1
|
Garliyev V, Lyssenko CA, Wiener JP, Praticò D, Lyssenko NN. Very low levels of ABCA7 in the cerebrum and Alzheimer's disease onset between the ages of 60 and 80 independently of APOE. J Neuropathol Exp Neurol 2024; 83:808-821. [PMID: 38900184 DOI: 10.1093/jnen/nlae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
This cross-sectional study addressed the ABCA7-Alzheimer's disease (AD) association. ABCA7 protein levels were quantified in 3 cerebral regions of brain donors with Braak neurofibrillary tangle (NFT) stages 0-V. Ordinal regression models were implemented to estimate the effect of ABCA7 on stopping in an earlier Braak NFT stage versus progressing to the later stages in 2 prespecified age segments. In the final model, high ABCA7 levels in the parietal cortex increased the odds of remaining cognitively healthy (ie, in stages 0/I) versus experiencing AD onset (ie, progressing to stages II-V) in the 61-80 age segment (OR = 2.87, adj 95% CI = 1.41-7.86, adj P = .007, n = 109), after controlling for APOE and other covariates. No ABCA7-AD association was found in the 81-98 age segment (n = 113). Parietal ABCA7 levels in 61-80-year-old with stages II-V were very low, even significantly lower than in 81-98-year-old with stages II-V. ABCA7 levels in the prefrontal cortex and hippocampus predicted AD onset in the 61-80 age segment after adjustment for APOE. ABCA7 levels were also the lowest in 61-80-year-old with frequent neuritic plaques. Thus, very low ABCA7 levels in the cerebrum are associated with AD onset in the 7th-8th decade of life.
Collapse
Affiliation(s)
- Viktor Garliyev
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Catherine A Lyssenko
- Office of Institutional Research & Analysis, University of Pennsylvania, Philadelphia, PA, United States
| | - Joel P Wiener
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Domenico Praticò
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Nicholas N Lyssenko
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Jayaraman S, Urdaneta A, Bullitt E, Fändrich M, Gursky O. Lipid clearance and amyloid formation by serum amyloid A: exploring the links between beneficial and pathologic actions of an enigmatic protein. J Lipid Res 2023; 64:100429. [PMID: 37604227 PMCID: PMC10509712 DOI: 10.1016/j.jlr.2023.100429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
Serum amyloid A (SAA) is named after a life-threatening disease, yet this small evolutionarily conserved protein must have played a vital role in host defense. Most circulating SAA binds plasma lipoproteins and modulates their metabolism. However, this hardly justifies the rapid and dramatic SAA upregulation in inflammation, which is concomitant with upregulation of secretory phospholipase A2 (sPLA2). We proposed that these proteins synergistically clear cell membrane debris from the sites of injury. The present study uses biochemical and biophysical approaches to further explore the beneficial function of SAA and its potential links to amyloid formation. We show that murine and human SAA1 are powerful detergents that solubilize diverse lipids, including mammalian biomembranes, converting them into lipoprotein-size nanoparticles. These nanoparticles provide ligands for cell receptors, such as scavenger receptor CD36 or heparin/heparan sulfate, act as substrates of sPLA2, and sequester toxic products of sPLA2. Together, these functions enable SAA to rapidly clear unprotected lipids. SAA can also adsorb, without remodeling, to lipoprotein-size nanoparticles such as exosomal liposomes, which are proxies for lipoproteins. SAA in complexes with zwitterionic phospholipids stabilizes α-helices, while SAA in complexes containing anionic lipids or micelle-forming sPLA2 products forms metastable β-sheet-rich species that readily aggregate to form amyloid. Consequently, the synergy between SAA and sPLA2 extends from the beneficial lipid clearance to the pathologic amyloid formation. Furthermore, we show that lipid composition alters SAA conformation and thereby can influence the metabolic fate of SAA-lipid complexes, including their proamyloidogenic and proatherogenic binding to heparan sulfate.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Pharmacology, Physiology & Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, USA.
| | - Angela Urdaneta
- Department of Pharmacology, Physiology & Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Esther Bullitt
- Department of Pharmacology, Physiology & Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Olga Gursky
- Department of Pharmacology, Physiology & Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, USA
| |
Collapse
|
3
|
Ralhan I, Chang J, Moulton MJ, Goodman LD, Lee NY, Plummer G, Pasolli HA, Matthies D, Bellen HJ, Ioannou MS. Autolysosomal exocytosis of lipids protect neurons from ferroptosis. J Cell Biol 2023; 222:e202207130. [PMID: 37036445 PMCID: PMC10098143 DOI: 10.1083/jcb.202207130] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/25/2022] [Accepted: 03/20/2023] [Indexed: 04/11/2023] Open
Abstract
During oxidative stress neurons release lipids that are internalized by glia. Defects in this coordinated process play an important role in several neurodegenerative diseases. Yet, the mechanisms of lipid release and its consequences on neuronal health are unclear. Here, we demonstrate that lipid-protein particle release by autolysosome exocytosis protects neurons from ferroptosis, a form of cell death driven by lipid peroxidation. We show that during oxidative stress, peroxidated lipids and iron are released from neurons by autolysosomal exocytosis which requires the exocytic machinery VAMP7 and syntaxin 4. We observe membrane-bound lipid-protein particles by TEM and demonstrate that these particles are released from neurons using cryoEM. Failure to release these lipid-protein particles causes lipid hydroperoxide and iron accumulation and sensitizes neurons to ferroptosis. Our results reveal how neurons protect themselves from peroxidated lipids. Given the number of brain pathologies that involve ferroptosis, defects in this pathway likely play a key role in the pathophysiology of neurodegenerative disease.
Collapse
Affiliation(s)
- Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
| | - Jinlan Chang
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Matthew J. Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Lindsey D. Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Nathanael Y.J. Lee
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
| | - Greg Plummer
- Faculty of Medicine & Dentistry Cell Imaging Core, University of Alberta, Edmonton, Canada
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Doreen Matthies
- Unit on Structural Biology, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Maria S. Ioannou
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
4
|
Picataggi A, Rodrigues A, Cromley DA, Wang H, Wiener JP, Garliyev V, Billheimer JT, Grabiner BC, Hurt JA, Chen AC, Han X, Rader DJ, Praticò D, Lyssenko NN. Specificity of ABCA7-mediated cell lipid efflux. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159157. [PMID: 35381375 PMCID: PMC9058236 DOI: 10.1016/j.bbalip.2022.159157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022]
Abstract
Adenosine triphosphate-binding cassette transporter subfamily A member 7 (ABCA7) performs incompletely understood biochemical functions that affect pathogenesis of Alzheimer's disease. ABCA7 is most similar in primary structure to ABCA1, the protein that mediates cell lipid efflux and formation of high-density lipoprotein (HDL). Lipid metabolic labeling/tracer efflux assays were employed to investigate lipid efflux in BHK-ABCA7(low expression), BHK-ABCA7(high expression) and BHK-ABCA1 cells. Shotgun lipid mass spectrometry was used to determine lipid composition of HDL synthesized by BHK-ABCA7 and BHK-ABCA1 cells. BHK-ABCA7(low) cells exhibited significant efflux only of choline-phospholipid and phosphatidylinositol. BHK-ABCA7(high) cells had significant cholesterol and choline-phospholipid efflux to apolipoprotein (apo) A-I, apo E, the 18A peptide, HDL, plasma and cerebrospinal fluid and significant efflux of sphingosine-lipid, serine-lipid (which is composed of phosphatidylserine and phosphatidylethanolamine in BHK cells) and phosphatidylinositol to apo A-I. In efflux assays to apo A-I, after adjustment to choline-phospholipid, ABCA7-mediated efflux removed ~4 times more serine-lipid and phosphatidylinositol than ABCA1-mediated efflux, while ABCA1-mediated efflux removed ~3 times more cholesterol than ABCA7-mediated efflux. Shotgun lipidomic analysis revealed that ABCA7-HDL had ~20 mol% less phosphatidylcholine and 3-5 times more serine-lipid and phosphatidylinositol than ABCA1-HDL, while ABCA1-HDL contained only ~6 mol% (or ~1.1 times) more cholesterol than ABCA7-HDL. The discrepancy between the tracer efflux assays and shotgun lipidomics with respect to cholesterol may be explained by an underestimate of ABCA7-mediated cholesterol efflux in the former approach. Overall, these results suggest that ABCA7 lacks specificity for phosphatidylcholine and releases significantly but not dramatically less cholesterol in comparison with ABCA1.
Collapse
Affiliation(s)
- Antonino Picataggi
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amrith Rodrigues
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Debra A Cromley
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hu Wang
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Joel P Wiener
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Viktor Garliyev
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jeffrey T Billheimer
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Nicholas N Lyssenko
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW Serum amyloid A (SAA) is a highly sensitive acute phase reactant that has been linked to a number of chronic inflammatory diseases. During a systemic inflammatory response, liver-derived SAA is primarily found on high-density lipoprotein (HDL). The purpose of this review is to discuss recent literature addressing the pathophysiological functions of SAA and the significance of its association with HDL. RECENT FINDINGS Studies in gene-targeted mice establish that SAA contributes to atherosclerosis and some metastatic cancers. Accumulating evidence indicates that the lipidation state of SAA profoundly affects its bioactivities, with lipid-poor, but not HDL-associated, SAA capable of inducing inflammatory responses in vitro and in vivo. Factors that modulate the equilibrium between lipid-free and HDL-associated SAA have been identified. HDL may serve to limit SAA's bioactivities in vivo. Understanding the factors leading to the release of systemic SAA from HDL may provide insights into chronic disease mechanisms.
Collapse
Affiliation(s)
- Nancy R Webb
- Department of Pharmacology and Nutritional Sciences, Saha Cardiovascular Research Center, and Barnstable Brown Diabetes Center, University of Kentucky, 553 Wethington Building, 900 South Limestone, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
6
|
The interaction between brain and liver regulates lipid metabolism in the TBI pathology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166078. [PMID: 33444711 DOI: 10.1016/j.bbadis.2021.166078] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/28/2020] [Accepted: 01/03/2021] [Indexed: 12/31/2022]
Abstract
To shed light on the impact of systemic physiology on the pathology of traumatic brain injury (TBI), we examine the effects of TBI (concussive injury) and dietary fructose on critical aspects of lipid homeostasis in the brain and liver of young-adult rats. Lipids are integral components of brain structure and function, and the liver has a role on the synthesis and metabolism of lipids. Fructose is mainly metabolized in the liver with potential implications for brain function. Lipidomic analysis accompanied by unbiased sparse partial least squares discriminant analysis (sPLS-DA) identified lysophosphatidylcholine (LPC) and cholesterol ester (CE) as the top lipid families impacted by TBI and fructose in the hippocampus, and only LPC (16:0) was associated with hippocampal-dependent memory performance. Fructose and TBI elevated liver pro-inflammatory markers, interleukin-1α (IL-1α), Interferon-γ (IFN-γ) that correlated with hippocampal-dependent memory dysfunction, and monocyte chemoattractant protein-1 (MCP-1) positively correlated with LPC levels in the hippocampus. The effects of fructose were more pronounced in the liver, in agreement with the role of liver on fructose metabolism and suggest that fructose could exacerbate liver inflammation caused by TBI. The overall results indicate that TBI and fructose interact to influence systemic and central inflammation by engaging liver lipids. The impact of TBI and fructose diet on the periphery provides a therapeutic target to counteract the TBI pathogenesis.
Collapse
|
7
|
ABCA7 links sterol metabolism to the host defense system: Molecular background for potential management measure of Alzheimer's disease. Gene 2020; 768:145316. [PMID: 33221536 DOI: 10.1016/j.gene.2020.145316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/20/2020] [Accepted: 11/13/2020] [Indexed: 01/10/2023]
Abstract
ATP-binding cassette transporter (ABC) A7 is a membrane protein that belongs to the large family of ABC transporters. It is 54% homologous in amino acid residue sequence to ABCA1 which mediates biogenesis of plasma high density lipoprotein (HDL) from cellular phospholipid and cholesterol with extracellular helical apolipoproteins such as apolipoprotein (apo) A-I. When transfected and expressed, ABCA7 also mediates generation of HDL-like particles but small and of less cholesterol content. However, endogenous ABCA7 is unlikely involved in HDL biogenesis and rather to regulate the host-defense system such as phagocytotic function of the cells. ABCA1 expression is regulated by cellular cholesterol levels, positively by the liver X receptor (LXR) in extrahepatic peripheral cells. However, it is modulated dually in the liver being relevant to transport of cholesterol for its catabolism; positively by LXR and negatively by sterol regulatory element binding protein (SREBP) or hepatic nuclear factor 4α (HNF4α). In contrast, ABCA7 expression was shown to be regulated negatively by the SREBP system so that decrease of cell cholesterol enhances ABCA7 function such as cellular phagocytotic reaction, suggesting that it links cholesterol metabolism to the host defense system. The interest is being build up in ABCA7 as its genomic diversity has been found related to a risk for late-onset Alzheimer's diseases. More recent findings indicate that ABCA7 is involved in metabolism of amyloid β peptide including its phagocytotic clearance. Accordingly, modulation of ABCA7 activity by manipulating cholesterol metabolism may open a new path for management of Alzheimer's disease.
Collapse
|
8
|
Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Curr Atheroscler Rep 2020; 22:69. [PMID: 32968930 PMCID: PMC7511256 DOI: 10.1007/s11883-020-00888-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review addresses normal and pathologic functions of serum amyloid A (SAA), an enigmatic biomarker of inflammation and protein precursor of AA amyloidosis, a life-threatening complication of chronic inflammation. SAA is a small, highly evolutionarily conserved acute-phase protein whose plasma levels increase up to one thousand-fold in inflammation, infection, or after trauma. The advantage of this dramatic but transient increase is unclear, and the complex role of SAA in immune response is intensely investigated. This review summarizes recent advances in our understanding of the structure-function relationship of this intrinsically disordered protein, outlines its newly emerging beneficial roles in lipid transport and inflammation control, and discusses factors that critically influence its misfolding in AA amyloidosis. RECENT FINDINGS High-resolution structures of lipid-free SAA in crystals and fibrils have been determined by x-ray crystallography and electron cryo-microscopy. Low-resolution structural studies of SAA-lipid complexes, together with biochemical, cell-based, animal model, genetic, and clinical studies, have provided surprising new insights into a wide range of SAA functions. An emerging vital role of SAA is lipid encapsulation to remove cell membrane debris from sites of injury. The structural basis for this role has been proposed. The lysosomal origin of AA amyloidosis has solidified, and its molecular and cellular mechanisms have emerged. Recent studies have revealed molecular underpinnings for understanding complex functions of this Cambrian protein in lipid transport, immune response, and amyloid formation. These findings help guide the search for much-needed targeted therapies to block the protein deposition in AA amyloidosis.
Collapse
|
9
|
Ji A, Wang X, Noffsinger VP, Jennings D, de Beer MC, de Beer FC, Tannock LR, Webb NR. Serum amyloid A is not incorporated into HDL during HDL biogenesis. J Lipid Res 2020; 61:328-337. [PMID: 31915139 DOI: 10.1194/jlr.ra119000329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/06/2020] [Indexed: 11/20/2022] Open
Abstract
Liver-derived serum amyloid A (SAA) is present in plasma where it is mainly associated with HDL and from which it is cleared more rapidly than are the other major HDL-associated apolipoproteins. Although evidence suggests that lipid-free and HDL-associated forms of SAA have different activities, the pathways by which SAA associates and disassociates with HDL are poorly understood. In this study, we investigated SAA lipidation by hepatocytes and how this lipidation relates to the formation of nascent HDL particles. We also examined hepatocyte-mediated clearance of lipid-free and HDL-associated SAA. We prepared hepatocytes from mice injected with lipopolysaccharide or an SAA-expressing adenoviral vector. Alternatively, we incubated primary hepatocytes from SAA-deficient mice with purified SAA. We analyzed conditioned media to determine the lipidation status of endogenously produced and exogenously added SAA. Examining the migration of lipidated species, we found that SAA is lipidated and forms nascent particles that are distinct from apoA-I-containing particles and that apoA-I lipidation is unaltered when SAA is overexpressed or added to the cells, indicating that SAA is not incorporated into apoA-I-containing HDL during HDL biogenesis. Like apoA-I formation, generation of SAA-containing particles was dependent on ABCA1, but not on scavenger receptor class B type I. Hepatocytes degraded significantly more SAA than apoA-I. Taken together, our results indicate that SAA's lipidation and metabolism by the liver is independent of apoA-I and that SAA is not incorporated into HDL during HDL biogenesis.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Xuebing Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | | | - Drew Jennings
- Departments of Agricultural and Medical Biotechnology, University of Kentucky, Lexington, KY
| | - Maria C de Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Physiology, University of Kentucky, Lexington, KY
| | - Frederick C de Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Internal Medicine, University of Kentucky, Lexington, KY
| | - Lisa R Tannock
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Internal Medicine, University of Kentucky, Lexington, KY
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY .,Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| |
Collapse
|
10
|
Choi S, Park YE, Cheon EJ, Kim KY, Kim M, Ann SJ, Noh HM, Lee J, Lee CJ, Lee ST, Lee C, Lee JE, Lee SH. Novel Associations between Related Proteins and Cellular Effects of High-Density Lipoprotein. Korean Circ J 2019; 50:236-247. [PMID: 31845554 PMCID: PMC7043958 DOI: 10.4070/kcj.2019.0195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Recent studies have examined the structure-function relationship of high-density lipoprotein (HDL). This study aimed to identify and rank HDL-associated proteins involved in several biological function of HDL. METHODS HDLs isolated from 48 participants were analyzed. Cholesterol efflux capacity, effect of HDL on nitric oxide production, and vascular cell adhesion molecule-1 expression were assessed. The relative abundance of identified proteins in the highest vs. lowest quartile was expressed using the normalized spectral abundance factor ratio. RESULTS After adjustment by multiple testing, six proteins, thyroxine-binding globulin, alpha-1B-glycoprotein, plasma serine protease inhibitor, vitronectin, angiotensinogen, and serum amyloid A-4, were more abundant (relative abundance ratio ≥2) in HDLs with the highest cholesterol efflux capacity. In contrast, three proteins, complement C4-A, alpha-2-macroglobulin, and immunoglobulin mu chain C region, were less abundant (relative abundance ratio <0.5). In terms of nitric oxide production and vascular cell adhesion molecule-1 expression, no proteins showed abundance ratios ≥2 or <0.5 after adjustment. Proteins correlated with the functional parameters of HDL belonged to diverse biological categories. CONCLUSIONS In summary, this study ranked proteins showing higher or lower abundance in HDLs with high functional capacities and newly identified multiple proteins linked to cholesterol efflux capacity.
Collapse
Affiliation(s)
- Seungbum Choi
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yae Eun Park
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Eun Jeong Cheon
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kyeong Yeon Kim
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,Department of Chemistry, Sookmyung Women's University, Seoul, Korea.,Proteometech Inc., Seoul, Korea
| | - Miso Kim
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jin Ann
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Min Noh
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeho Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Joo Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Cheolju Lee
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Korea
| | - Ji Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.
| | - Sang Hak Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
11
|
Milanović Z, Vekić J, Radonjić V, Ilić Božović A, Zeljković A, Janac J, Spasojević-Kalimanovska V, Buch J, Chandrashekar R, Bojić-Trbojević Ž, Hajduković L, Christopher MM, Kovačević Filipović M. Association of acute Babesia canis infection and serum lipid, lipoprotein, and apoprotein concentrations in dogs. J Vet Intern Med 2019; 33:1686-1694. [PMID: 31175698 PMCID: PMC6639482 DOI: 10.1111/jvim.15537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 05/15/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Babesia canis infection induces a marked acute phase response (APR) that might be associated with alteration in lipid and lipoprotein metabolism and disease prognosis. HYPOTHESIS Dogs with B. canis-induced APR develop dyslipidemia with altered lipoprotein concentration and morphology. ANIMALS Twenty-nine client-owned dogs with acute B. canis infection and 10 clinically healthy control dogs. METHODS Observational cross-sectional study. Serum amyloid A (SAA) was measured using ELISA. Cholesterol, phospholipids, and triglycerides were determined biochemically. Lipoproteins were separated using agarose gel electrophoresis. Lipoprotein diameter was assessed by polyacrylamide gradient gel electrophoresis; correlation with ApoA-1 (radioimmunoassay) and SAA was determined. RESULTS Dogs with B. canis infection had a marked APR (median SAA, 168.3 μg/mL; range, 98.1-716.2 μg/mL) compared with controls (3.2 μg/mL, 2.0-4.2 μg/mL) (P < .001). Dogs with B. canis infection had significantly lower median cholesterol (4.79 mmol/L, 1.89-7.64 mmol/L versus 6.15 mmol/L, 4.2-7.4 mmol/L) (P = .02), phospholipid (4.64 mmol/L, 2.6-6.6 mmol/L versus 5.72 mmol/L, 4.68-7.0 mmol/L) (P = .02), and α-lipoproteins (77.5%, 27.7%-93.5% versus 89.2%, 75.1%-93.5%) (P = .04), and higher ApoA-1 (1.36 U, 0.8-2.56 U versus 0.95 U, 0.73-1.54 U) concentrations (P = .02). Serum amyloid A correlated with high-density lipoproteins (HDLs) diameter (rho = .43; P = .03) and ApoA-1 (rho = .63, P < .001). CONCLUSIONS AND CLINICAL IMPORTANCE Major changes associated with B. canis-induced APR in dogs are related to concentration, composition, and morphology of HDL particles pointing to an altered reverse cholesterol transport. Parallel ApoA-1 and SAA concentration increase is a unique still unexplained pathophysiological finding.
Collapse
Affiliation(s)
- Zorana Milanović
- Faculty of Veterinary Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Vekić
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | - Anja Ilić Božović
- Faculty of Veterinary Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Jelena Janac
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | | | | | - Žanka Bojić-Trbojević
- INEP - Institute for the Application of Nuclear Energy, University of Belgrade, Zemun, Serbia
| | - Ljiljana Hajduković
- INEP - Institute for the Application of Nuclear Energy, University of Belgrade, Zemun, Serbia
| | - Mary M Christopher
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California
| | | |
Collapse
|
12
|
Abstract
Serum amyloid A (SAA) proteins were isolated and named over 50 years ago. They are small (104 amino acids) and have a striking relationship to the acute phase response with serum levels rising as much as 1000-fold in 24 hours. SAA proteins are encoded in a family of closely-related genes and have been remarkably conserved throughout vertebrate evolution. Amino-terminal fragments of SAA can form highly organized, insoluble fibrils that accumulate in “secondary” amyloid disease. Despite their evolutionary preservation and dynamic synthesis pattern SAA proteins have lacked well-defined physiologic roles. However, considering an array of many, often unrelated, reports now permits a more coordinated perspective. Protein studies have elucidated basic SAA structure and fibril formation. Appreciating SAA’s lipophilicity helps relate it to lipid transport and metabolism as well as atherosclerosis. SAA’s function as a cytokine-like protein has become recognized in cell-cell communication as well as feedback in inflammatory, immunologic, neoplastic and protective pathways. SAA likely has a critical role in control and possibly propagation of the primordial acute phase response. Appreciating the many cellular and molecular interactions for SAA suggests possibilities for improved understanding of pathophysiology as well as treatment and disease prevention.
Collapse
Affiliation(s)
- George H Sack
- Departments of Biological Chemistry and Medicine, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Physiology 615, Baltimore, MD, 21205, USA.
| |
Collapse
|
13
|
Frame NM, Jayaraman S, Gantz DL, Gursky O. Serum amyloid A self-assembles with phospholipids to form stable protein-rich nanoparticles with a distinct structure: A hypothetical function of SAA as a "molecular mop" in immune response. J Struct Biol 2017. [PMID: 28645735 DOI: 10.1016/j.jsb.2017.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Serum amyloid A (SAA) is an acute-phase protein whose action in innate immunity and lipid homeostasis is unclear. Most circulating SAA binds plasma high-density lipoproteins (HDL) and reroutes lipid transport. In vivo SAA binds existing lipoproteins or generates them de novo upon lipid uptake from cells. We explored the products of SAA-lipid interactions and lipoprotein remodeling in vitro. SAA complexes with palmitoyl-oleoyl phosphocholine (POPC) were analyzed for structure and stability using circular dichroism and fluorescence spectroscopy, electron microscopy, gel electrophoresis and gel filtration. The results revealed the formation of 8-11nm lipoproteins that were∼50% α-helical and stable at near-physiological conditions but were irreversibly remodeled at Tm∼52°C. Similar HDL-size nanoparticles formed spontaneously at ambient conditions or upon thermal remodeling of parent lipoproteins containing various amounts of proteins and lipids, including POPC and cholesterol. Therefore, such HDL-size particles formed stable kinetically accessible structures in a wide range of conditions. Based on their size and stoichiometry, each particle contained about 12 SAA and 72 POPC molecules, with a protein:lipid weight ratio circa 2.5:1, suggesting a structure distinct from HDL. High stability of these nanoparticles and their HDL-like size suggest that similar lipoproteins may form in vivo during inflammation or injury when SAA concentration is high and membranes from dead cells require rapid removal. We speculate that solubilization of membranes by SAA to generate lipoproteins in a spontaneous energy-independent process constitutes the primordial function of this ancient protein, providing the first line of defense in clearing cell debris from the injured sites.
Collapse
Affiliation(s)
- Nicholas M Frame
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Donald L Gantz
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| |
Collapse
|
14
|
Karaca I, Wagner H, Ramirez A. Suche nach Risikogenen bei der Alzheimer-Erkrankung. DER NERVENARZT 2017; 88:744-750. [DOI: 10.1007/s00115-017-0354-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Sun L, Ye RD. Serum amyloid A1: Structure, function and gene polymorphism. Gene 2016; 583:48-57. [PMID: 26945629 DOI: 10.1016/j.gene.2016.02.044] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023]
Abstract
Inducible expression of serum amyloid A (SAA) is a hallmark of the acute-phase response, which is a conserved reaction of vertebrates to environmental challenges such as tissue injury, infection and surgery. Human SAA1 is encoded by one of the four SAA genes and is the best-characterized SAA protein. Initially known as a major precursor of amyloid A (AA), SAA1 has been found to play an important role in lipid metabolism and contributes to bacterial clearance, the regulation of inflammation and tumor pathogenesis. SAA1 has five polymorphic coding alleles (SAA1.1-SAA1.5) that encode distinct proteins with minor amino acid substitutions. Single nucleotide polymorphism (SNP) has been identified in both the coding and non-coding regions of human SAA1. Despite high levels of sequence homology among these variants, SAA1 polymorphisms have been reported as risk factors of cardiovascular diseases and several types of cancer. A recently solved crystal structure of SAA1.1 reveals a hexameric bundle with each of the SAA1 subunits assuming a 4-helix structure stabilized by the C-terminal tail. Analysis of the native SAA1.1 structure has led to the identification of a competing site for high-density lipoprotein (HDL) and heparin, thus providing the structural basis for a role of heparin and heparan sulfate in the conversion of SAA1 to AA. In this brief review, we compares human SAA1 with other forms of human and mouse SAAs, and discuss how structural and genetic studies of SAA1 have advanced our understanding of the physiological functions of the SAA proteins.
Collapse
Affiliation(s)
- Lei Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Richard D Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, SAR, China.
| |
Collapse
|
16
|
Abdu-Allah AM, Tarhouny SAE, Baghdadi HH. Serum amyloid a gene polymorphism and its association with lipid profile in Saudi females with osteoporosis. Pak J Med Sci 2015; 31:1124-9. [PMID: 26648999 PMCID: PMC4641268 DOI: 10.12669/pjms.315.7981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background and Objective: Osteoporosis can be defined as a systemic skeletal disease characterized by low bone mass and micro architectural decline of bone tissue. Serum amyloid A (SAA) is a family of protein that increases up to 1,000-fold in blood during inflammation. In this study, we aimed to study the relationship between SAA1 gene polymorphism (rs12218) and lipid profile and osteoporosis. Methods: The study was performed on the female students of Taibah University in Al Medina, KSA during June 2014 to April 2015. According to BMD; osteoporosis group (138 students) and control group (128 students). All groups were subjected to; BMI, BMD, calcium, phosphorus, creatinine, lipid profile and SAA. Polymerase chain reaction and Real Time were done to determine the distribution of allele and genotype frequency of SAA (rs12218) C/T polymorphism. Results: This study shows that the TT genotype of rs12218 was more frequent in osteoporosis group than control group (P<0.001). Also, TT genotype and T allel was found to be associated with plasma total cholesterol, TG, LDLc, HDLc, Tscore, Zscore and SAA1 level in osteoporosis group (P=0.000, P=0.05, and P=0.000, P=0.000, P=0.01, P=0.02, P=0.000 respectively). The logistic regression analysis with and without lipid disorders in the osteoporosis group also show that the TT genotype of rs12218 still differed significantly between these two groups (P=0.001, OR=1.814, 95% CI: 0.719-4.577). Conclusion: The results of this study shows a significant association between TT genotype of rs12218 and both lipid level and osteoporosis in Saudi female population.
Collapse
Affiliation(s)
- Azza M Abdu-Allah
- Azza M. Abdu-Allah MD. Menofyia University, Egypt. Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Taibah University, Saudi Arabia
| | - Shereen A El Tarhouny
- Shereen A. El Tarhouny MD. Zagazig University, Egypt. Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Taibah University, Saudi Arabia
| | - Hussam Hussein Baghdadi
- Hussam Hussein Baghdadi, PhD. Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Taibah University, Saudi Arabia
| |
Collapse
|
17
|
Rached F, Lhomme M, Camont L, Gomes F, Dauteuille C, Robillard P, Santos RD, Lesnik P, Serrano CV, Chapman MJ, Kontush A. Defective functionality of small, dense HDL3 subpopulations in ST segment elevation myocardial infarction: Relevance of enrichment in lysophosphatidylcholine, phosphatidic acid and serum amyloid A. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1254-61. [PMID: 26037829 DOI: 10.1016/j.bbalip.2015.05.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 05/13/2015] [Accepted: 05/27/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Low plasma levels of high-density lipoprotein-cholesterol (HDL-C) are typical of acute myocardial infarction (MI) and predict risk of recurrent cardiovascular events. The potential relationships between modifications in the molecular composition and the functionality of HDL subpopulations in acute MI however remain indeterminate. METHODS AND RESULTS ST segment elevation MI (STEMI) patients were recruited within 24h after diagnosis (n=16) and featured low HDL-C (-31%, p<0.05) and acute-phase inflammation (determined as marked elevations in C-reactive protein, serum amyloid A (SAA) and interleukin-6) as compared to age- and sex-matched controls (n=10). STEMI plasma HDL and its subpopulations (HDL2b, 2a, 3a, 3b, 3c) displayed attenuated cholesterol efflux capacity from THP-1 cells (up to -32%, p<0.01, on a unit phospholipid mass basis) vs. CONTROLS Plasma HDL and small, dense HDL3b and 3c subpopulations from STEMI patients exhibited reduced anti-oxidative activity (up to -68%, p<0.05, on a unit HDL mass basis). HDL subpopulations in STEMI were enriched in two proinflammatory bioactive lipids, lysophosphatidylcholine (up to 3.0-fold, p<0.05) and phosphatidic acid (up to 8.4-fold, p<0.05), depleted in apolipoprotein A-I (up to -23%, p<0.05) and enriched in SAA (up to +10.2-fold, p<0.05); such changes were most marked in the HDL3b subfraction. In vitro HDL enrichment in both lysophosphatidylcholine and phosphatidic acid exerted deleterious effects on HDL functionality. CONCLUSIONS In the early phase of STEMI, HDL particle subpopulations display marked, concomitant alterations in both lipidome and proteome which are implicated in impaired HDL functionality. Such modifications may act synergistically to confer novel deleterious biological activities to STEMI HDL. SIGNIFICANCE Our present data highlight complex changes in the molecular composition and functionality of HDL particle subpopulations in the acute phase of STEMI, and for the first time, reveal that concomitant modifications in both the lipidome and proteome contribute to functional deficiencies in cholesterol efflux and antioxidative activities of HDL particles. These findings may provide new biomarkers and new insights in therapeutic strategy to reduce cardiovascular risk in this clinical setting where such net deficiency in HDL function, multiplied by low circulating HDL concentrations, can be expected to contribute to accelerated atherogenesis.
Collapse
Affiliation(s)
- Fabiana Rached
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France; Heart Institute-InCor, University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Marie Lhomme
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Laurent Camont
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Fernando Gomes
- Heart Institute-InCor, University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Carolane Dauteuille
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Paul Robillard
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Raul D Santos
- Heart Institute-InCor, University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Philippe Lesnik
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Carlos V Serrano
- Heart Institute-InCor, University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - M John Chapman
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France
| | - Anatol Kontush
- National Institute of Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6; Pitié-Salpétrière University Hospital, ICAN, Paris, France.
| |
Collapse
|
18
|
Jayaraman S, Haupt C, Gursky O. Thermal transitions in serum amyloid A in solution and on the lipid: implications for structure and stability of acute-phase HDL. J Lipid Res 2015; 56:1531-42. [PMID: 26022803 DOI: 10.1194/jlr.m059162] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Indexed: 12/20/2022] Open
Abstract
Serum amyloid A (SAA) is an acute-phase protein that circulates mainly on plasma HDL. SAA interactions with its functional ligands and its pathogenic deposition in reactive amyloidosis depend, in part, on the structural disorder of this protein and its propensity to oligomerize. In vivo, SAA can displace a substantial fraction of the major HDL protein, apoA-I, and thereby influence the structural remodeling and functions of acute-phase HDL in ways that are incompletely understood. We use murine SAA1.1 to report the first structural stability study of human plasma HDL that has been enriched with SAA. Calorimetric and spectroscopic analyses of these and other SAA-lipid systems reveal two surprising findings. First, progressive displacement of the exchangeable fraction of apoA-I by SAA has little effect on the structural stability of HDL and its fusion and release of core lipids. Consequently, the major determinant for HDL stability is the nonexchangeable apoA-I. A structural model explaining this observation is proposed, which is consistent with functional studies in acute-phase HDL. Second, we report an α-helix folding/unfolding transition in SAA in the presence of lipid at near-physiological temperatures. This new transition may have potentially important implications for normal functions of SAA and its pathogenic misfolding.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston MA 02118
| | - Christian Haupt
- Institute for Pharmaceutical Biotechnology, University of Ulm, 89081, Ulm, Germany
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston MA 02118
| |
Collapse
|
19
|
Hotta N, Abe-Dohmae S, Taguchi R, Yokoyama S. Preferential incorporation of shorter and less unsaturated acyl phospholipids into high density lipoprotein-like particles in the ABCA1- and ABCA7-mediated biogenesis with apoA-I. Chem Phys Lipids 2015; 187:1-9. [PMID: 25665932 DOI: 10.1016/j.chemphyslip.2015.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/24/2015] [Accepted: 01/27/2015] [Indexed: 11/27/2022]
Abstract
Molecular species of phosphatidylcholine (PC) and sphingomyelin (SPM) were globally analyzed for lipidomics in the nascent high-density lipoprotein (HDL)-like particles generated with human apolipoprotein A-I (apoA-I) form HEK293 cells where either human ATP binding cassette transporter (ABC) A1 or ABCA7 was transfected and overexpressed. SPM/PC ratio was higher in the ABCA1-mediated HDL than ABCA7-mediated HDL likely being related to their cholesterol content, while it was less than the ratio in the cell membrane in either case. Molecular species composition of hydrocarbon chain moiety in each phospholipid in the HDL largely reflected that in the cells the lipoprotein originated in, without remarkable difference between ABCA1 and ABCA7. Further analysis, however, revealed apparent preference for the molecules with shorter hydrocarbon chain length for both PC and SPM in their relative incorporation into HDL by ABCA1 and ABCA7. Likewise, it was in favor for less-unsaturated hydrocarbon chains of PC while this preference was not apparent for SPM. The results are consistent with the view that assembly of HDL particles with extracellular apoA-I is primarily with the cellular phospholipid molecules being regulated in part by their physicochemical nature.
Collapse
Affiliation(s)
- Noriko Hotta
- Graduate Schools of Pharmaceutical Sciences, Nagoya, Japan; Medical Sciences, Nagoya City University, Nagoya, Japan
| | | | - Ryo Taguchi
- Graduate Schools of Pharmaceutical Sciences, Nagoya, Japan; Nutritional Health Science Research Center, Chubu University, Kasugai, Japan
| | - Shinji Yokoyama
- Nutritional Health Science Research Center, Chubu University, Kasugai, Japan.
| |
Collapse
|
20
|
Gbelcová H, Svéda M, Laubertová L, Varga I, Vítek L, Kolář M, Strnad H, Zelenka J, Böhmer D, Ruml T. The effect of simvastatin on lipid droplets accumulation in human embryonic kidney cells and pancreatic cancer cells. Lipids Health Dis 2013; 12:126. [PMID: 23961716 PMCID: PMC3765626 DOI: 10.1186/1476-511x-12-126] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 08/11/2013] [Indexed: 01/09/2023] Open
Abstract
Background Statins (HMG-CoA reductase inhibitors) represent a major class of compounds for the treatment of hypercholesterolemia due to their ability to inhibit de novo cholesterol synthesis. In addition to their hypolipidemic effects, chemoprotective properties have been attributed to statins as well. These effects involve multiple mechanisms, which, however, are not known in detail. The aim of our study was to assess in non-malignant as well as cancer cells the impact of simvastatin on the amount of cytosolic lipid droplets (LDs) implicated in many biological processes including proliferation, inflammation, carcinogenesis, apoptosis, necrosis or growth arrest. Methods Human embryonic kidney cells HEK-293T and human pancreatic cancer cells MiaPaCa-2 were treated with simvastatin (6 and 12 μM) for 24 and 48 hours respectively. Neutral lipid probe Nile Red was used for detection of LDs by fluorescence microscopy. Cellular cholesterol content was determined by HPLC. Changes in expression of genes related to lipid metabolism in simvastatin-treated MiaPaCa-2 cells were examined by DNA microarray analysis. Validation of gene expression changes was performed using quantitative RT-PCR. Results The treatment of the cells with simvastatin increased their intracellular content of LDs in both non-malignant as well as cancer cells, partially due to the uptake of cholesterol and triacylglyceroles from medium; but in particular, due to enhanced synthesis of triacylglyceroles as proved by significant overexpression of genes related to de novo synthesis of triacylglyceroles and phospholipids. In addition, simvastatin also markedly influenced expression of genes directly affecting cell proliferation and signaling. Conclusions Simvastatin treatment led to accumulation of cytosolic LDs within the examined cells, a phenomenon which might contribute to the antiproliferative effects of statins.
Collapse
|
21
|
The Impairment of Macrophage-to-Feces Reverse Cholesterol Transport during Inflammation Does Not Depend on Serum Amyloid A. J Lipids 2013; 2013:283486. [PMID: 23431457 PMCID: PMC3572687 DOI: 10.1155/2013/283486] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 01/19/2023] Open
Abstract
Studies suggest that inflammation impairs reverse cholesterol transport (RCT). We investigated whether serum amyloid A (SAA) contributes to this impairment using an established macrophage-to-feces RCT model. Wild-type (WT) mice and mice deficient in SAA1.1 and SAA2.1 (SAAKO) were injected intraperitoneally with 3H-cholesterol-labeled J774 macrophages 4 hr after administration of LPS or buffered saline. 3H-cholesterol in plasma 4 hr after macrophage injection was significantly reduced in both WT and SAAKO mice injected with LPS, but this was not associated with a reduced capacity of serum from LPS-injected mice to promote macrophage cholesterol efflux in vitro. Hepatic accumulation of 3H-cholesterol was unaltered in either WT or SAAKO mice by LPS treatment. Radioactivity present in bile and feces of LPS-injected WT mice 24 hr after macrophage injection was reduced by 36% (P < 0.05) and 80% (P < 0.001), respectively. In contrast, in SAAKO mice, LPS did not significantly reduce macrophage-derived 3H-cholesterol in bile, and fecal excretion was reduced by only 45% (P < 0.05). Injection of cholesterol-loaded allogeneic J774 cells, but not syngeneic bone-marrow-derived macrophages, transiently induced SAA in C57BL/6 mice. Our study confirms reports that acute inflammation impairs steps in the RCT pathway and establishes that SAA plays only a minor role in this impairment.
Collapse
|
22
|
Berrougui H, Loued S, Khalil A. Purified human paraoxonase-1 interacts with plasma membrane lipid rafts and mediates cholesterol efflux from macrophages. Free Radic Biol Med 2012; 52:1372-81. [PMID: 22336243 DOI: 10.1016/j.freeradbiomed.2012.01.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 01/17/2012] [Accepted: 01/23/2012] [Indexed: 11/24/2022]
Abstract
Paraoxonase-1 (PON1) is a high-density lipoprotein (HDL)-associated serum enzyme thought to make a major contribution to the antioxidant and anti-inflammatory capacities of HDLs. However, the role of PON1 in the modulation of cholesterol efflux is poorly understood. The aim of our study was to investigate the involvement of PON1 in the regulation of cholesterol efflux, especially the mechanism by which it modulates HDL-mediated cholesterol transport. The enrichment of HDL(3) with human PON1 enhanced, in a dose-dependent manner, cholesterol efflux from THP-1 macrophage-like cells and ABCA1-enriched J774 macrophages. Moreover, an additive effect was observed when ABCA1-enriched J774 macrophages were incubated with both PON1 and apo-AI. Interestingly, PON1 alone was able to mediate cholesterol efflux from J774 macrophages and to upregulate ABCA1 expression on J774 macrophages. Immunofluorescence measurement showed an increase in PON1 levels in the cytoplasm of J774 macrophages overexpressing ABCA1. PON1 used an apo-AI-like mechanism to modulate cholesterol efflux from rapid and slow efflux pools derived from the lipid raft and nonraft domains of the plasma membrane, respectively. This was supported by the fact that ABCA1 protein was incrementally expressed by J774 macrophages within the first few hours of incubation with cholesterol-loaded J774 macrophages and that cyclodextrin significantly inhibited the capacity of PON1 to modulate cholesterol efflux from macrophages. This finding suggested that PON1 plays an important role in the antiatherogenic properties of HDLs and may exert its protective function outside the lipoprotein environment.
Collapse
Affiliation(s)
- Hicham Berrougui
- Research Center on Aging, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada QC J1H 4C4
| | | | | |
Collapse
|
23
|
Nagao K, Tomioka M, Ueda K. Function and regulation of ABCA1 - membrane meso-domain organization and reorganization. FEBS J 2011; 278:3190-203. [DOI: 10.1111/j.1742-4658.2011.08170.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
24
|
|
25
|
Kumon Y, Yasuoka Y, Yamanaka S, Wada A, Takeuchi H, Sugiura T. Acute-phase serum amyloid A is present in human colostrum and milk. Amyloid 2011; 18 Suppl 1:11-3. [PMID: 21838415 DOI: 10.3109/13506129.2011.574354003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Y Kumon
- Department of Laboratory Medicine, Kochi Medical School, Kochi University, Kochi, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
de Beer MC, Ji A, Jahangiri A, Vaughan AM, de Beer FC, van der Westhuyzen DR, Webb NR. ATP binding cassette G1-dependent cholesterol efflux during inflammation. J Lipid Res 2010; 52:345-53. [PMID: 21138980 DOI: 10.1194/jlr.m012328] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
ATP binding cassette transporter G1 (ABCG1) mediates the transport of cellular cholesterol to HDL, and it plays a key role in maintaining macrophage cholesterol homeostasis. During inflammation, HDL undergoes substantial remodeling, acquiring lipid changes and serum amyloid A (SAA) as a major apolipoprotein. In the current study, we investigated whether remodeling of HDL that occurs during acute inflammation impacts ABCG1-dependent efflux. Our data indicate that lipid free SAA acts similarly to apolipoprotein A-I (apoA-I) in mediating sequential efflux from ABCA1 and ABCG1. Compared with normal mouse HDL, acute phase (AP) mouse HDL containing SAA exhibited a modest but significant 17% increase in ABCG1-dependent efflux. Interestingly, AP HDL isolated from mice lacking SAA (SAAKO mice) was even more effective in promoting ABCG1 efflux. Hydrolysis with Group IIA secretory phospholipase A(2) (sPLA(2)-IIA) significantly reduced the ability of AP HDL from SAAKO mice to serve as a substrate for ABCG1-mediated cholesterol transfer, indicating that phospholipid (PL) enrichment, and not the presence of SAA, is responsible for alterations in efflux. AP human HDL, which is not PL-enriched, was somewhat less effective in mediating ABCG1-dependent efflux compared with normal human HDL. Our data indicate that inflammatory remodeling of HDL impacts ABCG1-dependent efflux independent of SAA.
Collapse
Affiliation(s)
- Maria C de Beer
- Departments of Physiology, University of Kentucky Medical Center, Lexington, KY, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang J, Cai S, Peterson BR, Kris-Etherton PM, Heuvel JPV. Development of a cell-based, high-throughput screening assay for cholesterol efflux using a fluorescent mimic of cholesterol. Assay Drug Dev Technol 2010; 9:136-46. [PMID: 21050070 DOI: 10.1089/adt.2010.0288] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Reverse cholesterol transport is the process by which extrahepatic cells, including macrophage-derived foam cells in arterial atherosclerotic plaque, transport excessive cholesterol back to the liver for bile acid synthesis and excretion, thus lowering the peripheral lipid burden. Cholesterol efflux from peripheral cells is the first step in this process, and finding drugs and interventions that promote this event is an important endeavor. Radioisotope-labeled cholesterol traditionally has been employed in measuring efflux efficiency, but this reagent has limitations for high-throughput screening. We developed an alternative method to measure cholesterol efflux in macrophage-derived foam cells using a novel fluorescent cholesterol mimic comprising the Pennsylvania Green fluorophore, attached by a linker containing a glutamic acid residue, to a derivative of N-alkyl-3β-cholesterylamine. Compared with the traditional radioisotope-based assay, this fluorescence-based assay gave similar results in the presence of known modulators of cholesterol efflux, such as cyclic AMP, and different cholesterol acceptors. When the fluorescent probe was employed in a high-throughput screening format, a variety of chemicals and bioactive compounds with known and unknown effects on cholesterol efflux could be tested simultaneously by plate-reader in a short period of time. Treatment of THP-1-derived macrophages with inhibitors of the membrane transporter ATP-binding cassette A1, such as glyburide or a specific antibody, significantly reduced the export of this fluorescent compound, indicating that ATP-binding cassette A1 represents the primary mediator of its cellular efflux. This fluorescent mimic of cholesterol provides a safe, sensitive, and reproducible alternative to radioactive assays in efflux experiments and has great potential as a valuable tool when incorporated into a drug discovery program.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, 16802, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Schizophrenia is one of the most severe psychiatric disorders affecting 1% of the world population. There is yet no empirical method to validate the diagnosis of the disease. The identification of an underlying molecular alteration could lead to an improved disease understanding and may yield an objective panel of biomarkers to aid in the diagnosis of this devastating disease. Presented is the largest reported liquid chromatography-mass spectrometry-based proteomic profiling study investigating serum samples taken from first-onset drug-naive patients compared with samples collected from healthy volunteers. The results of this large-scale study are presented along with enzyme-linked immunosorbent assay-based validation data.
Collapse
|
29
|
Moriarty PM, Luyendyk JP, Gibson CA, Backes JM. Effect of low-density lipoprotein apheresis on plasma levels of apolipoprotein e4. Am J Cardiol 2010; 105:1585-7. [PMID: 20494666 DOI: 10.1016/j.amjcard.2010.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 01/11/2010] [Accepted: 01/11/2010] [Indexed: 11/17/2022]
Abstract
Apolipoprotein E4 (apoE4) is a positively charged proinflammatory apolipoprotein bound to high-density lipoprotein (HDL) cholesterol and remnant lipoproteins. ApoE4 is associated with an increased risk of cardiovascular and cerebrovascular disease. Low-density lipoprotein (LDL) apheresis, a therapy for patients with familial hypercholesterolemia, removes apolipoprotein B and other positively charged plasma proteins but negatively charged proteins such as HDL cholesterol are generally spared. Despite their negative charge, LDL apheresis still removes 10% to 15% of HDL cholesterol, in particular, inflammatory HDL cholesterol. Patients with familial hypercholesterolemia have increased plasma levels of apoE4 and apoE4-bound HDL cholesterol. We tested the hypothesis that LDL apheresis would reduce the plasma levels of apoE4. We analyzed the plasma apoE4 levels using enzyme-linked immunosorbent assay immediately before and after LDL apheresis in 10 patients with familial hypercholesterolemia who had tested positive for the apoE4 isoform. After one treatment, the mean plasma apoE4 levels had been reduced by 39%, LDL cholesterol by 75%, triglycerides by 38%, and HDL cholesterol by 18%. The change in HDL cholesterol was significantly related to the apoE4 baseline values (r = -0.83, p = 0.001) and apoE4 levels after apheresis (r = 0.816, p = 0.004). In conclusion, LDL apheresis acutely reduced the plasma levels of apoE4. The mechanism of apoE4 reduction by LDL apheresis might be related to the selective reduction of a particular HDL cholesterol.
Collapse
Affiliation(s)
- Patrick M Moriarty
- Department of Internal Medicine, Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| | | | | | | |
Collapse
|
30
|
Yin K, Liao DF, Tang CK. ATP-binding membrane cassette transporter A1 (ABCA1): a possible link between inflammation and reverse cholesterol transport. Mol Med 2010; 16:438-49. [PMID: 20485864 DOI: 10.2119/molmed.2010.00004] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 05/11/2010] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is characterized by a chronic inflammatory condition that involves numerous cellular and molecular inflammatory components. A wide array of inflammatory mediators, such as cytokines and proteins produced by macrophages and other cells, play a critical role in the development and progression of the disease. ATP-binding membrane cassette transporter A1 (ABCA1) is crucial for cellular cholesterol efflux and reverse cholesterol transport (RCT) and is also identified as an important target in antiatherosclerosis treatment. Evidence from several recent studies indicates that inflammation, along with other atherogenic-related mediators, plays distinct regulating roles in ABCA1 expression. Proatherogenic cytokines such as interferon (IFN)-γ and interleukin (IL)-1β have been shown to inhibit the expression of ABCA1, while antiatherogenic cytokines, including IL-10 and transforming growth factor (TGF)-β1, have been shown to promote the expression of ABCA1. Moreover, some cytokines such as tumor necrosis factor (TNF)-α seem to regulate ABCA1 expression in species-specific and dose-dependent manners. Inflammatory proteins such as C-reactive protein (CRP) and cyclooxygenase (COX)-2 are likely to inhibit ABCA1 expression during inflammation, and inflammation induced by lipopolysaccharide (LPS) was also found to block the expression of ABCA1. Interestingly, recent experiments revealed ABCA1 can function as an antiinflammatory receptor to suppress the expression of inflammatory factors, suggesting that ABCA1 may be the molecular basis for the interaction between inflammation and RCT. This review aims to summarize recent findings on the role of inflammatory cytokines, inflammatory proteins, inflammatory lipids, and the endotoxin-mediated inflammatory process in expression of ABCA1. Also covered is the current understanding of the function of ABCA1 in modulating the immune response and inflammation through its direct and indirect antiinflammatory mechanisms including lipid transport, high-density lipoprotein (HDL) formation and apoptosis.
Collapse
Affiliation(s)
- Kai Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, China
| | | | | |
Collapse
|
31
|
Abstract
High density lipoprotein (HDL) possesses important anti-atherogenic properties and this review addresses the molecular mechanisms underlying these functions. The structures and cholesterol transport abilities of HDL particles are determined by the properties of their exchangeable apolipoprotein (apo) components. ApoA-I and apoE, which are the best characterized in structural terms, contain a series of amphipathic alpha-helical repeats. The helices located in the amino-terminal two-thirds of the molecule adopt a helix bundle structure while the carboxy-terminal segment forms a separately folded, relatively disorganized, domain. The latter domain initiates lipid binding and this interaction induces changes in conformation; the alpha-helix content increases and the amino-terminal helix bundle can open subsequently. These conformational changes alter the abilities of apoA-I and apoE to function as ligands for their receptors. The apoA-I and apoE molecules possess detergent-like properties and they can solubilize vesicular phospholipid to create discoidal HDL particles with hydrodynamic diameters of ~10 nm. In the case of apoA-I, such a particle is stabilized by two protein molecules arranged in an anti-parallel, double-belt, conformation around the edge of the disc. The abilities of apoA-I and apoE to solubilize phospholipid and stabilize HDL particles enable these proteins to be partners with ABCA1 in mediating efflux of cellular phospholipid and cholesterol, and the biogenesis of HDL particles. ApoA-I-containing nascent HDL particles play a critical role in cholesterol transport in the circulation whereas apoE-containing HDL particles mediate cholesterol transport in the brain. The mechanisms by which HDL particles are remodeled by lipases and lipid transfer proteins, and interact with SR-BI to deliver cholesterol to cells, are reviewed.
Collapse
|
32
|
Ohta S, Tanaka M, Sakakura K, Kawakami T, Aimoto S, Saito H. Defining lipid-binding regions of human serum amyloid A using its fragment peptides. Chem Phys Lipids 2009; 162:62-8. [DOI: 10.1016/j.chemphyslip.2009.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 06/30/2009] [Accepted: 07/31/2009] [Indexed: 11/30/2022]
|
33
|
Carty CL, Heagerty P, Heckbert SR, Enquobahrie DA, Jarvik GP, Davis S, Tracy RP, Reiner AP. Association of genetic variation in serum amyloid-A with cardiovascular disease and interactions with IL6, IL1RN, IL1beta and TNF genes in the Cardiovascular Health Study. J Atheroscler Thromb 2009; 16:419-30. [PMID: 19729864 DOI: 10.5551/jat.no968] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Since inflammation is an important contributor to atherosclerosis, gene variants mediating inflammation are of interest. We investigated gene variants in acute phase serum amyloid-A (SAA), a sensitive indicator of inflammatory activity, and their associations with cardiovascular disease (CVD) and HDL cholesterol. Interaction of the SAA genes with genetic variants of their regulators, IL-1, IL-6 and TNF-alpha in influencing CVD was also explored. METHODS SNPs characterizing common variation in the SAA1 and SAA2 genes were genotyped in European-(EA) and African-American (AA) participants (n=3969 and n=719) of the Cardiovascular Health Study. Using linear and Cox proportional hazards regression, we assessed associations of SNPs with baseline carotid artery intima-media thickness (cIMT) and risk of incident myocardial infarction, ischemic stroke, total CVD events or mortality during 14 years of follow-up. RESULTS No associations between SAA SNPs and outcomes were observed in EA, with the exception of total CVD events; each rs4638289 minor allele was associated with an increased risk in obese individuals, HR=1.2 (95%CI: 0.981.4; p=0.086) and decreased risk among non-obese, HR=0.9 (95%CI: 0.80.99; p=0.026). In AA, we observed modest associations between SAA SNPs and cIMT, potentially modified by HDL. SAA SNPs were also associated with lower HDL in EA and AA. Suggestive gene-gene interaction findings for cIMT in AA and CVD mortality in EA were not significant in subsequent model selection tests. CONCLUSION Associations of SAA SNPs with cIMT, HDL and total CVD events were identified, unadjusted for multiple testing. These findings should be regarded as hypothesis-generating until confirmed by other studies.
Collapse
Affiliation(s)
- Cara L Carty
- Department of Epidemiology, University of Washington, WA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The atheroprotective effects of HDL are mediated by several mechanisms, including its role in reverse cholesterol transport and via its antiinflammatory properties. However, not all HDL is functionally similar. HDL and apolipoprotein A-I may become dysfunctional or even proinflammatory and thus promote atherosclerosis. ApoAI posttranslational modification can have a large impact on its function. Myeloperoxidase modification of apoAI impairs its function as a cholesterol acceptor, and the molecular changes induced by myeloperoxidase have been studied in detail. These studies provide the basis for the development of an oxidant-resistant form of apoAI and clinical measures of HDL modification and dysfunction, which may be useful as a treatment criterion.
Collapse
Affiliation(s)
- Jonathan D Smith
- Department of Cell Biology, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
35
|
Villarroel F, Zambrano A, Amthauer R, Concha MI. Detection of up-regulated serum amyloid A transcript and of amyloid AA aggregates in skeletal muscle lesions of rainbow trout infected with Flavobacterium psychrophilum. Vet Immunol Immunopathol 2009; 130:120-4. [PMID: 19268373 DOI: 10.1016/j.vetimm.2009.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 01/27/2009] [Accepted: 02/03/2009] [Indexed: 10/21/2022]
Abstract
Serum amyloid A (SAA) is a family of acute-phase proteins, recognized as important effectors of innate immunity in higher vertebrates. Under pro-inflammatory conditions, up-regulation of saa transcripts occurs not only in the liver, but also in several extrahepatic tissues of a wide variety of vertebrates. SAA is also known as the precursor to amyloid A (AA), a major component of amyloid fibrils deposited in liver, kidney and spleen of humans suffering chronic inflammatory diseases. Here we show the up-regulation of saa transcription in lesions affecting skin, adipose tissue and skeletal muscle of rainbow trout naturally and experimentally infected with Flavobacterium psychrophilum, the causative agent of cold water disease (CWD). Using an antiserum against a trout acute SAA peptide that was previously shown to specifically recognize intact recombinant trout SAA and peptides derived from it, we showed by confocal microscopy analysis extensive colocalization of SAA and thioflavin T (ThT) staining in the skeletal muscle fibers of infected fish, suggesting for the first time the presence of AA-derived aggregates in the skeletal muscle of a lower vertebrate. These findings support the idea that SAA and/or its derivatives could constitute relevant markers for fish health and also for fish meat quality control.
Collapse
Affiliation(s)
- Franz Villarroel
- Instituto de Bioquímica, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.
| | | | | | | |
Collapse
|
36
|
Malle E, Sodin-Semrl S, Kovacevic A. Serum amyloid A: an acute-phase protein involved in tumour pathogenesis. Cell Mol Life Sci 2009; 66:9-26. [PMID: 18726069 PMCID: PMC4864400 DOI: 10.1007/s00018-008-8321-x] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The synthesis of acute-phase protein serum amyloid A (SAA) is largely regulated by inflammation- associated cytokines and a high concentration of circulating SAA may represent an ideal marker for acute and chronic inflammatory diseases. However, SAA is also synthesized in extrahepatic tissues, e.g. human carcinoma metastases and cancer cell lines. An increasing body of in vitro data supports the concept of involvement of SAA in carcinogenesis and neoplastic diseases. Accumulating evidence suggests that SAA might be included in a group of biomarkers to detect a pattern of physiological events that reflect the growth of malignancy and host response. This review is meant to provide a broad overview of the many ways that SAA could contribute to tumour development, and accelerate tumour progression and metastasis, and to gain a better understanding of this acute-phase reactant as a possible link between chronic inflammation and neoplasia.
Collapse
Affiliation(s)
- E Malle
- Center of Molecular Medicine, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, A-8010 Graz, Austria.
| | | | | |
Collapse
|
37
|
Villarroel F, Casado A, Vásquez J, Matamala E, Araneda B, Amthauer R, Enriquez R, Concha MI. Serum amyloid A: a typical acute-phase reactant in rainbow trout? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1160-9. [PMID: 18440634 DOI: 10.1016/j.dci.2008.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 03/06/2008] [Accepted: 03/07/2008] [Indexed: 05/20/2023]
Abstract
Acute serum amyloid A (A-SAA) has been considered a major acute-phase reactant and an effector of innate immunity in all vertebrates. The work presented here shows that the expression of A-SAA is strongly induced in a wide variety of immune-relevant tissues in rainbow trout, either naturally infected with Flavobacterium psychrophilum or challenged with lipopolysaccharide (LPS) or CpG oligonucleotides (CpG ODN). Nevertheless, A-SAA was undetectable by Western blot either in the plasma or in high-density lipoprotein (HDL) of infected or challenged fish, using either an anti-mouse SAA1 IgG or an anti-trout A-SAA peptide serum, which recognise both the intact recombinant trout A-SAA and fragments derived from it. However, the anti-peptide serum was the immunoreactive in all primary defence barriers and in mononuclear cells of head kidney, spleen and liver. These findings reveal that, unlike mammalian SAA, trout A-SAA does not increase significantly in the plasma of diseased fish, suggesting it is more likely to be involved in local defence.
Collapse
Affiliation(s)
- Franz Villarroel
- Instituto de Bioquímica, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja, Valdivia, Chile.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Hu W, Abe-Dohmae S, Tsujita M, Iwamoto N, Ogikubo O, Otsuka T, Kumon Y, Yokoyama S. Biogenesis of HDL by SAA is dependent on ABCA1 in the liver in vivo. J Lipid Res 2007; 49:386-93. [PMID: 18033752 DOI: 10.1194/jlr.m700402-jlr200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Serum amyloid A (SAA) was markedly increased in the plasma and in the liver upon acute inflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) in mice, and SAA in the plasma was exclusively associated with HDL. In contrast, no HDL was present in the plasma and only a small amount of SAA was found in the VLDL/LDL fraction (d < 1.063 g/ml) after the induction of inflammation in ABCA1-knockout (KO) mice, although SAA increased in the liver. Primary hepatocytes isolated from LPS-treated wild-type (WT) and ABCA1-KO mice both secreted SAA into the medium. SAA secreted from WT hepatocytes was associated with HDL, whereas SAA from ABCA1-KO hepatocytes was recovered in the fraction that was >1.21 g/ml. The behavior of apolipoprotein A-I (apoA-I) was the same as that of SAA in HDL biogenesis by WT and ABCA1-KO mouse hepatocytes. Lipid-free SAA and apoA-I both stabilized ABCA1 and caused cellular lipid release in WT mouse-derived fibroblasts, but not in ABCA1-KO mouse-derived fibroblasts, in vitro when added exogenously. We conclude that both SAA and apoA-I generate HDL largely in hepatocytes only in the presence of ABCA1, likely being secreted in a lipid-free form to interact with cellular ABCA1. In the absence of ABCA1, nonlipidated SAA is seemingly removed rapidly from the extracellular space.
Collapse
Affiliation(s)
- Wei Hu
- Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Westerterp M, Berbée JFP, Pires NMM, van Mierlo GJD, Kleemann R, Romijn JA, Havekes LM, Rensen PCN. Apolipoprotein C-I is crucially involved in lipopolysaccharide-induced atherosclerosis development in apolipoprotein E-knockout mice. Circulation 2007; 116:2173-81. [PMID: 17967778 DOI: 10.1161/circulationaha.107.693382] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lipopolysaccharide (LPS), which is released from gram-negative bacteria on multiplication or lysis, aggravates atherosclerosis in humans and rodents by inducing inflammation via toll-like receptors. Because apolipoprotein C-I (apoCI) enhances the LPS-induced inflammatory response in macrophages in vitro and in mice, we investigated the effect of endogenous apoCI expression on LPS-induced atherosclerosis in mice. METHODS AND RESULTS Twelve-week-old apoe-/- apoc1-/- and apoe-/- apoc1+/+ mice received weekly intraperitoneal injections of LPS (50 microg) or vehicle for a period of 10 weeks, and atherosclerosis development was assessed in the aortic root. LPS administration did not affect atherosclerotic lesion area in apoe-/- apoc1-/- mice but increased it in apoe-/- apoc1+/+ mice. In fact, apoCI expression increased the LPS-induced atherosclerotic lesion area by 60% (P<0.05), concomitant with an increase in LPS-induced plasma levels of fibrinogen and E-selectin. This indicated that apoCI increased the LPS-induced inflammatory state, both systemically (ie, fibrinogen) and at the level of the vessel wall (ie, E-selectin). In addition, both macrophage-derived apoCI and HDL-associated apoCI increased the LPS-induced tumor necrosis factor-alpha response by macrophages in vitro. CONCLUSIONS We conclude that apoCI is crucially involved in LPS-induced atherosclerosis in apoe-/- mice, which mainly relates to an increased inflammatory response toward LPS. We anticipate that apoCI plasma levels contribute to accelerated atherosclerosis development in individuals who have chronic infection.
Collapse
Affiliation(s)
- Marit Westerterp
- The Netherlands Organization for Applied Scientific Research-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW The aim of this article is to review recent advances made towards understanding how inflammation and acute phase proteins, particularly serum amyloid A and group IIa secretory phospholipase A2, may alter reverse cholesterol transport by HDL during inflammation and the acute phase response. RECENT FINDINGS Findings suggest that the decreased apoA-I content and markedly increased serum amyloid A content in HDL during the acute phase response result from reciprocal and coordinate transcriptional regulation of these proteins as well as HDL remodeling by group IIa secretory phospholipase A2. Serum amyloid A functions efficiently in a lipid-free or lipid-poor form to promote cholesterol efflux by ATP binding cassette protein ABCA1, evidently by functioning directly as an acceptor for cholesterol efflux as well as by increasing the availability of cellular free cholesterol. Serum amyloid A increases the ability of acute phase HDL to serve as an acceptor for SR-BI-dependent cellular cholesterol efflux. Altered remodeling of HDL by group IIa secretory phospholipase A2 in concert with cholesterol ester transfer protein may contribute to the generation of lipid-poor apoA-I and serum amyloid A acceptors for cholesterol efflux. SUMMARY Current data support a model for the acute phase response in which serum amyloid A and sPLA2-IIa, present at sites of inflammation and tissue damage, play a protective role by enhancing cellular cholesterol efflux, thereby promoting the removal of excess cholesterol from macrophages.
Collapse
Affiliation(s)
- Deneys R van der Westhuyzen
- Department of Internal Medicine, Cardiovascular Research Center and Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA.
| | | | | |
Collapse
|
41
|
Marsche G, Frank S, Raynes J, Kozarsky K, Sattler W, Malle E. The lipidation status of acute-phase protein serum amyloid A determines cholesterol mobilization via scavenger receptor class B, type I. Biochem J 2007; 402:117-24. [PMID: 17034364 PMCID: PMC1783981 DOI: 10.1042/bj20061406] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
During the acute-phase reaction, SAA (serum amyloid A) replaces apoA-I (apolipoprotein A-I) as the major HDL (high-density lipoprotein)-associated apolipoprotein. A remarkable portion of SAA exists in a lipid-free/lipid-poor form and promotes ABCA1 (ATP-binding cassette transporter A1)-dependent cellular cholesterol efflux. In contrast with lipid-free apoA-I and apoE, lipid-free SAA was recently reported to mobilize SR-BI (scavenger receptor class B, type I)-dependent cellular cholesterol efflux [Van der Westhuyzen, Cai, de Beer and de Beer (2005) J. Biol. Chem. 280, 35890-35895]. This unique property could strongly affect cellular cholesterol mobilization during inflammation. However, in the present study, we show that overexpression of SR-BI in HEK-293 cells (human embryonic kidney cells) (devoid of ABCA1) failed to mobilize cholesterol to lipid-free or lipid-poor SAA. Only reconstituted vesicles containing phospholipids and SAA promoted SR-BI-mediated cholesterol efflux. Cholesterol efflux from HEK-293 and HEK-293[SR-BI] cells to lipid-free and lipid-poor SAA was minimal, while efficient efflux was observed from fibroblasts and CHO cells (Chinese-hamster ovary cells) both expressing functional ABCA1. Overexpression of SR-BI in CHO cells strongly attenuated cholesterol efflux to lipid-free SAA even in the presence of an SR-BI-blocking IgG. This implies that SR-BI attenuates ABCA1-mediated cholesterol efflux in a way that is not dependent on SR-BI-mediated re-uptake of cholesterol. The present in vitro experiments demonstrate that the lipidation status of SAA is a critical factor governing cholesterol acceptor properties of this amphipathic apolipoprotein. In addition, we demonstrate that SAA mediates cellular cholesterol efflux via the ABCA1 and/or SR-BI pathway in a similar way to apoA-I.
Collapse
Affiliation(s)
- Gunther Marsche
- *Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Sǎsa Frank
- *Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - John G. Raynes
- †Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K
| | - Karen F. Kozarsky
- ‡GlaxoSmithKline, 709 Swedeland Rd, King of Prussia, PA 19406, U.S.A
| | - Wolfgang Sattler
- *Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Ernst Malle
- *Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, A-8010 Graz, Austria
- To whom correspondence should be addressed (email )
| |
Collapse
|
42
|
Heller M, Schlappritzi E, Stalder D, Nuoffer JM, Haeberli A. Compositional protein analysis of high density lipoproteins in hypercholesterolemia by shotgun LC-MS/MS and probabilistic peptide scoring. Mol Cell Proteomics 2007; 6:1059-72. [PMID: 17339632 DOI: 10.1074/mcp.m600326-mcp200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A protein of a biological sample is usually quantified by immunological techniques based on antibodies. Mass spectrometry offers alternative approaches that are not dependent on antibody affinity and avidity, protein isoforms, quaternary structures, or steric hindrance of antibody-antigen recognition in case of multiprotein complexes. One approach is the use of stable isotope-labeled internal standards; another is the direct exploitation of mass spectrometric signals recorded by LC-MS/MS analysis of protein digests. Here we assessed the peptide match score summation index based on probabilistic peptide scores calculated by the PHENYX protein identification engine for absolute protein quantification in accordance with the protein abundance index as proposed by Mann and co-workers (Rappsilber, J., Ryder, U., Lamond, A. I., and Mann, M. (2002) Large-scale proteomic analysis of the human spliceosome. Genome Res. 12, 1231-1245). Using synthetic protein mixtures, we demonstrated that this approach works well, although proteins can have different response factors. Applied to high density lipoproteins (HDLs), this new approach compared favorably to alternative protein quantitation methods like UV detection of protein peaks separated by capillary electrophoresis or quantitation of protein spots on SDS-PAGE. We compared the protein composition of a well defined HDL density class isolated from plasma of seven hypercholesterolemia subjects having low or high HDL cholesterol with HDL from nine normolipidemia subjects. The quantitative protein patterns distinguished individuals according to the corresponding concentration and distribution of cholesterol from serum lipid measurements of the same samples and revealed that hypercholesterolemia in unrelated individuals is the result of different deficiencies. The presented approach is complementary to HDL lipid analysis; does not rely on complicated sample treatment, e.g. chemical reactions, or antibodies; and can be used for projective clinical studies of larger patient groups.
Collapse
Affiliation(s)
- Manfred Heller
- Laboratory of Thrombosis Research, Department of Clinical Research, University of Bern, Switzerland.
| | | | | | | | | |
Collapse
|
43
|
Iwamoto N, Abe-Dohmae S, Sato R, Yokoyama S. ABCA7 expression is regulated by cellular cholesterol through the SREBP2 pathway and associated with phagocytosis. J Lipid Res 2006; 47:1915-27. [PMID: 16788211 DOI: 10.1194/jlr.m600127-jlr200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABCA7 is highly homologous to ABCA1 and mediates cellular cholesterol and phospholipid release by apolipoproteins when transfected in vitro. However, expression of ABCA7 was downregulated by increased cellular cholesterol while ABCA1 was upregulated, and the results were consistent by forced expression or downregulation of sterol-responsive/regulatory element (SRE) binding proteins (SREBPs). We analyzed the promoter of the ABCA7 gene and identified the new exon encoding 96 bp (mouse) and 95 bp (human) of the 5' untranslated region and the transcription start site at 1,122 bp (mouse) and 1,260 bp (human) upstream of the initiation methionine codon. At 5' upstream of this exon is the ABCA7 proximal promoter containing multiple binding sites of transcription factors for hematopoiesis and SRE of 9 bp at 212 bp (mouse) and 179 bp (human) upstream of the new exon. The apolipoprotein A-I-mediated lipid release was not influenced by suppression of the endogenous ABCA7 with small interfering RNA in mouse fibroblasts or by its increase in ABCA1-deficient mouse cells. In contrast, phagocytic activity was altered in parallel to the ABCA7 expression in these cells. When phagocytosis was induced, the messages increased for SREBP2, ABCA7, and other SREBP2-regulated proteins. The ABCA1 message decreased in this condition. We conclude that the ABCA7 gene is regulated by sterol in the opposite direction to ABCA1 through SRE/SREBP2 and that expression of ABCA7 by this regulation is associated with phagocytic activity.
Collapse
Affiliation(s)
- Noriyuki Iwamoto
- Biochemistry, Cell Biology, and Metabolism 1, Nagoya City University, Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | | | | |
Collapse
|