Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
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
2.
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
3.
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
4.
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
5.
Genes Dev ; 27(21): 2345-55, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24186979

ABSTRACT

The cJun N-terminal kinase (JNK) signaling pathway is a key mediator of metabolic stress responses caused by consuming a high-fat diet, including the development of obesity. To test the role of JNK, we examined diet-induced obesity in mice with targeted ablation of Jnk genes in the anterior pituitary gland. These mice exhibited an increase in the pituitary expression of thyroid-stimulating hormone (TSH), an increase in the blood concentration of thyroid hormone (T4), increased energy expenditure, and markedly reduced obesity compared with control mice. The increased amount of pituitary TSH was caused by reduced expression of type 2 iodothyronine deiodinase (Dio2), a gene that is required for T4-mediated negative feedback regulation of TSH expression. These data establish a molecular mechanism that accounts for the regulation of energy expenditure and the development of obesity by the JNK signaling pathway.


Subject(s)
Diet, High-Fat , Iodide Peroxidase/metabolism , MAP Kinase Signaling System/physiology , Obesity/physiopathology , Animals , Energy Metabolism/genetics , Feedback, Physiological , Gene Expression Regulation , MAP Kinase Signaling System/genetics , Mice , Obesity/genetics , Pituitary Gland, Anterior/metabolism , Thyroid Hormones/metabolism , Iodothyronine Deiodinase Type II
6.
Hum Mol Genet ; 21(7): 1521-33, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22186026

ABSTRACT

Lafora disease (LD), a fatal neurodegenerative disorder characterized by the presence of intracellular inclusions called Lafora bodies (LBs), is caused by loss-of-function mutations in laforin or malin. Previous studies suggested a role of these proteins in the regulation of glycogen biosynthesis, in glycogen dephosphorylation and in the modulation of the intracellular proteolytic systems. However, the contribution of each of these processes to LD pathogenesis is unclear. We have generated a malin-deficient (Epm2b-/-) mouse with a phenotype similar to that of LD patients. By 3-6 months of age, Epm2b-/- mice present neurological and behavioral abnormalities that correlate with a massive presence of LBs in the cortex, hippocampus and cerebellum. Sixteen-day-old Epm2b-/- mice, without detectable LBs, show an impairment of macroautophagy (hereafter called autophagy), which remains compromised in adult animals. These data demonstrate similarities between the Epm2a-/- and Epm2b-/- mice that provide further insights into LD pathogenesis. They illustrate that the dysfunction of autophagy is a consequence of the lack of laforin-malin complexes and a common feature of both mouse models of LD. Because this dysfunction precedes other pathological manifestations, we propose that decreased autophagy plays a primary role in the formation of LBs and it is critical in LD pathogenesis.


Subject(s)
Autophagy , Lafora Disease/pathology , Ubiquitin-Protein Ligases/genetics , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Dual-Specificity Phosphatases/analysis , Dual-Specificity Phosphatases/metabolism , Glucans/chemistry , Lafora Disease/genetics , Lafora Disease/physiopathology , Mice , Mice, Knockout , Motor Skills Disorders/genetics , Myocardium/ultrastructure , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin/analysis , Ubiquitin-Protein Ligases/deficiency
7.
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
8.
Nat Aging ; 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210150

ABSTRACT

Inhibition of S6 kinase 1 (S6K1) extends lifespan and improves healthspan in mice, but the underlying mechanisms are unclear. Cellular senescence is a stable growth arrest accompanied by an inflammatory senescence-associated secretory phenotype (SASP). Cellular senescence and SASP-mediated chronic inflammation contribute to age-related pathology, but the specific role of S6K1 has not been determined. Here we show that S6K1 deletion does not reduce senescence but ameliorates inflammation in aged mouse livers. Using human and mouse models of senescence, we demonstrate that reduced inflammation is a liver-intrinsic effect associated with S6K deletion. Specifically, we show that S6K1 deletion results in reduced IRF3 activation; impaired production of cytokines, such as IL1ß; and reduced immune infiltration. Using either liver-specific or myeloid-specific S6K knockout mice, we also demonstrate that reduced immune infiltration and clearance of senescent cells is a hepatocyte-intrinsic phenomenon. Overall, deletion of S6K reduces inflammation in the liver, suggesting that suppression of the inflammatory SASP by loss of S6K could underlie the beneficial effects of inhibiting this pathway on healthspan and lifespan.

9.
Hum Mol Genet ; 20(13): 2571-84, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21493628

ABSTRACT

Laforin is a dual specificity protein phosphatase involved in Lafora disease (LD), a fatal form of progressive myoclonus epilepsy characterized by neurodegeneration and the presence of intracellular polyglucosan inclusions (Lafora bodies) in different tissues. In this work, we describe that mice lacking laforin (epm2a-/-) have enhanced insulin response leading to altered whole-body energy balance. This enhanced insulin response overactivates the Akt pathway which increases glucose uptake in the heart, resulting in increased glycogen levels and the formation of polyglucosan inclusions. In addition, enhanced insulin response resulted in increased liver lipid biosynthesis, resulting in hepatic steatosis. On the contrary, overexpression in rat hepatoma FTO2B cells of native laforin but not of a form lacking phosphatase activity (C266S) resulted in attenuation of insulin signaling. These results define laforin as a new regulator of insulin sensitivity, which provides novel insights into LD pathogenesis and identifies this phosphatase as a potential novel component of the insulin signaling cascade.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Energy Metabolism , Insulin/metabolism , Lafora Disease/enzymology , Animals , Cell Line, Tumor , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Energy Metabolism/genetics , Female , Glucose/metabolism , Lafora Disease/genetics , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Knockout , Motor Activity/genetics , Myocardium/metabolism , Protein Tyrosine Phosphatases, Non-Receptor , Rats , Signal Transduction/genetics
10.
Biochem Biophys Res Commun ; 431(1): 36-40, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23291237

ABSTRACT

The bifunctional enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2) catalyzes the synthesis and degradation of fructose-2,6-bisphosphate, a key metabolite in the glucose homeostasis. Four genes, Pfkfb1-4, have been characterized in mammals that code for several isoforms generated by alternative splicing through the control of several promoters and 5' non-coding exons. Here, we characterize in fetal rat liver new mRNA variants which are transcribed from a new Pfkfb1 gene promoter. The long variant codes to a new isoform (FL-PFK-2) that would be of relevant function to modulate the transition of fetal to adult liver metabolism.


Subject(s)
Alternative Splicing , Fetus/enzymology , Liver/enzymology , Phosphofructokinase-2/genetics , RNA, Messenger/genetics , Animals , HEK293 Cells , Humans , Liver/embryology , Promoter Regions, Genetic , Rats
11.
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
12.
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
13.
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
14.
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
15.
Hum Mol Genet ; 19(14): 2867-76, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20453062

ABSTRACT

Lafora disease (LD) is an autosomal recessive, progressive myoclonus epilepsy, which is characterized by the accumulation of polyglucosan inclusion bodies, called Lafora bodies, in the cytoplasm of cells in the central nervous system and in many other organs. However, it is unclear at the moment whether Lafora bodies are the cause of the disease, or whether they are secondary consequences of a primary metabolic alteration. Here we describe that the major genetic lesion that causes LD, loss-of-function of the protein laforin, impairs autophagy. This phenomenon is confirmed in cell lines from human patients, mouse embryonic fibroblasts from laforin knockout mice and in tissues from such mice. Conversely, laforin expression stimulates autophagy. Laforin regulates autophagy via the mammalian target of rapamycin kinase-dependent pathway. The changes in autophagy mediated by laforin regulate the accumulation of diverse autophagy substrates and would be predicted to impact on the Lafora body accumulation and the cell stress seen in this disease that may eventually contribute to cell death.


Subject(s)
Autophagy/genetics , Lafora Disease/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lafora Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/genetics , Mutant Proteins/physiology , Phagosomes/genetics , Phagosomes/metabolism , Protein Processing, Post-Translational/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
16.
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.

17.
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
18.
BMC Evol Biol ; 11: 225, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798009

ABSTRACT

BACKGROUND: Malin is an E3-ubiquitin ligase that is mutated in Lafora disease, a fatal form of progressive myoclonus epilepsy. In order to perform its function, malin forms a functional complex with laforin, a glucan phosphatase that facilitates targeting of malin to its corresponding substrates. While laforin phylogeny has been studied, there are no data on the evolutionary lineage of malin. RESULTS: After an extensive search for malin orthologs, we found that malin is present in all vertebrate species and a cephalochordate, in contrast with the broader species distribution previously reported for laforin. These data suggest that in addition to forming a functional complex, laforin and perhaps malin may also have independent functions. In addition, we found that malin shares significant identity with the E3-ubiquitin ligase TRIM32, which belongs to the tripartite-motif containing family of proteins. We present experimental evidence that both malin and TRIM32 share some substrates for ubiquitination, although they produce ubiquitin chains with different topologies. However, TRIM32-specific substrates were not reciprocally ubiquitinated by the laforin-malin complex. CONCLUSIONS: We found that malin and laforin are not conserved in the same genomes. In addition, we found that malin shares significant identity with the E3-ubiquitin ligase TRIM32. The latter result suggests a common origin for malin and TRIM32 and provides insights into possible functional relationships between both proteins.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Evolution, Molecular , Lafora Disease/enzymology , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Humans , Lafora Disease/genetics , Molecular Sequence Data , Phylogeny , Sequence Alignment , Transcription Factors/chemistry , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitination , Vertebrates/classification , Vertebrates/genetics
19.
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
20.
Nat Metab ; 6(6): 983-986, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38822027
SELECTION OF CITATIONS
SEARCH DETAIL