Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 2457, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37117185

ABSTRACT

Understanding the factors and mechanisms involved in beta-cell development will guide therapeutic efforts to generate fully functional beta cells for diabetes. Neurogenin 3 (NGN3) is the key transcription factor that marks endocrine progenitors and drives beta-cell differentiation. Here we screen for binding partners of NGN3 and identify the deubiquitylating enzyme USP7 as a key regulator of NGN3 stability. Mechanistically, USP7 interacts with, deubiquitinates and stabilizes NGN3. In vivo, conditional knockout of Usp7 in the mouse embryonic pancreas causes a dramatic reduction in islet formation and hyperglycemia in adult mice, due to impaired NGN3-mediated endocrine specification during pancreatic development. Furthermore, pharmacological inhibition of USP7 during endocrine specification in human iPSC models of beta-cell differentiation decreases NGN3 expressing progenitor cell numbers and impairs beta cell differentiation. Thus, the USP7-NGN3 axis is an essential mechanism for driving endocrine development and beta-cell differentiation, which can be therapeutically exploited.


Subject(s)
Islets of Langerhans , Adult , Animals , Humans , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Islets of Langerhans/metabolism , Pancreas/metabolism , Transcription Factors/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/metabolism
2.
Arterioscler Thromb Vasc Biol ; 37(3): 423-432, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28082258

ABSTRACT

OBJECTIVE: The sterol-responsive nuclear receptors, liver X receptors α (LXRα, NR1H3) and ß (LXRß, NR1H2), are key determinants of cellular cholesterol homeostasis. LXRs are activated under conditions of high cellular sterol load and induce expression of the cholesterol efflux transporters ABCA1 and ABCG1 to promote efflux of excess cellular cholesterol. However, the full set of genes that contribute to LXR-stimulated cholesterol efflux is unknown, and their identification is the objective of this study. APPROACH AND RESULTS: We systematically compared the global transcriptional response of macrophages to distinct classes of LXR ligands. This allowed us to identify both common and ligand-specific transcriptional responses in macrophages. Among these, we identified endonuclease-exonuclease-phosphatase family domain containing 1 (EEPD1/KIAA1706) as a direct transcriptional target of LXRs in human and murine macrophages. EEPD1 specifically localizes to the plasma membrane owing to the presence of a myristoylation site in its N terminus. Accordingly, the first 10 amino acids of EEPD1 are sufficient to confer plasma membrane localization in the context of a chimeric protein with GFP. Functionally, we report that silencing expression of EEPD1 blunts maximal LXR-stimulated Apo AI-dependent efflux and demonstrate that this is the result of reduced abundance of ABCA1 protein in human and murine macrophages. CONCLUSIONS: In this study, we identify EEPD1 as a novel LXR-regulated gene in macrophages and propose that it promotes cellular cholesterol efflux by controlling cellular levels and activity of ABCA1.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cell Membrane/enzymology , Cholesterol/metabolism , Endodeoxyribonucleases/metabolism , Liver X Receptors/metabolism , Macrophages/enzymology , ATP Binding Cassette Transporter 1/genetics , Animals , Apolipoprotein A-I/metabolism , Biological Transport , COS Cells , Cell Membrane/drug effects , Chlorocebus aethiops , Endodeoxyribonucleases/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , HeLa Cells , Hep G2 Cells , Humans , Ligands , Liver X Receptors/agonists , Liver X Receptors/deficiency , Liver X Receptors/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , RNA Interference , Transcriptome , Transfection
3.
J Biol Chem ; 291(9): 4813-25, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26719329

ABSTRACT

Cholesterol metabolism is subject to complex transcriptional and nontranscriptional regulation. Herein, the role of ubiquitylation is emerging as an important post-translational modification that regulates cholesterol synthesis and uptake. Similar to other post-translational modifications, ubiquitylation is reversible in a process dependent on activity of deubiquitylating enzymes (DUBs). Yet whether these play a role in cholesterol metabolism is largely unknown. As a first step to test this possibility, we used pharmacological inhibition of cellular DUB activity. Short term (2 h) inhibition of DUBs resulted in accumulation of high molecular weight ubiquitylated proteins. This was accompanied by a dramatic decrease in abundance of the LDLR and attenuated LDL uptake into hepatic cells. Importantly, this occurred in the absence of changes in the mRNA levels of the LDLR or other SREBP2-regulated genes, in line with this phenotype being a post-transcriptional event. Mechanistically, we identify transcriptional induction of the E3 ubiquitin ligase IDOL in human and rodent cells as the underlying cause for ubiquitylation-dependent lysosomal degradation of the LDLR following DUB inhibition. In contrast to the established transcriptional regulation of IDOL by the sterol-responsive liver X receptor (LXR) transcription factors, induction of IDOL by DUB inhibition is LXR-independent and occurs in Lxrαß(-/-) MEFs. Consistent with the role of DUBs in transcriptional regulation, we identified a 70-bp region in the proximal promoter of IDOL, distinct from that containing the LXR-responsive element, which mediates the response to DUB inhibition. In conclusion, we identify a sterol-independent mechanism to regulate IDOL expression and IDOL-mediated lipoprotein receptor degradation.


Subject(s)
Lipoproteins, LDL/metabolism , Promoter Regions, Genetic , Receptors, LDL/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/metabolism , Absorption, Physiological/drug effects , Amino Acid Substitution , Animals , Cell Line , Cells, Cultured , Enzyme Inhibitors/pharmacology , Genes, Reporter , Human Umbilical Vein Endothelial Cells/cytology , Humans , Liver X Receptors , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/metabolism , Mice , Mutation , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic/drug effects , Proteolysis/drug effects , Receptors, LDL/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription, Genetic/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination/drug effects
4.
Circ Res ; 118(3): 410-9, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26666640

ABSTRACT

RATIONALE: The low-density lipoprotein (LDL) receptor (LDLR) is a central determinant of circulating LDL-cholesterol and as such subject to tight regulation. Recent studies and genetic evidence implicate the inducible degrader of the LDLR (IDOL) as a regulator of LDLR abundance and of circulating levels of LDL-cholesterol in humans. Acting as an E3-ubiquitin ligase, IDOL promotes ubiquitylation and subsequent lysosomal degradation of the LDLR. Consequently, inhibition of IDOL-mediated degradation of the LDLR represents a potential strategy to increase hepatic LDL-cholesterol clearance. OBJECTIVE: To establish whether deubiquitylases counteract IDOL-mediated ubiquitylation and degradation of the LDLR. METHODS AND RESULTS: Using a genetic screening approach, we identify the ubiquitin-specific protease 2 (USP2) as a post-transcriptional regulator of IDOL-mediated LDLR degradation. We demonstrate that both USP2 isoforms, USP2-69 and USP2-45, interact with IDOL and promote its deubiquitylation. IDOL deubiquitylation requires USP2 enzymatic activity and leads to a marked stabilization of IDOL protein. Paradoxically, this also markedly attenuates IDOL-mediated degradation of the LDLR and the ability of IDOL to limit LDL uptake into cells. Conversely, loss of USP2 reduces LDLR protein in an IDOL-dependent manner and limits LDL uptake. We identify a tri-partite complex encompassing IDOL, USP2, and LDLR and demonstrate that in this context USP2 promotes deubiquitylation of the LDLR and prevents its degradation. CONCLUSIONS: Our findings identify USP2 as a novel regulator of lipoprotein clearance owing to its ability to control ubiquitylation-dependent degradation of the LDLR by IDOL.


Subject(s)
Cholesterol, LDL/metabolism , Endopeptidases/metabolism , Receptors, LDL/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Endopeptidases/genetics , Enzyme Stability , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Mice, Knockout , Multienzyme Complexes , Protein Binding , Proteolysis , RNA Interference , Receptors, LDL/genetics , Transfection , Ubiquitin Thiolesterase , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...