Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 387(23): 2113-2125, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36477031

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors and targeted therapies have dramatically improved outcomes in patients with advanced melanoma, but approximately half these patients will not have a durable benefit. Phase 1-2 trials of adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) have shown promising responses, but data from phase 3 trials are lacking to determine the role of TILs in treating advanced melanoma. METHODS: In this phase 3, multicenter, open-label trial, we randomly assigned patients with unresectable stage IIIC or IV melanoma in a 1:1 ratio to receive TIL or anti-cytotoxic T-lymphocyte antigen 4 therapy (ipilimumab at 3 mg per kilogram of body weight). Infusion of at least 5×109 TILs was preceded by nonmyeloablative, lymphodepleting chemotherapy (cyclophosphamide plus fludarabine) and followed by high-dose interleukin-2. The primary end point was progression-free survival. RESULTS: A total of 168 patients (86% with disease refractory to anti-programmed death 1 treatment) were assigned to receive TILs (84 patients) or ipilimumab (84 patients). In the intention-to-treat population, median progression-free survival was 7.2 months (95% confidence interval [CI], 4.2 to 13.1) in the TIL group and 3.1 months (95% CI, 3.0 to 4.3) in the ipilimumab group (hazard ratio for progression or death, 0.50; 95% CI, 0.35 to 0.72; P<0.001); 49% (95% CI, 38 to 60) and 21% (95% CI, 13 to 32) of the patients, respectively, had an objective response. Median overall survival was 25.8 months (95% CI, 18.2 to not reached) in the TIL group and 18.9 months (95% CI, 13.8 to 32.6) in the ipilimumab group. Treatment-related adverse events of grade 3 or higher occurred in all patients who received TILs and in 57% of those who received ipilimumab; in the TIL group, these events were mainly chemotherapy-related myelosuppression. CONCLUSIONS: In patients with advanced melanoma, progression-free survival was significantly longer among those who received TIL therapy than among those who received ipilimumab. (Funded by the Dutch Cancer Society and others; ClinicalTrials.gov number, NCT02278887.).


Subject(s)
Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating , Melanoma , Humans , Cell- and Tissue-Based Therapy , Ipilimumab/adverse effects , Melanoma/drug therapy
2.
Atherosclerosis ; 315: 1-9, 2020 12.
Article in English | MEDLINE | ID: mdl-33190106

ABSTRACT

BACKGROUND AND AIMS: Cholesterol metabolism is tightly regulated by transcriptional and post-transcriptional mechanisms. Accordingly, dysregulation of cholesterol metabolism is a major risk factor for the development of coronary artery disease and associated complications. In recent years, it has become apparent that next to the liver, the intestine plays a key role in systemic cholesterol metabolism by governing cholesterol absorption, secretion, and incorporation into lipoprotein particles. We have previously demonstrated that the Liver X receptor (LXR)-regulated E3 ubiquitin ligase inducible degrader of LDLR (IDOL) is a regulator of cholesterol uptake owing to its ability to promote the ubiquitylation of the low-density lipoprotein receptor (LDLR). However, whether the LXR-IDOL-LDLR axis regulates the LDLR in the intestine and whether this influences intestinal cholesterol homeostasis is not known. METHODS: In this study, we evaluated the role of the LXR-IDOL-LDLR axis in enterocyte cell models and in primary enterocytes isolated from Idol(-/-) and wild type mice. Furthermore, we studied the regulation of intestinal LDLR in Idol(-/-) and in wild type mice treated with the LXR agonist GW3965. Finally, we assessed ezetimibe-induced trans-intestinal cholesterol efflux in Idol(-/-) mice. RESULTS: We show that in a wide range of intestinal cell lines LXR activation decreases LDLR protein abundance, cell surface occupancy, and LDL uptake in an IDOL-dependent manner. Similarly, we find that pharmacological dosing of C57BL6/N mice with the LXR agonist GW3965 increases Idol expression across the intestine with a concomitant reduction in Ldlr protein. Conversely, primary enterocytes isolated from Idol(-/-) mice have elevated Ldlr. To test whether these changes contribute to trans-intestinal cholesterol efflux, we measured fecal cholesterol in mice following ezetimibe dosing, but found no differences between Idol(-/-) and control mice in this setting. CONCLUSIONS: In conclusion, our study establishes that the LXR-IDOL-LDLR axis is active in the intestine and is part of the molecular circuitry that maintains cholesterol homeostasis in enterocytes.


Subject(s)
Orphan Nuclear Receptors , Receptors, LDL , Animals , Intestines , Liver X Receptors , Mice , Orphan Nuclear Receptors/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Nat Commun ; 11(1): 1128, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111832

ABSTRACT

The sterol-regulatory element binding proteins (SREBP) are central transcriptional regulators of lipid metabolism. Using haploid genetic screens we identify the SREBP Regulating Gene (SPRING/C12ORF49) as a determinant of the SREBP pathway. SPRING is a glycosylated Golgi-resident membrane protein and its ablation in Hap1 cells, Hepa1-6 hepatoma cells, and primary murine hepatocytes reduces SREBP signaling. In mice, Spring deletion is embryonic lethal yet silencing of hepatic Spring expression also attenuates the SREBP response. Mechanistically, attenuated SREBP signaling in SPRINGKO cells results from reduced SREBP cleavage-activating protein (SCAP) and its mislocalization to the Golgi irrespective of the cellular sterol status. Consistent with limited functional SCAP in SPRINGKO cells, reintroducing SCAP restores SREBP-dependent signaling and function. Moreover, in line with the role of SREBP in tumor growth, a wide range of tumor cell lines display dependency on SPRING expression. In conclusion, we identify SPRING as a previously unrecognized modulator of SREBP signaling.


Subject(s)
Cholesterol/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Cell Line , Embryonic Development/genetics , Endoplasmic Reticulum/metabolism , Gene Expression , Golgi Apparatus/metabolism , Haploidy , Hepatocytes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Proteins/genetics
4.
Diabetes ; 68(12): 2223-2234, 2019 12.
Article in English | MEDLINE | ID: mdl-31578192

ABSTRACT

Obesity is taking on worldwide epidemic proportions, yet effective pharmacological agents with long-term efficacy remain unavailable. Previously, we designed the iminosugar N-adamantine-methyloxypentyl-deoxynojirimycin (AMP-DNM), which potently improves glucose homeostasis by lowering excessive glycosphingolipids. Here we show that AMP-DNM promotes satiety and activates brown adipose tissue (BAT) in obese rodents. Moreover, we demonstrate that the mechanism mediating these favorable actions depends on oral, but not central, administration of AMP-DNM, which ultimately stimulates systemic glucagon-like peptide 1 (GLP1) secretion. We evidence an essential role of brain GLP1 receptors (GLP1r), as AMP-DNM fails to promote satiety and activate BAT in mice lacking the brain GLP1r as well as in mice treated intracerebroventricularly with GLP1r antagonist exendin-9. In conclusion, AMP-DNM markedly ameliorates metabolic abnormalities in obese rodents by restoring satiety and activating BAT through central GLP1r, while improving glucose homeostasis by mechanisms independent of central GLP1r.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Adamantane/analogs & derivatives , Adipose Tissue, Brown/drug effects , Glucagon-Like Peptide 1/physiology , Satiation/drug effects , 1-Deoxynojirimycin/pharmacology , Adamantane/pharmacology , Animals , Brain/drug effects , Brain/physiology , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
5.
Atherosclerosis ; 281: 137-142, 2019 02.
Article in English | MEDLINE | ID: mdl-30658189

ABSTRACT

BACKGROUND AND AIMS: Cholesterol is an essential lipid for cellular function and membrane integrity, and hence its cellular levels and distribution must be tightly regulated. Biosynthesis of cholesterol is ramped when its cellular levels are low. Herein, the ER-resident and rate-limiting enzymes 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and squalene monooxygenase (SQLE) play a prominent role. We have recently reported that MARCH6, an E3 ubiquitin ligase, specifically promotes cholesterol-stimulated ubiquitylation and subsequent proteasomal degradation of SQLE, but not of HMGCR. To further delineate how post-translational regulation of SQLE and HMGCR is differentially achieved, we hypothesized that their sterol-dependent degradation machinery makes use of distinct E2 ubiquitin conjugating enzymes. METHODS: To study this possibility, we therefore used a CRISPR/Cas9-based approach to screen for ER-associated degradation (ERAD)-associated E2 enzymes that are essential for MARCH6-dependent degradation of SQLE. RESULTS: We report here the identification of UBE2J2 as the primary E2 ubiquitin conjugating enzyme essential for this process in mammalian cells, in contrast to UBE2G2, which is essential for sterol-stimulated degradation of HMGCR. We demonstrate that ablating UBE2J2 disturbs cholesterol-accelerated SQLE degradation in multiple human cell types, including cells of hepatic origin, and that the ability of UBE2J2 to support SQLE degradation critically depends on its enzymatic activity. CONCLUSIONS: Our findings establish UBE2J2 as an important partner of MARCH6 in cholesterol-stimulated degradation of SQLE, thereby contributing to the complex regulation of cellular cholesterol homeostasis.


Subject(s)
Cholesterol/biosynthesis , Hepatocytes/enzymology , Hydroxymethylglutaryl CoA Reductases/metabolism , Membrane Proteins/metabolism , Squalene Monooxygenase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Enzyme Stability , HEK293 Cells , Hep G2 Cells , Humans , Membrane Proteins/genetics , Proteolysis , Time Factors , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
6.
Arterioscler Thromb Vasc Biol ; 38(8): 1785-1795, 2018 08.
Article in English | MEDLINE | ID: mdl-29903737

ABSTRACT

Objective- The E3 ubiquitin ligase IDOL (inducible degrader of the LDLR [LDL (low-density lipoprotein) receptor]) is a post-transcriptional regulator of LDLR abundance. Model systems and human genetics support a role for IDOL in regulating circulating LDL levels. Whether IDOL plays a broader metabolic role and affects development of metabolic syndrome-associated comorbidities is unknown. Approach and Results- We studied WT (wild type) and Idol(-/-) (Idol-KO) mice in 2 models: physiological aging and diet-induced obesity. In both models, deletion of Idol protected mice from metabolic dysfunction. On a Western-type diet, Idol loss resulted in decreased circulating levels of cholesterol, triglycerides, glucose, and insulin. This was accompanied by protection from weight gain in short- and long-term dietary challenges, which could be attributed to reduced hepatosteatosis and fat mass in Idol-KO mice. Although feeding and intestinal fat uptake were unchanged in Idol-KO mice, their brown adipose tissue was protected from lipid accumulation and had elevated expression of UCP1 (uncoupling protein 1) and TH (tyrosine hydroxylase). Indirect calorimetry indicated a marked increase in locomotion and suggested a trend toward increased cumulative energy expenditure and fat oxidation. An increase in in vivo clearance of reconstituted lipoprotein particles in Idol-KO mice may sustain this energetic demand. In the BXD mouse genetic reference population, hepatic Idol expression correlates with multiple metabolic parameters, thus providing support for findings in the Idol-KO mice. Conclusions- Our study uncovers an unrecognized role for Idol in regulation of whole body metabolism in physiological aging and on a Western-type diet. These findings support Idol inhibition as a therapeutic strategy to target multiple metabolic syndrome-associated comorbidities.


Subject(s)
Diet, High-Fat , Energy Metabolism , Liver/enzymology , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Ubiquitin-Protein Ligases/deficiency , Adipogenesis , Adipose Tissue, Brown/enzymology , Adiposity , Age Factors , Aging , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cholesterol/blood , Disease Models, Animal , Female , Insulin/blood , Locomotion , Male , Metabolic Syndrome/blood , Metabolic Syndrome/enzymology , Metabolic Syndrome/genetics , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Obesity/blood , Obesity/enzymology , Obesity/genetics , Triglycerides/blood , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin-Protein Ligases/genetics , Uncoupling Protein 1/metabolism
7.
Autophagy ; 14(3): 437-449, 2018.
Article in English | MEDLINE | ID: mdl-29455584

ABSTRACT

In recent years, the lysosome has emerged as a highly dynamic, transcriptionally regulated organelle that is integral to nutrient-sensing and metabolic rewiring. This is coordinated by a lysosome-to-nucleus signaling nexus in which MTORC1 controls the subcellular distribution of the microphthalmia-transcription factor E (MiT/TFE) family of "master lysosomal regulators". Yet, despite the importance of the lysosome in cellular metabolism, the impact of traditional in vitro culture media on lysosomal dynamics and/or MiT/TFE localization has not been fully appreciated. Here, we identify HEPES, a chemical buffering agent that is broadly applied in cell culture, as a potent inducer of lysosome biogenesis. Supplementation of HEPES to cell growth media is sufficient to decouple the MiT/TFE family members-TFEB, TFE3 and MITF-from regulatory mechanisms that control their cytosolic retention. Increased MiT/TFE nuclear import in turn drives the expression of a global network of lysosomal-autophagic and innate host-immune response genes, altering lysosomal dynamics, proteolytic capacity, autophagic flux, and inflammatory signaling. In addition, siRNA-mediated MiT/TFE knockdown effectively blunted HEPES-induced lysosome biogenesis and gene expression profiles. Mechanistically, we show that MiT/TFE activation in response to HEPES requires its macropinocytic ingestion and aberrant lysosomal storage/pH, but is independent of MTORC1 signaling. Altogether, our data underscore the cautionary use of chemical buffering agents in cell culture media due to their potentially confounding effects on experimental results.


Subject(s)
Autophagy/physiology , Gene Regulatory Networks/genetics , HEPES/metabolism , Lysosomes/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line , Humans , Signal Transduction/genetics , Signal Transduction/physiology
8.
Arterioscler Thromb Vasc Biol ; 37(11): 2064-2074, 2017 11.
Article in English | MEDLINE | ID: mdl-28882874

ABSTRACT

OBJECTIVE: The cellular demand for cholesterol requires control of its biosynthesis by the mevalonate pathway. Regulation of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), a rate-limiting enzyme in this pathway and the target of statins, is a key control point herein. Accordingly, HMGCR is subject to negative and positive regulation. In particular, the ability of oxysterols and intermediates of the mevalonate pathway to stimulate its proteasomal degradation is an exquisite example of metabolically controlled feedback regulation. To define the genetic determinants that govern this process, we conducted an unbiased haploid mammalian genetic screen. APPROACH AND RESULTS: We generated human haploid cells with mNeon fused to endogenous HMGCR using CRISPR/Cas9 and used these cells to interrogate regulation of HMGCR abundance in live cells. This resulted in identification of known and new regulators of HMGCR, and among the latter, UBXD8 (ubiquitin regulatory X domain-containing protein 8), a gene that has not been previously implicated in this process. We demonstrate that UBXD8 is an essential determinant of metabolically stimulated degradation of HMGCR and of cholesterol biosynthesis in multiple cell types. Accordingly, UBXD8 ablation leads to aberrant cholesterol synthesis due to loss of feedback control. Mechanistically, we show that UBXD8 is necessary for sterol-stimulated dislocation of ubiquitylated HMGCR from the endoplasmic reticulum membrane en route to proteasomal degradation, a function dependent on its UBX domain. CONCLUSIONS: We establish UBXD8 as a previously unrecognized determinant that couples flux across the mevalonate pathway to control of cholesterol synthesis and demonstrate the feasibility of applying mammalian haploid genetics to study metabolic traits.


Subject(s)
Blood Proteins/metabolism , Cholesterol/biosynthesis , Haploidy , Hydroxymethylglutaryl CoA Reductases/metabolism , Membrane Proteins/metabolism , Animals , Blood Proteins/genetics , CRISPR-Cas Systems , Endoplasmic Reticulum/enzymology , Enzyme Stability , Feedback, Physiological , Gene Expression Regulation, Enzymologic , Hep G2 Cells , Hepatocytes/enzymology , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Membrane Proteins/genetics , Mevalonic Acid/metabolism , Microscopy, Confocal , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Proteolysis , Rats , Recombinant Fusion Proteins/metabolism , Transfection , Ubiquitination
9.
ACS Chem Biol ; 12(7): 1830-1841, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28485919

ABSTRACT

Glucocerebrosidase (GBA) is a lysosomal ß-glucosidase that degrades glucosylceramide. Its deficiency results in Gaucher disease (GD). We examined the effects of active site occupancy of GBA on its structural stability. For this, we made use of cyclophellitol-derived activity-based probes (ABPs) that bind irreversibly to the catalytic nucleophile (E340), and for comparison, we used the potent reversible inhibitor isofagomine. We demonstrate that cyclophellitol ABPs improve the stability of GBA in vitro, as revealed by thermodynamic measurements (Tm increase by 21 °C), and introduce resistance to tryptic digestion. The stabilizing effect of cell-permeable cyclophellitol ABPs is also observed in intact cultured cells containing wild-type GBA, N370S GBA (labile in lysosomes), and L444P GBA (exhibits impaired ER folding): all show marked increases in lysosomal forms of GBA molecules upon exposure to ABPs. The same stabilization effect is observed for endogenous GBA in the liver of wild-type mice injected with cyclophellitol ABPs. Stabilization effects similar to those observed with ABPs were also noted at high concentrations of the reversible inhibitor isofagomine. In conclusion, we provide evidence that the increase in cellular levels of GBA by ABPs and by the reversible inhibitor is in part caused by their ability to stabilize GBA folding, which increases the resistance of GBA against breakdown by lysosomal proteases. These effects are more pronounced in the case of the amphiphilic ABPs, presumably due to their high lipophilic potential, which may promote further structural compactness of GBA through hydrophobic interactions. Our study provides further rationale for the design of chaperones for GBA to ameliorate Gaucher disease.


Subject(s)
Catalytic Domain/physiology , Enzyme Stability/physiology , Glucosylceramidase/chemistry , Glucosylceramidase/metabolism , Imino Pyranoses/metabolism , Animals , Binding Sites , Enzyme Stability/drug effects , Imino Pyranoses/chemistry , Imino Pyranoses/pharmacology , Liver/drug effects , Liver/enzymology , Mice , Molecular Structure , Temperature
10.
PLoS One ; 12(2): e0170268, 2017.
Article in English | MEDLINE | ID: mdl-28207759

ABSTRACT

Deficiency of glucocerebrosidase (GBA) causes Gaucher disease (GD). In the common non-neuronopathic GD type I variant, glucosylceramide accumulates primarily in the lysosomes of visceral macrophages. Supplementing storage cells with lacking enzyme is accomplished via chronic intravenous administration of recombinant GBA containing mannose-terminated N-linked glycans, mediating the selective uptake by macrophages expressing mannose-binding lectin(s). Two recombinant GBA preparations with distinct N-linked glycans are registered in Europe for treatment of type I GD: imiglucerase (Genzyme), contains predominantly Man(3) glycans, and velaglucerase (Shire PLC) Man(9) glycans. Activity-based probes (ABPs) enable fluorescent labeling of recombinant GBA preparations through their covalent attachment to the catalytic nucleophile E340 of GBA. We comparatively studied binding and uptake of ABP-labeled imiglucerase and velaglucerase in isolated dendritic cells, cultured human macrophages and living mice, through simultaneous detection of different GBAs by paired measurements. Uptake of ABP-labeled rGBAs by dendritic cells was comparable, as well as the bio-distribution following equimolar intravenous administration to mice. ABP-labeled rGBAs were recovered largely in liver, white-blood cells, bone marrow and spleen. Lungs, brain and skin, affected tissues in severe GD types II and III, were only poorly supplemented. Small, but significant differences were noted in binding and uptake of rGBAs in cultured human macrophages, in the absence and presence of mannan. Mannan-competed binding and uptake were largest for velaglucerase, when determined with single enzymes or as equimolar mixtures of both enzymes. Vice versa, imiglucerase showed more prominent binding and uptake not competed by mannan. Uptake of recombinant GBAs by cultured macrophages seems to involve multiple receptors, including several mannose-binding lectins. Differences among cells from different donors (n = 12) were noted, but the same trends were always observed. Our study suggests that further insight in targeting and efficacy of enzyme therapy of individual Gaucher patients could be obtained by the use of recombinant GBA, trace-labeled with an ABP, preferably equipped with an infrared fluorophore or other reporter tag suitable for in vivo imaging.


Subject(s)
Dendritic Cells/enzymology , Fluorescent Dyes/chemistry , Glucosylceramidase/metabolism , Macrophages/enzymology , Monocytes/enzymology , Animals , Benzofurans/chemistry , Cells, Cultured , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism
11.
PLoS One ; 12(2): e0172721, 2017.
Article in English | MEDLINE | ID: mdl-28231341

ABSTRACT

Cellular cholesterol metabolism is subject to tight regulation to maintain adequate levels of this central lipid molecule. Herein, the sterol-responsive Liver X Receptors (LXRs) play an important role owing to their ability to reduce cellular cholesterol load. In this context, identifying the full set of LXR-regulated genes will contribute to our understanding of their role in cholesterol metabolism. Using global transcriptional analysis we report here the identification of RNF145 as an LXR-regulated target gene. We demonstrate that RNF145 is regulated by LXRs in both human and mouse primary cells and cell lines, and in vivo in mice. Regulation of RNF145 by LXR depends on a functional LXR-element in its proximal promotor. Consistent with LXR-dependent regulation of Rnf145 we show that regulation is lost in macrophages and fibroblasts from Lxrαß(-/-) mice, and also in vivo in livers of Lxrα(-/-) mice treated with the LXR synthetic ligand T0901317. RNF145 is closely related to RNF139/TRC8, an E3 ligase implicated in control of SREBP processing. However, silencing of RNF145 in HepG2 or HeLa cells does not impair SREBP1/2 processing and sterol-responsive gene expression in these cells. Similar to TRC8, we demonstrate that RNF145 is localized to the ER and that it possesses intrinsic E3 ubiquitin ligase activity. In summary, we report the identification of RNF145 as an ER-resident E3 ubiquitin ligase that is transcriptionally controlled by LXR.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Liver X Receptors/genetics , Membrane Proteins/genetics , Transcription, Genetic , Animals , Cell Line , Cholesterol/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver/drug effects , Liver/metabolism , Liver X Receptors/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
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
13.
Chembiochem ; 18(4): 402-412, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28000364

ABSTRACT

Galactosylceramidase (GALC) is the lysosomal ß-galactosidase responsible for the hydrolysis of galactosylceramide. Inherited deficiency in GALC causes Krabbe disease, a devastating neurological disorder characterized by accumulation of galactosylceramide and its deacylated counterpart, the toxic sphingoid base galactosylsphingosine (psychosine). We report the design and application of a fluorescently tagged activity-based probe (ABP) for the sensitive and specific labeling of active GALC molecules from various species. The probe consists of a ß-galactopyranose-configured cyclophellitol-epoxide core, conferring specificity for GALC, equipped with a BODIPY fluorophore at C6 that allows visualization of active enzyme in cells and tissues. Detection of residual GALC in patient fibroblasts holds great promise for laboratory diagnosis of Krabbe disease. We further describe a procedure for in situ imaging of active GALC in murine brain by intra-cerebroventricular infusion of the ABP. In conclusion, this GALC-specific ABP should find broad applications in diagnosis, drug development, and evaluation of therapy for Krabbe disease.


Subject(s)
Galactosylceramidase/genetics , Galactosylceramidase/metabolism , Leukodystrophy, Globoid Cell/enzymology , Molecular Probes , Deficiency Diseases/enzymology , Deficiency Diseases/genetics , Galactosylceramidase/antagonists & inhibitors , Leukodystrophy, Globoid Cell/diagnosis , Leukodystrophy, Globoid Cell/genetics , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Molecular Structure , Mutation
14.
Chembiochem ; 17(18): 1698-704, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27383447

ABSTRACT

ß-Glucoside-configured cyclophellitols are activity-based probes (ABPs) that allow sensitive detection of ß-glucosidases. Their applicability to detect proteins fused with ß-glucosidase was investigated in the cellular context. The tag was Rhodococcus sp. M-777 endoglycoceramidase II (EGCaseII), based on its lack of glycans and ability to hydrolyze fluorogenic 4-methylumbelliferyl ß-d-lactoside (an activity absent in mammalian cells). Specific dual detection of fusion proteins was possible in vitro and in situ by using fluorescent ABPs and a fluorogenic substrate. Pre-blocking with conduritol ß-epoxide (a poor inhibitor of EGCaseII) eliminated ABP labeling of endogenous ß-glucosidases. ABPs equipped with biotin allowed convenient purification of the fusion proteins. Diversification of ABPs (distinct fluorophores, fluorogenic high-resolution detection moieties) should assist further research in living cells and organisms.


Subject(s)
Fluorescent Dyes/metabolism , Mannosidases/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Fluorescent Dyes/chemistry , Molecular Structure , Rhodococcus/enzymology
15.
Diabetes ; 65(9): 2591-605, 2016 09.
Article in English | MEDLINE | ID: mdl-27325287

ABSTRACT

Aberrant mitochondrial fission plays a pivotal role in the pathogenesis of skeletal muscle insulin resistance. However, fusion-fission dynamics are physiologically regulated by inherent tissue-specific and nutrient-sensitive processes that may have distinct or even opposing effects with respect to insulin sensitivity. Based on a combination of mouse population genetics and functional in vitro assays, we describe here a regulatory circuit in which peroxisome proliferator-activated receptor γ (PPARγ), the adipocyte master regulator and receptor for the thiazolidinedione class of antidiabetic drugs, controls mitochondrial network fragmentation through transcriptional induction of Bnip3. Short hairpin RNA-mediated knockdown of Bnip3 in cultured adipocytes shifts the balance toward mitochondrial elongation, leading to compromised respiratory capacity, heightened fatty acid ß-oxidation-associated mitochondrial reactive oxygen species generation, insulin resistance, and reduced triacylglycerol storage. Notably, the selective fission/Drp1 inhibitor Mdivi-1 mimics the effects of Bnip3 knockdown on adipose mitochondrial bioenergetics and glucose disposal. We further show that Bnip3 is reciprocally regulated in white and brown fat depots of diet-induced obesity and leptin-deficient ob/ob mouse models. Finally, Bnip3(-/-) mice trade reduced adiposity for increased liver steatosis and develop aggravated systemic insulin resistance in response to high-fat feeding. Together, our data outline Bnip3 as a key effector of PPARγ-mediated adipose mitochondrial network fragmentation, improving insulin sensitivity and limiting oxidative stress.


Subject(s)
Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Female , Glucose/metabolism , Immunoblotting , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/genetics , Obesity/genetics , Obesity/metabolism , PPAR gamma/genetics , Radioimmunoprecipitation Assay , Reverse Transcriptase Polymerase Chain Reaction
16.
FEBS Lett ; 590(6): 716-25, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26898341

ABSTRACT

Glycosphingoid bases are elevated in inherited lysosomal storage disorders with deficient activity of glycosphingolipid catabolizing glycosidases. We investigated the molecular basis of the formation of glucosylsphingosine and globotriaosylsphingosine during deficiency of glucocerebrosidase (Gaucher disease) and α-galactosidase A (Fabry disease). Independent genetic and pharmacological evidence is presented pointing to an active role of acid ceramidase in both processes through deacylation of lysosomal glycosphingolipids. The potential pathophysiological relevance of elevated glycosphingoid bases generated through this alternative metabolism in patients suffering from lysosomal glycosidase defects is discussed.


Subject(s)
Acid Ceramidase/metabolism , Fabry Disease/metabolism , Gaucher Disease/metabolism , Glycosphingolipids/metabolism , Acid Ceramidase/genetics , Acylation , Animals , Disease Models, Animal , Fabry Disease/genetics , Female , Gaucher Disease/genetics , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Glycosphingolipids/chemistry , HEK293 Cells , Humans , Lysosomes/metabolism , Male , Mice , Mice, Knockout , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
17.
J Lipid Res ; 57(3): 451-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26724485

ABSTRACT

The membrane lipid glucosylceramide (GlcCer) is continuously formed and degraded. Cells express two GlcCer-degrading ß-glucosidases, glucocerebrosidase (GBA) and GBA2, located in and outside the lysosome, respectively. Here we demonstrate that through transglucosylation both GBA and GBA2 are able to catalyze in vitro the transfer of glucosyl-moieties from GlcCer to cholesterol, and vice versa. Furthermore, the natural occurrence of 1-O-cholesteryl-ß-D-glucopyranoside (GlcChol) in mouse tissues and human plasma is demonstrated using LC-MS/MS and (13)C6-labeled GlcChol as internal standard. In cells, the inhibition of GBA increases GlcChol, whereas inhibition of GBA2 decreases glucosylated sterol. Similarly, in GBA2-deficient mice, GlcChol is reduced. Depletion of GlcCer by inhibition of GlcCer synthase decreases GlcChol in cells and likewise in plasma of inhibitor-treated Gaucher disease patients. In tissues of mice with Niemann-Pick type C disease, a condition characterized by intralysosomal accumulation of cholesterol, marked elevations in GlcChol occur as well. When lysosomal accumulation of cholesterol is induced in cultured cells, GlcChol is formed via lysosomal GBA. This illustrates that reversible transglucosylation reactions are highly dependent on local availability of suitable acceptors. In conclusion, mammalian tissues contain GlcChol formed by transglucosylation through ß-glucosidases using GlcCer as donor. Our findings reveal a novel metabolic function for GlcCer.


Subject(s)
Cholesterol/metabolism , beta-Glucosidase/metabolism , Animals , COS Cells , Chlorocebus aethiops , Female , Gaucher Disease/metabolism , Glycosylation , Humans , Male , Mice , Niemann-Pick Diseases/metabolism , RAW 264.7 Cells
18.
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
19.
Circ Res ; 118(2): 222-9, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26582775

ABSTRACT

RATIONALE: The (pro)renin receptor ([P]RR) interacts with (pro)renin at concentrations that are >1000× higher than observed under (patho)physiological conditions. Recent studies have identified renin-angiotensin system-independent functions for (P)RR related to its association with the vacuolar H(+)-ATPase. OBJECTIVE: To uncover renin-angiotensin system-independent functions of the (P)RR. METHODS AND RESULTS: We used a proteomics-based approach to purify and identify (P)RR-interacting proteins. This resulted in identification of sortilin-1 (SORT1) as a high-confidence (P)RR-interacting protein, a finding which was confirmed by coimmunoprecipitation of endogenous (P)RR and SORT1. Functionally, silencing (P)RR expression in hepatocytes decreased SORT1 and low-density lipoprotein (LDL) receptor protein abundance and, as a consequence, resulted in severely attenuated cellular LDL uptake. In contrast to LDL, endocytosis of epidermal growth factor or transferrin remained unaffected by silencing of the (P)RR. Importantly, reduction of LDL receptor and SORT1 protein abundance occurred in the absence of changes in their corresponding transcript level. Consistent with a post-transcriptional event, degradation of the LDL receptor induced by (P)RR silencing could be reversed by lysosomotropic agents, such as bafilomycin A1. CONCLUSIONS: Our study identifies a renin-angiotensin system-independent function for the (P)RR in the regulation of LDL metabolism by controlling the levels of SORT1 and LDL receptor.


Subject(s)
Endocytosis , Hepatocytes/metabolism , Lipoproteins, LDL/metabolism , Proteomics , Receptors, Cell Surface/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , CHO Cells , Chromatin Immunoprecipitation , Cricetulus , HEK293 Cells , Hep G2 Cells , Humans , Protein Processing, Post-Translational , Proteolysis , Proteomics/methods , RNA Interference , Receptors, Cell Surface/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Transfection , Vacuolar Proton-Translocating ATPases/genetics
20.
Mol Cell Biol ; 35(1): 52-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332231

ABSTRACT

The LIM-only protein FHL2 is expressed in smooth muscle cells (SMCs) and inhibits SMC-rich-lesion formation. To further elucidate the role of FHL2 in SMCs, we compared the transcriptomes of SMCs derived from wild-type (WT) and FHL2 knockout (KO) mice. This revealed that in addition to the previously recognized involvement of FHL2 in SMC proliferation, the cholesterol synthesis and liver X receptor (LXR) pathways are altered in the absence of FHL2. Using coimmunoprecipitation experiments, we found that FHL2 interacts with the two LXR isoforms, LXRα and LXRß. Furthermore, FHL2 strongly enhances transcriptional activity of LXR element (LXRE)-containing reporter constructs. Chromatin immunoprecipitation (ChIP) experiments on the ABCG1 promoter revealed that FHL2 enhances the association of LXRß with DNA. In line with these observations, we observed reduced basal transcriptional LXR activity in FHL2-KO SMCs compared to WT SMCs. This was also reflected in reduced expression of LXR target genes in intact aorta and aortic SMCs of FHL2-KO mice. Functionally, the absence of FHL2 resulted in attenuated cholesterol efflux to both ApoA-1 and high-density lipoprotein (HDL), in agreement with reduced LXR signaling. Collectively, our findings demonstrate that FHL2 is a transcriptional coactivator of LXRs and points toward FHL2 being an important determinant of cholesterol metabolism in SMCs.


Subject(s)
LIM-Homeodomain Proteins/metabolism , Lipid Metabolism , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Orphan Nuclear Receptors/metabolism , Transcription Factors/metabolism , Animals , Aorta/metabolism , Cell Proliferation , Cholesterol/metabolism , DNA/metabolism , HeLa Cells , Homeostasis/physiology , Humans , Lipoproteins, HDL/metabolism , Liver X Receptors , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...