Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters








Publication year range
1.
Diabetes ; 71(10): 2084-2093, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35772021

ABSTRACT

Skeletal muscle is a major regulator of glycemic control at rest, and glucose utilization increases drastically during exercise. Sustaining a high glucose utilization via glycolysis requires efficient replenishment of NAD+ in the cytosol. Apoptosis-inducing mitochondrion-associated factor 2 (AIFM2) was previously shown to be a NADH oxidoreductase domain-containing flavoprotein that promotes glycolysis for diet and cold-induced thermogenesis. Here, we find that AIFM2 is selectively and highly induced in glycolytic extensor digitorum longus (EDL) muscle during exercise. Overexpression (OE) of AIFM2 in myotubes is sufficient to elevate the NAD+-to-NADH ratio, increasing the glycolytic rate. Thus, OE of AIFM2 in skeletal muscle greatly increases exercise capacity, with increased glucose utilization. Conversely, muscle-specific Aifm2 depletion via in vivo transfection of hairpins against Aifm2 or tamoxifen-inducible haploinsufficiency of Aifm2 in muscles decreases exercise capacity and glucose utilization in mice. Moreover, muscle-specific introduction of NDE1, Saccharomyces cerevisiae external NADH dehydrogenase (NDE), ameliorates impairment in glucose utilization and exercise intolerance of the muscle-specific Aifm2 haploinsufficient mice. Together, we show a novel role for AIFM2 as a critical metabolic regulator for efficient utilization of glucose in glycolytic EDL muscles.


Subject(s)
Glucose , NAD , Animals , Glucose/metabolism , Glycolysis/physiology , Mice , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/metabolism , NAD/metabolism , NADH Dehydrogenase/metabolism , Tamoxifen/metabolism
2.
Commun Biol ; 5(1): 44, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027667

ABSTRACT

Kings and queens of eusocial termites can live for decades, while queens sustain a nearly maximal fertility. To investigate the molecular mechanisms underlying their long lifespan, we carried out transcriptomics, lipidomics and metabolomics in Macrotermes natalensis on sterile short-lived workers, long-lived kings and five stages spanning twenty years of adult queen maturation. Reproductives share gene expression differences from workers in agreement with a reduction of several aging-related processes, involving upregulation of DNA damage repair and mitochondrial functions. Anti-oxidant gene expression is downregulated, while peroxidability of membranes in queens decreases. Against expectations, we observed an upregulated gene expression in fat bodies of reproductives of several components of the IIS pathway, including an insulin-like peptide, Ilp9. This pattern does not lead to deleterious fat storage in physogastric queens, while simple sugars dominate in their hemolymph and large amounts of resources are allocated towards oogenesis. Our findings support the notion that all processes causing aging need to be addressed simultaneously in order to prevent it.


Subject(s)
Aging , DNA Repair , Insulin/physiology , Isoptera/physiology , Animals , Fertility , Longevity , Reproduction , Up-Regulation
3.
Front Endocrinol (Lausanne) ; 12: 698619, 2021.
Article in English | MEDLINE | ID: mdl-34239501

ABSTRACT

[This corrects the article DOI: 10.3389/fendo.2021.595020.].

4.
Front Endocrinol (Lausanne) ; 12: 595020, 2021.
Article in English | MEDLINE | ID: mdl-33841324

ABSTRACT

Obesity, an excess accumulation of white adipose tissue (WAT), has become a global epidemic and is associated with complex diseases, such as type 2 diabetes and cardiovascular diseases. Presently, there are no safe and effective therapeutic agents to treat obesity. In contrast to white adipocytes that store energy as triglycerides in unilocular lipid droplet, brown and brown-like or beige adipocytes utilize fatty acids (FAs) and glucose at a high rate mainly by uncoupling protein 1 (UCP1) action to uncouple mitochondrial proton gradient from ATP synthesis, dissipating energy as heat. Recent studies on the presence of brown or brown-like adipocytes in adult humans have revealed their potential as therapeutic targets in combating obesity. Classically, the main signaling pathway known to activate thermogenesis in adipocytes is ß3-adrenergic signaling, which is activated by norepinephrine in response to cold, leading to activation of the thermogenic program and browning. In addition to the ß3-adrenergic signaling, numerous other hormones and secreted factors have been reported to affect thermogenesis. In this review, we discuss several major pathways, ß3-adrenergic, insulin/IGF1, thyroid hormone and TGFß family, which regulate thermogenesis and browning of WAT.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Signal Transduction , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Humans , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
5.
Dev Cell ; 56(10): 1437-1451.e3, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33878347

ABSTRACT

Adipose tissue mass and adiposity change throughout the lifespan. During aging, while visceral adipose tissue (VAT) tends to increase, peripheral subcutaneous adipose tissue (SAT) decreases significantly. Unlike VAT, which is linked to metabolic diseases, including type 2 diabetes, SAT has beneficial effects. However, the molecular details behind the aging-associated loss of SAT remain unclear. Here, by comparing scRNA-seq of total stromal vascular cells of SAT from young and aging mice, we identify an aging-dependent regulatory cell (ARC) population that emerges only in SAT of aged mice and humans. ARCs express adipose progenitor markers but lack adipogenic capacity; they secrete high levels of pro-inflammatory chemokines, including Ccl6, to inhibit proliferation and differentiation of neighboring adipose precursors. We also found Pu.1 to be a driving factor for ARC development. We identify an ARC population and its capacity to inhibit differentiation of neighboring adipose precursors, correlating with aging-associated loss of SAT.


Subject(s)
Adipogenesis , Aging/physiology , Subcutaneous Fat/cytology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , CD36 Antigens/metabolism , Cell Proliferation , Chemokines/metabolism , Galectin 3/metabolism , Mice , Proto-Oncogene Proteins/metabolism , Stem Cells/cytology , Trans-Activators/metabolism
6.
Elife ; 92020 10 27.
Article in English | MEDLINE | ID: mdl-33107819

ABSTRACT

Brown adipose tissue is a metabolically beneficial organ capable of dissipating chemical energy into heat, thereby increasing energy expenditure. Here, we identify Dot1l, the only known H3K79 methyltransferase, as an interacting partner of Zc3h10 that transcriptionally activates the Ucp1 promoter and other BAT genes. Through a direct interaction, Dot1l is recruited by Zc3h10 to the promoter regions of thermogenic genes to function as a coactivator by methylating H3K79. We also show that Dot1l is induced during brown fat cell differentiation and by cold exposure and that Dot1l and its H3K79 methyltransferase activity is required for thermogenic gene program. Furthermore, we demonstrate that Dot1l ablation in mice using Ucp1-Cre prevents activation of Ucp1 and other target genes to reduce thermogenic capacity and energy expenditure, promoting adiposity. Hence, Dot1l plays a critical role in the thermogenic program and may present as a future target for obesity therapeutics.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Cell Differentiation , Energy Metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Methylation , Mice , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Uncoupling Protein 1/genetics
7.
Diabetes ; 69(4): 525-531, 2020 04.
Article in English | MEDLINE | ID: mdl-32198196

ABSTRACT

Hepatosteatosis, which is frequently associated with development of metabolic syndrome and insulin resistance, manifests when triglyceride (TG) input in the liver is greater than TG output, resulting in the excess accumulation of TG. Dysregulation of lipogenesis therefore has the potential to increase lipid accumulation in the liver, leading to insulin resistance and type 2 diabetes. Recently, efforts have been made to examine the epigenetic regulation of metabolism by histone-modifying enzymes that alter chromatin accessibility for activation or repression of transcription. For regulation of lipogenic gene transcription, various known lipogenic transcription factors, such as USF1, ChREBP, and LXR, interact with and recruit specific histone modifiers, directing specificity toward lipogenesis. Alteration or impairment of the functions of these histone modifiers can lead to dysregulation of lipogenesis and thus hepatosteatosis leading to insulin resistance and type 2 diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Epigenesis, Genetic , Fatty Liver/metabolism , Lipogenesis/physiology , Liver/metabolism , Animals , Humans , Insulin Resistance , Metabolic Syndrome/metabolism
8.
Biochem J ; 477(6): 1137-1148, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32219439

ABSTRACT

Brown adipose tissue (BAT) is a metabolically beneficial organ capable of burning fat by dissipating chemical energy into heat, thereby increasing energy expenditure. Moreover, subcutaneous white adipose tissue can undergo so-called browning/beiging. The recent recognition of the presence of brown or beige adipocytes in human adults has attracted much attention to elucidate the molecular mechanism underlying the thermogenic adipose program. Many key transcriptional regulators critical for the thermogenic gene program centering on activating the UCP1 promoter, have been discovered. Thermogenic gene expression in brown adipocytes rely on co-ordinated actions of a multitude of transcription factors, including EBF2, PPARγ, Zfp516 and Zc3h10. These transcription factors probably integrate into a cohesive network for BAT gene program. Moreover, these transcription factors recruit epigenetic factors, such as LSD1 and MLL3/4, for specific histone signatures to establish the favorable chromatin landscape. In this review, we discuss advances made in understanding the molecular mechanism underlying the thermogenic gene program, particularly epigenetic regulation.


Subject(s)
Adipocytes, Brown/metabolism , Epigenesis, Genetic/physiology , Histones/metabolism , Thermogenesis/physiology , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Histones/genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Nat Commun ; 11(1): 796, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034158

ABSTRACT

Fatty acid and triglyceride synthesis increases greatly in response to feeding and insulin. This lipogenic induction involves coordinate transcriptional activation of various enzymes in lipogenic pathway, including fatty acid synthase and glycerol-3-phosphate acyltransferase. Here, we show that JMJD1C is a specific histone demethylase for lipogenic gene transcription in liver. In response to feeding/insulin, JMJD1C is phosphorylated at T505 by mTOR complex to allow direct interaction with USF-1 for recruitment to lipogenic promoter regions. Thus, by demethylating H3K9me2, JMJD1C alters chromatin accessibility to allow transcription. Consequently, JMJD1C promotes lipogenesis in vivo to increase hepatic and plasma triglyceride levels, showing its role in metabolic adaption for activation of the lipogenic program in response to feeding/insulin, and its contribution to development of hepatosteatosis resulting in insulin resistance.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Lipogenesis/physiology , Oxidoreductases, N-Demethylating/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Diet, High-Fat/adverse effects , Eating/genetics , Eating/physiology , Female , Gene Expression Regulation , Genome-Wide Association Study , Hep G2 Cells , Histones/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance , Jumonji Domain-Containing Histone Demethylases/genetics , Lipogenesis/drug effects , Lipogenesis/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases, N-Demethylating/genetics , Phosphorylation , Promoter Regions, Genetic , Triglycerides/blood , Triglycerides/metabolism , Upstream Stimulatory Factors/metabolism
10.
Mol Cell ; 77(3): 600-617.e4, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31952989

ABSTRACT

Brown adipose tissue (BAT) is highly metabolically active tissue that dissipates energy via UCP1 as heat, and BAT mass is correlated negatively with obesity. The presence of BAT/BAT-like tissue in humans renders BAT as an attractive target against obesity and insulin resistance. Here, we identify Aifm2, a NADH oxidoreductase domain containing flavoprotein, as a lipid droplet (LD)-associated protein highly enriched in BAT. Aifm2 is induced by cold as well as by diet. Upon cold or ß-adrenergic stimulation, Aifm2 associates with the outer side of the mitochondrial inner membrane. As a unique BAT-specific first mammalian NDE (external NADH dehydrogenase)-like enzyme, Aifm2 oxidizes NADH to maintain high cytosolic NAD levels in supporting robust glycolysis and to transfer electrons to the electron transport chain (ETC) for fueling thermogenesis. Aifm2 in BAT and subcutaneous white adipose tissue (WAT) promotes oxygen consumption, uncoupled respiration, and heat production during cold- and diet-induced thermogenesis. Aifm2, thus, can ameliorate diet-induced obesity and insulin resistance.


Subject(s)
Adipose Tissue, Brown/metabolism , Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , Thermogenesis/physiology , Adipose Tissue, White/metabolism , Animals , Apoptosis Regulatory Proteins/physiology , Diet , Energy Metabolism , Glucose/metabolism , Glycolysis/physiology , HEK293 Cells , Humans , Insulin Resistance , Lipid Droplets/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/physiology , Multienzyme Complexes/metabolism , NAD/metabolism , NAD/physiology , NADH, NADPH Oxidoreductases/metabolism , Obesity/metabolism , Oxidation-Reduction , Oxygen Consumption , Uncoupling Protein 1/metabolism
11.
Cell Rep ; 29(9): 2621-2633.e4, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31775033

ABSTRACT

Brown adipose tissue harbors UCP1 to dissipate chemical energy as heat. However, the transcriptional network that governs the thermogenic gene program is incompletely understood. Zc3h10, a CCCH-type zinc finger protein, has recently been reported to bind RNA. However, we report here that Zc3h10 functions as a transcription factor to activate UCP1 not through the enhancer region, but by binding to a far upstream region of the UCP1 promoter. Upon sympathetic stimulation, Zc3h10 is phosphorylated at S126 by p38 mitogen-activated protein kinase (MAPK) to increase binding to the distal region of the UCP1 promoter. Zc3h10, as well as mutant Zc3h10, which cannot bind RNA, enhances thermogenic capacity and energy expenditure, protecting mice from diet-induced obesity. Conversely, Zc3h10 ablation in UCP1+ cells in mice impairs thermogenic capacity and lowers oxygen consumption, leading to weight gain. Hence, Zc3h10 plays a critical role in the thermogenic gene program and may present future targets for obesity therapeutics.


Subject(s)
Adipose Tissue, Brown/metabolism , Carrier Proteins/genetics , Thermogenesis/genetics , Transcription Factors/metabolism , Animals , Humans , Mice , Phosphorylation
12.
Nat Commun ; 10(1): 1760, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988300

ABSTRACT

The mammary gland experiences substantial remodeling and regeneration during development and reproductive life, facilitated by stem cells and progenitors that act in concert with physiological stimuli. While studies have focused on deciphering regenerative cells within the parenchymal epithelium, cell lineages in the stroma that may directly contribute to epithelial biology is unknown. Here we identify, in mouse, the transition of a PDGFRα+ mesenchymal cell population into mammary epithelial progenitors. In addition to being adipocyte progenitors, PDGFRα+ cells make a de novo contribution to luminal and basal epithelia during mammary morphogenesis. In the adult, this mesenchymal lineage primarily generates luminal progenitors within lobuloalveoli during sex hormone exposure or pregnancy. We identify cell migration as a key molecular event that is activated in mesenchymal progenitors in response to epithelium-derived chemoattractant. These findings demonstrate a stromal reservoir of epithelial progenitors and provide insight into cell origins and plasticity during mammary tissue growth.


Subject(s)
Adipocytes/cytology , Mammary Glands, Animal/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stromal Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Epithelial Cells , Humans , Mammary Glands, Animal/growth & development , Mice
13.
Article in English | MEDLINE | ID: mdl-29704660

ABSTRACT

White adipose tissue (WAT) is the primary energy storage organ and its excess contributes to obesity, while brown adipose tissue (BAT) and inducible thermogenic (beige/brite) adipocytes in WAT dissipate energy via Ucp1 to maintain body temperature. BAT and subcutaneous WAT develop perinatally while visceral WAT forms after birth from precursors expressing distinct markers, such as Myf5, Pref-1, Wt1, and Prx1, depending on the anatomical location. In addition to the embryonic adipose precursors, a pool of endothelial cells or mural cells expressing Pparγ, Pdgfrß, Sma and Zfp423 may become adipocytes during WAT expansion in adults. Several markers, such as Cd29, Cd34, Sca1, Cd24, Pdgfrα and Pref-1 are detected in adult WAT SVF cells that can be differentiated into adipocytes. However, potential heterogeneity and differences in developmental stage of these cells are not clear. Beige cells form in a depot- and condition-specific manner by de novo differentiation of precursors or by transdifferentiation. Thermogenic gene activation in brown and beige adipocytes relies on common transcriptional machinery that includes Prdm16, Zfp516, Pgc1α and Ebf2. Moreover, through changing the chromatin landscape, histone methyltransferases, such as Mll3/4 and Ehmt1, as well as demethylases, such as Lsd1, play an important role in regulating the thermogenic gene program. With the presence of BAT and beige/brite cells in human adults, increasing thermogenic activity of BAT and BAT-like tissues may help promote energy expenditure to combat obesity.


Subject(s)
Adipose Tissue, Brown/growth & development , Adipose Tissue, White/growth & development , Animals , Epigenesis, Genetic , Humans
14.
Cell Rep ; 25(4): 1002-1017.e4, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355480

ABSTRACT

Adipocytes arise from the commitment and differentiation of adipose precursors in white adipose tissue (WAT). In studying adipogenesis, precursor markers, including Pref-1 and PDGFRα, are used to isolate precursors from stromal vascular fractions of WAT, but the relation among the markers is not known. Here, we used the Pref-1 promoter-rtTA system in mice for labeling Pref-1+ cells and for inducible inactivation of the Pref-1 target Sox9. We show the requirement of Sox9 for the maintenance of Pref-1+ proliferative, early precursors. Upon Sox9 inactivation, these Pref-1+ cells become PDGFRα+ cells that express early adipogenic markers. Thus, we show that Pref-1+ cells precede PDGFRα+ cells in the adipogenic pathway and that Sox9 inactivation is required for WAT growth and expansion. Furthermore, we show that in maintaining early adipose precursors, Sox9 activates Meis1, which prevents adipogenic differentiation. Our study also demonstrates the Pref-1 promoter-rtTA system for inducible gene inactivation in early adipose precursor populations.


Subject(s)
Adipogenesis , Intercellular Signaling Peptides and Proteins/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOX9 Transcription Factor/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Base Sequence , Biomarkers/metabolism , Calcium-Binding Proteins , Male , Mice , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism
15.
Cell Metab ; 27(3): 602-615.e4, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29514068

ABSTRACT

The activation of brown/beige adipose tissue (BAT) metabolism and the induction of uncoupling protein 1 (UCP1) expression are essential for BAT-based strategies to improve metabolic homeostasis. Here, we demonstrate that BAT utilizes actomyosin machinery to generate tensional responses following adrenergic stimulation, similar to muscle tissues. The activation of actomyosin mechanics is critical for the acute induction of oxidative metabolism and uncoupled respiration in UCP1+ adipocytes. Moreover, we show that actomyosin-mediated elasticity regulates the thermogenic capacity of adipocytes via the mechanosensitive transcriptional co-activators YAP and TAZ, which are indispensable for normal BAT function. These biomechanical signaling mechanisms may inform future strategies to promote the expansion and activation of brown/beige adipocytes.


Subject(s)
Actomyosin/physiology , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Uncoupling Protein 1/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes, Beige/cytology , Adipocytes, Brown/cytology , Animals , Cell Cycle Proteins , Cell Respiration , Cells, Cultured , Disease Models, Animal , Homeostasis , Mice , Oxygen/metabolism , Phosphoproteins/metabolism , Signal Transduction , Thermogenesis , Trans-Activators , YAP-Signaling Proteins
16.
Oncotarget ; 9(3): 2969-2970, 2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29423020
17.
Sci Signal ; 10(467)2017 02 21.
Article in English | MEDLINE | ID: mdl-28223413

ABSTRACT

De novo lipogenesis is precisely regulated by nutritional and hormonal conditions. The genes encoding various enzymes involved in this process, such as fatty acid synthase (FASN), are transcriptionally activated in response to insulin. We showed that USF1, a key transcription factor for FASN activation, directly interacted with the Mediator subunit MED17 at the FASN promoter. This interaction recruited Mediator, which can bring POL II and other general transcription machinery to the complex. Moreover, we showed that MED17 was phosphorylated at Ser53 by casein kinase 2 (CK2) in the livers of fed mice or insulin-stimulated hepatocytes, but not in the livers of fasted mice or untreated hepatocytes. Furthermore, activation of the FASN promoter in response to insulin required this CK2-mediated phosphorylation event, which occurred only in the absence of p38 MAPK-mediated phosphorylation at Thr570 Overexpression of a nonphosphorylatable S53A MED17 mutant or knockdown of MED17, as well as CK2 knockdown or inhibition, impaired hepatic de novo fatty acid synthesis and decreased triglyceride content in mice. These results demonstrate that CK2-mediated phosphorylation of Ser53 in MED17 is required for the transcriptional activation of lipogenic genes in response to insulin.


Subject(s)
Casein Kinase II/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Lipogenesis , Mediator Complex/metabolism , Transcriptional Activation , Amino Acid Substitution , Animals , Casein Kinase II/genetics , Fatty Acid Synthase, Type I/metabolism , Fatty Acids/blood , Fatty Acids/genetics , Insulin/genetics , Male , Mediator Complex/genetics , Mice , Mice, Obese , Mutation, Missense , Phosphorylation
18.
Trends Endocrinol Metab ; 28(1): 19-31, 2017 01.
Article in English | MEDLINE | ID: mdl-27692461

ABSTRACT

In contrast to white adipose tissue (WAT), which stores energy in the form of triglycerides, brown adipose tissue (BAT) dissipates energy by producing heat to maintain body temperature by burning glucose and fatty acids in a process called adaptive thermogenesis. The presence of an inducible thermogenic adipose tissue, and its beneficial effects for maintaining body weight and glucose and lipid homeostasis, has raised intense interest in understanding the regulation of thermogenesis. Elucidating the regulatory mechanisms underlying the thermogenic adipose program may provide excellent targets for therapeutics against obesity and diabetes. Here we review recent research on the role of epigenetics in the thermogenic gene program, focusing on DNA methylation and histone modifications.


Subject(s)
Epigenesis, Genetic/genetics , Thermogenesis/genetics , Adipose Tissue, Brown/metabolism , Animals , DNA Methylation/genetics , DNA Methylation/physiology , Histones/metabolism , Humans , Thermogenesis/physiology
19.
Cell Rep ; 15(11): 2536-49, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264172

ABSTRACT

Zfp516, a brown fat (BAT)-enriched and cold-inducible transcription factor, promotes transcription of UCP1 and other BAT-enriched genes for non-shivering thermogenesis. Here, we identify lysine-specific demethylase 1 (LSD1) as a direct binding partner of Zfp516. We show that, through interaction with Zfp516, LSD1 is recruited to UCP1 and other BAT-enriched genes, such as PGC1α, to function as a coactivator by demethylating H3K9. We also show that LSD1 is induced during brown adipogenesis and that LSD1 and its demethylase activity is required for the BAT program. Furthermore, we show that LSD1 ablation in mice using Myf5-Cre alters embryonic BAT development. Moreover, BAT-specific deletion of LSD1 via the use of UCP1-Cre impairs the BAT program and BAT development, making BAT resemble WAT, reducing thermogenic activity and promoting obesity. Finally, we demonstrate an in vivo requirement of the Zfp516-LSD1 interaction for LSD1 function in BAT gene activation.


Subject(s)
Adipose Tissue, Brown/metabolism , Histone Demethylases/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Uncoupling Protein 1/genetics , 3T3-L1 Cells , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Cold Temperature , HEK293 Cells , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protein Binding , Thermogenesis/genetics
20.
Mol Cell Biol ; 36(14): 1961-76, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27185873

ABSTRACT

The role of AMP-activated protein kinase (AMPK) in promoting fatty acid (FA) oxidation in various tissues, such as liver and muscle, has been well understood. However, the role of AMPK in lipolysis and FA metabolism in adipose tissue has been controversial. To investigate the role of AMPK in the regulation of adipose lipolysis in vivo, we generated mice with adipose-tissue-specific knockout of both the α1 and α2 catalytic subunits of AMPK (AMPK-ASKO mice) by using aP2-Cre and adiponectin-Cre. Both models of AMPK-ASKO ablation show no changes in desnutrin/ATGL levels but have defective phosphorylation of desnutrin/ATGL at S406 to decrease its triacylglycerol (TAG) hydrolase activity, lowering basal lipolysis in adipose tissue. These mice also show defective phosphorylation of hormone-sensitive lipase (HSL) at S565, with higher phosphorylation at protein kinase A sites S563 and S660, increasing its hydrolase activity and isoproterenol-stimulated lipolysis. With higher overall adipose lipolysis, both models of AMPK-ASKO mice are lean, having smaller adipocytes with lower TAG and higher intracellular free-FA levels. Moreover, FAs from higher lipolysis activate peroxisome proliferator-activated receptor delta to induce FA oxidative genes and increase FA oxidation and energy expenditure. Overall, for the first time, we provide in vivo evidence of the role of AMPK in the phosphorylation and regulation of desnutrin/ATGL and HSL and thus adipose lipolysis.


Subject(s)
AMP-Activated Protein Kinases/genetics , Fatty Acids/metabolism , Lipase/metabolism , Sterol Esterase/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/metabolism , Animals , Gene Knockout Techniques , Lipid Metabolism , Lipolysis , Mice , Oxidation-Reduction , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL