Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Sci Total Environ ; 912: 169064, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38052392

ABSTRACT

The role of upflow velocity and Ca2+ concentration in controlling the type and rate of CaCO3 crystallization and their impacts on the anaerobic granular sludge (AnGS) formation and performance in an expanded granular sludge bed (EGSB) reactor were studied. The results showed that an improved upflow velocity could promote metastable CaCO3 crystals and achieve the optimized portion of vaterite with a value of 84 % at 10 m/h with a small amount of aragonite, thus limiting the scaling in the reactor. The removal efficiency of Ca2+ was to some extent positively correlated to the influent Ca2+ concentration, but declined when Ca2+ exceeded a specific threshold. Vaterite was dominant with the increase of Ca2+ concentrations of the influent. Compared with granules in R1 (Ca2+ 10 mg/L) and R2 (Ca2+ 100 mg/L), granules cultivated in R3 (Ca2+ 800 mg/L) revealed maximum amount of biomass with biggest particle size distribution and fastest average settling rate, with relative stable COD removal efficiency and the fast optimized reactor capacity at OLR of 16 kgCOD/m3d. A low upflow velocity and a higher Ca2+ concentration promoted nucleus formation and granules growth at the initial cultivation stage of the EGSB reactor. The Ca2+ concentration had a significant impact on the bacterial community and favoured the growth of Tolumonas and Anaeromousa Anaeroarcus. Archaea, rather than bacteria, was strengthened to contribute more to methane production at a relatively high Ca2+ concentration.


Subject(s)
Sewage , Wastewater , Sewage/microbiology , Calcium , Waste Disposal, Fluid/methods , Anaerobiosis , Crystallization , Bioreactors , Bacteria , Calcium Carbonate
2.
iScience ; 26(10): 108091, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37860762

ABSTRACT

Y-box binding protein 2 (YBX2) is an essential modulator of brown adipose tissue activation, yet the regulation on its own expression and the involved mechanism remains largely unknown. Herein, we report the YBX2 protein level, but not mRNA level, is induced in response to acute ß-adrenergic signaling. In this context, YBX2 is a dual substrate for both AMPK and Akt2. The phosphorylation at Thr115 by AMPK or at Ser137 by Akt2 facilitates YBX2 accumulation in brown adipocytes by decreasing ubiquitination-mediated degradation. Beyond stabilizing PGC1α mRNA, increased YBX2 upon thermogenic activation assists the expression of glycolytic enzymes, promotes glucose utilization and lactate production. Mechanistically, YBX2 modulates translation of glycolytic genes via direct binding to 5'-UTRs of these genes. Together these findings suggest YBX2 is responsive to thermogenic stimuli by phosphorylation modification, and stabilized YBX2 helps to boost glycolysis and thermogenesis in brown adipocytes.

3.
bioRxiv ; 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37745491

ABSTRACT

The endocrine control of food intake remains incompletely understood, and whether the leptin receptor-mediated anorexigenic pathway in the hypothalamus is negatively regulated by a humoral factor is unknown. Here we identify an appetite-stimulating factor - ASRA - that acts as a leptin receptor antagonist. ASRA encodes an 8 kD protein that is abundantly and selectively expressed in adipose tissue and to a lesser extent, in liver, and is upregulated during fasting and cold. ASRA protein associates with autophagosomes and its secretion is induced by energy deficiency. Overexpression of ASRA in mice attenuates leptin receptor signaling leading to elevated blood glucose and development of severe hyperphagic obesity, whereas either adipose- or liver-specific ASRA knockout mice display increased leptin sensitivity, improved glucose homeostasis, reduced food intake, and resistance to high fat diet-induced obesity. Furthermore, ASRA is indispensable for cold-evoked feeding response. Recombinant ASRA (rASRA) protein binds to leptin receptor and suppresses leptin receptor signaling in cultured cells. In vivo, rASRA promotes food intake and increases blood glucose in a leptin receptor signaling-dependent manner. Our studies collectively show that ASRA, acting as a peripheral signal of energy deficit, stimulates appetite and regulates glucose metabolism by antagonizing leptin receptor signaling, thus revealing a previously unknown endocrine mechanism that has important implications for our understanding of leptin resistance.

4.
Adv Sci (Weinh) ; 10(28): e2301855, 2023 10.
Article in English | MEDLINE | ID: mdl-37544897

ABSTRACT

Mitochondria are the pivot organelles to control metabolism and energy homeostasis. The capacity of mitochondrial metabolic adaptions to cold stress is essential for adipocyte thermogenesis. How brown adipocytes keep mitochondrial fitness upon a challenge of cold-induced oxidative stress has not been well characterized. This manuscript shows that IFI27 plays an important role in cristae morphogenesis, keeping intact succinate dehydrogenase (SDH) function and active fatty acid oxidation to sustain thermogenesis in brown adipocytes. IFI27 protein interaction map identifies SDHB and HADHA as its binding partners. IFI27 physically links SDHB to chaperone TNF receptor associated protein 1 (TRAP1), which shields SDHB from oxidative damage-triggered degradation. Moreover, IFI27 increases hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) catalytic activity in ß-oxidation pathway. The reduced SDH level and fatty acid oxidation in Ifi27-knockout brown fat results in impaired oxygen consumption and defective thermogenesis. Thus, IFI27 is a novel regulator of mitochondrial metabolism and thermogenesis.


Subject(s)
Adipocytes, Brown , Succinic Acid , Succinic Acid/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Fatty Acids/metabolism , Thermogenesis/physiology
5.
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
6.
Nat Commun ; 13(1): 7633, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36496438

ABSTRACT

The signaling mechanisms underlying adipose thermogenesis have not been fully elucidated. Particularly, the involvement of adipokines that are selectively expressed in brown adipose tissue (BAT) and beige adipocytes remains to be investigated. Here we show that a previously uncharacterized adipokine (UPF0687 protein / human C20orf27 homolog) we named as Adissp (Adipose-secreted signaling protein) is a key regulator for white adipose tissue (WAT) thermogenesis and glucose homeostasis. Adissp expression is adipose-specific and highly BAT-enriched, and its secretion is stimulated by ß3-adrenergic activation. Gain-of-functional studies collectively showed that secreted Adissp promotes WAT thermogenesis, improves glucose homeostasis, and protects against obesity. Adipose-specific Adissp knockout mice are defective in WAT browning, and are susceptible to high fat diet-induced obesity and hyperglycemia. Mechanistically, Adissp binds to a putative receptor on adipocyte surface and activates protein kinase A independently of ß-adrenergic signaling. These results establish BAT-enriched Adissp as a major upstream signaling component in thermogenesis and offer a potential avenue for the treatment of obesity and diabetes.


Subject(s)
Adipokines , Adipose Tissue, Brown , Mice , Animals , Humans , Adipose Tissue, Brown/metabolism , Thermogenesis , Adipose Tissue, White/metabolism , Obesity/metabolism , Glucose/metabolism , Adrenergic Agents/metabolism , Adipocytes, Brown/metabolism , Energy Metabolism
7.
Yi Chuan ; 44(10): 867-880, 2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36384724

ABSTRACT

The activation of brown adipose tissues and beige adipose tissues can utilize more substrates, including glucose and fatty acids, regulate the energy balance of the whole body and improve metabolic diseases such as obesity and type Ⅱ diabetes. Elucidating the regulatory mechanisms underlying the thermogenic adipose program may provide excellent targets for therapeutics against metabolic diseases. The current studies have indicated that epigenetic modifications are vital for regulating differentiation and thermogenesis of adipose tissues. In this review, we summarize the recent progress of epigenetic modifications in adipose tissue development and thermogenesis from the aspects of DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs in order to provide new ideas for further studying the activation of adipose tissues.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Diseases , Humans , Epigenesis, Genetic , Thermogenesis/genetics , Adipose Tissue, Brown/metabolism , Metabolic Diseases/metabolism
8.
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
9.
Nat Metab ; 3(11): 1536-1551, 2021 11.
Article in English | MEDLINE | ID: mdl-34782792

ABSTRACT

Beiging of white adipose tissue (WAT) is associated with an increase of anti-inflammatory M2-like macrophages in WAT. However, mechanisms through which M2-like macrophages affect beiging are incompletely understood. Here, we show that the macrophage cytokine Slit3 is secreted by adipose tissue macrophages and promotes cold adaptation by stimulating sympathetic innervation and thermogenesis in mice. Analysing the transcriptome of M2-like macrophages in murine inguinal WAT (iWAT) after cold exposure, we identify Slit3 as a secreted cytokine. Slit3 binds to the ROBO1 receptor on sympathetic neurons to stimulate Ca2+/calmodulin-dependent protein kinase II signalling and norepinephrine release, which enhances adipocyte thermogenesis. Adoptive transfer of Slit3-overexpressing M2 macrophages to iWAT promotes beiging and thermogenesis, whereas mice that lack Slit3 in myeloid cells are cold-intolerant and gain more weight. Our findings shed new light on the integral role of M2-like macrophages for adipose tissue homeostasis and uncover the macrophage-Slit3-sympathetic neuron-adipocyte signalling axis as a regulator of long-term cold adaptation.


Subject(s)
Adipose Tissue/innervation , Adipose Tissue/physiology , Adrenergic Fibers/physiology , Macrophages/metabolism , Membrane Proteins/biosynthesis , Thermogenesis , Adipose Tissue, White/innervation , Adipose Tissue, White/metabolism , Animals , Cell Plasticity , Energy Metabolism , Gene Expression Regulation , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , Nerve Tissue Proteins/metabolism , Organ Specificity/genetics , Phosphorylation , Protein Binding , Receptors, Immunologic/metabolism , Temperature , Thermogenesis/genetics , Roundabout Proteins
10.
EMBO J ; 40(24): e108069, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34704268

ABSTRACT

Brown and beige fat are specialized for energy expenditure by dissipating energy from glucose and fatty acid oxidation as heat. While glucose and fatty acid metabolism have been extensively studied in thermogenic adipose tissues, the involvement of amino acids in regulating adaptive thermogenesis remains little studied. Here, we report that asparagine supplementation in brown and beige adipocytes drastically upregulated the thermogenic transcriptional program and lipogenic gene expression, so that asparagine-fed mice showed better cold tolerance. In mice with diet-induced obesity, the asparagine-fed group was more responsive to ß3-adrenergic receptor agonists, manifesting in blunted body weight gain and improved glucose tolerance. Metabolomics and 13 C-glucose flux analysis revealed that asparagine supplement spurred glycolysis to fuel thermogenesis and lipogenesis in adipocytes. Mechanistically, asparagine stimulated the mTORC1 pathway, which promoted expression of thermogenic genes and key enzymes in glycolysis. These findings show that asparagine bioavailability affects glycolytic and thermogenic activities in adipose tissues, providing a possible nutritional strategy for improving systemic energy homeostasis.


Subject(s)
Asparagine/pharmacology , Glycolysis/drug effects , Signal Transduction/drug effects , Thermogenesis/drug effects , Animals , Cells, Cultured , Cold Temperature , Gene Expression Regulation/drug effects , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Metabolomics , Mice
11.
Sci Immunol ; 6(61)2021 07 30.
Article in English | MEDLINE | ID: mdl-34330814

ABSTRACT

IL-33-associated type 2 innate immunity has been shown to support beige fat formation and thermogenesis in subcutaneous inguinal white adipose tissue (iWAT), but little is known about how it is regulated in iWAT. Chemerin, as a newly identified adipokine, is clinically associated with obesity and metabolic disorders. We here show that cold exposure specifically reduces chemerin and its receptor chemerin chemokine-like receptor 1 (CMKLR1) expression in iWAT. Lack of chemerin or adipocytic CMKLR1 enhances cold-induced thermogenic beige fat via potentiating type 2 innate immune responses. Mechanistically, we identify adipocytes, particularly beige adipocytes, as the main source for cold-induced IL-33, which is restricted by the chemerin-CMKLR1 axis via dampening cAMP-PKA signaling, thereby interrupting a feed-forward circuit between beige adipocytes and type 2 innate immunity that is required for cold-induced beige fat and thermogenesis. Moreover, specific deletion of adipocytic IL-33 inhibits cold-induced beige fat and type 2 innate immune responses. Last, genetic blockade of adipocytic CMKLR1 protects against diet-induced obesity and enhances the metabolic benefits of cold stimulation in preestablished obese mice. Thus, our study identifies the chemerin-CMKLR1 axis as a physiological negative regulator of thermogenic beige fat via interrupting adipose-immune communication and suggests targeting adipose CMKLR1 as a potential therapeutic strategy for obesity-related metabolic disorders.


Subject(s)
Adipocytes, Beige/physiology , Chemokines/physiology , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-33/physiology , Receptors, Chemokine/physiology , Thermogenesis , Adipocytes/physiology , Adipocytes, Beige/immunology , Animals , Chemokines/genetics , Chemokines/immunology , Cold Temperature , Diet, High-Fat , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-33/immunology , Male , Mice, Transgenic , Obesity/immunology , Obesity/physiopathology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology
12.
Adv Sci (Weinh) ; 8(16): e2102060, 2021 08.
Article in English | MEDLINE | ID: mdl-34180153

ABSTRACT

Histone deacetylases (HDACs) are widely involved in many biological processes, as well as in control of brown and beige adipose physiology, but the precise molecular mechanisms by which HDACs are assembled into transcriptional machinery to fine-tune thermogenic program remain ill-defined. PWWP domain containing 2b (PWWP2B), which is identified as a component of the nucleosome remodeling and deacetylation complex (NuRD), interacts and stabilizes HDAC1/2 at the thermogenic gene promoters to suppress their expression. Ablation of Pwwp2b promotes adipocyte thermogenesis and ameliorates diet-induced obesity in vivo. Intriguingly, Pwwp2b is not only a brown fat-enriched gene but also dramatically induced by cold and sympathetic stimulation, which may serve as a physiological brake to avoid over-activation of thermogenesis in brown and beige fat cells.

13.
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
14.
Redox Biol ; 43: 101979, 2021 07.
Article in English | MEDLINE | ID: mdl-33895484

ABSTRACT

Loss of perivascular adipose tissue (PVAT) impairs endothelial function and enhances atherosclerosis. However, the roles of PVAT thermoregulation in vascular inflammation and the development of atherosclerosis remains unclear. Bone morphogenetic protein 4 (BMP4) transforms white adipocyte to beige adipocyte, while promotes a brown-to-white shift in inter-scapular brown adipose tissue (BAT). Here, we found that knockdown of BMP4 in PVAT reduced expression of brown adipocyte-characteristic genes and increased endothelial inflammation in vitro co-culture system. Ablating BMP4 expression either in adipose tissues or specifically in BAT in ApoE-/- mice demonstrated a marked exacerbation of atherosclerotic plaque formation in vivo. We further demonstrated that proinflammatory factors (especially IL-1ß) increased in the supernatant of BMP4 knockdown adipocytes. Overexpression of BMP4 in adipose tissues promotes browning of PVAT and protects against atherosclerosis in ApoE-/- mice. These findings uncover an organ crosstalk between PVAT and blood endothelial cells that is engaged in atherosclerosis.


Subject(s)
Atherosclerosis , Endothelial Cells , Adipose Tissue , Adipose Tissue, Brown , Adipose Tissue, White , Animals , Anti-Inflammatory Agents , Bone Morphogenetic Protein 4 , Mice
15.
Proc Natl Acad Sci U S A ; 117(36): 22413-22422, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32839323

ABSTRACT

Brown and beige adipocytes harbor the thermogenic capacity to adapt to environmental thermal or nutritional changes. Histone methylation is an essential epigenetic modification involved in the modulation of nonshivering thermogenesis in adipocytes. Here, we describe a molecular network leading by KMT5c, a H4K20 methyltransferase, that regulates adipocyte thermogenesis and systemic energy expenditure. The expression of Kmt5c is dramatically induced by a ß3-adrenergic signaling cascade in both brown and beige fat cells. Depleting Kmt5c in adipocytes in vivo leads to a decreased expression of thermogenic genes in both brown and subcutaneous (s.c.) fat tissues. These mice are prone to high-fat-diet-induced obesity and develop glucose intolerance. Enhanced transformation related protein 53 (Trp53) expression in Kmt5c knockout (KO) mice, that is due to the decreased repressive mark H4K20me3 on its proximal promoter, is responsible for the metabolic phenotypes. Together, these findings reveal the physiological role for KMT5c-mediated H4K20 methylation in the maintenance and activation of the thermogenic program in adipocytes.


Subject(s)
Adipocytes, Beige/physiology , Adipocytes, Brown/physiology , Histone-Lysine N-Methyltransferase , Thermogenesis/physiology , Tumor Suppressor Protein p53/metabolism , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Animals , Diet, High-Fat , Female , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Male , Mice , Mice, Knockout , Tumor Suppressor Protein p53/genetics
16.
Cell Discov ; 4: 52, 2018.
Article in English | MEDLINE | ID: mdl-30131868

ABSTRACT

Pericardial adipose tissue, which comprises both epicardial adipose tissue (EAT) and paracardial adipose tissue (PAT), has recently been recognized as a novel factor in the pathophysiology of cardiovascular diseases, especially coronary artery disease (CAD). The goal of this study was to evaluate differences in the brown-like characteristic and proteome among human EAT, PAT, and subcutaneous adipose tissue (SAT) to identify candidate molecules causing CAD. Uncoupling protein 1 (UCP-1) and other brown-related proteins were highly expressed in pericardial adipose tissue but was weakly expressed in SAT from the same non-CAD patient. Moreover, pericardial adipose tissues displayed a higher thermogenesis than SAT. However, brown-related genes were lower in CAD pericardial fat. Remarkably, there were lower levels of metabolic enzymes involved in glycolysis, tricarboxylic acid cycle, and fatty acid metabolism in pericardial adipose tissues of CAD. EAT is an organ adjacent to aortic root without anatomy barriers, which differs from PAT. We found that the expression of ribosomal protein S3A (RPS3A) was decreased in human EAT as well as in mouse perivascular adipose tissue (PVAT). Knockdown of RPS3A significantly inhibited adipocyte differentiation in preadipocytes and impaired the function of mitochondria in mature adipocytes. Moreover, RPS3A knockdown in mouse periaortic adipose tissue impaired browning of PVAT, accelerated vascular inflammation, and atherosclerosis progression. Mechanistically, RPS3A can migrate to the mitochondria to maintain the function of brown adipocytes. These findings provide compelling evidence that RPS3A was a key factor for modulating the brown fat-specific gene UCP-1 and carbon metabolic enzymes in EAT for preventing CAD.

17.
Biochem Biophys Res Commun ; 501(1): 273-279, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29730295

ABSTRACT

Brown adipose tissue (BAT) is specialized for energy expenditure, but the signaling pathways that regulate BAT metabolism and activity are incompletely understood. Interferon (IFN) signaling is a sophisticated defense mechanism to counteract viral infection. IFNs and interferon-stimulated genes (ISGs) are reported to exert profound effects on adipocytes. IFN-α inducible protein 27 (Ifi27/ISG12a) is a BAT-enriched gene, yet no any studies on its roles in BAT have been reported. Here, we show that Ifi27 protein localizes to mitochondria and the expression of Ifi27 can be induced by ß3-adrenergic activation in adipose tissues. Knockdown of Ifi27 leads to reduced expression of key enzymes of tricarboxylic acid cycle (TCA), the subunits of electron transport chain (ETC) and uncoupling protein 1 (Ucp1) in brown and beige adipocytes. Moreover, the browning of subcutaneous white fat induced by ß3-adrenergic agonist is also dramatically blocked. Ectopic expression of Ifi27 in brown adipocytes has the opposite effects. Together, these data indicate that Ifi27 regulates mitochondrial function and browning in adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Mitochondria/metabolism , Proteins/metabolism , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Citric Acid Cycle , Electron Transport , Energy Metabolism , Gene Knockdown Techniques , Interferon-alpha/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR delta/metabolism , Proteins/antagonists & inhibitors , Proteins/genetics , Signal Transduction , Thermogenesis , Uncoupling Protein 1/metabolism
18.
Cell Rep ; 22(11): 2860-2872, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29539416

ABSTRACT

Transcriptional co-activator Prdm16 controls brown fat development and white fat browning, but how this thermogenic function is modulated post-translationally is poorly understood. Here, we report that Cbx4, a Polycomb group protein, is a SUMO E3 ligase for Prdm16 and that Cbx4-mediated sumoylation of Prdm16 is required for thermogenic gene expression. Cbx4 expression is enriched in brown fat and is induced in adipose tissue by acute cold exposure. Sumoylation of Prdm16 at lysine 917 by Cbx4 blocks its ubiquitination-mediated degradation, thereby augmenting its stability and thermogenic function. Moreover, this sumoylation event primes Prdm16 to be further stabilized by methyltransferase Ehmt1. Heterozygous Cbx4-knockout mice develop metabolic phenotypes resembling those of Prdm16-knockout mice. Furthermore, fat-specific Cbx4 knockdown and overexpression produce remarkable, opposite effects on white fat remodeling. Our results identify a modifying enzyme for Prdm16, and they demonstrate a central role of Cbx4 in the control of Prdm16 stability and white fat browning.


Subject(s)
Adipocytes, Brown/metabolism , DNA-Binding Proteins/metabolism , Ligases/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/metabolism , Protamines/metabolism , Transcription Factors/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/physiology , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Ligases/genetics , Male , Mice , Mice, Knockout , Polycomb Repressive Complex 1/genetics , Sumoylation , Thermogenesis/physiology , Transfection
19.
Nat Commun ; 8(1): 68, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701693

ABSTRACT

Browning of subcutaneous white fat (iWAT) involves several reprograming events, but the underlying mechanisms are incompletely understood. Here we show that the transcription factor Hlx is selectively expressed in brown adipose tissue (BAT) and iWAT, and is translationally upregulated by ß3-adrenergic signaling-mediated suppression of the translational inhibitor 4E-BP1. Hlx interacts with and is co-activated by Prdm16 to control BAT-selective gene expression and mitochondrial biogenesis. Hlx heterozygous knockout mice have defects in brown-like adipocyte formation in iWAT, and develop glucose intolerance and high fat-induced hepatic steatosis. Conversely, transgenic expression of Hlx at a physiological level drives a full program of thermogenesis and converts iWAT to brown-like fat, which improves glucose homeostasis and prevents obesity and hepatic steatosis. The adipose remodeling phenotypes are recapitulated by fat-specific injection of Hlx knockdown and overexpression viruses, respectively. Our studies establish Hlx as a powerful regulator for systematic white adipose tissue browning and offer molecular insights into the underlying transcriptional mechanism.The transcriptional co-activator Prdm16 regulates browning of white adipose tissue (WAT). Here, the authors show that Prdm16 interacts with the transcription factor Hlx, which is stabilized in response to ß3-adrenergic signaling, to increase thermogenic gene expression and mitochondrial biogenesis in subcutaneous WAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cellular Reprogramming/genetics , Diet, High-Fat , Eukaryotic Initiation Factors , Fatty Liver/genetics , Glucose Intolerance/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Knockout , Organelle Biogenesis , Phosphoproteins/metabolism , Receptors, Adrenergic, beta-3/metabolism , Subcutaneous Fat/metabolism , Thermogenesis/genetics
20.
Dev Cell ; 35(5): 568-583, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26625958

ABSTRACT

Progression from brown preadipocytes to adipocytes engages two transcriptional programs: the expression of adipogenic genes common to both brown fat (BAT) and white fat (WAT), and the expression of BAT-selective genes. However, the dynamics of chromatin states and epigenetic enzymes involved remain poorly understood. Here we show that BAT development is selectively marked and guided by repressive H3K27me3 and is executed by its demethylase Jmjd3. We find that a significant subset of BAT-selective genes, but not common fat genes or WAT-selective genes, are demarcated by H3K27me3 in both brown and white preadipocytes. Jmjd3-catalyzed removal of H3K27me3, in part through Rreb1-mediated recruitment, is required for expression of BAT-selective genes and for development of beige adipocytes both in vitro and in vivo. Moreover, gain- and loss-of-function Jmjd3 transgenic mice show age-dependent body weight reduction and cold intolerance, respectively. Together, we identify an epigenetic mechanism governing BAT fate determination and WAT plasticity.


Subject(s)
Adipose Tissue, Brown/embryology , Adipose Tissue, White/embryology , Gene Expression Regulation, Developmental , Jumonji Domain-Containing Histone Demethylases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Body Weight , DNA-Binding Proteins/metabolism , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Point Mutation , Promoter Regions, Genetic , Sequence Analysis, RNA , Thermogenesis/genetics , Transcription Factors/metabolism , Transgenes , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...