Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nat Metab ; 6(2): 304-322, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38337096

ABSTRACT

Skeletal muscle is dynamically controlled by the balance of protein synthesis and degradation. Here we discover an unexpected function for the transcriptional repressor B cell lymphoma 6 (BCL6) in muscle proteostasis and strength in mice. Skeletal muscle-specific Bcl6 ablation in utero or in adult mice results in over 30% decreased muscle mass and force production due to reduced protein synthesis and increased autophagy, while it promotes a shift to a slower myosin heavy chain fibre profile. Ribosome profiling reveals reduced overall translation efficiency in Bcl6-ablated muscles. Mechanistically, tandem chromatin immunoprecipitation, transcriptomic and translational analyses identify direct BCL6 repression of eukaryotic translation initiation factor 4E-binding protein 1 (Eif4ebp1) and activation of insulin-like growth factor 1 (Igf1) and androgen receptor (Ar). Together, these results uncover a bifunctional role for BCL6 in the transcriptional and translational control of muscle proteostasis.


Subject(s)
Proteostasis , Proto-Oncogene Proteins c-bcl-6 , Transcription Factors , Animals , Mice , Chromatin Immunoprecipitation , Muscle, Skeletal/metabolism , Transcription Factors/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics
2.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35363257

ABSTRACT

The fat-muscle communication regulates metabolism and involves circulating signals like adiponectin. Modulation of this cross-talk could benefit muscle bioenergetics and exercise tolerance in conditions like obesity. Chronic daily intake of exogenous glucocorticoids produces or exacerbates metabolic stress, often leading to obesity. In stark contrast to the daily intake, we discovered that intermittent pulses of glucocorticoids improve dystrophic muscle metabolism. However, the underlying mechanisms, particularly in the context of obesity, are still largely unknown. Here we report that in mice with diet-induced obesity, intermittent once-weekly prednisone increased total and high-molecular weight adiponectin levels and improved exercise tolerance and energy expenditure. These effects were dependent upon adiponectin, as shown by genetic ablation of the adipokine. Upregulation of Adipoq occurred through the glucocorticoid receptor (GR), as this effect was blocked by inducible GR ablation in adipocytes. The treatment increased the muscle metabolic response of adiponectin through the CAMKK2-AMPK cascade. Our study demonstrates that intermittent glucocorticoids produce healthful metabolic remodeling in diet-induced obesity.


Subject(s)
Adiponectin , Exercise Tolerance , Adipocytes/metabolism , Adiponectin/genetics , Animals , Mice , Obesity/metabolism , Prednisone/pharmacology
3.
Nat Metab ; 3(12): 1621-1632, 2021 12.
Article in English | MEDLINE | ID: mdl-34903884

ABSTRACT

In mammals, circadian rhythms are entrained to the light cycle and drive daily oscillations in levels of NAD+, a cosubstrate of the class III histone deacetylase sirtuin 1 (SIRT1) that associates with clock transcription factors. Although NAD+ also participates in redox reactions, the extent to which NAD(H) couples nutrient state with circadian transcriptional cycles remains unknown. Here we show that nocturnal animals subjected to time-restricted feeding of a calorie-restricted diet (TRF-CR) only during night-time display reduced body temperature and elevated hepatic NADH during daytime. Genetic uncoupling of nutrient state from NADH redox state through transduction of the water-forming NADH oxidase from Lactobacillus brevis (LbNOX) increases daytime body temperature and blood and liver acyl-carnitines. LbNOX expression in TRF-CR mice induces oxidative gene networks controlled by brain and muscle Arnt-like protein 1 (BMAL1) and peroxisome proliferator-activated receptor alpha (PPARα) and suppresses amino acid catabolic pathways. Enzymatic analyses reveal that NADH inhibits SIRT1 in vitro, corresponding with reduced deacetylation of SIRT1 substrates during TRF-CR in vivo. Remarkably, Sirt1 liver nullizygous animals subjected to TRF-CR display persistent hypothermia even when NADH is oxidized by LbNOX. Our findings reveal that the hepatic NADH cycle links nutrient state to whole-body energetics through the rhythmic regulation of SIRT1.


Subject(s)
Energy Metabolism , Fasting , NAD/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcription, Genetic , Amino Acids/metabolism , Animals , Body Temperature , Circadian Rhythm , Diet , Fatty Acids/metabolism , Gene Expression Regulation , Liver/metabolism , Mice , Transcription Factors
4.
Hepatology ; 74(6): 3362-3375, 2021 12.
Article in English | MEDLINE | ID: mdl-34170527

ABSTRACT

BACKGROUND AND AIMS: The unfolded protein response (UPR) is a coordinated cellular response to endoplasmic reticulum (ER) stress that functions to maintain cellular homeostasis. When ER stress is unresolved, the UPR can trigger apoptosis. Pathways within the UPR influence bile acid metabolism in adult animal models and adult human liver diseases, however, the UPR has not been studied in young animal models or pediatric liver diseases. In this study we sought to determine whether weanling age mice had altered UPR activation compared with adult mice, which could lead to increased bile acid-induced hepatic injury. APPROACH AND RESULTS: We demonstrate that after 7 days of cholic acid (CA) feeding to wild-type animals, weanling age mice have a 2-fold greater serum alanine aminotransferase (ALT) levels compared with adult mice, with increased hepatic apoptosis. Weanling mice fed CA have increased hepatic nuclear X-box binding protein 1 spliced (XBP1s) expression, but cannot increase expression of its protective downstream target's ER DNA J domain-containing protein 4 and ER degradation enhancing α-mannoside. In response to tunicamycin induced ER stress, young mice have blunted expression of several UPR pathways compared with adult mice. CA feeding to adult liver-specific XBP1 knockout (LS-XBP1-/- ) mice, which are unable to resolve hepatic ER stress, leads to increased serum ALT and CCAAT/enhancer binding homologous protein, a proapoptotic UPR molecule, expression to levels similar to CA-fed LS-XBP1-/- weanlings. CONCLUSIONS: Weanling mice have attenuated hepatic XBP1 signaling and impaired UPR activation with resultant increased susceptibility to bile acid-induced injury.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Cholic Acid/adverse effects , Unfolded Protein Response , Animals , Animals, Newborn , Endoplasmic Reticulum Stress/drug effects , Male , Mice , Mice, Inbred C57BL
5.
Cell Rep ; 34(13): 108927, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789109

ABSTRACT

Understanding the epigenomic evolution and specificity of disease subtypes from complex patient data remains a major biomedical problem. We here present DeCET (decomposition and classification of epigenomic tensors), an integrative computational approach for simultaneously analyzing hierarchical heterogeneous data, to identify robust epigenomic differences among tissue types, differentiation states, and disease subtypes. Applying DeCET to our own data from 21 uterine benign tumor (leiomyoma) patients identifies distinct epigenomic features discriminating normal myometrium and leiomyoma subtypes. Leiomyomas possess preponderant alterations in distal enhancers and long-range histone modifications confined to chromatin contact domains that constrain the evolution of pathological epigenomes. Moreover, we demonstrate the power and advantage of DeCET on multiple publicly available epigenomic datasets representing different cancers and cellular states. Epigenomic features extracted by DeCET can thus help improve our understanding of disease states, cellular development, and differentiation, thereby facilitating future therapeutic, diagnostic, and prognostic strategies.


Subject(s)
Epigenome , Leiomyoma/classification , Leiomyoma/genetics , Uterine Neoplasms/classification , Uterine Neoplasms/genetics , Cell Differentiation/genetics , Chromatin/metabolism , Cluster Analysis , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Homeobox , HEK293 Cells , Humans , Leiomyoma/pathology , Myometrium/pathology , Nucleotide Motifs/genetics , Transcription Factors/metabolism , Uterine Neoplasms/pathology
6.
Sci Rep ; 11(1): 430, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33432099

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1) is a functional biomarker of the metabolic syndrome. Previous studies have demonstrated that PAI-1 is a mechanistic contributor to several elements of the syndrome, including obesity, hypertension and insulin resistance. Here we show that PAI-1 is also a critical regulator of hepatic lipid metabolism. RNA sequencing revealed that PAI-1 directly regulates the transcriptional expression of numerous genes involved in mammalian lipid homeostasis, including PCSK9 and FGF21. Pharmacologic or genetic reductions in plasma PAI-1 activity ameliorates hyperlipidemia in vivo. These experimental findings are complemented with the observation that genetic deficiency of PAI-1 is associated with reduced plasma PCSK9 levels in humans. Taken together, our findings identify PAI-1 as a novel contributor to mammalian lipid metabolism and provides a fundamental mechanistic insight into the pathogenesis of one of the most pervasive medical problems worldwide.


Subject(s)
Dyslipidemias/genetics , Fatty Liver/genetics , Plasminogen Activator Inhibitor 1/physiology , Animals , Cells, Cultured , Cohort Studies , Dyslipidemias/metabolism , Fatty Liver/metabolism , Female , Fibroblast Growth Factors/genetics , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proprotein Convertase 9/genetics
7.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31852847

ABSTRACT

In humans, chronic glucocorticoid use is associated with side effects like muscle wasting, obesity, and metabolic syndrome. Intermittent steroid dosing has been proposed in Duchenne Muscular Dystrophy patients to mitigate the side effects seen with daily steroid intake. We evaluated biomarkers from Duchenne Muscular Dystrophy patients, finding that, compared with chronic daily steroid use, weekend steroid use was associated with reduced serum insulin, free fatty acids, and branched chain amino acids, as well as reduction in fat mass despite having similar BMIs. We reasoned that intermittent prednisone administration in dystrophic mice would alter muscle epigenomic signatures, and we identified the coordinated action of the glucocorticoid receptor, KLF15 and MEF2C as mediators of a gene expression program driving metabolic reprogramming and enhanced nutrient utilization. Muscle lacking Klf15 failed to respond to intermittent steroids. Furthermore, coadministration of the histone acetyltransferase inhibitor anacardic acid with steroids in mdx mice eliminated steroid-specific epigenetic marks and abrogated the steroid response. Together, these findings indicate that intermittent, repeated exposure to glucocorticoids promotes performance in dystrophic muscle through an epigenetic program that enhances nutrient utilization.


Subject(s)
Glucocorticoids/administration & dosage , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Prednisone/administration & dosage , Anacardic Acids/administration & dosage , Animals , Biomarkers/blood , Biomarkers/metabolism , Child , Cross-Sectional Studies , Disease Models, Animal , Drug Therapy, Combination , Epigenesis, Genetic/drug effects , Epigenomics , Gene Expression Regulation/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MEF2 Transcription Factors/metabolism , Male , Metabolomics , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Nutrients/blood , Nutrients/metabolism , Pulse Therapy, Drug
8.
Mol Cell ; 76(4): 531-545.e5, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31706703

ABSTRACT

The glucocorticoid receptor (GR) is a potent metabolic regulator and a major drug target. While GR is known to play integral roles in circadian biology, its rhythmic genomic actions have never been characterized. Here we mapped GR's chromatin occupancy in mouse livers throughout the day and night cycle. We show how GR partitions metabolic processes by time-dependent target gene regulation and controls circulating glucose and triglycerides differentially during feeding and fasting. Highlighting the dominant role GR plays in synchronizing circadian amplitudes, we find that the majority of oscillating genes are bound by and depend on GR. This rhythmic pattern is altered by high-fat diet in a ligand-independent manner. We find that the remodeling of oscillatory gene expression and postprandial GR binding results from a concomitant increase of STAT5 co-occupancy in obese mice. Altogether, our findings highlight GR's fundamental role in the rhythmic orchestration of hepatic metabolism.


Subject(s)
Chromatin/metabolism , Circadian Clocks , Circadian Rhythm , Diet, High-Fat , Dietary Fats/metabolism , Energy Metabolism , Liver/metabolism , Obesity/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Blood Glucose/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Dietary Fats/administration & dosage , Dietary Fats/blood , Disease Models, Animal , Energy Metabolism/genetics , Fasting/metabolism , Gene Expression Regulation , Glucocorticoids/metabolism , Gluconeogenesis , Ligands , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , Postprandial Period , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Secretory Pathway , Signal Transduction , Time Factors , Transcription, Genetic , Triglycerides/blood
9.
PLoS Biol ; 17(10): e3000467, 2019 10.
Article in English | MEDLINE | ID: mdl-31589602

ABSTRACT

Skeletal muscles consist of fibers of differing metabolic activities and contractility, which become remodeled in response to chronic exercise, but the epigenomic basis for muscle identity and adaptation remains poorly understood. Here, we used chromatin immunoprecipitation sequencing of dimethylated histone 3 lysine 4 and acetylated histone 3 lysine 27 as well as transposase-accessible chromatin profiling to dissect cis-regulatory networks across muscle groups. We demonstrate that in vivo enhancers specify muscles in accordance with myofiber composition, show little resemblance to cultured myotube enhancers, and identify glycolytic and oxidative muscle-specific regulators. Moreover, we find that voluntary wheel running and muscle-specific peroxisome proliferator-activated receptor gamma coactivator-1 alpha (Pgc1a) transgenic (mTg) overexpression, which stimulate endurance performance in mice, result in markedly different changes to the epigenome. Exercise predominantly leads to enhancer hypoacetylation, whereas mTg causes hyperacetylation at different sites. Integrative analysis of regulatory regions and gene expression revealed that exercise and mTg are each associated with myocyte enhancer factor (MEF) 2 and estrogen-related receptor (ERR) signaling and transcription of genes promoting oxidative metabolism. However, exercise was additionally associated with regulation by retinoid X receptor (RXR), jun proto-oncogene (JUN), sine oculis homeobox factor (SIX), and other factors. Overall, our work defines the unique enhancer repertoires of skeletal muscles in vivo and reveals that divergent exercise-induced or PGC1α-driven epigenomic programs direct partially convergent transcriptional networks.


Subject(s)
Epigenesis, Genetic , Histones/genetics , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Physical Conditioning, Animal , Acetylation , Animals , Cellular Reprogramming , Chromatin/chemistry , Chromatin/metabolism , Enhancer Elements, Genetic , Glycolysis/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Cells/cytology , Muscle, Skeletal/cytology , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Signal Transduction , ERRalpha Estrogen-Related Receptor
10.
Elife ; 82019 04 15.
Article in English | MEDLINE | ID: mdl-30983568

ABSTRACT

Transcription is tightly regulated to maintain energy homeostasis during periods of feeding or fasting, but the molecular factors that control these alternating gene programs are incompletely understood. Here, we find that the B cell lymphoma 6 (BCL6) repressor is enriched in the fed state and converges genome-wide with PPARα to potently suppress the induction of fasting transcription. Deletion of hepatocyte Bcl6 enhances lipid catabolism and ameliorates high-fat-diet-induced steatosis. In Ppara-null mice, hepatocyte Bcl6 ablation restores enhancer activity at PPARα-dependent genes and overcomes defective fasting-induced fatty acid oxidation and lipid accumulation. Together, these findings identify BCL6 as a negative regulator of oxidative metabolism and reveal that alternating recruitment of repressive and activating transcription factors to shared cis-regulatory regions dictates hepatic lipid handling.


Subject(s)
Fasting , Fatty Liver/physiopathology , Gene Expression Regulation , Liver/physiology , Proto-Oncogene Proteins c-bcl-6/metabolism , Animals , Gene Deletion , Lipid Metabolism , Mice , Proto-Oncogene Proteins c-bcl-6/deficiency
11.
Cell Rep ; 25(12): 3283-3298.e6, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566857

ABSTRACT

Accumulation of visceral adiposity is directly linked to the morbidity of obesity, while subcutaneous body fat is considered more benign. We have identified an unexpected role for B cell lymphoma 6 (BCL6), a critical regulator of immunity, in the developmental expansion of subcutaneous adipose tissue. In adipocyte-specific knockout mice (Bcl6AKO), we found that Bcl6 deletion results in strikingly increased inguinal, but not perigonadal, adipocyte size and tissue mass in addition to marked insulin sensitivity. Genome-wide RNA expression and DNA binding analyses revealed that BCL6 controls gene networks involved in cell growth and fatty acid biosynthesis. Using deuterium label incorporation and comprehensive adipokine and lipid profiling, we discovered that ablation of adipocyte Bcl6 enhances subcutaneous adipocyte lipogenesis, increases levels of adiponectin and fatty acid esters of hydroxy fatty acids (FAHFAs), and prevents steatosis. Thus, our studies identify BCL6 as a negative regulator of subcutaneous adipose tissue expansion and metabolic health.


Subject(s)
Insulin Resistance , Obesity/genetics , Obesity/pathology , Proto-Oncogene Proteins c-bcl-6/metabolism , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adiponectin/blood , Adipose Tissue, Brown/metabolism , Adiposity , Animals , Cell Differentiation/genetics , DNA/metabolism , Diet, High-Fat , Fatty Liver/pathology , Fetus/metabolism , Gene Expression Regulation , Humans , Inflammation/pathology , Insulin/metabolism , Insulin Resistance/genetics , Lipids/biosynthesis , Lipogenesis/genetics , Male , Mice , Mice, Knockout , Obesity/blood , Protein Binding , Proto-Oncogene Proteins c-bcl-6/deficiency , Signal Transduction , Subcutaneous Fat/metabolism
12.
Genes Dev ; 32(21-22): 1367-1379, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366905

ABSTRACT

The mammalian circadian clock is encoded by an autoregulatory transcription feedback loop that drives rhythmic behavior and gene expression in the brain and peripheral tissues. Transcriptomic analyses indicate cell type-specific effects of circadian cycles on rhythmic physiology, although how clock cycles respond to environmental stimuli remains incompletely understood. Here, we show that activation of the inducible transcription factor NF-κB in response to inflammatory stimuli leads to marked inhibition of clock repressors, including the Period, Cryptochrome, and Rev-erb genes, within the negative limb. Furthermore, activation of NF-κB relocalizes the clock components CLOCK/BMAL1 genome-wide to sites convergent with those bound by NF-κB, marked by acetylated H3K27, and enriched in RNA polymerase II. Abrogation of NF-κB during adulthood alters the expression of clock repressors, disrupts clock-controlled gene cycles, and impairs rhythmic activity behavior, revealing a role for NF-κB in both unstimulated and activated conditions. Together, these data highlight NF-κB-mediated transcriptional repression of the clock feedback limb as a cause of circadian disruption in response to inflammation.


Subject(s)
Circadian Rhythm/genetics , NF-kappa B/physiology , ARNTL Transcription Factors/metabolism , Animals , Behavior, Animal , CLOCK Proteins/metabolism , Cell Line , Chromatin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Repressor Proteins/metabolism , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 115(21): E4910-E4919, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29735694

ABSTRACT

Mitochondrial dysfunction is increasingly recognized as a critical determinant of both hereditary and acquired kidney diseases. However, it remains poorly understood how mitochondrial metabolism is regulated to support normal kidney function and how its dysregulation contributes to kidney disease. Here, we show that the nuclear receptor estrogen-related receptor gamma (ERRγ) and hepatocyte nuclear factor 1 beta (HNF1ß) link renal mitochondrial and reabsorptive functions through coordinated epigenomic programs. ERRγ directly regulates mitochondrial metabolism but cooperatively controls renal reabsorption via convergent binding with HNF1ß. Deletion of ERRγ in renal epithelial cells (RECs), in which it is highly and specifically expressed, results in severe renal energetic and reabsorptive dysfunction and progressive renal failure that recapitulates phenotypes of animals and patients with HNF1ß loss-of-function gene mutations. Moreover, ERRγ expression positively correlates with renal function and is decreased in patients with chronic kidney disease (CKD). REC-ERRγ KO mice share highly overlapping renal transcriptional signatures with human patients with CKD. Together these findings reveal a role for ERRγ in directing independent and HNF1ß-integrated programs for energy production and use essential for normal renal function and the prevention of kidney disease.


Subject(s)
Cysts/prevention & control , Energy Metabolism , Epigenomics , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/genetics , Receptors, Estrogen/genetics , Renal Insufficiency, Chronic/prevention & control , Animals , Cysts/metabolism , Cysts/pathology , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocyte Nuclear Factor 1-beta/physiology , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
14.
Cell ; 165(1): 165-179, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26924576

ABSTRACT

Much has been learned about transcriptional cascades and networks from large-scale systems analyses of high-throughput datasets. However, analysis methods that optimize statistical power through simultaneous evaluation of thousands of ChIP-seq peaks or differentially expressed genes possess substantial limitations in their ability to uncover mechanistic principles of transcriptional control. By examining nascent transcript RNA-seq, ChIP-seq, and binding motif datasets from lipid A-stimulated macrophages with increased attention to the quantitative distribution of signals, we identified unexpected relationships between the in vivo binding properties of inducible transcription factors, motif strength, and transcription. Furthermore, rather than emphasizing common features of large clusters of co-regulated genes, our results highlight the extent to which unique mechanisms regulate individual genes with key biological functions. Our findings demonstrate the mechanistic value of stringent interrogation of well-defined sets of genes as a complement to broader systems analyses of transcriptional cascades and networks.


Subject(s)
Gene Regulatory Networks , Inflammation/genetics , Inflammation/immunology , Animals , Lipid A/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Receptor, Interferon alpha-beta/metabolism , Serum Response Factor/metabolism
15.
Science ; 350(6261): aac4250, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26542580

ABSTRACT

The mammalian transcription factors CLOCK and BMAL1 are essential components of the molecular clock that coordinate behavior and metabolism with the solar cycle. Genetic or environmental perturbation of circadian cycles contributes to metabolic disorders including type 2 diabetes. To study the impact of the cell-autonomous clock on pancreatic ß cell function, we examined pancreatic islets from mice with either intact or disrupted BMAL1 expression both throughout life and limited to adulthood. We found pronounced oscillation of insulin secretion that was synchronized with the expression of genes encoding secretory machinery and signaling factors that regulate insulin release. CLOCK/BMAL1 colocalized with the pancreatic transcription factor PDX1 within active enhancers distinct from those controlling rhythmic metabolic gene networks in liver. We also found that ß cell clock ablation in adult mice caused severe glucose intolerance. Thus, cell type-specific enhancers underlie the circadian control of peripheral metabolism throughout life and may help to explain its dysregulation in diabetes.


Subject(s)
Circadian Rhythm/genetics , Enhancer Elements, Genetic/physiology , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulin/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Exocytosis/genetics , Glucose Intolerance , Homeodomain Proteins/metabolism , Humans , Insulin Secretion , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Trans-Activators/metabolism , Transcription, Genetic
16.
Cell Metab ; 21(4): 628-36, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25863252

ABSTRACT

Neurons utilize mitochondrial oxidative phosphorylation (OxPhos) to generate energy essential for survival, function, and behavioral output. Unlike most cells that burn both fat and sugar, neurons only burn sugar. Despite its importance, how neurons meet the increased energy demands of complex behaviors such as learning and memory is poorly understood. Here we show that the estrogen-related receptor gamma (ERRγ) orchestrates the expression of a distinct neural gene network promoting mitochondrial oxidative metabolism that reflects the extraordinary neuronal dependence on glucose. ERRγ(-/-) neurons exhibit decreased metabolic capacity. Impairment of long-term potentiation (LTP) in ERRγ(-/-) hippocampal slices can be fully rescued by the mitochondrial OxPhos substrate pyruvate, functionally linking the ERRγ knockout metabolic phenotype and memory formation. Consistent with this notion, mice lacking neuronal ERRγ in cerebral cortex and hippocampus exhibit defects in spatial learning and memory. These findings implicate neuronal ERRγ in the metabolic adaptations required for memory formation.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Mitochondria/metabolism , Neurons/metabolism , Receptors, Estrogen/metabolism , Analysis of Variance , Animals , Chromatin Immunoprecipitation , Galactosides , Gene Knockout Techniques , Glycolysis/physiology , Hippocampus/metabolism , Indoles , Memory/physiology , Mice , Microarray Analysis , Pyruvic Acid , Real-Time Polymerase Chain Reaction , Spatial Learning/physiology
17.
FASEB J ; 29(2): 636-49, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25392268

ABSTRACT

Adiponectin (APN), a pleiotropic adipokine that exerts anti-inflammatory, antidiabetic, and antiatherogenic effects through its receptors (AdipoRs), AdipoR1 and AdipoR2, is an important therapeutic target. Factors regulating AdipoR expression in monocyte/macrophages are poorly understood, and the significance of polarized macrophage activation in controlling AdipoR expression and the APN-mediated inflammatory response has not been investigated. The aim of this study was to investigate whether the macrophage polarization phenotype controls the AdipoR expression and APN-mediated inflammatory response. With the use of mouse bone marrow and peritoneal macrophages, we demonstrate that classical activation (M1) of macrophages suppressed (40-60% of control) AdipoR expression, whereas alternative activation (M2) preserved it. Remarkably, the macrophage polarization phenotypes produced contrasting inflammatory responses to APN (EC50 5 µg/ml). In M1 macrophages, APN induced proinflammatory cytokines, TNF-α, IL-6, and IL-12 (>10-fold of control) and AdipoR levels. In contrast, in M2 macrophages, APN induced the anti-inflammatory cytokine IL-10 without altering AdipoR expression. Furthermore, M1 macrophages adapt to a cytokine environment by reversing AdipoR expression. APN induced AdipoR mRNA and protein expression by up-regulating liver X receptor-α (LXRα) in macrophages. These results provide the first evidence that macrophage polarization is a key determinant regulating AdipoR expression and differential APN-mediated macrophage inflammatory responses, which can profoundly influence their pathogenic role in inflammatory and metabolic disorders.


Subject(s)
Adiponectin/metabolism , Macrophages/cytology , Receptors, Adiponectin/metabolism , Animals , Atherosclerosis , Cell Line , Cytokines/metabolism , Gene Expression Regulation , Humans , Inflammation , Insulin Resistance , Interleukin-10/metabolism , Liver X Receptors , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Orphan Nuclear Receptors/metabolism , Phenotype
18.
Mol Cell ; 54(4): 613-25, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24793694

ABSTRACT

Upon androgen stimulation, PKN1-mediated histone H3 threonine 11 phosphorylation (H3T11P) promotes AR target gene activation. However, the underlying mechanism is not completely understood. Here, we show that WDR5, a subunit of the SET1/MLL complex, interacts with H3T11P, and this interaction facilitates the recruitment of the MLL1 complex and subsequent H3K4 tri-methylation (H3K4me3). Using ChIP-seq, we find that androgen stimulation results in a 6-fold increase in the number of H3T11P-marked regions and induces WDR5 colocalization to one third of H3T11P-enriched promoters, thus establishing a genome-wide relationship between H3T11P and recruitment of WDR5. Accordingly, PKN1 knockdown or chemical inhibition severely blocks WDR5 chromatin association and H3K4me3 on AR target genes. Finally, WDR5 is critical in prostate cancer cell proliferation and is hyperexpressed in human prostate cancers. Together, these results identify WDR5 as a critical epigenomic integrator of histone phosphorylation and methylation and as a major driver of androgen-dependent prostate cancer cell proliferation.


Subject(s)
Androgens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Prostatic Neoplasms/metabolism , Protein Kinase C/metabolism , Receptors, Androgen/metabolism , Threonine/metabolism , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Methylation , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase C/genetics , Receptors, Androgen/genetics , Signal Transduction , Threonine/genetics
19.
PLoS One ; 9(1): e86404, 2014.
Article in English | MEDLINE | ID: mdl-24466075

ABSTRACT

Adiponectin (APN), an adipocytokine produced by adipose tissue, exerts pleiotropic actions regulating inflammation, metabolism and vascular homeostasis. APN levels are inversely correlated with obesity, type-2 diabetes, hypertension and cardiovascular disease. Although renin angiotensin system (RAS) activation in these interrelated metabolic syndrome components increases angiotensin II (AngII) levels leading to vascular damage, it is unknown whether APN under these conditions provides atheroprotection. We investigated whether increasing plasma APN provides atheroprotection in a hypertensive and accelerated atherosclerosis model. Using adenoviral gene transfer, sustained APN expression increased plasma levels of total and high-molecular weight APN, leading to a significant elevation of plasma HDL-cholesterol (HDL-C). Elevated APN levels were strongly atheroprotective, yet had no impact on blood pressure. Notably, gene expression analyses revealed that APN significantly inhibited the expression of pro-inflammatory and atherogenic genes while it increased the expression of the anti-inflammatory cytokine, IL-10 and the cholesterol efflux transporters, ABCA1 and ABCG1 in the artery wall. These findings suggest that increasing APN levels may be an effective therapeutic strategy to inhibit vascular inflammation and accelerated atherosclerosis associated with RAS activation in the metabolic syndrome.


Subject(s)
Adiponectin/genetics , Angiotensin II/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Gene Expression , Inflammation/genetics , Inflammation/metabolism , Adiponectin/blood , Adiponectin/chemistry , Adiponectin/metabolism , Angiotensin II/adverse effects , Animals , Apolipoproteins/genetics , Apolipoproteins/metabolism , Atherosclerosis/pathology , Blood Pressure/genetics , Cholesterol/metabolism , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Protein Multimerization , Protein Transport , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Receptors, LDL/genetics , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism
20.
Proc Natl Acad Sci U S A ; 110(47): 18820-5, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24191050

ABSTRACT

Molecular targeting of the two receptor interaction domains of the epigenetic repressor silencing mediator of retinoid and thyroid hormone receptors (SMRT(mRID)) produced a transplantable skeletal syndrome that reduced radial bone growth, increased numbers of bone-resorbing periosteal osteoclasts, and increased bone fracture risk. Furthermore, SMRT(mRID) mice develop spontaneous primary myelofibrosis, a chronic, usually idiopathic disorder characterized by progressive bone marrow fibrosis. Frequently linked to polycythemia vera and chronic myeloid leukemia, myelofibrosis displays high patient morbidity and mortality, and current treatment is mostly palliative. To decipher the etiology of this disease, we identified the thrombopoietin (Tpo) gene as a target of the SMRT-retinoic acid receptor signaling pathway in bone marrow stromal cells. Chronic induction of Tpo in SMRT(mRID) mice results in up-regulation of TGF-ß and PDGF in megakaryocytes, uncontrolled proliferation of bone marrow reticular cells, and fibrosis of the marrow compartment. Of therapeutic relevance, we show that this syndrome can be rescued by retinoid antagonists, demonstrating that the physical interface between SMRT and retinoic acid receptor can be a potential therapeutic target to block primary myelofibrosis disease progression.


Subject(s)
Bone Marrow/metabolism , Cytokines/metabolism , Epigenetic Repression/physiology , Nuclear Receptor Co-Repressor 2/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Signal Transduction/physiology , Thrombopoietin/genetics , Alkaline Phosphatase/blood , Animals , Benzothiazoles , Calcium/blood , Cell Proliferation/drug effects , DNA Primers/genetics , Diamines , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Knock-In Techniques , Luciferases , Megakaryocytes/metabolism , Mice , Nuclear Receptor Co-Repressor 2/genetics , Organic Chemicals , Platelet-Derived Growth Factor/metabolism , Polymerase Chain Reaction , Primary Myelofibrosis/etiology , Quinolines , Thrombopoietin/biosynthesis , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL