1
|
Lin X, Zhang W, Yang C, Ma P, He K, Chen G, Tao Y, Yan H, Yang Z, Zhang L, Fan J, Cui Q, Huang W, Liu G, Xian X, Wang Y. Depleting LCAT Aggravates Atherosclerosis in LDLR-deficient Hamster with Reduced LDL-Cholesterol Level. J Adv Res 2024; 63:187-194. [PMID: 37923248 PMCID: PMC11379981 DOI: 10.1016/j.jare.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023] Open
Abstract
INTRODUCTION Lecithin cholesterol acyltransferase (LCAT) plays a crucial role in acyl-esterifying cholesterol in plasma, which is essential for reverse cholesterol transport (RCT). Previous studies indicated that its activity on both α and β lipoproteins interpret its effects on lipoproteins for many controversial investigations of atherosclerosis. OBJECTIVES To better understand the relationship between LCAT, diet-induced dyslipidemia and atherosclerosis, we developed a double knockout (LCAT-/-&LDLR-/-, DKO) hamster model to evaluate the specific role of LCAT independent of LDL clearance effects. METHODS Plasma triglyceride (TG), total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), and free cholesterol (FC) levels were measured using biochemical reagent kits. FPLC was performed to analyze the components of lipoproteins. Apolipoprotein content was assessed using western blotting (WB). The hamsters were fed a high cholesterol/high fat diet (HCHFD) to induce atherosclerosis. Oil Red O staining was employed to detect plaque formation. Peritoneal macrophages were studied to investigate the effects of LCAT on cholesterol uptake and efflux. RESULTS On HCHFD, DKO hamsters exhibited significantly elevated levels of TG and FC, while HDL-C was nearly undetectable without affecting TC levels, as compared to low-density lipoprotein receptor (LDLR)-deficient (LDLR-/-, LKO) hamsters. Lipoprotein profiling revealed a marked increase in plasma chylomicron/very low-density lipoprotein (CM/VLDL) fractions, along with an unexpected reduction in LDL fraction in DKO hamsters. Furthermore, DKO hamsters displayed aggravated atherosclerotic lesions in the aorta, aortic root, and coronary artery relative to LKO hamsters, attributed to a pro-atherogenic lipoprotein profile and impaired cholesterol efflux in macrophages. CONCLUSIONS Our study demonstrates the beneficial role of LCAT in inhibiting atherosclerotic development and highlights the distinctive lipid metabolism characteristics in hamsters with familial hypercholesterolemia.
Collapse
Affiliation(s)
- Xiao Lin
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing 100191, China
| | - Wei Zhang
- Peking University Hepatology Institute, Peking University People's Hospital, Beijing, China
| | - Chun Yang
- Department of General Practice, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ping Ma
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Kunxiang He
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Gonglie Chen
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Yijun Tao
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Haizhao Yan
- Department of Molecular Pathology, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Zhao Yang
- Faculty of Art and Science, University of Toronto, Toronto, Canada
| | - Ling Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Jianglin Fan
- Department of Molecular Pathology, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Qinghua Cui
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing 100191, China
| | - Wei Huang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - George Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China.
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China.
| |
Collapse
|
2
|
Reyes-Soffer G, Matveyenko A, Lignos J, Matienzo N, Santos Baez LS, Hernandez-Ono A, Yung L, Nandakumar R, Singh SA, Aikawa M, George R, Ginsberg HN. Effects of Recombinant Human Lecithin Cholesterol Acyltransferase on Lipoprotein Metabolism in Humans. Arterioscler Thromb Vasc Biol 2024; 44:1407-1418. [PMID: 38695168 DOI: 10.1161/atvbaha.123.320387] [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: 11/06/2023] [Accepted: 03/28/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND LCAT (lecithin cholesterol acyl transferase) catalyzes the conversion of unesterified, or free cholesterol, to cholesteryl ester, which moves from the surface of HDL (high-density lipoprotein) into the neutral lipid core. As this iterative process continues, nascent lipid-poor HDL is converted to a series of larger, spherical cholesteryl ester-enriched HDL particles that can be cleared by the liver in a process that has been termed reverse cholesterol transport. METHODS We conducted a randomized, placebocontrolled, crossover study in 5 volunteers with atherosclerotic cardiovascular disease, to examine the effects of an acute increase of recombinant human (rh) LCAT via intravenous administration (300-mg loading dose followed by 150 mg at 48 hours) on the in vivo metabolism of HDL APO (apolipoprotein)A1 and APOA2, and the APOB100-lipoproteins, very low density, intermediate density, and low-density lipoproteins. RESULTS As expected, recombinant human LCAT treatment significantly increased HDL-cholesterol (34.9 mg/dL; P≤0.001), and this was mostly due to the increase in cholesteryl ester content (33.0 mg/dL; P=0.014). This change did not affect the fractional clearance or production rates of HDL-APOA1 and HDL-APOA2. There were also no significant changes in the metabolism of APOB100-lipoproteins. CONCLUSIONS Our results suggest that an acute increase in LCAT activity drives greater flux of cholesteryl ester through the reverse cholesterol transport pathway without significantly altering the clearance and production of the main HDL proteins and without affecting the metabolism of APOB100-lipoproteins. Long-term elevations of LCAT might, therefore, have beneficial effects on total body cholesterol balance and atherogenesis.
Collapse
Affiliation(s)
- Gissette Reyes-Soffer
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Anastasiya Matveyenko
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - James Lignos
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Nelsa Matienzo
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Leinys S Santos Baez
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Antonio Hernandez-Ono
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Lau Yung
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| | - Renu Nandakumar
- Irving Institute for Clinical and Translations Research (R.N.) and Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine (S.A.S., M.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine (S.A.S., M.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine (M.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine (M.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Richard George
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (R.G.)
| | - Henry N Ginsberg
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., A.M., J.L., N.M., L.S.S.B., A.H.-O., L.Y., H.N.G.)
| |
Collapse
|
3
|
Hou Y, Zhang X, Sun X, Qin Q, Chen D, Jia M, Chen Y. Genetically modified rabbit models for cardiovascular medicine. Eur J Pharmacol 2022; 922:174890. [PMID: 35300995 DOI: 10.1016/j.ejphar.2022.174890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/23/2022] [Accepted: 03/09/2022] [Indexed: 01/19/2023]
Abstract
Genetically modified (GM) rabbits are outstanding animal models for studying human genetic and acquired diseases. As such, GM rabbits that express human genes have been extensively used as models of cardiovascular disease. Rabbits are genetically modified via prokaryotic microinjection. Through this process, genes are randomly integrated into the rabbit genome. Moreover, gene targeting in embryonic stem (ES) cells is a powerful tool for understanding gene function. However, rabbits lack stable ES cell lines. Therefore, ES-dependent gene targeting is not possible in rabbits. Nevertheless, the RNA interference technique is rapidly becoming a useful experimental tool that enables researchers to knock down specific gene expression, which leads to the genetic modification of rabbits. Recently, with the emergence of new genetic technology, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), clustered regularly interspaced short palindromic repeats (CRISPR), and CRISPR-associated protein 9 (CRISPR/Cas9), major breakthroughs have been made in rabbit gene targeting. Using these novel genetic techniques, researchers have successfully modified knockout (KO) rabbit models. In this paper, we aimed to review the recent advances in GM technology in rabbits and highlight their application as models for cardiovascular medicine.
Collapse
Affiliation(s)
- Ying Hou
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Xin Zhang
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Xia Sun
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China; School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Qiaohong Qin
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Di Chen
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China; School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Min Jia
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Yulong Chen
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, 710021, China.
| |
Collapse
|
4
|
LCAT- targeted therapies: Progress, failures and future. Biomed Pharmacother 2022; 147:112677. [PMID: 35121343 DOI: 10.1016/j.biopha.2022.112677] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
Lecithin: cholesterol acyltransferase (LCAT) is the only enzyme in plasma which is able to esterify cholesterol and boost cholesterol esterify with phospholipid-derived acyl chains. In order to better understand the progress of LCAT research, it is always inescapable that it is linked to high-density lipoprotein (HDL) metabolism and reverse cholesterol transport (RCT). Because LCAT plays a central role in HDL metabolism and RCT, many animal studies and clinical studies are currently aimed at improving plasma lipid metabolism by increasing LCAT activity in order to find better treatment options for familial LCAT deficiency (FLD), fish eye disease (FED), and cardiovascular disease. Recombinant human LCAT (rhLCAT) injections, cells and gene therapy, and small molecule activators have been carried out with promising results. Recently rhLCAT therapies have entered clinical phase II trials with good prospects. In this review, we discuss the diseases associated with LCAT and therapies that use LCAT as a target hoping to find out whether LCAT can be an effective therapeutic target for coronary heart disease and atherosclerosis. Also, probing the mechanism of action of LCAT may help better understand the heterogeneity of HDL and the action mechanism of dynamic lipoprotein particles.
Collapse
|
5
|
Vitali C, Khetarpal SA, Rader DJ. HDL Cholesterol Metabolism and the Risk of CHD: New Insights from Human Genetics. Curr Cardiol Rep 2017; 19:132. [PMID: 29103089 DOI: 10.1007/s11886-017-0940-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Elevated high-density lipoprotein cholesterol levels in the blood (HDL-C) represent one of the strongest epidemiological surrogates for protection against coronary heart disease (CHD), but recent human genetic and pharmacological intervention studies have raised controversy about the causality of this relationship. Here, we review recent discoveries from human genome studies using new analytic tools as well as relevant animal studies that have both addressed, and in some cases, fueled this controversy. RECENT FINDINGS Methodologic developments in genotyping and sequencing, such as genome-wide association studies (GWAS), exome sequencing, and exome array genotyping, have been applied to the study of HDL-C and risk of CHD in large, multi-ethnic populations. Some of these efforts focused on population-wide variation in common variants have uncovered new polymorphisms at novel loci associated with HDL-C and, in some cases, CHD risk. Other efforts have discovered loss-of-function variants for the first time in genes previously implicated in HDL metabolism through common variant studies or animal models. These studies have allowed the genetic relationship between these pathways, HDL-C and CHD to be explored in humans for the first time through analysis tools such as Mendelian randomization. We explore these discoveries for selected key HDL-C genes CETP, LCAT, LIPG, SCARB1, and novel loci implicated from GWAS including GALNT2, KLF14, and TTC39B. Recent human genetics findings have identified new nodes regulating HDL metabolism while reshaping our current understanding of known candidate genes to HDL and CHD risk through the study of critical variants across model systems. Despite their effect on HDL-C, variants in many of the reviewed genes were found to lack any association with CHD. These data collectively indicate that HDL-C concentration, which represents a static picture of a very dynamic and heterogeneous metabolic milieu, is unlikely to be itself causally protective against CHD. In this context, human genetics represent an extremely valuable tool to further explore the biological mechanisms regulating HDL metabolism and investigate what role, if any, HDL plays in the pathogenesis of CHD.
Collapse
Affiliation(s)
- Cecilia Vitali
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Sumeet A Khetarpal
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA. .,Departments of Genetics and Medicine, Cardiovascular Institute, and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, 11-125 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Miller NE, Olszewski WL, Miller IP, Nanjee MN. Mechanism and Physiologic Significance of the Suppression of Cholesterol Esterification in Human Interstitial Fluid. Front Pharmacol 2016; 7:216. [PMID: 27471469 PMCID: PMC4945636 DOI: 10.3389/fphar.2016.00216] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/05/2016] [Indexed: 11/13/2022] Open
Abstract
Cholesterol esterification in high density lipoproteins (HDLs) by lecithin:cholesterol acyltransferase (LCAT) promotes unesterified cholesterol (UC) transfer from red cell membranes to plasma in vitro. However, it does not explain the transfer of UC from most peripheral cells to interstitial fluid in vivo, as HDLs in afferent peripheral lymph are enriched in UC. Having already reported that the endogenous cholesterol esterification rate (ECER) in lymph is only 5% of that in plasma, we have now explored the underlying mechanism. In peripheral lymph from 20 healthy men, LCAT concentration, LCAT activity (assayed using an optimized substrate), and LCAT specific activity averaged, respectively, 11.8, 10.3, and 84.9% of plasma values. When recombinant human LCAT was added to lymph, the increments in enzyme activity were similar to those when LCAT was added to plasma. Addition of apolipoprotein AI (apo AI), fatty acid-free albumin, Intralipid, or the d < 1.006 g/ml plasma fraction had no effect on ECER. During incubation of lymph plus plasma, the ECER was similar to that observed with buffer plus plasma. When lymph was added to heat-inactivated plasma, the ECER was 11-fold greater than with lymph plus buffer. Addition of discoidal proteoliposomes of apo AI and phosphatidycholine (PC) to lymph increased ECER 10-fold, while addition of apo AI/PC/UC disks did so by only six-fold. We conclude that the low ECER in lymph is due to a property of the HDLs, seemingly substrate inhibition of LCAT by excess cell-derived UC. This is reversed when lymph enters plasma, consequent upon redistribution of UC from lymph HDLs to plasma lipoproteins.
Collapse
Affiliation(s)
| | - Waldemar L Olszewski
- Department of Surgical Research and Transplantology, Medical Research Centre, Polish Academy of Sciences, Warsaw Poland
| | - Irina P Miller
- Department of Cardiovascular Biochemistry, Queen Mary University of London, London UK
| | - Mahmud N Nanjee
- Cardiovascular Genetics Unit, School of Medicine, University of Utah, Salt Lake City, UT USA
| |
Collapse
|
7
|
Gunawardane RN, Fordstrom P, Piper DE, Masterman S, Siu S, Liu D, Brown M, Lu M, Tang J, Zhang R, Cheng J, Gates A, Meininger D, Chan J, Carlson T, Walker N, Schwarz M, Delaney J, Zhou M. Agonistic Human Antibodies Binding to Lecithin-Cholesterol Acyltransferase Modulate High Density Lipoprotein Metabolism. J Biol Chem 2015; 291:2799-811. [PMID: 26644477 PMCID: PMC4742745 DOI: 10.1074/jbc.m115.672790] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Indexed: 11/28/2022] Open
Abstract
Drug discovery opportunities where loss-of-function alleles of a target gene link to a disease-relevant phenotype often require an agonism approach to up-regulate or re-establish the activity of the target gene. Antibody therapy is increasingly recognized as a favored drug modality due to multiple desirable pharmacological properties. However, agonistic antibodies that enhance the activities of the target enzymes are rarely developed because the discovery of agonistic antibodies remains elusive. Here we report an innovative scheme of discovery and characterization of human antibodies capable of binding to and agonizing a circulating enzyme lecithin cholesterol acyltransferase (LCAT). Utilizing a modified human LCAT protein with enhanced enzymatic activity as an immunogen, we generated fully human monoclonal antibodies using the XenoMouseTM platform. One of the resultant agonistic antibodies, 27C3, binds to and substantially enhances the activity of LCAT from humans and cynomolgus macaques. X-ray crystallographic analysis of the 2.45 Å LCAT-27C3 complex shows that 27C3 binding does not induce notable structural changes in LCAT. A single administration of 27C3 to cynomolgus monkeys led to a rapid increase of plasma LCAT enzymatic activity and a 35% increase of the high density lipoprotein cholesterol that was observed up to 32 days after 27C3 administration. Thus, this novel scheme of immunization in conjunction with high throughput screening may represent an effective strategy for discovering agonistic antibodies against other enzyme targets. 27C3 and other agonistic human anti-human LCAT monoclonal antibodies described herein hold potential for therapeutic development for the treatment of dyslipidemia and cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Stephanie Masterman
- Therapeutic Discovery, Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Sophia Siu
- From Therapeutic Discovery, Amgen Inc., Seattle, Washington 98119
| | | | - Mike Brown
- From Therapeutic Discovery, Amgen Inc., Seattle, Washington 98119
| | - Mei Lu
- Therapeutic Discovery, and
| | | | | | - Janet Cheng
- From Therapeutic Discovery, Amgen Inc., Seattle, Washington 98119
| | - Andrew Gates
- From Therapeutic Discovery, Amgen Inc., Seattle, Washington 98119
| | - David Meininger
- From Therapeutic Discovery, Amgen Inc., Seattle, Washington 98119
| | | | - Tim Carlson
- PKDM Department, Amgen Inc., South San Francisco, California 94080, and
| | | | | | - John Delaney
- Therapeutic Discovery, Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | | |
Collapse
|
8
|
Elshourbagy NA, Meyers HV, Abdel-Meguid SS. Cholesterol: the good, the bad, and the ugly - therapeutic targets for the treatment of dyslipidemia. Med Princ Pract 2013; 23:99-111. [PMID: 24334831 PMCID: PMC5586853 DOI: 10.1159/000356856] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 10/27/2013] [Indexed: 01/06/2023] Open
Abstract
Maintaining cholesterol and triglyceride (TG) levels within healthy limits is critical for decreasing the risk of heart disease. Dyslipidemia refers to the abnormal levels of lipids in the blood, including low high-density lipoprotein cholesterol (HDL-C), also known as good cholesterol, high low-density lipoprotein cholesterol (LDL-C), also known as bad cholesterol, and/or high TG levels that contribute to the development and progression of atherosclerosis. In this article we reviewed some of the current therapeutic targets for the treatment of dyslipidemia, with a primary focus on endothelial lipase and lecithin cholesterol acyl transferase for raising HDL-C, and the proprotein convertase subtilisin-like kexin type 9 (PCSK9), microsomal triglyceride transfer protein, and the messenger RNA of apolipoprotein B for lowering LDL-C. In addition, we reviewed the role of apolipoprotein AI (apoAI) in raising HDL-C, where we discuss three apoAI-based drugs under development. These are its mutated dimer (apoAI-Milano), a complex with phospholipids, and a mimetic peptide. Atherosclerosis, mainly because of dyslipidemia, is a leading cause of cardiovascular disease. Regarding the title of this article, the 'good' refers to HDL-C, the 'bad' refers to LDL-C, and the 'ugly' refers to atherosclerosis.
Collapse
|
9
|
Abstract
Multiple human population studies have established the concentration of high density lipoprotein (HDL) cholesterol as an independent, inverse predictor of the risk of having a cardiovascular event. Furthermore, HDLs have several well-documented functions with the potential to protect against cardiovascular disease. These include an ability to promote the efflux of cholesterol from macrophages in the artery wall, inhibit the oxidative modification of low density lipoproteins (LDLs), inhibit vascular inflammation, inhibit thrombosis, promote endothelial repair, promote angiogenesis, enhance endothelial function, improve diabetic control, and inhibit hematopoietic stem cell proliferation. There are undoubtedly other beneficial functions of HDLs yet to be identified. The HDL fraction in human plasma is heterogeneous, consisting of several subpopulations of particles of varying size, density, and composition. The functions of the different HDL subpopulations remain largely unknown. Given that therapies that increase the concentration of HDL cholesterol have varying effects on the levels of specific HDL subpopulations, it is of great importance to understand how distribution of different HDL subpopulations contribute to the potentially cardioprotective functions of this lipoprotein fraction. This review summarizes current understanding of the relationship of HDL subpopulations to their cardioprotective properties and highlights the gaps in current knowledge regarding this important aspect of HDL biology.
Collapse
Affiliation(s)
- Kerry-Anne Rye
- Lipid Research Group, Centre for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia 2052
| | | |
Collapse
|
10
|
Lecithin/cholesterol acyltransferase modulates diet-induced hepatic deposition of triglycerides in mice. J Nutr Biochem 2013; 24:567-77. [DOI: 10.1016/j.jnutbio.2012.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Revised: 01/10/2012] [Accepted: 02/20/2012] [Indexed: 11/20/2022]
|
11
|
Remaley AT. Apolipoprotein A-II: still second fiddle in high-density lipoprotein metabolism? Arterioscler Thromb Vasc Biol 2013; 33:166-7. [PMID: 23325471 PMCID: PMC3864680 DOI: 10.1161/atvbaha.112.300921] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
12
|
Kunnen S, Van Eck M. Lecithin:cholesterol acyltransferase: old friend or foe in atherosclerosis? J Lipid Res 2012; 53:1783-99. [PMID: 22566575 PMCID: PMC3413220 DOI: 10.1194/jlr.r024513] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/23/2012] [Indexed: 11/20/2022] Open
Abstract
Lecithin:cholesterol acyltransferase (LCAT) is a key enzyme that catalyzes the esterification of free cholesterol in plasma lipoproteins and plays a critical role in high-density lipoprotein (HDL) metabolism. Deficiency leads to accumulation of nascent preβ-HDL due to impaired maturation of HDL particles, whereas enhanced expression is associated with the formation of large, apoE-rich HDL(1) particles. In addition to its function in HDL metabolism, LCAT was believed to be an important driving force behind macrophage reverse cholesterol transport (RCT) and, therefore, has been a subject of great interest in cardiovascular research since its discovery in 1962. Although half a century has passed, the importance of LCAT for atheroprotection is still under intense debate. This review provides a comprehensive overview of the insights that have been gained in the past 50 years on the biochemistry of LCAT, the role of LCAT in lipoprotein metabolism and the pathogenesis of atherosclerosis in animal models, and its impact on cardiovascular disease in humans.
Collapse
Affiliation(s)
- Sandra Kunnen
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Gorlaeus Laboratories, Leiden University, Leiden, The Netherlands
| | - Miranda Van Eck
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Gorlaeus Laboratories, Leiden University, Leiden, The Netherlands
| |
Collapse
|
13
|
Rosenson RS, Brewer HB, Davidson WS, Fayad ZA, Fuster V, Goldstein J, Hellerstein M, Jiang XC, Phillips MC, Rader DJ, Remaley AT, Rothblat GH, Tall AR, Yvan-Charvet L. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation 2012; 125:1905-19. [PMID: 22508840 PMCID: PMC4159082 DOI: 10.1161/circulationaha.111.066589] [Citation(s) in RCA: 716] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Robert S Rosenson
- Mount Sinai Heart, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
AAV8-mediated long-term expression of human LCAT significantly improves lipid profiles in hCETP;Ldlr(+/-) mice. J Cardiovasc Transl Res 2011; 4:801-10. [PMID: 21822774 DOI: 10.1007/s12265-011-9309-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 07/11/2011] [Indexed: 12/17/2022]
Abstract
Lecithin:cholesterol acyltransferase (LCAT) is the key circulating enzyme responsible for high-density lipoprotein (HDL) cholesterol esterification, HDL maturation, and potentially reverse cholesterol transport. To further explore LCAT's mechanism of action on lipoprotein metabolism, we employed adeno-associated viral vector (AAV) serotype 8 to achieve long-term (32-week) high level expression of human LCAT in hCETP;Ldlr(+/-) mice, and characterized the lipid profiles in detail. The mice had a marked increase in HDL cholesterol, HDL particle size, and significant reduction in low-density lipoprotein (LDL) cholesterol, plasma triglycerides, and plasma apoB. Plasma LCAT activity significantly increased with humanized substrate specificity. HDL cholesteryl esters increased in a fashion that fits human LCAT specificity. HDL phosphatidylcholines trended toward decrease, with no change observed for HDL lysophosphatidylcholines. Triglycerides reduction appeared to reside in all lipoprotein particles (very low-density lipoprotein (VLDL), LDL, and HDL), with HDL triglycerides composition highly reflective of VLDL, suggesting that changes in HDL triglycerides were primarily driven by the altered triglycerides metabolism in VLDL. In summary, in this human-like model for lipoprotein metabolism, AAV8-mediated overexpression of human LCAT resulted in profound changes in plasma lipid profiles. Detailed lipid analyses in the lipoprotein particles suggest that LCAT's beneficial effect on lipid metabolism includes not only enhanced HDL cholesterol esterification but also improved metabolism of apoB-containing particles and triglycerides. Our findings thus shed new light on LCAT's mechanism of action and lend support to its therapeutic potential in treating dyslipidemia.
Collapse
|
15
|
Sider KL, Blaser MC, Simmons CA. Animal models of calcific aortic valve disease. Int J Inflam 2011; 2011:364310. [PMID: 21826258 PMCID: PMC3150155 DOI: 10.4061/2011/364310] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 04/27/2011] [Indexed: 11/20/2022] Open
Abstract
Calcific aortic valve disease (CAVD), once thought to be a degenerative disease, is now recognized to be an active pathobiological process, with chronic inflammation emerging as a predominant, and possibly driving, factor. However, many details of the pathobiological mechanisms of CAVD remain to be described, and new approaches to treat CAVD need to be identified. Animal models are emerging as vital tools to this end, facilitated by the advent of new models and improved understanding of the utility of existing models. In this paper, we summarize and critically appraise current small and large animal models of CAVD, discuss the utility of animal models for priority CAVD research areas, and provide recommendations for future animal model studies of CAVD.
Collapse
Affiliation(s)
- Krista L Sider
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
| | | | | |
Collapse
|
16
|
Hypertriglyceridemia and delayed clearance of fat load in transgenic rabbits expressing human apolipoprotein CIII. Transgenic Res 2010; 20:867-75. [DOI: 10.1007/s11248-010-9467-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Accepted: 11/15/2010] [Indexed: 10/18/2022]
|
17
|
Zerfas PM, Brinster LR, Starost MF, Burkholder TH, Raffeld M, Eckhaus MA. Amelanotic melanoma in a New Zealand White Rabbit (Oryctolagus cuniculus). Vet Pathol 2010; 47:977-81. [PMID: 20460451 DOI: 10.1177/0300985810369898] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A 3.5-year-old intact male double-transgenic New Zealand white rabbit (Oryctolagus cuniculus), apoA-I and LCAT (apolipoprotein and lecithin:cholesterol acyltransferase), was presented with a discrete, raised facial mass (0.5 x 1.0 x 1.0 cm). The mass was surgically excised, with reoccurrence to the same site 88 days later. A second surgical excision was performed, and the rabbit died 3 weeks later from respiratory distress. At necropsy, multiple varying-sized masses were observed in the ventral mandibular region and throughout the lungs, pleura, and diaphragm. On histopathology, the masses were composed of moderately anisocytotic and anisokaryotic polygonal to spindloid cells with moderate finely granular, lightly eosinophilic cytoplasm, having round to oval nuclei with one to several nucleoli and finely stippled chromatin. Mitotic figures were frequent. Lymphatic and venous invasion were noted with neoplastic cells metastasized to the submandibular lymph nodes, lungs, liver, and adventitial surface of the aorta. Fontana-Masson stain was negative for melanin, thereby necessitating immunohistochemistry and transmission electron microscopy. Positive staining with MART-1 (a melanocyte protein marker) combined with transmission electron microscopy revealing type II melanosomes confirmed the diagnosis of an amelanotic melanoma.
Collapse
Affiliation(s)
- P M Zerfas
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Vergeer M, Holleboom AG, Kastelein JJP, Kuivenhoven JA. The HDL hypothesis: does high-density lipoprotein protect from atherosclerosis? J Lipid Res 2010; 51:2058-73. [PMID: 20371550 DOI: 10.1194/jlr.r001610] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is unequivocal evidence of an inverse association between plasma high-density lipoprotein (HDL) cholesterol concentrations and the risk of cardiovascular disease, a finding that has led to the hypothesis that HDL protects from atherosclerosis. This review details the experimental evidence for this "HDL hypothesis". In vitro studies suggest that HDL has a wide range of anti-atherogenic properties but validation of these functions in humans is absent to date. A significant number of animal studies and clinical trials support an atheroprotective role for HDL; however, most of these findings were obtained in the context of marked changes in other plasma lipids. Finally, genetic studies in humans have not provided convincing evidence that HDL genes modulate cardiovascular risk. Thus, despite a wealth of information on this intriguing lipoprotein, future research remains essential to prove the HDL hypothesis correct.
Collapse
Affiliation(s)
- Menno Vergeer
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
19
|
The diameter of liver sinusoidal fenestrae is not a major determinant of lipoprotein levels and atherosclerosis in cholesterol-fed rabbits. Cardiovasc Pathol 2009; 20:44-50. [PMID: 19914092 DOI: 10.1016/j.carpath.2009.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2008] [Revised: 06/12/2009] [Accepted: 09/04/2009] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The liver is a key organ in lipid and lipoprotein metabolism. It has been postulated that a small diameter of sinusoidal fenestrae retards clearance of chylomicron remnants, resulting in hypercholesterolemia and atherosclerosis. However, this hypothesis has not been rigorously tested hitherto. METHODS In the current study, we compared plasma levels of proatherogenic lipoproteins and assessed the development of atherosclerosis at distinct locations throughout the arterial tree in heterozygous New Zealand White and Dutch Belt rabbits that are deficient in low-density lipoprotein receptor and with an average fenestrae size of 103 and 124 nm, respectively. RESULTS Feeding of a 0.15% cholesterol diet for 4 months resulted in similar total plasma cholesterol levels in New Zealand White (420±20 mg/dl) and Dutch Belt (380±30 mg/dl) rabbits. Following isolation of lipoproteins by ultracentrifugation, no biologically significant differences in very-low-density lipoprotein, intermediate-density lipoprotein, and low-density lipoprotein cholesterol levels were observed between cholesterol-fed New Zealand White and Dutch Belt rabbits. Furthermore, the relative amount of intestinally derived apolipoprotein-B48-containing lipoproteins did not differ significantly between both strains (7.3±0.42% vs. 8.0±0.54%). Atherosclerosis was more pronounced in the thoracic aorta in New Zealand White rabbits than in Dutch Belt rabbits, but the reverse was observed with the abdominal aorta. These topographic differences cannot be explained by circulating lipoprotein levels. CONCLUSIONS The data presented in this study do not support the hypothesis that the diameter of fenestrae is an important determinant of chylomicron remnant levels, diet-induced hypercholesterolemia, and atherosclerosis in cholesterol-fed rabbits.
Collapse
|
20
|
Shiomi M, Ito T. The Watanabe heritable hyperlipidemic (WHHL) rabbit, its characteristics and history of development: A tribute to the late Dr. Yoshio Watanabe. Atherosclerosis 2009; 207:1-7. [DOI: 10.1016/j.atherosclerosis.2009.03.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 03/13/2009] [Accepted: 03/17/2009] [Indexed: 11/28/2022]
|
21
|
Chen X, Burton C, Song X, McNamara L, Langella A, Cianetti S, Chang CH, Wang J. An apoA-I mimetic peptide increases LCAT activity in mice through increasing HDL concentration. Int J Biol Sci 2009; 5:489-99. [PMID: 19680471 PMCID: PMC2726446 DOI: 10.7150/ijbs.5.489] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Accepted: 07/20/2009] [Indexed: 01/23/2023] Open
Abstract
Lecithin cholesterol acyltransferase (LCAT) plays a key role in the reverse cholesterol transport (RCT) process by converting cholesterol to cholesteryl ester to form mature HDL particles, which in turn deliver cholesterol back to the liver for excretion and catabolism. HDL levels in human plasma are negatively correlated with cardiovascular risk and HDL functions are believed to be more important in atheroprotection. This study investigates whether and how D-4F, an apolipoprotein A-I (apoA-I) mimetic peptide, influences LCAT activity in the completion of the RCT process. We demonstrated that the apparent rate constant value of the LCAT enzyme reaction gives a measure of LCAT activity and determined the effects of free metals and a reducing agent on LCAT activity, showing an inhibition hierarchy of Zn(2+)>Mg(2+)>Ca(2+) and no inhibition with beta-mercaptoethanol up to 10 mM. We reconstituted nano-disc particles using apoA-I or D-4F with phospholipids. These particles elicited good activity in vitro in the stimulation of cholesterol efflux from macrophages through the ATP-binding cassette transporter A1 (ABCA1). With these particles we studied the LCAT activity and demonstrated that D-4F did not activate LCAT in vitro. Furthermore, we have done in vivo experiments with apoE-null mice and demonstrated that D-4F (20 mg/kg body weight, once daily subcutaneously) increased LCAT activity and HDL level as well as apoA-I concentration at 72 hours post initial dosing. Finally, we have established a correlation between HDL concentration and LCAT activity in the D-4F treated mice.
Collapse
Affiliation(s)
- Xun Chen
- Merck Research Laboratories, Rahway, New Jersey 07065, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Rousset X, Vaisman B, Amar M, Sethi AA, Remaley AT. Lecithin: cholesterol acyltransferase--from biochemistry to role in cardiovascular disease. Curr Opin Endocrinol Diabetes Obes 2009; 16:163-71. [PMID: 19306528 PMCID: PMC2910390 DOI: 10.1097/med.0b013e328329233b] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW We discuss the latest findings on the biochemistry of lecithin : cholesterol acyltransferase (LCAT), the effect of LCAT on atherosclerosis, clinical features of LCAT deficiency, and the impact of LCAT on cardiovascular disease from human studies. RECENT FINDINGS Although there has been much recent progress in the biochemistry of LCAT and its effect on high-density lipoprotein metabolism, its role in the pathogenesis of atherosclerosis is still not fully understood. Studies from various animal models have revealed a complex interaction between LCAT and atherosclerosis that may be modified by diet and by other proteins that modify lipoproteins. Furthermore, the ability of LCAT to lower apoB appears to be the best way to predict its effect on atherosclerosis in animal models. Recent studies on patients with LCAT deficiency have shown a modest but significant increase in incidence of cardiovascular disease consistent with a beneficial effect of LCAT on atherosclerosis. The role of LCAT in the general population, however, has not revealed a consistent association with cardiovascular disease. SUMMARY Recent research findings from animal and human studies have revealed a potential beneficial role of LCAT in reducing atherosclerosis but additional studies are necessary to better establish the linkage between LCAT and cardiovascular disease.
Collapse
Affiliation(s)
- Xavier Rousset
- National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, Bethesda, MD. 20814
| | - Boris Vaisman
- National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, Bethesda, MD. 20814
| | - Marcelo Amar
- National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, Bethesda, MD. 20814
| | - Amar A. Sethi
- National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, Bethesda, MD. 20814
| | - Alan T. Remaley
- National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, Bethesda, MD. 20814
- To whom correspondence should be addressed: National Institutes of Health, National Heart, Lung and Blood Institute, Pulmonary and Vascular Medicine Branch, Lipoprotein Metabolism Section, 10 Center Dr. Bldg. 10/2C-433, Bethesda, MD. 20814, , 301-402-9796
| |
Collapse
|
23
|
Amar MJA, Shamburek RD, Vaisman B, Knapper CL, Foger B, Hoyt RF, Santamarina-Fojo S, Brewer HB, Remaley AT. Adenoviral expression of human lecithin-cholesterol acyltransferase in nonhuman primates leads to an antiatherogenic lipoprotein phenotype by increasing high-density lipoprotein and lowering low-density lipoprotein. Metabolism 2009; 58:568-75. [PMID: 19303980 PMCID: PMC3703629 DOI: 10.1016/j.metabol.2008.11.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 11/26/2008] [Indexed: 01/30/2023]
Abstract
Lecithin-cholesterol acyltransferase (LCAT), a key enzyme in high-density lipoprotein (HDL) metabolism, has been proposed to have atheroprotective properties by promoting reverse cholesterol transport. Overexpression of LCAT in various animal models, however, has led to conflicting results on its overall effect on lipoproteins and atherosclerosis. In this study, the effect of overexpression of LCAT in nonhuman primates on lipoprotein metabolism is examined. Human LCAT was expressed with adenovirus in squirrel monkeys (n = 8), resulting on day 4 in a 22-fold increase of LCAT activity (257 +/- 23 vs 5618 +/- 799 nmol mL(-1) h(-1), P < .0001). At its peak, LCAT was found to nearly double the level of HDL cholesterol from baseline (113 +/- 7 vs 260 +/- 24 mg/dL, P < .01). High-density lipoprotein formed after treatment with the adenovirus was larger in size, as assessed by fast protein liquid chromatography (FPLC) analysis. By kinetic studies, it was determined that there was a decrease in apolipoprotein (Apo) A-I resident time (0.373 +/- 0.027 vs 0.685 +/- 0.045 d(-1), P < .0001) and almost a doubling in the ApoA-I synthetic rate (22 +/- 2 vs 41 +/- 3 mg kg(-1) d(-1), P < .0001), but no overall change in ApoA-I levels. In addition, increased expression of LCAT was associated with a 37% reduction of ApoB levels (12 +/- 1 vs 19 +/- 1 mg/dL, P < .05) due to increased low-density lipoprotein catabolism (fractional catabolic rate = 1.7 +/- 0.1 d(-1) in controls vs 4.2 +/- 0.3 d(-1) in LCAT-treated group, P < .05). In summary, overexpression of LCAT in nonhuman primates leads to an antiatherogenic lipoprotein profile by increasing HDL cholesterol and lowering ApoB, thus making LCAT a potential drug target for reducing atherosclerosis.
Collapse
Affiliation(s)
- Marcelo J A Amar
- Lipoprotein Metabolism Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Apolipoprotein A-I and lecithin:cholesterol acyltransferase transfer induce cholesterol unloading in complex atherosclerotic lesions. Gene Ther 2009; 16:757-65. [PMID: 19242527 DOI: 10.1038/gt.2009.8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Plasma levels of high-density lipoprotein (HDL) cholesterol and its major apolipoprotein (apo), apo A-I, are inversely correlated with the incidence of ischemic cardiovascular diseases. Reverse cholesterol transport is likely the main mechanism underlying the atheroprotective effects of HDL. Here, we investigated whether increased HDL cholesterol following hepatocyte-directed adenoviral rabbit apo A-I (AdrA-I) or rabbit lecithin-cholesterol acyltransferase (LCAT) (AdrLCAT) transfer may induce cholesterol unloading in complex atherosclerotic lesions in heterozygous low-density lipoprotein receptor-deficient rabbits fed a 0.15% cholesterol diet for 420 days before and for 120 days after transfer. HDL cholesterol levels increased 2.0-fold (P<0.001) and 1.9-fold (P<0.001) in the 120 days after transfer with AdrA-I and AdrLCAT, respectively, compared to levels just before transfer whereas non-HDL cholesterol remained unchanged. Increased HDL cholesterol following AdrA-I and AdrLCAT transfer resulted in a 31% (P<0.05) reduction of the intima/media ratio in comparison with the control progression group. Compared to the baseline group killed after 420 days of cholesterol diet, AdrA-I and AdrLCAT transfer reduced the percentage of Oil Red O area 1.6-fold (P<0.001) and 1.4-fold (P<0.001), respectively. In conclusion, increased HDL cholesterol after AdrA-I and AdrLCAT transfer inhibits progression of atherosclerosis and induces cholesterol unloading in complex lesions in rabbits.
Collapse
|
25
|
Padoveze A, Maniero F, Oliveira T, Vitorio T, Couto R, Maranhão R. Effect of a cholesterol-rich diet on the metabolism of the free and esterified cholesterol components of a nanoemulsion that resembles LDL in rabbits. Braz J Med Biol Res 2009; 42:172-8. [DOI: 10.1590/s0100-879x2009000200005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Accepted: 01/15/2009] [Indexed: 11/21/2022] Open
Affiliation(s)
| | | | | | | | - R.D. Couto
- Universidade de São Paulo, Brasil; Universidade Federal da Bahia, Brasil
| | - R.C. Maranhão
- Universidade de São Paulo, Brasil; Universidade de São Paulo, Brasil
| |
Collapse
|
26
|
Remaley AT, Amar M, Sviridov D. HDL-replacement therapy: mechanism of action, types of agents and potential clinical indications. Expert Rev Cardiovasc Ther 2009; 6:1203-15. [PMID: 18939908 DOI: 10.1586/14779072.6.9.1203] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
HDL-replacement therapy is a promising new treatment strategy involving the acute administration of HDL to rapidly stabilize patients at imminent risk for developing a myocardial infarction, such as those with acute coronary syndrome. This review will first focus on the anti-atherogenic mechanisms for HDL, such as the stimulation of the reverse cholesterol transport pathway, and then discuss the other potential beneficial biological effects of HDL on atherosclerosis. The various types of HDL-replacement therapies that are being investigated and developed will be reviewed and ongoing clinical trials and other possible clinical indications for HDL-replacement therapy besides the prevention of myocardial infarction will also be described. Finally, HDL-replacement therapy will be put into perspective by summarizing the current gaps in our knowledge of HDL metabolism and identifying challenges for future research in this area.
Collapse
Affiliation(s)
- Alan T Remaley
- National Institutes of Health, Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, National Heart, Lung, and Blood Institute, Bethesda, MD 20892-21508, USA.
| | | | | |
Collapse
|
27
|
Degrace P, Moindrot B, Mohamed I, Gresti J, Clouet P. Moderate consumption of beer reduces liver triglycerides and aortic cholesterol deposit in LDLr−/− apoB100/100 mice. Atherosclerosis 2006; 189:328-35. [PMID: 16487531 DOI: 10.1016/j.atherosclerosis.2006.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 01/11/2006] [Accepted: 01/16/2006] [Indexed: 10/25/2022]
Abstract
This study was designed to address the effects of a moderate consumption of beer on serum and liver lipid parameters and on the development of aortic lesions in a mouse model associated with a human atherogenic lipoprotein profile. LDLr(-/-) apoB(100/100) mice received each day during 12 weeks either water, mild beer (0.570g of ethanol/kg of body weight) or ethanol-free beer in a single pure dose. Serum and liver lipid parameters were analyzed and atherosclerotic lesions were estimated in heart and aorta through their total cholesterol content. mRNA levels of enzymes and receptors involved in lipoprotein uptake, in fatty acid esterification and oxidation, and in reverse cholesterol transport were also measured in the liver. Serum glucose, triglyceride (TG) and cholesterol levels were altered neither by ethanol-free beer nor by mild beer. Nevertheless, both beer treatments significantly increased HDL-cholesterol (HDL-C) and VLDL-C levels by reference to controls with no change in LDL-C levels. Liver TG contents were significantly decreased by either beer treatment. Cholesterol accumulation was attenuated in the whole aorta of mice treated with mild beer at p<0.05 and not significantly with ethanol-free beer. Heart cholesterol contents were comparable in the three series. Among the genes studied, only scavenger receptor-B1 was downregulated by both beer-based beverages. LDL receptor related protein, lecithin-cholesterol acyltransferase and sterol regulatory element-binding protein 2 were downregulated only by mild beer. The expression of other genes assayed was not altered. When administered in chronic and moderate dose, unidentified components of beer may exert beneficial effects towards atherosclerosis development through alteration of lipoprotein metabolism in LDLr(-/-) apoB(100/100) mice. This effect was slightly amplified by the presence of ethanol in beer.
Collapse
MESH Headings
- Alcohol Drinking
- Animals
- Aorta, Thoracic/metabolism
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Apolipoproteins B/deficiency
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Beer
- Cholesterol, HDL/metabolism
- Cholesterol, LDL/metabolism
- Cholesterol, VLDL/metabolism
- Chromatography, High Pressure Liquid
- Down-Regulation
- Female
- Liver/metabolism
- Mice
- Mice, Inbred C57BL
- Mitochondria, Liver/metabolism
- Oxidation-Reduction
- Palmitates/metabolism
- Phosphatidylcholine-Sterol O-Acyltransferase/genetics
- Polymerase Chain Reaction
- RNA, Messenger/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Scavenger Receptors, Class B/genetics
- Sterol Regulatory Element Binding Protein 2/genetics
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Pascal Degrace
- UPRES Lipides et Nutrition EA2422, Faculté des Sciences Gabriel, Université de Bourgogne, 6 Bd Gabriel, 21000 Dijon, France.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Atherosclerosis and related diseases are the leading cause of death in Western world. The disease process begins with the formation of fatty streaks already during the first decade of life but does not manifest clinically until several decades later. Gene therapy is a potential new way to target multiple factors playing a role in the development and progression of atherosclerosis. A great number of genes involved in the development of atherosclerosis have been identified and have been tested both in vitro and in vivo as potential new targets for therapy. Pre-clinical experiments have shown the feasibility and safety of several gene therapy applications for the treatment of atherosclerosis and clinical trials have also provided evidence for the applicability of gene therapy for the treatment of cardiovascular diseases. In this review we discuss vectors and potential gene therapy approaches for intervention and therapy of atherosclerosis.
Collapse
Affiliation(s)
- E Vähäkangas
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | | |
Collapse
|
29
|
Abstract
Genetically modified laboratory animals provide a powerful approach for studying gene expression and regulation and allow one to directly examine structure-function and cause-and-effect relationships in pathophysiological processes. Today, transgenic mice are available as a research tool in almost every research institution. On the other hand, the development of a relatively large mammalian transgenic model, transgenic rabbits, has provided unprecedented opportunities for investigators to study the mechanisms of human diseases and has also provided an alternative way to produce therapeutic proteins to treat human diseases. Transgenic rabbits expressing human genes have been used as a model for cardiovascular disease, AIDS, and cancer research. The recombinant proteins can be produced from the milk of transgenic rabbits not only at lower cost but also on a relatively large scale. One of the most promising and attractive recombinant proteins derived from transgenic rabbit milk, human alpha-glucosidase, has been successfully used to treat the patients who are genetically deficient in this enzyme. Although the pronuclear microinjection is still the major and most popular method for the creation of transgenic rabbits, recent progress in gene targeting and animal cloning has opened new avenues that should make it possible to produce transgenic rabbits by somatic cell nuclear transfer in the future. Based on a computer-assisted search of the studies of transgenic rabbits published in the English literature here, we introduce to the reader the achievements made thus far with transgenic rabbits, with emphasis on the application of these rabbits as human disease models and live bioreactors for producing human therapeutic proteins and on the recent progress in cloned rabbits.
Collapse
Affiliation(s)
- Jianglin Fan
- Laboratory of Cardiovascular Disease, Department of Pathology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba 305-8575, Japan.
| | | |
Collapse
|
30
|
Fischer TH, Brittain J, Trabalzini L, Banes AJ, White GC, Smith CJ, Nichols TC. The ras-binding domain of ral GDS-like protein-2 as a ras inhibitor in smooth muscle cells. Biochem Biophys Res Commun 2003; 305:934-40. [PMID: 12767920 DOI: 10.1016/s0006-291x(03)00878-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This study was undertaken to determine whether the response of smooth muscle cells to mitogens can be inhibited by inactivating ras with the ral GDS like protein-2 ras-binding domain (RGL2-RBD). RGL2 is a member of the ral GDS family of proteins that contains a carboxy terminal ras-binding domain which binds the GTP ligated form of ras and rap and a CDC25 homology domain with the structural features of a guanine nucleotide exchange factor. The effect of ras signaling on the smooth muscle cell growth factor response was studied using rat aortic A10 smooth muscle cells transfected with a plasmid that encoded the RGL2-RBD. RGL2-RBD transfection resulted in a 12-fold reduction in the number of clonal colonies that were obtained after selection, and dramatically slowed cell cycle progression. RGBL2-RBD reduced DNA synthesis and inhibited platelet derived growth factor (PDGF)-mediated activation of the MAPK pathway. These findings indicated that interfering with ras signaling inhibits smooth muscle cell proliferation and raise the possibility that ras signaling inhibition might be used therapeutically to control smooth muscle proliferation after vascular injury.
Collapse
Affiliation(s)
- Thomas H Fischer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Sun H, Usui SI, Shiomi M, Watanabe T, Fan J. A rapid PCR method of genotyping LDL receptor mutations in WHHL rabbits. J Atheroscler Thromb 2003; 9:145-8. [PMID: 12226556 DOI: 10.5551/jat.9.145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
WHHL rabbits are a valuable model for the study of human familial hypercholesterolemia and atherosclerosis. To use this animal model, it is often necessary to confirm LDL receptor status in WHHL rabbits. Here, we described a simple and rapid PCR method to detect LDL mutations in WHHL rabbits.
Collapse
Affiliation(s)
- Huijun Sun
- Department of Pathology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | |
Collapse
|
32
|
Goto A, Sasai K, Suzuki S, Fukutomi T, Ito S, Matsushita T, Okamoto M, Suzuki T, Itoh M, Okumura-Noji K, Yokoyama S. Plasma concentrations of LPL and LCAT are in putative association with females and alcohol use which are independent negative risk factors for coronary atherosclerosis among Japanese. Clin Chim Acta 2003; 329:69-76. [PMID: 12589967 DOI: 10.1016/s0009-8981(03)00021-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Coronary heart disease (CHD) prevalence appears low among Japanese. Analysis of their negative risk factors is therefore important for public health strategy. METHODS We analyzed the impact on coronary atherosclerosis of sex, alcohol intake, plasma lipoproteins and enzymes to regulate cholesterol transport, lipoprotein lipase (LPL) and lecithin:cholesterol acyltransferase (LCAT) for the 110 patients who underwent coronary angiography, consecutively enrolled by excluding those >70 years or under hypolipidemic-drug treatment. Subgroup combinations compared were males vs. females in non-drinkers, and drinkers vs. non-drinkers in males. RESULTS Coronary stenosis was less in females and in drinkers, accompanied by high-density lipoprotein (HDL) in the respective comparison. LPL associated with sex (females>males) and LCAT with alcohol intake (drinkers>non-drinkers) although neither enzyme demonstrated direct correlation with coronary stenosis. LPL positively associated only with HDL in most of subgroups and LCAT correlated with low-density lipoprotein (LDL) in all subgroups but with HDL only in males. CONCLUSIONS Among non-drinkers, females are at lower risk for coronary atherosclerosis than males mainly due to higher HDL in potential association with high LPL, and that drinkers are protected among males also by high HDL that is in apparent association with LCAT.
Collapse
Affiliation(s)
- Akitomo Goto
- Internal Medicine and Bioregulation, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
For gene therapy, the last few years have been an exciting period. Encouraging results from several successful gene therapy trials were reported. Children born with a life-threatening immune system disorder, severe combined immune deficiency (SCID), were cured after receiving gene therapy for replacement of their defective adenosine deaminase (ADA) gene. Gene therapy successes related to vascular complications were also reported. The first human gene therapy trial for a blood-vessel disorder was performed successfully, in which copies of an angiogenic gene, the vascular endothelial growth factor (VEGF) gene, were directly delivered to the area surrounding the diseased artery of the leg of a patient with peripheral artery disease. Within a few days, this stimulated the growth of new blood vessels around the blockage in the ailing blood vessel and helped avoid amputation. In 1998, a patient with genetically small arteries became the first to receive VEGF gene therapy in the heart. Multiple copies of a plasmid with the VEGF gene were delivered into the damaged area of the heart, and a few days later angiogenesis ensued that helped bypass the blocked vessel, with markedly reduced chest pain in the patient. Gene therapy is becoming a reality and, more importantly, it appears to be safe and does not require supplementary immuno-suppressing drugs. Gene therapy seems to have begun delivering on its promises.
Collapse
Affiliation(s)
- Sayon Roy
- Department of Ophthlamology, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | |
Collapse
|
34
|
Furbee JW, Parks JS. Transgenic overexpression of human lecithin: cholesterol acyltransferase (LCAT) in mice does not increase aortic cholesterol deposition. Atherosclerosis 2002; 165:89-100. [PMID: 12208474 DOI: 10.1016/s0021-9150(02)00201-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Results from several atherosclerosis studies using morphometric procedures have proven controversial with regard to whether over-expression of human LCAT in transgenic (Tg) mice is atherogenic. The purpose of the present study was to determine the effect of 10-fold over-expression of human LCAT on aortic free and esterified cholesterol (EC) deposition as well as plasma lipoprotein cholesteryl ester (CE) fatty acid composition in mice fed an atherogenic diet containing cholic acid. C57Bl/6 (control) and human LCAT-Tg mice were fed chow or an atherogenic diet (15% of calories from palm oil, 1.0% cholesterol and 0.5% cholic acid) for 24 weeks before measurement of aortic cholesterol content. Compared with the chow diet, control and LCAT-Tg mice fed the atherogenic diet had a 2-fold increase in plasma total, free and EC, a 7-fold increase in plasma apoB lipoprotein cholesterol, and a 40-50-fold increase in hepatic cholesterol content. The aortic EC content was increased in control (0.7 vs. 1.2 mg/g protein) and LCAT-Tg (0.3 vs. 1.5 mg/g protein) mice fed the atherogenic diet compared with those consuming the chow diet; however, there was no difference in aortic free (14.4+/-6.8 vs. 18.5+/-7.7 mg/g protein) or esterified (1.2+/-1.0 vs. 1.5+/-1.2 mg/g protein) cholesterol content between atherogenic diet-fed control and LCAT-Tg mice, respectively. LCAT-Tg mice fed the atherogenic diet had a 2-fold increase in the ratio of saturated+monounsaturated to polyunsaturated CE species in plasma apoB lipoproteins compared with control mice (9.4+/-2.4 vs. 4.9+/-0.7). We conclude that over-expression of human LCAT in Tg mice fed an atherogenic diet containing cholic acid does not result in increased aortic cholesterol deposition compared with control mice, even though the CE fatty acid saturation index of plasma apoB lipoproteins was doubled.
Collapse
Affiliation(s)
- James W Furbee
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1040, USA
| | | |
Collapse
|
35
|
Abstract
High-density lipoproteins (HDLs) are strongly related to risk of atherosclerotic cardiovascular disease. Low levels of HDL cholesterol are a major cardiovascular risk factor, and overexpression of the major HDL protein, apolipoprotein (apo) A-I, markedly inhibits progression and even induces regression of atherosclerosis in animal models. Clinical data regarding the effect of increasing HDL cholesterol on vascular events are limited. HDL remains an important potential target for therapeutic intervention. A variety of gene products are involved in the regulation of HDL metabolism. Yet, the mechanisms by which HDL inhibits atherosclerosis are not yet fully understood. There remains much to be learned about HDL metabolism and its relation to atherosclerosis and other cardiovascular risk factors.
Collapse
Affiliation(s)
- Daniel J Rader
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
36
|
Abstract
Plasma levels of high-density lipoprotein (HDL) cholesterol are strongly inversely associated with atherosclerotic cardiovascular disease, and overexpression of HDL proteins, such as apolipoprotein A-I in animals, reduces progression and even induces regression of atherosclerosis. Therefore, HDL metabolism is recognized as a potential target for therapeutic intervention of atherosclerotic vascular diseases. The antiatherogenic properties of HDL include promotion of cellular cholesterol efflux and reverse cholesterol transport, as well as antioxidant, anti-inflammatory and anticoagulant properties. The molecular regulation of HDL metabolism is not fully understood, but it is influenced by several extracellular lipases. Here, we focus on new developments and insights into the role of secreted lipases on HDL metabolism and their relationship to atherosclerosis.
Collapse
Affiliation(s)
- Weijun Jin
- University of Pennsylvania School of Medicine, 654 BRB II/III, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
37
|
Furbee JW, Sawyer JK, Parks JS. Lecithin:cholesterol acyltransferase deficiency increases atherosclerosis in the low density lipoprotein receptor and apolipoprotein E knockout mice. J Biol Chem 2002; 277:3511-9. [PMID: 11719520 DOI: 10.1074/jbc.m109883200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to test the hypothesis that lecithin:cholesterol acyltransferase (LCAT) deficiency would accelerate atherosclerosis development in low density lipoprotein (LDL) receptor (LDLr-/-) and apoE (apoE-/-) knockout mice. After 16 weeks of atherogenic diet (0.1% cholesterol, 10% calories from palm oil) consumption, LDLr-/- LCAT-/- double knockout mice, compared with LDLr-/- mice, had similar plasma concentrations of free (FC), esterified (EC), and apoB lipoprotein cholesterol, increased plasma concentrations of phospholipid and triglyceride, decreased HDL cholesterol, and 2-fold more aortic FC (142 +/- 28 versus 61 +/- 20 mg/g protein) and EC (102 +/- 27 versus 61+/- 27 mg/g). ApoE-/- LCAT-/- mice fed the atherogenic diet, compared with apoE-/- mice, had higher concentrations of plasma FC, EC, apoB lipoprotein cholesterol, and phospholipid, and significantly more aortic FC (149 +/- 62 versus 109 +/- 33 mg/g) and EC (101 +/- 23 versus 69 +/- 20 mg/g) than did the apoE-/- mice. LCAT deficiency resulted in a 12-fold increase in the ratio of saturated + monounsaturated to polyunsaturated cholesteryl esters in apoB lipoproteins in LDLr-/- mice and a 3-fold increase in the apoE-/- mice compared with their counterparts with active LCAT. We conclude that LCAT deficiency in LDLr-/- and apoE-/- mice fed an atherogenic diet resulted in increased aortic cholesterol deposition, likely due to a reduction in plasma HDL, an increased saturation of cholesteryl esters in apoB lipoproteins and, in the apoE-/- background, an increased plasma concentration of apoB lipoproteins.
Collapse
Affiliation(s)
- James W Furbee
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, North Carolina 27157-1040, USA
| | | | | |
Collapse
|
38
|
Moghadasian MH, Frohlich JJ, McManus BM. Advances in experimental dyslipidemia and atherosclerosis. J Transl Med 2001; 81:1173-83. [PMID: 11555665 DOI: 10.1038/labinvest.3780331] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Among the models of dyslipidemia and atherosclerosis, a number of wild-type, naturally defective, and genetically modified animals (rabbits, mice, pigeons, dogs, pigs, and monkeys) have been characterized. In particular, their similarities to and differences from humans in respect to relevant biochemical, physiologic, and pathologic conditions have been evaluated. Features of atherosclerotic lesions and their specific relationship to plasma lipoprotein particles have been critically reviewed and summarized. All animal models studied have limitations: the most significant advantages and disadvantages of using a specific animal species are outlined here. New insights in lipid metabolism and genetic background with regard to variations in pathogenesis of dyslipidemia-associated atherogenesis have also been reviewed. Evidence suggests that among wild-type species, strains of White Carneau pigeons and Watanabe Heritable Hyperlipidemic and St. Thomas's Hospital rabbits are preferable to the cholesterol-fed wild-type animal species in dyslipidemia and atherosclerosis research. Evidence for the usefulness of both wild-type and transgenic animals in studying the involvement of inflammatory pathways and Chlamydia pneumoniae infection in pathogenesis of atherosclerosis has also been summarized. Transgenic mice and rabbits are excellent tools for studying specific gene-related disorders. However, despite these significant achievements in animal experimentation, there are no suitable animal models for several rare types of fatal dyslipidemia-associated disorders such as phytosterolemia and cerebrotendinous xanthomatosis. An excellent model of diabetic atherosclerosis is unavailable. The question of reversibility of atherosclerosis still remains unanswered. Further work is needed to overcome these deficiencies.
Collapse
Affiliation(s)
- M H Moghadasian
- Healthy Heart Program, Department of Pathology and Laboratory Medicine, St. Paul's Hospital and the University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|
39
|
Santamarina-Fojo S, Lambert G, Hoeg JM, Brewer HB. Lecithin-cholesterol acyltransferase: role in lipoprotein metabolism, reverse cholesterol transport and atherosclerosis. Curr Opin Lipidol 2000; 11:267-75. [PMID: 10882342 DOI: 10.1097/00041433-200006000-00007] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the past several years significant advances have been made in our understanding of lecithin-cholesterol acyltransferase (LCAT) function. LCAT beneficially alters the plasma concentrations of apolipoprotein B-containing lipoproteins, as well as HDL. In addition, its proposed role in facilitating reverse cholesterol transport and modulating atherosclerosis has been demonstrated in vivo. Analysis of LCAT transgenic animals has established the importance of evaluating HDL function, as well as HDL plasma levels, to predict atherogenic risk.
Collapse
Affiliation(s)
- S Santamarina-Fojo
- Molecular Disease Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, USA.
| | | | | | | |
Collapse
|