Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(13): e23743, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38877852

ABSTRACT

Kisspeptin signaling regulates energy homeostasis. Adiposity is the principal source and receiver of peripheral Kisspeptin, and adipose Kiss1 metastasis suppressor (Kiss1) gene expression is stimulated by exercise. However, whether the adipose Kiss1 gene regulates energy homeostasis and plays a role in adaptive alterations during prolonged exercise remains unknown. Here, we investigated the role of Kiss1 role in mice and adipose tissues and the adaptive changes it induces after exercise, using adipose-specific Kiss1 knockout (Kiss1adipoq-/-) and adeno-associated virus-induced adipose tissue Kiss1-overexpressing (Kiss1adipoq over) mice. We found that adipose-derived kisspeptin signal regulates lipid and glucose homeostasis to maintain systemic energy homeostasis, but in a sex-dependent manner, with more pronounced metabolic changes in female mice. Kiss1 regulated adaptive alterations of genes and proteins in tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OxPhos) pathways in female gWAT following prolonged aerobic exercise. We could further show that adipose Kiss1 deficiency leads to reduced peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) protein content of soleus muscle and maximum oxygen uptake (VO2 max) of female mice after prolonged exercise. Therefore, adipose Kisspeptin may be a novel adipokine that increases organ sensitivity to glucose, lipids, and oxygen following exercise.


Subject(s)
Adipose Tissue , Energy Metabolism , Homeostasis , Kisspeptins , Mice, Knockout , Physical Conditioning, Animal , Animals , Kisspeptins/metabolism , Kisspeptins/genetics , Female , Mice , Physical Conditioning, Animal/physiology , Male , Adipose Tissue/metabolism , Mice, Inbred C57BL , Adaptation, Physiological
2.
Cell Metab ; 34(3): 473-486.e9, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35120589

ABSTRACT

Fasting metabolism and immunity are tightly linked; however, it is largely unknown how immune cells contribute to metabolic homeostasis during fasting in healthy subjects. Here, we combined cell-type-resolved genomics and computational approaches to map crosstalk between hepatocytes and liver macrophages during fasting. We identified the glucocorticoid receptor (GR) as a key driver of fasting-induced reprogramming of the macrophage secretome including fasting-suppressed cytokines and showed that lack of macrophage GR impaired induction of ketogenesis during fasting as well as endotoxemia. Mechanistically, macrophage GR suppressed the expression of tumor necrosis factor (TNF) and promoted nuclear translocation of hepatocyte GR to activate a fat oxidation/ketogenesis-related gene program, cooperatively induced by GR and peroxisome proliferator-activated receptor alpha (PPARα) in hepatocytes. Together, our results demonstrate how resident liver macrophages directly influence ketogenesis in hepatocytes, thereby also outlining a strategy by which the immune system can set the metabolic tone during inflammatory disease and infection.


Subject(s)
Fasting , Receptors, Glucocorticoid , Animals , Fasting/metabolism , Hepatocytes/metabolism , Humans , Ketone Bodies/metabolism , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , PPAR alpha/metabolism , Receptors, Glucocorticoid/metabolism
3.
STAR Protoc ; 2(3): 100805, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34527961

ABSTRACT

Isolation of nuclei tagged in specific cell types (INTACT) allows for stress-free and high-throughput analyses of cellular subpopulations. Here, we present an improved protocol for isolation of pure and high-quality GFP-labeled nuclei from frozen livers of INTACT mice, as well as protocols for downstream sequencing analyses. The adaptation to frozen tissue provides a pause point that allows sampling at multiple time points and/or phenotypic characterization of livers prior to nuclei isolation and downstream analyses. For complete details on the use of this protocol, please refer to Loft et al. (2021).


Subject(s)
Cell Nucleus/chemistry , Chromatin Immunoprecipitation Sequencing , Cytological Techniques/methods , Green Fluorescent Proteins/chemistry , Liver/cytology , RNA-Seq , Animals , Cell Nucleus/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Liver/chemistry , Male , Mice
4.
Cell Metab ; 33(8): 1685-1700.e9, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34237252

ABSTRACT

Liver fibrosis is a strong predictor of long-term mortality in individuals with metabolic-associated fatty liver disease; yet, the mechanisms underlying the progression from the comparatively benign fatty liver state to advanced non-alcoholic steatohepatitis (NASH) and liver fibrosis are incompletely understood. Using cell-type-resolved genomics, we show that comprehensive alterations in hepatocyte genomic and transcriptional settings during NASH progression, led to a loss of hepatocyte identity. The hepatocyte reprogramming was under tight cooperative control of a network of fibrosis-activated transcription factors, as exemplified by the transcription factor Elf-3 (ELF3) and zinc finger protein GLIS2 (GLIS2). Indeed, ELF3- and GLIS2-controlled fibrosis-dependent hepatokine genes targeting disease-associated hepatic stellate cell gene programs. Thus, interconnected transcription factor networks not only promoted hepatocyte dysfunction but also directed the intra-hepatic crosstalk necessary for NASH and fibrosis progression, implying that molecular "hub-centered" targeting strategies are superior to existing mono-target approaches as currently used in NASH therapy.


Subject(s)
Gene Regulatory Networks , Non-alcoholic Fatty Liver Disease , Communication , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
5.
J Cachexia Sarcopenia Muscle ; 11(6): 1459-1475, 2020 12.
Article in English | MEDLINE | ID: mdl-33090732

ABSTRACT

BACKGROUND: Cancer cachexia (CCx) is a multifactorial energy-wasting syndrome reducing the efficiency of anti-cancer therapies, quality of life, and survival of cancer patients. In the past years, most studies focused on the identification of tumour and host-derived proteins contributing to CCx. However, there is still a lack of studies addressing the changes in bioactive lipids. The aim of this study was to identify specific lipid species as a hallmark of CCx by performing a broad range lipid analysis of plasma from well-established CCx mouse models as well as cachectic and weight stable cancer patients. METHODS: Plasma from non-cachectic (PBS-injected mice, NC26 tumour-bearing mice), pre-cachectic and cachectic mice (C26 and LLC tumour-bearing mice, ApcMin/+ mutant mice), and plasma from weight stable and cachectic patients with gastrointestinal cancer, were analysed using the Lipidyzer™ platform. In total, 13 lipid classes and more than 1100 lipid species, including sphingolipids, neutral and polar glycerolipids, were covered by the analysis. Correlation analysis between specific lipid species and readouts of CCx were performed. Lipidomics data were confirmed by gene expression analysis of metabolic organs to analyse enzymes involved in sphingolipid synthesis and degradation. RESULTS: A decrease in several lysophosphatidylcholine (LPC) species and an increase in numerous sphingolipids including sphingomyelins (SMs), ceramides (CERs), hexosyl-ceramides (HCERs) and lactosyl-ceramides (LCERs), were mutual features of CCx in both mice and cancer patients. Notably, sphingolipid levels gradually increased during cachexia development. Key enzymes involved in ceramide synthesis were elevated in liver but not in adipose, muscle, or tumour tissues, suggesting that ceramide turnover in the liver is a major contributor to elevated sphingolipid levels in CCx. LPC(16:1), LPC(20:3), SM(16:0), SM(24:1), CER(16:0), CER(24:1), HCER(16:0), and HCER(24:1) were the most consistently affected lipid species between mice and humans and correlated negatively (LPCs) or positively (SMs, CERs and HCERs) with the severity of body weight loss. CONCLUSIONS: High levels of sphingolipids, specifically ceramides and modified ceramides, are a defining feature of murine and human CCx and may contribute to tissue wasting and skeletal muscle atrophy through the inhibition of anabolic signals. The progressive increase in sphingolipids during cachexia development supports their potential as early biomarkers for CCx.


Subject(s)
Cachexia , Ceramides , Neoplasms , Animals , Cachexia/etiology , Ceramides/metabolism , Humans , Mice , Muscular Atrophy , Neoplasms/complications , Quality of Life
6.
Trends Cancer ; 4(12): 849-860, 2018 12.
Article in English | MEDLINE | ID: mdl-30470306

ABSTRACT

Cancer cachexia is a multifactorial condition characterized by body weight loss that negatively affects quality of life and survival of patients with cancer. Despite the clinical relevance, there is currently no defined standard of care to effectively counteract cancer-associated progressive tissue wasting. Skeletal muscle atrophy represents the main manifestation of cancer cachexia. However, cancer cachexia is increasingly seen as a systemic phenomenon affecting and/or influenced by various organs. Here, we describe recent developments elucidating the roles of different tissues as well as tissue crosstalk in this wasting syndrome, including potential links to other cancer-associated morbidities. A more comprehensive understanding of cancer cachexia etiology and heterogeneity may enable the development of intervention strategies to prevent or reverse this devastating condition.


Subject(s)
Antineoplastic Agents/therapeutic use , Cachexia/physiopathology , Neoplasms/complications , Nutritional Support/methods , Paraneoplastic Syndromes/physiopathology , Antineoplastic Agents/pharmacology , Cachexia/etiology , Cachexia/mortality , Cachexia/prevention & control , Combined Modality Therapy/methods , Humans , Muscle, Skeletal/physiopathology , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/physiopathology , Paraneoplastic Syndromes/etiology , Paraneoplastic Syndromes/mortality , Paraneoplastic Syndromes/prevention & control , Quality of Life , Treatment Outcome
7.
Genome Res ; 28(2): 243-255, 2018 02.
Article in English | MEDLINE | ID: mdl-29233921

ABSTRACT

The ability to predict transcription factors based on sequence information in regulatory elements is a key step in systems-level investigation of transcriptional regulation. Here, we have developed a novel tool, IMAGE, for precise prediction of causal transcription factors based on transcriptome profiling and genome-wide maps of enhancer activity. High precision is obtained by combining a near-complete database of position weight matrices (PWMs), generated by compiling public databases and systematic prediction of PWMs for uncharacterized transcription factors, with a state-of-the-art method for PWM scoring and a novel machine learning strategy, based on both enhancers and promoters, to predict the contribution of motifs to transcriptional activity. We applied IMAGE to published data obtained during 3T3-L1 adipocyte differentiation and showed that IMAGE predicts causal transcriptional regulators of this process with higher confidence than existing methods. Furthermore, we generated genome-wide maps of enhancer activity and transcripts during human mesenchymal stem cell commitment and adipocyte differentiation and used IMAGE to identify positive and negative transcriptional regulators of this process. Collectively, our results demonstrate that IMAGE is a powerful and precise method for prediction of regulators of gene expression.


Subject(s)
Enhancer Elements, Genetic , Nucleotide Motifs/genetics , Software , Transcription Factors/genetics , Binding Sites/genetics , Computational Biology/methods , Gene Expression Regulation/genetics , Humans , Position-Specific Scoring Matrices , Promoter Regions, Genetic
8.
Cell Rep ; 16(9): 2359-72, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545881

ABSTRACT

Glucose is an important inducer of insulin secretion, but it also stimulates long-term adaptive changes in gene expression that can either promote or antagonize the proliferative potential and function of ß cells. Here, we have generated time-resolved profiles of enhancer and transcriptional activity in response to glucose in the INS-1E pancreatic ß cell line. Our data outline a biphasic response with a first transcriptional wave during which metabolic genes are activated, and a second wave where cell-cycle genes are activated and ß cell identity genes are repressed. The glucose-sensing transcription factor ChREBP directly activates first wave enhancers, whereas repression and activation of second wave enhancers are indirect. By integrating motif enrichment within late-regulated enhancers with expression profiles of the associated transcription factors, we have identified multiple putative regulators of the second wave. These include RORγ, the activity of which is important for glucose-induced proliferation of both INS-1E and primary rat ß cells.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation/genetics , Dose-Response Relationship, Drug , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Expression Regulation , Genomics , Glucose/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Promoter Regions, Genetic , Rats , Transcription, Genetic
9.
Bioessays ; 38(7): 618-26, 2016 07.
Article in English | MEDLINE | ID: mdl-27273739

ABSTRACT

Cofactor squelching is the term used to describe competition between transcription factors (TFs) for a limited amount of cofactors in a cell with the functional consequence that TFs in a given cell interfere with the activity of each other. Since cofactor squelching was proposed based primarily on reporter assays some 30 years ago, it has remained controversial, and the idea that it could be a physiologically relevant mechanism for transcriptional repression has not received much support. However, recent genome-wide studies have demonstrated that signal-dependent TFs are very often absent from the enhancers that are acutely repressed by those signals, which is consistent with an indirect mechanism of repression such as squelching. Here we review these recent studies in the light of the classical studies of cofactor squelching, and we discuss how TF cooperativity in so-called hotspots and super-enhancers may sensitize these to cofactor squelching.


Subject(s)
DNA/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Eukaryota/genetics , Eukaryota/metabolism , Humans , Protein Binding
10.
Article in English | MEDLINE | ID: mdl-26432526

ABSTRACT

The ability to modify the transcriptional program in response to external signals provides a way for mammalian cells to alter their biological fate and properties, thereby adapting to changes in the environment. Adipocytes are excellent examples of differentiated cells that possess a striking transcriptional plasticity when exposed to physiological and metabolic stimuli. In our work, we have focused on understanding the processes responsible for modulating the genomic programming in response to different external signals. Thus, we have shown that browning of human adipocytes with rosiglitazone, an antidiabetic agonist of the key adipocyte transcription factor peroxisome proliferator-activated receptor γ (PPARγ), involves redistribution of PPARγ binding to form browning-selective PPARγ super-enhancers that drive expression of key browning genes. These include genes encoding transcriptional regulators, such as Krüppel-like factor 11 (KLF11) that are essential for modulating the genomic program in white adipocytes to induce browning. Furthermore, we have shown that acute suppression of adipocyte genes by the proinflammatory cytokine, tumor necrosis factor (TNF), involves redistribution of cofactors to enhancers activated by the master inflammatory regulator, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Interestingly, this redistribution occurs selectively from enhancers with high-cofactor occupancies, thereby predominantly affecting super-enhancers and their associated genes. We propose that this is a general mechanism contributing to transcriptional repression associated with activation of signal-dependent transcription factors.


Subject(s)
Adipocytes/metabolism , Cellular Reprogramming/genetics , Gene Expression Regulation , Adipocytes/drug effects , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Cycle Proteins/metabolism , Cell Plasticity , Cellular Reprogramming Techniques , Humans , Hypoglycemic Agents/pharmacology , NF-kappa B/metabolism , PPAR gamma/agonists , Repressor Proteins/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology , Tumor Necrosis Factor-alpha/metabolism
11.
Genome Res ; 25(9): 1281-94, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26113076

ABSTRACT

The proinflammatory cytokine tumor necrosis factor (TNF) plays a central role in low-grade adipose tissue inflammation and development of insulin resistance during obesity. In this context, nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) is directly involved and required for the acute activation of the inflammatory gene program. Here, we show that the major transactivating subunit of NFκB, v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA), is also required for acute TNF-induced suppression of adipocyte genes. Notably, this repression does not involve RELA binding to the associated enhancers but rather loss of cofactors and enhancer RNA (eRNA) selectively from high-occupancy sites within super-enhancers. Based on these data, we have developed models that, with high accuracy, predict which enhancers and genes are repressed by TNF in adipocytes. We show that these models are applicable to other cell types where TNF represses genes associated with super-enhancers in a highly cell-type-specific manner. Our results propose a novel paradigm for NFκB-mediated repression, whereby NFκB selectively redistributes cofactors from high-occupancy enhancers, thereby specifically repressing super-enhancer-associated cell identity genes.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Cell Cycle Proteins , Cell Differentiation , Cellular Reprogramming/genetics , Humans , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/metabolism , Organ Specificity/genetics , Protein Binding , Protein Transport , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Transcriptome
12.
Nucleic Acids Res ; 43(6): e40, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25564527

ABSTRACT

RNA-seq is a sensitive and accurate technique to compare steady-state levels of RNA between different cellular states. However, as it does not provide an account of transcriptional activity per se, other technologies are needed to more precisely determine acute transcriptional responses. Here, we have developed an easy, sensitive and accurate novel computational method, IRNA-SEQ: , for genome-wide assessment of transcriptional activity based on analysis of intron coverage from total RNA-seq data. Comparison of the results derived from iRNA-seq analyses with parallel results derived using current methods for genome-wide determination of transcriptional activity, i.e. global run-on (GRO)-seq and RNA polymerase II (RNAPII) ChIP-seq, demonstrate that iRNA-seq provides similar results in terms of number of regulated genes and their fold change. However, unlike the current methods that are all very labor-intensive and demanding in terms of sample material and technologies, iRNA-seq is cheap and easy and requires very little sample material. In conclusion, iRNA-seq offers an attractive novel alternative to current methods for determination of changes in transcriptional activity at a genome-wide level.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Cell Line , Chromatin Immunoprecipitation/methods , Chromatin Immunoprecipitation/statistics & numerical data , Gene Expression Profiling/statistics & numerical data , Gene Expression Regulation , Genome, Human , Humans , Introns , Sequence Analysis, RNA/statistics & numerical data
13.
Genes Dev ; 29(1): 7-22, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25504365

ABSTRACT

Long-term exposure to peroxisome proliferator-activated receptor γ (PPARγ) agonists such as rosiglitazone induces browning of rodent and human adipocytes; however, the transcriptional mechanisms governing this phenotypic switch in adipocytes are largely unknown. Here we show that rosiglitazone-induced browning of human adipocytes activates a comprehensive gene program that leads to increased mitochondrial oxidative capacity. Once induced, this gene program and oxidative capacity are maintained independently of rosiglitazone, suggesting that additional browning factors are activated. Browning triggers reprogramming of PPARγ binding, leading to the formation of PPARγ "superenhancers" that are selective for brown-in-white (brite) adipocytes. These are highly associated with key brite-selective genes. Based on such an association, we identified an evolutionarily conserved metabolic regulator, Kruppel-like factor 11 (KLF11), as a novel browning transcription factor in human adipocytes that is required for rosiglitazone-induced browning, including the increase in mitochondrial oxidative capacity. KLF11 is directly induced by PPARγ and appears to cooperate with PPARγ in a feed-forward manner to activate and maintain the brite-selective gene program.


Subject(s)
Adipocytes/metabolism , Cell Cycle Proteins/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Repressor Proteins/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes, Brown/cytology , Apoptosis Regulatory Proteins , Cell Cycle Proteins/genetics , Cellular Reprogramming , Chromatin/metabolism , Gene Expression Regulation , Humans , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Oxidation-Reduction , Protein Binding , Repressor Proteins/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Transcriptional Activation/drug effects
14.
Proc Natl Acad Sci U S A ; 111(3): 1055-9, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395784

ABSTRACT

Type 1 diabetes is due to destruction of pancreatic ß-cells. Lysine deacetylase inhibitors (KDACi) protect ß-cells from inflammatory destruction in vitro and are promising immunomodulators. Here we demonstrate that the clinically well-tolerated KDACi vorinostat and givinostat revert diabetes in the nonobese diabetic (NOD) mouse model of type 1 diabetes and counteract inflammatory target cell damage by a mechanism of action consistent with transcription factor--rather than global chromatin--hyperacetylation. Weaning NOD mice received low doses of vorinostat and givinostat in their drinking water until 100-120 d of age. Diabetes incidence was reduced by 38% and 45%, respectively, there was a 15% increase in the percentage of islets without infiltration, and pancreatic insulin content increased by 200%. Vorinostat treatment increased the frequency of functional regulatory T-cell subsets and their transcription factors Gata3 and FoxP3 in parallel to a decrease in inflammatory dendritic cell subsets and their cytokines IL-6, IL-12, and TNF-α. KDACi also inhibited LPS-induced Cox-2 expression in peritoneal macrophages from C57BL/6 and NOD mice. In insulin-producing ß-cells, givinostat did not upregulate expression of the anti-inflammatory genes Socs1-3 or sirtuin-1 but reduced levels of IL-1ß + IFN-γ-induced proinflammatory Il1a, Il1b, Tnfα, Fas, Cxcl2, and reduced cytokine-induced ERK phosphorylation. Further, NF-κB genomic iNos promoter binding was reduced by 50%, and NF-κB-dependent mRNA expression was blocked. These effects were associated with NF-κB subunit p65 hyperacetylation. Taken together, these data provide a rationale for clinical trials of safety and efficacy of KDACi in patients with autoimmune disease such as type 1 diabetes.


Subject(s)
Chromatin/metabolism , Diabetes Mellitus, Type 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Insulin-Secreting Cells/cytology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Epigenesis, Genetic , Female , GATA3 Transcription Factor/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Inflammation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Rats , Time Factors , Vorinostat
15.
J Biol Chem ; 286(15): 13214-25, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21282101

ABSTRACT

Chronic exposure to elevated levels of glucose and fatty acids leads to dysfunction of pancreatic ß-cells by mechanisms that are only partly understood. The transcription factor peroxisome proliferator-activated receptor α (PPARα) is an important regulator of genes involved in fatty acid metabolism and has been shown to protect against lipid-induced ß-cell dysfunction. We and others have previously shown that expression of the PPARα gene in ß-cells is rapidly repressed by glucose. Here we show that the PPARα gene is transcribed from five alternative transcription start sites, resulting in three alternative first exons that are spliced to exon 2. Expression of all PPARα transcripts is repressed by glucose both in insulinoma cells and in isolated pancreatic islets. The observation that the dynamics of glucose repression of PPARα transcription are very similar to those of glucose activation of target genes by the carbohydrate response element-binding protein (ChREBP) prompted us to investigate the potential role of ChREBP in the regulation of PPARα expression. We show that a constitutively active ChREBP lacking the N-terminal domain efficiently represses PPARα expression in insulinoma cells and in rodent and human islets. In addition, we demonstrate that siRNA-mediated knockdown of ChREBP abrogates glucose repression of PPARα expression as well as induction of well established ChREBP target genes in insulinoma cells. In conclusion, this work shows that ChREBP is a critical and direct mediator of glucose repression of PPARα gene expression in pancreatic ß-cells, suggesting that ChREBP may be important for glucose suppression of the fatty acid oxidation capacity of ß-cells.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Nuclear Proteins/metabolism , PPAR alpha/biosynthesis , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Exons/physiology , Fatty Acids/genetics , Fatty Acids/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Glucose/pharmacology , HEK293 Cells , Humans , Insulin-Secreting Cells/cytology , Mice , Nuclear Proteins/genetics , Oxidation-Reduction , PPAR alpha/genetics , Protein Structure, Tertiary , Rats , Rats, Wistar , Repressor Proteins/genetics , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...