Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Metab ; 85: 101962, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815625

ABSTRACT

OBJECTIVE: p63 is a transcription factor involved in multiple biological functions. In the liver, the TAp63 isoform induces lipid accumulation in hepatocytes. However, the role of liver TAp63 in the progression of metabolic dysfunction-associated steatohepatitis (MASH) with fibrosis is unknown. METHODS: We evaluated the hepatic p63 levels in different mouse models of steatohepatitis with fibrosis induced by diet. Next, we used virogenetic approaches to manipulate the expression of TAp63 in adult mice under diet-induced steatohepatitis with fibrosis and characterized the disease condition. Finally, we performed proteomics analysis in mice with overexpression and knockdown of hepatic TAp63. RESULTS: Levels of TAp63, but not of ΔN isoform, are increased in the liver of mice with diet-induced steatohepatitis with fibrosis. Both preventive and interventional strategies for the knockdown of hepatic TAp63 significantly ameliorated diet-induced steatohepatitis with fibrosis in mice fed a methionine- and choline-deficient diet (MCDD) and choline deficient and high fat diet (CDHFD). The overexpression of hepatic TAp63 in mice aggravated the liver condition in mice fed a CDHFD. Proteomic analysis in the liver of these mice revealed alteration in multiple proteins and pathways, such as oxidative phosphorylation, antioxidant activity, peroxisome function and LDL clearance. CONCLUSIONS: These results indicate that liver TAp63 plays a critical role in the progression of diet-induced steatohepatitis with fibrosis, and its inhibition ameliorates the disease.


Subject(s)
Fatty Liver , Liver Cirrhosis , Liver , Mice, Inbred C57BL , Animals , Mice , Liver/metabolism , Liver/pathology , Male , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Disease Models, Animal , Diet, High-Fat/adverse effects , Trans-Activators/metabolism , Trans-Activators/genetics , Proteomics , Methionine/deficiency , Methionine/metabolism
2.
Hepatology ; 2024 May 19.
Article in English | MEDLINE | ID: mdl-38761407

ABSTRACT

BACKGROUND AND AIMS: Mitochondrial antiviral signaling protein (MAVS) is a critical regulator that activates the host's innate immunity against RNA viruses, and its signaling pathway has been linked to the secretion of proinflammatory cytokines. However, the actions of MAVS on inflammatory pathways during the development of metabolic dysfunction-associated steatotic liver disease (MASLD) have been little studied. APPROACH AND RESULTS: Liver proteomic analysis of mice with genetically manipulated hepatic p63, a transcription factor that induces liver steatosis, revealed MAVS as a target downstream of p63. MAVS was thus further evaluated in liver samples from patients and in animal models with MASLD. Genetic inhibition of MAVS was performed in hepatocyte cell lines, primary hepatocytes, spheroids, and mice. MAVS expression is induced in the liver of both animal models and people with MASLD as compared with those without liver disease. Using genetic knockdown of MAVS in adult mice ameliorates diet-induced MASLD. In vitro, silencing MAVS blunts oleic and palmitic acid-induced lipid content, while its overexpression increases the lipid load in hepatocytes. Inhibiting hepatic MAVS reduces circulating levels of the proinflammatory cytokine TNFα and the hepatic expression of both TNFα and NFκß. Moreover, the inhibition of ERK abolished the activation of TNFα induced by MAVS. The posttranslational modification O -GlcNAcylation of MAVS is required to activate inflammation and to promote the high lipid content in hepatocytes. CONCLUSIONS: MAVS is involved in the development of steatosis, and its inhibition in previously damaged hepatocytes can ameliorate MASLD.

3.
Cell Metab ; 35(9): 1630-1645.e5, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37541251

ABSTRACT

Neddylation is a post-translational mechanism that adds a ubiquitin-like protein, namely neural precursor cell expressed developmentally downregulated protein 8 (NEDD8). Here, we show that neddylation in mouse liver is modulated by nutrient availability. Inhibition of neddylation in mouse liver reduces gluconeogenic capacity and the hyperglycemic actions of counter-regulatory hormones. Furthermore, people with type 2 diabetes display elevated hepatic neddylation levels. Mechanistically, fasting or caloric restriction of mice leads to neddylation of phosphoenolpyruvate carboxykinase 1 (PCK1) at three lysine residues-K278, K342, and K387. We find that mutating the three PCK1 lysines that are neddylated reduces their gluconeogenic activity rate. Molecular dynamics simulations show that neddylation of PCK1 could re-position two loops surrounding the catalytic center into an open configuration, rendering the catalytic center more accessible. Our study reveals that neddylation of PCK1 provides a finely tuned mechanism of controlling glucose metabolism by linking whole nutrient availability to metabolic homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Mice , Animals , Phosphoenolpyruvate/metabolism , Diabetes Mellitus, Type 2/metabolism , Proteins/metabolism , Liver/metabolism , Lysine/metabolism , Glucose/metabolism
4.
Mol Metab ; 75: 101776, 2023 09.
Article in English | MEDLINE | ID: mdl-37453647

ABSTRACT

OBJECTIVE: O-GlcNAcylation is a post-translational modification that directly couples the processes of nutrient sensing, metabolism, and signal transduction, affecting protein function and localization, since the O-linked N-acetylglucosamine moiety comes directly from the metabolism of glucose, lipids, and amino acids. The addition and removal of O-GlcNAc of target proteins are mediated by two highly conserved enzymes: O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) and O-GlcNAcase (OGA), respectively. Deregulation of O-GlcNAcylation has been reported to be associated with various human diseases such as cancer, diabetes, and cardiovascular diseases. The contribution of deregulated O-GlcNAcylation to the progression and pathogenesis of NAFLD remains intriguing, and a better understanding of its roles in this pathophysiological context is required to uncover novel avenues for therapeutic intervention. By using a translational approach, our aim is to describe the role of OGT and O-GlcNAcylation in the pathogenesis of NAFLD. METHODS: We used primary mouse hepatocytes, human hepatic cell lines and in vivo mouse models of steatohepatitis to manipulate O-GlcNAc transferase (OGT). We also studied OGT and O-GlcNAcylation in liver samples from different cohorts of people with NAFLD. RESULTS: O-GlcNAcylation was upregulated in the liver of people and animal models with steatohepatitis. Downregulation of OGT in NAFLD-hepatocytes improved diet-induced liver injury in both in vivo and in vitro models. Proteomics studies revealed that mitochondrial proteins were hyper-O-GlcNAcylated in the liver of mice with steatohepatitis. Inhibition of OGT is able to restore mitochondrial oxidation and decrease hepatic lipid content in in vitro and in vivo models of NAFLD. CONCLUSIONS: These results demonstrate that deregulated hyper-O-GlcNAcylation favors NAFLD progression by reducing mitochondrial oxidation and promoting hepatic lipid accumulation.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Down-Regulation , Acetylglucosamine/metabolism , Mitochondria/metabolism , Hepatocytes/metabolism , Lipids
5.
Biochem Soc Trans ; 51(2): 783-796, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37013972

ABSTRACT

Ageing is a conserved and unavoidable biological process characterized by progressive decline of physiological functions with time. Despite constituting the greatest risk factor for most human diseases, little is known about the molecular mechanisms driving the ageing process. More than 170 chemical RNA modifications, also known as the epitranscriptome, decorate eukaryotic coding and non-coding RNAs and have emerged as novel regulators of RNA metabolism, modulating RNA stability, translation, splicing or non-coding RNA processing. Studies on short-lived organisms such as yeast or worms connect mutations on RNA modifying enzymes with lifespan changes, and dysregulation of the epitranscriptome has been linked to age-related diseases and ageing hallmarks themselves in mammals. Moreover, transcriptome-wide analyses are starting to reveal changes in messenger RNA modifications in neurodegenerative diseases and in the expression of some RNA modifiers with age. These studies are starting to put the focus on the epitranscriptome as a potential novel regulator of ageing and lifespan, and open new avenues for the identification of targets to treat age-related diseases. In this review, we discuss the connection between RNA modifications and the enzymatic machinery regulating their deposition in coding and non-coding RNAs, and ageing and hypothesize about the potential role of RNA modifications in the regulation of other ncRNAs playing a key role in ageing, such as transposable elements and tRNA fragments. Finally, we reanalyze available datasets of mouse tissues during ageing and report a wide transcriptional dysregulation of proteins involved in the deposition, removal or decoding of several of the best-known RNA modifications.


Subject(s)
RNA , Transcriptome , Humans , Animals , Mice , RNA/metabolism , RNA, Transfer/metabolism , Gene Expression Profiling , RNA Processing, Post-Transcriptional , Mammals/metabolism
7.
Cell Reprogram ; 25(1): 7-8, 2023 02.
Article in English | MEDLINE | ID: mdl-36695734

ABSTRACT

Single-cell transcriptomics and in situ imaging of murine pancreas upon partial reprogramming in vivo reveal transcriptional dynamics upon Oct4, Sox2, Klf4, and cMyc (OSKM) induction. Interestingly, transcriptomic signatures of partial reprogramming observed in pancreas are shared by several tissues upon OSKM induction as well as during in vitro reprogramming of fibroblasts, pointing to the existence of conserved pathways critical for early reprogramming, regeneration, and rejuvenation.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Animals , Mice , Fibroblasts/metabolism , Gene Expression Profiling , Transcriptome , Induced Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , Octamer Transcription Factor-3/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
8.
Gut ; 72(3): 472-483, 2023 03.
Article in English | MEDLINE | ID: mdl-35580962

ABSTRACT

OBJECTIVE: p63 is a transcription factor within the p53 protein family that has key roles in development, differentiation and prevention of senescence, but its metabolic actions remain largely unknown. Herein, we investigated the physiological role of p63 in glucose metabolism. DESIGN: We used cell lines and mouse models to genetically manipulate p63 in hepatocytes. We also measured p63 in the liver of patients with obesity with or without type 2 diabetes (T2D). RESULTS: We show that hepatic p63 expression is reduced on fasting. Mice lacking the specific isoform TAp63 in the liver (p63LKO) display higher postprandial and pyruvate-induced glucose excursions. These mice have elevated SIRT1 levels, while SIRT1 knockdown in p63LKO mice normalises glycaemia. Overexpression of TAp63 in wild-type mice reduces postprandial, pyruvate-induced blood glucose and SIRT1 levels. Studies carried out in hepatocyte cell lines show that TAp63 regulates SIRT1 promoter by repressing its transcriptional activation. TAp63 also mediates the inhibitory effect of insulin on hepatic glucose production, as silencing TAp63 impairs insulin sensitivity. Finally, protein levels of TAp63 are reduced in obese persons with T2D and are negatively correlated with fasting glucose and homeostasis model assessment index. CONCLUSIONS: These results demonstrate that p63 physiologically regulates glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Sirtuin 1 , Trans-Activators , Animals , Mice , Glucose/metabolism , Liver/metabolism , Pyruvates/metabolism , Sirtuin 1/metabolism , Trans-Activators/metabolism
9.
Nat Metab ; 4(7): 901-917, 2022 07.
Article in English | MEDLINE | ID: mdl-35879461

ABSTRACT

Early-life determinants are thought to be a major factor in the rapid increase of obesity. However, while maternal nutrition has been extensively studied, the effects of breastfeeding by the infant on the reprogramming of energy balance in childhood and throughout adulthood remain largely unknown. Here we show that delayed weaning in rat pups protects them against diet-induced obesity in adulthood, through enhanced brown adipose tissue thermogenesis and energy expenditure. In-depth metabolic phenotyping in this rat model as well as in transgenic mice reveals that the effects of prolonged suckling are mediated by increased hepatic fibroblast growth factor 21 (FGF21) production and tanycyte-controlled access to the hypothalamus in adulthood. Specifically, FGF21 activates GABA-containing neurons expressing dopamine receptor 2 in the lateral hypothalamic area and zona incerta. Prolonged breastfeeding thus constitutes a protective mechanism against obesity by affecting long-lasting physiological changes in liver-to-hypothalamus communication and hypothalamic metabolic regulation.


Subject(s)
Breast Feeding , Obesity , Animals , Female , Fibroblast Growth Factors , Humans , Hypothalamus/metabolism , Liver/metabolism , Mice , Obesity/metabolism , Obesity/prevention & control , Rats
10.
Cell Rep ; 39(10): 110928, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35675764

ABSTRACT

TET1 maintains hypomethylation at bivalent promoters through its catalytic activity in embryonic stem cells (ESCs). However, TET1 catalytic activity-independent function in regulating bivalent genes is not well understood. Using a proteomics approach, we map the TET1 interactome in ESCs and identify PSPC1 as a TET1 partner. Genome-wide location analysis reveals that PSPC1 functionally associates with TET1 and Polycomb repressive complex-2 (PRC2). We establish that PSPC1 and TET1 repress, and the lncRNA Neat1 activates, bivalent gene expression. In ESCs, Neat1 is preferentially bound to PSPC1 alongside its PRC2 association at bivalent promoters. During the ESC-to-epiblast-like stem cell (EpiLC) transition, PSPC1 and TET1 maintain PRC2 chromatin occupancy at bivalent gene promoters, while Neat1 facilitates the activation of certain bivalent genes by promoting PRC2 binding to their mRNAs. Our study demonstrates a TET1-PSPC1-Neat1 molecular axis that modulates PRC2-binding affinity to chromatin and bivalent gene transcripts in controlling stem cell bivalency.


Subject(s)
Embryonic Stem Cells , Polycomb Repressive Complex 2 , Cell Differentiation/genetics , Chromatin/metabolism , DNA Methylation , Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics
11.
Methods Mol Biol ; 2454: 743-754, 2022.
Article in English | MEDLINE | ID: mdl-33689163

ABSTRACT

The reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) has proven to be a powerful system creating new opportunities to interrogate molecular mechanisms controlling cell fate determination. Under standard conditions, the generation of iPSCs upon overexpression of OCT4, SOX2, KLF4, and c-MYC (OSKM) is generally slow and inefficient due to the presence of barriers that confer resistance to cell fate changes. Hyperactivated endoplasmic reticulum (ER) stress has emerged as a major reprogramming barrier that impedes the initial mesenchymal-to-epithelial transition (MET) step to form iPSCs from mesenchymal somatic cells. Here, we describe several systems to detect ER stress in the context of OSKM reprogramming and chemical interventions to modulate this process for improving iPSC formation.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Cellular Reprogramming/genetics , Octamer Transcription Factor-3/genetics
12.
Methods Mol Biol ; 2520: 199-213, 2022.
Article in English | MEDLINE | ID: mdl-34611821

ABSTRACT

Embryonic stem cells (ESCs) are derived from the inner cell mass of the preimplantation blastocyst and can be maintained indefinitely in vitro without losing their properties. Given their self-renewal and pluripotency, ESCs not only represent a key tool to study early embryonic development in a dish, but also an unlimited source of material for tissue replacement in regenerative medicine. Loss-of-function assays using RNA interference are a powerful tool to understand the roles of specific genes and are facilitated by lentiviral-mediated delivery of vector-encoded shRNAs which allows long-term silencing of single or multiple genes. Here, we describe the steps for rapid and cost-effective production and testing of lentiviral particles with vector-encoded shRNAs for gene silencing in ESCs. This protocol can be easily adapted for loss-of-function assays in other pluripotent cells or culture conditions of interest.


Subject(s)
Embryonic Stem Cells , Pluripotent Stem Cells , Cell Differentiation/genetics , Cost-Benefit Analysis , Gene Silencing , RNA, Small Interfering/genetics
13.
J Hepatol ; 76(1): 11-24, 2022 01.
Article in English | MEDLINE | ID: mdl-34555423

ABSTRACT

BACKGROUND & AIMS: Autophagy-related gene 3 (ATG3) is an enzyme mainly known for its actions in the LC3 lipidation process, which is essential for autophagy. Whether ATG3 plays a role in lipid metabolism or contributes to non-alcoholic fatty liver disease (NAFLD) remains unknown. METHODS: By performing proteomic analysis on livers from mice with genetic manipulation of hepatic p63, a regulator of fatty acid metabolism, we identified ATG3 as a new target downstream of p63. ATG3 was evaluated in liver samples from patients with NAFLD. Further, genetic manipulation of ATG3 was performed in human hepatocyte cell lines, primary hepatocytes and in the livers of mice. RESULTS: ATG3 expression is induced in the liver of animal models and patients with NAFLD (both steatosis and non-alcoholic steatohepatitis) compared with those without liver disease. Moreover, genetic knockdown of ATG3 in mice and human hepatocytes ameliorates p63- and diet-induced steatosis, while its overexpression increases the lipid load in hepatocytes. The inhibition of hepatic ATG3 improves fatty acid metabolism by reducing c-Jun N-terminal protein kinase 1 (JNK1), which increases sirtuin 1 (SIRT1), carnitine palmitoyltransferase 1a (CPT1a), and mitochondrial function. Hepatic knockdown of SIRT1 and CPT1a blunts the effects of ATG3 on mitochondrial activity. Unexpectedly, these effects are independent of an autophagic action. CONCLUSIONS: Collectively, these findings indicate that ATG3 is a novel protein implicated in the development of steatosis. LAY SUMMARY: We show that autophagy-related gene 3 (ATG3) contributes to the progression of non-alcoholic fatty liver disease in humans and mice. Hepatic knockdown of ATG3 ameliorates the development of NAFLD by stimulating mitochondrial function. Thus, ATG3 is an important factor implicated in steatosis.


Subject(s)
Autophagy-Related Proteins/antagonists & inhibitors , Fatty Liver/prevention & control , Mitochondria, Liver/metabolism , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Animals , Autophagy-Related Proteins/pharmacology , Disease Models, Animal , Fatty Liver/physiopathology , Lipid Metabolism/genetics , Mice , Mitochondria, Liver/physiology , Proteomics/methods , Ubiquitin-Conjugating Enzymes/pharmacology
14.
Nat Commun ; 12(1): 5068, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417460

ABSTRACT

p53 regulates several signaling pathways to maintain the metabolic homeostasis of cells and modulates the cellular response to stress. Deficiency or excess of nutrients causes cellular metabolic stress, and we hypothesized that p53 could be linked to glucose maintenance. We show here that upon starvation hepatic p53 is stabilized by O-GlcNAcylation and plays an essential role in the physiological regulation of glucose homeostasis. More specifically, p53 binds to PCK1 promoter and regulates its transcriptional activation, thereby controlling hepatic glucose production. Mice lacking p53 in the liver show a reduced gluconeogenic response during calorie restriction. Glucagon, adrenaline and glucocorticoids augment protein levels of p53, and administration of these hormones to p53 deficient human hepatocytes and to liver-specific p53 deficient mice fails to increase glucose levels. Moreover, insulin decreases p53 levels, and over-expression of p53 impairs insulin sensitivity. Finally, protein levels of p53, as well as genes responsible of O-GlcNAcylation are elevated in the liver of type 2 diabetic patients and positively correlate with glucose and HOMA-IR. Overall these results indicate that the O-GlcNAcylation of p53 plays an unsuspected key role regulating in vivo glucose homeostasis.


Subject(s)
Acetylglucosamine/metabolism , Glucose/metabolism , Liver/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Caloric Restriction , Cell Line , Colforsin/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Epinephrine/metabolism , Glucagon/metabolism , Glucocorticoids/metabolism , Gluconeogenesis/drug effects , Glycosylation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrocortisone/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Insulin Resistance , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Obesity/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Stability/drug effects , Pyruvic Acid/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/genetics
15.
Cell Stem Cell ; 28(10): 1868-1883.e11, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34038708

ABSTRACT

Topological-associated domains (TADs) are thought to be relatively stable across cell types, although some TAD reorganization has been observed during cellular differentiation. However, little is known about the mechanisms through which TAD reorganization affects cell fate or how master transcription factors affect TAD structures during cell fate transitions. Here, we show extensive TAD reorganization during somatic cell reprogramming, which is correlated with gene transcription and changes in cellular identity. Manipulating TAD reorganization promotes reprogramming, and the dynamics of concentrated chromatin loops in OCT4 phase separated condensates contribute to TAD reorganization. Disrupting OCT4 phase separation attenuates TAD reorganization and reprogramming, which can be rescued by fusing an intrinsically disordered region (IDR) to OCT4. We developed an approach termed TAD reorganization-based multiomics analysis (TADMAN), which identified reprogramming regulators. Together, these findings elucidate a role and mechanism of TAD reorganization, regulated by OCT4 phase separation, in cellular reprogramming.


Subject(s)
Cellular Reprogramming , Chromatin , Octamer Transcription Factor-3/metabolism , Cell Differentiation
16.
Hepatology ; 73(2): 606-624, 2021 02.
Article in English | MEDLINE | ID: mdl-32329085

ABSTRACT

BACKGROUND AND AIMS: G protein-coupled receptor (GPR) 55 is a putative cannabinoid receptor, and l-α-lysophosphatidylinositol (LPI) is its only known endogenous ligand. Although GPR55 has been linked to energy homeostasis in different organs, its specific role in lipid metabolism in the liver and its contribution to the pathophysiology of nonalcoholic fatty liver disease (NAFLD) remains unknown. APPROACH AND RESULTS: We measured (1) GPR55 expression in the liver of patients with NAFLD compared with individuals without obesity and without liver disease, as well as animal models with steatosis and nonalcoholic steatohepatitis (NASH), and (2) the effects of LPI and genetic disruption of GPR55 in mice, human hepatocytes, and human hepatic stellate cells. Notably, we found that circulating LPI and liver expression of GPR55 were up-regulated in patients with NASH. LPI induced adenosine monophosphate-activated protein kinase activation of acetyl-coenzyme A carboxylase (ACC) and increased lipid content in human hepatocytes and in the liver of treated mice by inducing de novo lipogenesis and decreasing ß-oxidation. The inhibition of GPR55 and ACCα blocked the effects of LPI, and the in vivo knockdown of GPR55 was sufficient to improve liver damage in mice fed a high-fat diet and in mice fed a methionine-choline-deficient diet. Finally, LPI promoted the initiation of hepatic stellate cell activation by stimulating GPR55 and activation of ACC. CONCLUSIONS: The LPI/GPR55 system plays a role in the development of NAFLD and NASH by activating ACC.


Subject(s)
Lysophospholipids/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Receptors, Cannabinoid/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Adult , Aged , Animals , Biopsy , Cannabinoid Receptor Agonists/pharmacology , Cell Line , Cohort Studies , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Knockdown Techniques , Hepatic Stellate Cells , Hepatocytes , Humans , Lipogenesis/drug effects , Liver/pathology , Lysophospholipids/blood , Male , Mice , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/blood , Obesity/metabolism , Receptors, Cannabinoid/genetics , Up-Regulation
17.
Nat Commun ; 11(1): 4956, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009383

ABSTRACT

Tet-enzyme-mediated 5-hydroxymethylation of cytosines in DNA plays a crucial role in mouse embryonic stem cells (ESCs). In RNA also, 5-hydroxymethylcytosine (5hmC) has recently been evidenced, but its physiological roles are still largely unknown. Here we show the contribution and function of this mark in mouse ESCs and differentiating embryoid bodies. Transcriptome-wide mapping in ESCs reveals hundreds of messenger RNAs marked by 5hmC at sites characterized by a defined unique consensus sequence and particular features. During differentiation a large number of transcripts, including many encoding key pluripotency-related factors (such as Eed and Jarid2), show decreased cytosine hydroxymethylation. Using Tet-knockout ESCs, we find Tet enzymes to be partly responsible for deposition of 5hmC in mRNA. A transcriptome-wide search further reveals mRNA targets to which Tet1 and Tet2 bind, at sites showing a topology similar to that of 5hmC sites. Tet-mediated RNA hydroxymethylation is found to reduce the stability of crucial pluripotency-promoting transcripts. We propose that RNA cytosine 5-hydroxymethylation by Tets is a mark of transcriptome flexibility, inextricably linked to the balance between pluripotency and lineage commitment.


Subject(s)
5-Methylcytosine/analogs & derivatives , Cell Differentiation , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , RNA/metabolism , 5-Methylcytosine/metabolism , Animals , Antibody Specificity/immunology , Base Sequence , Dioxygenases , Embryoid Bodies/metabolism , Mice , Models, Biological , Pluripotent Stem Cells/metabolism , Protein Binding , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics
18.
STAR Protoc ; 1(2)2020 09 18.
Article in English | MEDLINE | ID: mdl-32995755

ABSTRACT

RNA-binding proteins are key regulators of cell identity and function, which underscores the need for unbiased and versatile protocols to identify and characterize novel protein-RNA interactions. Here, we describe a simple and cost-effective in vitro RNA immunoprecipitation (iv-RIP) method to assess the direct or indirect RNA-binding ability of any protein of interest. The versatility of this method relies on the adaptability of the immunoprecipitation conditions and the choice of the RNA, which exponentially broadens the range of potential applications. For complete details on the use and execution of this protocol, please refer to Guallar et al. (2020).


Subject(s)
Immunoprecipitation/methods , RNA-Binding Proteins , RNA , Animals , Mice , Protein Binding , RNA/analysis , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
19.
Cell Stem Cell ; 27(2): 300-314.e11, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32396862

ABSTRACT

RNA editing of adenosine to inosine (A to I) is catalyzed by ADAR1 and dramatically alters the cellular transcriptome, although its functional roles in somatic cell reprogramming are largely unexplored. Here, we show that loss of ADAR1-mediated A-to-I editing disrupts mesenchymal-to-epithelial transition (MET) during induced pluripotent stem cell (iPSC) reprogramming and impedes acquisition of induced pluripotency. Using chemical and genetic approaches, we show that absence of ADAR1-dependent RNA editing induces aberrant innate immune responses through the double-stranded RNA (dsRNA) sensor MDA5, unleashing endoplasmic reticulum (ER) stress and hindering epithelial fate acquisition. We found that A-to-I editing impedes MDA5 sensing and sequestration of dsRNAs encoding membrane proteins, which promote ER homeostasis by activating the PERK-dependent unfolded protein response pathway to consequently facilitate MET. This study therefore establishes a critical role for ADAR1 and its A-to-I editing activity during cell fate transitions and delineates a key regulatory layer underlying MET to control efficient reprogramming.


Subject(s)
Induced Pluripotent Stem Cells , RNA Editing , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Induced Pluripotent Stem Cells/metabolism , Inosine/metabolism , RNA, Double-Stranded
20.
Life Sci Alliance ; 3(5)2020 05.
Article in English | MEDLINE | ID: mdl-32284354

ABSTRACT

BMAL1 is essential for the regulation of circadian rhythms in differentiated cells and adult stem cells, but the molecular underpinnings of its function in pluripotent cells, which hold a great potential in regenerative medicine, remain to be addressed. Here, using transient and permanent loss-of-function approaches in mouse embryonic stem cells (ESCs), we reveal that although BMAL1 is dispensable for the maintenance of the pluripotent state, its depletion leads to deregulation of transcriptional programs linked to cell differentiation commitment. We further confirm that depletion of Bmal1 alters the differentiation potential of ESCs in vitro. Mechanistically, we demonstrate that BMAL1 participates in the regulation of energy metabolism maintaining a low mitochondrial function which is associated with pluripotency. Loss-of-function of Bmal1 leads to the deregulation of metabolic gene expression associated with a shift from glycolytic to oxidative metabolism. Our results highlight the important role that BMAL1 plays at the exit of pluripotency in vitro and provide evidence implicating a non-canonical circadian function of BMAL1 in the metabolic control for cell fate determination.


Subject(s)
ARNTL Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , ARNTL Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Circadian Rhythm/genetics , Energy Metabolism/physiology , Gene Expression/genetics , Glycolysis , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mitochondria/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...