Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Chem Biol ; 16(2): 206-213, 2020 02.
Article in English | MEDLINE | ID: mdl-31932720

ABSTRACT

Genetic screens in cultured human cells represent a powerful unbiased strategy to identify cellular pathways that determine drug efficacy, providing critical information for clinical development. We used insertional mutagenesis-based screens in haploid cells to identify genes required for the sensitivity to lasonolide A (LasA), a macrolide derived from a marine sponge that kills certain types of cancer cells at low nanomolar concentrations. Our screens converged on a single gene, LDAH, encoding a member of the metabolite serine hydrolase family that is localized on the surface of lipid droplets. Mechanistic studies revealed that LasA accumulates in lipid droplets, where it is cleaved into a toxic metabolite by LDAH. We suggest that selective partitioning of hydrophobic drugs into the oil phase of lipid droplets can influence their activation and eventual toxicity to cells.


Subject(s)
Drug Evaluation, Preclinical/methods , Lipid Droplets/metabolism , Macrolides/pharmacokinetics , Macrolides/toxicity , Proteins/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Haploidy , Humans , Inactivation, Metabolic , Lipid Droplets/drug effects , Macrolides/metabolism , Proteins/genetics
2.
Int J Mol Sci ; 20(15)2019 Aug 02.
Article in English | MEDLINE | ID: mdl-31382484

ABSTRACT

Arterial foam cells are central players of atherogenesis. Cholesterol acceptors, apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL), take up cholesterol and phospholipids effluxed from foam cells into the circulation. Due to the high abundance of cholesterol in foam cells, most previous studies focused on apoA-I/HDL-mediated free cholesterol (FC) transport. However, recent lipidomics of human atherosclerotic plaques also identified that oxidized sterols (oxysterols) and non-sterol lipid species accumulate as atherogenesis progresses. While it is known that these lipids regulate expression of pro-inflammatory genes linked to plaque instability, how cholesterol acceptors impact the foam cell lipidome, particularly oxysterols and non-sterol lipids, remains unexplored. Using lipidomics analyses, we found cholesterol acceptors remodel foam cell lipidomes. Lipid subclass analyses revealed various oxysterols, sphingomyelins, and ceramides, species uniquely enriched in human plaques were significantly reduced by cholesterol acceptors, especially by apoA-I. These results indicate that the function of lipid-poor apoA-I is not limited to the efflux of cholesterol and phospholipids but suggest that apoA-I serves as a major regulator of the foam cell lipidome and might play an important role in reducing multiple lipid species involved in the pathogenesis of atherosclerosis.


Subject(s)
Cholesterol/metabolism , Foam Cells/metabolism , Plaque, Atherosclerotic/metabolism , Animals , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Cells, Cultured , Humans , Lipidomics , Lipoproteins, LDL/metabolism , Male , Mice, Inbred C57BL , Oxysterols/metabolism
3.
Arterioscler Thromb Vasc Biol ; 34(2): 386-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24357060

ABSTRACT

OBJECTIVE: Lipid-laden macrophages or foam cells are characterized by massive cytosolic lipid droplet (LD) deposition containing mostly cholesterol ester (CE) derived from the lipoproteins cleared from the arterial wall. Cholesterol efflux from foam cells is considered to be atheroprotective. Because cholesterol is effluxed as free cholesterol, CE accumulation in LDs may limit free cholesterol efflux. Our objective was to identify proteins that regulate cholesterol trafficking through LDs. APPROACH AND RESULTS: In a proteomic analysis of the LD fraction of RAW 264.7 macrophages, we identified an evolutionarily conserved protein with a canonical GXSXG lipase catalytic motif and a predicted α/ß-hydrolase fold, the RIKEN cDNA 1110057K04 gene, which we named LD-associated hydrolase (LDAH). LDAH association with LDs was confirmed by immunoblotting and immunocytochemistry. LDAH was labeled with a probe specific for active serine hydrolases. LDAH showed relatively weak in vitro CE hydrolase activity. However, cholesterol measurements in intact cells supported a significant role of LDAH in CE homeostasis because LDAH upregulation and downregulation decreased and increased, respectively, intracellular cholesterol and CE in human embryonic kidney-293 cells and RAW 264.7 macrophages. Mutation of the putative nucleophilic serine impaired active hydrolase probe binding, in vitro CE hydrolase activity, and cholesterol-lowering effect in cells, whereas this mutant still localized to the LD. LDAH upregulation increased CE hydrolysis and cholesterol efflux from macrophages, and, interestingly, LDAH is highly expressed in macrophage-rich areas within mouse and human atherosclerotic lesions. CONCLUSIONS: The data identify a candidate target to promote reverse cholesterol transport from atherosclerotic lesions.


Subject(s)
Cholesterol/metabolism , Foam Cells/enzymology , Serine Proteases/metabolism , Amino Acid Sequence , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Biological Transport , Cholesterol Esters/metabolism , Disease Models, Animal , Foam Cells/pathology , Gene Expression Regulation, Enzymologic , HEK293 Cells , HeLa Cells , Humans , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Plaque, Atherosclerotic , Proteomics/methods , RNA Interference , Serine Proteases/chemistry , Serine Proteases/genetics , Substrate Specificity , Time Factors , Transfection
4.
Nat Commun ; 15(1): 6540, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095402

ABSTRACT

Foam cells in atheroma are engorged with lipid droplets (LDs) that contain esters of regulatory lipids whose metabolism remains poorly understood. LD-associated hydrolase (LDAH) has a lipase structure and high affinity for LDs of foam cells. Using knockout and transgenic mice of both sexes, here we show that LDAH inhibits atherosclerosis development and promotes stable lesion architectures. Broad and targeted lipidomic analyzes of primary macrophages and comparative lipid profiling of atheroma identified a broad impact of LDAH on esterified sterols, including natural liver X receptor (LXR) sterol ligands. Transcriptomic analyzes coupled with rescue experiments show that LDAH modulates the expression of prototypical LXR targets and leads macrophages to a less inflammatory phenotype with a profibrotic gene signature. These studies underscore the role of LDs as reservoirs and metabolic hubs of bioactive lipids, and suggest that LDAH favorably modulates macrophage activation and protects against atherosclerosis via lipolytic mobilization of regulatory sterols.


Subject(s)
Atherosclerosis , Lipid Droplets , Liver X Receptors , Macrophages , Mice, Knockout , Animals , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/pathology , Liver X Receptors/metabolism , Liver X Receptors/genetics , Mice , Male , Ligands , Female , Lipid Droplets/metabolism , Macrophages/metabolism , Sterols/metabolism , Foam Cells/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Humans , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Macrophage Activation , Sterol Esterase
5.
Nucleic Acids Res ; 39(7): 2769-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21109529

ABSTRACT

Muscleblind-like 1 (MBNL1) is a splicing regulator that controls developmentally regulated alternative splicing of a large number of exons including exon 11 of the Insulin Receptor (IR) gene and exon 5 of the cardiac Troponin T (cTNT) gene. There are three paralogs of MBNL in humans, all of which promote IR exon 11 inclusion and cTNT exon 5 skipping. Here, we identify a cluster of three binding sequences located downstream of IR exon 11 that constitute the MBNL1 response element and a weaker response element in the upstream intron. In addition, we used sequential deletions to define the functional domains of MBNL1 and MBNL3. We demonstrate that the regions required for splicing regulation are separate from the two pairs of zinc-finger RNA-binding domains. MBNL1 and MBNL3 contain core regulatory regions for both activation and repression located within an 80-amino-acid segment located downstream of the N-terminal zinc-finger pair. Deletions of these regions abolished regulation without preventing RNA binding. These domains have common features with the CUG-BP and ETR3-like Factor (CELF) family of splicing regulators. These results have identified protein domains required for splicing repression and activation and provide insight into the mechanism of splicing regulation by MBNL proteins.


Subject(s)
Alternative Splicing , RNA-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Exons , Humans , Molecular Sequence Data , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Regulatory Sequences, Ribonucleic Acid , Sequence Deletion
6.
Sci Rep ; 13(1): 19588, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37949969

ABSTRACT

Arterial macrophage foam cells are filled with cholesterol ester (CE) stored in cytosolic lipid droplets (LDs). Foam cells are central players in progression of atherosclerosis as regulators of lipid metabolism and inflammation, two major driving forces of atherosclerosis development. Thus, foam cells are considered plausible targets for intervention in atherosclerosis. However, a compound that directly regulates the lipid metabolism of LDs in the arterial foam cells has not yet been identified. In this study, we screened compounds that inhibit macrophage foam cell formation using a library of 2697 FDA-approved drugs. From the foam cells generated via loading of human oxidized low-density lipoprotein (oxLDL), we found 21 and 6 compounds that reduced and enhanced accumulations of lipids respectively. Among them, verteporfin most significantly reduced oxLDL-induced foam cell formation whereas it did not display a significant impact on foam cell formation induced by fatty acid. Mechanistically our data demonstrate that verteporfin acts via inhibition of oxLDL association with macrophages, reducing accumulation of CE. Interestingly, while other drugs that reduced foam cell formation did not have impact on pre-existing foam cells, verteporfin treatment significantly reduced their total lipids, CE, and pro-inflammatory gene expression. Together, our study identifies verteporfin as a novel regulator of foam cell lipid metabolism and inflammation and a potential compound for intervention in atherosclerosis.


Subject(s)
Atherosclerosis , Foam Cells , Humans , Foam Cells/metabolism , Verteporfin/pharmacology , Verteporfin/metabolism , Lipid Metabolism , Macrophages/metabolism , Atherosclerosis/metabolism , Lipoproteins, LDL/metabolism , Cholesterol Esters/metabolism , Inflammation/metabolism
7.
Nucleic Acids Res ; 37(13): 4275-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19443441

ABSTRACT

CUGBP2 (ETR-3/NAPOR/BRUNOL3) promotes inclusion of cardiac troponin T (cTNT) exon 5 via binding between positions 21 and 74 of the downstream intron. The molecular mechanism by which CUGBP2 activates cTNT exon 5 inclusion is unknown. Our results suggest that CUGBP2 promotes exon inclusion by a novel mechanism in which CUGBP2 directly interacts with components of the activated U2 snRNP and enhances binding of U2 snRNP to the branch site located upstream of the exon. Using an in vitro splicing assay, we show that recombinant CUGBP2 enhances complex A formation of a cTNT pre-mRNA. Enhanced complex A assembly requires both the upstream and downstream introns consistent with dual requirements for the downstream CUGBP2-binding site and an upstream branch site for U2 snRNP binding. We also show that CUGBP2 enhances binding of U2 snRNA to the cTNT pre-mRNA consistent with enhanced complex A assembly. Purification of CUGBP2-interacting proteins using tandem affinity purification leads to the demonstration that the core 17S U2 snRNP components, SF3b145 and SF3b49 bind directly to CUGBP2. We conclude that CUGBP2 activates exon inclusion by forming direct interactions with components of the 17S snRNP complex and recruits and/or stabilizes binding of U2 snRNP.


Subject(s)
RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Troponin T/genetics , Animals , Chickens , HeLa Cells , Humans , Introns , RNA Splicing Factors , Troponin T/metabolism
8.
J Am Heart Assoc ; 10(2): e018151, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33401929

ABSTRACT

Background Despite compelling epidemiological evidence that circadian disruption inherent to long-term shift work enhances atherosclerosis progression and vascular events, the underlying mechanisms remain poorly understood. A challenge to the use of mouse models for mechanistic and interventional studies involving light-dark patterns is that the spectral and absolute sensitivities of the murine and human circadian systems are very different, and light stimuli in nocturnal mice should be scaled to represent the sensitivities of the human circadian system. Methods and Results We used calibrated devices to deliver to low-density lipoprotein receptor knockout mice light-dark patterns representative of that experienced by humans working day shifts or rotating shift schedules. Mice under day shifts were maintained under regular 12 hours of light and 12 hours of dark cycles. Mice under rotating shift schedules were subjected for 11 weeks to reversed light-dark patterns 4 days in a row per week, followed by 3 days of regular light-dark patterns. In both protocols the light phases consisted of monochromatic green light at an irradiance of 4 µW/cm2. We found that the shift work paradigm disrupts the foam cell's molecular clock and increases endoplasmic reticulum stress and apoptosis. Lesions of mice under rotating shift schedules were larger and contained less prostabilizing fibrillar collagen and significantly increased areas of necrosis. Conclusions Low-density lipoprotein receptor knockout mice under light-dark patterns analogous to that experienced by rotating shift workers develop larger and more vulnerable plaques and may represent a valuable model for further mechanistic and/or interventional studies against the deleterious vascular effects of rotating shift work.


Subject(s)
Apoptosis/physiology , Atherosclerosis , Circadian Clocks/physiology , Endoplasmic Reticulum Stress/physiology , Foam Cells , Plaque, Atherosclerotic , Shift Work Schedule , Animals , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Circadian Rhythm/physiology , Foam Cells/metabolism , Foam Cells/pathology , Humans , Lipoproteins, LDL/genetics , Mice , Mice, Knockout , Models, Animal , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
9.
Clin Transl Med ; 7(1): 4, 2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29374337

ABSTRACT

Dysregulation of lipid metabolism is responsible for pathologies of human diseases including metabolic diseases. Recent advances in lipidomics analysis allow for the targeted and untargeted identification of lipid species and for their quantification in normal and diseased conditions. Herein, this review provides a brief introduction to lipidomics, highlights its application to characterize the lipidome at the cellular and physiological levels under different biological conditions, and discusses the potential for the use of lipidomics in the discovery of biomarkers.

10.
Mol Cell Biol ; 22(24): 8409-14, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12446761

ABSTRACT

ASC-2, a recently isolated transcriptional coactivator molecule, stimulates transactivation by multiple transcription factors, including nuclear receptors. We generated a potent dominant negative fragment of ASC-2, encompassing the N-terminal LXXLL motif that binds a broad range of nuclear receptors. This fragment, termed DN1, specifically inhibited endogenous ASC-2 from binding these receptors in vivo, whereas DN1/m, in which the LXXLL motif was mutated to LXXAA to abolish the receptor interactions, was inert. Interestingly, DN1 transgenic mice but not DN1/m transgenic mice exhibited severe microphthalmia and posterior lenticonus with cataract as well as a variety of pathophysiological phenotypes in many other organs. Our results provide a novel insight into the molecular and histopathological mechanism of posterior lenticonus with cataract and attest to the importance of ASC-2 as a pivotal transcriptional coactivator of nuclear receptors in vivo.


Subject(s)
Congenital Abnormalities , Intracellular Signaling Peptides and Proteins , Lens Diseases/metabolism , Peptide Fragments/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/pathology , Embryo, Mammalian/physiology , Eye/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, Lethal , Genes, Reporter , Humans , Lens Diseases/genetics , Mice , Mice, Transgenic , Nuclear Receptor Coactivators , Peptide Fragments/genetics , Phenotype , Pregnancy , Receptors, Retinoic Acid/metabolism , Signal Transduction/physiology , Transcription Factors/genetics
11.
Mol Cell Biol ; 23(1): 140-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12482968

ABSTRACT

Many transcription coactivators interact with nuclear receptors in a ligand- and C-terminal transactivation function (AF2)-dependent manner. These include activating signal cointegrator 2 (ASC-2), a recently isolated transcriptional coactivator molecule, which is amplified in human cancers and stimulates transactivation by nuclear receptors and numerous other transcription factors. In this report, we show that ASC-2 belongs to a steady-state complex of approximately 2 MDa (ASC-2 complex [ASCOM]) in HeLa nuclei. ASCOM contains retinoblastoma-binding protein RBQ-3, alpha/beta-tubulins, and trithorax group proteins ALR-1, ALR-2, HALR, and ASH2. In particular, ALR-1/2 and HALR contain a highly conserved 130- to 140-amino-acid motif termed the SET domain, which was recently implicated in histone H3 lysine-specific methylation activities. Indeed, recombinant ALR-1, HALR, and immunopurified ASCOM exhibit very weak but specific H3-lysine 4 methylation activities in vitro, and transactivation by retinoic acid receptor appears to involve ligand-dependent recruitment of ASCOM and subsequent transient H3-lysine 4 methylation of the promoter region in vivo. Thus, ASCOM may represent a distinct coactivator complex of nuclear receptors. Further characterization of ASCOM will lead to a better understanding of how nuclear receptors and other transcription factors mediate transcriptional activation.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cation Transport Proteins , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HeLa Cells , Histones/metabolism , Humans , Lysine/metabolism , Macromolecular Substances , Methylation , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Coactivators , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Sequence Homology, Amino Acid , Transcription Factors/genetics , Tubulin/metabolism
12.
Sci Rep ; 7(1): 2743, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28578400

ABSTRACT

Lipid droplet (LD)-associated hydrolase (LDAH) is a newly identified LD protein abundantly expressed in tissues that predominantly store triacylglycerol (TAG). However, how LDAH regulates TAG metabolism remains unknown. We found that upon oleic acid loading LDAH translocalizes from the ER to newly formed LDs, and induces LD coalescence in a tubulin-dependent manner. LDAH overexpression and downregulation in HEK293 cells increase and decrease, respectively, TAG levels. Pulse and chase experiments show that LDAH enhances TAG biogenesis, but also decreases TAG turnover and fatty acid release from cells. Mutations in predicted catalytic and acyltransferase motifs do not influence TAG levels, suggesting that the effect is independent of LDAH's enzymatic activity. However, a LDAH alternative-splicing variant missing 90 amino acids at C-terminus does not promote LD fusion or TAG accumulation, while it still localizes to LDs. Interestingly, LDAH enhances polyubiquitination and proteasomal degradation of adipose triglyceride lipase (ATGL), a rate limiting enzyme of TAG hydrolysis. Co-expression of ATGL reverses the changes in LD phenotype induced by LDAH, and both proteins counterbalance their effects on TAG stores. Together, these studies support that under conditions of TAG storage in LDs LDAH plays a primarily lipogenic role, inducing LD growth and enhancing degradation of ATGL.


Subject(s)
Lipase/metabolism , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Triglycerides/metabolism , Alternative Splicing/genetics , Fatty Acids/metabolism , HEK293 Cells , HeLa Cells , Humans , Hydrolysis , Lipase/genetics , Mutation , Oleic Acid/metabolism , Triglycerides/genetics
13.
Mol Med Rep ; 13(6): 4527-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27082419

ABSTRACT

Accumulation of atherosclerotic plaques in arterial walls leads to major cardiovascular diseases and stroke. Macrophages/foam cells are central components of atherosclerotic plaques, which populate the arterial wall in order to remove harmful modified low­density lipoprotein (LDL) particles, resulting in the accumulation of lipids, mostly LDL­derived cholesterol ester, in cytosolic lipid droplets (LDs). At present, LDs are recognized as dynamic organelles that govern cellular metabolic processes. LDs consist of an inner core of neutral lipids surrounded by a monolayer of phospholipids and free cholesterol, and contain LD­associated proteins (LDAPs) that regulate LD functions. Foam cells are characterized by an aberrant accumulation of cytosolic LDs, and are considered a hallmark of atherosclerotic lesions through all stages of development. Previous studies have investigated the mechanisms underlying foam cell formation, aiming to discover therapeutic strategies that target foam cells and intervene against atherosclerosis. It is well established that LDAPs have a major role in the pathogenesis of metabolic diseases caused by dysfunction of lipid metabolism, and several studies have linked LDAPs to the development of atherosclerosis. In this review, several foam cell­targeting pathways have been described, with an emphasis on the role of LDAPs in cholesterol mobilization from macrophages. In addition, the potential of LDAPs as therapeutic targets to prevent the progression and/or facilitate the regression of the disease has been discussed.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Lipid Droplet Associated Proteins/metabolism , Animals , Atherosclerosis/pathology , Biological Transport , Cholesterol/metabolism , Cholesterol Esters/metabolism , Disease Models, Animal , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hydrolysis , Lipid Droplet Associated Proteins/genetics , Lipid Droplets/metabolism , Lipid Metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
14.
Genom Data ; 9: 37-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27408807

ABSTRACT

Hypercholesterolemia is a main risk factor for atherosclerosis development. Arterial macrophages, or foam cells, take-up and process lipoprotein particles deposited in arteries, and store much of the cholesterol carried by these particles in their cytoplasm. However, the effects of exposure to different cholesterol levels on foam cells remain poorly understood. Given the remarkable plasticity of macrophages in response to environmental variables, studies on macrophage biology should ideally be performed in the environment where they exert their physiological functions, namely atherosclerotic lesions in the case of foam cells. We used a mouse model of atherosclerosis, the apolipoprotein E-deficient mouse, to study in vivo the transcriptional response of foam cells to short- and long-term elevations in plasma cholesterol, induced by feeding mice a western type diet. The microarray data sets from this study have been deposited in NCBI's Gene Expression Omnibus under the accession number GSE70619. Here we provide detailed information on the experimental set-up, on the isolation of RNA by laser capture microdissection, and on the methodology used for RNA amplification and analysis by microarray and quantitative real-time PCR.

15.
J Am Heart Assoc ; 5(4): e002663, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27091181

ABSTRACT

BACKGROUND: Foam cells are central to two major pathogenic processes in atherogenesis: cholesterol buildup in arteries and inflammation. The main underlying cause of cholesterol deposition in arteries is hypercholesterolemia. This study aimed to assess, in vivo, whether elevated plasma cholesterol also alters the inflammatory balance of foam cells. METHODS AND RESULTS: Apolipoprotein E-deficient mice were fed regular mouse chow through the study or were switched to a Western-type diet (WD) 2 or 14 weeks before death. Consecutive sections of the aortic sinus were used for lesion quantification or to isolate RNA from foam cells by laser-capture microdissection (LCM) for microarray and quantitative polymerase chain reaction analyses. WD feeding for 2 or 14 weeks significantly increased plasma cholesterol, but the size of atherosclerotic lesions increased only in the 14-week WD group. Expression of more genes was affected in foam cells of mice under prolonged hypercholesterolemia than in mice fed WD for 2 weeks. However, most transcripts coding for inflammatory mediators remained unchanged in both WD groups. Among the main players in inflammatory or immune responses, chemokine (C-X-C motif) ligand 13 was induced in foam cells of mice under WD for 2 weeks. The interferon-inducible GTPases, guanylate-binding proteins (GBP)3 and GBP6, were induced in the 14-week WD group, and other GBP family members were moderately increased. CONCLUSIONS: Our results indicate that acceleration of atherosclerosis by hypercholesterolemia is not linked to global changes in the inflammatory balance of foam cells. However, induction of GBPs uncovers a novel family of immune modulators with a potential role in atherogenesis.


Subject(s)
Atherosclerosis/etiology , Diet, Western/adverse effects , Foam Cells/chemistry , GTP-Binding Proteins/analysis , Inflammation/physiopathology , Animals , Atherosclerosis/chemically induced , Female , Foam Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Laser Capture Microdissection , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Sinus of Valsalva/chemistry
16.
Cardiovasc Res ; 109(2): 294-304, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26487692

ABSTRACT

AIMS: Atherosclerosis development can be ameliorated by promoting reverse cholesterol transport (RCT) from arteries. The process involves cholesterol efflux from foam cells to extracellular acceptors such as apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL) that mediate transport to the liver. Perilipin-2 (PLIN2) is a lipid droplet (LD)-associated protein that in macrophages facilitates cholesterol storage and prevents efflux. We hypothesized that atheroprotection would be enhanced by concurrently targeting PLIN2 to increase the efflux capacity of foam cells and increasing plasma apoA-I and HDL. METHODS AND RESULTS: PLIN2-knockout and wild-type mice lacking apolipoprotein E (PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-)) were treated with a helper-dependent adenoviral vector encoding human apoA-I (HDAd-AI) or with control empty vector. Treatment with HDAd-AI increased hepatic apoA-I production, plasma apoA-I and HDL-cholesterol (HDL-C), and apoA-I deposition in lesions to a similar extent in PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-) mice. However, atherosclerosis development at the aortic sinus was considerably lower in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice. A more stable lesion phenotype, with increased collagen content, was primarily associated to treatment with HDAd-AI, but was enhanced under PLIN2 deficiency. PLIN2 deficiency and apoA-I cumulatively reduced LDs and cholesterol ester content in cultured macrophages. Neutral lipid in atheroma was significantly reduced in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice, and RCT from macrophages to feces was enhanced in PLIN2(-/-) macrophages. CONCLUSION: These studies demonstrate a mutually beneficial relationship between PLIN2 deficiency and elevated apoA-I/HDL-C in preventing atherosclerosis development. The data support that targeting foam cell components to mobilize cholesterol may be a promising strategy to enhance the atheroprotection of plasma cholesterol acceptors.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/metabolism , Foam Cells/metabolism , Perilipin-2/metabolism , Animals , Apolipoprotein A-I/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Biological Transport/genetics , Biological Transport/physiology , Cholesterol Esters/genetics , Cholesterol Esters/metabolism , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Mice, Knockout , Perilipin-2/genetics
17.
PLoS One ; 7(3): e33063, 2012.
Article in English | MEDLINE | ID: mdl-22427949

ABSTRACT

Interventions on macrophages/foam cells to redirect intracellular cholesterol towards efflux pathways could become a very valuable addition to our therapeutic arsenal against atherosclerosis. However, certain manipulations of the cholesteryl ester cycle, such as the inhibition of ACAT1, an ER-resident enzyme that re-esterifies cholesterol, are not well tolerated. Previously we showed that targeting perilipin-2 (PLIN2), a major lipid droplet (LD)-associated protein in macrophages, prevents foam cell formation and protects against atherosclerosis. Here we have assessed the tolerance of PLIN2-deficient bone marrow derived macrophages (BMM) to several lipid loading conditions similar to the found during atherosclerosis development, including exposure to modified low-density lipoprotein (mLDL) and 7-ketocholesterol (7-KC), a free cholesterol (FC) metabolite, in media with or without cholesterol acceptors. BMM isolated from mice that do or do not express PLIN2 were tested for apoptosis (TUNEL and cleaved caspase-3), ER stress (CHOP induction and XBP-1 splicing), and inflammation (TNF-α and IL-6 mRNA levels). Like in other cell types, PLIN2 deficiency impairs LD buildup in BMM. However, while most stress parameters were elevated in macrophages under ACAT inhibition and 7-KC loading, PLIN2 inactivation was well tolerated. The data support the safety of targeting PLIN2 to prevent foam cell formation and atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Cholesterol, LDL/metabolism , Macrophages/metabolism , Membrane Proteins/deficiency , Analysis of Variance , Animals , Apoptosis/physiology , Atherosclerosis/prevention & control , Azo Compounds , Blotting, Western , Cell Culture Techniques , Cholesterol, LDL/toxicity , DNA Primers/genetics , Endoplasmic Reticulum Stress/physiology , Foam Cells/metabolism , In Situ Nick-End Labeling , Interleukin-6/metabolism , Ketocholesterols/metabolism , Mice , Mice, Inbred C57BL , Perilipin-2 , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
18.
Mol Endocrinol ; 23(5): 610-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19221051

ABSTRACT

Nuclear receptor (NR) transactivation involves multiple coactivators, and the molecular basis for how these are functionally integrated needs to be determined to fully understand the NR action. Activating signal cointegrator-2 (ASC-2), a transcriptional coactivator of many NRs and transcription factors, forms a steady-state complex, ASCOM (for ASC-2 complex), which contains histone H3-lysine-4 (H3K4) methyltransferase MLL3 or its paralog MLL4. Here, we show that ASCOM requires a functional cross talk with the ATPase-dependent chromatin remodeling complex Swi/Snf for efficient NR transactivation. Our results reveal that ASCOM and Swi/Snf are tightly colocalized in the nucleus and that ASCOM and Swi/Snf promote each other's binding to NR target genes. We further show that the C-terminal SET domain of MLL3 and MLL4 directly interacts with INI1, an integral subunit of Swi/Snf. Our mutational analysis demonstrates that this interaction underlies the mutual facilitation of ASCOM and Swi/Snf recruitment to NR target genes. Importantly, this study uncovers a specific protein-protein interaction as a novel venue to couple two distinct enzymatic coactivator complexes during NR transactivation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/chemistry , DNA-Binding Proteins/chemistry , Fluorescent Antibody Technique, Indirect , HeLa Cells , Histone-Lysine N-Methyltransferase , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Nuclear Receptor Coactivators , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , SMARCB1 Protein , Transcription Factors/chemistry
19.
J Biol Chem ; 279(8): 7131-5, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14645241

ABSTRACT

Activating signal cointegrator-2 (ASC-2), a cancer-amplified transcription coactivator of nuclear receptors and numerous other transcription factors, was previously shown to contain two LXXLL motifs, each of which interacts with a distinct set of nuclear receptors. In this work, we showed that ASC-2 has an indirect, separate binding site for androgen receptor (AR). Interestingly, this region overlapped with the direct interaction interfaces with the tumor suppressor retinoblastoma (Rb). Although ASC-2 alone stimulated AR transactivation in cotransfections of HeLa cells, ectopic expression of Rb effected ASC-2 to act as a transcription coactivator of AR in Rb-null Saos2 cells. These results, along with the previous report in which AR was shown to directly interact with Rb (Yeh, S., Miyamoto, H., Nishimura, K., Kang, H., Ludlow, J., Hsiao, P., Wang, C., Su, C., and Chang C. (1998) Biochem. Biophys. Res. Commun. 248, 361-367), suggest that the AR-ASC-2 interactions in vivo may involve Rb. Thus, ASC-2 appears to contain at least three distinct nuclear receptor interaction domains.


Subject(s)
Intracellular Signaling Peptides and Proteins , Receptors, Androgen/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Amino Acid Motifs , Animals , Chromatin/metabolism , Fibroblasts/metabolism , Glutathione Transferase/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Nuclear Receptor Coactivators , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Androgen/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL