Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 64(5): 100362, 2023 05.
Article in English | MEDLINE | ID: mdl-36958722

ABSTRACT

Cholesterol biosynthesis is a highly regulated pathway, with over 20 enzymes controlled at the transcriptional and posttranslational levels. While some enzymes remain stable, increased sterol levels can trigger degradation of several synthesis enzymes via the ubiquitin-proteasome system. Of note, we previously identified four cholesterol synthesis enzymes as substrates for one E3 ubiquitin ligase, membrane-associated RING-CH-type finger 6 (MARCHF6). Whether MARCHF6 targets the cholesterol synthesis pathway at other points is unknown. In addition, the posttranslational regulation of many cholesterol synthesis enzymes, including the C4-demethylation complex (sterol-C4-methyl oxidase-like, SC4MOL; NAD(P)-dependent steroid dehydrogenase-like, NSDHL; hydroxysteroid 17-beta dehydrogenase, HSD17B7), is largely uncharacterized. Using cultured mammalian cell lines (human-derived and Chinese hamster ovary cells), we show SC4MOL, the first acting enzyme of C4-demethylation, is a MARCHF6 substrate and is rapidly turned over and sensitive to sterols. Sterol depletion stabilizes SC4MOL protein levels, while sterol excess downregulates both transcript and protein levels. Furthermore, we found SC4MOL depletion by siRNA results in a significant decrease in total cell cholesterol. Thus, our work indicates SC4MOL is the most regulated enzyme in the C4-demethylation complex. Our results further implicate MARCHF6 as a crucial posttranslational regulator of cholesterol synthesis, with this E3 ubiquitin ligase controlling levels of at least five enzymes of the pathway.


Subject(s)
Phytosterols , Sterols , Cricetinae , Animals , Humans , Sterols/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , CHO Cells , Cricetulus , Cholesterol/metabolism , Oxidoreductases , 3-Hydroxysteroid Dehydrogenases
2.
EMBO J ; 42(5): e109032, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36715213

ABSTRACT

Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Humans , Mice , Animals , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Gene Expression Regulation , Endothelium, Vascular , Transcription Factors/metabolism , Lymphangiogenesis/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
3.
Nucleic Acids Res ; 49(19): 10931-10955, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34570228

ABSTRACT

Few genetically dominant mutations involved in human disease have been fully explained at the molecular level. In cases where the mutant gene encodes a transcription factor, the dominant-negative mode of action of the mutant protein is particularly poorly understood. Here, we studied the genome-wide mechanism underlying a dominant-negative form of the SOX18 transcription factor (SOX18RaOp) responsible for both the classical mouse mutant Ragged Opossum and the human genetic disorder Hypotrichosis-lymphedema-telangiectasia-renal defect syndrome. Combining three single-molecule imaging assays in living cells together with genomics and proteomics analysis, we found that SOX18RaOp disrupts the system through an accumulation of molecular interferences which impair several functional properties of the wild-type SOX18 protein, including its target gene selection process. The dominant-negative effect is further amplified by poisoning the interactome of its wild-type counterpart, which perturbs regulatory nodes such as SOX7 and MEF2C. Our findings explain in unprecedented detail the multi-layered process that underpins the molecular aetiology of dominant-negative transcription factor function.


Subject(s)
Glomerulonephritis/genetics , Hypotrichosis/genetics , Lymphedema/genetics , SOXF Transcription Factors/genetics , Telangiectasis/genetics , Transcription, Genetic , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Gene Expression Regulation , Gene Regulatory Networks , Genes, Reporter , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Hypotrichosis/metabolism , Hypotrichosis/pathology , Luciferases/genetics , Luciferases/metabolism , Lymphedema/metabolism , Lymphedema/pathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mutation , SOXF Transcription Factors/metabolism , Single Molecule Imaging , Telangiectasis/metabolism , Telangiectasis/pathology
4.
Biochem J ; 477(2): 541-555, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31904814

ABSTRACT

Cholesterol synthesis is a tightly controlled pathway, with over 20 enzymes involved. Each of these enzymes can be distinctly regulated, helping to fine-tune the production of cholesterol and its functional intermediates. Several enzymes are degraded in response to increased sterol levels, whilst others remain stable. We hypothesised that an enzyme at a key branch point in the pathway, lanosterol 14α-demethylase (LDM) may be post-translationally regulated. Here, we show that the preceding enzyme, lanosterol synthase is stable, whilst LDM is rapidly degraded. Surprisingly, this degradation is not triggered by sterols. However, the E3 ubiquitin ligase membrane-associated ring-CH-type finger 6 (MARCH6), known to control earlier rate-limiting steps in cholesterol synthesis, also control levels of LDM and the terminal cholesterol synthesis enzyme, 24-dehydrocholesterol reductase. Our work highlights MARCH6 as the first example of an E3 ubiquitin ligase that targets multiple steps in a biochemical pathway and indicates new facets in the control of cholesterol synthesis.


Subject(s)
Cholesterol/biosynthesis , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Sterol 14-Demethylase/genetics , Ubiquitin-Protein Ligases/genetics , Animals , CHO Cells , Cholesterol/genetics , Cricetulus , HeLa Cells , Humans , Intramolecular Transferases/genetics , Intramolecular Transferases/metabolism , Lipogenesis/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Protein Processing, Post-Translational/genetics , Proteolysis , Sterol 14-Demethylase/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
5.
J Biol Chem ; 294(7): 2436-2448, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30545937

ABSTRACT

The E3 ligase membrane-associated ring-CH-type finger 6 (MARCH6) is a polytopic enzyme bound to the membranes of the endoplasmic reticulum. It controls levels of several known protein substrates, including a key enzyme in cholesterol synthesis, squalene monooxygenase. However, beyond its own autodegradation, little is known about how MARCH6 itself is regulated. Using CRISPR/Cas9 gene-editing, MARCH6 overexpression, and immunoblotting, we found here that cholesterol stabilizes MARCH6 protein endogenously and in HEK293 cells that stably express MARCH6. Conversely, MARCH6-deficient HEK293 and HeLa cells lost their ability to degrade squalene monooxygenase in a cholesterol-dependent manner. The ability of cholesterol to boost MARCH6 did not seem to involve a putative sterol-sensing domain in this E3 ligase, but was abolished when either membrane extraction by valosin-containing protein (VCP/p97) or proteasomal degradation was inhibited. Furthermore, cholesterol-mediated stabilization was absent in two MARCH6 mutants that are unable to degrade themselves, indicating that cholesterol stabilizes MARCH6 protein by preventing its autodegradation. Experiments with chemical chaperones suggested that this likely occurs through a conformational change in MARCH6 upon cholesterol addition. Moreover, cholesterol reduced the levels of at least three known MARCH6 substrates, indicating that cholesterol-mediated MARCH6 stabilization increases its activity. Our findings highlight an important new role for cholesterol in controlling levels of proteins, extending the known repertoire of cholesterol homeostasis players.


Subject(s)
Cholesterol/metabolism , Membrane Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , CRISPR-Cas Systems , Cholesterol/genetics , Enzyme Activation/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Domains , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
6.
Methods Mol Biol ; 1583: 41-52, 2017.
Article in English | MEDLINE | ID: mdl-28205165

ABSTRACT

Cellular cholesterol levels are intricately controlled to maintain homeostasis. Here, we describe ways in which cellular cholesterol status can be manipulated for the study of cholesterol homeostasis, including sterol starvation (by culturing cells in lipoprotein-deficient serum and pretreating/treating with the cholesterol-lowering drug, statin) and sterol enrichment (using cholesterol complexed to cyclodextrin, and low-density lipoprotein). We also describe how to prepare lipoprotein-deficient serum and complex cholesterol to cyclodextrin.


Subject(s)
Cholesterol/metabolism , Cyclodextrins/pharmacology , Homeostasis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/pharmacology , Animals , Cell Line , Cyclodextrins/chemistry , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Lipoproteins, LDL/chemistry
7.
Methods Mol Biol ; 1583: 211-219, 2017.
Article in English | MEDLINE | ID: mdl-28205177

ABSTRACT

The development of gas chromatography/mass spectrometry (GC/MS) technology has improved the ease and efficiency with which sterols in biological samples can be analyzed. Its advantages include that it needs only a small amount of sample, a short analysis time, and has enhanced specificity over traditional methods. Furthermore, a major benefit is its nonselective properties, which means that a complete scan of the sample will display the relative abundance of every sterol in the sample. This property has made it possible to define the abnormal, but distinctive, sterol profiles in a number of inborn errors of cholesterol synthesis. Here, we describe a semiquantitative method to determine relative activity of cholesterol synthesis enzymes. As an example, we measure the relative abundance of the substrate and product sterols of a cholesterol synthetic enzyme, 24-dehydrocholesterol reductase (DHCR24), which is defective in the hereditary developmental disease desmosterolosis.


Subject(s)
Cholesterol , Gas Chromatography-Mass Spectrometry/methods , Nerve Tissue Proteins , Oxidoreductases Acting on CH-CH Group Donors , Abnormalities, Multiple/enzymology , Animals , Cholesterol/biosynthesis , Cholesterol/chemistry , Humans , Lipid Metabolism, Inborn Errors/enzymology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/analysis , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism
8.
J Steroid Biochem Mol Biol ; 165(Pt B): 363-368, 2017 01.
Article in English | MEDLINE | ID: mdl-27520299

ABSTRACT

Cholesterol is essential for survival, but too much or too little can cause disease. Thus, cholesterol levels must be kept within close margins. 7-dehydrocholesterol reductase (DHCR7) is a terminal enzyme of cholesterol synthesis, and is essential for embryonic development. Largely, DHCR7 research is associated with the developmental disease Smith-Lemli-Opitz syndrome, which is caused by mutations in the DHCR7 gene. However, little is known about what regulates DHCR7 activity. Here we provide evidence that phosphorylation plays a role in controlling DHCR7 activity, which may provide a means to divert flux from cholesterol synthesis to vitamin D production. DHCR7 activity was significantly decreased when we used pharmacological inhibitors against two important kinases, AMP-activated protein kinase and protein kinase A. Moreover, mutating a known phosphorylated residue, S14, also decreased DHCR7 activity. Thus, we demonstrate that phosphorylation modulates DHCR7 activity in cells, and contributes to the overall synthesis of cholesterol, and probably vitamin D.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cholesterol/biosynthesis , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Gas Chromatography-Mass Spectrometry , Humans , Mutagenesis, Site-Directed , Mutation , Phosphorylation , RNA, Small Interfering/metabolism , Smith-Lemli-Opitz Syndrome/metabolism , Vitamin D/metabolism
9.
Prog Lipid Res ; 64: 138-151, 2016 10.
Article in English | MEDLINE | ID: mdl-27697512

ABSTRACT

The conversion of 7-dehydrocholesterol to cholesterol, the final step of cholesterol synthesis in the Kandutsch-Russell pathway, is catalyzed by the enzyme 7-dehydrocholesterol reductase (DHCR7). Homozygous or compound heterozygous mutations in DHCR7 lead to the developmental disease Smith-Lemli-Opitz syndrome, which can also result in fetal mortality, highlighting the importance of this enzyme in human development and survival. Besides serving as a substrate for DHCR7, 7-dehydrocholesterol is also a precursor of vitamin D via the action of ultraviolet light on the skin. Thus, DHCR7 exerts complex biological effects, involved in both cholesterol and vitamin D production. Indeed, we argue that DHCR7 can act as a switch between cholesterol and vitamin D synthesis. This review summarizes current knowledge about the critical enzyme DHCR7, highlighting recent findings regarding its structure, transcriptional and post-transcriptional regulation, and its links to vitamin D synthesis. Greater understanding about DHCR7 function, regulation and its place within cellular metabolism will provide important insights into its biological roles.


Subject(s)
Cholesterol/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Vitamin D/metabolism , Animals , Dehydrocholesterols/metabolism , Embryo, Nonmammalian/metabolism , Humans , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/genetics , Protein Domains , Smith-Lemli-Opitz Syndrome/metabolism , Smith-Lemli-Opitz Syndrome/pathology , Xenopus/growth & development , Xenopus/metabolism
10.
J Biol Chem ; 291(16): 8363-73, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26887953

ABSTRACT

Cholesterol is detrimental to human health in excess but is also essential for normal embryogenesis. Hence, enzymes involved in its synthesis possess many layers of regulation to achieve balanced cholesterol levels. 7-Dehydrocholesterol reductase (DHCR7) is the terminal enzyme of cholesterol synthesis in the Kandutsch-Russell pathway, converting 7-dehydrocholesterol (7DHC) to cholesterol. In the absence of functional DHCR7, accumulation of 7DHC and a lack of cholesterol production leads to the devastating developmental disorder, Smith-Lemli-Opitz syndrome. This study identifies that statin treatment can ameliorate the low DHCR7 expression seen with common Smith-Lemli-Opitz syndrome mutations. Furthermore, we show that wild-type DHCR7 is also relatively labile. In an example of end-product inhibition, cholesterol accelerates the proteasomal degradation of DHCR7, resulting in decreased protein levels and activity. The loss of enzymatic activity results in the accumulation of the substrate 7DHC, which leads to an increased production of vitamin D. Thus, these findings highlight DHCR7 as an important regulatory switch between cholesterol and vitamin D synthesis.


Subject(s)
Dehydrocholesterols/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Vitamin D/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Oxidoreductases Acting on CH-CH Group Donors/genetics , Proteasome Endopeptidase Complex/genetics , Smith-Lemli-Opitz Syndrome/enzymology , Smith-Lemli-Opitz Syndrome/genetics , Smith-Lemli-Opitz Syndrome/pathology , Vitamin D/genetics
11.
Annu Rev Pharmacol Toxicol ; 56: 447-67, 2016.
Article in English | MEDLINE | ID: mdl-26738477

ABSTRACT

Oxysterols have long been known for their important role in cholesterol homeostasis, where they are involved in both transcriptional and posttranscriptional mechanisms for controlling cholesterol levels. However, they are increasingly associated with a wide variety of other, sometimes surprising cell functions. They are activators of the Hedgehog pathway (important in embryogenesis), and they act as ligands for a growing list of receptors, including some that are of importance to the immune system. Oxysterols have also been implicated in several diseases such as neurodegenerative diseases and atherosclerosis. Here, we explore the latest research into the roles oxy-sterols play in different areas, and we evaluate the current evidence for these roles. In addition, we outline critical concepts to consider when investigating the roles of oxysterols in various situations, which includes ensuring that the concentration and form of the oxysterol are relevant in that context--a caveat with which many studies have struggled.


Subject(s)
Sterols/metabolism , Animals , Cholesterol/metabolism , Hedgehog Proteins/metabolism , Homeostasis/physiology , Humans
12.
J Nutr Sci Vitaminol (Tokyo) ; 61 Suppl: S154-6, 2015.
Article in English | MEDLINE | ID: mdl-26598836

ABSTRACT

Cholesterol is vital for human life, but its levels must be tightly regulated. Too little cholesterol leads to developmental disorders, but too much is widely appreciated as contributing to heart disease. Levels are regulated through the coordinated control of cholesterol synthesis, uptake and efflux. Here, we focus on cholesterol synthesis. The cholesterol synthesis pathway involves more than twenty enzymes, but most research so far has focused on a very early enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), a well characterised control point. This is largely because HMGCR is the target of the successful cholesterol-lowering drugs, the statins. Our recent work has examined several other enzymes in the pathway and revealed complex regulatory mechanisms that also contribute to the control of cholesterol synthesis. In this review, we discuss the transcriptional regulation of the two terminal enzymes, 7- and 24-dehydrocholesterol reductase (DHCR7 and DHCR24), where we have found that a cooperative transcriptional program exists. We also discuss the post-translational regulation of another critical enzyme, squalene monooxygenase (SM), which has its protein levels controlled by cholesterol, and DHCR24, which has its activity affected by sterols and related compounds, as well as via phosphorylation/signalling. There is an unforeseen complexity in the regulation of cholesterol synthesis which requires further investigation.


Subject(s)
Biosynthetic Pathways , Cholesterol/biosynthesis , Gene Expression Regulation , Hydroxymethylglutaryl CoA Reductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phosphorylation , Squalene Monooxygenase/metabolism
13.
J Lipid Res ; 56(4): 888-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637936

ABSTRACT

Cholesterol is essential to human health, and its levels are tightly regulated by a balance of synthesis, uptake, and efflux. Cholesterol synthesis requires the actions of more than twenty enzymes to reach the final product, through two alternate pathways. Here we describe a physical and functional interaction between the two terminal enzymes. 24-Dehydrocholesterol reductase (DHCR24) and 7-dehydrocholesterol reductase (DHCR7) coimmunoprecipitate, and when the DHCR24 gene is knocked down by siRNA, DHCR7 activity is also ablated. Conversely, overexpression of DHCR24 enhances DHCR7 activity, but only when a functional form of DHCR24 is used. DHCR7 is important for both cholesterol and vitamin D synthesis, and we have identified a novel layer of regulation, whereby its activity is controlled by DHCR24. This suggests the existence of a cholesterol "metabolon", where enzymes from the same metabolic pathway interact with each other to provide a substrate channeling benefit. We predict that other enzymes in cholesterol synthesis may similarly interact, and this should be explored in future studies.


Subject(s)
Cholesterol/biosynthesis , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Gene Knockdown Techniques , Gene Silencing , Humans , Immunoprecipitation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Oxidoreductases Acting on CH-CH Group Donors/genetics , Protein Binding , Proteomics , RNA, Small Interfering/genetics
14.
Biochem J ; 461(3): 435-42, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24840124

ABSTRACT

SM (squalene mono-oxygenase) catalyses the first oxygenation step in cholesterol synthesis, immediately before the formation of the steroid backbone at lanosterol. SM is an important control point in the pathway, and is regulated at the post-translational level by accelerated cholesterol-dependent ubiquitination and proteasomal degradation, which is associated with the accumulation of squalene. Using model cell systems, we report that SM is stabilized by unsaturated fatty acids. Treatment with unsaturated fatty acids such as oleate, but not saturated fatty acids, increased protein levels of SM or SM-N100-GFP (the first 100 amino acids of SM fused to GFP) at the post-translational level and partially overcame cholesterol-dependent degradation, as well as reversing cholesterol-dependent squalene accumulation. Maximum stabilization required activation of fatty acids, but not triacylglycerol or phosphatidylcholine synthesis. The mechanism of oleate-mediated stabilization appeared to occur through reduced ubiquitination by the E3 ubiquitin ligase MARCH6. Stabilization of a cholesterol biosynthetic enzyme by unsaturated fatty acids may help maintain a constant cholesterol/phospholipid ratio.


Subject(s)
Acyl Coenzyme A/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Unsaturated/metabolism , Membrane Proteins/antagonists & inhibitors , Squalene Monooxygenase/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Enzyme Stability , Gene Silencing , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Kinetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oleic Acid/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Squalene Monooxygenase/chemistry , Squalene Monooxygenase/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
15.
J Lipid Res ; 55(3): 410-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24363437

ABSTRACT

The role of signaling in regulating cholesterol homeostasis is gradually becoming more widely recognized. Here, we explored how kinases and phosphorylation sites regulate the activity of the enzyme involved in the final step of cholesterol synthesis, 3ß-hydroxysterol Δ24-reductase (DHCR24). Many factors are known to regulate DHCR24 transcriptionally, but little is known about its posttranslational regulation. We developed a system to specifically test human ectopic DHCR24 activity in a model cell-line (Chinese hamster ovary-7) using siRNA targeted only to hamster DHCR24, thus ensuring that all activity could be attributed to the human enzyme. We determined the effect of known phosphorylation sites and found that mutating certain residues (T110, Y299, and Y507) inhibited DHCR24 activity. In addition, inhibitors of protein kinase C ablated DHCR24 activity, although not through a known phosphorylation site. Our data indicate a novel mechanism whereby DHCR24 activity is regulated by signaling.


Subject(s)
Cholesterol/metabolism , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Signal Transduction , Animals , Binding Sites/genetics , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Desmosterol/metabolism , Gene Expression , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Mutation , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Tyrosine/genetics , Tyrosine/metabolism
16.
IUBMB Life ; 65(8): 675-84, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23847008

ABSTRACT

Cholesterol is a vital lipid and performs diverse functions on a whole body and cellular level. However, excess cellular cholesterol is toxic, and thus, elegant mechanisms have evolved to tightly regulate this important lipid. The regulation of cholesterol homeostasis is an area of intense research, and the role that signalling plays is gradually becoming more widely recognised. Cholesterol homeostasis is achieved through intricate mechanisms involving synthesis, uptake, and efflux. Although there is a large body of work elucidating these cholesterol-related pathways, less is known about the role of signalling in these processes. Here, we discuss the variety of ways that signalling impacts on these modes and levels of cholesterol homeostasis, including transcriptional regulation. Most work thus far has investigated the role of kinases in cholesterol efflux (especially on ATP-binding cassette transporter A1, ABCA1), and therefore constitutes a major focus of this review. We also indicate further avenues to explore in the area of signalling in cellular cholesterol homeostasis.


Subject(s)
Cholesterol/metabolism , Homeostasis , Signal Transduction/physiology , ATP Binding Cassette Transporter 1/physiology , Animals , Calcineurin/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Janus Kinase 2/metabolism , Liver X Receptors , Mitogen-Activated Protein Kinases/metabolism , Orphan Nuclear Receptors/physiology , Phosphorylation , Protein Kinase C/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/physiology
17.
Atherosclerosis ; 228(2): 362-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23582768

ABSTRACT

OBJECTIVE: ATP-binding cassette transporter (ABC)-A1 is an important protein of cholesterol homoeostasis and atherosclerosis as it is the major lipid transporter responsible for the export of cholesterol from cells. Many studies have examined kinase regulation of ABCA1 expression. In contrast, very little is known about whether dephosphorylation events play a role in ABCA1 expression. In this study, we explored the involvement of phosphatases in the regulation of ABCA1 expression. METHODS AND RESULTS: We observed that general protein tyrosine phosphatase inhibitors ablated ABCA1 protein and mRNA when stimulated with synthetic ligands. This effect is transcriptional, and appears to involve the nuclear receptor, retinoid X receptor (RXR). CONCLUSION: Our data demonstrate that inhibition of protein tyrosine phosphatases down-regulates ABCA1 expression, indicating a new level of regulation of a key protein in cholesterol export.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Enzyme Inhibitors/pharmacology , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/antagonists & inhibitors , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Genes, Reporter , HeLa Cells , Humans , Ligands , Liver X Receptors , Mice , Orphan Nuclear Receptors/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/metabolism , Retinoid X Receptors/metabolism , Time Factors , Transcription, Genetic/drug effects , Transfection
18.
J Biol Chem ; 287(40): 33897-904, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22869373

ABSTRACT

How cholesterol is sensed to maintain homeostasis has been explained by direct binding to a specific protein, Scap, or through altering the physical properties of the membrane. The enantiomer of cholesterol (ent-cholesterol) is a valuable tool in distinguishing between these two models because it shares nonspecific membrane effects with native cholesterol (nat-cholesterol), but not specific binding interactions. This is the first study to compare ent- and nat-cholesterol directly on major molecular parameters of cholesterol homeostasis. We found that ent-cholesterol suppressed activation of the master transcriptional regulator of cholesterol metabolism, SREBP-2, almost as effectively as nat-cholesterol. Importantly, ent-cholesterol induced a conformational change in the cholesterol-sensing protein Scap in isolated membranes in vitro, even when steps were taken to eliminate potential confounding effects from endogenous cholesterol. Ent-cholesterol also accelerated proteasomal degradation of the key cholesterol biosynthetic enzyme, squalene monooxygenase. Together, these findings provide compelling evidence that cholesterol maintains its own homeostasis not only via direct protein interactions, but also by altering membrane properties.


Subject(s)
Cholesterol/chemistry , Cholesterol/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Chromatography, Thin Layer/methods , Cricetinae , Homeostasis , Lipid Metabolism , Lipids/chemistry , Protein Binding , Squalene Monooxygenase/chemistry , Stereoisomerism , Sterol Regulatory Element Binding Proteins/metabolism , Trypsin/chemistry
19.
Biochim Biophys Acta ; 1823(2): 458-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22005015

ABSTRACT

Akt is an essential protein kinase for cell growth, proliferation, and survival. Perturbed Akt regulation is associated with a number of human diseases, such as cancer and diabetes. Recently, evidence has emerged that Akt is involved in the regulation of the sterol-regulatory element binding proteins, which are master transcriptional regulators of lipid metabolism. This offers a means by which synthesis of new membrane can be coordinated with cell growth and proliferation. However, the link between Akt and sterol-regulatory element binding protein-2, the major isoform participating in cholesterol regulation, is relatively unexplored. In the present study, we employed insulin-like growth factor-1 as an inducer of Akt signalling, and showed that it increased sterol-regulatory element binding protein-2 activation acutely (within 1h). This insulin-like growth factor-1-induced sterol-regulatory element binding protein-2 activation was blunted when Akt was inhibited pharmacologically or molecularly with small interfering RNA. Furthermore, we employed a rapalog heterodimerisation system to specifically and rapidly activate Akt, and found that sterol-regulatory element binding protein-2 activation was increased in response to Akt activation. Together, this study provides compelling evidence that Akt contributes to the acute regulation of cholesterol metabolism through activating sterol-regulatory element binding protein-2.


Subject(s)
Cholesterol/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation , Humans , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 2/genetics
20.
Traffic ; 12(1): 19-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20950345

ABSTRACT

Regulation of protein transport within the early secretory pathway is a relatively unexplored area. Here, we propose a new player in the control of protein transport from the endoplasmic reticulum (ER) to the Golgi. Akt is an important signaling kinase whose functioning is perturbed in diseases such as cancer and diabetes. We discovered that Akt phosphorylates Sec24, an essential coat protein II (COPII) component involved in mediating cargo selection for ER-to-Golgi trafficking. We discuss how this finding may provide new insights into the regulation of protein transport.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vesicular Transport Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , HeLa Cells , Humans , Mice , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Vesicular Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...