Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Sci Rep ; 14(1): 9157, 2024 04 22.
Article En | MEDLINE | ID: mdl-38644456

Brown adipose tissue (BAT) which is a critical regulator of energy homeostasis, and its activity is inhibited by obesity and low-grade chronic inflammation. Ginsenoside Rg3, the primary constituent of Korean red ginseng (steamed Panax ginseng CA Meyer), has shown therapeutic potential in combating inflammatory and metabolic diseases. However, it remains unclear whether Rg3 can protect against the suppression of browning or activation of BAT induced by inflammation. In this study, we conducted a screening of ginsenoside composition in red ginseng extract (RGE) and explored the anti-adipogenic effects of both RGE and Rg3. We observed that RGE (exist 0.25 mg/mL of Rg3) exhibited significant lipid-lowering effects in adipocytes during adipogenesis. Moreover, treatment with Rg3 (60 µM) led to the inhibition of triglyceride accumulation, subsequently promoting enhanced fatty acid oxidation, as evidenced by the conversion of radiolabeled 3H-fatty acids into 3H-H2O with mitochondrial activation. Rg3 alleviated the attenuation of browning in lipopolysaccharide (LPS)-treated beige adipocytes and primary brown adipocytes by recovered by uncoupling protein 1 (UCP1) and the oxygen consumption rate compared to the LPS-treated group. These protective effects of Rg3 on inflammation-induced inhibition of beige and BAT-derived thermogenesis were confirmed in vivo by treating with CL316,243 (a beta-adrenergic receptor agonist) and LPS to induce browning and inflammation, respectively. Consistent with the in vitro data, treatment with Rg3 (2.5 mg/kg, 8 weeks) effectively reversed the LPS-induced inhibition of brown adipocyte features in C57BL/6 mice. Our findings confirm that Rg3-rich foods are potential browning agents that counteract chronic inflammation and metabolic complications.


Adipose Tissue, Brown , Ginsenosides , Lipopolysaccharides , Mitochondria , Panax , Plant Extracts , Thermogenesis , Ginsenosides/pharmacology , Animals , Thermogenesis/drug effects , Panax/chemistry , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Plant Extracts/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/drug effects , Mice, Inbred C57BL , Male , Adipogenesis/drug effects
2.
Toxicology ; 463: 152972, 2021 11.
Article En | MEDLINE | ID: mdl-34606950

The effects of Endocrine Disrupting Chemicals (EDCs) on the current obesity epidemic is a growing field of interest. Numerous EDCs have shown the potential to alter energy metabolism, which may increase the risk of obesity, in part, through direct actions on adipose tissue. While white adipose tissue has historically been the primary focus of this work, evidence of the EDC-induced disruption of brown and beige adipose tissues continues to build. Both brown and beige fat are thermogenic adipose depots rich in mitochondria that dispense heat when activated. Due to these properties, brown and beige fat are implicated in metabolic diseases such as obesity, diabetes, and cachexia. This review delves into the current literature of different EDCs, including bisphenols, dioxins, air pollutants, phthalates, and phytochemicals. The possible implications that these EDCs have on thermogenic adipose tissues are covered. This review also introduces the possibility of using brown and beige fat as a therapeutic target organ by taking advantage of some of the properties of EDCs. Collectively, we provide a comprehensive discussion of the evidence of EDC disruption in white, brown, and beige fat and highlight gaps worthy of further exploration.


Adipose Tissue, Beige/drug effects , Adipose Tissue, Brown/drug effects , Endocrine Disruptors/pharmacology , Adipose Tissue, White/drug effects , Animals , Endocrine Disruptors/toxicity , Energy Metabolism/drug effects , Humans , Mitochondria/drug effects , Obesity/metabolism , Thermogenesis/drug effects
3.
Biochem Biophys Res Commun ; 558: 154-160, 2021 06 18.
Article En | MEDLINE | ID: mdl-33915329

Genistein, a naturally occurring phytoestrogen and a member of the large class of compounds known as isoflavones, exerts protective effects in several diseases. Recent studies indicate that genistein plays a critical role in controlling body weight, obesity-associated insulin resistance, and metabolic disorders, but its target organs in reversing obesity and related pathological conditions remain unclear. In this study, we showed that mice supplemented with 0.2% genistein in a high-fat diet for 12 weeks showed enhanced metabolic homeostasis, including reduced obesity, improved glucose uptake and insulin sensitivity, and alleviated hepatic steatosis. We also observed a beiging phenomenon in the white adipose tissue and reversal of brown adipose tissue whitening in these mice. These changes led to enhanced resistance to cold stress. Altogether, our data suggest that the improved metabolic profile in mice treated with genistein is likely a result of enhanced adipose tissue function.


Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Cold-Shock Response/drug effects , Cold-Shock Response/physiology , Genistein/pharmacology , Adipocytes, White/cytology , Adipocytes, White/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Body Weight/drug effects , Cell Enlargement/drug effects , Diet, High-Fat/adverse effects , Eating/drug effects , Energy Metabolism/drug effects , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Phytoestrogens/pharmacology , Protective Agents/pharmacology
4.
JCI Insight ; 6(6)2021 03 22.
Article En | MEDLINE | ID: mdl-33571166

BACKGROUNDBeige and brown adipose tissue (BAT) are associated with improved metabolic homeostasis. We recently reported that the ß3-adrenergic receptor agonist mirabegron induced beige adipose tissue in obese insulin-resistant subjects, and this was accompanied by improved glucose metabolism. Here we evaluated pioglitazone treatment with a combination pioglitazone and mirabegron treatment and compared these with previously published data evaluating mirabegron treatment alone. Both drugs were used at FDA-approved dosages.METHODSWe measured BAT by PET CT scans, measured beige adipose tissue by immunohistochemistry, and comprehensively characterized glucose and lipid homeostasis and insulin sensitivity by euglycemic clamp and oral glucose tolerance tests. Subcutaneous white adipose tissue, muscle fiber type composition and capillary density, lipotoxicity, and systemic inflammation were evaluated by immunohistochemistry, gene expression profiling, mass spectroscopy, and ELISAs.RESULTSTreatment with pioglitazone or the combination of pioglitazone and mirabegron increased beige adipose tissue protein marker expression and improved insulin sensitivity and glucose homeostasis, but neither treatment induced BAT in these obese subjects. When the magnitude of the responses to the treatments was evaluated, mirabegron was found to be the most effective at inducing beige adipose tissue. Although monotherapy with either mirabegron or pioglitazone induced adipose beiging, combination treatment resulted in less beiging than either alone. The 3 treatments also had different effects on muscle fiber type switching and capillary density.CONCLUSIONThe addition of pioglitazone to mirabegron treatment does not enhance beiging or increase BAT in obese insulin-resistant research participants.TRIAL REGISTRATIONClinicalTrials.gov NCT02919176.FUNDINGNIH DK112282 and P20GM103527 and Clinical and Translational Science Awards grant UL1TR001998.


Acetanilides/pharmacology , Adipose Tissue, Beige/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Pioglitazone/pharmacology , Thiazoles/pharmacology , Acetanilides/administration & dosage , Drug Synergism , Female , Glucose Tolerance Test , Humans , Hypoglycemic Agents/administration & dosage , Insulin Resistance , Male , Middle Aged , Obesity/metabolism , Pioglitazone/administration & dosage , Thiazoles/administration & dosage
5.
Biochem Biophys Res Commun ; 537: 109-117, 2021 01 22.
Article En | MEDLINE | ID: mdl-33388413

Dissipating energy by activating thermogenic adipose to combating obesity attracts many interests. Ski-interacting protein (Skip) has been known to play an important role in cell proliferation and differentiation, but whether it participates in energy metabolism is not known. Our previous study revealed that BTM-0512 could induce beige adipose formation, accompanying with up-regulation of Skip, but the role of Skip in metabolism was unknown. In this study, we mainly investigated whether Skip was involved in beige remodeling of subcutaneous white preadipocytes as well as in lipid metabolism of differentiated beige adipocytes. The results showed that in high fat diet-induced obesity mice, the protein levels of Skip in subcutaneous and visceral white adipose as well as in brown adipose were all down-regulated, especially in subcutaneous white adipose. Then we cultured subcutaneous adipose derived-stem cells (ADSCs) and found knock-down of Skip (siSkip) inhibited the expressions of thermogenic adipose specific genes including PRDM16 and UCP1 in both undifferentiated ADSCs and differentiated beige adipocytes, which could abolish the effects of BTM-0512 on beige remodeling. We further observed that siSkip affected multiple rate-limiting enzymes in lipid metabolism. The expressions of ACC, GPAT-1, HSL and ATGL were down-regulated, while CPT1α expression was up-regulated by siSkip. The expression of AMPK was also decreased by siSkip. In conclusion, our study demonstrated that Skip might play an important role in the beige remodeling of white adipocytes as well as lipid metabolism of beige adipose.


Adipose Tissue, Beige/metabolism , Lipid Metabolism , Phosphoric Monoester Hydrolases/metabolism , Sirtuin 1/metabolism , Stilbenes/pharmacology , Adipose Tissue, Beige/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Diet , Down-Regulation/drug effects , Down-Regulation/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice, Inbred C57BL , Obesity/genetics , Phosphoric Monoester Hydrolases/genetics , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Thermogenesis/drug effects , Thermogenesis/genetics , Uncoupling Protein 1/metabolism
6.
J Agric Food Chem ; 68(49): 14513-14522, 2020 Dec 09.
Article En | MEDLINE | ID: mdl-33231468

Obesity is an important health issue nowadays. 3'-Hydroxydaidzein (OHD) is a metabolite of daidzein (DAI) that can be found in fermented soybean products, such as miso. DAI has been known to affect lipid accumulation, but the effect of OHD on lipid accumulation still needs to be investigated. In this study, we investigated the effects of OHD on mice with obesity induced by a high-fat diet (HFD). The results showed that mice treated with 0.1% OHD (HOHD) significantly reduced their body weight and inguinal fat without altering their food intake compared with the HFD group. The HOHD and DAI groups' hyperlipidemia were alleviated through decreased serum triacylglycerols and total cholesterol levels. The adipocyte sizes in inguinal fat were significantly smaller in the HOHD and DAI groups compared with the HFD group. Both the HOHD and DAI groups had increased PRDM16, C/EBP ß, p-p38, SIRT1, PGC1 α, and UCP1 protein expression in their inguinal adipose tissue compared with the HFD group. Moreover, the OHD and DAI groups had significantly lower amounts of Lachnospira and GCA_900066225 compared with the HFD group. Collectively, OHD can ameliorate HFD-induced obesity in mice by stimulating the browning of the white adipose tissue and modulating gut microbiota.


Adipose Tissue, Beige/drug effects , Gastrointestinal Microbiome/drug effects , Isoflavones/administration & dosage , Obesity/drug therapy , Adipose Tissue, Beige/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Obesity/microbiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
7.
Pharm Res ; 37(10): 185, 2020 Sep 04.
Article En | MEDLINE | ID: mdl-32888109

PURPOSE: Inhibition of Notch signaling has been recently demonstrated to promote beige adipocyte biogenesis. However, most γ-secretase inhibitors (GSIs) used to achieve pharmacological inhibition of Notch signaling are at the basic research or preclinical stage, limiting the translation of fundamental findings into clinical practice. This present study aimed to evaluate the potential of several clinical candidates of GSIs as browning agents for the treatment of obesity. METHODS: Seven GSIs that are clinical candidates for the treatment of Alzheimer's disease or cancer were selected and their impacts on Notch inhibition as well as promoting beige biogenesis were compared using in vitro culture of 3T3-L1 preadipocytes. RESULTS: Four compounds (i.e.RO4929097, PF-03084014, LY3039478, and BMS-906024) that efficiently inhibited the expression of Notch target genes in 3T3-L1 preadipocytes were identified. Moreover, these compounds were optimized for dose-dependent effects at three gradient concentrations (0.5, 1, and 10 µM) to promote beige adipogenesis and mitochondrial biogenesis in 3T3-L1 preadipocytes without causing severe cytotoxicity. CONCLUSIONS: Our findings not only highlight the potential of cross-therapeutic application of these GSIs for obesity treatment via inhibition of γ-secretase-mediated processing of Notch signaling, but also provide important experimental evidence to support further design and development of clinically translatable Notch-inhibiting drug delivery systems.


Adipogenesis/drug effects , Adipose Tissue, Beige/drug effects , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Anti-Obesity Agents/pharmacology , Enzyme Inhibitors/pharmacology , Organelle Biogenesis , Receptors, Notch/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Anti-Obesity Agents/chemistry , Apoptosis/drug effects , Cell Proliferation , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Lipid Droplets/chemistry , Mice
8.
Cardiovasc Pathol ; 49: 107259, 2020.
Article En | MEDLINE | ID: mdl-32692664

Perivascular adipose tissue (PVAT) is a fat tissue deposit that encircles the vasculature. PVAT is traditionally known to protect the vasculature from external stimuli that could cause biological stress. In addition to the protective role of PVAT, it secretes certain biologically active substances known as adipokines that induce paracrine effects on proximate blood vessels. These adipokines influence vascular tones. There are different types of PVAT and they are phenotypically and functionally distinct. These are the white and brown PVATs. Under certain conditions, white PVAT could undergo phenotypic switch to attain a brown PVAT-like phenotype. This type of PVAT is referred to as Beige PVAT. The morphology of adipose tissue is influenced by species, age, and sex. These factors play significant roles in adipose tissue mass, functionality, paracrine activity, and predisposition to vascular diseases. The difficulty that is currently experienced in extrapolating animal models to human physiology could be traceable to these factors. Up till now, the involvement of PVAT in the development of vascular pathology is still not well understood. Brown and white PVAT contribute differently to vascular pathology. Thus, the PVAT could be a therapeutic target in curbing certain vascular diseases. In this review, knowledge would be updated on the multifaceted involvement of PVAT in vascular pathology and also explore its vascular therapeutic potential.


Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Arteries/pathology , Vascular Diseases/pathology , Adipokines/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/physiopathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiopathology , Adiposity , Animals , Arteries/drug effects , Arteries/metabolism , Arteries/physiopathology , Cardiovascular Agents/therapeutic use , Hemodynamics , Humans , Inflammation Mediators/metabolism , Paracrine Communication , Signal Transduction , Vascular Diseases/drug therapy , Vascular Diseases/metabolism , Vascular Diseases/physiopathology
9.
Clin Sci (Lond) ; 134(12): 1537-1553, 2020 06 26.
Article En | MEDLINE | ID: mdl-32556103

Hyperuricaemia (HUA) significantly increases the risk of metabolic syndrome and is strongly associated with the increased prevalence of high serum free fatty acids (FFAs) and insulin resistance. However, the underlying mechanisms are not well established, especially the effect of uric acid (UA) on adipose tissue, a vital organ in regulating whole-body energy and FFA homeostasis. In the present study, we noticed that adipocytes from the white adipose tissue of patients with HUA were hypertrophied and had decreased UCP1 expression. To test the effects of UA on adipose tissue, we built both in vitro and in vivo HUA models and elucidated that a high level of UA could induce hypertrophy of adipocytes, inhibit their hyperplasia and reduce their beige-like characteristics. According to mRNA-sequencing analysis, UA significantly decreased the expression of leptin in adipocytes, which was closely related to fatty acid metabolism and the AMPK signalling pathway, as indicated by KEGG pathway analysis. Moreover, lowering UA using benzbromarone (a uricosuric agent) or metformin-induced activation of AMPK expression significantly attenuated UA-induced FFA metabolism impairment and adipose beiging suppression, which subsequently alleviated serum FFA elevation and insulin resistance in HUA mice. Taken together, these observations confirm that UA is involved in the aetiology of metabolic abnormalities in adipose tissue by regulating leptin-AMPK pathway, and metformin could lessen HUA-induced serum FFA elevation and insulin resistance by improving adipose tissue function via AMPK activation. Therefore, metformin could represent a novel treatment strategy for HUA-related metabolic disorders.


Adipocytes/pathology , Adipose Tissue, Beige/pathology , Adipose Tissue, White/pathology , Fatty Acids, Nonesterified/blood , Hyperuricemia/blood , Hyperuricemia/drug therapy , Insulin Resistance , Metformin/therapeutic use , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, White/drug effects , Adult , Animals , Enzyme Activation , Female , Humans , Hypertrophy , Leptin/metabolism , Lipogenesis , Lipolysis , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction , Triglycerides/metabolism , Uric Acid/blood
10.
Article En | MEDLINE | ID: mdl-32351446

Brown fat and beige fat are known as thermogenic fat due to their contribution to non-shivering thermogenesis in mammals following cold stimulation. Beige fat is unique due to its origin and its development in white fat. Subsequently, both brown fat and beige fat have become viable targets to combat obesity. Over the last few decades, most therapeutic strategies have been focused on the canonical pathway of thermogenic fat activation via the ß3-adrenergic receptor (AR). Notwithstanding, administering ß3-AR agonists often leads to side effects including hypertension and particularly cardiovascular disease. It is thus imperative to search for alternative therapeutic approaches to combat obesity. In this review, we discuss the current challenges in the field with respect to stimulating brown/beige fat thermogenesis. Additionally, we include a summary of other newly discovered pathways, including non-AR signaling- and non-UCP1-dependent mechanisms, which could be potential targets for the treatment of obesity and its related metabolic diseases.


Adipose Tissue, Beige/physiology , Adipose Tissue, Brown/physiology , Obesity/therapy , Thermogenesis/physiology , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Agonists/therapeutic use , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Humans , Obesity/metabolism , Receptors, Adrenergic, beta-3/metabolism , Receptors, Adrenergic, beta-3/physiology , Signal Transduction/drug effects , Thermogenesis/drug effects
11.
Metabolism ; 103: 154048, 2020 02.
Article En | MEDLINE | ID: mdl-31843339

OBJECTIVE: Angiotensin-(1-7) [Ang-(1-7)], a component of the renin angiotensin system, is a vasodilator that exerts its effects primarily through the Mas receptor. The discovery of the Mas receptor in white adipose tissue (WAT) suggests an additional role for this peptide. The aim of the present study was to assess whether Ang-(1-7) can induce the expression of thermogenic genes in white adipose tissue and increase mitochondrial respiration in adipocytes. MATERIALS/METHODS: Stromal Vascular fraction (SVF)-derived from mice adipose tissue was stimulated for one week with Ang-(1-7), then expression of beige markers and mitochondrial respiration were assessed. Mas+/+ and Mas-/- mice fed a control diet or a high fat-sucrose diet (HFSD) were exposed to a short or long term infusion of Ang-(1-7) and body weight, body fat, energy expenditure, cold resistance and expression of beige markers were assessed. Also, transgenic rats overexpressing Ang-(1-7) were fed with a control diet or a high fat-sucrose diet and the same parameters were assessed. Ang-(1-7) circulating levels from human subjects with different body mass index (BMI) or age were measured. RESULTS: Incubation of adipocytes derived from SVF with Ang-(1-7) increased the expression of beige markers. Infusion of Ang-(1-7) into lean and obese Mas+/+mice also induced the expression of Ucp1 and some beige markers, an effect not observed in Mas-/- mice. Mas-/- mice had increased body weight gain and decreased cold resistance, whereas rats overexpressing Ang-(1-7) showed the opposite effects. Overexpressing rats exposed to cold developed new thermogenic WAT in the anterior interscapular area. Finally, in human subjects the higher the BMI, low circulating concentration of Ang-(1-7) levels were detected. Similarly, the circulating levels of Ang-(1-7) peptide were reduced with age. CONCLUSION: These data indicate that Ang-(1-7) stimulates beige markers and thermogenesis via the Mas receptor, and this evidence suggests a potential therapeutic use to induce thermogenesis of WAT, particularly in obese subjects that have reduced circulating concentration of Ang-(1-7).


Adipose Tissue, Beige/drug effects , Angiotensin I/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Thermogenesis/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adult , Animals , Cell Respiration/drug effects , Cell Respiration/genetics , Cells, Cultured , Energy Metabolism/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Rats , Rats, Transgenic , Receptors, G-Protein-Coupled/genetics , Thermogenesis/genetics , Young Adult
12.
Nutrients ; 11(9)2019 Sep 10.
Article En | MEDLINE | ID: mdl-31509935

The alteration of white adipose tissue (WAT) "browning", a change of white into beige fat, has been considered as a new therapeutic strategy to treat obesity. In this study, we investigated the browning effect of black raspberry (Rubus coreanus Miquel) using in vitro and in vivo models. Black raspberry water extract (BRWE) treatment inhibited lipid accumulation in human mesenchymal stem cells (hMSCs) and zebrafish. To evaluate the thermogenic activity, BRWE was orally administered for 2 weeks, and then, the mice were placed in a 4 °C environment. As a result, BRWE treatment increased rectal temperature and inguinal WAT (iWAT) thermogenesis by inducing the expression of beige fat specific markers such as PR domain zinc-finger protein 16 (PRDM16), uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), and t-box protein 1 (TBX1) in cold-exposed mice. Furthermore, ellagic acid (EA), a constituent of BRWE, markedly promoted beige specific markers: UCP1, PGC1α, TBX1, and nuclear respiratory factor 1 in beige differentiation media (DM)-induced 3T3-L1 adipocytes. Our findings indicate that BRWE can promote beige differentiation/activation, and EA is the active compound responsible for such effect. Thus, we suggest the nature-derived agents BRWE and EA as potential agents for obesity treatment.


Adipocytes, Beige/drug effects , Adipocytes, White/drug effects , Adipogenesis/drug effects , Adipose Tissue, Beige/drug effects , Adipose Tissue, White/drug effects , Anti-Obesity Agents/pharmacology , Plant Extracts/pharmacology , Thermogenesis/drug effects , 3T3-L1 Cells , Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Animals , Anti-Obesity Agents/isolation & purification , Cold Temperature , Gene Expression Regulation , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Plant Extracts/isolation & purification , Rubus/chemistry , Signal Transduction , Zebrafish
13.
Sci Rep ; 9(1): 9104, 2019 06 24.
Article En | MEDLINE | ID: mdl-31235722

Brown adipose tissue (BAT) is able to rapidly generate heat and metabolise macronutrients, such as glucose and lipids, through activation of mitochondrial uncoupling protein 1 (UCP1). Diet can modulate UCP1 function but the capacity of individual nutrients to promote the abundance and activity of UCP1 is not well established. Caffeine consumption has been associated with loss of body weight and increased energy expenditure, but whether it can activate UCP1 is unknown. This study examined the effect of caffeine on BAT thermogenesis in vitro and in vivo. Stem cell-derived adipocytes exposed to caffeine (1 mM) showed increased UCP1 protein abundance and cell metabolism with enhanced oxygen consumption and proton leak. These functional responses were associated with browning-like structural changes in mitochondrial and lipid droplet content. Caffeine also increased peroxisome proliferator-activated receptor gamma coactivator 1-alpha expression and mitochondrial biogenesis, together with a number of BAT selective and beige gene markers. In vivo, drinking coffee (but not water) stimulated the temperature of the supraclavicular region, which co-locates to the main region of BAT in adult humans, and is indicative of thermogenesis. Taken together, these results demonstrate that caffeine can promote BAT function at thermoneutrality and may have the potential to be used therapeutically in adult humans.


Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Caffeine/pharmacology , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Animals , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/cytology , Mice , Organelle Biogenesis , Temperature , Uncoupling Protein 1/genetics
14.
Cell Metab ; 30(1): 174-189.e5, 2019 07 02.
Article En | MEDLINE | ID: mdl-31155495

The precursor cells for metabolically beneficial beige adipocytes can alternatively become fibrogenic and contribute to adipose fibrosis. We found that cold exposure or ß3-adrenergic agonist treatment of mice decreased the fibrogenic profile of precursor cells and stimulated beige adipocyte differentiation. This fibrogenic-to-adipogenic transition was impaired in aged animals, correlating with reduced adipocyte expression of the transcription factor PRDM16. Genetic loss of Prdm16 mimicked the effect of aging in promoting fibrosis, whereas increasing PRDM16 in aged mice decreased fibrosis and restored beige adipose development. PRDM16-expressing adipose cells secreted the metabolite ß-hydroxybutyrate (BHB), which blocked precursor fibrogenesis and facilitated beige adipogenesis. BHB catabolism in precursor cells, mediated by BDH1, was required for beige fat differentiation in vivo. Finally, dietary BHB supplementation in aged animals reduced adipose fibrosis and promoted beige fat formation. Together, our results demonstrate that adipocytes secrete a metabolite signal that controls beige fat remodeling.


Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , 3-Hydroxybutyric Acid/pharmacology , Adipocytes/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Animals , Blotting, Western , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , In Vitro Techniques , Male , Mass Spectrometry , Mice , Transcription Factors/genetics
15.
J Nutr Biochem ; 68: 59-68, 2019 06.
Article En | MEDLINE | ID: mdl-31030168

White adipose tissue (WAT) can differentiate into beige adipose tissue by the browning process. Some polyphenols, including isoflavones, particularly genistein, are suggested to increase the expression of browning markers. There is evidence that consumption of genistein can attenuate body weight gain and improve glucose tolerance and blood lipid levels. The aim of the present study was to investigate the potential mechanisms of stimulation by which genistein activates the browning of WAT. We studied the stimulation of the expression of browning markers in the following models: mice fed genistein; preadipocytes from 3 T3-L1 cells; and the stromal vascular fraction (SVF) from the inguinal adipose tissue of mice. The results indicated that genistein can stimulate the browning process by at least two mechanisms. An indirect mechanism was involved in the induction of PGC-1α/FNDC5 in skeletal muscle leading to an increase in the myokine irisin. In preadipocytes, irisin was able to increase the expression of Ucp1 and Tmem26, markers of browning, to increase energy expenditure. Interestingly, genistein was also able to activate browning by a direct mechanism. Incubation of preadipocytes with genistein increased UCP1 expression as well as some biomarkers of browning in a concentration-dependent manner, possibly via phosphorylation of AMPK. The effect of genistein was accompanied by an increase in the number of mitochondria as well as in the maximum respiration rate of the adipocytes. In conclusion, this study indicated that genistein can increase energy expenditure by stimulating the browning process directly in preadipocytes and indirectly by increasing the circulating levels of irisin.


Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Energy Metabolism/drug effects , Genistein/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Beige/drug effects , Animals , Cell Differentiation/drug effects , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Thermogenesis/drug effects
16.
J Biol Chem ; 294(17): 6751-6761, 2019 04 26.
Article En | MEDLINE | ID: mdl-30824545

Beige fat is a potential therapeutic target for obesity and other metabolic diseases due to its inducible brown fat-like functions. Inguinal white adipose tissue (iWAT) can undergo robust brown remodeling with appropriate stimuli and is therefore widely considered as a representative beige fat depot. However, adipose tissues residing in different anatomic depots exhibit a broad range of plasticity, raising the possibility that better beige fat depots with greater plasticity may exist. Here we identified and characterized a novel, naturally-existing beige fat depot, thigh adipose tissue (tAT). Unlike classic WATs, tAT maintains beige fat morphology at room temperature, whereas high-fat diet (HFD) feeding or aging promotes the development of typical WAT features, namely unilocular adipocytes. The brown adipocyte gene expression in tAT is consistently higher than in iWAT under cold exposure, HFD feeding, and rosiglitazone treatment conditions. Our molecular profiling by RNA-Seq revealed up-regulation of energy expenditure pathways and repressed inflammation in tAT relative to eWAT and iWAT. Furthermore, we demonstrated that the master fatty acid oxidation regulator peroxisome proliferator-activated receptor α is dispensable for maintaining and activating the beige character of tAT. Therefore, we have identified tAT as a natural beige adipose depot in mice with a unique molecular profile that does not require peroxisome proliferator-activated receptor α.


Adipose Tissue, Beige/anatomy & histology , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Animals , Biomarkers/metabolism , Diet, High-Fat , Hyperplasia/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , PPAR alpha/metabolism , Sequence Analysis, RNA , Thiazolidinediones/pharmacology , Thigh , Transcriptome
17.
Food Funct ; 10(2): 836-848, 2019 Feb 20.
Article En | MEDLINE | ID: mdl-30681105

Obesity is a worldwide public health concern requiring safe and effective strategies. Recent studies suggest that bioactive compounds from soybeans have beneficial effects on weight loss and reducing fat accumulation. However, despite the biochemical and nutritional changes during germination, the biological effects of germinated soy germ have not been fully investigated. In this article, germinated soy germ extract (GSGE) was evaluated as a potential treatment option for obesity using 3T3-L1 pre-adipocyte and high-fat diet (HFD)-induced obese mice. In vitro studies demonstrated that GSGE suppressed the differentiation of 3T3-L1 cells into mature adipocytes, along with reductions in lipid accumulation and lipid droplet formation. In vivo studies also showed that a daily dose of 1 mg kg-1 of GSGE reduced weight gain, adipocyte area, serum triglyceride, and LDL-cholesterol in HFD-fed mice. The GSGE treatment promoted browning, which was associated with increased UCP1 expression in vitro and in vivo. In addition, GSGE treatment induced beige fat activation by upregulation of lipolysis and beta-oxidation. Furthermore, gene and protein expression levels of endocannabinoid system-related factors such as NAPE-PLD, FAAH, DAGL-α, and CB2 were altered along with browning and beige fat activation by GSGE. The present study indicates that GSGE effectively inhibits lipid accumulation and promotes beige fat transition and activation. Therefore, we suggest that GSGE treatment could be a promising strategy for the prevention of obesity by promoting weight loss, reducing fat accumulation, and improving obesity-related metabolic disorders.


Adipose Tissue, Beige/drug effects , Glycine max/chemistry , Obesity/prevention & control , Plant Extracts/pharmacology , Saponins/pharmacology , 3T3-L1 Cells , Adipose Tissue, Beige/physiology , Animals , Cell Survival , Diet, High-Fat/adverse effects , Female , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Plant Extracts/chemistry , Saponins/chemistry
18.
Int J Obes (Lond) ; 43(5): 1058-1069, 2019 05.
Article En | MEDLINE | ID: mdl-30018312

BACKGROUND/OBJECTIVES: The browning of white adipose tissue (WAT) has been in the spotlight during the last years, becoming an attractive approach to combat obesity. Melanocortin neuropeptides, such as α-melanocyte-stimulating hormone (α-MSH), are well-known regulators of appetite at the central nervous system, but its role in adipocyte metabolism is poorly elucidated. This study sought to verify if α-MSH can induce transdifferentiation of white to brown/beige adipocytes and to determine whether it can ameliorate the obesity phenotype. METHODS: The browning effect of α-MSH was determined in isolated adipocytes using the 3T3-L1 cell line and in inguinal subcutaneous adipose tissue (ingWAT) of diet-induced obese (DIO) mice by quantifying the expression of browning hallmark genes, oxygen consumption, and mitochondrial biogenesis. α-MSH protection from diet-induced obesity was evaluated by analyzing mice body weight, fat mass, and lipid and glucose serum profiles. RESULTS: Here, we report that α-MSH activates a thermogenic gene program and increases the mitochondrial respiratory rate in 3T3-L1 adipocytes and ingWAT of DIO mice. Without affecting food intake, peripheral administration of α-MSH decreases body weight and ingWAT mass, promoting a significant rise in the number of smaller adipocytes, whereas it lowered the larger ones. Additionally, there was an increase in the mass of brown adipose tissue. Browning activation occurs concomitantly with improvement on serum lipid profile, insulin resistance, and glucose homeostasis. CONCLUSIONS: This study highlights the anti-obesity properties of melanocortins by promoting ingWAT browning and provides new perspectives for future designing of more effective therapeutic strategies.


Adipocytes/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Melanocortins/pharmacology , Obesity/prevention & control , Thermogenesis/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Mice , Mice, Inbred C57BL , Obesity/metabolism , Thermogenesis/physiology
19.
J Physiol Biochem ; 75(1): 1-10, 2019 Feb.
Article En | MEDLINE | ID: mdl-30506389

Mammalian adipose tissue is traditionally categorized into white and brown relating to their function and morphology: while white serves as an energy storage, brown adipose tissue acts as the heat generator maintaining the core body temperature. The most recently identified type of fat, beige adipocyte tissue, resembles brown fat by morphology and function but is developmentally more related to white. The synthesis of beige fat, so-called browning of white fat, has developed into a topical issue in diabetes and metabolism research. This is due to its favorable effect on whole-body energy metabolism and the fact that it can be recruited during adult life. Indeed, brown and beige adipose tissues have been demonstrated to play a role in glucose homeostasis, insulin sensitivity, and lipid metabolism-all factors related to pathogenesis of type 2 diabetes. Many agents capable of initiating browning have been identified so far and tested widely in humans and animal models including in vitro and in vivo experiments. Interestingly, several agents demonstrated to have browning activity are in fact secreted as adipokines from brown and beige fat tissue, suggesting a physiological relevance both in beige adipocyte recruitment processes and in maintenance of metabolic homeostasis. The newest findings on agents driving beige fat recruitment, their mechanisms, and implications on type 2 diabetes are discussed in this review.


Adipose Tissue, Beige/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Diabetes Mellitus, Type 2/drug therapy , Lipotropic Agents/pharmacology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Glucagon-Like Peptide 1/pharmacology , Glucose/metabolism , Humans , Insulin Resistance , Leptin/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Melatonin/pharmacology , Natriuretic Peptides/pharmacology , Thermogenesis/drug effects , Thermogenesis/genetics , Tretinoin/pharmacology
20.
Endocrine ; 64(2): 271-283, 2019 05.
Article En | MEDLINE | ID: mdl-30535743

PURPOSE: Glucagon like peptide-1 (GLP-1) is produced to induce postprandial insulin secretion. Liraglutide, a full agonist of the GLP-1 receptor, has a protective effect on weight gain in obese subjects. Brown adipose tissue plays a major role in the control of energy balance and is known to be involved in the weight loss regulated by liraglutide. The putative anti-obesity properties of liraglutide and the cell signaling pathways involved were examined. METHODS: Four groups of C57/BL6 mice fed with chow or HFHS diet were injected with either liraglutide or vehicle for four weeks. Western blotting was used to analyze protein expression. RESULTS: Liraglutide significantly attenuated the weight gain in mice fed with HFHS diet and was associated with significant reductions of epididymal fat and inguinal fat mass. Furthermore, liraglutide significantly upregulated the expression of brown adipose-specific markers in perigonadal fat in association with upregulation of AMPK-SIRT-1-PGC1-α cell signaling. However, elevation of brown fat markers in skeletal muscle was only observed in HFHS diet fed mice after liraglutide treatment, and AMPK-SIRT-1 cell signaling is not involved in this process. CONCLUSIONS: the anti-obesity effect of liraglutide occurs through adaptive thermogenesis and may act through different cell signaling pathways in fat and skeletal muscle tissue. Liraglutide induces beige fat development partially through the AMPK-SIRT-1-PGC1-α cell signaling pathway. Therefore, liraglutide is a potential medication for obesity prevention and in targeting pre-diabetics.


Adipose Tissue, Beige/drug effects , Hypoglycemic Agents/pharmacology , Liraglutide/pharmacology , Mitochondria/drug effects , Obesity/metabolism , Signal Transduction/drug effects , Adenylate Kinase/metabolism , Adipose Tissue, Beige/metabolism , Adiposity/drug effects , Animals , Diet, High-Fat , Lipogenesis/drug effects , Male , Mice , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/metabolism
...