Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
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
2.
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
3.
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
4.
Cell Mol Life Sci ; 73(21): 4101-20, 2016 11.
Article in English | MEDLINE | ID: mdl-27141937

ABSTRACT

Glial fibrillary acidic protein (GFAP) is the characteristic intermediate filament (IF) protein in astrocytes. Expression of its main isoforms, GFAPα and GFAPδ, varies in astrocytes and astrocytoma implying a potential regulatory role in astrocyte physiology and pathology. An IF-network is a dynamic structure and has been functionally linked to cell motility, proliferation, and morphology. There is a constant exchange of IF-proteins with the network. To study differences in the dynamic properties of GFAPα and GFAPδ, we performed fluorescence recovery after photobleaching experiments on astrocytoma cells with fluorescently tagged GFAPs. Here, we show for the first time that the exchange of GFP-GFAPδ was significantly slower than the exchange of GFP-GFAPα with the IF-network. Furthermore, a collapsed IF-network, induced by GFAPδ expression, led to a further decrease in fluorescence recovery of both GFP-GFAPα and GFP-GFAPδ. This altered IF-network also changed cell morphology and the focal adhesion size, but did not alter cell migration or proliferation. Our study provides further insight into the modulation of the dynamic properties and functional consequences of the IF-network composition.


Subject(s)
Astrocytes/cytology , Cell Shape , Focal Adhesions/metabolism , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Actins/metabolism , Adult , Aged , Astrocytes/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Green Fluorescent Proteins/metabolism , Humans , Imaging, Three-Dimensional , Microtubules/metabolism , Nestin/metabolism , Protein Isoforms/metabolism , Vimentin/metabolism
5.
FASEB J ; 28(7): 2942-54, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24696300

ABSTRACT

Glial fibrillary acidic protein (GFAP) is an intermediate filament protein expressed in astrocytes and neural stem cells. The GFAP gene is alternatively spliced, and expression of GFAP is highly regulated during development, on brain damage, and in neurodegenerative diseases. GFAPα is the canonical splice variant and is expressed in all GFAP-positive cells. In the human brain, the alternatively spliced transcript GFAPδ marks specialized astrocyte populations, such as subpial astrocytes and the neurogenic astrocytes in the human subventricular zone. We here show that shifting the GFAP isoform ratio in favor of GFAPδ in astrocytoma cells, by selectively silencing the canonical isoform GFAPα with short hairpin RNAs, induced a change in integrins, a decrease in plectin, and an increase in expression of the extracellular matrix component laminin. Together, this did not affect cell proliferation but resulted in a significantly decreased motility of astrocytoma cells. In contrast, a down-regulation of all GFAP isoforms led to less cell spreading, increased integrin expression, and a >100-fold difference in the adhesion of astrocytoma cells to laminin. In summary, isoform-specific silencing of GFAP revealed distinct roles of a specialized GFAP network in regulating the interaction of astrocytoma cells with the extracellular matrix through laminin.-Moeton, M., Kanski, R., Stassen, O. M. J. A., Sluijs, J. A., Geerts, D., van Tijn, P., Wiche, G., van Strien, M. E., Hol, E. M. Silencing GFAP isoforms in astrocytoma cells disturbs laminin dependent motility and cell adhesion.


Subject(s)
Astrocytoma/metabolism , Cell Adhesion/genetics , Cell Movement/genetics , Glial Fibrillary Acidic Protein/metabolism , Laminin/metabolism , Protein Isoforms/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Astrocytoma/genetics , Astrocytoma/pathology , Brain/metabolism , Brain/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Down-Regulation/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Glial Fibrillary Acidic Protein/genetics , HEK293 Cells , Humans , Integrins/genetics , Integrins/metabolism , Laminin/genetics , Protein Isoforms/genetics
6.
J Neuroinflammation ; 10: 35, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-23452918

ABSTRACT

BACKGROUND: Macrophages play a dual role in multiple sclerosis (MS) pathology. They can exert neuroprotective and growth promoting effects but also contribute to tissue damage by production of inflammatory mediators. The effector function of macrophages is determined by the way they are activated. Stimulation of monocyte-derived macrophages in vitro with interferon-γ and lipopolysaccharide results in classically activated (CA/M1) macrophages, and activation with interleukin 4 induces alternatively activated (AA/M2) macrophages. METHODS: For this study, the expression of a panel of typical M1 and M2 markers on human monocyte derived M1 and M2 macrophages was analyzed using flow cytometry. This revealed that CD40 and mannose receptor (MR) were the most distinctive markers for human M1 and M2 macrophages, respectively. Using a panel of M1 and M2 markers we next examined the activation status of macrophages/microglia in MS lesions, normal appearing white matter and healthy control samples. RESULTS: Our data show that M1 markers, including CD40, CD86, CD64 and CD32 were abundantly expressed by microglia in normal appearing white matter and by activated microglia and macrophages throughout active demyelinating MS lesions. M2 markers, such as MR and CD163 were expressed by myelin-laden macrophages in active lesions and perivascular macrophages. Double staining with anti-CD40 and anti-MR revealed that approximately 70% of the CD40-positive macrophages in MS lesions also expressed MR, indicating that the majority of infiltrating macrophages and activated microglial cells display an intermediate activation status. CONCLUSIONS: Our findings show that, although macrophages in active MS lesions predominantly display M1 characteristics, a major subset of macrophages have an intermediate activation status.


Subject(s)
Brain/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Adult , Aged , Brain/pathology , CD40 Antigens/metabolism , Cells, Cultured , Female , Humans , Inflammation Mediators/physiology , Macrophage Activation/physiology , Male , Middle Aged
7.
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
8.
Cell Rep ; 32(5): 107944, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755570

ABSTRACT

The endothelial monolayer forms a barrier between the lumen of blood vessels and the underlying tissues. Stable VE-cadherin-based adherens junctions are essential for maintaining this barrier, whereas their remodeling is required for angiogenesis in health and disease. Here, we position the ERAD-associated ubiquitin ligase MARCH6 as a determinant of angiogenic sprouting and barrier integrity through its ability to promote the degradation of the rate-limiting cholesterol biosynthetic enzyme squalene epoxidase (SQLE). Accordingly, MARCHF6 ablation in endothelial cells increases SQLE protein and cholesterol load. This leads to altered membrane order, disorganized adherens junctions, decreased endothelial barrier function, and impaired SQLE-dependent sprouting angiogenesis. Akin to MARCHF6 silencing, the overexpression of SQLE impairs angiogenesis. However, angiogenesis is also attenuated when SQLE is silenced, indicating that fine-tuning cholesterol biosynthesis is a determinant of healthy endothelial function. In summary, we propose a mechanistic link between regulation of cholesterol homeostasis by the MARCH6-SQLE axis and endothelial integrity and angiogenesis.


Subject(s)
Cholesterol/metabolism , Homeostasis , Human Umbilical Vein Endothelial Cells/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Squalene Monooxygenase/metabolism , Ubiquitin-Protein Ligases/metabolism , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Antigens, CD/metabolism , Cadherins/metabolism , Gene Silencing , HEK293 Cells , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans
9.
Brain Commun ; 2(2): fcaa150, 2020.
Article in English | MEDLINE | ID: mdl-33376983

ABSTRACT

Neurogenesis continues throughout adulthood in specialized regions of the brain. One of these regions is the subventricular zone. During brain development, neurogenesis is regulated by a complex interplay of intrinsic and extrinsic cues that control stem-cell survival, renewal and cell lineage specification. Cerebrospinal fluid (CSF) is an integral part of the neurogenic niche in development as it is in direct contact with radial glial cells, and it is important in regulating proliferation and migration. Yet, the effect of CSF on neural stem cells in the subventricular zone of the adult human brain is unknown. We hypothesized a persistent stimulating effect of ventricular CSF on neural stem cells in adulthood, based on the literature, describing bulging accumulations of subventricular cells where CSF is in direct contact with the subventricular zone. Here, we show by immunohistochemistry on post-mortem adult human subventricular zone sections that neural stem cells are in close contact with CSF via protrusions through both intact and incomplete ependymal layers. We are the first to systematically quantify subventricular glial nodules denuded of ependyma and consisting of proliferating neural stem and progenitor cells, and showed that they are present from foetal age until adulthood. Neurosphere, cell motility and differentiation assays as well as analyses of RNA expression were used to assess the effects of CSF of adult humans on primary neural stem cells and a human immortalized neural stem cell line. We show that human ventricular CSF increases proliferation and decreases motility of neural stem cells. Our results also indicate that adult CSF pushes neural stem cells from a relative quiescent to a more active state and promotes neuronal over astrocytic lineage differentiation. Thus, CSF continues to stimulate neural stem cells throughout aging.

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.
Nat Commun ; 8(1): 445, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874658

ABSTRACT

Advanced prostate cancer (PCa) is a clinical challenge as no curative therapeutic is available. In this context, a better understanding of metastasis and resistance mechanisms in PCa is an important issue. As phosphatase and tensin homolog (PTEN) loss is the most common genetic lesion in such cancer, we investigate human data sets for mechanisms that can constrain cancer evolution in this setting. Here we report a liver X receptor (LXR) signature, which tightly correlates with PTEN loss, in PCa. Accordingly, the LXR pathway is deregulated in prostate carcinomas in Pten-null mice. Genetic ablation of LXRs in Pten-null mice, exacerbates PCa invasiveness and metastatic dissemination, which involves mesenchymal transition and accumulation of matrix metalloproteinases. Mechanistically, PTEN deletion governed LXR transcriptional activity through deregulation of cholesterol de novo synthesis, resulting in accumulation of endogenous LXR ligands. Our study therefore reveals a functional circuit linking PTEN and LXR, and highlights LXRs as metabolic gatekeepers that are able to constrain PCa progression.Treatment of prostate cancer, especially in its advanced stage, is still challenging; therefore, strategies to prevent metastatic dissemination are of great interest. Here the authors reveal a crucial role for liver X receptors in suppressing prostate carcinogenesis and metastatic progression in PTEN-null tumors.


Subject(s)
Liver X Receptors/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Cholesterol/metabolism , Disease Progression , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Liver X Receptors/deficiency , Male , Mice, Knockout , Neoplasm Metastasis , PTEN Phosphohydrolase/deficiency , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
13.
Oncotarget ; 8(50): 88104-88121, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152145

ABSTRACT

Astrocytomas are the most common malignant brain tumours and are to date incurable. It is unclear how astrocytomas progress into higher malignant grades. The intermediate filament cytoskeleton is emerging as an important regulator of malignancy in several tumours. The majority of the astrocytomas express the intermediate filament protein Glial Fibrillary Acidic Protein (GFAP). Several GFAP splice variants have been identified and the main variants expressed in human astrocytoma are the GFAPα and GFAPδ isoforms. Here we show a significant downregulation of GFAPα in grade IV astrocytoma compared to grade II and III, resulting in an increased GFAPδ/α ratio. Mimicking this increase in GFAPδ/α ratio in astrocytoma cell lines and comparing the subsequent transcriptomic changes with the changes in the patient tumours, we have identified a set of GFAPδ/α ratio-regulated high-malignant and low-malignant genes. These genes are involved in cell proliferation and protein phosphorylation, and their expression correlated with patient survival. We additionally show that changing the ratio of GFAPδ/α, by targeting GFAP expression, affected expression of high-malignant genes. Our data imply that regulating GFAP expression and splicing are novel therapeutic targets that need to be considered as a treatment for astrocytoma.

14.
Mol Cell Biol ; 36(2): 285-94, 2016 01 15.
Article in English | MEDLINE | ID: mdl-26527619

ABSTRACT

Cholesterol synthesis and lipoprotein uptake are tightly coordinated to ensure that the cellular level of cholesterol is adequately maintained. Hepatic dysregulation of these processes is associated with pathological conditions, most notably cardiovascular disease. Using a genetic approach, we have recently identified the E3 ubiquitin ligase MARCH6 as a regulator of cholesterol biosynthesis, owing to its ability to promote degradation of the rate-limiting enzymes 3-hydroxy-3-methyl-glutaryl coenzyme A reductase (HMGCR) and squalene epoxidase (SQLE). Here, we present evidence for MARCH6 playing a multifaceted role in the control of cholesterol homeostasis in hepatocytes. We identify MARCH6 as an endogenous inhibitor of the sterol regulatory element binding protein (SREBP) transcriptional program. Accordingly, loss of MARCH6 increases expression of SREBP-regulated genes involved in cholesterol biosynthesis and lipoprotein uptake. Unexpectedly, this is associated with a decrease in cellular lipoprotein uptake, induced by enhanced lysosomal degradation of the low-density lipoprotein receptor (LDLR). Finally, we provide evidence that induction of the E3 ubiquitin ligase IDOL represents the molecular mechanism underlying this MARCH6-induced phenotype. Our study thus highlights a MARCH6-dependent mechanism to direct cellular cholesterol accretion that relies on uncoupling of cholesterol synthesis from lipoprotein uptake.


Subject(s)
Cholesterol/metabolism , Hepatocytes/metabolism , Lipoproteins/metabolism , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Biosynthetic Pathways , Cell Line , Gene Silencing , Hep G2 Cells , Humans , Membrane Proteins/genetics , Receptors, LDL/metabolism , Ubiquitin-Protein Ligases/genetics
15.
PLoS One ; 10(9): e0138107, 2015.
Article in English | MEDLINE | ID: mdl-26418157

ABSTRACT

Gaucher disease is characterized by lysosomal accumulation of glucosylceramide due to deficient activity of lysosomal glucocerebrosidase (GBA). In cells, glucosylceramide is also degraded outside lysosomes by the enzyme glucosylceramidase 2 (GBA2) of which inherited deficiency is associated with ataxias. The interest in GBA and glucosylceramide metabolism in the brain has grown following the notion that mutations in the GBA gene impose a risk factor for motor disorders such as α-synucleinopathies. We earlier developed a ß-glucopyranosyl-configured cyclophellitol-epoxide type activity based probe (ABP) allowing in vivo and in vitro visualization of active molecules of GBA with high spatial resolution. Labeling occurs through covalent linkage of the ABP to the catalytic nucleophile residue in the enzyme pocket. Here, we describe a method to visualize active GBA molecules in rat brain slices using in vivo labeling. Brain areas related to motor control, like the basal ganglia and motor related structures in the brainstem, show a high content of active GBA. We also developed a ß-glucopyranosyl cyclophellitol-aziridine ABP allowing in situ labeling of GBA2. Labeled GBA2 in brain areas can be identified and quantified upon gel electrophoresis. The distribution of active GBA2 markedly differs from that of GBA, being highest in the cerebellar cortex. The histological findings with ABP labeling were confirmed by biochemical analysis of isolated brain areas. In conclusion, ABPs offer sensitive tools to visualize active GBA and to study the distribution of GBA2 in the brain and thus may find application to establish the role of these enzymes in neurodegenerative disease conditions such as α-synucleinopathies and cerebellar ataxia.


Subject(s)
Brain/enzymology , Gaucher Disease/genetics , Glucosylceramidase/metabolism , Glucosylceramides/metabolism , Neurodegenerative Diseases/genetics , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Fluorescent Antibody Technique , Fluorescent Dyes/chemistry , Gaucher Disease/pathology , Glucosylceramidase/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/metabolism , Microscopy, Confocal , Neurodegenerative Diseases/pathology , Purkinje Cells/metabolism , Rats , Rats, Wistar
16.
Neurobiol Aging ; 35(3): 492-510, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24269023

ABSTRACT

In Alzheimer's disease (AD), amyloid plaques are surrounded by reactive astrocytes with an increased expression of intermediate filaments including glial fibrillary acidic protein (GFAP). Different GFAP isoforms have been identified that are differentially expressed by specific subpopulations of astrocytes and that impose different properties to the intermediate filament network. We studied transcript levels and protein expression patterns of all known GFAP isoforms in human hippocampal AD tissue at different stages of the disease. Ten different transcripts for GFAP isoforms were detected at different abundancies. Transcript levels of most isoforms increased with AD progression. GFAPδ-immunopositive astrocytes were observed in subgranular zone, hilus, and stratum-lacunosum-moleculare. GFAPδ-positive cells also stained for GFAPα. In AD donors, astrocytes near plaques displayed increased staining of both GFAPα and GFAPδ. The reading-frame-shifted isoform, GFAP(+1), staining was confined to a subset of astrocytes with long processes, and their number increased in the course of AD. In conclusion, the various GFAP isoforms show differential transcript levels and are upregulated in a concerted manner in AD. The GFAP(+1) isoform defines a unique subset of astrocytes, with numbers increasing with AD progression. These data indicate the need for future exploration of underlying mechanisms concerning the functions of GFAPδ and GFAP(+1) isoforms in astrocytes and their possible role in AD pathology.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Astrocytes/metabolism , Astrocytes/pathology , Glial Fibrillary Acidic Protein/metabolism , Plaque, Amyloid/metabolism , Cells, Cultured , Disease Progression , Gene Expression , Hippocampus/metabolism , Humans , Protein Isoforms , Severity of Illness Index , Transcription, Genetic , Up-Regulation
17.
PLoS One ; 7(8): e42823, 2012.
Article in English | MEDLINE | ID: mdl-22912745

ABSTRACT

Glial fibrillary acidic protein (GFAP) is the main astrocytic intermediate filament (IF). GFAP splice isoforms show differential expression patterns in the human brain. GFAPδ is preferentially expressed by neurogenic astrocytes in the subventricular zone (SVZ), whereas GFAP(+1) is found in a subset of astrocytes throughout the brain. In addition, the expression of these isoforms in human brain material of epilepsy, Alzheimer and glioma patients has been reported. Here, for the first time, we present a comprehensive study of GFAP isoform expression in both wild-type and Alzheimer Disease (AD) mouse models. In cortex, cerebellum, and striatum of wild-type mice, transcripts for Gfap-α, Gfap-ß, Gfap-γ, Gfap-δ, Gfap-κ, and a newly identified isoform Gfap-ζ, were detected. Their relative expression levels were similar in all regions studied. GFAPα showed a widespread expression whilst GFAPδ distribution was prominent in the SVZ, rostral migratory stream (RMS), neurogenic astrocytes of the subgranular zone (SGZ), and subpial astrocytes. In contrast to the human SVZ, we could not establish an unambiguous GFAPδ localization in proliferating cells of the mouse SVZ. In APPswePS1dE9 and 3xTgAD mice, plaque-associated reactive astrocytes had increased transcript levels of all detectable GFAP isoforms and low levels of a new GFAP isoform, Gfap-ΔEx7. Reactive astrocytes in AD mice showed enhanced GFAPα and GFAPδ immunolabeling, less frequently increased vimentin and nestin, but no GFAPκ or GFAP(+1) staining. In conclusion, GFAPδ protein is present in SVZ, RMS, and neurogenic astrocytes of the SGZ, but also outside neurogenic niches. Furthermore, differential GFAP isoform expression is not linked with aging or reactive gliosis. This evidence points to the conclusion that differential regulation of GFAP isoforms is not involved in the reorganization of the IF network in reactive gliosis or in neurogenesis in the mouse brain.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Aging/genetics , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Animals , Antibody Specificity , Brain/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein , Intermediate Filament Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Pharmacogenet Genomics ; 19(1): 67-76, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18987562

ABSTRACT

BACKGROUND: In major depressive disorder, selective serotonin reuptake inhibitors target the serotonin transporter (SERT). Their response rates (30-50%) are modified by SERT promotor polymorphisms (5-HTTLPR). OBJECTIVES: To quantify the relationship between SERT occupancy and response, and whether 5-HTTLPR is a modifier. METHODS: Drug-free depressed outpatients (n=49; both sexes; aged 25-55 years), received paroxetine (20 mg/day). We quantified SERT occupancy with iodine-123-labeled 2beta-carbomethoxy-3beta-(4-iodophenyl)-tropane single-photon emission computed tomography imaging at baseline and after 6 weeks; we genotyped 5-HTTLPR (S, L(G), L(A)). PRIMARY OUTCOMES: percentage decrease in 17-item Hamilton Depression Rating Scale and response (> or =50% decrease of 17-item Hamilton Depression Rating Scale). RESULTS: A significant positive relationship between SERT occupancy and clinical response existed only in the L(A)/L(A) genotype (P<0.002). Relative to paroxetine serum concentrations maximal midbrain SERT occupancy was numerically higher for L(A)/L(A) compared with other genotypes, but this difference was nonsignificant (P=0.188). CONCLUSION: Higher SERT occupancy is only associated with more clinical improvement in the L(A)/L(A) genotype. We hypothesize that the L(A)/L(A) carriers have a more dynamic serotonergic system, which seems more responsive to selective serotonin reuptake inhibitors. (ISRCTN Trial Register ISRCTN44111488; http://www.trialregister.nl/trialreg/admin/rctview.asp?TC=193).


Subject(s)
Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Paroxetine/metabolism , Paroxetine/therapeutic use , Polymorphism, Genetic , Promoter Regions, Genetic , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Adult , Antidepressive Agents, Second-Generation/metabolism , Antidepressive Agents, Second-Generation/therapeutic use , Base Sequence , DNA Primers/genetics , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/metabolism , Diencephalon/diagnostic imaging , Diencephalon/metabolism , Female , Genotype , Humans , Male , Mesencephalon/diagnostic imaging , Mesencephalon/metabolism , Middle Aged , Pharmacogenetics , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/therapeutic use , Tomography, Emission-Computed, Single-Photon
SELECTION OF CITATIONS
SEARCH DETAIL