as obesity treatments. We evaluated the synergistic antiangiogenesis, antiadipogenic and antilipogenic efficacy of standardized rebaudioside A, sativoside, and theasaponin E1 formulations (RASE1) in vitro in human umbilical vein endothelial cells (HUVECs), 3T3-L1 preadipocytes respectively, and in vivo using a high-fat and carbohydrate diet-induced obesity mouse model. Orlistat was used as a positive control, while untreated cells and animals were normal controls (NCs). Adipose tissue, liver, and blood were analyzed after dissection. Extracted stevia compounds and green tea seed saponin E1 exhibited pronounced antiobesity effects when combined. RASE1 inhibited HUVEC proliferation and tube formation by suppressing VEGFR2, NF-κB, PIK3, and-catenin beta-1 expression levels. RASE1 inhibited 3T3-L1 adipocyte differentiation and lipid accumulation by downregulating adipogenesis- and lipogenesis-promoting genes. RASE1 oral administration reduced mouse body and body fat pad weight and blood cholesterol, TG, ALT, AST, glucose, insulin, and adipokine levels. RASE1 suppressed adipogenic and lipid metabolism gene expression in mouse adipose and liver tissues and enhanced AMP-activated protein kinase levels in liver and adipose tissues and in serum adiponectin. RASE1 suppressed the NF-κB pathway and proinflammatory cytokines IL-10, IL-6, and TNF-α levels in mice which involve inflammation and progression of obesity. The overall results indicate RASE1 is a potential therapeutic formulation and functional food for treating or preventing obesity and inflammation.


Subject(s)
Biological Products/therapeutic use , Inflammation/drug therapy , Obesity/drug therapy , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Angiogenesis Inhibitors/therapeutic use , Animals , Biological Products/administration & dosage , Biological Products/toxicity , Disease Models, Animal , Diterpenes, Kaurane/administration & dosage , Drug Compounding , Drug Synergism , Female , Glucosides/administration & dosage , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/metabolism , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , Lipolysis/drug effects , Mice , Mice, Inbred ICR , Obesity/genetics , Obesity/metabolism , Oleanolic Acid/administration & dosage , Oleanolic Acid/analogs & derivatives , Phytotherapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saponins/administration & dosage , Signal Transduction/drug effects , Stevia/chemistry , Tea/chemistry
12.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768840

ABSTRACT

Obesity is a lipid metabolism disorder caused by genetic, medicinal, nutritional, and other environmental factors. It is characterized by a complex condition of excess lipid accumulation in adipocytes. Adipogenesis is a differentiation process that converts preadipocytes into mature adipocytes and contributes to excessive fat deposition. Saikosaponin A (SSA) and saikosaponin D (SSD) are triterpenoid saponins separated from the root of the Bupleurum chinensis, which has long been used to treat inflammation, fever, and liver diseases. However, the effects of these constituents on lipid accumulation and obesity are poorly understood. We investigated the anti-obesity effects of SSA and SSD in mouse 3T3-L1 adipocytes. The MTT assay was performed to measure cell viability, and Oil Red O staining was conducted to determine lipid accumulation. Various adipogenic transcription factors were evaluated at the protein and mRNA levels by Western blot assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Here, we showed that SSA and SSD significantly inhibited lipid accumulation without affecting cell viability within the range of the tested concentrations (0.938-15 µM). SSA and SSD also dose-dependently suppressed the expression of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), sterol regulatory element binding protein-1c (SREBP-1c), and adiponectin. Furthermore, the decrease of these transcriptional factors resulted in the repressed expression of several lipogenic genes including fatty acid binding protein (FABP4), fatty acid synthase (FAS), and lipoprotein lipase (LPL). In addition, SSA and SSD enhanced the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and its substrate, acetyl-CoA carboxylase (ACC), and inhibited the phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) and p38, but not c-Jun-N-terminal kinase (JNK). These results suggest that SSA and SSD inhibit adipogenesis through the AMPK or mitogen-activated protein kinase (MAPK) pathways in the early stages of adipocyte differentiation. This is the first study on the anti-adipogenic effects of SSA and SSD, and further research in animals and humans is necessary to confirm the potential of saikosaponins as therapeutic agents for obesity.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/drug effects , Adenylate Kinase/metabolism , Adipogenesis/genetics , Adiponectin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Obesity Agents/pharmacology , Bupleurum , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Gene Expression/drug effects , Lipogenesis/physiology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Obesity/drug therapy , Oleanolic Acid/pharmacology , PPAR gamma/drug effects , PPAR gamma/metabolism , Phosphorylation/drug effects , Plant Extracts/pharmacology , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/metabolism
13.
Nutrients ; 13(10)2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34684660

ABSTRACT

Decreased energy expenditure and chronically positive energy balance contribute to the prevalence of obesity and associated metabolic dysfunctions, such as dyslipidemia, hepatic fat accumulation, inflammation, and muscle mitochondrial defects. We investigated the effects of Chrysanthemum morifolium Ramat flower extract (CE) on obesity-induced inflammation and muscle mitochondria changes. Sprague-Dawley rats were randomly divided into four groups and fed either a normal diet, 45% high-fat diet (HF), HF containing 0.2% CE, or 0.4% CE for 13 weeks. CE alleviated HF-increased adipose tissue mass and size, dyslipidemia, hepatic fat deposition, and systematic inflammation, and increased energy expenditure. CE significantly decreased gene expression involved in adipogenesis, pro-inflammation, and the M1 macrophage phenotype, as well as glycerol-3-phosphate dehydrogenase (GPDH) and nuclear factor-kappa B (NF-kB) activities in epididymal adipose tissue. Moreover, CE supplementation improved hepatic fat accumulation and modulated gene expression related to fat synthesis and oxidation with an increase in adenosine monophosphate-activated protein kinase (AMPK) activity in the liver. Furthermore, CE increased muscle mitochondrial size, mitochondrial DNA (mtDNA) content, and gene expression related to mitochondrial biogenesis and function, including sirtuin 1 (SIRT1), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), and PGC-1α-target genes, along with AMPK-SIRT1 activities in the skeletal muscle. These results suggest that CE attenuates obesity-associated inflammation by modulating the muscle AMPK-SIRT1 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chrysanthemum/chemistry , Flowers/chemistry , Inflammation/drug therapy , Mitochondria, Muscle/metabolism , Obesity/complications , Plant Extracts/therapeutic use , Sirtuin 1/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue, White/metabolism , Animals , Body Weight/drug effects , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Diet, High-Fat , Dyslipidemias/complications , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Hypertrophy , Inflammation/etiology , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Plant Extracts/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
14.
BMC Complement Med Ther ; 21(1): 255, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34620154

ABSTRACT

BACKGROUND: Adipose tissue is a critical regulator of lipid storage and endocrine function. Impairment of the recruitment of new adipocytes in the adipose tissue is associated with ectopic fat accumulation, diabetes and insulin resistance. Torreya nucifera, an evergreen conifer that grows in warm temperate climates, has been found to exert beneficial effects against inflammation, infection and diabetes. However, the molecular mechanisms responsible for these effects at the cellular level remain unknown. This study aimed to investigate effects of Torreya nucifera seed oil (TNSO) on 3T3-L1 adipocyte differentiation and its underlying regulatory mechanism. METHODS: To investigate the effects of TNSO on adipocyte differentiation, 3T3-L1 cells were induced to differentiate for 5 days in the presence of 0.75 µL/mL TNSO. Oil Red O staining and an assay for intracellular triglyceride were performed to determine the extent of lipid accumulation in 3T3-L1 cells. To elucidate the underlying mechanism of TNSO, adipogenic gene expression was analyzed using quantitative real-time PCR. Moreover, we monitored TNSO-derived activation of PPARγ and STAT3 with 3T3-L1 reporter cell lines engineered to secrete Gaussia luciferase upon the interaction of a transcription factor to its DNA binding element. RESULTS: Oil Red O staining revealed that TNSO improved the differentiation of 3T3-L1 preadipocytes into mature adipocytes. The mRNA levels of adipogenic genes, including adiponectin, fatty acid synthase (FAS) and adipocyte fatty acid-binding protein (FABP4), were upregulated and intracellular triglyceride levels increased upon TNSO treatment. We also established that adipocyte differentiation was improved by TNSO-derived activation of PPARγ and STAT3. CONCLUSIONS: Our results suggest that TNSO improves adipocyte differentiation by regulating the activation of adipogenic transcription factors, indicating that it may serve as a potential treatment strategy for adipocyte dysfunction.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Taxaceae/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Fatty Acid-Binding Proteins , Gene Expression Regulation , Mice , PPAR gamma , Plant Oils/pharmacology , STAT3 Transcription Factor
15.
PLoS One ; 16(9): e0249438, 2021.
Article in English | MEDLINE | ID: mdl-34473703

ABSTRACT

Muscle derived stem cells (MDSCs) and myoblast play an important role in myotube regeneration when muscle tissue is injured. However, these cells can be induced to differentiate into adipocytes once exposed to PPARγ activator like EPA and DHA that are highly suggested during pregnancy. The objective of this study aims at determining the identity of trans-differentiated cells by exploring the effect of EPA and DHA on C2C12 undergoing differentiation into brown and white adipocytes. DHA but not EPA committed C2C12 cells reprograming into white like adipocyte phenotype. Also, DHA promoted the expression of lipolysis regulating genes but had no effect on genes regulating ß-oxidation referring to its implication in lipid re-esterification. Furthermore, DHA impaired C2C12 cells differentiation into brown adipocytes through reducing the thermogenic capacity and mitochondrial biogenesis of derived cells independent of UCP1. Accordingly, DHA treated groups showed an increased accumulation of lipid droplets and suppressed mitochondrial maximal respiration and spare respiratory capacity. EPA, on the other hand, reduced myogenesis regulating genes, but no significant differences were observed in the expression of adipogenesis key genes. Likewise, EPA suppressed the expression of WAT signature genes indicating that EPA and DHA have an independent role on white adipogensis. Unlike DHA treatment, EPA supplementation had no effect on the differential of C2C12 cells into brown adipocytes. In conclusion, DHA is a potent adipogenic and lipogenic factor that can change the metabolic profile of muscle cells by increasing myocellular fat.


Subject(s)
Adipocytes, White/drug effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Adipocytes, Brown/drug effects , Adipocytes, White/cytology , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , DNA, Mitochondrial , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipolysis/drug effects , Mice , Myoblasts/cytology , Myoblasts/drug effects
16.
J Microbiol Biotechnol ; 31(7): 1011-1021, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34099594

ABSTRACT

The root bark of Ulmus davidiana var. japonica (Japanese elm) is used in Korea and other East Asian countries as a traditional herbal remedy to treat a variety of inflammatory diseases and ailments such as edema, gastric cancer and mastitis. For this study, we investigated the lipid metabolism and anti-obesity efficacy of ethyl alcohol extract of Ulmus davidiana var. japonica root bark (UDE). First, HPLC was performed to quantify the level of (+)-catechin, the active ingredient of UDE. In the following experiments, cultured 3T3-L1 pre-adipocytes and high-fat diet (HFD)-fed murine model were studied for anti-obesity efficacy by testing the lipid metabolism effects of UDE and (+)-catechin. In the test using 3T3-L1 pre-adipocytes, treatment with UDE inhibited adipocyte differentiation and significantly reduced the production of adipogenic genes and transcription factors PPARγ, C/EBPα and SREBP-1c. HFD-fed, obese mice were administered with UDE (200 mg/kg per day) and (+)-catechin (30 mg/kg per day) by oral gavage for 4 weeks. Weight gain, epididymal and abdominal adipose tissue mass were significantly reduced, and a change in adipocyte size was observed in the UDE and (+)-catechin treatment groups compared to the untreated control group (***p < 0.001). Significantly lower total cholesterol and triglyceride levels were detected in UDE-treated HFD mice compared to the control, revealing the efficacy of UDE. In addition, it was found that lipid accumulation in hepatocytes was also significantly reduced after administration of UDE. These results suggest that UDE has significant anti-obesity and lipid metabolism effects through inhibition of adipocyte differentiation and adipogenesis.


Subject(s)
Anti-Obesity Agents/pharmacology , Diet, High-Fat/adverse effects , Lipid Metabolism/drug effects , Obesity/drug therapy , Ulmus/chemistry , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/chemistry , Catechin/administration & dosage , Catechin/pharmacology , Cell Differentiation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipids/blood , Mice , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Plant Bark/chemistry , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Weight Gain/drug effects
17.
Food Funct ; 12(10): 4621-4629, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33908983

ABSTRACT

Annona muricata (graviola) is a medicinal plant that can be used to alleviate chronic human diseases by providing antioxidants and inducing immunomodulation. In this study, we found that treatment of AML12 hepatocytes with steam (SGE) and ethanol (EGE) extracts of graviola leaf downregulated the expression of fatty acid (FA) oxidation genes, including ACOX1, CPT1, and PPARα, with no change in the expression of FA synthesis genes. However, whereas EGE inhibited the differentiation and lipid accumulation of 3T3-L1 adipocytes and downregulated FA synthesis genes, no similar changes were observed in response to treatment with SGE. In an in vivo experiment using mice fed a high-fat diet (HFD), body weight was reduced in response to treatment with EGE, which also dose-dependently alleviated liver hepatocyte ballooning induced by the consumption of a HFD. However, genes involved in FA oxidation and the secretion of very low density lipoprotein (VLDL) were downregulated. We also found that the size of adipocytes was reduced in response to EGE treatment, and that there was a downregulated expression of genes involved in adipogenesis and FA synthesis. Furthermore, we detected increases in the levels of cholesterol in the plasma, whereas ALT activity was reduced. Collectively, these results indicates that EGE inhibits lipid influx into the liver and adipogenesis in adipose tissues. These bioactive properties of EGE indicate its potential as a natural ingredient that can be used to prevent obesity.


Subject(s)
Adipogenesis/drug effects , Annona/chemistry , Lipogenesis/drug effects , Liver/metabolism , Plant Extracts/pharmacology , 3T3-L1 Cells , Acyl-CoA Oxidase/genetics , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Cell Differentiation/drug effects , Diet, High-Fat , Down-Regulation , Gene Expression Regulation/drug effects , Lipid Metabolism , Lipogenesis/genetics , Male , Mice , Mice, Inbred C57BL , PPAR alpha
18.
Nutr Res ; 86: 50-59, 2021 02.
Article in English | MEDLINE | ID: mdl-33482598

ABSTRACT

Citrus fruits contain an abundance of nutrients, including vitamins C and B6 and hesperidin, which attribute to its beneficial health effects. Previously, kimchi with Jeju citrus concentrate (CK) elicited anti-obesity effects in 3T3-L1 adipocytes. Here, we aimed to investigate whether CK exhibits anti-obesity effects by reducing serum and hepatic lipid concentrations and anti-obesity-associated gene expression in high-fat diet (HFD)-induced obese C57BL/6N mice. Low-dose CK (LDCK, 50 mg/kg) and high-dose CK (HDCK, 200 mg/kg) were orally administered 3 times per week over 8 weeks with HFD diet. Body weight gain, food efficiency ratio, and tissue weight were measured. Serum glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, fasting glucose, fasting insulin, homeostatic model assessment-insulin resistance, leptin, and adiponectin concentrations were also assessed. The effect of CK on the lipid profile and lipid accumulation was analyzed. Body and white adipose tissue masses were significantly lower in the LDCK and HDCK groups than in the HFD group. Orally administered CK significantly decreased serum lipid, fasting glucose, fasting insulin, homeostatic model assessment-insulin resistance, glutamic oxaloacetic transaminase, and glutamic pyruvic transaminase levels. Hepatic lipid content also decreased in the LDCK and HDCK groups. Serum leptin concentrations decreased, whereas serum adiponectin concentrations increased, confirming the anti-obesity effects of LDCK and HDCK. The decrease of hepatic vacuoles and stained lipid droplets indicated inhibition of lipid accumulation. These results support the hypothesis that CK exhibits anti-obesity effects in vivo by reducing lipid accumulation and by regulating anti-obesity-related genes.


Subject(s)
Citrus , Diet, High-Fat/adverse effects , Fermented Foods , Fruit , Lipid Metabolism , Obesity/diet therapy , Adipogenesis/genetics , Adiponectin/blood , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Weight , Gene Expression Regulation , Insulin Resistance , Leptin/blood , Lipids/blood , Lipogenesis/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism
19.
Eur J Pharmacol ; 892: 173689, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33127360

ABSTRACT

Portulaca oleracea L. is used as a folk medicine in many countries because of its wide range of pharmacological effects. HM-chromanone, isolated from P. oleracea using bioassay-guided fractionation and HPLC, belongs to the homoisoflavonoid group and has been shown to exert several biological effects. In this study, we evaluated whether HM-chromanone inhibits adipogenesis by regulating adipogenic transcription factors in 3T3-L1 adipocytes. The results showed that HM-chromanone suppresses adipocyte differentiation and adipogenesis in a dose-dependent manner in 3T3-L1 adipocytes. The HM-chromanone-treated adipocytes exhibited lower triglyceride accumulation and leptin secretion, and higher glycerol and adiponectin secretion than the control adipocytes. Microscopic observation using oil red O staining revealed a dose-dependent reduction in the number of lipid droplets in the HM-chromanone-treated adipocytes compared to the control group. HM-chromanone significantly down-regulated the protein expression of major adipogenic transcription factors sterol regulatory element binding protein-1c (SREBP-1c), peroxisome proliferator-activated receptor γ (PPARγ), and CCAAT/enhancer binding protein α (C/EBPα) and markedly inhibited several key adipogenic enzymes including fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC). In addition, adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) were both more activated in the HM-chromanone-treated adipocytes than in the control adipocytes. HM-chromanone also promoted the phosphorylation of 5' Adenosine monophosphate-activated protein kinase (AMPK), which inhibits adipogenesis through the regulation of adipogenic transcription factors. These results suggest that HM-chromanone may be an effective anti-adipogenesis agent that functions via the suppression of adipogenic transcription factors and the activation of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Isoflavones/pharmacology , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/pathology , Adipogenesis/genetics , Animals , Gene Expression Regulation , Lipid Metabolism/drug effects , Mice , Phosphorylation , Signal Transduction , Transcription Factors/genetics
20.
Eur J Pharmacol ; 891: 173686, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33121949

ABSTRACT

Adipogenesis, the maturation process of preadipocytes, is closely associated with the development of obesity and other complex metabolic syndromes. Herein, we investigated the effect of 7- methoxy-3-methyl-5-((E)- prop-1-enyl)-2-(3,4,5-trimethoxyphenyl)-2,3-dihydrobenzofuran (TM), a benzofuran, isolated from the mace of Myristica fragrans Houtt on adipogenesis in 3T3-L1 preadipocytes to extrapolate whether this compound has any anti-obesity potential. For this, 3T3-L1 preadipocytes were induced to differentiate in the presence of various concentrations of TM (1, 5, 10 µM) and analyzed for triglyceride (TG) accumulation and the expression of proteins and genes involved in lipogenesis and lipolysis associated with adipogenesis. Results showed that TM significantly reduced TG accumulation and expression of marker proteins of adipocyte differentiation (peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α, and fatty acid-binding protein 4) and increased the secretion of glycerol in a dose-dependent manner. There was a significant dose-dependent decrease in the expression of fatty acid synthase, stearoyl-CoA desaturase-1, sterol regulatory element-binding transcription factor 1c, and acetyl-CoA carboxylase 1 and an increase in carnitine palmitoyltransferase 1, acyl-CoA oxidase, and peroxisome proliferator-activated receptor α in TM treated cells. The phosphorylation of cAMP-activated protein kinase was also increased, which in turn activated the phosphorylation of acetyl-CoA carboxylase in mature adipocytes. Also, there was an increase in glucose uptake by TM, suggesting its insulin-sensitizing potential. This is the first report on the anti-obesity effects of TM from Myristica fragrans on adipogenesis and lipid metabolism in 3T3-L1 adipocytes and demands detailed in vivo study for developing TM as anti-obesity therapeutics.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Benzofurans/pharmacology , Lipid Metabolism/drug effects , Myristica , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Anti-Obesity Agents/isolation & purification , Benzofurans/isolation & purification , Gene Expression Regulation , Glucose/metabolism , Lipid Metabolism/genetics , Lipogenesis/drug effects , Lipolysis/drug effects , Mice , Myristica/chemistry , Plant Extracts/isolation & purification , Signal Transduction , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL