Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Nat Metab ; 6(6): 983-986, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38822027
2.
Nat Commun ; 15(1): 1041, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310113

ABSTRACT

Cellular senescence is a stress response with broad pathophysiological implications. Senotherapies can induce senescence to treat cancer or eliminate senescent cells to ameliorate ageing and age-related pathologies. However, the success of senotherapies is limited by the lack of reliable ways to identify senescence. Here, we use nuclear morphology features of senescent cells to devise machine-learning classifiers that accurately predict senescence induced by diverse stressors in different cell types and tissues. As a proof-of-principle, we use these senescence classifiers to characterise senolytics and to screen for drugs that selectively induce senescence in cancer cells but not normal cells. Moreover, a tissue senescence score served to assess the efficacy of senolytic drugs and identified senescence in mouse models of liver cancer initiation, ageing, and fibrosis, and in patients with fatty liver disease. Thus, senescence classifiers can help to detect pathophysiological senescence and to discover and validate potential senotherapies.


Subject(s)
Aging , Cellular Senescence , Animals , Mice , Humans , Aging/physiology , Cellular Senescence/physiology , Fibrosis
3.
Nat Cell Biol ; 25(12): 1804-1820, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38012402

ABSTRACT

Drugs that selectively kill senescent cells (senolytics) improve the outcomes of cancer, fibrosis and age-related diseases. Despite their potential, our knowledge of the molecular pathways that affect the survival of senescent cells is limited. To discover senolytic targets, we performed RNAi screens and identified coatomer complex I (COPI) vesicle formation as a liability of senescent cells. Genetic or pharmacological inhibition of COPI results in Golgi dispersal, dysfunctional autophagy, and unfolded protein response-dependent apoptosis of senescent cells, and knockdown of COPI subunits improves the outcomes of cancer and fibrosis in mouse models. Drugs targeting COPI have poor pharmacological properties, but we find that N-myristoyltransferase inhibitors (NMTi) phenocopy COPI inhibition and are potent senolytics. NMTi selectively eliminated senescent cells and improved outcomes in models of cancer and non-alcoholic steatohepatitis. Our results suggest that senescent cells rely on a hyperactive secretory apparatus and that inhibiting trafficking kills senescent cells with the potential to treat various senescence-associated diseases.


Subject(s)
Neoplasms , Senotherapeutics , Mice , Animals , Golgi Apparatus/metabolism , Cellular Senescence , Neoplasms/metabolism , Fibrosis
4.
Trends Endocrinol Metab ; 34(12): 823-837, 2023 12.
Article in English | MEDLINE | ID: mdl-37673766

ABSTRACT

The liver plays a key role in sensing nutritional and hormonal inputs to maintain metabolic homeostasis. Recent studies into pre-mRNA splicing and alternative splicing (AS) and their effects on gene expression have revealed considerable transcriptional complexity in the liver, both in health and disease. While the contribution of these mechanisms to cell and tissue identity is widely accepted, their role in physiological and pathological contexts within tissues is just beginning to be appreciated. In this review, we showcase recent studies on the splicing and AS of key genes in metabolic pathways in the liver, the effect of metabolic signals on the spliceosome, and therapeutic intervention points based on RNA splicing.


Subject(s)
Liver Diseases , RNA Splicing , Humans , RNA Splicing/genetics , Alternative Splicing/genetics , Liver Diseases/genetics , Homeostasis/genetics
5.
Nature ; 621(7980): 821-829, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37586410

ABSTRACT

Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.


Subject(s)
Endothelial Cells , Receptors, Aryl Hydrocarbon , Humans , Animals , Mice , Receptors, Aryl Hydrocarbon/metabolism , Endothelial Cells/metabolism , Intestines , Signal Transduction , Homeostasis , Ligands
6.
Nanomedicine (Lond) ; 18(1): 67-84, 2023 01.
Article in English | MEDLINE | ID: mdl-36896958

ABSTRACT

Nanomedicines are revolutionizing healthcare as recently demonstrated by the Pfizer/BioNTech and Moderna COVID-2019 vaccines, with billions of doses administered worldwide in a safe manner. Nonalcoholic fatty liver disease is the most common noncommunicable chronic liver disease, posing a major growing challenge to global public health. However, due to unmet diagnostic and therapeutic needs, there is great interest in the development of novel translational approaches. Nanoparticle-based approaches offer novel opportunities for efficient and specific drug delivery to liver cells, as a step toward precision medicines. In this review, the authors highlight recent advances in nanomedicines for the generation of novel diagnostic and therapeutic tools for nonalcoholic fatty liver disease and related liver diseases.


Chronic liver diseases are a growing concern for global public health since they can affect up to 25% of the global adult population. Currently, there is no effective treatment or cure for these diseases. Nanometer-sized capsules can be loaded with drugs and more accurately deliver these drugs to their sites of action. They help improve the availability of medicines to the liver and have the potential to reduce their side effects. Here, the authors discuss recent advances to explain how nanotechnology can help improve the benefits of existing medicines for liver disease therapy.


Subject(s)
COVID-19 , Nanoparticles , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/diagnosis , Nanomedicine , Drug Delivery Systems , Nanoparticles/therapeutic use
7.
Biomaterials ; 293: 121982, 2023 02.
Article in English | MEDLINE | ID: mdl-36640555

ABSTRACT

Human pluripotent stem cell-derived hepatocytes (hPSC-Heps) may be suitable for treating liver diseases, but differentiation protocols often fail to yield adult-like cells. We hypothesised that replicating healthy liver niche biochemical and biophysical cues would produce hepatocytes with desired metabolic functionality. Using 2D synthetic hydrogels which independently control mechanical properties and biochemical cues, we found that culturing hPSC-Heps on surfaces matching the stiffness of fibrotic liver tissue upregulated expression of genes for RGD-binding integrins, and increased expression of YAP/TAZ and their transcriptional targets. Alternatively, culture on soft, healthy liver-like substrates drove increases in cytochrome p450 activity and ureagenesis. Knockdown of ITGB1 or reducing RGD-motif-containing peptide concentration in stiff hydrogels reduced YAP activity and improved metabolic functionality; however, on soft substrates, reducing RGD concentration had the opposite effect. Furthermore, targeting YAP activity with verteporfin or forskolin increased cytochrome p450 activity, with forskolin dramatically enhancing urea synthesis. hPSC-Heps could also be successfully encapsulated within RGD peptide-containing hydrogels without negatively impacting hepatic functionality, and compared to 2D cultures, 3D cultured hPSC-Heps secreted significantly less fetal liver-associated alpha-fetoprotein, suggesting furthered differentiation. Our platform overcomes technical hurdles in replicating the liver niche, and allowed us to identify a role for YAP/TAZ-mediated mechanosensing in hPSC-Hep differentiation.


Subject(s)
Hepatocytes , Oligopeptides , Humans , Colforsin/metabolism , Colforsin/pharmacology , Cell Differentiation , Oligopeptides/pharmacology , Oligopeptides/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/pharmacology , Hydrogels/chemistry
8.
Nat Metab ; 4(12): 1812-1829, 2022 12.
Article in English | MEDLINE | ID: mdl-36536133

ABSTRACT

RNA alternative splicing (AS) expands the regulatory potential of eukaryotic genomes. The mechanisms regulating liver-specific AS profiles and their contribution to liver function are poorly understood. Here, we identify a key role for the splicing factor RNA-binding Fox protein 2 (RBFOX2) in maintaining cholesterol homeostasis in a lipogenic environment in the liver. Using enhanced individual-nucleotide-resolution ultra-violet cross-linking and immunoprecipitation, we identify physiologically relevant targets of RBFOX2 in mouse liver, including the scavenger receptor class B type I (Scarb1). RBFOX2 function is decreased in the liver in diet-induced obesity, causing a Scarb1 isoform switch and alteration of hepatocyte lipid homeostasis. Our findings demonstrate that specific AS programmes actively maintain liver physiology, and underlie the lipotoxic effects of obesogenic diets when dysregulated. Splice-switching oligonucleotides targeting this network alleviate obesity-induced inflammation in the liver and promote an anti-atherogenic lipoprotein profile in the blood, underscoring the potential of isoform-specific RNA therapeutics for treating metabolism-associated diseases.


Subject(s)
Alternative Splicing , RNA-Binding Proteins , Mice , Animals , Alternative Splicing/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA/genetics , Liver/metabolism , Homeostasis , Cholesterol/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
9.
Proc Natl Acad Sci U S A ; 119(44): e2210434119, 2022 11.
Article in English | MEDLINE | ID: mdl-36282921

ABSTRACT

The cJun NH2-terminal kinase (JNK) signaling pathway in the liver promotes systemic changes in metabolism by regulating peroxisome proliferator-activated receptor α (PPARα)-dependent expression of the hepatokine fibroblast growth factor 21 (FGF21). Hepatocyte-specific gene ablation studies demonstrated that the Mapk9 gene (encoding JNK2) plays a key mechanistic role. Mutually exclusive inclusion of exons 7a and 7b yields expression of the isoforms JNK2α and JNK2ß. Here we demonstrate that Fgf21 gene expression and metabolic regulation are primarily regulated by the JNK2α isoform. To identify relevant substrates of JNK2α, we performed a quantitative phosphoproteomic study of livers isolated from control mice, mice with JNK deficiency in hepatocytes, and mice that express only JNK2α or JNK2ß in hepatocytes. We identified the JNK substrate retinoid X receptor α (RXRα) as a protein that exhibited JNK2α-promoted phosphorylation in vivo. RXRα functions as a heterodimeric partner of PPARα and may therefore mediate the effects of JNK2α signaling on Fgf21 expression. To test this hypothesis, we established mice with hepatocyte-specific expression of wild-type or mutated RXRα proteins. We found that the RXRα phosphorylation site Ser260 was required for suppression of Fgf21 gene expression. Collectively, these data establish a JNK-mediated signaling pathway that regulates hepatic Fgf21 expression.


Subject(s)
Metabolic Syndrome , PPAR alpha , Animals , Mice , Carrier Proteins/metabolism , Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , Liver/metabolism , Metabolic Syndrome/metabolism , Mice, Knockout , Phosphorylation , PPAR alpha/genetics , PPAR alpha/metabolism , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , MAP Kinase Kinase 4/metabolism
10.
Biomedicines ; 10(5)2022 May 19.
Article in English | MEDLINE | ID: mdl-35625914

ABSTRACT

Patients with high cholesterol and glucose levels are at high risk for cardiovascular disease. The Sterol Regulatory Element Binding Protein (SREBP) system regulates genes involved in lipid, cholesterol and glucose pathways. Autosomal Dominant Hypercholesterolemias (ADHs) are a group of diseases with increased cholesterol levels. They affect 1 out of every 500 individuals. About 20-30% of patients do not present any mutation in the known genes (LDLR, APOB and PCSK9). ADHs constitute a good model to identify the genes involved in the alteration of lipid levels or possible therapeutic targets. In this paper, we studied whether a mutation in the SREBP system could be responsible for ADH and other metabolic alterations present in these patients. Forty-one ADH patients without mutations in the main responsible genes were screened by direct sequencing of SREBP system genes. A luciferase reporter assay of the found mutation and an oral glucose tolerance test in carriers and non-carriers were performed. We found a novel mutation in the SREBF2 gene that increases transcription levels and cosegregates with hypercholesterolemia, and we found increased glucose levels in one family. SREBP2 is known to be involved in cholesterol synthesis, plasma levels and glucose metabolism in humans. The found mutation may involve the SREBF2 gene in hypercholesterolemia combined with hyperglycemia.

11.
Nucleic Acids Res ; 50(6): 3379-3393, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35293570

ABSTRACT

Pre-mRNA processing is an essential mechanism for the generation of mature mRNA and the regulation of gene expression in eukaryotic cells. While defects in pre-mRNA processing have been implicated in a number of diseases their involvement in metabolic pathologies is still unclear. Here, we show that both alternative splicing and alternative polyadenylation, two major steps in pre-mRNA processing, are significantly altered in non-alcoholic fatty liver disease (NAFLD). Moreover, we find that Serine and Arginine Rich Splicing Factor 10 (SRSF10) binding is enriched adjacent to consensus polyadenylation motifs and its expression is significantly decreased in NAFLD, suggesting a role mediating pre-mRNA dysregulation in this condition. Consistently, inactivation of SRSF10 in mouse and human hepatocytes in vitro, and in mouse liver in vivo, was found to dysregulate polyadenylation of key metabolic genes such as peroxisome proliferator-activated receptor alpha (PPARA) and exacerbate diet-induced metabolic dysfunction. Collectively our work implicates dysregulated pre-mRNA polyadenylation in obesity-induced liver disease and uncovers a novel role for SRSF10 in this process.


Subject(s)
Cell Cycle Proteins/metabolism , Non-alcoholic Fatty Liver Disease , Polyadenylation , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Animals , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing
12.
Genes Dev ; 35(5-6): 379-391, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33602872

ABSTRACT

Senescence is a key barrier to neoplastic transformation. To identify senescence regulators relevant to cancer, we screened a genome-wide shRNA library. Here, we describe exportin 7 (XPO7) as a novel regulator of senescence and validate its function in telomere-induced, replicative, and oncogene-induced senescence (OIS). XPO7 is a bidirectional transporter that regulates the nuclear-cytoplasmic shuttling of a broad range of substrates. Depletion of XPO7 results in reduced levels of TCF3 and an impaired induction of the cyclin-dependent kinase inhibitor p21CIP1 during OIS. Deletion of XPO7 correlates with poorer overall survival in several cancer types. Moreover, depletion of XPO7 alleviated OIS and increased tumor formation in a mouse model of liver cancer. Our results suggest that XPO7 is a novel tumor suppressor that regulates p21CIP1 expression to control senescence and tumorigenesis.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Karyopherins/genetics , Karyopherins/metabolism , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Humans , Mice , Neoplasms/physiopathology , Telomeric Repeat Binding Protein 2/genetics
13.
Methods Mol Biol ; 2164: 129-143, 2020.
Article in English | MEDLINE | ID: mdl-32607890

ABSTRACT

Transgenic mouse are reliable, convenient models for studying human hepatocellular carcinoma (HCC). The development of a synthetically engineered Sleeping Beauty (SB) transposon system further enables the viral-free, efficient delivery of desired oncogenes to mouse tissues. Here, we describe an SB transposon-based approach to induce HCC in mice by expressing a hyperactive form of N-RAS, N-RASG12V, while silencing the endogenous Trp53 gene via hydrodynamic tail vein injection, a method to rapidly deliver naked plasmids to mouse liver.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Transposable Elements/genetics , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Therapy/methods , Hydrodynamics , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmids/genetics , Tail
14.
Nat Metab ; 1(11): 1074-1088, 2019 11.
Article in English | MEDLINE | ID: mdl-31799499

ABSTRACT

Senescence is a cellular stress response that results in the stable arrest of old, damaged or preneoplastic cells. Oncogene-induced senescence is tumor suppressive but can also exacerbate tumorigenesis through the secretion of pro-inflammatory factors from senescent cells. Drugs that selectively kill senescent cells, termed senolytics, have proved beneficial in animal models of many age-associated diseases. Here, we show that the cardiac glycoside, ouabain, is a senolytic agent with broad activity. Senescent cells are sensitized to ouabain-induced apoptosis, a process mediated in part by induction of the pro-apoptotic Bcl2-family protein NOXA. We show that cardiac glycosides synergize with anti-cancer drugs to kill tumor cells and eliminate senescent cells that accumulate after irradiation or in old mice. Ouabain also eliminates senescent preneoplastic cells. Our findings suggest that cardiac glycosides may be effective anti-cancer drugs by acting through multiple mechanism. Given the broad range of senescent cells targeted by cardiac glycosides their use against age-related diseases warrants further exploration.


Subject(s)
Cardiac Glycosides/pharmacology , Cellular Senescence/drug effects , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Humans , Mice , Ouabain/pharmacology , Quercetin/pharmacology , Rats
15.
Mol Metab ; 30: 221-229, 2019 12.
Article in English | MEDLINE | ID: mdl-31767173

ABSTRACT

OBJECTIVE: Coupling metabolic and reproductive pathways is essential for the survival of species. However, the functions of steroidogenic enzymes expressed in metabolic tissues are largely unknown. METHODS AND RESULTS: Here, we show that in the liver, the classical steroidogenic enzyme Cyp17a1 forms an essential nexus for glucose and ketone metabolism during feed-fast cycles. Both gain- and loss-of-function approaches are used to show that hepatic Cyp17a1 is induced by fasting, catalyzes the production of at least one hormone-ligand (DHEA) for the nuclear receptor PPARα, and is ultimately required for maintaining euglycemia and ketogenesis during nutrient deprivation. The feedback-loop that terminates Cyp17a1-PPARα activity, and re-establishes anabolic liver metabolism during re-feeding is mapped to postprandial bile acid-signaling, involving the receptors FXR, SHP and LRH-1. CONCLUSIONS: Together, these findings represent a novel paradigm of homeostatic control in which nutritional cues feed-forward on to metabolic pathways by influencing extragonadal steroidogenesis.


Subject(s)
Liver/metabolism , PPAR alpha/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Animals , Bile Acids and Salts/metabolism , Glucose/metabolism , HEK293 Cells , Hepatocytes/metabolism , Homeostasis , Humans , Ketones/metabolism , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Steroid 17-alpha-Hydroxylase/physiology
16.
Autophagy ; 14(9): 1586-1595, 2018.
Article in English | MEDLINE | ID: mdl-29950132

ABSTRACT

Autophagy is required for cellular homeostasis and can determine cell viability in response to stress. It is established that MTOR is a master regulator of starvation-induced macroautophagy/autophagy, but recent studies have also implicated an essential role for the MAPK8/cJun NH2-terminal kinase 1 signal transduction pathway. We found that MAPK8/JNK1 and MAPK9/JNK2 were not required for autophagy caused by starvation or MTOR inhibition in murine fibroblasts and epithelial cells. These data demonstrate that MAPK8/9 has no required role in starvation-induced autophagy. We conclude that the role of MAPK8/9 in autophagy may be context-dependent and more complex than previously considered. ABBREVIATIONS: AKT: thymoma viral proto-oncogene;ALB: albumin; ATG4: autophagy related 4; BCL2: B cell leukemia/lymphoma 2; BECN1: beclin 1, autophagy related; BNIP3: BCL2/adenovirus E1B interacting protein 3; CQ: chloroquine diphosphate; DMEM: Dulbecco's modified Eagle's medium; EDTA: ethylenediaminetetraacetic acid; EBSS: Earle's balanced salt solution; FBS: fetal bovine serum; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HRAS: Harvey rat sarcoma virus oncogene; IgG: Immunoglobulin G; MAPK3/ERK1: mitogen-activated protein kinase 3; MAPK8/JNK1: mitogen-activated protein kinase 8; MAPK9/JNK2: mitogen-activated protein kinase 9; MAPK10/JNK3: mitogen-activated protein kinase 10; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MEFs: mouse embryonic fibroblasts; MTOR: mechanistic target of rapamycin kinase; RPS6KB1/p70: ribosomal protein S6 kinase, polypeptide 1; PPARA: peroxisome proliferator activated receptor alpha; SEM: standard error of the mean; SQSTM1/p62: sequestosome 1; TORC1: target of rapamycin complex 1; TORC2: target of rapamycin complex 2; TRP53: transforming related protein 53; TUBA: tubulin alpha; UV: ultraviolet; WT: wild-type.


Subject(s)
Amino Acids/deficiency , Autophagy , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Autophagy/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , MAP Kinase Signaling System/drug effects , Mice , Naphthyridines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism
17.
Sci Rep ; 7(1): 174, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28282965

ABSTRACT

Diet plays a crucial role in shaping human health and disease. Diets promoting obesity and insulin resistance can lead to severe metabolic diseases, while calorie-restricted (CR) diets can improve health and extend lifespan. In this work, we fed mice either a chow diet (CD), a 16 week high-fat diet (HFD), or a CR diet to compare and contrast the effects of these diets on mouse liver biology. We collected transcriptomic and epigenomic datasets from these mice using RNA-Seq and DNase-Seq. We found that both CR and HFD induce extensive transcriptional changes, in some cases altering the same genes in the same direction. We used our epigenomic data to infer transcriptional regulatory proteins bound near these genes that likely influence their expression levels. In particular, we found evidence for critical roles played by PPARα and RXRα. We used ChIP-Seq to profile the binding locations for these factors in HFD and CR livers. We found extensive binding of PPARα near genes involved in glycolysis/gluconeogenesis and uncovered a role for this factor in regulating anaerobic glycolysis. Overall, we generated extensive transcriptional and epigenomic datasets from livers of mice fed these diets and uncovered new functions and gene targets for PPARα.


Subject(s)
Caloric Restriction/adverse effects , Diet, High-Fat/adverse effects , Epigenomics/methods , Gene Expression Profiling/methods , Liver/chemistry , PPAR alpha/genetics , Anaerobiosis , Animals , Epigenesis, Genetic , Gene Expression Regulation , Glycolysis , Male , Mice , Nutritional Status , Sequence Analysis, DNA , Sequence Analysis, RNA
18.
Elife ; 52016 09 16.
Article in English | MEDLINE | ID: mdl-27635635

ABSTRACT

Alternative pre-mRNA splicing expands the complexity of the transcriptome and controls isoform-specific gene expression. Whether alternative splicing contributes to metabolic regulation is largely unknown. Here we investigated the contribution of alternative splicing to the development of diet-induced obesity. We found that obesity-induced changes in adipocyte gene expression include alternative pre-mRNA splicing. Bioinformatics analysis associated part of this alternative splicing program with sequence specific NOVA splicing factors. This conclusion was confirmed by studies of mice with NOVA deficiency in adipocytes. Phenotypic analysis of the NOVA-deficient mice demonstrated increased adipose tissue thermogenesis and improved glycemia. We show that NOVA proteins mediate a splicing program that suppresses adipose tissue thermogenesis. Together, these data provide quantitative analysis of gene expression at exon-level resolution in obesity and identify a novel mechanism that contributes to the regulation of adipose tissue function and the maintenance of normal glycemia.


Subject(s)
Adipose Tissue/physiology , Alternative Splicing , Antigens, Neoplasm/analysis , RNA-Binding Proteins/analysis , Thermogenesis , Animals , Antigens, Neoplasm/genetics , Computational Biology , Hyperglycemia , Mice, Inbred C57BL , Mice, Knockout , Neuro-Oncological Ventral Antigen , Obesity/physiopathology , RNA-Binding Proteins/genetics
19.
Nat Commun ; 7: 11365, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27098609

ABSTRACT

Adipose tissue de novo lipogenesis (DNL) positively influences insulin sensitivity, is reduced in obesity, and predicts insulin resistance. Therefore, elucidating mechanisms controlling adipose tissue DNL could lead to therapies for type 2 diabetes. Here, we report that mechanistic target of rapamycin complex 2 (mTORC2) functions in white adipose tissue (WAT) to control expression of the lipogenic transcription factor ChREBPß. Conditionally deleting the essential mTORC2 subunit Rictor in mature adipocytes decreases ChREBPß expression, which reduces DNL in WAT, and impairs hepatic insulin sensitivity. Mechanistically, Rictor/mTORC2 promotes ChREBPß expression in part by controlling glucose uptake, but without impairing pan-AKT signalling. High-fat diet also rapidly decreases adipose tissue ChREBPß expression and insulin sensitivity in wild-type mice, and does not further exacerbate insulin resistance in adipose tissue Rictor knockout mice, implicating adipose tissue DNL as an early target in diet-induced insulin resistance. These data suggest mTORC2 functions in WAT as part of an extra-hepatic nutrient-sensing mechanism to control glucose homeostasis.


Subject(s)
Adipose Tissue, White/metabolism , Carrier Proteins/genetics , Liver/metabolism , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Obesity/genetics , Protein Subunits/genetics , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/genetics , Adipose Tissue, White/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Carrier Proteins/metabolism , Diet, High-Fat/adverse effects , Female , Gene Deletion , Gene Expression Regulation , Glucose/metabolism , Insulin Resistance/genetics , Lipogenesis/genetics , Liver/pathology , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
20.
Cell Rep ; 14(10): 2273-80, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947074

ABSTRACT

The cJun NH2-terminal kinase (JNK)-signaling pathway is implicated in metabolic syndrome, including dysregulated blood glucose concentration and insulin resistance. Fibroblast growth factor 21 (FGF21) is a target of the hepatic JNK-signaling pathway and may contribute to the regulation of glycemia. To test the role of FGF21, we established mice with selective ablation of the Fgf21 gene in hepatocytes. FGF21 deficiency in the liver caused marked loss of FGF21 protein circulating in the blood. Moreover, the protective effects of hepatic JNK deficiency to suppress metabolic syndrome in high-fat diet-fed mice were not observed in mice with hepatocyte-specific FGF21 deficiency, including reduced blood glucose concentration and reduced intolerance to glucose and insulin. Furthermore, we show that JNK contributes to the regulation of hepatic FGF21 expression during fasting/feeding cycles. These data demonstrate that the hepatokine FGF21 is a key mediator of JNK-regulated metabolic syndrome.


Subject(s)
Fibroblast Growth Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Metabolic Diseases/etiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose/analysis , Cells, Cultured , Diet, High-Fat , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Insulin/blood , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Leptin/blood , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism , Protein Kinase Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Resistin/blood , Signal Transduction , Mitogen-Activated Protein Kinase Kinase Kinase 11
SELECTION OF CITATIONS
SEARCH DETAIL
...