Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Physiol Behav ; 273: 114401, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37939828

ABSTRACT

AIM: The present study aimed to investigate the effect of the intracerebroventricular (icv) administration of spexin on the hypothalamus-pituitary-thyroid (HPT) axis (TRH, TSH, T4 and T3 hormones) and energy expenditure (PGC-1α and UCP1 genes) in white adipose (WAT) and brown adipose tissues (BAT) in rats. Furthermore, the study aimed to determine the effects of spexin on food-water consumption and body weight of rats. MATERIAL AND METHOD: The study was conducted with 40 male rats that were divided into 4 groups: Control, Sham, Spexin 30 and Spexin 100 (n = 10). Spexin (1 µl/hour) was administered to rats other than those in the control group for 7 days with osmotic minipumps intracerebroventricularly, artificial cerebrospinal fluid (vehicle) was administered to the Sham group, and 30 nMol and 100 nMol spexin was infused to the Spexin 30 and Spexin 100 groups, respectively. Food-water consumption and body weight of the rats were monitored during the experiments. After the seven-day infusion, the rats were decapitated and serum TSH, fT4 and fT3 levels were determined with ELISA on rat blood samples. Also, TRH gene expression levels from the hypothalamus tissues and PGC-1α and UCP1 expression levels from WAT and BAT were determined by real-time PCR. FINDINGS: It was determined that icv spexin infusion reduced daily food consumption and body weight without leading to a significant change in water consumption (p < 0.05). Icv spexin infusion significantly decreased serum TSH, and increased fT4 and fT3 levels when compared to control and sham groups (p < 0.05). Moreover, icv spexin infusion increased the TRH expressions in the hypothalamus tissues and PGC-1α UCP1 in the WAT and BAT (p < 0.05). CONCLUSION: Icv Spexin infusion may have effects on food consumption and body weight as well as, thyroid hormones and energy metabolism.


Subject(s)
Thyroid Gland , Thyroxine , Rats , Male , Animals , Thyroid Gland/metabolism , Triiodothyronine , Adipocytes, Brown , Organelle Biogenesis , Hypothalamus/metabolism , Body Weight , Thyrotropin/metabolism , Thyrotropin/pharmacology
2.
Int J Mol Sci ; 24(22)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-38003364

ABSTRACT

Mammary fat plays a profound role in the postnatal development of mammary glands. However, the specific types (white, brown, or beige) of adipocytes in mammary fat and their potential regulatory effects on modulating mammary gland development remain poorly understood. This study aimed to investigate the role of the browning of mammary fat on pubertal mammary gland development and explore the underlying mechanisms. Thus, the mammary gland development and the serum lipid profile were evaluated in mice treated with CL316243, a ß3-adrenoceptor agonist, to induce mammary fat browning. In addition, the proliferation of HC11 cells co-cultured with brown adipocytes or treated with the altered serum lipid metabolite was determined. Our results showed that the browning of mammary fat by injection of CL316243 suppressed the pubertal development of mice mammary glands, accompanied by the significant elevation of serum dioleoylphosphocholine (DOPC). In addition, the proliferation of HC11 was repressed when co-cultured with brown adipocytes or treated with DOPC. Furthermore, DOPC suppressed the activation of the PI3K/Akt pathway, while the DOPC-inhibited HC11 proliferation was reversed by SC79, an Akt activator, suggesting the involvement of the PI3K/Akt pathway in the DOPC-inhibited proliferation of HC11. Together, the browning of mammary fat suppressed the development of the pubertal mammary gland, which was associated with the elevated serum DOPC and the inhibition of the PI3K/Akt pathway.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Animals , Mice , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Adipocytes, Brown/metabolism , Lecithins/pharmacology
3.
Endocrinology ; 164(4)2023 02 11.
Article in English | MEDLINE | ID: mdl-36825874

ABSTRACT

Berberine (BBR), which is a compound derived from the Chinese medicinal plant Coptis chinensis, promotes weight loss, but the molecular mechanisms are not well understood. Here, we show that BBR increases the serum level of growth differentiation factor 15 (GDF15), which is a stress response cytokine that can reduce food intake and lower body weight in diet-induced obese (DIO) mice. The body weight and food intake of DIO mice were decreased after BBR treatment, and the weight change was negatively correlated with the serum GDF15 level. Further studies show that BBR induced GDF15 mRNA expression and secretion in the brown adipose tissue (BAT) of DIO mice and primary mouse brown adipocytes. In addition, we found that BBR upregulates GDF15 mRNA expression and secretion by activating the integrated stress response (ISR) in primary mouse brown adipocytes. Overall, our findings show that BBR lowers body weight by inducing GDF15 secretion via the activation of the ISR in BAT.


Subject(s)
Adipocytes, Brown , Berberine , Animals , Mice , Adipocytes, Brown/metabolism , Berberine/metabolism , Berberine/pharmacology , Growth Differentiation Factor 15/genetics , Obesity/metabolism , Body Weight , Adipose Tissue, Brown/metabolism , RNA, Messenger/metabolism
4.
Int J Biol Macromol ; 222(Pt B): 1963-1973, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36252624

ABSTRACT

BACKGROUND: Obesity, fatty liver, type 2 diabetes, and Non-alcoholic fatty liver disease (NAFLD) are all metabolic diseases caused by excess food consumption. Existing drug molecules had negative side effects and caused other diseases to develop (Orlistat causes angioedema, and menstrual irregularities; megestrol acetate causes hypertension, and insomnia). By enhancing lipid consumption and increasing nonshivering thermogenesis, targeting mitochondrial uncoupling protein-1 (UCP1) expression in adipocytes could be an auspicious treatment strategy against obesity or metabolic disorders associated with obesity. METHODS: We used previously produced UCP1-A-GFP reporter cell lines in this investigation to find new pharmacological compounds against obesity or metabolic syndrome, which we then tested in cellular analysis, cytotoxicity, mitochondrial function, mitochondrial DNA quantification, mitochondrial ATP production, and in-silico models. RESULTS: Baicalein was discovered to play a critical role in obesity prevention via altering mitochondrial function. Baicalein lowers ATP generation while increasing considerable UCP1 gene expression in brown adipocytes. As a result, cellular thermogenesis is boosted. The HEK293T cell line is harmless by baicalein. The investigation by the in-silico study revealed drug-protein interaction and UCP1 binding. Thus, our research clarifies baicalein's therapeutic role in metabolic and obesity-related illnesses via modulating mitochondrial activity (Supplementary Fig. 2). CONCLUSIONS: Further studies are required in both murine and human models to understand the full mechanism of action by mitochondrial modulation. Drug development investigation also requires to development of a precise formulation.


Subject(s)
Adipocytes, Brown , Diabetes Mellitus, Type 2 , Humans , Mice , Animals , Adipocytes, Brown/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Type 2/metabolism , HEK293 Cells , Mitochondria , Obesity/metabolism , Adenosine Triphosphate/metabolism , Mitochondrial Proteins/metabolism
5.
Eur J Pharmacol ; 935: 175345, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36283451

ABSTRACT

Dihydromyricetin (DHM) is a natural bioactive flavonoid extracted from Ampelopsis Grossedentata, a commonly used Chinese herbal medicine. It has multiple beneficial pharmacological effects including lowering blood glucose and lipid, as well as anti-inflammation, anti-oxidation and hepato-protection. In this study, we elucidated its actions on mitochondrial dynamics and browning of white adipose. In the experiments in vivo, six-week-old male C57BL/6 mice were fed with normal diet (ND), high-fat diet (HFD), or HFD with intragastric administration of DHM (250 mg/kg.d-1); in the experiments in vitro, 3T3-L1 and mouse primary preadipocytes were induced and treated with various concentrations of DHM. The mouse metabolic phenotype, lipid accumulation, the browning and mitochondrial dynamics of white adipocytes were examined. It was found that DHM treatment reduced body weight and fat mass, improved glucose tolerance, insulin resistance and cold tolerance in mice with obesity. DHM treatment increased the expressions of classical brown adipocyte markers (UCP-1, PGC-1α, PRDM16) and mitochondrial dynamics-related proteins (DRP1, FIS1, OPA1, MFN2) in adipose tissue. Likewise, DHM treatment induced the differentiation of mature 3T3-L1 cells into brown-like adipocytes and also enhanced the expressions of mitochondrial dynamics-related proteins in vitro. Moreover, the pro-browning effect of DHM can be abrogated by mitochondrial fission inhibitor Mdivi-1. These findings indicate that DHM treatment induces the browning-remodeling of white adipose by enhancing mitochondrial fission and manifests an anti-obesity property via pro-browning mediated by mitochondrial fission, which implies it may play important roles in prevention and therapy of obesity and related diseases.


Subject(s)
Diet, High-Fat , Mitochondrial Dynamics , Male , Mice , Animals , Mice, Inbred C57BL , 3T3-L1 Cells , Diet, High-Fat/adverse effects , Adipocytes, Brown , Adipose Tissue/metabolism , Obesity/metabolism , Weight Loss , Lipids , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism
6.
Am J Chin Med ; 50(5): 1315-1329, 2022.
Article in English | MEDLINE | ID: mdl-35642460

ABSTRACT

The aim of this study is to investigate the molecular mechanism of Smilax china L. polyphenols (SCLPs) in enhancing lipid metabolism and stimulating browning to reduce lipid accumulation in 3T3-L1 adipocytes. SCLP treatment obviously decreased lipid content in a dose-dependent manner (10-40 µg/mL) in adipocytes. SCLP treatment cooperated with noradrenalin to increase lipolysis. SCLPs reduced the gene expressions of C/EBP[Formula: see text] and Ap2 and enhanced the expressions of ACO, CPT, pHSL/HSL, ATGL, and PKA in adipocytes. Furthermore, SCLPs increased mRNA and protein expressions of brown adipocyte-specific factors (UCP-1, PRDM16, PGC-1α, and PPARγ) and mRNA expressions of beige adipocyte-specific markers (CD137, Tbx1, and Tmem26) in 3T3-L1 adipocytes, as well as mitochondrial biogenesis genes (Nrf1 and Tfam). In addition, according to the immunofluorescence staining, the mitochondria number was increased by SCLP. Moreover, ß3-AR or AMPK agonist synergistic SCLPs enhanced the expressions of UCP-1, PRDM16, and PGC-1α. While ß3-AR or AMPK antagonist significantly decreased the expressions of these brown adipocyte-specific factors, SCLP treatment inhibited the effect of antagonist to improve the expression of UCP-1, PRDM16, and PGC-1α. These results indicated that SCLPs may regulate lipid metabolism and stimulate browning via the ß3-AR/AMPKα signaling pathway. Thus, SCLPs likely have potential therapeutic effects on obesity.


Subject(s)
AMP-Activated Protein Kinases , Smilax , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/metabolism , Animals , China , Lipids , Mice , Polyphenols/metabolism , Polyphenols/pharmacology , RNA, Messenger/metabolism , Receptors, Adrenergic/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Phytomedicine ; 96: 153857, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34840022

ABSTRACT

BACKGROUND: Natural compounds with medicinal properties are part of a strategic trend in the treatment of obesity. The vitamin A agent, ß-carotene, is a well-known carotenoid, and its numerous functions in metabolism have been widely studied. The activation of thermogenesis by stimulating white fat browning (beiging) has been identified as a treatment for obese individuals. PURPOSE: The current study was undertaken to unveil the browning activity of ß-carotene in 3T3-L1 white adipocytes. METHODS: The effects of ß-carotene were evaluated in 3T3-L1 white adipocytes, and gene/protein expressions were determined by performing quantitative real-time PCR, immunoblot analysis, immunofluorescence assessment, and molecular docking techniques. RESULTS: ß-carotene strikingly increased the expression levels of brown-fat-specific marker proteins (UCP1, PRDM16, and PGC-1α) and beige-fat-specific genes (Cd137, Cidea, Cited1, andTbx1) in 3T3-L1 cells. Exposure to ß-carotene also elevated the expressions of key adipogenic transcription factors C/EBPα and PPARγ in white adipocytes but decreased the expressions of lipogenic marker proteins ACC and FAS. Moreover, lipolysis and fat oxidation were regulated by ß-carotene via upregulation of ATGL, pHSL, ACOX, and CPT1. In addition, molecular docking studies revealed ß-carotene activation of the adenosine A2A receptor and ß3-AR. ß-Carotene increased the expressions of mitochondrial biogenic markers, stimulated the ß3-AR and p38 MAPK signaling pathways and its downstream signaling molecules (SIRTs and ATF2), thereby inducing browning. CONCLUSIONS: Taken together, our results indicate the potential of ß-carotene as a natural-source therapeutic anti-obesity agent.


Subject(s)
Sirtuins , beta Carotene , 3T3-L1 Cells , Adipocytes, Brown , Adipocytes, White , Animals , Humans , Mice , Molecular Docking Simulation , Signal Transduction , Thermogenesis , p38 Mitogen-Activated Protein Kinases
8.
Am J Chin Med ; 49(8): 1929-1948, 2021.
Article in English | MEDLINE | ID: mdl-34961413

ABSTRACT

Although gomisin A (GA) alleviates cancer and inflammation, its anti-obesity effect and the underlying mechanism have not yet been elucidated. Therefore, in this study, we aimed to elucidate the anti-obesity effects of GA by investigating the phenotypic changes involved in the browning and whitening of adipocytes. Here, obesity was induced to C57BL/6J mice using a high-fat diet (HFD). We administrated GA and checked weight changes for 12 weeks. We found that GA decreased the weight of weight gain, epididymal white adipose tissue (eWAT), and liver in the mice. In addition, the administration of GA elevated the levels of high-density lipoprotein (HDL)-cholesterol in the mice serum. Moreover, even after 12 weeks of treatment with GA, it did not cause any hepatic and renal toxicity. However, we found that GA induced the browning of eWAT and inhibited the whitening of brown adipose tissue. We further confirmed the anti-obesity mechanism of GA using 3T3-L1 cells, the human adipose mesenchymal stem cells (hAMSCs), and primary brown adipocytes (BAs) in vitroexperiments. We found that GA suppressed adipogenesis via the activation of AMP-activated protein kinase (AMPK). Furthermore, GA-induced browning by increasing the expression levels of uncoupling protein 1 (UCP1) in hAMSCs. The results of our study indicate that GA can inhibit weight gain by regulating the phenotypic changes involved in the browning and whitening of adipose tissues, which makes it a potential therapeutic agent for the treatment of obesity.


Subject(s)
Adipocytes, Brown , Obesity , 3T3-L1 Cells , Adipose Tissue, Brown , Animals , Cyclooctanes , Diet, High-Fat/adverse effects , Dioxoles , Lignans , Mice , Mice, Inbred C57BL , Obesity/drug therapy
9.
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
10.
Phytother Res ; 35(11): 6281-6294, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34523169

ABSTRACT

Microbial fermentation of grape-skin extracts is found to synthesize anthocyanin oligomers (AO), which are more active than the monomeric anthocyanins that are effective for some metabolic diseases such as diabetes and obesity. This study investigated the functional role of AO in 3T3-L1 white adipocyte metabolism, with a focus on inducing browning. To achieve this, we determined the expressions of core genes and protein markers responsible for browning and lipid metabolism in response to AO treatment of 3T3-L1 white adipocytes. AO exposure significantly increases the expressions of beige-specific genes (Cidea, Cited1, Ppargc1α, Prdm16, Tbx1, Tmem26, and Ucp1) and brown-fat signature proteins (UCP1, PRDM16, and PGC-1α), and suppresses the expressions of lipogenic marker proteins while enhancing the protein levels of lipolysis in white adipocytes. The mechanistic study revealed stimulation of white fat browning via activation of the ß3-AR/PKA/p38 axis and ERK/CREB signaling pathway subsequent to AO treatment. In conclusion, our current findings indicate the beneficial effects of AO for the treatment of obesity with interesting properties such as regulating the browning of adipocytes and increasing thermogenic activity. Although further research based on animal models or clinical trials remains, AO treatment can bring more insights into the treatment of obesity and metabolic syndrome.


Subject(s)
Adipocytes, White , Anthocyanins , 3T3-L1 Cells , Adipocytes, Brown , Animals , Anthocyanins/pharmacology , MAP Kinase Signaling System , Mice , Receptors, Adrenergic , Signal Transduction , Thermogenesis
11.
Sci Rep ; 11(1): 2008, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479386

ABSTRACT

Obesity is associated with the growth and expansion of adipocytes which could be decreased via several mechanisms. Cissus Quadrangularis (CQ) extract has been shown to reduce obesity in humans; however, its effect on human white adipocytes (hWA) has not been elucidated. This study aimed to investigate the effects of CQ on obesity, lipolysis, and browning of hWA. CQ treatment in obese humans significantly decreased waist circumference at week 4 and week 8 when compared with the baseline values (p < 0.05 all) and significantly decreased hip circumference at week 8 when compared with the baseline and week 4 values (p < 0.05 all). Serum leptin levels of the CQ-treated group were significantly higher at week 8 compared to baseline levels (p < 0.05). In hWA, glycerol release was reduced in the CQ-treated group when compared with the vehicle-treated group. In the browning experiment, pioglitazone, the PPAR-γ agonist, increased UCP1 mRNA when compared to vehicle (p < 0.01). Interestingly, 10, 100, and 1000 ng/ml CQ extract treatment on hWA significantly enhanced UCP1 expression in a dose-dependent manner when compared to pioglitazone treatment (p < 0.001 all). In conclusion, CQ decreased waist and hip circumferences in obese humans and enhanced UCP1 mRNA in hWA suggestive of its action via browning of hWA.


Subject(s)
Cissus/chemistry , Obesity, Abdominal/drug therapy , Plant Extracts/administration & dosage , Uncoupling Protein 1/genetics , Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Adult , Female , Humans , Leptin/genetics , Lipolysis/drug effects , Male , Obesity, Abdominal/pathology , Plant Extracts/chemistry , RNA, Messenger/genetics
12.
Mol Nutr Food Res ; 65(2): e2000681, 2021 01.
Article in English | MEDLINE | ID: mdl-33274552

ABSTRACT

SCOPE: Brown and brite adipocytes within the mammalian adipose organ provide non-shivering thermogenesis and thus, have an exceptional capacity to dissipate chemical energy as heat. Polyunsaturated fatty acids (PUFA) of the n3-series, abundant in fish oil, have been repeatedly demonstrated to enhance the recruitment of thermogenic capacity in these cells, consequently affecting body adiposity and glucose tolerance. These effects are scrutinized in mice housed in a thermoneutral environment and in a human dietary intervention trial. METHODS AND RESULTS: Mice are housed in a thermoneutral environment eliminating the superimposing effect of mild cold-exposure on thermogenic adipocyte recruitment. Dietary fish oil supplementation in two different inbred mouse strains neither affects body mass trajectory nor enhances the recruitment of brown and brite adipocytes, both in the presence and absence of a ß3-adrenoreceptor agonist imitating the effect of cold-exposure on adipocytes. In line with these findings, dietary fish oil supplementation of persons with overweight or obesity fails to recruit thermogenic adipocytes in subcutaneous adipose tissue. CONCLUSION: Thus, the authors' data question the hypothesized potential of n3-PUFA as modulators of adipocyte-based thermogenesis and energy balance regulation.


Subject(s)
Adipocytes, Beige/drug effects , Adipocytes, Brown/drug effects , Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Subcutaneous Fat/drug effects , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Adult , Animals , Dietary Supplements , Fatty Acids, Omega-3/metabolism , Female , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Humans , Male , Mice, Inbred C57BL , Mice, Inbred Strains , Middle Aged , Palm Oil/pharmacology , Plant Oils/pharmacology , Subcutaneous Fat/physiology , Thermogenesis/drug effects , Thermogenesis/physiology , gamma-Linolenic Acid/pharmacology
13.
Phytother Res ; 35(2): 920-931, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32840919

ABSTRACT

In this study, we investigated the effect of Biochanin A (BioA), an O-methylated isoflavone on the brown-fat phenotype formation and on the associated thermogenic program including mitochondrial biogenesis and lipolysis in C3H10T1/2 MSCs. Our data demonstrates that Treatment with BioA in an adipogenic differentiation cocktail induced formation of brown-fat-like adipocytes from C3H10T1/2 MSCs without treatment with a known browning inducer (rosiglitazone or T3) at an early stage of differentiation. The formation of brown-fat-like adipocytes by BioA treatment was evidenced by upregulation of key thermogenic markers: Ucp1, Pgc1α, Prdm16, and Pparγ. BioA also increased the expression of beige (Cd137 and Fgf21) and brown (Elovl3 and Zic1)-specific markers. Additionally, BioA treatment promoted mitochondrial biogenesis, judging by the upregulation of genes; Cox8b, Cidea, Dio2, Sirt1, Opa1, and Fis1. BioA treatment increased the amount of mitochondrial DNA and its encoded proteins: oxidative phosphorylation complexes (I-V); this change was associated with high oxygen consumption by C3H10T1/2 MSCs. A small-interfering-RNA-induced gene knockdown and experiments with dorsomorphin-driven competitive inhibition revealed that BioA exerts the thermogenic action via activation of AMPK signaling. Our study shows the mechanism of BioA-induced promotion of a brown-fat phenotype. Nonetheless, clinical research is necessary to validate BioA as a brown-fat-like signature inducer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/drug effects , Anticarcinogenic Agents/therapeutic use , Genistein/therapeutic use , Mesenchymal Stem Cells/drug effects , Animals , Anticarcinogenic Agents/pharmacology , Cell Differentiation , Genistein/pharmacology , Mice , Organelle Biogenesis , Signal Transduction , Transfection
14.
Phytother Res ; 35(2): 1113-1124, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33015893

ABSTRACT

Trigonelline, a major alkaloid component of fenugreek, has been demonstrated to have several biological activities, including antidiabetic and anticancer effects. This study aimed to examine the possible application of trigonelline as an anti-obesity compound based on an investigation of its enhancement of lipid catabolism and induction of browning in white adipocytes. Trigonelline induces browning of 3T3-L1 white adipocytes by enhancing the expressions of brown-fat signature proteins and genes as well as beige-specific genes, including Cd137, Cited1, Tbx1, and Tmem26. Trigonelline also improves lipid metabolism in white adipocytes by decreasing adipogenesis and lipogenesis as well as promotes lipolysis and fatty acid oxidation. Moreover, trigonelline increases the expression of Cox4, Nrf1, and Tfam genes that are responsible for mitochondrial biogenesis. Mechanistic studies revealed that the browning effect of trigonelline in 3T3-L1 white adipocytes is mediated by activating ß3-AR and inhibiting PDE4, thereby stimulating the p38 MAPK/ATF-2 signaling pathway. Considering its high bioavailability in humans and the results of this study, trigonelline may have potential as an anti-obesity compound.


Subject(s)
3T3-L1 Cells/metabolism , Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Alkaloids/therapeutic use , Obesity/drug therapy , Alkaloids/pharmacology , Animals , Humans , Mice
15.
Exp Biol Med (Maywood) ; 246(2): 163-176, 2021 01.
Article in English | MEDLINE | ID: mdl-33045853

ABSTRACT

Obesity is one of major risk factors increasing chronic diseases including type II diabetes, cardiovascular diseases, and hypertension. The effects of epigallocatechin gallate (EGCG), the major active compound in green tea, on reduced obesity and improved metabolic profiles are still controversial. Furthermore, the effects of EGCG on human adipocyte lipolysis and browning of white adipocytes have not been elucidated. This study aimed to investigate the effects of EGCG on obesity, lipolysis, and browning of human white adipocytes. The results showed that, when compared to the baseline values, EGCG significantly decreased fasting plasma triglyceride levels (P < 0.05), systolic blood pressure (P < 0.05), diastolic blood pressure (P < 0.05), and serum kisspeptin levels (P < 0.05) after 8 weeks of supplement. On the other hand, supplement of EGCG in obese human subjects for 4 or 8 weeks did not decrease body weight, body mass index, waist and hip circumferences, nor total body fat mass or percentage when compared to their baseline values. The study in human adipocytes showed that EGCG did not increase the glycerol release when compared to vehicle, suggesting that it had no lipolytic effect. Furthermore, treatment of EGCG did not enhance uncoupling protein 1 (UCP1) mRNA expression in human white adipocytes when compared with treatment of pioglitazone, the peroxisome proliferator-activated receptor γ (PPAR-γ) agonist, suggesting that EGCG did not augment the browning effect of PPAR-γ on white adipocytes. This study revealed that EGCG reduced 2 metabolic risk factors which are triglyceride and blood pressure in the human experiment. We also showed a novel evidence that EGCG decreased kisspeptin levels. However, EGCG had no effects on obesity reduction in humans, lipolysis, nor browning of human white adipocytes.


Subject(s)
Blood Pressure/drug effects , Catechin/analogs & derivatives , Kisspeptins/blood , Obesity/blood , Obesity/physiopathology , Triglycerides/blood , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adiponectin/blood , Adult , Blood Glucose/metabolism , Catechin/pharmacology , Humans , Kidney/drug effects , Kidney/physiopathology , Leptin/blood , Lipolysis , Liver/drug effects , Liver/physiopathology , Middle Aged , Obesity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
16.
Int J Mol Sci ; 21(23)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287103

ABSTRACT

The effects of insulin on the bioenergetic and thermogenic capacity of brown adipocyte mitochondria were investigated by focusing on key mitochondrial proteins. Two-month-old male Wistar rats were treated acutely or chronically with a low or high dose of insulin. Acute low insulin dose increased expression of all electron transport chain complexes and complex IV activity, whereas high dose increased complex II expression. Chronic low insulin dose decreased complex I and cyt c expression while increasing complex II and IV expression and complex IV activity. Chronic high insulin dose decreased complex II, III, cyt c, and increased complex IV expression. Uncoupling protein (UCP) 1 expression was decreased after acute high insulin but increased following chronic insulin treatment. ATP synthase expression was increased after acute and decreased after chronic insulin treatment. Only a high dose of insulin increased ATP synthase activity in acute and decreased it in chronic treatment. ATPase inhibitory factor protein expression was increased in all treated groups. Confocal microscopy showed that key mitochondrial proteins colocalize differently in different mitochondria within a single brown adipocyte, indicating mitochondrial mosaicism. These results suggest that insulin modulates the bioenergetic and thermogenic capacity of rat brown adipocytes in vivo by modulating mitochondrial mosaicism.


Subject(s)
Adipocytes, Brown/metabolism , Energy Metabolism , Insulin/metabolism , Mitochondria/metabolism , Thermogenesis , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Biomarkers , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism/drug effects , Enzyme Activation , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Expression , Insulin/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mosaicism , Rats , Thermogenesis/drug effects , Thermogenesis/genetics
17.
Cells ; 9(11)2020 11 07.
Article in English | MEDLINE | ID: mdl-33171828

ABSTRACT

Thermogenic brown and brite adipocytes convert chemical energy from nutrients into heat. Therapeutics that regulate brown adipocyte recruitment and activity represent interesting strategies to control fat mass such as in obesity or cachexia. The peroxisome proliferator-activated receptor (PPAR) family plays key roles in the maintenance of adipose tissue and in the regulation of thermogenic activity. Activation of these receptors induce browning of white adipocyte. The purpose of this work was to characterize the role of carnosic acid (CA), a compound used in traditional medicine, in the control of brown/brite adipocyte formation and function. We used human multipotent adipose-derived stem (hMADS) cells differentiated into white or brite adipocytes. The expression of key marker genes was determined using RT-qPCR and western blotting. We show here that CA inhibits the browning of white adipocytes and favors decreased gene expression of thermogenic markers. CA treatment does not affect ß-adrenergic response. Importantly, the effects of CA are fully reversible. We used transactivation assays to show that CA has a PPARα/γ antagonistic action. Our data pinpoint CA as a drug able to control PPAR activity through an antagonistic effect. These observations shed some light on the development of natural PPAR antagonists and their potential effects on thermogenic response.


Subject(s)
Abietanes/pharmacology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Rosmarinus/chemistry , Adipocytes, Beige/drug effects , Adipocytes, Beige/metabolism , Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Animals , Biomarkers/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Lipolysis/drug effects , Mice , Peroxisome Proliferator-Activated Receptors/metabolism , Rosiglitazone/pharmacology , Thermogenesis/drug effects , Thermogenesis/genetics
18.
Sci Rep ; 10(1): 14175, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843714

ABSTRACT

Patients with chronic kidney disease (CKD) are often 25(OH)D3 and 1,25(OH)2D3 insufficient. We studied whether vitamin D repletion could correct aberrant adipose tissue and muscle metabolism in a mouse model of CKD-associated cachexia. Intraperitoneal administration of 25(OH)D3 and 1,25(OH)2D3 (75 µg/kg/day and 60 ng/kg/day respectively for 6 weeks) normalized serum concentrations of 25(OH)D3 and 1,25(OH)2D3 in CKD mice. Vitamin D repletion stimulated appetite, normalized weight gain, and improved fat and lean mass content in CKD mice. Vitamin D supplementation attenuated expression of key molecules involved in adipose tissue browning and ameliorated expression of thermogenic genes in adipose tissue and skeletal muscle in CKD mice. Furthermore, repletion of vitamin D improved skeletal muscle fiber size and in vivo muscle function, normalized muscle collagen content and attenuated muscle fat infiltration as well as pathogenetic molecular pathways related to muscle mass regulation in CKD mice. RNAseq analysis was performed on the gastrocnemius muscle. Ingenuity Pathway Analysis revealed that the top 12 differentially expressed genes in CKD were correlated with impaired muscle and neuron regeneration, enhanced muscle thermogenesis and fibrosis. Importantly, vitamin D repletion normalized the expression of those 12 genes in CKD mice. Vitamin D repletion may be an effective therapeutic strategy for adipose tissue browning and muscle wasting in CKD patients.


Subject(s)
Adipocytes, Beige/drug effects , Cachexia/drug therapy , Calcifediol/therapeutic use , Calcitriol/therapeutic use , Renal Insufficiency, Chronic/complications , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Animals , Cachexia/etiology , Cachexia/physiopathology , Calcifediol/blood , Calcifediol/deficiency , Calcifediol/pharmacology , Calcitriol/blood , Calcitriol/deficiency , Calcitriol/pharmacology , Disease Models, Animal , Eating/drug effects , Fibrosis/genetics , Gene Expression Regulation/drug effects , Hand Strength , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Nephrectomy , Parathyroid Hormone/blood , RNA, Messenger/biosynthesis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/drug therapy , Rotarod Performance Test , Sequence Analysis, RNA , Thermogenesis/drug effects , Weight Gain/drug effects
19.
Phytomedicine ; 78: 153292, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32777487

ABSTRACT

BACKGROUND: Brown adipose tissue (BAT) activation is a promising therapeutic target to treat hyperlipidemia with obesity. Huang-Qi San (HQS), an traditional Chinese medicine, can ameliorate hyperlipidemia with obesity, but its mechanism of action (MOA) is not understood. PURPOSE: To articulate the MOA for HQS with animal models. METHODS: The main chemical constituents of HQS were identified by high-performance liquid chromatography (HPLC) based assay. Hyperlipidemia with obesity rat models induced by high-fat diet were employed in the study. The levels of the fasting plasma glucose (FPG), triglyceride (TG), total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C) and high-density lipoprotein-cholesterol (HDL-C) were measured to evaluate the ability of HQS to ameliorate hyperlipidemia with obesity. Pathological analyses of organs were conducted with Oil Red O staining, hematoxylin-eosin (H&E) staining and transmission electron microscopy. The expression of mRNAs related to thermogenic genes, fatty acid oxidation-related genes and mitochondria biogenic genes were examined by quantitative real-time PCR. The protein expressions of uncoupling protein 1 (UCP1) were investigated by immunohistochemistry and western blot. Simultaneously, the protein expression of PR domain containing 16 (PRDM16), ATP synthase F1 subunit alpha (ATP5A) was detected by western blot. RESULTS: HQS ameliorates metabolic disorder, lipid ectopic deposition, obesity and maintained glucose homeostasis in hyperlipidemia with obesity rats. HQS can significantly increase the number of mitochondria and reduced the size of the intracellular lipid droplets in BAT, and increase the expression of BAT activation-related genes (UCP1, PGC1α, PGC1ß, Prdm16, CD137, TBX1, CPT1a, PPARα, Tfam, NRF1 and NRF2) in vivo. Furthermore, UCP1, PRDM16 and ATP5A proteins of BAT were increased. CONCLUSION: HQS can activate BAT and browning of S-WAT (subcutaneous white adipose tissue) through activating the PRDM16/PGC1α/UCP1 pathway, augmenting mitochondrial biogenesis and fatty acid oxidation to increase thermogenesis and energy expenditure, resulting in a significant amelioration of hyperlipidemia with obesity. Therefore, HQS is an effective therapeutic medicine for the treatment of hyperlipidemia with obesity.


Subject(s)
Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Drugs, Chinese Herbal/pharmacology , Hyperlipidemias/drug therapy , Adipocytes, Brown/physiology , Adipocytes, White/metabolism , Adipocytes, White/physiology , Animals , Astragalus propinquus , Diet, High-Fat/adverse effects , Drugs, Chinese Herbal/chemistry , Energy Metabolism/drug effects , Fatty Acids/metabolism , Gene Expression Regulation/drug effects , Hyperlipidemias/etiology , Hyperlipidemias/pathology , Lipid Metabolism/drug effects , Male , Obesity/drug therapy , Obesity/etiology , Rats, Sprague-Dawley , Thermogenesis/drug effects
20.
EMBO Rep ; 21(9): e49807, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32657019

ABSTRACT

This study investigated the role of CDK4 in the oxidative metabolism of brown adipose tissue (BAT). BAT from Cdk4-/- mice exhibited fewer lipids and increased mitochondrial volume and expression of canonical thermogenic genes, rendering these mice more resistant to cold exposure. Interestingly, these effects were not BAT cell-autonomous but rather driven by increased sympathetic innervation. In particular, the ventromedial hypothalamus (VMH) is known to modulate BAT activation via the sympathetic nervous system. We thus examined the effects of VMH neuron-specific Cdk4 deletion. These mice display increased sympathetic innervation and enhanced cold tolerance, similar to Cdk4-/- mice, in addition to browning of scWAT. Overall, we provide evidence showing that CDK4 modulates thermogenesis by regulating sympathetic innervation of adipose tissue depots through hypothalamic nuclei, including the VMH. This demonstrates that CDK4 not only negatively regulates oxidative pathways, but also modulates the central regulation of metabolism through its action in the brain.


Subject(s)
Adipose Tissue, White , Thermogenesis , Adipocytes, Brown , Adipose Tissue, Brown , Animals , Hypothalamus , Mice , Thermogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL