Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Science ; 384(6695): 563-572, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38696572

ABSTRACT

A molecular clock network is crucial for daily physiology and maintaining organismal health. We examined the interactions and importance of intratissue clock networks in muscle tissue maintenance. In arrhythmic mice showing premature aging, we created a basic clock module involving a central and a peripheral (muscle) clock. Reconstituting the brain-muscle clock network is sufficient to preserve fundamental daily homeostatic functions and prevent premature muscle aging. However, achieving whole muscle physiology requires contributions from other peripheral clocks. Mechanistically, the muscle peripheral clock acts as a gatekeeper, selectively suppressing detrimental signals from the central clock while integrating important muscle homeostatic functions. Our research reveals the interplay between the central and peripheral clocks in daily muscle function and underscores the impact of eating patterns on these interactions.


Subject(s)
Aging, Premature , Aging , Brain , Circadian Rhythm , Muscle, Skeletal , Animals , Male , Mice , Aging/genetics , Aging/physiology , Aging, Premature/genetics , Aging, Premature/prevention & control , Brain/physiology , Circadian Clocks/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Homeostasis , Muscle, Skeletal/physiology , Mice, Knockout , ARNTL Transcription Factors/genetics
2.
Cell Stem Cell ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38701785

ABSTRACT

In mammals, the circadian clock network drives daily rhythms of tissue-specific homeostasis. To dissect daily inter-tissue communication, we constructed a mouse minimal clock network comprising only two nodes: the peripheral epidermal clock and the central brain clock. By transcriptomic and functional characterization of this isolated connection, we identified a gatekeeping function of the peripheral tissue clock with respect to systemic inputs. The epidermal clock concurrently integrates and subverts brain signals to ensure timely execution of epidermal daily physiology. Timely cell-cycle termination in the epidermal stem cell compartment depends upon incorporation of clock-driven signals originating from the brain. In contrast, the epidermal clock corrects or outcompetes potentially disruptive feeding-related signals to ensure the optimal timing of DNA replication. Together, we present an approach for cataloging the systemic dependencies of daily temporal organization in a tissue and identify an essential gate-keeping function of peripheral circadian clocks that guarantees tissue homeostasis.

3.
Sci Adv ; 10(1): eadi2689, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38170777

ABSTRACT

Defects in adipocyte lipolysis drive multiple aspects of cardiometabolic disease, but the transcriptional framework controlling this process has not been established. To address this, we performed a targeted perturbation screen in primary human adipocytes. Our analyses identified 37 transcriptional regulators of lipid mobilization, which we classified as (i) transcription factors, (ii) histone chaperones, and (iii) mRNA processing proteins. On the basis of its strong relationship with multiple readouts of lipolysis in patient samples, we performed mechanistic studies on one hit, ZNF189, which encodes the zinc finger protein 189. Using mass spectrometry and chromatin profiling techniques, we show that ZNF189 interacts with the tripartite motif family member TRIM28 and represses the transcription of an adipocyte-specific isoform of phosphodiesterase 1B (PDE1B2). The regulation of lipid mobilization by ZNF189 requires PDE1B2, and the overexpression of PDE1B2 is sufficient to attenuate hormone-stimulated lipolysis. Thus, our work identifies the ZNF189-PDE1B2 axis as a determinant of human adipocyte lipolysis and highlights a link between chromatin architecture and lipid mobilization.


Subject(s)
Adipocytes , Lipid Mobilization , Humans , Adipocytes/metabolism , Lipolysis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/genetics , Chromatin/metabolism
4.
Trends Endocrinol Metab ; 34(12): 813-822, 2023 12.
Article in English | MEDLINE | ID: mdl-37716877

ABSTRACT

A thorough understanding of the mechanisms controlling energy homeostasis is needed to prevent and treat metabolic morbidities. While the contribution of organs such as the liver, muscle, adipose tissue, and pancreas to the regulation of energy has received wide attention, less is known about the interplay with the nervous system. Here, we highlight the role of the nervous systems in regulating metabolism beyond the classic hypothalamic endocrine signaling models and discuss the contribution of circadian rhythms, higher brain regions, and sociodemographic variables in the energy equation. We infer that interdisciplinary approaches are key to conceptually advancing the current research frontier and devising innovative therapies to prevent and treat metabolic disease.


Subject(s)
Adipose Tissue , Circadian Rhythm , Humans , Circadian Rhythm/physiology , Homeostasis , Hypothalamus , Brain , Energy Metabolism/physiology
5.
Cell Rep ; 42(6): 112588, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37267101

ABSTRACT

Physiology is regulated by interconnected cell and tissue circadian clocks. Disruption of the rhythms generated by the concerted activity of these clocks is associated with metabolic disease. Here we tested the interactions between clocks in two critical components of organismal metabolism, liver and skeletal muscle, by rescuing clock function either in each organ separately or in both organs simultaneously in otherwise clock-less mice. Experiments showed that individual clocks are partially sufficient for tissue glucose metabolism, yet the connections between both tissue clocks coupled to daily feeding rhythms support systemic glucose tolerance. This synergy relies in part on local transcriptional control of the glucose machinery, feeding-responsive signals such as insulin, and metabolic cycles that connect the muscle and liver. We posit that spatiotemporal mechanisms of muscle and liver play an essential role in the maintenance of systemic glucose homeostasis and that disrupting this diurnal coordination can contribute to metabolic disease.


Subject(s)
Circadian Clocks , Mice , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Liver/metabolism , Muscle, Skeletal/metabolism , Glucose/metabolism
6.
Mol Metab ; 64: 101556, 2022 10.
Article in English | MEDLINE | ID: mdl-35914650

ABSTRACT

OBJECTIVE: The circadian clock aligns physiology with the 24-hour rotation of Earth. Light and food are the main environmental cues (zeitgebers) regulating circadian rhythms in mammals. Yet, little is known about the interaction between specific dietary components and light in coordinating circadian homeostasis. Herein, we focused on the role of essential amino acids. METHODS: Mice were fed diets depleted of specific essential amino acids and their behavioral rhythms were monitored and tryptophan was selected for downstream analyses. The role of tryptophan metabolism in modulating circadian homeostasis was studied using isotope tracing as well as transcriptomic- and metabolomic- analyses. RESULTS: Dietary tryptophan depletion alters behavioral rhythms in mice. Furthermore, tryptophan metabolism was shown to be regulated in a time- and light- dependent manner. A multi-omics approach and combinatory diet/light interventions demonstrated that tryptophan metabolism modulates temporal regulation of metabolism and transcription programs by buffering photic cues. Specifically, tryptophan metabolites regulate central circadian functions of the suprachiasmatic nucleus and the core clock machinery in the liver. CONCLUSIONS: Tryptophan metabolism is a modulator of circadian homeostasis by integrating environmental cues. Our findings propose tryptophan metabolism as a potential point for pharmacologic intervention to modulate phenotypes associated with disrupted circadian rhythms.


Subject(s)
Circadian Clocks , Circadian Rhythm , Animals , Circadian Rhythm/physiology , Liver/metabolism , Mammals , Mice , Suprachiasmatic Nucleus/metabolism , Tryptophan/metabolism
7.
Sci Adv ; 8(26): eabo2896, 2022 07.
Article in English | MEDLINE | ID: mdl-35767612

ABSTRACT

Life on Earth anticipates recurring 24-hour environmental cycles via genetically encoded molecular clocks active in all mammalian organs. Communication between these clocks controls circadian homeostasis. Intertissue communication is mediated, in part, by temporal coordination of metabolism. Here, we characterize the extent to which clocks in different organs control systemic metabolic rhythms, an area that remains largely unexplored. We analyzed the metabolome of serum from mice with tissue-specific expression of the clock gene Bmal1. Having functional hepatic and muscle clocks can only drive a minority (13%) of systemic metabolic rhythms. Conversely, limiting Bmal1 expression to the central pacemaker in the brain restores rhythms to 57% of circulatory metabolites. Rhythmic feeding imposed on clockless mice resulted in a similar rescue, indicating that the central clock mainly regulates metabolic rhythms via behavior. These findings explicate the circadian communication between tissues and highlight the importance of the central clock in governing those signals.

8.
Life Sci ; 303: 120601, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35561749

ABSTRACT

The gut microbiome influences cognition and behavior in mammals, yet its metabolic impact on the brain is only starting to be defined. Using metabolite profiling of antibiotics-treated mice, we reveal the microbiome as a key input controlling circadian metabolic cycles in the brain. Intra and inter-region analyses characterise the influence of the microbiome on the suprachiasmatic nucleus, containing the central clockwork, as well as the hippocampus and cortex, regions involved in learning and behavior.


Subject(s)
Anti-Bacterial Agents , Gastrointestinal Microbiome , Animals , Anti-Bacterial Agents/pharmacology , Brain/metabolism , Mammals , Mice , Suprachiasmatic Nucleus
9.
Methods Mol Biol ; 2482: 211-215, 2022.
Article in English | MEDLINE | ID: mdl-35610429

ABSTRACT

Rhythmic locomotor activity is a commonly used readout of general circadian function in animals. For instance, measuring the activity of rodents in their home cages can provide information about circadian phase and period in response to genetic, pharmacological, and environmental manipulations. Herein, the use of infrared light sensors to measure circadian locomotor activity is described. Furthermore, we provide information about data handling, analysis and software use as well as points to consider when performing the experiment.


Subject(s)
Circadian Rhythm , Rodentia , Animals , Circadian Rhythm/physiology , Locomotion
10.
Sci Adv ; 7(39): eabi7828, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34550736

ABSTRACT

The mammalian circadian clock, expressed throughout the brain and body, controls daily metabolic homeostasis. Clock function in peripheral tissues is required, but not sufficient, for this task. Because of the lack of specialized animal models, it is unclear how tissue clocks interact with extrinsic signals to drive molecular oscillations. Here, we isolated the interaction between feeding and the liver clock by reconstituting Bmal1 exclusively in hepatocytes (Liver-RE), in otherwise clock-less mice, and controlling timing of food intake. We found that the cooperative action of BMAL1 and the transcription factor CEBPB regulates daily liver metabolic transcriptional programs. Functionally, the liver clock and feeding rhythm are sufficient to drive temporal carbohydrate homeostasis. By contrast, liver rhythms tied to redox and lipid metabolism required communication with the skeletal muscle clock, demonstrating peripheral clock cross-talk. Our results highlight how the inner workings of the clock system rely on communicating signals to maintain daily metabolism.

11.
Adipocyte ; 9(1): 620-625, 2020 12.
Article in English | MEDLINE | ID: mdl-33043853

ABSTRACT

A chronic low-grade inflammation of white adipose tissue (WAT) is one of the hallmarks of obesity and is proposed to contribute to insulin resistance and type 2 diabetes. Despite this, the causal mechanisms underlying WAT inflammation remain unclear. Based on metabolomic analyses of human WAT, Petrus et al. showed that the amino acid glutamine was the most markedly reduced polar metabolite in the obese state. Reduced glutamine levels in adipocytes induce an increase of Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) levels via induction of glycolysis and the hexosamine biosynthetic pathways. This promotes nuclear O-GlcNAcylation, a posttranslational modification that activates the transcription of pro-inflammatory genes. Conversely, glutamine supplementation in vitro and in vivo, reversed these effects. Altogether, dysregulation of intracellular glutamine metabolism in WAT establishes an epigenetic link between adipocytes and inflammation. This commentary discusses these findings and their possibly therapeutic relevance in relation to insulin resistance and type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Glutamine/metabolism , Adipose Tissue, White/metabolism , Animals , Chromatin Assembly and Disassembly , Disease Susceptibility , Epigenesis, Genetic , Glycolysis , Humans , Immunomodulation , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance , Metabolome , Metabolomics/methods , Obesity/metabolism
12.
Proc Nutr Soc ; 79(1): 42-46, 2020 02.
Article in English | MEDLINE | ID: mdl-31122299

ABSTRACT

Nutrition is a major variable factor in human environments. The composition of nutrients has changed markedly in recent decades which may contribute to the increased prevalence of metabolic diseases, such as obesity and type 2 diabetes. Fat is an important component of the diet which comes in various forms with fatty acids (FA) of different carbon chain lengths and saturation degrees. In addition to being an energy supply, FA function as potent signalling molecules and influence transcriptional activity. Among other tissues, dietary FA target white adipose tissue function, which is central in maintaining metabolic health. This review focuses on the possible role of dietary FA composition and its effect on human white adipose tissue expandability and transcriptional response. Altogether, the existing literature suggests that unsaturated fat has more benign effects on adipose tissue distribution when compared to long-chain saturated fat. However, the mechanisms of action remain poorly characterised.


Subject(s)
Adipose Tissue, White , Dietary Fats/metabolism , Fatty Acids/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiology , Humans
13.
Cell Metab ; 31(2): 375-390.e11, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31866443

ABSTRACT

While obesity and associated metabolic complications are linked to inflammation of white adipose tissue (WAT), the causal factors remain unclear. We hypothesized that the local metabolic environment could be an important determinant. To this end, we compared metabolites released from WAT of 81 obese and non-obese women. This identified glutamine to be downregulated in obesity and inversely associated with a pernicious WAT phenotype. Glutamine administration in vitro and in vivo attenuated both pro-inflammatory gene and protein levels in adipocytes and WAT and macrophage infiltration in WAT. Metabolomic and bioenergetic analyses in human adipocytes suggested that glutamine attenuated glycolysis and reduced uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) levels. UDP-GlcNAc is the substrate for the post-translational modification O-linked ß-N-acetylglucosamine (O-GlcNAc) mediated by the enzyme O-GlcNAc transferase. Functional studies in human adipocytes established a mechanistic link between reduced glutamine, O-GlcNAcylation of nuclear proteins, and a pro-inflammatory transcriptional response. Altogether, glutamine metabolism is linked to WAT inflammation in obesity.


Subject(s)
Adipocytes , Adipose Tissue, White , Glutamine , Inflammation/metabolism , Obesity/metabolism , Acetylglucosamine , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adult , Animals , Cells, Cultured , Cohort Studies , Female , Glucose/metabolism , Glutamine/metabolism , Glutamine/pharmacology , Glycosylation/drug effects , Humans , Male , Mice, Inbred C57BL , Middle Aged , N-Acetylglucosaminyltransferases/metabolism
14.
EBioMedicine ; 44: 489-501, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31221584

ABSTRACT

BACKGROUND: A positive energy balance promotes white adipose tissue (WAT) expansion which is characterized by activation of a repertoire of events including hypoxia, inflammation and extracellular matrix remodelling. The transmembrane glycoprotein CD248 has been implicated in all these processes in different malignant and inflammatory diseases but its potential impact in WAT and metabolic disease has not been explored. METHODS: The role of CD248 in adipocyte function and glucose metabolism was evaluated by omics analyses in human WAT, gene knockdowns in human in vitro differentiated adipocytes and by adipocyte-specific and inducible Cd248 gene knockout studies in mice. FINDINGS: CD248 is upregulated in white but not brown adipose tissue of obese and insulin-resistant individuals. Gene ontology analyses showed that CD248 expression associated positively with pro-inflammatory/pro-fibrotic pathways. By combining data from several human cohorts with gene knockdown experiments in human adipocytes, our results indicate that CD248 acts as a microenvironmental sensor which mediates part of the adipose tissue response to hypoxia and is specifically perturbed in white adipocytes in the obese state. Adipocyte-specific and inducible Cd248 knockouts in mice, both before and after diet-induced obesity and insulin resistance/glucose intolerance, resulted in increased microvascular density as well as attenuated hypoxia, inflammation and fibrosis without affecting fat cell volume. This was accompanied by significant improvements in insulin sensitivity and glucose tolerance. INTERPRETATION: CD248 exerts detrimental effects on WAT phenotype and systemic glucose homeostasis which may be reversed by suppression of adipocyte CD248. Therefore, CD248 may constitute a target to treat obesity-associated co-morbidities.


Subject(s)
Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Energy Metabolism/genetics , Hypoxia/metabolism , Panniculitis/genetics , Panniculitis/metabolism , Adult , Animals , Disease Models, Animal , Extracellular Matrix , Female , Fibrosis , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Mice, Transgenic , Middle Aged , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Panniculitis/pathology , Signal Transduction
15.
Curr Genomics ; 20(6): 428-437, 2019 Sep.
Article in English | MEDLINE | ID: mdl-32477000

ABSTRACT

According to the Developmental Origin of Health and Disease (DOHaD) concept, maternal obesity and the resulting accelerated growth in neonates predispose offspring to obesity and associated metabolic diseases that may persist across generations. In this context, the adipose tissue has emerged as an important player due to its involvement in metabolic health, and its high potential for plasticity and adaptation to environmental cues. Recent years have seen a growing interest in how maternal obesity induces long-lasting adipose tissue remodeling in offspring and how these modifications could be transmitted to subsequent generations in an inter- or transgenerational manner. In particular, epigenetic mechanisms are thought to be key players in the developmental programming of adipose tissue, which may partially mediate parts of the transgenerational inheritance of obesity. This review presents data supporting the role of maternal obesity in the developmental programming of adipose tissue through epigenetic mechanisms. Inter- and transgenerational effects on adipose tissue expansion are also discussed in this review.

16.
Cell Rep ; 25(3): 551-560.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332637

ABSTRACT

White adipose tissue (WAT) mass is determined by adipocyte size and number. While adipocytes are continuously turned over, the mechanisms controlling fat cell number in WAT upon weight changes are unclear. Herein, prospective studies of human subcutaneous WAT demonstrate that weight gain increases both adipocyte size and number, but the latter remains unaltered after weight loss. Transcriptome analyses associate changes in adipocyte number with the expression of 79 genes. This gene set is enriched for growth factors, out of which one, transforming growth factor-ß3 (TGFß3), stimulates adipocyte progenitor proliferation, resulting in a higher number of cells undergoing differentiation in vitro. The relevance of these observations was corroborated in vivo where Tgfb3+/- mice, in comparison with wild-type littermates, display lower subcutaneous adipocyte progenitor proliferation, WAT hypertrophy, and glucose intolerance. TGFß3 is therefore a regulator of subcutaneous adipocyte number and may link WAT morphology to glucose metabolism.


Subject(s)
Adipogenesis , Adipose Tissue, White/pathology , Glucose Intolerance/etiology , Obesity/complications , Subcutaneous Fat/pathology , Transforming Growth Factor beta3/physiology , Adipose Tissue, White/metabolism , Adolescent , Animals , Case-Control Studies , Cell Differentiation , Female , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies , Subcutaneous Fat/metabolism
17.
Trends Endocrinol Metab ; 29(10): 675-685, 2018 10.
Article in English | MEDLINE | ID: mdl-30104112

ABSTRACT

An adverse nutritional environment during the perinatal period increases the risk of adult-onset metabolic diseases, such as obesity, which may persist across generations. Adipose tissue (AT) from offspring of malnourished dams has been shown to display altered adipogenesis, lipogenesis, and adipokine expression, impaired thermogenesis, and low-grade inflammation. Although the exact mechanisms underlying these alterations remain unclear, epigenetic processes are believed to have an important role. In this review, we focus on epigenetic mechanisms in AT that may account for transgenerational dysregulation of adipocyte formation and adipose function. Understanding the complex interactions between maternal diet and epigenetic regulation of the AT in offspring may be valuable in improving preventive strategies against the obesity pandemic.


Subject(s)
Adipogenesis/physiology , Adipose Tissue , Epigenesis, Genetic/physiology , Maternal Nutritional Physiological Phenomena/physiology , Nutrition Disorders/metabolism , Pregnancy Complications/metabolism , Adipogenesis/genetics , Adipose Tissue/embryology , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Epigenesis, Genetic/genetics , Female , Humans , Nutrition Disorders/complications , Nutrition Disorders/etiology , Nutrition Disorders/genetics , Pregnancy
18.
Int J Obes (Lond) ; 42(12): 2047-2056, 2018 12.
Article in English | MEDLINE | ID: mdl-29453465

ABSTRACT

BACKGROUND: Adipokines are peptides secreted from white adipose tissue (WAT), which have been linked to WAT dysfunction and metabolic complications of obesity. We set out to identify novel adipokines in subcutaneous WAT (sWAT) linked to insulin resistance (IR). METHODS: Gene expression was determined by microarray and qPCR in obese and non-obese subjects with varying degree of IR. WAT-secreted and circulating protein levels were measured by ELISA. RESULTS: In sWAT of 80 obese women discordant for IR, 44 genes encoding potential adipose-secreted proteins were differentially expressed. Among these, merely two proteins, S100A4 and MXRA5 were released from sWAT in a time-dependent manner (criterion for true adipokines) but only the circulating levels of S100A4 were higher in IR. In two additional cohorts (n = 29 and n = 56), sWAT S100A4 secretion was positively and BMI-independently associated with IR (determined by clamp or HOMA-IR), ATP-III risk score and adipocyte size (hypertrophy). In non-obese (n = 20) and obese subjects before and after bariatric surgery (n = 21), circulating and sWAT-secreted levels were highest in the obese and normalized following weight loss. Serum S100A4 concentrations were higher in subjects with type 2 diabetes. S100A4 sWAT expression associated positively with genes involved in inflammation/extracellular matrix formation and inversely with genes in metabolic pathways. Although S100A4 was expressed in both stromal cells and adipocytes, only the expression in adipocytes associated with BMI. CONCLUSIONS: S100A4 is a novel adipokine associated with IR and sWAT inflammation/adipocyte hypertrophy independently of BMI. Its value as a circulating marker for dysfunctional WAT and IR needs to be validated in larger cohorts.


Subject(s)
Adipokines/metabolism , Adipose Tissue, White/metabolism , Insulin Resistance/physiology , Obesity/metabolism , S100 Calcium-Binding Protein A4/metabolism , Adipokines/blood , Adipose Tissue, White/chemistry , Adult , Biomarkers , Cohort Studies , Female , Humans , Inflammation/metabolism , Male , Middle Aged , Obesity/blood , Obesity/epidemiology , S100 Calcium-Binding Protein A4/blood
19.
J Clin Endocrinol Metab ; 103(2): 710-721, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29121255

ABSTRACT

Context: Insulin resistance (IR) is promoted by a chronic low-grade inflammation in white adipose tissue (WAT). The latter might be regulated through epigenetic mechanisms such as DNA methylation. The one carbon cycle (1CC) is a central metabolic process governing DNA methylation. Objective: To identify adipocyte-expressed 1CC genes linked to WAT inflammation, IR, and their causal role. Design: Cohort study. Setting: Outpatient academic clinic. Participants: Obese and nonobese subjects. Methods: Gene expression and DNA methylation arrays were performed in subcutaneous WAT and isolated adipocytes. In in vitro differentiated human adipocytes, gene knockdown was achieved by small interfering RNA, and analyses included microarray, quantitative polymerase chain reaction, DNA methylation by enzyme-linked immunosorbent assay and pyrosequencing, protein secretion by enzyme-linked immunosorbent assay, targeted metabolomics, and luciferase reporter and thermal shift assays. Main Outcome Measures: Effects on adipocyte inflammation. Results: In adipocytes from obese individuals, global DNA hypermethylation was associated positively with gene expression of proinflammatory pathways. Among the 1CC genes, IR in vivo and proinflammatory gene expression in WAT were most strongly and inversely associated with SLC19A1, a gene encoding a membrane folate carrier. SLC19A1 knockdown in human adipocytes perturbed intracellular 1CC metabolism, induced global DNA hypermethylation, and increased expression of proinflammatory genes. Several CpG loci linked SLC19A1 to inflammation; validation studies were focused on the chemokine C-C motif chemokine ligand 2 (CCL2) in which methylation in the promoter (cg12698626) regulated CCL2 expression and CCL2 secretion through altered transcriptional activity. Conclusions: Reduced SLC19A1 expression in human adipocytes induces DNA hypermethylation, resulting in increased expression of specific proinflammatory genes, including CCL2. This constitutes an epigenetic mechanism that might link dysfunctional adipocytes to WAT inflammation and IR.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/pathology , DNA Methylation/genetics , Inflammation/genetics , Insulin Resistance/genetics , Reduced Folate Carrier Protein/genetics , Adipocytes/pathology , Adipose Tissue/metabolism , Adult , Case-Control Studies , Cohort Studies , Epigenesis, Genetic/physiology , Female , Gene Expression Profiling , Humans , Inflammation/metabolism , Microarray Analysis , Middle Aged , Obesity/complications , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Panniculitis/genetics , Panniculitis/metabolism , Panniculitis/pathology , Reduced Folate Carrier Protein/metabolism , Young Adult
20.
Sci Rep ; 7(1): 10152, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860604

ABSTRACT

Increased adipocyte lipolysis links obesity to insulin resistance. The lipid droplet coating-protein Perilipin participates in regulation of lipolysis and is implicated in obesity. In the present study we investigate epigenetic regulation of the PLIN1 gene by correlating PLIN1 CpG methylation to gene expression and lipolysis, and functionally evaluating PLIN1 promoter methylation. PLIN1 CpG methylation in adipocytes and gene expression in white adipose tissue (WAT) was quantified in two cohorts by array. Basal lipolysis in WAT explants and adipocytes was quantified by measuring glycerol release. CpG-methylation of the PLIN1 promoter in adipocytes from obese women was higher as compared to never-obese women. PLIN1 promoter methylation was inversely correlated with PLIN1 mRNA expression and the lipolytic activity. Human mesenchymal stem cells (hMSCs) differentiated in vitro into adipocytes and harboring methylated PLIN1 promoter displayed decreased reporter gene activity as compared to hMSCs harboring unmethylated promoter. Treatment of hMSCs differentiated in vitro into adipocytes with a DNA methyltransferase inhibitor increased levels of PLIN1 mRNA and protein. In conclusion, the PLIN1 gene is epigenetically regulated and promoter methylation is inversely correlated with basal lipolysis in women suggesting that epigenetic regulation of PLIN1 is important for increased adipocyte lipolysis in insulin resistance states.


Subject(s)
Epigenesis, Genetic , Lipolysis , Obesity/genetics , Perilipin-1/genetics , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adult , CpG Islands , DNA Methylation , Female , Glycerol/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Middle Aged , Obesity/metabolism , Perilipin-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...