49 results on '"Kiss RS"'
Search Results
2. Corrigendum to "PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease" [Metabolism Volume 150, January 2024, 155736, PMID: 7967646].
- Author
-
Sachan V, LeDévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Sesiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQ, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, and Seidah NG
- Published
- 2024
- Full Text
- View/download PDF
3. Foam fractionation studies of recombinant human apolipoprotein A-I.
- Author
-
Lethcoe K, Fox CA, Hafiane A, Kiss RS, Liu J, Ren G, and Ryan RO
- Subjects
- Humans, Recombinant Proteins genetics, Recombinant Proteins metabolism, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Lipoproteins, HDL metabolism, Lipoproteins, HDL chemistry, Lipoproteins, HDL genetics, Apolipoprotein A-I genetics, Apolipoprotein A-I chemistry, Apolipoprotein A-I metabolism, Escherichia coli genetics, Escherichia coli metabolism, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism
- Abstract
Apolipoprotein A-I (apoA-I), the primary protein component of plasma high-density lipoproteins (HDL), is comprised of two structural regions, an N-terminal amphipathic α-helix bundle domain (residues 1-184) and a hydrophobic C-terminal domain (residues 185-243). When a recombinant fusion protein construct [bacterial pelB leader sequence - human apoA-I (1-243)] was expressed in Escherichia coli shaker flask cultures, apoA-I was recovered in the cell lysate. By contrast, when the C-terminal domain was deleted from the construct, large amounts of the truncated protein, apoA-I (1-184), were recovered in the culture medium. Consequently, following pelB leader sequence cleavage in the E. coli periplasmic space, apoA-I (1-184) was secreted from the bacteria. When the pelB-apoA-I (1-184) fusion construct was expressed in a 5 L bioreactor, substantial foam production (~30 L) occurred. Upon foam collection and collapse into a liquid foamate, SDS-PAGE revealed that apoA-I (1-184) was the sole major protein present. Incubation of apoA-I (1-184) with phospholipid vesicles yielded reconstituted HDL (rHDL) particles that were similar in size and cholesterol efflux capacity to those generated with full-length apoA-I. Mass spectrometry analysis confirmed that pelB leader sequence cleavage occurred and that foam fractionation did not result in unwanted protein modifications. The facile nature and scalability of bioreactor-based apolipoprotein foam fractionation provide a novel means to generate a versatile rHDL scaffold protein., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease.
- Author
-
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQ, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, and Seidah NG
- Subjects
- Mice, Animals, Subtilisin metabolism, Triglycerides metabolism, Liver metabolism, Apolipoproteins B genetics, Apolipoproteins B metabolism, Proprotein Convertases metabolism, Apolipoprotein B-100 genetics, Apolipoprotein B-100 metabolism, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Background: Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7., Methods: We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7
-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7., Results: Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results., Conclusions: Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Nabil G Seidah has patent METHOD FOR REDUCING HEPATIC TRIGLYCERIDES pending to Nabil G Seidah., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
5. Comparative proximity biotinylation implicates the small GTPase RAB18 in sterol mobilization and biosynthesis.
- Author
-
Kiss RS, Chicoine J, Khalil Y, Sladek R, Chen H, Pisaturo A, Martin C, Dale JD, Brudenell TA, Kamath A, Kyei-Boahen J, Hafiane A, Daliah G, Alecki C, Hopes TS, Heier M, Aligianis IA, Lebrun JJ, Aspden J, Paci E, Kerksiek A, Lütjohann D, Clayton P, Wills JC, von Kriegsheim A, Nilsson T, Sheridan E, and Handley MT
- Subjects
- Humans, Cells, Cultured, Cholesterol biosynthesis, Cholesterol metabolism, Gene Knockdown Techniques, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors metabolism, HeLa Cells, Protein Transport genetics, rab3 GTP-Binding Proteins metabolism, Calcium Channels genetics, Calcium Channels metabolism, Biotinylation, rab GTP-Binding Proteins genetics, rab GTP-Binding Proteins metabolism, Sterols biosynthesis, Sterols metabolism
- Abstract
Loss of functional RAB18 causes the autosomal recessive condition Warburg Micro syndrome. To better understand this disease, we used proximity biotinylation to generate an inventory of potential RAB18 effectors. A restricted set of 28 RAB18 interactions were dependent on the binary RAB3GAP1-RAB3GAP2 RAB18-guanine nucleotide exchange factor complex. Twelve of these 28 interactions are supported by prior reports, and we have directly validated novel interactions with SEC22A, TMCO4, and INPP5B. Consistent with a role for RAB18 in regulating membrane contact sites, interactors included groups of microtubule/membrane-remodeling proteins, membrane-tethering and docking proteins, and lipid-modifying/transporting proteins. Two of the putative interactors, EBP and OSBPL2/ORP2, have sterol substrates. EBP is a Δ8-Δ7 sterol isomerase, and ORP2 is a lipid transport protein. This prompted us to investigate a role for RAB18 in cholesterol biosynthesis. We found that the cholesterol precursor and EBP-product lathosterol accumulates in both RAB18-null HeLa cells and RAB3GAP1-null fibroblasts derived from an affected individual. Furthermore, de novo cholesterol biosynthesis is impaired in cells in which RAB18 is absent or dysregulated or in which ORP2 expression is disrupted. Our data demonstrate that guanine nucleotide exchange factor-dependent Rab interactions are highly amenable to interrogation by proximity biotinylation and may suggest that Micro syndrome is a cholesterol biosynthesis disorder., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
6. Isolation of recombinant apolipoprotein E4 N-terminal domain by foam fractionation.
- Author
-
Lethcoe K, Fox CA, Hafiane A, Kiss RS, and Ryan RO
- Subjects
- Humans, Carrier Proteins, Recombinant Proteins chemistry, Apolipoprotein E4 genetics, Apolipoprotein E4 metabolism, Apolipoproteins E genetics, Apolipoproteins E chemistry, Apolipoproteins E metabolism
- Abstract
Apolipoprotein (apo) E functions in lipoprotein metabolism as a low density lipoprotein receptor ligand. ApoE is comprised of two structural domains, a 22 kDa N-terminal (NT) domain that adopts a helix bundle conformation and a 10 kDa C-terminal domain with strong lipid binding affinity. The NT domain is capable of transforming aqueous phospholipid dispersions into discoidal reconstituted high density lipoprotein (rHDL) particles. Given the utility of apoE-NT as a structural component of rHDL, expression studies were conducted. A plasmid construct encoding a pelB leader sequence fused to the N-terminus of human apoE4 (residues 1-183) was transformed into Escherichia coli. Upon expression, the fusion protein is directed to the periplasmic space where leader peptidase cleaves the pelB sequence, generating mature apoE4-NT. In shaker flask expression cultures, apoE4-NT escapes the bacteria and accumulates in the medium. In a bioreactor setting, however, apoE4-NT was found to combine with gas and liquid components in the culture medium to generate large quantities of foam. When this foam was collected in an external vessel and collapsed into a liquid foamate, analysis revealed that apoE4-NT was the sole major protein present. The product protein was further isolated by heparin affinity chromatography (60-80 mg/liter bacterial culture), shown to be active in rHDL formulation, and documented to serve as an acceptor of effluxed cellular cholesterol. Thus, foam fractionation provides a streamlined process to produce recombinant apoE4-NT for biotechnology applications., Competing Interests: Declaration of competing interest None., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. High Density Lipoprotein-Based Therapeutics: Novel Mechanism of Probucol in Foam Cells.
- Author
-
Hafiane A, Ronca A, Kiss RS, and Favari E
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
8. Probucol treatment is associated with an ABCA1-independent mechanism of cholesterol efflux to lipid poor apolipoproteins from foam cell macrophages.
- Author
-
Hafiane A, Pisaturo A, Ronca A, Incerti M, Kiss RS, and Favari E
- Abstract
Objective: Probucol is a cholesterol-lowering agent whose ability to prevent atherosclerosis is currently under study. Herein, we investigate the putative mechanism of probucol by observation of changes in cellular cholesterol efflux and lipid droplet morphology in macrophages., Results: The inhibitory activity of probucol was assessed in non-foam or foam cell macrophages expressing ABCA1 generated by treatment with fetal calf serum (FCS) alone or in combination with acetylated LDL, respectively. Probucol inhibited cholesterol efflux to apolipoprotein A-I (apoA-I) by 31.5±0.1% in THP-1 non-foam cells and by 18.5±0.2% in foam cells. In probucol-treated non-foam THP-1 cells, nascent high density lipoprotein (nHDL) particles with a diameter < 7 nm were generated, while in probucol-treated THP-1 foam cells nHDL particles of > 7 nm in diameter containing cholesterol were produced. Foam cells also displayed a significant accumulation of free cholesterol at the plasma membrane, as measured by percent cholestenone formed. Intracellularly, there was a significant decrease in lipid droplet number and an increase in size in probucol-treated THP-1 foam cells when compared to non-treated cells., Conclusions: We report for the first time that probucol is unable to completely inhibit cholesterol efflux in foam cells to the same extent as in non-foam cells. Indeed, functional nHDL is released from foam cells in the presence of probucol. This difference in inhibitory effect could potentially be explained by changes in the plasma membrane pool as well as intracellular cholesterol storage independently of ABCA1., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2021 The Authors. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
9. Proprotein convertase 7 (PCSK7) reduces apoA-V levels.
- Author
-
Ashraf Y, Duval S, Sachan V, Essalmani R, Susan-Resiga D, Roubtsova A, Hamelin J, Gerhardy S, Kirchhofer D, Tagliabracci VS, Prat A, Kiss RS, and Seidah NG
- Subjects
- Animals, Apolipoprotein A-V blood, Cell Line, Tumor, Endoplasmic Reticulum metabolism, Hepatocytes metabolism, Humans, Lysosomes metabolism, Mice, Inbred C57BL, Mice, Knockout, Polymorphism, Single Nucleotide, Subtilisins metabolism, Triglycerides blood, Exome Sequencing methods, Apolipoprotein A-V metabolism, Liver metabolism, Subtilisins genetics, Triglycerides metabolism
- Abstract
The locus of the human proprotein convertase subtilisin-kexin type-7 (PC7) gene (PCSK7) is on chromosome 11q23.3 close to the gene cluster APOA5/APOA4/APOC3/APOA1, a region implicated in the regulation of lipoprotein metabolism. A GWAS reported the association of PCSK7 SNPs with plasma triglyceride (TG), and exome sequencing of African Americans revealed the association of a low-frequency coding variant of PC7 (R504H; SNP rs142953140) with a ~ 30% TG reduction. Another PCSK7 SNP rs508487 is in linkage disequilibrium with a promoter variant of the liver-derived apolipoprotein A-V (apoA-V), an indirect activator of the lipoprotein lipase (LpL), and is associated with elevated TG levels. We thus hypothesized that PC7 regulates the levels/activity of apoA-V. Studies in the human hepatic cell line HuH7 revealed that wild-type (WT) PC7 and its endoplasmic reticulum (ER)-retained forms bind to and enhance the degradation of human apoA-V in acidic lysosomes in a nonenzymatic fashion. PC7-induced degradation of apoA-V is inhibited by bafilomycin A1 and the alkalinizing agents: chloroquine and NH
4 Cl. Thus, the PC7-induced apoA-V degradation implicates an ER-lysosomal communication inhibited by bafilomycin A1. In vitro, the natural R504H mutant enhances PC7 Ser505 phosphorylation at the structurally exposed Ser-X-Glu507 motif recognized by the secretory kinase Fam20C. Co-expression of the phosphomimetic PC7-S505E with apoA-V resulted in lower degradation compared to WT, suggesting that Ser505 phosphorylation of PC7 lowers TG levels via reduced apoA-V degradation. In agreement, in Pcsk7-/- mice fed high-fat diet, plasma apoA-V levels and adipocyte LpL activity are increased, providing an in vivo mechanistic link for a role of liver PC7 in enhanced TG storage in adipocytes., (© 2020 Federation of European Biochemical Societies.)- Published
- 2020
- Full Text
- View/download PDF
10. C. elegans rab-18 mutants display reduced lipid content under fed and fasted conditions.
- Author
-
Ratemi Z, Kiss RS, and Rocheleau CE
- Published
- 2019
- Full Text
- View/download PDF
11. A facile method for isolation of recombinant human apolipoprotein A-I from E. coli.
- Author
-
Ikon N, Shearer J, Liu J, Tran JJ, Feng S, Kamei A, Beckstead JA, Kiss RS, Weers PM, Ren G, and Ryan RO
- Subjects
- Biological Transport, Active drug effects, Cell Line, Cholesterol metabolism, Escherichia coli chemistry, Escherichia coli genetics, Humans, Macrophages metabolism, Protein Structure, Secondary, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Recombinant Proteins pharmacology, Apolipoprotein A-I biosynthesis, Apolipoprotein A-I chemistry, Apolipoprotein A-I isolation & purification, Apolipoprotein A-I pharmacokinetics, Escherichia coli metabolism, Protein Refolding
- Abstract
Apolipoprotein (apo) A-I is the major protein component of high-density lipoprotein (HDL) and plays key roles in the Reverse Cholesterol Transport pathway. In the past decade, reconstituted HDL (rHDL) has been employed as a therapeutic agent for treatment of atherosclerosis. The ability of rHDL to promote cholesterol efflux from peripheral cells has been documented to reduce the size of atherosclerotic plaque lesions. However, development of apoA-I rHDL-based therapeutics for human use requires a cost effective process to generate an apoA-I product that meets "Good Manufacturing Practice" standards. Methods available for production and isolation of unmodified recombinant human apoA-I at scale are cumbersome, laborious and complex. To overcome this obstacle, a streamlined two-step procedure has been devised for isolation of recombinant untagged human apoA-I from E. coli that takes advantage of its ability to re-fold to a native conformation following denaturation. Heat treatment of a sonicated E. coli supernatant fraction induced precipitation of a large proportion of host cell proteins (HCP), yielding apoA-I as the major soluble protein. Reversed-phase HPLC of this material permitted recovery of apoA-I largely free of HCP and endotoxin. Purified apoA-I possessed α-helix secondary structure, formed rHDL upon incubation with phospholipid and efficiently promoted cholesterol efflux from cholesterol loaded J774 macrophages., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
12. Palmitate mediated diacylglycerol accumulation causes endoplasmic reticulum stress, Plin2 degradation, and cell death in H9C2 cardiomyoblasts.
- Author
-
Akoumi A, Haffar T, Mousterji M, Kiss RS, and Bousette N
- Subjects
- Animals, Cell Death drug effects, Cell Line, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum metabolism, Fatty Acids metabolism, Leupeptins pharmacology, Lipid Droplets drug effects, Lipid Droplets metabolism, Mice, Myocytes, Cardiac drug effects, Oleic Acid pharmacology, Oxidation-Reduction drug effects, Proteasome Inhibitors pharmacology, Proteolysis drug effects, Rats, Diglycerides metabolism, Endoplasmic Reticulum Stress drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Palmitic Acid pharmacology, Perilipin-2 metabolism
- Abstract
We have previously shown that palmitate causes ER stress in primary cardiomyocytes and this was associated with a diffuse lipid staining histology. In contrast, oleate, which was non-toxic, led to the formation of abundant, clearly delineated lipid droplets. The aberrant lipid histology in palmitate treated cells led us to hypothesize that perhaps there was an impairment in lipid droplet formation, which could lead to accumulation of lipids in the ER and consequent ER stress. To test this hypothesis we treated H9C2s (a cardiomyoblast cell line) with either 300µM oleate or palmitate for 8h. We found that palmitate resulted in significantly less lipid droplet abundance despite elevated intracellular lipid accumulation. Next we showed that palmitate was packaged primarily as diacylglycerol (DAG), in contrast oleate formed primarily triacylglycerol (TAG). Furthermore, the palmitate induced DAG accumulated mostly in the ER, while oleate treatment resulted in accumulation of TAG primarily in lipid droplets. The palmitate-induced accumulation of lipid in the ER was associated with a strong ER stress response. Interestingly, we found that ER stress induced by either palmitate, tunicamycin, or thapsigargin led to the degradation of Plin2, an important lipid droplet binding protein. In contrast palmitate had little effect on either Plin3 or Plin5. Furthermore, we found that acute MG132 administration significantly attenuated palmitate mediated ER stress and cell death. This protection was associated with a moderate attenuation of Plin2 degradation., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
13. Statins, PCSK9 inhibitors and cholesterol homeostasis: a view from within the hepatocyte.
- Author
-
Sniderman AD, Kiss RS, Reid T, Thanassoulis G, and Watts GF
- Subjects
- Cholesterol blood, Hepatocytes metabolism, Humans, Lipid Metabolism drug effects, Lipoproteins, LDL blood, Lipoproteins, LDL metabolism, Liver cytology, Liver drug effects, Liver metabolism, Models, Biological, Proprotein Convertase 9 metabolism, Receptors, LDL metabolism, Cholesterol metabolism, Hepatocytes drug effects, Homeostasis drug effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, PCSK9 Inhibitors
- Abstract
Statins and PCSK9 inhibitors dramatically lower plasma LDL levels and dramatically increase LDL receptor number within hepatocyte cell membranes. It seems self-evident that total clearance of LDL particles from plasma and total delivery of cholesterol to the liver must increase in consequence. However, based on the results of stable isotope tracer studies, this analysis demonstrates the contrary to be the case. Statins do not change the production rate of LDL particles. Accordingly, at steady state, the clearance rate cannot change. Because LDL particles contain less cholesterol on statin therapy, the delivery of cholesterol to the liver must, therefore, be reduced. PCSK9 inhibitors reduce the production of LDL particles and this further reduces cholesterol delivery to the liver. With both agents, a larger fraction of a smaller pool is removed per unit time. These findings are inconsistent with the conventional model of cholesterol homeostasis within the liver, but are consistent with a new model of regulation, the multi-channel model, which postulates that different lipoprotein particles enter the hepatocyte by different routes and have different metabolic fates within the hepatocyte. The multi-channel model, but not the conventional model, may explain how statins and PCSK9 inhibitors can produce sustained increases in LDL receptor number., (© 2017 The Author(s); published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2017
- Full Text
- View/download PDF
14. The Proprotein Convertase Subtilisin/Kexin Type 9-resistant R410S Low Density Lipoprotein Receptor Mutation: A NOVEL MECHANISM CAUSING FAMILIAL HYPERCHOLESTEROLEMIA.
- Author
-
Susan-Resiga D, Girard E, Kiss RS, Essalmani R, Hamelin J, Asselin MC, Awan Z, Butkinaree C, Fleury A, Soldera A, Dory YL, Baass A, and Seidah NG
- Subjects
- Amino Acid Substitution, Endosomes genetics, Female, Humans, Lysosomes genetics, Male, Mutation, Missense, Protein Binding, Endosomes metabolism, Hyperlipoproteinemia Type II genetics, Hyperlipoproteinemia Type II metabolism, Lipoproteins, LDL metabolism, Lysosomes metabolism, Proprotein Convertase 9 metabolism, Receptors, LDL genetics, Receptors, LDL metabolism
- Abstract
Familial hypercholesterolemia (FH) is characterized by severely elevated low density lipoprotein (LDL) cholesterol. Herein, we identified an FH patient presenting novel compound heterozygote mutations R410S and G592E of the LDL receptor (LDLR). The patient responded modestly to maximum rosuvastatin plus ezetimibe therapy, even in combination with a PCSK9 monoclonal antibody injection. Using cell biology and molecular dynamics simulations, we aimed to define the underlying mechanism(s) by which these LDLR mutations affect LDL metabolism and lead to hypercholesterolemia. Our data showed that the LDLR-G592E is a class 2b mutant, because it mostly failed to exit the endoplasmic reticulum and was degraded. Even though LDLR-R410S and LDLR-WT were similar in levels of cell surface and total receptor and bound equally well to LDL or extracellular PCSK9, the LDLR-R410S was resistant to exogenous PCSK9-mediated degradation in endosomes/lysosomes and showed reduced LDL internalization and degradation relative to LDLR-WT. Evidence is provided for a tighter association of LDL with LDLR-R410S at acidic pH, a reduced LDL delivery to late endosomes/lysosomes, and an increased release in the medium of the bound/internalized LDL, as compared with LDLR-WT. These data suggested that LDLR-R410S recycles loaded with its LDL-cargo. Our findings demonstrate that LDLR-R410S represents an LDLR loss-of-function through a novel class 8 FH-causing mechanism, thereby rationalizing the observed phenotype., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
15. PGC-1 coactivators in β-cells regulate lipid metabolism and are essential for insulin secretion coupled to fatty acids.
- Author
-
Oropeza D, Jouvet N, Bouyakdan K, Perron G, Ringuette LJ, Philipson LH, Kiss RS, Poitout V, Alquier T, and Estall JL
- Abstract
Objectives: Peroxisome proliferator-activated receptor γ coactivator 1 (PPARGCA1, PGC-1) transcriptional coactivators control gene programs important for nutrient metabolism. Islets of type 2 diabetic subjects have reduced PGC-1α expression and this is associated with decreased insulin secretion, yet little is known about why this occurs or what role it plays in the development of diabetes. Our goal was to delineate the role and importance of PGC-1 proteins to β-cell function and energy homeostasis., Methods: We investigated how nutrient signals regulate coactivator expression in islets and the metabolic consequences of reduced PGC-1α and PGC-1β in primary and cultured β-cells. Mice with inducible β-cell specific double knockout of Pgc-1α/Pgc-1β (βPgc-1 KO) were created to determine the physiological impact of reduced Pgc1 expression on glucose homeostasis., Results: Pgc-1α and Pgc-1β expression was increased in primary mouse and human islets by acute glucose and palmitate exposure. Surprisingly, PGC-1 proteins were dispensable for the maintenance of mitochondrial mass, gene expression, and oxygen consumption in response to glucose in adult β-cells. However, islets and mice with an inducible, β-cell-specific PGC-1 knockout had decreased insulin secretion due in large part to loss of the potentiating effect of fatty acids. Consistent with an essential role for PGC-1 in lipid metabolism, β-cells with reduced PGC-1s accumulated acyl-glycerols and PGC-1s controlled expression of key enzymes in lipolysis and the glycerolipid/free fatty acid cycle., Conclusions: These data highlight the importance of PGC-1s in coupling β-cell lipid metabolism to promote efficient insulin secretion.
- Published
- 2015
- Full Text
- View/download PDF
16. Apoptotic cells trigger a membrane-initiated pathway to increase ABCA1.
- Author
-
Fond AM, Lee CS, Schulman IG, Kiss RS, and Ravichandran KS
- Subjects
- ATP Binding Cassette Transporter 1 deficiency, ATP Binding Cassette Transporter 1 genetics, Adaptor Proteins, Signal Transducing deficiency, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Angiogenic Proteins genetics, Angiogenic Proteins metabolism, Animals, Cell Membrane metabolism, Cholesterol metabolism, Female, Humans, Jurkat Cells, Lipid Metabolism, Liver X Receptors, Macrophages cytology, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Neuropeptides deficiency, Neuropeptides genetics, Neuropeptides metabolism, Orphan Nuclear Receptors metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction, Up-Regulation, rac1 GTP-Binding Protein deficiency, rac1 GTP-Binding Protein genetics, rac1 GTP-Binding Protein metabolism, ATP Binding Cassette Transporter 1 biosynthesis, Apoptosis physiology
- Abstract
Macrophages clear millions of apoptotic cells daily and, during this process, take up large quantities of cholesterol. The membrane transporter ABCA1 is a key player in cholesterol efflux from macrophages and has been shown via human genetic studies to provide protection against cardiovascular disease. How the apoptotic cell clearance process is linked to macrophage ABCA1 expression is not known. Here, we identified a plasma membrane-initiated signaling pathway that drives a rapid upregulation of ABCA1 mRNA and protein. This pathway involves the phagocytic receptor brain-specific angiogenesis inhibitor 1 (BAI1), which recognizes phosphatidylserine on apoptotic cells, and the intracellular signaling intermediates engulfment cell motility 1 (ELMO1) and Rac1, as ABCA1 induction was attenuated in primary macrophages from mice lacking these molecules. Moreover, this apoptotic cell-initiated pathway functioned independently of the liver X receptor (LXR) sterol-sensing machinery that is known to regulate ABCA1 expression and cholesterol efflux. When placed on a high-fat diet, mice lacking BAI1 had increased numbers of apoptotic cells in their aortic roots, which correlated with altered lipid profiles. In contrast, macrophages from engineered mice with transgenic BAI1 overexpression showed greater ABCA1 induction in response to apoptotic cells compared with those from control animals. Collectively, these data identify a membrane-initiated pathway that is triggered by apoptotic cells to enhance ABCA1 within engulfing phagocytes and with functional consequences in vivo.
- Published
- 2015
- Full Text
- View/download PDF
17. Rab proteins implicated in lipid storage and mobilization.
- Author
-
Kiss RS and Nilsson T
- Abstract
Abnormal intracellular accumulation or transport of lipids contributes greatly to the pathogenesis of human diseases. In the liver, excess accumulation of triacylglycerol (TG) leads to fatty liver disease encompassing steatosis, steatohepatitis and fibrosis. This places individuals at risk of developing cirrhosis, hepatocellular carcinoma or hepatic decompensation and also contributes to the emergence of insulin resistance and dyslipidemias affecting many other organs. Excessive accumulation of TG in adipose tissue contributes to insulin resistance as well as to the release of cytokines attracting leucocytes leading to a pro-inflammatory state. Pathological accumulation of cholesteryl ester (CE) in macrophages in the arterial wall is the progenitor of atherosclerotic plaques and heart disease. Overconsumption of dietary fat, cholesterol and carbohydrates explains why these diseases are on the increase yet offers few clues for how to prevent or treat individuals. Dietary regimes have proven futile and barring surgery, no realistic alternatives are at hand as effective drugs are few and not without side effects. Overweight and obesity-related diseases are no longer restricted to the developed world and as such, constitute a global problem. Development of new drugs and treatment strategies are a priority yet requires as a first step, elucidation of the molecular pathophysiology underlying each associated disease state. The lipid droplet (LD), an up to now overlooked intracellular organelle, appears at the heart of each pathophysiology linking key regulatory and metabolic processes as well as constituting the site of storage of both TGs and CEs. As the molecular machinery and mechanisms of LDs of each cell type are being elucidated, regulatory proteins used to control various cellular processes are emerging. Of these and the subject of this review, small GTPases belonging to the Rab protein family appear as important molecular switches used in the regulation of the intracellular trafficking and storage of lipids.
- Published
- 2014
- Full Text
- View/download PDF
18. Hepatic cholesterol homeostasis: is the low-density lipoprotein pathway a regulatory or a shunt pathway?
- Author
-
Sniderman AD, Qi Y, Ma CI, Wang RH, Naples M, Baker C, Zhang J, Adeli K, and Kiss RS
- Subjects
- Animals, Cholesterol Esters biosynthesis, Cholesterol Esters metabolism, Cholesterol, HDL biosynthesis, Cholesterol, HDL metabolism, Cholesterol, LDL biosynthesis, Cholesterol, VLDL biosynthesis, Chylomicrons metabolism, Cricetinae, Fibroblasts metabolism, Homeostasis genetics, Hydroxymethylglutaryl CoA Reductases genetics, Hydroxymethylglutaryl CoA Reductases metabolism, Lipid Metabolism genetics, Receptors, LDL genetics, Receptors, LDL metabolism, Cholesterol, LDL metabolism, Cholesterol, VLDL metabolism, Hepatocytes metabolism, Homeostasis physiology, Lipid Metabolism physiology
- Abstract
Objective: The hypothesis that cholesterol that enters the cell within low-density lipoprotein (LDL) particles rapidly equilibrates with the regulatory pool of intracellular cholesterol and maintains cholesterol homeostasis by reducing cholesterol and LDL receptor synthesis was validated in the fibroblast but not in the hepatocyte. Accordingly, the present studies were designed to compare the effects of cholesterol that enters the hepatocyte within an LDL particle with those of cholesterol that enters via other lipoprotein particles., Approach and Results: We measured cholesterol synthesis and esterification in hamster hepatocytes treated with LDL and other lipoprotein particles, including chylomicron remnants and VLDL. Endogenous cholesterol synthesis was not significantly reduced by uptake of LDL, but cholesterol esterification (280%) and acyl CoA:cholesterol acyltransferase 2 expression (870%) were increased. In contrast, cholesterol synthesis was significantly reduced (70% decrease) with other lipoprotein particles. Furthermore, more cholesterol that entered the hepatocyte within LDL particles was secreted within VLDL particles (480%) compared with cholesterol from other sources., Conclusions: Much of the cholesterol that enters the hepatocyte within LDL particles is shunted through the cell and resecreted within VLDL particles without reaching equilibrium with the regulatory pool.
- Published
- 2013
- Full Text
- View/download PDF
19. Cholesterol trapping in Niemann-Pick disease type B fibroblasts can be relieved by expressing the phosphotyrosine binding domain of GULP.
- Author
-
Lee CY, Ruel I, Denis M, Genest J, and Kiss RS
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters metabolism, Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing genetics, Animals, CHO Cells, Cells, Cultured, Cricetinae, Cricetulus, Esterification, Female, Fibroblasts cytology, Heterozygote, Humans, Lysosomes metabolism, Male, Microscopy, Fluorescence, Middle Aged, Mutation, Niemann-Pick Disease, Type B metabolism, Niemann-Pick Disease, Type B pathology, Phosphotyrosine chemistry, Protein Binding, Sphingomyelin Phosphodiesterase genetics, Sphingomyelins metabolism, Sterol Regulatory Element Binding Protein 2 metabolism, Transfection, Adaptor Proteins, Signal Transducing metabolism, Cholesterol metabolism, Fibroblasts metabolism, Phosphotyrosine metabolism
- Abstract
Background: Impairment of acid sphingomyelinase (SMase) results in accumulation of sphingomyelin (SM) and cholesterol in late endosomes, the hallmarks of a lysosomal storage disease., Objective: We describe cellular lipid metabolism in fibroblasts from two patients with novel compound heterozygote mutations in the sphingomyelin phosphodiesterase 1 (SMPD1) gene manifesting as Niemann-Pick disease type B (NPB) and demonstrate mechanisms to overcome the storage defect., Methods: Using biochemical assays and confocal microscopy, we provide evidence that accumulated lysosomal SM and cholesterol can be released by different treatments., Results: Defective SMase activity in these fibroblasts results in a 2.5-fold increased cellular mass of SM and cholesterol, increased de novo endogenous cholesterol synthesis, and decreased cholesterol esterification, demonstrating impaired intracellular cholesterol homeostasis. Depletion of exogenous addition of cholesterol for 24 hours or addition of the cholesterol acceptor apolipoprotein A-I are sufficient to restore normal homeostatic responses. In an effort to correct the lysosomal storage phenotype of NPB, we infected the fibroblasts with a lentivirus expressing the phosphotyrosine binding domain of the adapter protein GULP (PTB-GULP). We have previously shown that expression of PTB-GULP in Chinese hamster ovary cells promotes intracellular cholesterol trafficking and ABCA1-mediated cholesterol efflux. We find that expression of PTB-GULP in NPB fibroblasts results in increased ABCA1 expression, increased cellular cholesterol efflux and lysosomal cholesterol redistribution, independent of the impaired SMase and cholesterol presence., Conclusion: We provide extensive functional characterization of a novel compound heterozygote mutation and provide a novel functional mechanism to overcome lysosomal storage disease defects., (Copyright © 2013 National Lipid Association. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
20. Tweaking the cholesterol efflux capacity of reconstituted HDL.
- Author
-
Ma CI, Beckstead JA, Thompson A, Hafiane A, Wang RH, Ryan RO, and Kiss RS
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Animals, Apolipoprotein A-I genetics, Apolipoprotein A-I metabolism, CHO Cells, Cells, Cultured, Cricetinae, Transfection, Cholesterol metabolism, Lipoproteins, HDL metabolism
- Abstract
Mechanisms to increase plasma high-density lipoprotein (HDL) or to promote egress of cholesterol from cholesterol-loaded cells (e.g., foam cells from atherosclerotic lesions) remain an important target to regress heart disease. Reconstituted HDL (rHDL) serves as a valuable vehicle to promote cellular cholesterol efflux in vitro and in vivo. rHDL were prepared with wild type apolipoprotein (apo) A-I and the rare variant, apoA-I Milano (M), and each apolipoprotein was reconstituted with phosphatidylcholine (PC) or sphingomyelin (SM). The four distinct rHDL generated were incubated with CHO cells, J774 macrophages, and BHK cells in cellular cholesterol efflux assays. In each cell type, apoA-I(M) SM-rHDL promoted the greatest cholesterol efflux. In BHK cells, the cholesterol efflux capacities of all four distinct rHDL were greatly enhanced by increased expression of ABCG1. Efflux to PC-containing rHDL was stimulated by transfection of a nonfunctional ABCA1 mutant (W590S), suggesting that binding to ABCA1 represents a competing interaction. This interpretation was confirmed by binding experiments. The data show that cholesterol efflux activity is dependent upon the apoA-I protein employed, as well as the phospholipid constituent of the rHDL. Future studies designed to optimize the efflux capacity of therapeutic rHDL may improve the value of this emerging intervention strategy.
- Published
- 2012
- Full Text
- View/download PDF
21. Engulfment protein GULP is regulator of transforming growth factor-β response in ovarian cells.
- Author
-
Ma CI, Martin C, Ma Z, Hafiane A, Dai M, Lebrun JJ, and Kiss RS
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adenocarcinoma genetics, Adenocarcinoma pathology, Animals, CHO Cells, Cricetinae, Cricetulus, Endosomes genetics, Endosomes metabolism, Endosomes pathology, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Mice, Neoplasm Invasiveness, Neoplasm Proteins genetics, Ovarian Neoplasms pathology, Smad3 Protein genetics, Smad3 Protein metabolism, Transforming Growth Factor beta genetics, Adaptor Proteins, Signal Transducing biosynthesis, Adenocarcinoma metabolism, Cell Movement, Neoplasm Proteins metabolism, Ovarian Neoplasms metabolism, Transforming Growth Factor beta metabolism
- Abstract
Transforming growth factor β (TGF-β) is a key regulatory molecule with pleiotropic effects on cell growth, migration, and invasion. As a result, impairment of proper TGF-β signaling is central to tumorigenesis and metastasis. The TGF-β receptor V (TGFBRV or LRP1) has been shown to be responsible for TGF-β-mediated cell growth inhibition in Chinese hamster ovary (CHO) cells. The LRP1 adapter protein GULP mediates internalization of the various LRP1-specific ligands, and we hypothesize that GULP acts as a novel regulator of TGF-β signaling in ovarian cells. CHO cells that overexpress exogenous GULP (FL) demonstrate enhancement in growth inhibition, migration, and invasion from TGF-β treatment, whereas cells that lack GULP (AS) show impairment of growth inhibition and decreased migration and invasion. The enhanced TGF-β response in FL cells was confirmed by a prolonged TGF-β-induced SMAD3 phosphorylation, whereas a shortening of the phosphorylation event is observed in AS cells. Mechanistically, the presence of GULP retains the TGF-β in a signaling-competent early endosome for enhanced signaling. To address this mechanism in a physiological setting, TGF-β insensitive ovarian adenocarcinoma cells (HEY) have a very low GULP expression level, similar to the observation made in a wide selection of human ovarian adenocarcinomas. Transfection of GULP into the HEY cells restored the TGF-β responsiveness, as measured by SMAD3 phosphorylation and impairment of cell growth. Because GULP expression positively regulates TGF-β signaling leading to growth inhibition, this may represent an attractive target to achieve TGF-β responsiveness in ovarian cells.
- Published
- 2012
- Full Text
- View/download PDF
22. Urotensin II differentially regulates macrophage and hepatic cholesterol homeostasis.
- Author
-
Kiss RS, You Z, Genest J Jr, Behm DJ, and Giaid A
- Subjects
- Acetyl-CoA C-Acetyltransferase metabolism, Animals, Blotting, Western, Cells, Cultured, Cholesterol Esters metabolism, Cholesterol, LDL metabolism, Chromatography, High Pressure Liquid, Hepatocytes drug effects, Hepatocytes metabolism, Mice, Mice, Knockout, Cholesterol metabolism, Homeostasis drug effects, Liver drug effects, Liver metabolism, Macrophages drug effects, Macrophages metabolism, Urotensins pharmacology
- Abstract
Urotensin II (UII) is a vasoactive peptide with pleiotropic activity. Interestingly, UII levels are elevated in hyperlipidemic patients, and UII induces lipase activity in some species. However, the exact role UII plays in cholesterol homeostasis remains to be elucidated. UII knockout (UII KO) mice were generated and a plasma lipoprotein profile, and hepatocytes and macrophages cholesterol uptake, storage and synthesis was determined. UII KO had a decreased LDL cholesterol profile and liver steatosis compared to wildtype mice (WT). UII KO macrophages demonstrated enhanced ACAT activity and LDL uptake in the short term (up to 4h), of which more LDL-delivered exogenously derived cholesterol was incorporated into cholesteryl ester (CE) than the WT macrophages. UII KO macrophages generated more than two times the amount of de novo endogenously synthesized cholesterol, and of this cholesterol more than two times the relative amount was esterified to CE. In comparison, results in hepatocytes demonstrated that far more exogenously derived cholesterol was incorporated into CE in the WT cells, generating almost ten times the amount of CE than UII KO. WT cells synthesize de novo almost ten times the amount of cholesterol than UIIKO, and of that cholesterol, almost two times the amount of CE in WT than UII KO hepatocytes. In addition, more ApoB lipoproteins were secreted from WT than UII KO hepatocytes. These results demonstrate a fundamental difference between macrophages and hepatocytes in terms of cholesterol homeostasis, and suggest an important role for UII in modulating cholesterol regulation., (Crown Copyright © 2011. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
23. Novel N-terminal mutation of human apolipoprotein A-I reduces self-association and impairs LCAT activation.
- Author
-
Weers PM, Patel AB, Wan LC, Guigard E, Kay CM, Hafiane A, McPherson R, Marcel YL, and Kiss RS
- Subjects
- Humans, Kinetics, Male, Middle Aged, Phosphatidylcholine-Sterol O-Acyltransferase chemistry, Phosphatidylcholines metabolism, Structure-Activity Relationship, Apolipoprotein A-I chemistry, Apolipoprotein A-I genetics, Mutation, Phosphatidylcholine-Sterol O-Acyltransferase metabolism
- Abstract
We have identified a novel mutation in apoA-I (serine 36 to alanine; S36A) in a human subject with severe hypoalphalipoproteinemia. The mutation is located in the N-terminal region of the protein, which has been implicated in several functions, including lipid binding and lecithin:cholesterol acyltransferase (LCAT) activity. In the present study, the S36A protein was produced recombinantly and characterized both structurally and functionally. While the helical content of the mutant protein was lower compared with wild-type (WT) apoA-I, it retained its helical character. The protein stability, measured as the resistance to guanidine-induced denaturation, decreased significantly. Interestingly, native gel electrophoresis, cross-linking, and sedimentation equilibrium analysis showed that the S36A mutant was primarily present as a monomer, notably different from the WT protein, which showed considerable oligomeric forms. Although the ability of S36A apoA-I to solubilize phosphatidylcholine vesicles and bind to lipoprotein surfaces was not altered, a significantly impaired LCAT activation compared with the WT protein was observed. These results implicate a region around S36 in apoA-I self-association, independent of the intact C terminus. Furthermore, the region around S36 in the N-terminus of human apoA-I is necessary for LCAT activation.
- Published
- 2011
- Full Text
- View/download PDF
24. The helix bundle: a reversible lipid binding motif.
- Author
-
Narayanaswami V, Kiss RS, and Weers PM
- Subjects
- Animals, Apolipoproteins metabolism, Humans, Lipid Metabolism, Models, Molecular, Protein Binding, Protein Conformation, Apolipoproteins chemistry, Lipids chemistry, Protein Folding, Protein Structure, Secondary
- Abstract
Apolipoproteins are the protein components of lipoproteins that have the innate ability to inter convert between a lipid-free and a lipid-bound form in a facile manner, a remarkable property conferred by the helix bundle motif. Composed of a series of four or five amphipathic alpha-helices that fold to form a helix bundle, this motif allows the en face orientation of the hydrophobic faces of the alpha-helices in the protein interior in the lipid-free state. A conformational switch then permits helix-helix interactions to be substituted by helix-lipid interactions upon lipid binding interaction. This review compares the apolipoprotein high-resolution structures and the factors that trigger this switch in insect apolipophorin III and the mammalian apolipoproteins, apolipoprotein E and apolipoprotein A-I, pointing out the commonalities and key differences in the mode of lipid interaction. Further insights into the lipid-bound conformation of apolipoproteins are required to fully understand their functional role under physiological conditions., (2009 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
25. Regulation of plasma LDL: the apoB paradigm.
- Author
-
Sniderman AD, De Graaf J, Couture P, Williams K, Kiss RS, and Watts GF
- Subjects
- Cholesterol metabolism, Hepatocytes metabolism, Homeostasis physiology, Humans, Receptors, LDL physiology, Signal Transduction physiology, Apolipoproteins B blood, Lipoproteins, LDL blood, Models, Biological
- Abstract
The objectives of this analysis are to re-examine the foundational studies of the in vivo metabolism of plasma LDL (low-density lipoprotein) particles in humans and, based on them, to reconstruct our understanding of the governance of the concentration of plasma LDL and the maintenance of cholesterol homoeostasis in the hepatocyte. We believe that regulation of cholesterol homoeostasis within the hepatocyte is demonstrably more complex than envisioned by the LDL receptor paradigm, the conventional model to explain the regulation of plasma LDL and the fluxes of cholesterol into the liver, a model which was generated in the fibroblast but has never been fully validated in the hepatocyte. We suggest that the LDL receptor paradigm should be reconfigured as the apoB (apolipoprotein B) paradigm, which states that the rate at which LDL particles are produced is at least an important determinant of their concentration in plasma as the rate at which they are cleared from plasma and that secretion of cholesterol within VLDL (very-low-density lipoprotein) particles is an important mechanism of maintaining cholesterol homoeostasis within the hepatocyte. These two paradigms are not mutually exclusive. The LDL receptor paradigm, however, includes only one critical aspect of the regulation of plasma LDL, namely the rate at which LDL particles are cleared through the LDL receptor pathway, but ignores another - the rate at which LDL particles are added to the plasma compartment. The apoB paradigm includes both and points to a different model of how the hepatocyte achieves cholesterol homoeostasis in a complex metabolic environment.
- Published
- 2009
- Full Text
- View/download PDF
26. Urotensin II receptor knockout mice on an ApoE knockout background fed a high-fat diet exhibit an enhanced hyperlipidemic and atherosclerotic phenotype.
- Author
-
Bousette N, D'Orleans-Juste P, Kiss RS, You Z, Genest J, Al-Ramli W, Qureshi ST, Gramolini A, Behm D, Ohlstein EH, Harrison SM, Douglas SA, and Giaid A
- Subjects
- Acetyl-CoA C-Acetyltransferase metabolism, Animals, Aorta drug effects, Aorta pathology, Apolipoproteins E genetics, Atherosclerosis genetics, Atherosclerosis pathology, Atherosclerosis physiopathology, Blood Glucose metabolism, Blood Pressure, Cells, Cultured, Cholesterol Esters metabolism, Dietary Fats administration & dosage, Disease Models, Animal, Fatty Liver metabolism, Fatty Liver prevention & control, Genotype, Hyperlipidemias genetics, Hyperlipidemias pathology, Hyperlipidemias physiopathology, Insulin blood, Lipids blood, Liver drug effects, Liver pathology, Macrophages, Peritoneal metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Phenotype, Protein Kinase Inhibitors pharmacology, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, G-Protein-Coupled genetics, Receptors, Scavenger metabolism, Time Factors, Aorta metabolism, Apolipoproteins E deficiency, Atherosclerosis metabolism, Hyperlipidemias metabolism, Liver metabolism, Receptors, G-Protein-Coupled deficiency, Urotensins metabolism
- Abstract
Rationale: Expression of the vasoactive peptide Urotensin II (UII) is elevated in a number of cardiovascular diseases., Objective: Here, we sought to determine the effect of UII receptor (UT) gene deletion in a mouse model of atherosclerosis., Methods and Results: UT knockout (KO) mice were crossed with ApoE KO mice to generate UT/ApoE double knockout (DKO) mice. Mice were placed on a high-fat Western-type diet for 12 weeks. We evaluated the degree of atherosclerosis and hepatic steatosis by histology. In addition, serum glucose, insulin, and lipids were determined. DKO mice exhibited significantly increased atherosclerosis compared to ApoE KO mice (P<0.05). This was associated with a significant increase in serum insulin and lipids (P<0.001) but a decrease in hepatic steatosis (P<0.001). UT gene deletion led to a significant increase in systolic pressure and pulse pressure. RT-PCR and immunoblot analyses showed significant reductions in hepatic scavenger receptors, nuclear receptors, and acyl-CoA:cholesterol acyltransferase (ACAT1) expression in DKO mice. UII induced a significant increase in intracellular cholesteryl ester formation in primary mouse hepatocytes, which was blocked by the MEK inhibitor, PD98059. Hepatocytes of UTKO mice showed a significant reduction in lipoprotein uptake compared to wild-type mice., Conclusions: We propose that UT gene deletion in an ApoE-deficient background promotes downregulation of ACAT1, which in turn attenuates hepatic lipoprotein receptor-mediated uptake and lipid transporter expression. As the liver is the main organ for uptake of lipoprotein-derived lipids, DKO leads to an increase in hyperlipidemia, with a concomitant decrease in hepatic steatosis, and consequently increased atherosclerotic lesion formation. Furthermore, the hypertension associated with UT gene deletion is likely to contribute to the increased atherosclerotic burden.
- Published
- 2009
- Full Text
- View/download PDF
27. The strengths and limitations of the apoB/apoA-I ratio to predict the risk of vascular disease: a Hegelian analysis.
- Author
-
Sniderman AD and Kiss RS
- Subjects
- Cholesterol, HDL blood, Cholesterol, LDL blood, Humans, Predictive Value of Tests, Risk Assessment, Apolipoprotein A-I blood, Apolipoproteins B blood, Vascular Diseases blood
- Abstract
The levels of pro- and antiatherogenic lipoproteins are the most important risk factors for vascular disease, and there is now compelling evidence that the apolipoprotein (apo) B/apoA-I ratio is a better index of the likelihood of vascular events than any of the corresponding cholesterol indices: the total cholesterol/high-density lipoprotein cholesterol (HDL-C) ratio, non-HDL-C/HDL-C ratio, or low-density lipoprotein cholesterol (LDL-C)/HDL-C ratio. But are there any restrictions on the application of the apoB/apoA-I ratio to clinical practice? This article suggests that the answer is yes. Based on the available biologic and epidemiologic data, the relation between risk and apoB is continuous, whereas at the extremes of HDL concentration in plasma the relation to risk is not certain. Moreover, LDL plays a causal role in atherogenesis whereas HDL plays a contingent role. Appreciating these distinctions should allow appropriate use of the apoB/apoA-I ratio as a simple, single, summary index of the lipoprotein-related risk of vascular disease.
- Published
- 2007
- Full Text
- View/download PDF
28. Differential regulation of ATP binding cassette protein A1 expression and ApoA-I lipidation by Niemann-Pick type C1 in murine hepatocytes and macrophages.
- Author
-
Wang MD, Franklin V, Sundaram M, Kiss RS, Ho K, Gallant M, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, Animals, Cathepsin D metabolism, Intracellular Signaling Peptides and Proteins, Liver metabolism, Mice, Niemann-Pick C1 Protein, Pepstatins metabolism, Protein Transport, RNA Processing, Post-Transcriptional, ATP-Binding Cassette Transporters metabolism, Apolipoprotein A-I metabolism, Gene Expression Regulation, Hepatocytes metabolism, Lipids chemistry, Macrophages metabolism, Niemann-Pick Diseases metabolism, Proteins metabolism
- Abstract
Niemann-Pick type C1 (Npc1) protein inactivation results in lipid accumulation in late endosomes and lysosomes, leading to a defect of ATP binding cassette protein A1 (Abca1)-mediated lipid efflux to apolipoprotein A-I (apoA-I) in macrophages and fibroblasts. However, the role of Npc1 in Abca1-mediated lipid efflux to apoA-I in hepatocytes, the major cells contributing to HDL formation, is still unknown. Here we show that, whereas lipid efflux to apoA-I in Npc1-null macrophages is impaired, the lipidation of endogenously synthesized apoA-I by low density lipoprotein-derived cholesterol or de novo synthesized cholesterol or phospholipids in Npc1-null hepatocytes is significantly increased by about 1-, 3-, and 8-fold, respectively. The increased cholesterol efflux reflects a major increase of Abca1 protein in Npc1-null hepatocytes, which contrasts with the decrease observed in Npc1-null macrophages. The increased Abca1 expression is largely post-transcriptional, because Abca1 mRNA is only slightly increased and Lxr alpha mRNA is not changed, and Lxr alpha target genes are reduced. This differs from the regulation of Abcg1 expression, which is up-regulated at both mRNA and protein levels in Npc1-null cells. Abca1 protein translation rate is higher in Npc1-null hepatocytes, compared with wild type hepatocytes as measured by [(35)S]methionine incorporation, whereas there is no difference for the degradation of newly synthesized Abca1 in these two types of hepatocytes. Cathepsin D, which we recently identified as a positive modulator of Abca1, is markedly increased at both mRNA and protein levels by Npc1 inactivation in hepatocytes but not in macrophages. Consistent with this, inhibition of cathepsin D with pepstatin A reduced the Abca1 protein level in both Npc1-inactivated and WT hepatocytes. Therefore, Abca1 expression is specifically regulated in hepatocytes, where Npc1 activity modulates cathepsin D expression and Abca1 protein translation rate.
- Published
- 2007
- Full Text
- View/download PDF
29. Genetic etiology of isolated low HDL syndrome: incidence and heterogeneity of efflux defects.
- Author
-
Kiss RS, Kavaslar N, Okuhira K, Freeman MW, Walter S, Milne RW, McPherson R, and Marcel YL
- Subjects
- Apolipoprotein A-I metabolism, Biomarkers blood, Blotting, Western, Cholesterol, LDL blood, Electrophoresis, Agar Gel, Genetic Predisposition to Disease, Humans, Hypercholesterolemia blood, Middle Aged, Phenotype, Phospholipid Transfer Proteins blood, Phospholipid Transfer Proteins genetics, Sterol O-Acyltransferase blood, Sterol O-Acyltransferase genetics, Syndrome, Apolipoprotein A-I genetics, Cholesterol, HDL blood, Hypercholesterolemia genetics, Mutation, RNA genetics
- Abstract
Objective: We have used a multitiered approach to identify genetic and cellular contributors to high-density lipoprotein (HDL) deficiency in 124 human subjects., Methods and Results: We resequenced 4 candidate genes for HDL regulation and identified several functional nonsynonymous mutations including 2 in apolipoprotein A-I (APOA1), 4 in lecithin:cholesterol acyltransferase (LCAT), 1 in phospholipid transfer protein (PLTP), and 7 in the ATP-binding cassette transporter ABCA1, leaving 88% (110/124) of HDL deficient subjects without a genetic diagnosis. Cholesterol efflux assays performed using cholesterol-loaded monocyte-derived macrophages from the 124 low HDL subjects and 48 control subjects revealed that 33% (41/124) of low HDL subjects had low efflux, despite the fact that the majority of these subjects (34/41) were not carriers of dysfunctional ABCA1 alleles. In contrast, only 2% of control subjects presented with low efflux (1/48). In 3 families without ABCA1 mutations, efflux defects were found to cosegregate with low HDL., Conclusions: Efflux defects are frequent in low HDL syndromes, but the majority of HDL deficient subjects with cellular cholesterol efflux defects do not harbor ABCA1 mutations, suggesting that novel pathways contribute to this phenotype.
- Published
- 2007
- Full Text
- View/download PDF
30. Predominance of a proinflammatory phenotype in monocyte-derived macrophages from subjects with low plasma HDL-cholesterol.
- Author
-
Sarov-Blat L, Kiss RS, Haidar B, Kavaslar N, Jaye M, Bertiaux M, Steplewski K, Hurle MR, Sprecher D, McPherson R, and Marcel YL
- Subjects
- Atherosclerosis blood, Atherosclerosis etiology, Biomarkers blood, Cholesterol, HDL blood, Fatty Acid-Binding Proteins biosynthesis, Fatty Acid-Binding Proteins genetics, Genotype, Humans, Hypolipoproteinemias complications, Hypolipoproteinemias genetics, Interleukin-1beta biosynthesis, Interleukin-1beta genetics, Interleukin-8 biosynthesis, Interleukin-8 genetics, Membrane Proteins biosynthesis, Membrane Proteins genetics, Microarray Analysis, Mutation, PPAR delta biosynthesis, PPAR gamma biosynthesis, Perilipin-2, Phenotype, Polymerase Chain Reaction, Risk Factors, Tumor Necrosis Factor-alpha biosynthesis, Tumor Necrosis Factor-alpha genetics, Cholesterol, HDL deficiency, Gene Expression, Hypolipoproteinemias blood, Macrophages metabolism, PPAR delta genetics, PPAR gamma genetics, RNA genetics
- Abstract
Objective: Reduced plasma concentrations of high-density lipoprotein-cholesterol (HDL-C) are a significant risk factor for cardiovascular disease. Mechanisms that regulate HDL-C concentrations represent an important area of investigation., Methods and Results: Comparative transcriptome analyses of monocyte-derived macrophages (MDM) from a large population of low HDL-C subjects and age- and sex-matched controls revealed a cluster of inflammatory genes highly expressed in low HDL-C subjects. The expression levels of peroxisome proliferator activated receptor (PPAR) gamma and several antioxidant metallothionein genes were decreased in MDM from all low HDL-C groups compared with controls, as was the expression of other genes regulated by PPARgamma, including CD36, adipocyte fatty acid binding protein (FABP4), and adipophilin (ADFP). In contrast, PPARdelta expression was increased in MDM from low HDL-C groups. Quantitative RT-PCR corroborated all major findings from the microarray analysis in two separate patient cohorts. Expression of several inflammatory cytokine genes including interleukin 1beta, interleukin 8, and tumor necrosis factor alpha were highly increased in low HDL-C subjects., Conclusions: The activated proinflammatory state of monocytes and MDM in low HDL-C subjects constitutes a novel parameter of risk associated with HDL deficiency, related to altered expression of metallothionein genes and the reciprocal regulation of PPARgamma and PPARdelta.
- Published
- 2007
- Full Text
- View/download PDF
31. Different cellular traffic of LDL-cholesterol and acetylated LDL-cholesterol leads to distinct reverse cholesterol transport pathways.
- Author
-
Wang MD, Kiss RS, Franklin V, McBride HM, Whitman SC, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Animals, Biological Transport, Blotting, Western, Caveolin 1 genetics, Caveolin 1 metabolism, Caveolin 1 physiology, Cells, Cultured, Female, Intracellular Signaling Peptides and Proteins, Liver metabolism, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Niemann-Pick C1 Protein, Proteins genetics, Proteins metabolism, Proteins physiology, Receptors, LDL genetics, Receptors, LDL metabolism, Receptors, LDL physiology, ATP-Binding Cassette Transporters physiology, Cholesterol metabolism, Cholesterol, LDL metabolism, Lipoproteins, LDL metabolism
- Abstract
Endocytosis of LDL and modified LDL represents regulated and unregulated cholesterol delivery to macrophages. To elucidate the mechanisms of cellular cholesterol transport and egress under both conditions, various primary macrophages were labeled and loaded with cholesterol or cholesteryl ester from LDL or acetylated low density lipoprotein (AcLDL), and the cellular cholesterol traffic pathways were examined. Confocal microscopy using fluorescently labeled 3,3'-dioctyldecyloxacarbocyanine perchlorate-labeled LDL and 1,1'-dioctyldecyl-3,3,3',3'-tetramethylindodicarbocyanine perchlorate-labeled AcLDL demonstrated their discrete traffic pathways and accumulation in distinct endosomes. ABCA1-mediated cholesterol efflux to apolipoprotein A-I (apoA-I) was much greater for AcLDL-loaded macrophages compared with LDL. Treatment with the liver X receptor ligand 22-OH increased efflux to apoA-I in AcLDL-loaded but not LDL-loaded cells. In contrast, at a level equivalent to AcLDL, LDL-derived cholesterol was preferentially effluxed to HDL, in keeping with increased ABCG1. In vivo studies of reverse cholesterol transport (RCT) from cholesterol-labeled macrophages injected intraperitoneally demonstrated that LDL-derived cholesterol was more efficiently transported to the liver and secreted into bile than AcLDL-derived cholesterol. This indicates a greater efficiency of HDL than lipid-poor apoA-I in interstitial fluid in controlling in vivo RCT. These assays, taken together, emphasize the importance of mediators of diffusional cholesterol efflux in RCT.
- Published
- 2007
- Full Text
- View/download PDF
32. Cathepsin D, a lysosomal protease, regulates ABCA1-mediated lipid efflux.
- Author
-
Haidar B, Kiss RS, Sarov-Blat L, Brunet R, Harder C, McPherson R, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters physiology, Animals, Biological Transport, Active physiology, CHO Cells, Cathepsin D antagonists & inhibitors, Cathepsin D biosynthesis, Cathepsin D genetics, Cell Line, Chlorocebus aethiops, Cholesterol, HDL metabolism, Cricetinae, Gene Expression Regulation genetics, Humans, Intracellular Fluid enzymology, Lipoproteins, HDL biosynthesis, Lipoproteins, HDL metabolism, Macrophages enzymology, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes enzymology, Monocytes metabolism, ATP-Binding Cassette Transporters metabolism, Cathepsin D physiology, Lipid Metabolism physiology, Lysosomes enzymology, Peptide Hydrolases physiology
- Abstract
To identify genes involved in the regulation of plasma high density lipoprotein (HDL) cholesterol (HDL-C) levels, patients with low HDL-C and age- and sex-matched controls (normal HDL-C) were extensively characterized. Comparative transcriptome analysis was carried out in cholesterol-loaded monocyte-derived macrophages from low HDL subjects segregated into groups with or without cholesterol efflux defects or ABCA1 mutations. Clusters of differentially regulated genes were evident in the low HDL groups as compared with controls. Of particular note, expression of cathepsin D (CTSD), a lysosomal proteinase, was reduced by approximately 50% in monocyte-derived macrophages of low HDL-C subjects, most significantly those with cholesterol efflux defects but without mutations in ABCA1 (p < 0.01). These results were verified by reverse transcription-PCR and replicated in a second cohort. We show here that blocking the activity or expression of CTSD, by pepstatin or CTSD small interfering RNA, respectively, reduced ABCA1 expression and protein abundance in both macrophages and CHO cells and apolipoprotein A-I-mediated lipid efflux by more than 70%. Conversely, expression of CTSD increased both ABCA1 mRNA expression and cellular ABCA1 protein. Consistent with its role in the proteolytic processing of prosaposin, inactivation of CTSD function resulted in the accumulation of glycosphingo-lipid and free cholesterol in late endosomes/lysosomes, a phenotype similar to NPC1 deficiency. Inhibition of CTSD also caused retention of ABCA1 in lysosomal compartments, reducing its trafficking to the plasma membrane. These studies demonstrate a novel and potentially important role for CTSD in intracellular cholesterol trafficking and ABCA1-mediated efflux. Therefore, decreased CTSD expression may contribute to low plasma HDL-C levels.
- Published
- 2006
- Full Text
- View/download PDF
33. Apoptotic cells induce a phosphatidylserine-dependent homeostatic response from phagocytes.
- Author
-
Kiss RS, Elliott MR, Ma Z, Marcel YL, and Ravichandran KS
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters metabolism, Animals, Blotting, Western, Cholesterol metabolism, Humans, Jurkat Cells, Macrophages, Mice, Polymerase Chain Reaction, Signal Transduction physiology, Apoptosis physiology, Homeostasis physiology, Phagocytes metabolism, Phagocytosis physiology, Phosphatidylserines metabolism
- Abstract
Engulfment of apoptotic cells by phagocytes is important throughout development and adult life. When phagocytes engulf apoptotic cells, they increase their cellular contents including cholesterol and phospholipids, but how the phagocytes respond to this increased load is poorly understood. Here, we identify one type of a phagocyte response, wherein the recognition of apoptotic cells triggers enhanced cholesterol efflux (to apolipoprotein A-I) from macrophages. Phosphatidylserine (PS) exposed on apoptotic cells was necessary and sufficient to stimulate the efflux response. A major mechanism for this enhanced efflux by macrophages was the upregulation of the mRNA and protein for ABCA1, a membrane transporter independently linked to cholesterol efflux as well as engulfment of apoptotic cells. This increase in phagocyte ABCA1 levels required the function of nuclear receptor LXRalpha/beta, a known regulator of cholesterol homeostasis in humans and mice. Taken together, these data reveal a "homeostatic program" initiated in phagocytes that include a proximal membrane signaling event initiated by PS recognition, a downstream signaling event acting through nuclear receptors, and an effector arm involving upregulation of ABCA1, in turn promoting reverse cholesterol transport from the phagocytes. These data also have implications for macrophage handling of contents derived from apoptotic versus necrotic cells in atherosclerotic lesions.
- Published
- 2006
- Full Text
- View/download PDF
34. The lipoprotein receptor-related protein-1 (LRP) adapter protein GULP mediates trafficking of the LRP ligand prosaposin, leading to sphingolipid and free cholesterol accumulation in late endosomes and impaired efflux.
- Author
-
Kiss RS, Ma Z, Nakada-Tsukui K, Brugnera E, Vassiliou G, McBride HM, Ravichandran KS, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters metabolism, Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing genetics, Animals, CHO Cells metabolism, Cricetinae, Immunoblotting, Ligands, Lysosomes metabolism, Mice, Protein Transport, Saposins antagonists & inhibitors, Saposins genetics, alpha-Macroglobulins metabolism, Adaptor Proteins, Signal Transducing metabolism, Cholesterol metabolism, Endosomes metabolism, Glycosphingolipids metabolism, Saposins metabolism
- Abstract
One of the conserved functional pathways linked to engulfment of apoptotic corpses involves two membrane proteins low density lipoprotein receptor-related protein-1 (LRP) and ABCA1 and the LRP adapter protein GULP. Because LRP and ABCA1 play roles in cellular lipid trafficking and efflux, here we addressed whether the third member, the LRP adapter protein GULP, also affects cellular lipid transport. Several lines of evidence show that overexpression of GULP causes glycosphingolipid and free cholesterol accumulation in the late endosome/lysosome compartment that is accompanied by down-regulation of ABCA1 and decreased efflux. Conversely, knockdown of endogenous GULP expression promoted cholesterol flux through the late endosomes and up-regulation of ABCA1, even in the context of a disease state such as Niemann-Pick Type C disease. Mechanistically, we were able to show that trafficking of the LRP ligands alpha2-macroglobulin and prosaposin, a protein cofactor necessary for glycosphingolipid degradation, are impaired in cells expressing full-length GULP protein, resulting in glycosphingolipid and free cholesterol accumulation in the late endosome/lysosome compartment. On the other hand, knockdown of endogenous GULP results in enhanced targeting of prosaposin and enhanced clearance of glycosphingolipids and cholesterol from the late endosomes. Taken together, these data reveal that GULP/LRP/ABCA1 represents a triad of molecules involved in engulfment and cellular lipid homeostasis.
- Published
- 2006
- Full Text
- View/download PDF
35. Replacement of helix 1' enhances the lipid binding activity of apoE3 N-terminal domain.
- Author
-
Redmond KA, Murphy C, Narayanaswami V, Kiss RS, Hauser P, Guigard E, Kay CM, and Ryan RO
- Subjects
- Apolipoprotein E3, Apolipoproteins E isolation & purification, Escherichia coli metabolism, Fluorescent Dyes chemistry, Gene Expression Regulation, Humans, Phosphatidylglycerols chemistry, Protein Binding physiology, Protein Conformation, Protein Structure, Secondary, Protein Structure, Tertiary physiology, Receptors, LDL chemistry, Receptors, LDL physiology, Solubility, Time Factors, Apolipoproteins E chemistry, Apolipoproteins E metabolism, Lipids chemistry, Lipids physiology
- Abstract
The N-terminal domain of human apolipoprotein E (apoE-NT) harbors residues critical for interaction with members of the low-density lipoprotein receptor (LDLR) family. Whereas lipid free apoE-NT adopts a stable four-helix bundle conformation, a lipid binding induced conformational adaptation is required for manifestation of LDLR binding ability. To investigate the structural basis for this conformational change, the short helix connecting helix 1 and 2 in the four-helix bundle was replaced by the sequence NPNG, introducing a beta-turn. Recombinant helix-to-turn (HT) variant apoE3-NT was produced in Escherichia coli, isolated and characterized. Stability studies revealed a denaturation transition midpoint of 1.9 m guanidine hydrochloride for HT apoE3-NT vs. 2.5 M for wild-type apoE3-NT. Wild-type and HT apoE3-NT form dimers in solution via an intermolecular disulfide bond. Native PAGE showed that reconstituted high-density lipoprotein prepared with HT apoE3-NT have a diameter in the range of 9 nm and possess binding activity for the LDLR on cultured human skin fibroblasts. In phospholipid vesicle solubilization assays, HT apoE3-NT was more effective than wild-type apoE3-NT at inducing a time dependent decrease in dimyristoylphosphatidylglycerol vesicle light scattering intensity. In lipoprotein binding assays, HT apoE3-NT protected human low-density lipoprotein from phospholipase C induced aggregation to a greater extent that wild-type apoE3-NT. The results indicate that a mutation at one end of the apoE3-NT four-helix bundle markedly enhances the lipid binding activity of this protein. In the context of lipoprotein associated full-length apoE, increased lipid binding affinity of the N-terminal domain may alter the balance between receptor-active and -inactive conformational states.
- Published
- 2006
- Full Text
- View/download PDF
36. Intracellular lipidation of newly synthesized apolipoprotein A-I in primary murine hepatocytes.
- Author
-
Maric J, Kiss RS, Franklin V, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters metabolism, Animals, Blotting, Western, Cell Membrane metabolism, Cells, Cultured, Cholesterol metabolism, Cholesterol, LDL chemistry, Choline chemistry, Chromatography, Gas, Chromatography, Thin Layer, Cycloheximide pharmacology, Endoplasmic Reticulum metabolism, Golgi Apparatus metabolism, Hepatocytes cytology, Immunoprecipitation, Iohexol pharmacology, Lipid Metabolism, Lipoproteins chemistry, Lipoproteins, HDL chemistry, Lipoproteins, LDL chemistry, Lipoproteins, LDL metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Phosphatidylcholines chemistry, Protein Synthesis Inhibitors pharmacology, Sphingomyelins chemistry, Subcellular Fractions metabolism, Time Factors, Apolipoprotein A-I chemistry, Hepatocytes metabolism, Lipids chemistry
- Abstract
Hepatocytes, which are the main site of apolipoprotein (apo)A-I and ATP-binding cassette transporter A1 (ABCA1) expression, are also the main source of circulating high density lipoprotein. Here we have characterized the intracellular lipidation of newly synthesized apoA-I, in primary hepatocytes cultured with [3H]choline to label choline-phospholipids, low density lipoprotein-[3H]cholesterol to label the cell surface, or [3H]mevalonate to label de novo synthesized cholesterol. Phospholipidation of apoA-I is significant and most evident in endoplasmic reticulum (ER) and medial Golgi, both in the lumen and on the membrane fractions of the ER and medial Golgi. In the presence of cycloheximide, endogenous apoA-I is substantially phospholipidated intracellularly but acquires some additional lipid after export out of the cell. In cells labeled with low density lipoprotein-[3H]cholesterol, intracellular cholesterol lipidation of apoA-I is entirely absent, but the secreted apoA-I rapidly accumulates cholesterol after secretion from the cell in the media. On the other hand, de novo synthesized cholesterol can lipidate apoA-I intracellularly. We also showed the interaction between apoA-I and ABCA1 in ER and Golgi fractions. In hepatocytes lacking ABCA1, lipidation by low density lipoprotein-cholesterol was significantly reduced at the plasma membrane, phospholipidation and lipidation by de novo synthesized sterols were both reduced in Golgi compartments, whereas ER lipidation remained mostly unchanged. Therefore, the early lipidation in ER is ABCA1 independent, but in contrast, the lipidation of apoA-I in Golgi and at the plasma membrane requires ABCA1. Thus, we demonstrated that apoA-I phospholipidation starts early in the ER and is partially dependent on ABCA1, with the bulk of lipidation by phospholipids and cholesterol occurring in the Golgi and at the plasma membrane, respectively. Finally, we showed that the previously reported association of newly synthesized apoA-I and apoB (Zheng, H., Kiss, R. S., Franklin, V., Wang, M. D., Haidar, B., and Marcel, Y. L. (2005) J. Biol. Chem. 280, 21612-21621) occurs after secretion at the cell surface.
- Published
- 2005
- Full Text
- View/download PDF
37. Lipid efflux in human and mouse macrophagic cells: evidence for differential regulation of phospholipid and cholesterol efflux.
- Author
-
Kiss RS, Maric J, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters physiology, Animals, Anisomycin pharmacology, Apolipoprotein A-I metabolism, Cell Line, Cell Line, Transformed, Cyclic AMP pharmacology, Cyclic AMP-Dependent Protein Kinases metabolism, GTP-Binding Protein alpha Subunits, Gi-Go physiology, Humans, Intercellular Signaling Peptides and Proteins, Mice, Peptides pharmacology, Pertussis Toxin pharmacology, Protein Kinase C metabolism, Wasp Venoms pharmacology, Cholesterol metabolism, Homeostasis, Lipid Metabolism, Macrophages metabolism, Phospholipids metabolism
- Abstract
ABCA1 is a critical regulator of lipid efflux from cells, which is highly regulated at the transcriptional and posttranslational levels. However, cells from different species and different tissues, and primary versus immortalized cells, show different modes of regulation. We have carried out a comparative analysis of basic signaling pathways of lipid efflux in mouse J774 cells, mouse peritoneal macrophages (MPMs), human THP-1 cells, and human monocyte-derived macrophages. Cyclic AMP (cAMP) was a potent stimulator of lipid efflux in mouse macrophages, but not in human macrophages. Moreover, this cAMP-inducible component of efflux from MPMs was inhibitable by H89 [a protein kinase A (PKA) inhibitor], but H89 did not affect basal efflux. On the other hand, cAMP failed to show any stimulatory effect in human macrophages, but basal efflux was inhibitable by H89. In MPMs and THP-1 cells, protein kinase C (PKC) inhibitors blocked cholesterol efflux but had no effect on phospholipid efflux, demonstrating the separation of the regulation of phospholipid efflux and cholesterol efflux in macrophages. We conclude that: 1) cAMP regulates lipid efflux predominantly in a PKA-dependent fashion; 2) cholesterol efflux is modulated by a PKC-dependent mechanism; and 3) mouse and human macrophages exhibit different modes of regulation of lipid efflux.
- Published
- 2005
- Full Text
- View/download PDF
38. ApoA-I lipidation in primary mouse hepatocytes. Separate controls for phospholipid and cholesterol transfers.
- Author
-
Zheng H, Kiss RS, Franklin V, Wang MD, Haidar B, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters genetics, Adenoviridae genetics, Animals, Brefeldin A pharmacology, Immunoprecipitation, Lipoproteins metabolism, Lipoproteins, HDL metabolism, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Monensin pharmacology, Phospholipids metabolism, Phosphorylation, Progesterone metabolism, Temperature, Time Factors, Transgenes, Apolipoprotein A-I metabolism, Cholesterol metabolism, Hepatocytes metabolism, Lipid Metabolism, Phospholipids chemistry
- Abstract
The liver is the major site of both apolipoprotein A-I (apoA-I) synthesis and ATP-binding cassette transporter A1 (ABCA1) expression. Here, we compare the lipidation with cholesterol and phospholipid of newly synthesized human apoA-I (hapoA-I) using adenoviral vector-mediated endogenous expression or exogenously added hapoA-I in wild type and ABCA1-null hepatocytes. Hepatocytes were labeled with [3H]cholesterol (delivered with LDL or methyl-beta-cyclodextrin), [3H]mevalonate, or [3H]choline. ABCA1 deficiency decreased apoA-I phospholipidation by 80%, but acquisition of de novo synthesized and exogenous cholesterol only decreased by 40-60%. The transfer of de novo synthesized cholesterol to apoA-I was decreased at all time points, but that of exogenously delivered cholesterol was independent of ABCA1 activity at the early time points. Progesterone does not affect apoA-I synthesis or its lipidation but inhibited the early phase of apoA-I cholesterol lipidation in both wild type and ABCA1-null hepatocytes. Fast protein liquid chromatography analysis of medium lipoproteins confirmed that with ABCA1 deficiency, the proportion of secreted high density lipoprotein-associated apoA-I and cholesterol decreased by about 50%. The very low density lipoprotein (VLDL)/LDL size fraction also contained a significant level of cholesterol in ABCA1 deficiency, consistent with the result of immunoprecipitations showing the presence of lipoproteins with both apoA-I and murine apoB. ApoA-I lipidation with newly synthesized cholesterol in ABCA1-null hepatocytes was significantly decreased by brefeldin A and monensin. In conclusion, we demonstrate that: (i) whereas most hepatic phospholipidation of apoA-I is mediated by ABCA1, acquisition of cholesterol depends on active transfer from intracellular compartments by ABCA1-dependent and -independent pathways, both sensitive to progesterone and (ii) there is separate regulation of phospholipid and cholesterol lipidation of apoA-I in hepatocytes.
- Published
- 2005
- Full Text
- View/download PDF
39. Multiple rare alleles contribute to low plasma levels of HDL cholesterol.
- Author
-
Cohen JC, Kiss RS, Pertsemlidis A, Marcel YL, McPherson R, and Hobbs HH
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters chemistry, ATP-Binding Cassette Transporters metabolism, Adult, Aged, Amino Acid Substitution, Apolipoprotein A-I chemistry, Apolipoprotein A-I metabolism, Black People genetics, Female, Haplotypes, Humans, Male, Middle Aged, Phosphatidylcholine-Sterol O-Acyltransferase chemistry, Phosphatidylcholine-Sterol O-Acyltransferase metabolism, Quantitative Trait, Heritable, White People genetics, ATP-Binding Cassette Transporters genetics, Alleles, Apolipoprotein A-I genetics, Cholesterol, HDL blood, Genetic Variation, Phosphatidylcholine-Sterol O-Acyltransferase genetics
- Abstract
Heritable variation in complex traits is generally considered to be conferred by common DNA sequence polymorphisms. We tested whether rare DNA sequence variants collectively contribute to variation in plasma levels of high density lipoprotein cholesterol (HDL-C). We sequenced three candidate genes (ABCA1, APOA1, and LCAT) that cause Mendelian forms of low HDL-C levels in individuals from a population-based study. Nonsynonymous sequence variants were significantly more common (16% versus 2%) in individuals with low HDL-C (
95th percentile). Similar findings were obtained in an independent population, and biochemical studies indicated that most sequence variants in the low HDL-C group were functionally important. Thus, rare alleles with major phenotypic effects contribute significantly to low plasma HDL-C levels in the general population. - Published
- 2004
- Full Text
- View/download PDF
40. Structure-guided protein engineering modulates helix bundle exchangeable apolipoprotein properties.
- Author
-
Kiss RS, Weers PM, Narayanaswami V, Cohen J, Kay CM, and Ryan RO
- Subjects
- Animals, Apolipoproteins E metabolism, Cattle, Cell Line, Circular Dichroism, Dimyristoylphosphatidylcholine chemistry, Dose-Response Relationship, Drug, Electrophoresis, Polyacrylamide Gel, Escherichia coli metabolism, Fibroblasts metabolism, Fluorescent Dyes pharmacology, Genetic Vectors, Heparin chemistry, Heparin metabolism, Humans, Immunoblotting, Lipid Bilayers, Lipid Metabolism, Manduca, Models, Molecular, Protein Binding, Protein Engineering, Receptors, LDL metabolism, Recombinant Fusion Proteins metabolism, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Skin cytology, Skin metabolism, Spectrometry, Fluorescence, Time Factors, Ultraviolet Rays, Urea pharmacology, Apolipoproteins chemistry
- Abstract
Apolipoprotein (apo) E plays a major role in lipid metabolism by mediating cellular uptake of lipoprotein particles through interaction with members of the low density lipoprotein (LDL) receptor family. The primary region of apoE responsible for receptor binding has been limited to a cluster of basic amino acids between residues 134 and 150, located in the fourth helix of the N-terminal domain globular helix bundle structure. To investigate structural and functional requirements of this "receptor binding region" we engineered an apolipoprotein chimera wherein residues 131-151 of human apoE were substituted for residues 146-166 (helix 5) of Manduca sexta apolipophorin III (apoLp-III). Recombinant hybrid apolipoprotein was expressed in Escherichia coli, isolated, and characterized. Hybrid apolipoprotein and apoE3-N-terminal, but not apoLp-III, bound to heparin-Sepharose. Far UV circular dichroism spectroscopy revealed the presence of predominantly alpha-helix secondary structure, and stability studies revealed a urea denaturation midpoint of 1.05 m, similar to wild-type apoLp-III. Hybrid apolipoprotein-induced dimyristoylphosphatidylcholine (DMPC) bilayer vesicle solubilization activity was significantly enhanced compared with either parent protein, consistent with detection of solvent-exposed hydrophobic regions on the protein in fluorescent dye binding experiments. Unlike wild-type apoLp-III.DMPC complexes, disc particles bearing the hybrid apolipoprotein competed with 125ILDL for binding to the LDL receptor on cultured human skin fibroblasts. We conclude that a hybrid apolipoprotein containing a key receptor recognition element of apoE preserves the structural integrity of the parent protein while conferring a new biological activity, illustrating the potential of helix swapping to introduce desirable biological properties into unrelated or engineered apolipoproteins.
- Published
- 2003
- Full Text
- View/download PDF
41. Structure-function relationships of apolipoprotein A-I: a flexible protein with dynamic lipid associations.
- Author
-
Marcel YL and Kiss RS
- Subjects
- Humans, Protein Conformation, Protein Structure, Tertiary, Structure-Activity Relationship, Apolipoprotein A-I chemistry, Apolipoprotein A-I metabolism, Lipid Metabolism
- Abstract
Purpose of Review: Apolipoprotein A-I is the major structural protein of HDL. Its physicochemical properties maintain a delicate balance between maintenance of stable lipoproteins and the ability to associate with and dissociate from the lipid transported. Here we review the progress made in the last 2-3 years on the structure-function relationships of apolipoprotein A-I, including elements related to the ATP binding cassette transporter A1., Recent Findings: Current evidence now supports the so-called 'belt' or 'hairpin' models for apolipoprotein A-I conformation when bound to discoidal lipoproteins. In-vivo expression of apolipoprotein A-I mutant proteins has shown that both the N- and C-terminal domains are important for lipid association as well as for the esterification reaction, particularly binding of cholesteryl esters and formation of mature alpha-migrating lipoproteins. This property is apparently quite distinct from the activation of the enzyme lecithin cholesterol acyl transferase, which requires interaction with the central helix 6. The interaction of apolipoprotein A-I with the ATP binding cassette transporter A1 has been shown to require the C-terminal domain, which is proposed to mediate the opening of the helix bundle formed by lipid-free or lipid-poor apolipoprotein A-I and allow its association with hydrophobic binding sites., Summary: Significant progress has been made in the understanding of the molecular mechanisms controlling the folding of apolipoprotein A-I and its interaction with lipids and various other protein factors involved in HDL metabolism.
- Published
- 2003
- Full Text
- View/download PDF
42. The lipidation by hepatocytes of human apolipoprotein A-I occurs by both ABCA1-dependent and -independent pathways.
- Author
-
Kiss RS, McManus DC, Franklin V, Tan WL, McKenzie A, Chimini G, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, Alitretinoin, Animals, Cholesterol metabolism, Chromatography, Liquid, Humans, Lipoproteins, HDL metabolism, Lipoproteins, LDL metabolism, Lipoproteins, VLDL metabolism, Mice, Mice, Inbred C57BL, Phospholipids metabolism, Receptors, Retinoic Acid physiology, Retinoid X Receptors, Transcription Factors physiology, Tretinoin pharmacology, ATP-Binding Cassette Transporters physiology, Apolipoprotein A-I metabolism, Hepatocytes metabolism, Lipid Metabolism
- Abstract
The pathways of hepatic intra- and peri-cellular lipidation of apolipoprotein A-I (apoA-I) were studied by infecting primary mouse hepatocytes from either apoA-I-deficient or ABCA1-deficient mice with a recombinant adenovirus expressing the human apoA-I (hapoA-I) cDNA (endo apoA-I) or incubating the hepatocytes with exogenously added hapoA-I (exo apoA-I) and examining the hapoA-I-containing lipoproteins formed. The cells, maintained in serum-free medium, were labeled with [(3)H]choline, and the cell medium was separated by fast protein liquid chromatography or immunoprecipitated to quantify labeled choline phospholipids specifically associated with hapoA-I. With the apoA-I-deficient hepatocytes, the high density lipoprotein fraction formed with endo apoA-I contained proportionally more phospholipids than that formed with exo apoA-I. However, the lipoprotein size and electrophoretic mobility and phospholipid profiles were similar for exo apoA-I and endo apoA-I. Taken together, these data demonstrate that a significant proportion of hapoA-I is secreted from hepatocytes in a phospholipidated state but that hapoA-I is also phospholipidated peri-cellularly. With primary hepatocytes from ABCA1-deficient mice, the expression and net secretion of adenoviral-generated endogenous apoA-I was unchanged compared with control mice, but (3)H-phospholipids associated with endo apoA-I and exo apoA-I decreased by 63 and 25%, respectively. The lipoprotein size and electrophoretic migration and their phospholipid profiles remained unchanged. In conclusion, we demonstrated that intracellular and peri-cellular lipidation of apoA-I represent distinct and additive pathways that may be regulated independently. Hepatocyte expression of ABCA1 is central to the lipidation of newly synthesized apoA-I but also contributes to the lipidation of exogenous apoA-I. However, a significant basal level of phospholipidation occurs in the absence of ABCA1.
- Published
- 2003
- Full Text
- View/download PDF
43. Trypsin-sensitive and lipid-containing sites of the macrophage extracellular matrix bind apolipoprotein A-I and participate in ABCA1-dependent cholesterol efflux.
- Author
-
Burgess JW, Kiss RS, Zheng H, Zachariah S, and Marcel YL
- Subjects
- ATP Binding Cassette Transporter 1, Animals, Apolipoprotein A-I chemistry, Binding Sites, Cell Line, Cells, Cultured, Fibroblasts metabolism, Fibronectins metabolism, Humans, Mice, Organ Specificity, Skin metabolism, Tetradecanoylphorbol Acetate pharmacology, ATP-Binding Cassette Transporters metabolism, Apolipoprotein A-I metabolism, Cholesterol metabolism, Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Laminin metabolism, Macrophages metabolism, Phospholipids metabolism, Trypsin metabolism
- Abstract
A unique property of the extracellular matrix of J774 and THP-1 cells has been identified, which contributes to the ability of these cells to promote cholesterol efflux. We demonstrate high level apolipoprotein (apo) A-I binding to macrophage cells (THP-1 and J774) and to their extracellular matrix (ECM). However, high level apoA-I binding is not observed on fibroblasts, HepG2 cells, or U937 cells (a macrophage cell line that does not efflux cholesterol to apoA-I or bind apoA-I on their respective ECM). Binding to the ECM of THP-1 or J774 macrophages depends on the presence of apoA-I C-terminal helices and is markedly reduced with a mutant lacking residues 187-243 (apoA-I Delta(187-243)), suggesting that the hydrophobic C terminus forms a hydrophobic interaction with the ECM. ApoA-I binding is lost upon trypsin treatment or with Triton X-100, a preparation method that de-lipidates the ECM. However, binding is recovered with re-lipidation, and is preserved with ECM prepared using cytochalasin B, which conserves the endogenous phospholipid levels of the ECM. We also demonstrate that specific cholesterol efflux to apoA-I is much reduced in cells released from their native ECM, but fully restored when ECM-depleted cells are added back to ECM in the presence of apoA-I. The apoA-I-mediated efflux is deficient in plated or suspension U937 macrophages, but is restored to high levels when the suspension U937 cells are reconstituted with the ECM of J774 cells. The ECM-dependent activity was much reduced in the presence of glyburide, indicating participation of ABCA1 (ATP-binding cassette transporter 1) in the efflux mechanism. These studies establish a novel binding site for apoA-I on the macrophage ECM that may function together with ABCA1 in promoting cholesterol efflux.
- Published
- 2002
- Full Text
- View/download PDF
44. Functional similarities of human and chicken apolipoprotein A-I: dependence on secondary and tertiary rather than primary structure.
- Author
-
Kiss RS, Ryan RO, and Francis GA
- Subjects
- Amino Acid Sequence, Animals, Apolipoprotein A-I chemistry, Cells, Cultured, Chickens, Cholesterol analysis, Cholesterol metabolism, Fibroblasts drug effects, Fibroblasts metabolism, Humans, Lipid Bilayers chemistry, Lipoproteins, HDL pharmacology, Lipoproteins, LDL metabolism, Molecular Sequence Data, Phospholipids chemistry, Sequence Alignment, Type C Phospholipases, Apolipoprotein A-I pharmacology, Protein Conformation
- Abstract
To investigate the sequence requirements for apolipoprotein (apo) AI functions, comparisons of human and chicken apoAI were performed. In lipid binding assays, chicken apoAI was capable of transforming phospholipid vesicles into discoidal bilayer structures, similar in both size and apolipoprotein content to those produced with human apoAI under the same conditions. Human and chicken apoAI were indistinguishable in their relative abilities to prevent phospholipase C-induced aggregation of human low density lipoprotein. This activity, which is dependent upon formation of a stable interaction with the modified lipoprotein, represents a sensitive measure of apolipoprotein association with spherical lipoprotein particles. The ability of chicken versus human apoAI to mobilize the regulatory pool of cholesterol available for esterification by acyl-CoA:cholesterol acyltransferase by human fibroblasts was also assessed. Lipid-free chicken and human apoAI were equivalent in their ability to deplete cholesterol from this pool, as were intact chicken high density lipoprotein (HDL) and human HDL(3). Based on the overall sequence identity of chicken and human apoAI (48%), and comparison of regions thought to be responsible for key apoAI functions, these data indicate that amphipathic alpha-helical structure, rather than specific amino acid sequence, is the major determinant of apoAI lipid binding and ability to mobilize the regulatory pool of cellular cholesterol.
- Published
- 2001
- Full Text
- View/download PDF
45. Uptake of lipoproteins for axonal growth of sympathetic neurons.
- Author
-
Posse De Chaves EI, Vance DE, Campenot RB, Kiss RS, and Vance JE
- Subjects
- Animals, Animals, Newborn, Anticholesteremic Agents pharmacology, Axons metabolism, Brain metabolism, CD36 Antigens biosynthesis, Cell Division, Cells, Cultured, Electrophoresis, Polyacrylamide Gel, Humans, Immunoblotting, LDL-Receptor Related Proteins, Lipoproteins, HDL pharmacokinetics, Lipoproteins, LDL pharmacokinetics, Liver metabolism, Low Density Lipoprotein Receptor-Related Protein-1, Microscopy, Fluorescence, Models, Biological, Neurons metabolism, Pravastatin pharmacology, Rats, Rats, Sprague-Dawley, Receptors, Immunologic biosynthesis, Receptors, LDL biosynthesis, Receptors, Lipoprotein biosynthesis, Receptors, Scavenger, Scavenger Receptors, Class B, Tissue Distribution, Axons physiology, Lipoproteins pharmacokinetics, Membrane Proteins, Membrane Transport Proteins, Neurons cytology, Sympathetic Nervous System metabolism
- Abstract
Lipoproteins originating from axon and myelin breakdown in injured peripheral nerves are believed to supply cholesterol to regenerating axons. We have used compartmented cultures of rat sympathetic neurons to investigate the utilization of lipids from lipoproteins for axon elongation. Lipids and proteins from human low density lipoproteins (LDL) and high density lipoproteins (HDL) were taken up by distal axons and transported to cell bodies, whereas cell bodies/proximal axons internalized these components from only LDL, not HDL. Consistent with these observations, the impairment of axonal growth, induced by inhibition of cholesterol synthesis, was reversed when LDL or HDL were added to distal axons or when LDL, but not HDL, were added to cell bodies. LDL receptors (LDLRs) and LR7/8B (apoER2) were present in cell bodies/proximal axons and distal axons, with LDLRs being more abundant in the former. Inhibition of cholesterol biosynthesis increased LDLR expression in cell bodies/proximal axons but not distal axons. LR11 (SorLA) was restricted to cell bodies/proximal axons and was undetectable in distal axons. Neither the LDL receptor-related protein nor the HDL receptor, SR-B1, was detected in sympathetic neurons. These studies demonstrate for the first time that lipids are taken up from lipoproteins by sympathetic neurons for use in axonal regeneration.
- Published
- 2000
- Full Text
- View/download PDF
46. Amphipathic alpha-helix bundle organization of lipid-free chicken apolipoprotein A-I.
- Author
-
Kiss RS, Kay CM, and Ryan RO
- Subjects
- Acrylamide chemistry, Anilino Naphthalenesulfonates chemistry, Animals, Cesium chemistry, Chickens, Chlorides chemistry, Circular Dichroism, Fatty Acids chemistry, Fluorescence Polarization, Fluorescent Dyes chemistry, Humans, Potassium Iodide chemistry, Protein Structure, Secondary, Spectrometry, Fluorescence, Spin Labels, Trifluoroethanol chemistry, Tryptophan chemistry, Apolipoprotein A-I chemistry, Lipids chemistry
- Abstract
Apolipoprotein A-I (apoA-I), the major protein component of plasma high-density lipoprotein (HDL), exists in alternate lipid-free and lipid-bound states. Among various species, chicken apoA-I possesses unique structural properties: it is a monomer in the lipid-free state and it is virtually the sole protein component of HDL. Near-UV circular dichroism (CD) spectroscopic studies provide evidence that chicken apoA-I undergoes a major conformational change upon binding to lipid, while far-UV CD data indicate its overall alpha-helix content is maintained during this transition. The fluorescence emission wavelength maximum (excitation 295 nm) of the tryptophans in apoA-I (W74 and W107) displayed a marked blue shift in both the lipid-free (331 nm) and HDL-bound (329 nm) states, compared to free tryptophan in solution. The effect of aqueous quenchers on tryptophan fluorescence was determined in lipid-free, dimyristoylphosphatidylcholine (DMPC)- and HDL-bound states. The most effective quencher in the lipid-free and HDL-bound states was acrylamide, giving rise to Ksv values of 1.6 +/- 0.1 and 1.2 +/- 0.1 M-1, respectively. Together, these data suggest that a hydrophobic environment around the two tryptophan residues (W74 and W107) is maintained in alternate conformations of the protein. To further probe the molecular organization of lipid-free apoA-I, its effect on the fluorescence properties of 8-anilino-1-naphthalenesulfonic acid (ANS) was determined. Human and chicken apoA-I induced a similar increase in ANS fluorescence quantum yield, in keeping with the hypothesis that these proteins adopt a similar global fold in the absence of lipid. When considered with near- and far-UV CD experiments, the data support a model in which lipid-free chicken apoA-I is organized as an amphipathic alpha-helix bundle. In other studies, lipid-soluble quenchers, 5-, 7-, 10-, and 12-DOXYL stearic acid (DSA), were employed to investigate the depth of penetration of apoA-I into the surface monolayer of spherical HDL particles. 5-DSA was the most effective quencher, suggesting that apoA-I tryptophan residues localize near the surface monolayer, providing a structural rationale for the reversibility of apoA-I-lipoprotein particle interactions.
- Published
- 1999
- Full Text
- View/download PDF
47. Human apolipoprotein E N-terminal domain displacement of apolipophorin III from insect low density lipophorin creates a receptor-competent hybrid lipoprotein.
- Author
-
Fisher CA, Kiss RS, Francis GA, Gao P, and Ryan RO
- Subjects
- Animals, Apolipoprotein A-I physiology, Binding, Competitive, Humans, Insect Proteins chemistry, Manduca chemistry, Protein Binding, Recombinant Fusion Proteins, Apolipoproteins physiology, Apolipoproteins E chemistry, Lipoproteins, LDL physiology
- Abstract
The surface of Manduca sexta low density lipophorin (LDLp) particles was employed as a template to examine the relative lipid binding affinity of the 22 kDa receptor binding domain (residues 1-183) of human apolipoprotein E3 (apo E3). Isolated LDLp was incubated with exogenous apolipoprotein and, following re-isolation by density gradient ultracentrifugation, particle apolipoprotein content was determined. Incubation of recombinant human apo E3(1-183) with LDLp resulted in a saturable displacement of apolipophorin III (apo Lp-III) from the particle surface, creating a hybrid apo E3(1-183)-LDLp. Although subsequent incubation with excess exogenous apo Lp-III failed to reverse the process, human apolipoprotein A-I (apo A-I) effectively displaced apo E3(1-183) from the particle surface. We conclude that human apo E N-terminal domain possesses a higher intrinsic lipid binding affinity than apo Lp-III but has a lower affinity than human apo A-I. The apo E3(1-183)-LDLp hybrid was competent to bind to the low density lipoprotein receptor on cultured fibroblasts. The system described is useful for characterizing the relative lipid binding affinities of wild type and mutant exchangeable apolipoproteins and evaluation of their biological properties when associated with the surface of a spherical lipoprotein.
- Published
- 1999
- Full Text
- View/download PDF
48. Bacterial expression and characterization of chicken apolipoprotein A-I.
- Author
-
Kiss RS, Kay CM, and Ryan RO
- Subjects
- Animals, Apolipoprotein A-I biosynthesis, Apolipoprotein A-I chemistry, Apolipoprotein A-I isolation & purification, Biological Assay, Chickens, Chromatography, High Pressure Liquid, Circular Dichroism, DNA, Bacterial chemistry, DNA, Bacterial genetics, Electrophoresis, Polyacrylamide Gel, Escherichia coli genetics, Escherichia coli metabolism, Genetic Vectors, Humans, Immunoblotting, Protein Sorting Signals chemistry, Protein Sorting Signals genetics, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins immunology, Recombinant Proteins isolation & purification, Spectrometry, Fluorescence, Apolipoprotein A-I genetics, Gene Expression Regulation, Bacterial genetics
- Abstract
Apolipoprotein (apo) A-I is a 28-kDa exchangeable apolipoprotein that plays a key role in lipoprotein metabolism. It is widely distributed among animal species and is rich in alpha-helical secondary structure. Unlike human apoA-I, which aggregates in the absence of lipid, chicken apoA-I is monomeric in the lipid-free state. To take advantage of this physical characteristic, a bacterial expression system for production of recombinant chicken apoA-I has been developed. The cDNA-encoding chicken apoA-I was cloned into the pET expression vector under the regulation of the lac operon and transformed into Escherichia coli. Recombinant apoA-I protein recovered from the soluble fraction of the bacterial cell pellet was purified to greater than 95% homogeneity by reversed-phase high-performance liquid chromatography. Although immunoblot analysis confirmed the identity of the overexpressed protein, its migration on denaturing polyacrylamide gel electrophoresis was slower than its natural counterpart. To determine if the vector-encoded 18 residue pelB N-terminal leader sequence was not cleaved by the bacterial leader peptidase, isolated recombinant chicken apoA-I was incubated with exogenous leader peptidase. This treatment resulted in an increased electrophoretic mobility, with migration to a position corresponding to plasma-derived chicken apoA-I. Electrospray mass spectrometry indicated a mass of 27,961 +/- 4 Da, in agreement with that predicted for natural chicken apoA-I. Far-UV circular dichroism spectroscopy indicated an alpha-helical content similar to apoA-I isolated from chicken plasma, suggesting that the protein is folded in solution. Fluorescence studies showed that the wavelength of maximum fluorescence emission of the two tryptophan residues in the protein was 331 nm, with no shift occurring following complexation with lipid. Recombinant apoA-I was shown to be functional in lipoprotein binding as well as to possess an ability to transform bilayer vesicles of dimyristoylphosphatidylcholine into discoidal complexes. This is the first report of bacterial expression of an avian apoA-I. Increased availability and the potential for site-directed mutagenesis of this protein will aid in further characterization of apoA-I and the mechanism whereby it functions in cholesterol transport., (Copyright 1998 Academic Press.)
- Published
- 1998
- Full Text
- View/download PDF
49. Physical properties of apolipoprotein A-I from the chicken, Gallus domesticus.
- Author
-
Kiss RS, Ryan RO, Hicks LD, Oikawa K, and Kay CM
- Subjects
- Animals, Apolipoprotein A-I isolation & purification, Apolipoprotein A-I metabolism, Carrier Proteins metabolism, Chemical Phenomena, Chemistry, Physical, Circular Dichroism, Humans, Lipid Metabolism, Models, Chemical, Protein Denaturation, Protein Structure, Secondary, Spectrometry, Fluorescence, Ultracentrifugation, Apolipoprotein A-I chemistry, Chickens blood, Lipoproteins
- Abstract
The amphipathic alpha-helices of exchangeable apolipoproteins (apo) function to simultaneously facilitate interaction with lipid surfaces and the aqueous environment. In contrast to mammalian apoA-I's, which self-associate in the absence of lipid, chicken apoA-I, which shares 66% sequence homology with human apoA-I, exists as a monomeric protein when dissociated from high-density lipoprotein (HDL). Sedimentation equilibrium studies conducted in the analytical ultracentrifuge yielded a weight-average molecular weight of 28,170. Corresponding sedimentation velocity and diffusion experiments gave rise to s0(20,w) = 2.23 S and D0(20,w) = 6.39 x 10(-7) cm2/s. A translational frictional ratio (f/fmin) of 1.18 and an axial ratio of 4.0 were also determined from this data. The Stokes radius (Rs,sed = 2.80 nm) and translational frictional ratio were subsequently used to calculate estimated molecular dimensions of 25.2 x 100.8 A for chicken apoA-I. Circular dichroism (CD) studies revealed a highly alpha-helical structure predicted to be 74% by Provencher-Glöckner analysis. Denaturation studies performed on lipid-free apoA-I and monitored by CD revealed a midpoint of denaturation of 0.64 M guanidine hydrochloride. From plots of delta G(app) versus guanidine hydrochloride concentration, a delta GDH2O of 1.86 kcal/mol was determined. In other studies, a midpoint of temperature-induced denaturation for apoA-I of 57 degrees C was obtained. The effect of solvent pH on the secondary structure content of apoA-I revealed a significant loss of alpha-helix below pH 4.0 and above pH 10, suggesting that lipid-free apoA-I may by partially stabilized by the formation of intra- or interhelix salt bridges between oppositely charged amino acid side chains.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.