Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Metab ; 81: 101891, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38307386

ABSTRACT

OBJECTIVE: Brown adipose tissue (BAT) development and function are essential for maintaining energy balance. However, the key factors that specifically regulate brown adipogenesis require further identification. Here, we demonstrated that the nuclear receptor subfamily 2 group F member 6 (NR2F6) played a pivotal role in brown adipogenesis and energy homeostasis. METHODS: We examined the differentiation of immortalized brown adipocytes and primary brown adipocytes when NR2F6 were deleted, and explored the mechanism through which NR2F6 regulated adipogenesis using ChIP-qPCR in vitro. Male wild type (WT) and Pdgfra-Cre-mediated deletion of Nr2f6 in preadipocytes (NR2F6-PKO) mice were fed with high fat diet (HFD) for 12 weeks, and adiposity, glucose intolerance, insulin resistance and inflammation were assessed. RESULTS: NR2F6 exhibited abundant expression in BAT, while its expression was minimal in white adipose tissue (WAT). Within BAT, NR2F6 was highly expressed in preadipocytes, experienced a transient increase in the early stage of brown adipocyte differentiation, and significantly decreased in the mature adipocytes. Depletion of NR2F6 in preadipocytes inhibited brown adipogenesis, caused hypertrophy of brown adipocytes, and impaired thermogenic function of BAT, but without affecting WAT development. NR2F6 transcriptionally regulated PPARγ expression to promote adipogenic process in brown adipocytes. Loss of NR2F6 in preadipocytes led to increased susceptibility to diet-induced metabolic disorders. CONCLUSIONS: Our findings unveiled NR2F6 as a novel key regulator of brown adipogenesis, potentially opening up new avenues for maintaining metabolic homeostasis by targeting NR2F6.


Subject(s)
Adipocytes, Brown , Adipose Tissue, Brown , Animals , Male , Mice , Adipocytes, Brown/metabolism , Adipogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Homeostasis
2.
Proc Natl Acad Sci U S A ; 120(33): e2305717120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37549287

ABSTRACT

Great progress has been made in identifying positive regulators that activate adipocyte thermogenesis, but negative regulatory signaling of thermogenesis remains poorly understood. Here, we found that cardiotrophin-like cytokine factor 1 (CLCF1) signaling led to loss of brown fat identity, which impaired thermogenic capacity. CLCF1 levels decreased during thermogenic stimulation but were considerably increased in obesity. Adipocyte-specific CLCF1 transgenic (CLCF1-ATG) mice showed impaired energy expenditure and severe cold intolerance. Elevated CLCF1 triggered whitening of brown adipose tissue by suppressing mitochondrial biogenesis. Mechanistically, CLCF1 bound and activated ciliary neurotrophic factor receptor (CNTFR) and augmented signal transducer and activator of transcription 3 (STAT3) signaling. STAT3 transcriptionally inhibited both peroxisome proliferator-activated receptor-γ coactivator (PGC) 1α and 1ß, which thereafter restrained mitochondrial biogenesis in adipocytes. Inhibition of CNTFR or STAT3 could diminish the inhibitory effects of CLCF1 on mitochondrial biogenesis and thermogenesis. As a result, CLCF1-TG mice were predisposed to develop metabolic dysfunction even without external metabolic stress. Our findings revealed a brake signal on nonshivering thermogenesis and suggested that targeting this pathway could be used to restore brown fat activity and systemic metabolic homeostasis in obesity.


Subject(s)
Adipocytes, Brown , Organelle Biogenesis , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Homeostasis , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Thermogenesis/physiology
3.
Cell Rep ; 42(1): 111948, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640325

ABSTRACT

Obesity, particularly increased visceral fat, positively correlates with various metabolic challenges, including atherosclerosis, but the mechanism is not fully understood. The aim of this study is to determine the role of visceral-fat-derived exosomes (Exo) in endothelial cells and atherosclerosis. We show that obesity changes the miRNA profile of visceral adipose exosomes in mice. Importantly, exosomal miR-27b-3p efficiently enters into the vascular endothelial cells and activates the NF-κB pathway by downregulating PPARα. Mechanistically, miR-27b-3p binds directly to the CDS region of PPARα mRNA, thereby promoting mRNA degradation and suppressing translation. In ApoE-deficient mice, administration of miR-27b-3p mimic increases inflammation and atherogenesis, while overexpression of PPARα protects against atherosclerosis. Thus, obesity-induced exosomal miR-27b-3p promotes endothelial inflammation and facilitates atherogenesis by PPARα suppression. We reveal an exosomal pathway by which obesity aggravates atherosclerosis and proposed therapeutic strategies for atherosclerosis in people with obesity.


Subject(s)
Atherosclerosis , Exosomes , MicroRNAs , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Endothelial Cells/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Adipocytes/metabolism , Inflammation/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Obesity/metabolism , Exosomes/metabolism
4.
Front Cardiovasc Med ; 9: 1038176, 2022.
Article in English | MEDLINE | ID: mdl-36457800

ABSTRACT

Background: Perivascular adipose tissue (PVAT), an active endocrine organ, exerts direct effect on vascular tone through paracrine. Activation of PVAT metabolism plays an inhibitory role in atherosclerosis via secreting relaxing factors. The present studies were designed to investigate the role of PVAT metabolism in regulation of hypertension. Materials and methods: Apolipoprotein E (ApoE) knockout mice with BMP4 knockout in adipose tissue or brown adipose tissue (aP2-DKO or UCP1-DKO, respectively) were used for exploring the role of impaired PVAT metabolism in hypertension. Vascular function was assessed using wire myography. The potential regulatory factor of vascular function was explored using qPCR and ELISA and further confirmed in perivascular fat cell line. Results: Knockout of BMP4 either in adipose tissue or specifically in BAT aggravates high-fat diet (HFD, 40% fat)-induced hypertension and endothelial dysfunction in ApoE-/- mice. In the meanwhile, deficiency of BMP4 also aggravates Ang II (angiotensin II) -induced hypertension and vascular remodeling in ApoE-/- mice. Moreover, deficiency of BMP4 inhibits NO release and induces ROS production. In vitro system, aortic rings pretreated with PVAT extracts from BMP4-DKO mice showed increased vasoconstriction and reduced endothelial-dependent relaxation compared with the controls. We further demonstrated that PVAT of BMP4-DKO mice expressed higher level of angiotensinogen (AGT) and Ang II compared with the controls. Conclusion: Impaired PVAT metabolism aggravates hypertension, and this effect is dependent on the activation of local renin-angiotensin-aldosterone system (RAAS). The results of this study first demonstrate the regulatory role of PVAT metabolism in hypertension.

5.
Diabetes ; 71(9): 1862-1879, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35709007

ABSTRACT

Brown and beige adipocytes dissipate energy in a nonshivering thermogenesis manner, exerting beneficial effects on metabolic homeostasis. CHCHD10 is a nuclear-encoded mitochondrial protein involved in cristae organization; however, its role in thermogenic adipocytes remains unknown. We identify CHCHD10 as a novel regulator for adipocyte thermogenesis. CHCHD10 is dramatically upregulated during thermogenic adipocyte activation by PPARγ-PGC1α and positively correlated with UCP1 expression in adipose tissues from humans and mice. We generated adipocyte-specific Chchd10 knockout mice (Chchd10-AKO) and found that depleting CHCHD10 leads to impaired UCP1-dependent thermogenesis and energy expenditure in the fasting state, with no effect in the fed state. Lipolysis in adipocytes is disrupted by CHCHD10 deficiency, while augmented lipolysis through ATGL overexpression recovers adipocyte thermogenesis in Chchd10-AKO mice. Consistently, overexpression of Chchd10 activates thermogenic adipocytes. Mechanistically, CHCHD10 deficiency results in the disorganization of mitochondrial cristae, leading to impairment of oxidative phosphorylation complex assembly in mitochondria, which in turn inhibits ATP generation. Decreased ATP results in downregulation of lipolysis by reducing nascent protein synthesis of ATGL, thereby suppressing adipocyte thermogenesis. As a result, Chchd10-AKO mice are prone to develop high-fat diet-induced metabolic disorders. Together, our findings reveal an essential role of CHCHD10 in regulating lipolysis and the thermogenic program in adipocytes.


Subject(s)
Adipocytes, Beige , Adipocytes, Brown , Lipolysis , Mitochondrial Proteins , Thermogenesis , Adenosine Triphosphate/metabolism , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Animals , Humans , Lipolysis/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
6.
J Biol Chem ; 298(2): 101544, 2022 02.
Article in English | MEDLINE | ID: mdl-34971706

ABSTRACT

Uncontrolled gluconeogenesis results in elevated hepatic glucose production in type 2 diabetes (T2D). The small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is known to catalyze deSUMOylation of target proteins, with broad effects on cell growth, signal transduction, and developmental processes. However, the role of SENP2 in hepatic gluconeogenesis and the occurrence of T2D remain unknown. Herein, we established SENP2 hepatic knockout mice and found that SENP2 deficiency could protect against high-fat diet-induced hyperglycemia. Pyruvate- or glucagon-induced elevation in blood glucose was attenuated by disruption of SENP2 expression, whereas overexpression of SENP2 in the liver facilitated high-fat diet-induced hyperglycemia. Using an in vitro assay, we showed that SENP2 regulated hepatic glucose production. Mechanistically, the effects of SENP2 on gluconeogenesis were found to be mediated by the cellular fuel sensor kinase, 5'-AMP-activated protein kinase alpha (AMPKα), which is a negative regulator of gluconeogenesis. SENP2 interacted with and deSUMOylated AMPKα, thereby promoting its ubiquitination and reducing its protein stability. Inhibition of AMPKα kinase activity dramatically reversed impaired hepatic gluconeogenesis and reduced blood glucose levels in SENP2-deficient mice. Our study highlights the novel role of hepatic SENP2 in regulating gluconeogenesis and furthers our understanding of the pathogenesis of T2D.


Subject(s)
AMP-Activated Protein Kinases , Cysteine Endopeptidases , Diabetes Mellitus, Type 2 , Hyperglycemia , Sumoylation , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Blood Glucose/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gluconeogenesis , Glucose/metabolism , Hyperglycemia/metabolism , Liver/metabolism , Mice , Peptide Hydrolases/metabolism
7.
Hepatology ; 74(4): 1864-1883, 2021 10.
Article in English | MEDLINE | ID: mdl-33934381

ABSTRACT

BACKGROUND AND AIMS: NAFLD, characterized by aberrant triglyceride accumulation in liver, affects the metabolic remodeling of hepatic and nonhepatic tissues by secreting altered hepatokines. Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is responsible for de-SUMOylation of target protein, with broad effects on cell growth, signal transduction, and developmental processes. However, the role of SENP2 in hepatic metabolism remains unclear. APPROACH AND RESULTS: We found that SENP2 was the most dramatically increased SENP in the fatty liver and that its level was modulated by fed/fasted conditions. To define the role of hepatic SENP2 in metabolic regulation, we generated liver-specific SENP2 knockout (Senp2-LKO) mice. Senp2-LKO mice exhibited resistance to high-fat diet-induced hepatic steatosis and obesity. RNA-sequencing analysis showed that Senp2 deficiency up-regulated genes involved in fatty acid oxidation and down-regulated genes in lipogenesis in the liver. Additionally, ablation of hepatic SENP2 activated thermogenesis of adipose tissues. Improved energy homeostasis of both the liver and adipose tissues by SENP2 disruption prompted us to detect the hepatokines, with FGF21 identified as a key factor markedly elevated in Senp2-LKO mice that maintained metabolic homeostasis. Loss of FGF21 obviously reversed the positive effects of SENP2 deficiency on metabolism. Mechanistically, by screening transcriptional factors of FGF21, peroxisome proliferator-activated receptor alpha (PPARα) was defined as the mediator for SENP2 and FGF21. SENP2 interacted with PPARα and deSUMOylated it, thereby promoting ubiquitylation and subsequent degradation of PPARα, which in turn inhibited FGF21 expression and fatty acid oxidation. Consistently, SENP2 overexpression in liver facilitated development of metabolic disorders. CONCLUSIONS: Our finding demonstrated a key role of hepatic SENP2 in governing metabolic balance by regulating liver-adipose tissue crosstalk, linking the SUMOylation process to metabolic regulation.


Subject(s)
Adipose Tissue/metabolism , Cysteine Endopeptidases/genetics , Fibroblast Growth Factors/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , PPAR alpha/metabolism , Animals , Cysteine Endopeptidases/metabolism , Diet, High-Fat , Energy Metabolism/genetics , Fatty Acids/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Humans , Lipogenesis/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , Obesity/metabolism , Sumoylation , Thermogenesis/genetics , Ubiquitination
8.
J Lipid Res ; 61(12): 1589-1604, 2020 12.
Article in English | MEDLINE | ID: mdl-32978274

ABSTRACT

Beiging of white adipose tissue (WAT) has beneficial effects on metabolism. Although it is known that beige adipocytes are active in lipid catabolism and thermogenesis, how they are regulated deserves more explorations. In this study, we demonstrate that stearoyl-CoA desaturase 1 (SCD1) in subcutaneous WAT (scWAT) responded to cold stimulation and was able to promote mobilization of triacylglycerol [TAG (triglyceride)]. In vitro studies showed that SCD1 promoted lipolysis in C3H10T1/2 white adipocytes. The lipolytic effect was contributed by one of SCD1's products, oleic acid (OA). OA upregulated adipose TAG lipase and hormone-sensitive lipase expression. When SCD1 was overexpressed in the scWAT of mice, lipolysis was enhanced, and oxygen consumption and heat generation were increased. These effects were also demonstrated by the SCD1 knockdown experiments in mice. In conclusion, our study suggests that SCD1, known as an enzyme for lipid synthesis, plays a role in upregulating lipid mobilization through its desaturation product, OA.


Subject(s)
Adipose Tissue, White/metabolism , Lipid Metabolism , Stearoyl-CoA Desaturase/metabolism , Subcutaneous Fat/metabolism , Animals , Mice
9.
Am J Physiol Endocrinol Metab ; 317(6): E1158-E1171, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31550180

ABSTRACT

Although many studies have shown that histamine and its signaling regulate energy homeostasis through the central nervous system, their roles in adipose tissues remain poorly understood. Here, we identified that the histamine H4 receptor (HrH4) was highly expressed in adipocytes at a level higher than that of the other three receptors (i.e., HrH1, HrH2, and HrH3). The HrH4 expression in adipocytes responded to cold through thermogenesis and lipolysis, supported by results from both mouse and cell models. When HrH4 expression was knocked down in the subcutaneous white adipose tissue (scWAT), browning and lipolysis effects triggered by cold were ablated, and the oxygen consumption was also lowered both at the normal and cold conditions. Moreover, mice exhibited browned scWAT, accelerated metabolic rates, and tolerance to hypothermia when 4-methylhistamine (4MH), a selective HrH4 agonist, was adjacently injected to the scWAT. Consistent with these findings, 4MH also triggered the browning and lipolytic effects in cultured C3H10T1/2 adipocytes. Mechanically, we demonstrated that p38/MAPK and ERK/MAPK pathways were involved in these processes. In conclusion, our findings have uncovered an effective role of HrH4 in adipose tissue browning.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cold Temperature , Oxygen Consumption/genetics , Receptors, Histamine H4/genetics , Subcutaneous Fat/metabolism , Thermogenesis/genetics , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/drug effects , Animals , Basal Metabolism/drug effects , Basal Metabolism/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Histamine Agonists/pharmacology , Lipolysis/drug effects , Lipolysis/genetics , MAP Kinase Signaling System , Methylhistamines/pharmacology , Mice , Oxygen Consumption/drug effects , Receptors, Histamine H4/agonists , Receptors, Histamine H4/metabolism , Subcutaneous Fat/drug effects , Thermogenesis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Mol Cell Biol ; 11(1): 14-25, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29462349

ABSTRACT

Thermogenic beige fat improves metabolism and prevents obesity. Emerging evidence shows that the activation of M2 macrophages stimulates beige adipogenesis, whereas the activation of M1 macrophages, which play a major role in inflammation, impedes beige adipogenesis. Thus, the identification of factors that regulate adipose tissue macrophages (ATMs) will help clarify the mechanism involved in beiging. Here, we found that one of the secreted proteins in adipose tissue, namely, BMP4, alters the ATM profile in subcutaneous adipose tissue by activating M2 and inhibiting M1 macrophages. Mechanistically, the BMP4-stimulated p38/MAPK/STAT6/PI3K-AKT signalling pathway is involved. Meanwhile, BMP4 improved the potency of M2 macrophages to induce beige fat biogenesis. Considering that the overexpression of BMP4 in adipose tissue promotes the beiging of subcutaneous adipose tissue and improves insulin sensitivity, these findings provide evidence that BMP4 acts as an activator of beige fat by targeting immuno-metabolic pathways.


Subject(s)
Adipose Tissue, Beige/metabolism , Bone Morphogenetic Protein 4/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/pharmacology , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Insulin Resistance , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Obesity/metabolism , Obesity/pathology , Phosphatidylinositol 3-Kinases/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction , Subcutaneous Fat/metabolism
11.
Mol Cell Biol ; 37(16)2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28559430

ABSTRACT

White adipose tissue (WAT) serves as a reversible energy storage depot in the form of lipids in response to nutritional status. Cavin-1, an essential component in the biogenesis of caveolae, is a positive regulator of lipolysis in adipocytes. However, molecular mechanisms of cavin-1 in the modulation of lipolysis remain poorly understood. Here, we showed that cavin-1 was acetylated at lysines 291, 293, and 298 (3K), which were under nutritional regulation in WAT. We further identified GCN5 as the acetyltransferase and Sirt1 as the deacetylase of cavin-1. Acetylation-mimetic 3Q mutants of cavin-1 augmented fat mobilization in 3T3-L1 adipocytes and zebrafish. Mechanistically, acetylated cavin-1 preferentially interacted with hormone-sensitive lipase and recruited it to the caveolae, thereby promoting lipolysis. Our findings shed light on the essential role of cavin-1 in regulating lipolysis in an acetylation-dependent manner in WAT.


Subject(s)
Adipose Tissue, White/metabolism , Lipolysis , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , 3T3-L1 Cells , Acetylation/drug effects , Adipose Tissue, White/drug effects , Amino Acid Sequence , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , HEK293 Cells , Humans , Lipase/metabolism , Lipolysis/drug effects , Lysine/metabolism , Male , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Niacinamide/pharmacology , Protein Binding/drug effects , RNA-Binding Proteins/chemistry , Sirtuin 1/metabolism , Zebrafish , p300-CBP Transcription Factors/metabolism
12.
J Biol Chem ; 292(28): 11740-11750, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28572510

ABSTRACT

Polycystic ovary syndrome is a common endocrine disorder and a major cause of anovulatory sterility in women at reproductive age. Most patients with polycystic ovary syndrome have hyperandrogenism, caused by excess androgen synthesis. Bone morphogenetic protein 4 (BMP4) is an essential regulator of embryonic development and organ formation, and recent studies have also shown that BMP4 may be involved in female steroidogenesis process. However, the effect of BMP4 on hyperandrogenism remains unknown. Here, using a female mouse model of hyperandrogenism, we found that ovarian BMP4 levels were significantly decreased in hyperandrogenism. Elevated androgens inhibited BMP4 expression via activation of androgen receptors. Moreover, BMP4 treatment suppressed androgen synthesis in theca cells and promoted estrogen production in granulosa cells by regulating the expression of steroidogenic enzymes, including CYP11A, HSD3B2, CYP17A1, and CYP19A1 Consistently, knockdown of BMP4 augmented androgen levels and inhibited estrogen levels. Mechanistically, Smad signaling rather than the p38 MAPK pathway regulated androgen and estrogen formation, thereby mediating the effect of BMP4. Of note, BMP4-transgenic mice were protected against hyperandrogenism. Our observations clarify a vital role of BMP4 in controlling sex hormone levels and offer new insights into intervention for managing hyperandrogenism by targeting the BMP4-Smad signaling pathway.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Disease Models, Animal , Hyperandrogenism/etiology , Ovary/metabolism , Polycystic Ovary Syndrome/physiopathology , Signal Transduction , Smad4 Protein/metabolism , Androgens/metabolism , Androgens/pharmacology , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/genetics , Cells, Cultured , Dehydroepiandrosterone , Down-Regulation/drug effects , Estrogens/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Granulosa Cells/pathology , Mice, Inbred C57BL , Mice, Transgenic , Ovary/drug effects , Ovary/pathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , RNA Interference , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Smad4 Protein/antagonists & inhibitors , Smad4 Protein/genetics , Theca Cells/drug effects , Theca Cells/metabolism , Theca Cells/pathology
13.
Biochem Biophys Res Commun ; 491(3): 814-820, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28647369

ABSTRACT

Dysregulation of insulin signaling leads to type 2 diabetes mellitus (T2DM) and other metabolic disorders. Obesity is an important contributor to insulin resistance, and although the understanding of this relationship has improved in recent years, the mechanism of obesity-induced insulin resistance is not completely understood. Disorders of copper metabolism tend to accompany the development of obesity, which increases the risk of insulin resistance. Synthesis of cytochrome c oxidase 1 (SCO1) functions in the assembly of cytochrome c oxidase (COX) and cellular copper homeostasis. However, the role of SCO1 in the regulation of metabolism remains unknown. Here, we found that obese mice had higher expression of SCO1 and lower levels of copper in white adipose tissue (WAT) than did the control mice. Overexpression of SCO1 in adipocytes was associated with copper deficiency. Copper increased insulin sensitivity by decreasing the level of phosphatase and tensin homolog (PTEN) protein. Ectopic expression of SCO1 led to insulin resistance and was accompanied by a decrease in intracellular copper level, and addition of copper abolished the inhibitory effect of SCO1 on insulin sensitivity. Our results demonstrated a novel role of SCO1 in modulating insulin sensitivity via the regulation of copper concentration in WAT and suggested a potential therapeutic target for T2DM.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Copper/metabolism , Electron Transport Complex IV/biosynthesis , Insulin Resistance , Insulin/metabolism , Obesity/metabolism , Adipocytes/pathology , Adipose Tissue, White/pathology , Animals , Cells, Cultured , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones , Obesity/pathology
15.
EBioMedicine ; 11: 91-100, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27522322

ABSTRACT

Similar to estrogens, bone morphogenetic protein 4 (BMP4) promotes the accumulation of more metabolically active subcutaneous fat and reduction of visceral fat. However, whether there is a cross-talk between BMP4 and estrogen signaling remained unknown. Herein, we found that BMP4 deficiency in white adipose tissue (WAT) increased the estrogen receptor α (ERα) level and its signaling, which prevented adult female mice from developing high fat diet (HFD)-induced obesity and insulin resistance; estrogens depletion up regulated BMP4 expression to overcome overt adiposity and impaired insulin sensitivity with aging, and failure of BMP4 regulation due to genetic knockout led to more fat gain in aged female mice. This mutual regulation between BMP4 and estrogen/ERα signaling may also happen in adipose tissue of women, since the BMP4 level significantly increased after menopause, and was inversely correlated with body mass index (BMI). These findings suggest a counterbalance between BMP4 and estrogen/ERα signaling in the regulation of adiposity and relative metabolism in females.


Subject(s)
Adiposity , Bone Morphogenetic Protein 4/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Glucose/metabolism , Signal Transduction , Adipocytes/metabolism , Adipose Tissue/metabolism , Adiposity/genetics , Age Factors , Animals , Body Mass Index , Bone Morphogenetic Protein 4/genetics , Cell Line , Diet, High-Fat , Estrogens/pharmacology , Female , Gene Expression Regulation , Humans , Insulin Resistance , Metabolic Diseases/metabolism , Mice , Mice, Knockout , Models, Animal , Obesity/metabolism , Protein Binding , Protein Stability
16.
Mol Cell Biol ; 36(20): 2553-67, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27457618

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease, and decreased fatty acid oxidation is one of the important contributors to NAFLD. Mitochondrial trifunctional protein α-subunit (MTPα) functions as a critical enzyme for fatty acid ß-oxidation, but whether dysregulation of MTPα is pathogenically connected to NAFLD is poorly understood. We show that MTPα is acetylated at lysine residues 350, 383, and 406 (MTPα-3K), which promotes its protein stability by antagonizing its ubiquitylation on the same three lysines (MTPα-3K) and blocking its subsequent degradation. Sirtuin 4 (SIRT4) has been identified as the deacetylase, deacetylating and destabilizing MTPα. Replacement of MTPα-3K with either MTPα-3KR or MTPα-3KQ inhibits cellular lipid accumulation both in free fatty acid (FFA)-treated alpha mouse liver 12 (AML12) cells and primary hepatocytes and in the livers of high-fat/high-sucrose (HF/HS) diet-fed mice. Moreover, knockdown of SIRT4 could phenocopy the effects of MTPα-3K mutant expression in mouse livers, and MTPα-3K mutants more efficiently attenuate SIRT4-mediated hepatic steatosis in HF/HS diet-fed mice. Importantly, acetylation of both MTPα and MTPα-3K is decreased while SIRT4 is increased in the livers of mice and humans with NAFLD. Our study reveals a novel mechanism of MTPα regulation by acetylation and ubiquitylation and a direct functional link of this regulation to NAFLD.


Subject(s)
Fatty Acids/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/chemistry , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Sirtuins/metabolism , Acetylation , Animals , Disease Models, Animal , HEK293 Cells , Humans , Lipid Metabolism , Lysine/metabolism , Mice , Oxidation-Reduction , Protein Stability , Ubiquitination
17.
J Mol Cell Biol ; 8(4): 302-12, 2016 08.
Article in English | MEDLINE | ID: mdl-27030507

ABSTRACT

The expansion of subcutaneous (SC) white adipose tissue (WAT) has beneficial effects on metabolic health. Our previous work showed an increased number of bone morphogenetic protein 4 (BMP4)-activated beige adipocytes in SC WAT, indicating a potential role of BMP4 in adipocyte recruitment. It was also demonstrated that BMP4 committed multipotent mesodermal C3H10T1/2 stem cells to the adipocyte lineage ex vivo However, the mechanism by which BMP4 regulates adipogenesis in vivo has not been clarified. In this study, we found that BMP4 stimulated de novo adipogenesis in SC WAT concomitant with enhanced blood vessel formation, thus promoting adipose tissue angiogenesis. Platelet-derived growth factor receptor-ß-positive (PDGFRß(+)) multipotent stem cells within the neoangiogenic vessels were found to be adipocyte progenitors. Moreover, BMP4 downregulated PDGFRß by stimulating the lysosome-dependent degradation, which efficiently initiated adipogenic differentiation. These results suggest how BMP4 regulates adipocyte recruitment in SC WAT, and thus promote its beneficial metabolic effects.


Subject(s)
Adipogenesis , Bone Morphogenetic Protein 4/metabolism , Neovascularization, Physiologic , Subcutaneous Fat/growth & development , Subcutaneous Fat/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Blood Vessels/metabolism , Cell Differentiation , Cytokines/metabolism , Down-Regulation , Inguinal Canal/anatomy & histology , Lysosomes/metabolism , Mice, Knockout , Pericytes/metabolism , Proteolysis , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stem Cells/metabolism , Up-Regulation
18.
Diabetes ; 64(12): 4061-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26324179

ABSTRACT

Obesity is associated with chronic low-level inflammation, especially in fat tissues, which contributes to insulin resistance and type 2 diabetes mellitus (T2DM). Protein inhibitor of activated STAT 1 (PIAS1) modulates a variety of cellular processes such as cell proliferation and DNA damage responses. Particularly, PIAS1 functions in the innate immune system and is a key regulator of the inflammation cascade. However, whether PIAS1 is involved in the regulation of insulin sensitivity remains unknown. Here, we demonstrated that PIAS1 expression in white adipose tissue (WAT) was downregulated by c-Jun N-terminal kinase in prediabetic mice models. Overexpression of PIAS1 in inguinal WAT of prediabetic mice significantly improved systemic insulin sensitivity, whereas knockdown of PIAS1 in wild-type mice led to insulin resistance. Mechanistically, PIAS1 inhibited the activation of stress-induced kinases and the expression of nuclear factor-κB target genes in adipocytes, mainly including proinflammatory and chemotactic factors. In doing so, PIAS1 inhibited macrophage infiltration in adipose tissue, thus suppressing amplification of the inflammation cascade, which in turn improved insulin sensitivity. These results were further verified in a fat transplantation model. Our findings shed light on the critical role of PIAS1 in controlling insulin sensitivity and suggest a therapeutic potential of PIAS1 in T2DM.


Subject(s)
Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/etiology , Insulin Resistance , Macrophages/metabolism , Obesity/metabolism , Prediabetic State/etiology , Protein Inhibitors of Activated STAT/metabolism , Adipose Tissue, White/immunology , Animals , Chemotaxis , Diabetes Mellitus, Type 2/prevention & control , Diet, High-Fat/adverse effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Macrophage Activation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/immunology , Obesity/physiopathology , Obesity/therapy , Prediabetic State/prevention & control , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Protein Inhibitors of Activated STAT/genetics , RAW 264.7 Cells , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
J Hepatol ; 63(6): 1466-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26272872

ABSTRACT

BACKGROUND & AIMS: Due to the protective effect of estrogen against hepatic fat accumulation, the prevalence of non-alcoholic fatty liver disease (NAFLD) in premenopausal women is lower than that in men at the same age and in postmenopausal women. Our study was to further elucidate an underlying mechanism by which estrogen prevents NAFLD from miRNA perspective in female mice. METHODS: miRNA expression was evaluated by TaqMan miRNA assay. Luciferase and ChIP assay were done to validate regulation of miR-125b by estrogen via estrogen receptor alpha (ERα). Nile red and Oil red O staining were used to check lipid content. Overexpressing or inhibiting the physiological role of miR-125b in the liver of mice through injecting adenovirus were used to identify the function of miR-125b in vivo. RESULTS: miR-125b expression was activated by estrogen via ERα in vitro and in vivo. miR-125b inhibited lipid accumulation both in HepG2 cells and primary mouse hepatocytes. Consistently, ovariectomized or liver-specific ERα knockdown mice treated with miR-125b overexpressing adenoviruses were resistant to hepatic steatosis induced by high-fat diet, due to decreased fatty acid uptake and synthesis and decreased triglyceride synthesis. Conversely, inhibiting the physiological role of miR-125b with a sponge decoy slightly promoted liver steatosis with a high-fat diet. Notably, we provided evidence showing that fatty acid synthase was a functional target of miR-125b. CONCLUSION: Our findings identify a novel mechanism by which estrogen protects against hepatic steatosis in female mice via upregulating miR-125b expression.


Subject(s)
Estrogens/metabolism , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Diet, High-Fat/adverse effects , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Ovariectomy , Up-Regulation
20.
Mol Cell Biol ; 34(12): 2330-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24732798

ABSTRACT

Previous studies have shown that CCAAT/enhancer-binding protein α (C/EBPα) plays a very important role during adipocyte terminal differentiation and that AP-2α (activator protein 2α) acts as a repressor to delay the expression of C/EBPα. However, the mechanisms by which AP-2α prevents the expression of C/EBPα are not fully understood. Here, we present evidence that Suv39h1, a histone H3 lysine 9 (H3K9)-specific trimethyltransferase, and G9a, a euchromatic methyltransferase, both interact with AP-2α and enhance AP-2α-mediated transcriptional repression of C/EBPα. Interestingly, we discovered that G9a mediates dimethylation of H3K9, thus providing the substrate, which is methylated by Suv39h1, to H3K9me3 on the C/EBPα promoter. The expression level of AP-2α was consistent with enrichment of H3K9me2 and H3K9me3 on the C/EBPα promoter in 3T3-L1 preadipocytes. Knockdown of Suv39h markedly increased C/EBPα expression and promoted adipogenesis. Conversely, ectopic expression of Suv39h1 delayed C/EBPα expression and impaired the accumulation of triglyceride, while simultaneous knockdown of AP-2α or G9a partially rescued this process. These findings indicate that Suv39h1 enhances AP-2α-mediated transcriptional repression of C/EBPα in an epigenetic manner and further inhibits adipocyte differentiation.


Subject(s)
Adipogenesis , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Methyltransferases/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-2/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation , Epigenesis, Genetic , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/metabolism , Mice , Models, Biological , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...