Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2662: 157-166, 2023.
Article in English | MEDLINE | ID: mdl-37076679

ABSTRACT

Excessive fat accumulation is a risk factor for metabolic diseases. Activating non-shivering thermogenesis in adipose tissue increases energy expenditure and potentially reverses obesity-related metabolic dysfunctions. While brown/beige adipocytes specialize in non-shivering thermogenesis and catabolic lipid metabolism, thermogenic stimuli and pharmacological intervention can induce the recruitment and metabolic activation of these cell types in adipose tissue. Thus, these adipocytes are attractive therapeutic targets to combat obesity, and there is an increasing need for efficient screening strategies for thermogenic drugs. Cell death-inducing DNA fragmentation factor-like effector A (CIDEA) is a well-known marker of the thermogenic capacity of brown and beige adipocytes. We recently developed a CIDEA reporter mouse model that expresses multicistronic mRNAs encoding CIDEA, luciferase 2, and tdTomato proteins under endogenous Cidea promoter control. Here, we introduce the CIDEA reporter model system as a tool for in vitro and in vivo screening of drug candidate molecules with thermogenic effects and provide a detailed protocol to monitor CIDEA reporter expression.


Subject(s)
Adipocytes, Brown , Adipose Tissue , Mice , Animals , Adipose Tissue/metabolism , Adipocytes, Brown/metabolism , Proteins/metabolism , Obesity/metabolism , Thermogenesis/genetics , Apoptosis Regulatory Proteins/metabolism
2.
J Nutr Biochem ; 111: 109173, 2023 01.
Article in English | MEDLINE | ID: mdl-36228975

ABSTRACT

The antidiabetic effects of green tea have been demonstrated in clinical trials and epidemiological studies. This study investigated the antidiabetic effects of green tea extract (GTE) and its underlying molecular mechanisms using a leptin receptor-deficient db/db mouse model (Leprdb/db). Treatment with GTE for 2 weeks improved glucose tolerance and insulin sensitivity in Leprdb/db mice. In addition, GTE treatment reduced the body weight and adiposity of Leprdb/db mice. Furthermore, GTE treatment reduced pro-inflammatory gene expression, including nuclear factor kappa B (NF-κB) in white adipose tissue (WAT), and also reduced dipeptidyl peptidase-4 (DPP4) expression levels in WAT as well as in the serum. The promoter region of Dpp4 contains the NF-κB binding site, and DPP4 was found to be a direct target of NF-κB. Consistently, in vitro treatment of cells with GTE or its main constituent epigallocatechin gallate reduced lipopolysaccharide-induced NF-κB/DPP4 expression in 3T3-L1 adipocytes and RAW264.7 cells. Overall, our data demonstrated that GTE exerts an anti-diabetic effect by regulating the expression levels of NF-κB and DPP4 in WAT.


Subject(s)
Dipeptidyl Peptidase 4 , Hypoglycemic Agents , Mice , Animals , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/metabolism , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , NF-kappa B/metabolism , Plant Extracts/therapeutic use , Adipose Tissue/metabolism , Tea/chemistry
3.
Int J Obes (Lond) ; 46(12): 2128-2136, 2022 12.
Article in English | MEDLINE | ID: mdl-36167764

ABSTRACT

BACKGROUND AND OBJECTIVES: A2A adenosine receptor (A2AAR)-mediated signaling in adipose tissues has been investigated as a potential target for obesity-related metabolic diseases. LJ-4378 has been developed as a dual-acting ligand with A2AAR agonist and A3 adenosine receptor (A3AR) antagonist activity. The current study aimed to investigate the anti-obesity effects of LJ-4378 and its underlying molecular mechanisms. METHODS: Immortalized brown adipocytes were used for in vitro analysis. A high-fat diet (HFD)-induced obesity and cell death-inducing DFFA-like effector A reporter mouse models were used for in vivo experiments. The effects of LJ-4378 on lipolysis and mitochondrial metabolism were evaluated using immunoblotting, mitochondrial staining, and oxygen consumption rate analyses. The in vivo anti-obesity effects of LJ-4378 were evaluated using indirect calorimetry, body composition analyses, glucose tolerance tests, and histochemical analyses. RESULTS: In vitro LJ-4378 treatment increased the levels of brown adipocyte markers and mitochondrial proteins, including uncoupling protein 1. The effects of LJ-4378 on lipolysis of adipocytes were more potent than those of the A2AAR agonist or A3AR antagonist. In vivo, LJ-4378 treatment increased energy expenditure by 17.0% (P value < 0.0001) compared to vehicle controls. LJ-4378 (1 mg/kg, i.p.) treatment for 10 days reduced body weight and fat content by 8.24% (P value < 0.0001) and 24.2% (P value = 0.0044), respectively, and improved glucose tolerance in the HFD-fed mice. LJ-4378 increased the expression levels of brown adipocyte markers and mitochondrial proteins in interscapular brown and inguinal white adipose tissue. CONCLUSION: These findings support the in vivo anti-obesity effects of LJ-4378, and suggest a novel therapeutic approach to combat obesity and related metabolic diseases.


Subject(s)
Adenosine , Metabolic Diseases , Animals , Mice , Adenosine/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Diet, High-Fat , Ligands , Metabolic Diseases/metabolism , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Uncoupling Protein 1/metabolism , Receptors, Purinergic P1/metabolism
4.
Nat Commun ; 13(1): 4084, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835749

ABSTRACT

Dysregulation of adipose tissue plasmalogen metabolism is associated with obesity-related metabolic diseases. We report that feeding mice a high-fat diet reduces adipose tissue lysoplasmalogen levels and increases transmembrane protein 86 A (TMEM86A), a putative lysoplasmalogenase. Untargeted lipidomic analysis demonstrates that adipocyte-specific TMEM86A-knockout (AKO) increases lysoplasmalogen content in adipose tissue, including plasmenyl lysophosphatidylethanolamine 18:0 (LPE P-18:0). Surprisingly, TMEM86A AKO increases protein kinase A signalling pathways owing to inhibition of phosphodiesterase 3B and elevation of cyclic adenosine monophosphate. TMEM86A AKO upregulates mitochondrial oxidative metabolism, elevates energy expenditure, and protects mice from metabolic dysfunction induced by high-fat feeding. Importantly, the effects of TMEM86A AKO are largely reproduced in vitro and in vivo by LPE P-18:0 supplementation. LPE P-18:0 levels are significantly lower in adipose tissue of human patients with obesity, suggesting that TMEM86A inhibition or lysoplasmalogen supplementation might be therapeutic approaches for preventing or treating obesity-related metabolic diseases.


Subject(s)
Plasmalogens , Thermogenesis , Adipocytes/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/physiology , Homeostasis , Humans , Hydrolases , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Plasmalogens/metabolism , Thermogenesis/physiology
5.
Nutr Metab (Lond) ; 19(1): 14, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35241108

ABSTRACT

BACKGROUND: Adipose tissue thermogenesis is a potential therapeutic target to increase energy expenditure and thereby combat obesity. The aim of the present study was to investigate the thermogenic and anti-obesity effects of heat-transformed green tea extract (HTGT) and enzymatically modified isoquercetin (EMIQ). METHODS: Immortalized brown pre-adipocytes and C3H10T1/2 cells were used for in vitro analyses. A high-fat diet (HFD)-induced obesity mouse model and CIDEA-reporter mice were used for in vivo experiments. The effects of HTGT and EMIQ on mitochondrial metabolism were evaluated by immunoblot, mitochondrial staining, and oxygen consumption rate analyses. In vivo anti-obesity effects of HTGT and EMIQ were measured using indirect calorimetry, body composition analyses, glucose tolerance tests, and histochemical analyses. RESULTS: Co-treatment with HTGT and EMIQ (50 µg/mL each) for 48 h increased brown adipocyte marker and mitochondrial protein levels (UCP1 and COXIV) in brown adipocytes by 2.9-fold, while the maximal and basal oxygen consumption rates increased by 1.57- and 1.39-fold, respectively. Consistently, HTGT and EMIQ treatment increased the fluorescence intensity of mitochondrial staining in C3H10T1/2 adipocytes by 1.68-fold. The combination of HTGT and EMIQ (100 mg/kg each) increased the expression levels of brown adipocyte markers and mitochondrial proteins in adipose tissue. Two weeks of HTGT and EMIQ treatment (100 mg/kg each) led to a loss of 3% body weight and 7.09% of body fat. Furthermore, the treatment increased energy expenditure by 8.95% and improved glucose tolerance in HFD-fed mice. CONCLUSIONS: The current study demonstrated that HTGT and EMIQ have in vivo anti-obesity effects partly by increasing mitochondrial metabolism in adipocytes. Our findings suggest that a combination of HTGT and EMIQ is a promising therapeutic agent for the treatment of obesity and related metabolic diseases.

6.
Metabolism ; 130: 155159, 2022 05.
Article in English | MEDLINE | ID: mdl-35150731

ABSTRACT

INTRODUCTION: The mobilization and catabolism of lipid energy is a central function of adipocytes that is under the control of the ß-adrenergic signaling pathway, and defects in ß-adrenergic signaling in adipocytes have been linked to obesity and obesity-related metabolic diseases. Receptor expression-enhancing proteins (REEPs) are endoplasmic reticulum (ER) proteins that play critical roles in subcellular targeting of receptor signaling complexes. Examination of gene expression profiles indicates that, among REEPs expressed in adipocytes, REEP6 expression is uniquely upregulated by sympathetic nervous system activation, suggesting involvement in regulating adrenergic signal transduction. OBJECTIVE: The aim of this study was to assess the contribution of REEP6 to the thermogenic activation of adipocytes and characterize the metabolic consequences of REEP6 deficiency in vivo. METHODS: Expression levels of Reep6 in adipose tissue were examined by using public transcriptomic data and validated by Western blot and qPCR analyses. Adipocyte-specific regulatory roles of REEP6 were investigated in vitro in C3H10T1/2 adipocytes and in primary adipocytes obtained from REEP6 KO mice. Effects of in vivo REEP6 deficiency on energy expenditure were measured by indirect calorimetry. Mitochondrial content in adipose tissue was accessed by immunoblot, mitochondrial DNA analysis, and confocal and electron microscopy. Effects of REEP6 KO on obesity-induced metabolic dysfunction were tested in a high-fat diet-induced obesity mouse model by glucose tolerance test, Western blot, and histological analyses. RESULTS: REEP6 expression is highly enriched in murine adipocytes and is sharply upregulated upon adipocyte differentiation and by cold exposure. Inactivation of REEP6 in mice increased adiposity, and reduced energy expenditure and cold tolerance. REEP6 KO severely reduced protein kinase A-mediated signaling in BAT and greatly reduced mitochondrial mass. The effect of REEP6 inactivation on diminished ß-adrenergic signaling was reproduced in cultured adipocytes, indicating that this effect is cell-autonomous. REEP6 KO also suppressed expression of adenylate cyclase 3 (Adcy3) in brown adipose tissue and knockdown of REEP6 in adipocytes reduced targeting of ADCY3 to the plasma membrane. Lastly, REEP6 KO exacerbated high-fat diet-induced insulin resistance and inflammation in adipose tissue. CONCLUSIONS: This study indicates that REEP6 plays an important role in ß-adrenergic signal transduction in adipocytes involving the expression and trafficking of Adcy3. Genetic inactivation of REEP6 reduces energy expenditure, increases adiposity, and the susceptibility to obesity-related metabolic dysfunction.


Subject(s)
Adipocytes , Adrenergic Agents , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adrenergic Agents/metabolism , Animals , Diet, High-Fat , Energy Metabolism/genetics , Eye Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Signal Transduction , Thermogenesis/genetics
7.
Sci Rep ; 11(1): 18429, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531447

ABSTRACT

Cell death-inducing DNA fragmentation factor-like effector A (CIDEA) is a lipid droplet-associated protein and is a known marker of the thermogenic capacity of brown/beige adipocytes. To monitor the expression of CIDEA in live mice in a non-invasive manner, we generated CIDEA reporter mice expressing multicistronic mRNAs encoding CIDEA, luciferase 2, and tdTomato proteins under the control of the Cidea promoter. The expression level of endogenous CIDEA protein in adipose tissue was not affected by the expression of polycistronic reporters. The two CIDEA reporters, luciferase 2 and tdTomato, correctly reflected CIDEA protein levels. Importantly, luciferase activity was induced by cold exposure and the treatment with ß3-adrenergic receptor agonist CL316,243 in interscapular and inguinal adipose tissue, which was detectable by in vivo bioluminescence imaging. We further evaluated the effects of candidate brown adipogenic agents using this CIDEA reporter system and demonstrated a positive correlation between drug-induced luciferase activity and thermogenic gene expression levels both in vitro and in vivo. Collectively, we established a dual CIDEA reporter mouse model in which fluorescence and luminescence signals correctly reflect CIDEA expression, and therefore, suggested that this reporter system can be used to evaluate the thermogenic efficacy of candidate molecules.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Drug Discovery/methods , Genetic Engineering/methods , Thermogenesis/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL
8.
Diabetes ; 70(9): 2000-2013, 2021 09.
Article in English | MEDLINE | ID: mdl-34187836

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) functions as a sensor for lysosomal arginine levels and activates the mammalian target of rapamycin complex 1 (mTORC1). While the mTORC1 signaling pathway plays a key role in adipose tissue metabolism, the regulatory function of TM4SF5 in adipocytes remains unclear. In this study we aimed to establish a TM4SF5 knockout (KO) mouse model and investigated the effects of TM4SF5 KO on mTORC1 signaling-mediated autophagy and mitochondrial metabolism in adipose tissue. TM4SF5 expression was higher in inguinal white adipose tissue (iWAT) than in brown adipose tissue and significantly upregulated by a high-fat diet (HFD). TM4SF5 KO reduced mTORC1 activation and enhanced autophagy and lipolysis in adipocytes. RNA sequencing analysis of TM4SF5 KO mouse iWAT showed that the expression of genes involved in peroxisome proliferator-activated receptor α signaling pathways and mitochondrial oxidative metabolism was upregulated. Consequently, TM4SF5 KO reduced adiposity and increased energy expenditure and mitochondrial oxidative metabolism. TM4SF5 KO prevented HFD-induced glucose intolerance and inflammation in adipose tissue. Collectively, the results of our study demonstrate that TM4SF5 regulates autophagy and lipid catabolism in adipose tissue and suggest that TM4SF5 could be therapeutically targeted for the treatment of obesity-related metabolic diseases.


Subject(s)
Adipose Tissue/metabolism , Autophagy/genetics , Membrane Proteins/genetics , Obesity/genetics , Animals , Diet, High-Fat , Energy Metabolism/genetics , Female , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Obesity/metabolism , Signal Transduction/genetics
9.
Nat Metab ; 3(3): 428-441, 2021 03.
Article in English | MEDLINE | ID: mdl-33758424

ABSTRACT

Obesity reduces adipocyte mitochondrial function, and expanding adipocyte oxidative capacity is an emerging strategy to improve systemic metabolism. Here, we report that serine/threonine-protein kinase 3 (STK3) and STK4 are key physiological suppressors of mitochondrial capacity in brown, beige and white adipose tissues. Levels of STK3 and STK4, kinases in the Hippo signalling pathway, are greater in white than brown adipose tissues, and levels in brown adipose tissue are suppressed by cold exposure and greatly elevated by surgical denervation. Genetic inactivation of Stk3 and Stk4 increases mitochondrial mass and function, stabilizes uncoupling protein 1 in beige adipose tissue and confers resistance to metabolic dysfunction induced by high-fat diet feeding. Mechanistically, STK3 and STK4 increase adipocyte mitophagy in part by regulating the phosphorylation and dimerization status of the mitophagy receptor BNIP3. STK3 and STK4 expression levels are elevated in human obesity, and pharmacological inhibition improves metabolic profiles in a mouse model of obesity, suggesting STK3 and STK4 as potential targets for treating obesity-related diseases.


Subject(s)
Adipocytes/metabolism , Energy Metabolism , Mitophagy , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Obesity/prevention & control , Obesity/therapy , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3
10.
Int J Obes (Lond) ; 45(1): 122-129, 2021 01.
Article in English | MEDLINE | ID: mdl-32467614

ABSTRACT

BACKGROUND/OBJECTIVES: Polymethoxyselenoflavone (PMSF) is a compound that substitutes the oxygen atom in a flavonoid with selenium. This study aimed to investigate the effects of PMSFs on lipid metabolism in adipocytes and their anti-obesity potential. SUBJECTS/METHODS: To test lipolytic and thermogenic effects of the compounds in vitro, adipocytes differentiated from immortalized pre-brown adipocyte progenitors and pre-white adipocyte cell lines were treated with 19 PMSFs. The expression levels of brown adipocyte markers and genes related to mitochondrial metabolism were analyzed by qPCR and western blot. In vivo anti-obesity effect was investigated using diet-induced obesity mouse models and adipocyte-specific ATGL knockout mice. RESULTS: The qPCR analysis identified 2-(3,4-dimethoxyphenyl)-4H-selenochromen-4-one (DMPSC) as the most potent brown adipogenic candidate among the 19 compounds tested in this study. DMPSC treatment significantly increased the mitochondrial content and oxidative metabolism in adipocytes in vitro. Mechanistically, DMPSC treatment increased lipolysis through activation of PKA downstream signaling. Consistently, the in vivo treatment of DMPSC increased energy consumption, reduced body weight, and improved glucose tolerance in mice fed with high-fat diets. Moreover, DMPSC treatment increased brown adipocyte marker expression and mitochondrial content in adipose tissue of mice. The anti-obesity effects were absent in adipocyte-specific ATGL knockout mice, indicating that the DMPSC effect is mediated by cytosolic lipase-dependent mechanisms. CONCLUSIONS: Collectively, our results indicated that DMPSC exerted anti-obesity effects partially through the PKA signaling-mediated activation of lipolysis and brown adipose tissue metabolism.


Subject(s)
Adipocytes, Brown/drug effects , Anti-Obesity Agents/pharmacology , Flavonoids/pharmacology , Lipolysis/drug effects , Selenium Compounds/pharmacology , 3T3-L1 Cells , Adipocytes, Brown/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Knockout , Obesity/metabolism
11.
Biomolecules ; 10(10)2020 09 30.
Article in English | MEDLINE | ID: mdl-33008006

ABSTRACT

Soy isoflavones are bioactive phytoestrogens with known health benefits. Soybean embryo extract (SEE) has been consumed as a source of isoflavones, mainly daidzein, glycitein, and genistein. While previous studies have reported the anti-obesity effects of SEE, this study investigates their molecular mechanisms and the synergistic effects of co-treatment with SEE and enzymatically modified isoquercitrin (EMIQ). SEE upregulated genes involved in lipolysis and brown adipocyte markers and increased mitochondrial content in differentiated C3H10T1/2 adipocytes in vitro. Next, we use a high-fat diet-induced obesity mouse model to determine the anti-obesity effect of SEE. Two weeks of single or combined treatment with SEE and EMIQ significantly reduced body weight gain and improved glucose tolerance. Mechanistically, SEE treatment increased mitochondrial content and upregulated genes involved in lipolysis in adipose tissue through the cAMP/PKA-dependent signaling pathway. These effects required a cytosolic lipase adipose triglyceride lipase (ATGL) expression, confirmed by an adipocyte-specific ATGL knockout mouse study. Collectively, this study demonstrates that SEE exerts anti-obesity effects through the activation of adipose tissue metabolism and exhibits a synergistic effect of co-treatment with EMIQ. These results improve our understanding of the mechanisms underlying the anti-obesity effects of SEE related to adipose tissue metabolism.


Subject(s)
Glycine max/chemistry , Lipolysis/drug effects , Obesity/drug therapy , Quercetin/analogs & derivatives , Adipocytes/drug effects , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Cell Differentiation/drug effects , Diet, High-Fat/adverse effects , Genistein/chemistry , Genistein/pharmacology , Humans , Isoflavones/chemistry , Isoflavones/pharmacology , Mice , Obesity/etiology , Obesity/genetics , Obesity/pathology , Phytoestrogens/chemistry , Phytoestrogens/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Quercetin/chemistry , Quercetin/pharmacology , Seeds/chemistry
12.
Nutrients ; 12(10)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33050029

ABSTRACT

Epigallocatechin-3-gallate (EGCG) is a primary bioactive phytochemical in green tea. Its therapeutic potential in metabolic diseases has been reported; however, the molecular mechanisms of the anti-obesity effect of EGCG have not been fully elucidated. In this study, we examined the effects of EGCG on lipid metabolism and autophagy in adipose tissue. After 8 weeks of high-fat diet feeding, mice were treated with EGCG (20 mg/kg/day) for 2 weeks to test in vivo anti-obesity effects of EGCG. EGCG treatment improved glucose tolerance and caused body weight loss. Interestingly, reduced adipose tissue mass was more prominent in visceral compared to subcutaneous white adipose tissue. Mechanistically, EGCG treatment increased autophagic flux in white adipose tissue through the AMP-activated protein kinase-mediated signaling pathway. Adipocyte-specific knockout of Beclin1 mitigated the effects of EGCG on visceral adipose tissue mass and glucose tolerance, indicating that the anti-obesity effect of EGCG requires Beclin1-dependent autophagy. Collectively, our data demonstrated that EGCG has anti-obesity effects through the upregulation of Beclin1-dependent autophagy and lipid catabolism in white adipose tissue (WAT).


Subject(s)
Adipose Tissue, White/metabolism , Adiposity/drug effects , Adiposity/genetics , Autophagy/genetics , Beclin-1/physiology , Catechin/analogs & derivatives , Intra-Abdominal Fat/metabolism , Obesity/genetics , Animals , Autophagy/physiology , Beclin-1/metabolism , Catechin/administration & dosage , Catechin/pharmacology , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression Regulation, Developmental/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism
13.
Aging Dis ; 11(3): 575-587, 2020 May.
Article in English | MEDLINE | ID: mdl-32489703

ABSTRACT

Aging-related adipose tissue dysfunction contributes to the progression of chronic metabolic diseases. We investigated the role of age-dependent expression of a neurotrophin, brain-derived neurotrophic factor (BDNF) in adipose tissue. Pro-BDNF expression was elevated in epididymal white adipose tissue (eWAT) with advanced age, which was associated with the reduction in sympathetic innervation. Interestingly, BDNF expression was enriched in PDGFRα+ adipocyte progenitors isolated from eWAT, with age-dependent increase in expression. In vitro pro-BDNF treatment caused apoptosis in adipocytes differentiated from C3H10T1/2 cells, and siRNA knockdown of sortilin mitigated these effects. Tamoxifen-inducible PDGFRα+ cell-specific deletion of BDNF (BDNFPdgfra KO) reduced pro-BDNF expression in eWAT, prevented age-associated declines in sympathetic innervation and mitochondrial content in eWAT, and improved insulin sensitivity. Moreover, BDNFPdgfra KO mice showed reduced expression of aging-induced inflammation and senescence markers in eWAT. Collectively, these results identified the upregulation of pro-BDNF expression in adipocyte progenitors as a feature of visceral white adipose tissue aging and suggested that inhibition of BDNF expression in adipocyte progenitors is potentially beneficial to prevent aging-related adipose tissue dysfunction.

14.
Mol Metab ; 39: 101005, 2020 09.
Article in English | MEDLINE | ID: mdl-32344065

ABSTRACT

OBJECTIVE: Beclin1 is a core molecule of the macroautophagy machinery. Although dysregulation of macroautophagy is known to be involved in metabolic disorders, the function of Beclin1 in adipocyte metabolism has not been investigated. In the present study, we aimed to study the role of Beclin1 in lipolysis and mitochondrial homeostasis of adipocytes. METHODS: Autophagic flux during lipolysis was examined in adipocytes cultured in vitro and in the adipose tissue of mice. Adipocyte-specific Beclin1 knockout (KO) mice were used to investigate the activities of Beclin1 in adipose tissues. RESULTS: cAMP/PKA signaling increased the autophagic flux in adipocytes differentiated from C3H10T1/2 cells. In vivo autophagic flux was higher in the brown adipose tissue (BAT) than that in the white adipose tissue and was further increased by the ß3 adrenergic receptor agonist CL316243. In addition, surgical denervation of BAT greatly reduced autophagic flux, indicating that sympathetic nerve activity is a major regulator of tissue autophagy. Adipocyte-specific KO of Beclin1 led to a hypertrophic enlargement of lipid droplets in BAT and impaired CL316243-induced lipolysis/lipid mobilization and energy expenditure. While short-term effects of Beclin1 deletion were characterized by an increase in mitochondrial proteins, long-term Beclin1 deletion led to severe disruption of autophagy, resulting in mitochondrial loss, and dramatically reduced the expression of genes involved in lipid metabolism. Consequently, adipose tissue underwent increased activation of cell death signaling pathways, macrophage recruitment, and inflammation, particularly in BAT. CONCLUSIONS: The present study demonstrates the critical roles of Beclin1 in the maintenance of lipid metabolism and mitochondrial homeostasis in adipose tissues.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Beclin-1/genetics , Gene Deletion , Lipolysis/genetics , Mitochondria/genetics , Mitochondria/metabolism , Adipocytes/ultrastructure , Adipose Tissue, Brown/metabolism , Animals , Autophagy/genetics , Beclin-1/metabolism , Cell Line , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Copy Number Variations , Immunity , Lipid Metabolism , Mice , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Thermogenesis/genetics
15.
J Nutr Biochem ; 76: 108300, 2020 02.
Article in English | MEDLINE | ID: mdl-31812908

ABSTRACT

Coumestrol is a dietary phytoestrogen with estrogen-mimicking characteristics. This study investigated the molecular mechanisms of antiobesity effects of coumestrol. Two weeks of coumestrol treatment reduced body weight and improved glucose tolerance of high-fat diet (HFD)-fed mice. Notably, coumestrol treatment reduced adiposity but expanded brown adipose tissue mass. In addition, coumestrol treatment induced up-regulation of brown adipocyte markers and lipolytic gene expression in adipose tissue. Mechanistically, coumestrol induced an increase in mitochondrial contents of brown adipose tissue, which was associated with up-regulation of adenosine monophosphate-activated protein kinase and sirtuin 1. In vitro knockdown of estrogen receptor 1 inhibited the effect of coumestrol on brown adipogenic marker expression, increase in mitochondrial contents and oxygen consumption rate in brown adipocytes. Furthermore, lineage tracing of platelet-derived growth factor receptor A-positive (PDGFRA+) adipocyte progenitors confirmed increased levels of de novo brown adipogenesis from PDGFRA+ cells by coumestrol treatment. In conclusion, our results indicate that coumestrol has antiobesity effects through the expansion and activation of brown adipose tissue metabolism.


Subject(s)
Adipose Tissue, Brown/metabolism , Coumestrol/pharmacology , Obesity/drug therapy , Obesity/metabolism , Adipocytes, Beige/drug effects , Adipogenesis , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Weight , Diet, High-Fat , Glucose Tolerance Test , Lipolysis , Male , Mice , Mice, Inbred C57BL , Phytoestrogens/pharmacology
16.
Mol Metab ; 29: 86-98, 2019 11.
Article in English | MEDLINE | ID: mdl-31668395

ABSTRACT

OBJECTIVE: This study investigated the role of microRNAs generated from adipose tissue macrophages (ATMs) during adipose tissue remodeling induced by pharmacological and nutritional stimuli. METHODS: Macrophage-specific Dicer knockout (KO) mice were used to determine the roles of microRNA generated in macrophages in adipose tissue remodeling induced by the ß3-adrenergic receptor agonist CL316,243 (CL). RNA-seq was performed to characterize microRNA and mRNA expression profiles in isolated macrophages and PDGFRα+ adipocyte stem cells (ASCs). The role of miR-10a-5p was further investigated in cell culture, and in adipose tissue remodeling induced by CL treatment and high fat feeding. RESULTS: Macrophage-specific deletion of Dicer elevated pro-inflammatory gene expression and prevented CL-induced de novo beige adipogenesis in gonadal white adipose tissue (gWAT). Co-culture of ASCs with ATMs of wild type mice promoted brown adipocyte gene expression upon differentiation, but co-culture with ATMs of Dicer KO mice did not. Bioinformatic analysis of RNA expression profiles identified miR-10a-5p as a potential regulator of inflammation and differentiation in ATMs and ASCs, respectively. CL treatment increased levels of miR-10a-5p in ATMs and ASCs in gWAT. Interestingly, CL treatment elevated levels of pre-mir-10a in ATMs but not in ASCs, suggesting possible transfer from ATMs to ASCs. Elevating miR-10a-5p levels inhibited proinflammatory gene expression in cultured RAW 264.7 macrophages and promoted the differentiation of C3H10T1/2 cells into brown adipocytes. Furthermore, treatment with a miR-10a-5p mimic in vivo rescued CL-induced beige adipogenesis in Dicer KO mice. High fat feeding reduced miR-10a-5p levels in ATMs of gWAT, and treatment of mice with a miR-10a-5p mimic suppressed pro-inflammatory responses, promoted the appearance of new white adipocytes in gWAT, and improved systemic glucose tolerance. CONCLUSIONS: These results demonstrate an important role of macrophage-generated microRNAs in adipogenic niches and identify miR-10a-5p as a key regulator that reduces adipose tissue inflammation and promotes therapeutic adipogenesis.


Subject(s)
Adipose Tissue, White/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Adipocytes, Brown/metabolism , Adipogenesis , Animals , Antagomirs/metabolism , Cell Differentiation , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Dioxoles/pharmacology , Down-Regulation/drug effects , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , RAW 264.7 Cells , Receptors, Adrenergic, beta-3/chemistry , Receptors, Adrenergic, beta-3/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics , Stem Cells/cytology , Stem Cells/metabolism
17.
Nutrients ; 10(12)2018 Dec 12.
Article in English | MEDLINE | ID: mdl-30545041

ABSTRACT

Myricetin is a biologically active natural polyphenol with beneficial effects on metabolic health. This study aimed to examine the effects of myricetin on the expression levels of genes involved in lipolysis and mitochondrial respiration in adipocytes and the anti-obesity potential of myricetin. The results indicated that myricetin reduced triglyceride (TG) content and increased mitochondrial content and oxygen consumption rate (OCR) in adipocytes in vitro. To determine anti-obesity effect of myricetin, C57BL6/J mice were fed a high-fat diet (HFD) for eight weeks and then treated with myricetin (10 mg/kg) for 2 weeks. The in vivo treatment of myricetin reduced body weight by 11%. Furthermore, it improved the glucose tolerance, and increased fatty acid consumption of HFD-fed mice. Myricetin treatment increased Sirt3 expression and reduced the acetylation of mitochondrial proteins in adipose tissue. Finally, the knockdown of Sirt3 in adipocytes reduced the myricetin-induced increase in mitochondrial oxygen consumption rate by about 27% compared to controls. Our results indicated that myricetin exerted anti-obesity effects through the upregulation of Sirt3 expression and mitochondrial metabolism in adipose tissue.


Subject(s)
Anti-Obesity Agents/pharmacology , Flavonoids/pharmacology , Sirtuin 3/metabolism , Up-Regulation/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Cell Line , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Sirtuin 3/analysis , Sirtuin 3/genetics
18.
Sci Rep ; 8(1): 8856, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891910

ABSTRACT

15-lipoxygenase is involved in the generation of specialized pro-resolving lipid mediators that play essential roles in resolution and inflammatory responses. Here, we investigated anti-inflammatory role of Alox15 in skin homeostasis. We demonstrated that knockout (KO) of Alox15 led to hair loss and disrupted the structural integrity of the dorsal skin. Alox15 KO resulted in loss of hair follicle stem cells and abnormal transition of dermal adipocytes into fibroblasts. Alox15 deficiency increased infiltration of proinflammatory macrophages and upregulated proinflammatory and necroptotic signaling in dermal adipose tissue in the dorsal skin. Lipidomic analysis revealed severe loss of resolvin D2 in the dorsal skin of Alox15 KO mice compared to wild type controls. Treatment with resolvin D2 reduced skin inflammation in Alox15 KO mice. Collectively, these results indicate that Alox15-mediated production of resolvin D2 is required to maintain skin integrity by suppressing dermal inflammation.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Docosahexaenoic Acids/metabolism , Inflammation , Skin/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Alopecia , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Cell Death , Fibroblasts/cytology , Gene Knockout Techniques , Hair Follicle/pathology , Homeostasis , Inflammation/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Sci Rep ; 7(1): 7159, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28769076

ABSTRACT

We investigated the role of connexin 43 (Cx43) in maintaining the integrity of mitochondria in brown adipose tissue (BAT). The functional effects of Cx43 were evaluated using inducible, adipocyte-specific Cx43 knockout in mice (Gja1 adipoq KO) and by overexpression and knockdown of Cx43 in cultured adipocytes. Mitochondrial morphology was evaluated by electron microscopy and mitochondrial function and autophagy were assessed by immunoblotting, immunohistochemistry, and qPCR. The metabolic effects of adipocyte-specific knockout of Cx43 were assessed during cold stress and following high fat diet feeding. Cx43 expression was higher in BAT compared to white adipose tissue. Treatment with the ß3-adrenergic receptor agonist CL316,243 increased Cx43 expression and mitochondrial localization. Gja1 adipoq KO mice reduced mitochondrial density and increased the presence of damaged mitochondria in BAT. Moreover, metabolic activation with CL316,243 further reduced mitochondrial integrity and upregulated autophagy in the BAT of Gja1 adipoq KO mice. Inhibition of Cx43 in cultured adipocytes increased the generation of reactive oxygen species and induction of autophagy during ß-adrenergic stimulation. Gja1 adipoq KO mice were cold intolerant, expended less energy in response to ß3-adrenergic receptor activation, and were more insulin resistant after a high-fat diet challenge. Collectively, our data demonstrate that Cx43 is required for maintaining the mitochondrial integrity and metabolic activity of BAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Connexin 43/metabolism , Mitochondria/metabolism , Adipocytes/metabolism , Animals , Connexin 43/genetics , Connexins/metabolism , Gene Knockdown Techniques , Immunohistochemistry , Male , Mice , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Oxidative Stress , Phenotype , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...