Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Adv ; 7(44): eabh2007, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34705514

ABSTRACT

Circadian disruption, as occurs in shift work, is associated with metabolic diseases often attributed to a discordance between internal clocks and environmental timekeepers. REV-ERB nuclear receptors are key components of the molecular clock, but their specific role in the SCN master clock is unknown. We report here that mice lacking circadian REV-ERB nuclear receptors in the SCN maintain free-running locomotor and metabolic rhythms, but these rhythms are notably shortened by 3 hours. When housed under a 24-hour light:dark cycle and fed an obesogenic diet, these mice gained excess weight and accrued more liver fat than controls. These metabolic disturbances were corrected by matching environmental lighting to the shortened endogenous 21-hour clock period, which decreased food consumption. Thus, SCN REV-ERBs are not required for rhythmicity but determine the free-running period length. Moreover, these results support the concept that dissonance between environmental conditions and endogenous time periods causes metabolic disruption.

2.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33021965

ABSTRACT

Obesity occurs when energy expenditure is outweighed by energy intake. Tuberal hypothalamic nuclei, including the arcuate nucleus (ARC), ventromedial nucleus (VMH), and dorsomedial nucleus (DMH), control food intake and energy expenditure. Here we report that, in contrast with females, male mice lacking circadian nuclear receptors REV-ERBα and -ß in the tuberal hypothalamus (HDKO mice) gained excessive weight on an obesogenic high-fat diet due to both decreased energy expenditure and increased food intake during the light phase. Moreover, rebound food intake after fasting was markedly increased in HDKO mice. Integrative transcriptomic and cistromic analyses revealed that such disruption in feeding behavior was due to perturbed REV-ERB-dependent leptin signaling in the ARC. Indeed, in vivo leptin sensitivity was impaired in HDKO mice on an obesogenic diet in a diurnal manner. Thus, REV-ERBs play a crucial role in hypothalamic control of food intake and diurnal leptin sensitivity in diet-induced obesity.


Subject(s)
Circadian Rhythm , Diet/adverse effects , Hypothalamus/metabolism , Leptin/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Obesity/metabolism , Signal Transduction , Animals , Female , Leptin/genetics , Male , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Obesity/chemically induced , Obesity/genetics
3.
Proc Natl Acad Sci U S A ; 116(37): 18691-18699, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451658

ABSTRACT

Ambient temperature influences the molecular clock and lipid metabolism, but the impact of chronic cold exposure on circadian lipid metabolism in thermogenic brown adipose tissue (BAT) has not been studied. Here we show that during chronic cold exposure (1 wk at 4 °C), genes controlling de novo lipogenesis (DNL) including Srebp1, the master transcriptional regulator of DNL, acquired high-amplitude circadian rhythms in thermogenic BAT. These conditions activated mechanistic target of rapamycin 1 (mTORC1), an inducer of Srebp1 expression, and engaged circadian transcriptional repressors REV-ERBα and ß as rhythmic regulators of Srebp1 in BAT. SREBP was required in BAT for the thermogenic response to norepinephrine, and depletion of SREBP prevented maintenance of body temperature both during circadian cycles as well as during fasting of chronically cold mice. By contrast, deletion of REV-ERBα and ß in BAT allowed mice to maintain their body temperature in chronic cold. Thus, the environmental challenge of prolonged noncircadian exposure to cold temperature induces circadian induction of SREBP1 that drives fuel synthesis in BAT and is necessary to maintain circadian body temperature during chronic cold exposure. The requirement for BAT fatty acid synthesis has broad implications for adaptation to cold.


Subject(s)
Acclimatization , Adipose Tissue, Brown/metabolism , Circadian Rhythm/physiology , Lipogenesis/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Thermogenesis/genetics , Animals , Body Temperature , Cold Temperature/adverse effects , Gene Expression Regulation/physiology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Models, Animal , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
4.
Genes Dev ; 31(12): 1202-1211, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28747429

ABSTRACT

Liver lipid metabolism is under intricate temporal control by both the circadian clock and feeding. The interplay between these two mechanisms is not clear. Here we show that liver-specific depletion of nuclear receptors RORα and RORγ, key components of the molecular circadian clock, up-regulate expression of lipogenic genes only under fed conditions at Zeitgeber time 22 (ZT22) but not under fasting conditions at ZT22 or ad libitum conditions at ZT10. RORα/γ controls circadian expression of Insig2, which keeps feeding-induced SREBP1c activation under check. Loss of RORα/γ causes overactivation of the SREBP-dependent lipogenic response to feeding, exacerbating diet-induced hepatic steatosis. These findings thus establish ROR/INSIG2/SREBP as a molecular pathway by which circadian clock components anticipatorily regulate lipogenic responses to feeding. This highlights the importance of time of day as a consideration in the treatment of liver metabolic disorders.


Subject(s)
Circadian Clocks/genetics , Gene Expression Regulation , Lipogenesis/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , Feeding Behavior/physiology , Gene Knockout Techniques , Lipid Metabolism/genetics , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Transcriptional Activation
5.
Nature ; 546(7659): 544-548, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614293

ABSTRACT

Brown adipose tissue is a thermogenic organ that dissipates chemical energy as heat to protect animals against hypothermia and to counteract metabolic disease. However, the transcriptional mechanisms that determine the thermogenic capacity of brown adipose tissue before environmental cold are unknown. Here we show that histone deacetylase 3 (HDAC3) is required to activate brown adipose tissue enhancers to ensure thermogenic aptitude. Mice with brown adipose tissue-specific genetic ablation of HDAC3 become severely hypothermic and succumb to acute cold exposure. Uncoupling protein 1 (UCP1) is nearly absent in brown adipose tissue lacking HDAC3, and there is also marked downregulation of mitochondrial oxidative phosphorylation genes resulting in diminished mitochondrial respiration. Remarkably, although HDAC3 acts canonically as a transcriptional corepressor, it functions as a coactivator of oestrogen-related receptor α (ERRα) in brown adipose tissue. HDAC3 coactivation of ERRα is mediated by deacetylation of PGC-1α and is required for the transcription of Ucp1, Ppargc1a (encoding PGC-1α), and oxidative phosphorylation genes. Importantly, HDAC3 promotes the basal transcription of these genes independently of adrenergic stimulation. Thus, HDAC3 uniquely primes Ucp1 and the thermogenic transcriptional program to maintain a critical capacity for thermogenesis in brown adipose tissue that can be rapidly engaged upon exposure to dangerously cold temperature.


Subject(s)
Adipose Tissue, Brown/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Thermogenesis , Animals , Cell Respiration , Cold Temperature , Enhancer Elements, Genetic/genetics , Hot Temperature , Humans , Male , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Estrogen/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , ERRalpha Estrogen-Related Receptor
6.
J Clin Invest ; 127(4): 1451-1462, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28240605

ABSTRACT

Obesity causes insulin resistance, and PPARγ ligands such as rosiglitazone are insulin sensitizing, yet the mechanisms remain unclear. In C57BL/6 (B6) mice, obesity induced by a high-fat diet (HFD) has major effects on visceral epididymal adipose tissue (eWAT). Here, we report that HFD-induced obesity in B6 mice also altered the activity of gene regulatory elements and genome-wide occupancy of PPARγ. Rosiglitazone treatment restored insulin sensitivity in obese B6 mice, yet, surprisingly, had little effect on gene expression in eWAT. However, in subcutaneous inguinal fat (iWAT), rosiglitazone markedly induced molecular signatures of brown fat, including the key thermogenic gene Ucp1. Obesity-resistant 129S1/SvImJ mice (129 mice) displayed iWAT browning, even in the absence of rosiglitazone. The 129 Ucp1 locus had increased PPARγ binding and gene expression that were preserved in the iWAT of B6x129 F1-intercrossed mice, with an imbalance favoring the 129-derived alleles, demonstrating a cis-acting genetic difference. Thus, B6 mice have genetically defective Ucp1 expression in iWAT. However, when Ucp1 was activated by rosiglitazone, or by iWAT browning in cold-exposed or young mice, expression of the B6 version of Ucp1 was no longer defective relative to the 129 version, indicating epigenomic rescue. These results provide a framework for understanding how environmental influences like drugs can affect the epigenome and potentially rescue genetically determined disease phenotypes.


Subject(s)
Epigenesis, Genetic , Obesity/metabolism , PPAR gamma/physiology , Animals , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Intra-Abdominal Fat/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Protein Binding , Regulatory Elements, Transcriptional , Rosiglitazone , Subcutaneous Fat, Abdominal/metabolism , Thiazolidinediones/pharmacology , Transcriptional Activation , Transcriptome , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
7.
J Biol Chem ; 291(20): 10867-75, 2016 May 13.
Article in English | MEDLINE | ID: mdl-27002153

ABSTRACT

FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified ßKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of ßKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Fibroblast Growth Factors/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Signal Transduction/physiology , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Female , Fibroblast Growth Factors/genetics , Klotho Proteins , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , PPAR gamma/genetics , PPAR gamma/metabolism
8.
Proc Natl Acad Sci U S A ; 110(46): 18656-61, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24167256

ABSTRACT

Adipose tissue is an important metabolic organ, the dysfunction of which is associated with the development of obesity, diabetes mellitus, and cardiovascular disease. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is considered the master regulator of adipocyte differentiation and function. Although its cell-autonomous role in adipogenesis has been clearly demonstrated in cell culture, previous fat-specific knockouts of the murine PPARγ gene did not demonstrate a dramatic phenotype in vivo. Here, using Adipoq-Cre mice to drive adipose-specific recombination, we report a unique fat-specific PPARγ knockout (PPARγ FKO) mouse model with almost no visible brown and white adipose tissue at age 3 mo. As a consequence, PPARγ FKO mice had hugely enlarged pancreatic islets, massive fatty livers, and dramatically elevated levels of blood glucose and serum insulin accompanied by extreme insulin resistance. PPARγ FKO mice also exhibited delayed hair coat formation associated with absence of dermal fat, disrupted mammary gland development with loss of mammary fat pads, and high bone mass with loss of bone marrow fat, indicating the critical roles of adipose PPARγ in these tissues. Together, our data reveal the necessity of fat PPARγ in adipose formation, whole-body metabolic homeostasis, and normal development of fat-containing tissues.


Subject(s)
Adipocytes/metabolism , Insulin Resistance/genetics , Obesity/metabolism , PPAR gamma/deficiency , Animals , Azo Compounds , Immunoblotting , Immunohistochemistry , Insulin Resistance/physiology , Mice , PPAR gamma/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
9.
Mol Endocrinol ; 27(1): 127-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23192980

ABSTRACT

Adipose-specific gene deletion in mice is crucial in determining gene function in adipocyte homeostasis and the development of obesity. We noted 100% mortality when the Hdac3 gene was conditionally deleted using Fabp4-Cre mice, the most commonly used model of adipose-targeted Cre recombinase. However, this surprising result was not reproduced using other models of adipose targeting of Cre, including a novel Retn-Cre mouse. These findings underscore the need for caution when interpreting data obtained using Fabp4-Cre mice and should encourage the use of additional or alternative adipose-targeting Cre mouse models before drawing conclusions about in vivo adipocyte-specific functions.


Subject(s)
Adipose Tissue/enzymology , Disease Models, Animal , Gene Deletion , Histone Deacetylases/genetics , Obesity/enzymology , Adipose Tissue/physiopathology , Animals , Epididymis/enzymology , Epididymis/physiopathology , Fatty Acid-Binding Proteins/genetics , Genes, Lethal , Genetic Engineering , Male , Mice , Mice, Transgenic , Obesity/genetics , Organ Specificity , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...