1
|
Moll T, Farber SA. Zebrafish ApoB-Containing Lipoprotein Metabolism: A Closer Look. Arterioscler Thromb Vasc Biol 2024; 44:1053-1064. [PMID: 38482694 PMCID: PMC11042983 DOI: 10.1161/atvbaha.123.318287] [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: 04/26/2024]
Abstract
Zebrafish have become a powerful model of mammalian lipoprotein metabolism and lipid cell biology. Most key proteins involved in lipid metabolism, including cholesteryl ester transfer protein, are conserved in zebrafish. Consequently, zebrafish exhibit a human-like lipoprotein profile. Zebrafish with mutations in genes linked to human metabolic diseases often mimic the human phenotype. Zebrafish larvae develop rapidly and externally around the maternally deposited yolk. Recent work revealed that any disturbance of lipoprotein formation leads to the accumulation of cytoplasmic lipid droplets and an opaque yolk, providing a visible phenotype to investigate disturbances of the lipoprotein pathway, already leading to discoveries in MTTP (microsomal triglyceride transfer protein) and ApoB (apolipoprotein B). By 5 days of development, the digestive system is functional, making it possible to study fluorescently labeled lipid uptake in the transparent larvae. These and other approaches enabled the first in vivo description of the STAB (stabilin) receptors, showing lipoprotein uptake in endothelial cells. Various zebrafish models have been developed to mimic human diseases by mutating genes known to influence lipoproteins (eg, ldlra, apoC2). This review aims to discuss the most recent research in the zebrafish ApoB-containing lipoprotein and lipid metabolism field. We also summarize new insights into lipid processing within the yolk cell and how changes in lipid flux alter yolk opacity. This curious new finding, coupled with the development of several techniques, can be deployed to identify new players in lipoprotein research directly relevant to human disease.
Collapse
|
2
|
Mast N, El-Darzi N, Li Y, Pikuleva IA. Quantitative characterizations of the cholesterol-related pathways in the retina and brain of hamsters. J Lipid Res 2023:100401. [PMID: 37330011 PMCID: PMC10394389 DOI: 10.1016/j.jlr.2023.100401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
The retina and brain are separated from the systemic circulation by the anatomical barriers, which are permeable (the outer blood-retinal barrier) and impermeable (the blood-brain and inner blood-retina barriers) to cholesterol. Herein we investigated whether the whole-body cholesterol maintenance affects cholesterol homeostasis in the retina and brain. We used hamsters, whose whole-body cholesterol handling is more similar to those in humans than in mice and conducted separate administrations of deuterated water and deuterated cholesterol. We assessed the quantitative significance of the retinal and brain pathways of cholesterol input and compared the results with those from our previous studies in mice. The utility of the measurements in the plasma of deuterated 24-hydroxycholesterol, the major cholesterol elimination product from the brain, was investigated as well. We established that despite a 7-fold higher serum LDL to HDL ratio and other cholesterol-related differences, in situ biosynthesis remained the major source of cholesterol for hamster retina, although its quantitative significance was reduced to 53% as compared to 72-78% in mouse retina. In the brain, the principal pathway of cholesterol input was also the same, in situ biosynthesis, accounting for 94% of the total brain cholesterol input (96% in mice); the interspecies differences pertained to the absolute rates of the total cholesterol input and turnover. We documented the correlations between deuterium enrichments of the brain 24-hydroxycholesterol, brain cholesterol, and plasma 24-hydroxycholesterol, which suggested that deuterium enrichment of plasma 24-hydroxycholesteol could be an in vivo marker of cholesterol elimination and turnover in the brain.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Nicole El-Darzi
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Yong Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA.
| |
Collapse
|
3
|
Oteng AB, Higuchi S, Banks AS, Haeusler RA. Cyp2c-deficiency depletes muricholic acids and protects against high-fat diet-induced obesity in male mice but promotes liver damage. Mol Metab 2021; 53:101326. [PMID: 34438105 PMCID: PMC8449133 DOI: 10.1016/j.molmet.2021.101326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Murine-specific muricholic acids (MCAs) are reported to protect against obesity and associated metabolic disorders. However, the response of mice with genetic depletion of MCA to an obesogenic diet has not been evaluated. We used Cyp2c-deficient (Cyp2c−/−) mice, which lack MCAs and thus have a human-like bile acid (BA) profile, to directly investigate the potential role of MCAs in diet-induced obesity. Methods Male and female Cyp2c−/− mice and wild-type (WT) littermate controls were fed a standard chow diet or a high-fat diet (HFD) for 18 weeks. We measured BA composition from a pool of liver, gallbladder, and intestine, as well as weekly body weight, food intake, lean and fat mass, systemic glucose homeostasis, energy expenditure, intestinal lipid absorption, fecal lipid, and energy content. Results Cyp2c-deficiency depleted MCAs and caused other changes in BA composition, namely a decrease in the ratio of 12α-hydroxylated (12α-OH) BAs to non-12α-OH BAs, without altering the total BA levels. While WT male mice became obese after HFD feeding, Cyp2c−/− male mice were protected from obesity and associated metabolic dysfunctions. Cyp2c−/− male mice also showed reduced intestinal lipid absorption and increased lipid excretion, which was reversed by oral gavage with the 12α-OH BA and taurocholic acid (TCA). Cyp2c−/− mice also showed increased liver damage, which appeared stronger in females. Conclusions MCA does not protect against diet-induced obesity but may protect against liver injury. Reduced lipid absorption in Cyp2c-deficient male mice is potentially due to a reduced ratio of 12α-OH/non-12α-OH BAs. Presence of MCA does not necessarily protect against diet-induced obesity. Cyp2c deficiency promotes resistance to diet-induced obesity in males. Cyp2c-knockout mice have decreased the ratio of 12α-OH/non-12α-OH BAs that promotes decreased intestinal lipid absorption. Cyp2c-knockout mice have improved glucose homeostasis. Cyp2c-deficiency promotes mild and severe liver injury in male and female mice, respectively.
Collapse
Affiliation(s)
- Antwi-Boasiako Oteng
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sei Higuchi
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Alexander S Banks
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
Dorighello GG, Rovani JC, Paim BA, Rentz T, Assis LHP, Vercesi AE, Oliveira HCF. Mild Mitochondrial Uncoupling Decreases Experimental Atherosclerosis, A Proof of Concept. J Atheroscler Thromb 2021; 29:825-838. [PMID: 34092712 PMCID: PMC9174088 DOI: 10.5551/jat.62796] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aim: Atherosclerosis is responsible for high morbidity and mortality rates around the world. Local arterial oxidative stress is involved in all phases of atherosclerosis development. Mitochondria is a relevant source of the oxidants, particularly under certain risky conditions, such as hypercholesterolemia. The aim of this study was to test whether lowering the production of mitochondrial oxidants by induction of a mild uncoupling can reduce atherosclerosis in hypercholesterolemic LDL receptor knockout mice.
Methods: The mice were chronically treated with very low doses of DNP (2,4-dinitrophenol) and metabolic, inflammatory and redox state markers and atherosclerotic lesion sizes were determined.
Results: The DNP treatment did not change the classical atherosclerotic risk markers, such as plasma lipids, glucose homeostasis, and fat mass, as well as systemic inflammatory markers. However, the DNP treatment diminished the production of mitochondrial oxidants, systemic and tissue oxidative damage markers, peritoneal macrophages and aortic rings oxidants generation. Most importantly, development of spontaneous and diet-induced atherosclerosis (lipid and macrophage content) were significantly decreased in the DNP-treated mice. In vitro, DNP treated peritoneal macrophages showed decreased H2O2 production, increased anti-inflammatory cytokines gene expression and secretion, increased phagocytic activity, and decreased LDL-cholesterol uptake.
Conclusions: These findings are a proof of concept that activation of mild mitochondrial uncoupling is sufficient to delay the development of atherosclerosis under the conditions of hypercholesterolemia and oxidative stress. These results promote future approaches targeting mitochondria for the prevention or treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gabriel G Dorighello
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas.,Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas
| | - Juliana C Rovani
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas
| | - Bruno A Paim
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas
| | - Thiago Rentz
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas
| | - Leandro H P Assis
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas
| | - Anibal E Vercesi
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas
| |
Collapse
|
5
|
Pownall HJ, Liu J, Gillard BK, Yelamanchili D, Rosales C. Physico-chemical and physiological determinants of lipo-nanoparticle stability. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102361. [PMID: 33540069 DOI: 10.1016/j.nano.2021.102361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022]
Abstract
Liposome-based nanoparticles (NPs) comprised mostly of phospholipids (PLs) have been developed to deliver diagnostic and therapeutic agents. Whereas reassembled plasma lipoproteins have been tested as NP carriers of hydrophobic molecules, they are unstable because the components can spontaneously transfer to other PL surfaces-cell membranes and lipoproteins-and can be degraded by plasma lipases. Here we review two strategies for NP stabilization. One is to use PLs that contain long acyl-chains: according to a quantitative thermodynamic model and in vivo tests, increasing the chain length of a PL reduces the spontaneous transfer rate and increases plasma lifetime. A second strategy is to substitute ether for ester bonds which makes the PLs lipase resistant. We conclude with recommendations of simple ex vivo and in vitro tests of NP stability that should be conducted before in vivo tests are begun.
Collapse
Affiliation(s)
- Henry J Pownall
- Center for Bioenergetics, Department of Medicine, Houston Methodist Academic Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA.
| | - Jing Liu
- Center for Bioenergetics, Department of Medicine, Houston Methodist Academic Institute, Houston, TX, USA; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Baiba K Gillard
- Center for Bioenergetics, Department of Medicine, Houston Methodist Academic Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| | - Dedipya Yelamanchili
- Center for Bioenergetics, Department of Medicine, Houston Methodist Academic Institute, Houston, TX, USA
| | - Corina Rosales
- Center for Bioenergetics, Department of Medicine, Houston Methodist Academic Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
6
|
Kakiyama G, Marques D, Martin R, Takei H, Rodriguez-Agudo D, LaSalle SA, Hashiguchi T, Liu X, Green R, Erickson S, Gil G, Fuchs M, Suzuki M, Murai T, Nittono H, Hylemon PB, Zhou H, Pandak WM. Insulin resistance dysregulates CYP7B1 leading to oxysterol accumulation: a pathway for NAFL to NASH transition. J Lipid Res 2020; 61:1629-1644. [PMID: 33008924 PMCID: PMC7707165 DOI: 10.1194/jlr.ra120000924] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
NAFLD is an important public health issue closely associated with the pervasive epidemics of diabetes and obesity. Yet, despite NAFLD being among the most common of chronic liver diseases, the biological factors responsible for its transition from benign nonalcoholic fatty liver (NAFL) to NASH remain unclear. This lack of knowledge leads to a decreased ability to find relevant animal models, predict disease progression, or develop clinical treatments. In the current study, we used multiple mouse models of NAFLD, human correlation data, and selective gene overexpression of steroidogenic acute regulatory protein (StarD1) in mice to elucidate a plausible mechanistic pathway for promoting the transition from NAFL to NASH. We show that oxysterol 7α-hydroxylase (CYP7B1) controls the levels of intracellular regulatory oxysterols generated by the "acidic/alternative" pathway of cholesterol metabolism. Specifically, we report data showing that an inability to upregulate CYP7B1, in the setting of insulin resistance, results in the accumulation of toxic intracellular cholesterol metabolites that promote inflammation and hepatocyte injury. This metabolic pathway, initiated and exacerbated by insulin resistance, offers insight into approaches for the treatment of NAFLD.
Collapse
Affiliation(s)
- Genta Kakiyama
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA.
| | - Dalila Marques
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA
| | - Rebecca Martin
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | - Hajime Takei
- Junshin Clinic Bile Acid Institute, Tokyo, Japan
| | - Daniel Rodriguez-Agudo
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA
| | - Sandra A LaSalle
- Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA
| | | | - Xiaoying Liu
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Richard Green
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Sandra Erickson
- School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Gregorio Gil
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael Fuchs
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Tsuyoshi Murai
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Hokkaido, Japan
| | | | - Phillip B Hylemon
- Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Huiping Zhou
- Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - William M Pandak
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Veterans Affairs, McGuire Veterans Administration Medical Center, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
7
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
8
|
Fuior EV, Gafencu AV. Apolipoprotein C1: Its Pleiotropic Effects in Lipid Metabolism and Beyond. Int J Mol Sci 2019; 20:ijms20235939. [PMID: 31779116 PMCID: PMC6928722 DOI: 10.3390/ijms20235939] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
Apolipoprotein C1 (apoC1), the smallest of all apolipoproteins, participates in lipid transport and metabolism. In humans, APOC1 gene is in linkage disequilibrium with APOE gene on chromosome 19, a proximity that spurred its investigation. Apolipoprotein C1 associates with triglyceride-rich lipoproteins and HDL and exchanges between lipoprotein classes. These interactions occur via amphipathic helix motifs, as demonstrated by biophysical studies on the wild-type polypeptide and representative mutants. Apolipoprotein C1 acts on lipoprotein receptors by inhibiting binding mediated by apolipoprotein E, and modulating the activities of several enzymes. Thus, apoC1 downregulates lipoprotein lipase, hepatic lipase, phospholipase A2, cholesterylester transfer protein, and activates lecithin-cholesterol acyl transferase. By controlling the plasma levels of lipids, apoC1 relates directly to cardiovascular physiology, but its activity extends beyond, to inflammation and immunity, sepsis, diabetes, cancer, viral infectivity, and-not last-to cognition. Such correlations were established based on studies using transgenic mice, associated in the recent years with GWAS, transcriptomic and proteomic analyses. The presence of a duplicate gene, pseudogene APOC1P, stimulated evolutionary studies and more recently, the regulatory properties of the corresponding non-coding RNA are steadily emerging. Nonetheless, this prototypical apolipoprotein is still underexplored and deserves further research for understanding its physiology and exploiting its therapeutic potential.
Collapse
Affiliation(s)
- Elena V. Fuior
- Institute of Cellular Biology and Pathology “N. Simionescu”, 050568 Bucharest, Romania;
| | - Anca V. Gafencu
- Institute of Cellular Biology and Pathology “N. Simionescu”, 050568 Bucharest, Romania;
- Correspondence:
| |
Collapse
|
9
|
Lee SX, Heine M, Schlein C, Ramakrishnan R, Liu J, Belnavis G, Haimi I, Fischer AW, Ginsberg HN, Heeren J, Rinninger F, Haeusler RA. FoxO transcription factors are required for hepatic HDL cholesterol clearance. J Clin Invest 2018; 128:1615-1626. [PMID: 29408809 DOI: 10.1172/jci94230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022] Open
Abstract
Insulin resistance and type 2 diabetes are associated with low levels of high-density lipoprotein cholesterol (HDL-C). The insulin-repressible FoxO transcription factors are potential mediators of the effect of insulin on HDL-C. FoxOs mediate a substantial portion of insulin-regulated transcription, and poor FoxO repression is thought to contribute to the excessive glucose production in diabetes. In this work, we show that mice with liver-specific triple FoxO knockout (L-FoxO1,3,4), which are known to have reduced hepatic glucose production, also have increased HDL-C. This was associated with decreased expression of the HDL-C clearance factors scavenger receptor class B type I (SR-BI) and hepatic lipase and defective selective uptake of HDL cholesteryl ester by the liver. The phenotype could be rescued by re-expression of SR-BI. These findings demonstrate that hepatic FoxOs are required for cholesterol homeostasis and HDL-mediated reverse cholesterol transport to the liver.
Collapse
Affiliation(s)
- Samuel X Lee
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Rajasekhar Ramakrishnan
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Jing Liu
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Gabriella Belnavis
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Ido Haimi
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Alexander W Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Henry N Ginsberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Franz Rinninger
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,Department of Internal Medicine III, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
10
|
Sun H, Krauss RM, Chang JT, Teng BB. PCSK9 deficiency reduces atherosclerosis, apolipoprotein B secretion, and endothelial dysfunction. J Lipid Res 2018; 59:207-223. [PMID: 29180444 PMCID: PMC5794417 DOI: 10.1194/jlr.m078360] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/22/2017] [Indexed: 01/05/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) interacts directly with cytoplasmic apoB and prevents its degradation via the autophagosome/lysosome pathway. This process affects VLDL and LDL production and influences atherogenesis. Here, we investigated the molecular machinery by which PCSK9 modulates autophagy and affects atherogenesis. We backcrossed Pcsk9-/- mice with atherosclerosis-prone Ldlr-/-Apobec1-/- (LDb) mice to generate Ldlr-/-Apobec1-/-Pcsk9-/- (LTp) mice. Deletion of PCSK9 resulted in decreased hepatic apoB secretion, increased autophagic flux, and decreased plasma levels of IDL and LDL particles. The LDLs from LTp mice (LTp-LDLs) were less atherogenic and contained less cholesteryl ester and phospholipids than LDb-LDLs. Moreover LTp-LDLs induced lower endothelial expression of the genes encoding TLR2, Lox-1, ICAM-1, CCL2, CCL7, IL-6, IL-1β, Beclin-1, p62, and TRAF6 Collectively, these effects were associated with substantially less atherosclerosis development (>4-fold) in LTp mice. The absence of PCSK9 in LDb mice results in decreased lipid and apoB levels, fewer atherogenic LDLs, and marked reduction of atherosclerosis. The effect on atherogenesis may be mediated in part by the effects of modified LDLs on endothelial cell receptors and proinflammatory and autophagy molecules. These findings suggest that there may be clinical benefits of PCSK9 inhibition due to mechanisms unrelated to increased LDL receptor activity.
Collapse
Affiliation(s)
- Hua Sun
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine University of Texas Health Science Center at Houston, Houston, TX
| | | | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX
- University of Texas MD Anderson Cancer Center, University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX
| | - Ba-Bie Teng
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine University of Texas Health Science Center at Houston, Houston, TX
- University of Texas MD Anderson Cancer Center, University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX
| |
Collapse
|
11
|
Gordon SM, Li H, Zhu X, Tso P, Reardon CA, Shah AS, Lu LJ, Davidson WS. Impact of genetic deletion of platform apolipoproteins on the size distribution of the murine lipoproteome. J Proteomics 2016; 146:184-94. [PMID: 27385375 DOI: 10.1016/j.jprot.2016.06.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/01/2016] [Accepted: 06/29/2016] [Indexed: 01/16/2023]
Abstract
UNLABELLED Given their association with cardiovascular disease protection, there has been intense interest in understanding the biology of high density lipoproteins (HDL). HDL is actually a family of diverse particle types, each made up of discrete - but as yet undetermined - combinations of proteins drawn from up to 95 lipophilic plasma proteins. The abundant apolipoproteins (apo) of the A class (apoA-I, apoA-II and apoA-IV) have been proposed to act as organizing platforms for auxiliary proteins, but this concept has not been systematically evaluated. We assessed the impact of genetic knock down of each platform protein on the particle size distribution of auxiliary HDL proteins. Loss of apoA-I or apoA-II massively reduced HDL lipids and changed the plasma size pattern and/or abundance of several plasma proteins. Surprisingly though, many HDL proteins were not affected, suggesting they assemble on lipid particles in the absence of apoA-I or apoA-II. In contrast, apoA-IV ablation had minor effects on plasma lipids and proteins, suggesting that it forms particles that largely exclude other apolipoproteins. Overall, the data indicate that distinct HDL subpopulations exist that do not contain, nor depend on, apoA-I, apoA-II or apoA-IV and these contribute substantially to the proteomic diversity of HDL. BIOLOGICAL SIGNIFICANCE Plasma levels of high density lipoproteins (HDL) are inversely correlated with cardiovascular disease. These particles are becoming known as highly heterogeneous entities that have diverse compositions and functions that may impact disease. Unfortunately, we know little about the forces that maintain the composition of each particle in plasma. It has been suggested that certain 'scaffold' proteins, such as apolipoprotein (apo) A-I, apoA-II and apoA-IV, may act as organizing centers for the docking of myriad accessory proteins. To test this hypothesis, we took advantage of the genetic tractability of the mouse model and ablated these three proteins individually. We then tracked the abundance and size profile of the remaining HDL proteins by gel filtration chromatography combined with mass spectrometry. The results clearly show that certain cohorts of proteins depend on each scaffold molecule to assemble normal sized HDL particles under wild-type conditions. This work forms the basis for more detailed studies that will define the specific compositions of HDL subspecies with the possibility of connecting them to specific functions or roles in disease.
Collapse
Affiliation(s)
- Scott M Gordon
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, OH 45237-0507, USA.
| | - Hailong Li
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, OH 45229-3039, USA.
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, OH 45229-3039, USA.
| | - Patrick Tso
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, OH 45237-0507, USA.
| | | | - Amy S Shah
- Department of Pediatrics, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, MLC 7012, Cincinnati, OH 45229-3039, USA.
| | - L Jason Lu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, OH 45229-3039, USA.
| | - W Sean Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, OH 45237-0507, USA.
| |
Collapse
|
12
|
Link JC, Chen X, Prien C, Borja MS, Hammerson B, Oda MN, Arnold AP, Reue K. Increased high-density lipoprotein cholesterol levels in mice with XX versus XY sex chromosomes. Arterioscler Thromb Vasc Biol 2015; 35:1778-86. [PMID: 26112012 DOI: 10.1161/atvbaha.115.305460] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/08/2015] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The molecular mechanisms underlying sex differences in dyslipidemia are poorly understood. We aimed to distinguish genetic and hormonal regulators of sex differences in plasma lipid levels. APPROACH AND RESULTS We assessed the role of gonadal hormones and sex chromosome complement on lipid levels using the four core genotypes mouse model (XX females, XX males, XY females, and XY males). In gonadally intact mice fed a chow diet, lipid levels were influenced by both male-female gonadal sex and XX-XY chromosome complement. Gonadectomy of adult mice revealed that the male-female differences are dependent on acute effects of gonadal hormones. In both intact and gonadectomized animals, XX mice had higher HDL cholesterol (HDL-C) levels than XY mice, regardless of male-female sex. Feeding a cholesterol-enriched diet produced distinct patterns of sex differences in lipid levels compared with a chow diet, revealing the interaction of gonadal and chromosomal sex with diet. Notably, under all dietary and gonadal conditions, HDL-C levels were higher in mice with 2 X chromosomes compared with mice with an X and Y chromosome. By generating mice with XX, XY, and XXY chromosome complements, we determined that the presence of 2 X chromosomes, and not the absence of the Y chromosome, influences HDL-C concentration. CONCLUSIONS We demonstrate that having 2 X chromosomes versus an X and Y chromosome complement drives sex differences in HDL-C. It is conceivable that increased expression of genes escaping X-inactivation in XX mice regulates downstream processes to establish sexual dimorphism in plasma lipid levels.
Collapse
Affiliation(s)
- Jenny C Link
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Xuqi Chen
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Christopher Prien
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Mark S Borja
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Bradley Hammerson
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Michael N Oda
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Arthur P Arnold
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Karen Reue
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.).
| |
Collapse
|
13
|
Gordon SM, Li H, Zhu X, Shah AS, Lu LJ, Davidson WS. A comparison of the mouse and human lipoproteome: suitability of the mouse model for studies of human lipoproteins. J Proteome Res 2015; 14:2686-95. [PMID: 25894274 PMCID: PMC4712022 DOI: 10.1021/acs.jproteome.5b00213] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Plasma levels of low density lipoproteins (LDL) and high density lipoproteins (HDL) exhibit opposing associations with cardiovascular disease in human populations and mouse models have been heavily used to derive a mechanistic understanding of these relationships. In humans, recent mass spectrometry studies have revealed that the plasma lipoproteome is significantly more complex than originally appreciated. This is particularly true for HDL which contains some 90 distinct proteins, a majority of which play functional roles that go beyond those expected for simple lipid transport. Unfortunately, the mouse lipoproteome remains largely uncharacterized-a significant gap given the heavy reliance on the model. Using a gel filtration chromatography and mass spectrometry analysis that targets phospholipid-bound plasma proteins, we compared the mouse lipoproteome and its size distribution to a previous, identical human analysis. We identified 113 lipid associated proteins in the mouse. In general, the protein diversity in the LDL and HDL size ranges was similar in mice versus humans, though some distinct differences were noted. For the majority of proteins, the size distributions, that is, whether a given protein was associated with large versus small HDL particles, for example, were also similar between species. Again, however, there were clear differences exhibited by a minority of proteins that may reflect metabolic differences between species. Finally, by correlating the lipid and protein size profiles, we identified five proteins that closely track with the major HDL protein, apolipoprotein A-I across both species. Thus, mice have most of the minor proteins identified in human lipoproteins that play key roles in inflammation, innate immunity, proteolysis and its inhibition, and vitamin transport. This provides support for the continued use of the mouse as a model for many aspects of human lipoprotein metabolism.
Collapse
Affiliation(s)
- Scott M. Gordon
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, Ohio 45237-0507, United States
| | - Hailong Li
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - Amy S. Shah
- Department of Pediatrics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7012, Cincinnati, Ohio 45229-3039, United States
| | - L. Jason Lu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - W. Sean Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, Ohio 45237-0507, United States
| |
Collapse
|
14
|
Fotakis P, Kuivenhoven JA, Dafnis E, Kardassis D, Zannis VI. The Effect of Natural LCAT Mutations on the Biogenesis of HDL. Biochemistry 2015; 54:3348-59. [PMID: 25948084 DOI: 10.1021/acs.biochem.5b00180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have investigated how the natural LCAT[T147I] and LCAT[P274S] mutations affect the pathway of biogenesis of HDL. Gene transfer of WT LCAT in LCAT(-/-) mice increased 11.8-fold the plasma cholesterol, whereas the LCAT[T147I] and LCAT[P274S] mutants caused a 5.2- and 2.9-fold increase, respectively. The LCAT[P274S] and the WT LCAT caused a monophasic distribution of cholesterol in the HDL region, whereas the LCAT[T147I] caused a biphasic distribution of cholesterol in the LDL and HDL region. Fractionation of plasma showed that the expression of WT LCAT increased plasma apoE and apoA-IV levels and shifted the distribution of apoA-I to lower densities. The LCAT[T147I] and LCAT[P274S] mutants restored partially apoA-I in the HDL3 fraction and LCAT[T147I] increased apoE in the VLD/IDL/LDL fractions. The in vivo functionality of LCAT was further assessed based on is its ability to correct the aberrant HDL phenotype that was caused by the apoA-I[L159R]FIN mutation. Co-infection of apoA-I(-/-) mice with this apoA-I mutant and either of the two mutant LCAT forms restored only partially the HDL biogenesis defect that was caused by the apoA-I[L159R]FIN and generated a distinct aberrant HDL phenotype.
Collapse
Affiliation(s)
- Panagiotis Fotakis
- †Molecular Genetics, Boston University School of Medicine, 700 Albany Street, W509, Boston, Massachusetts 02118-2394, United States.,‡Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, GR-74100 Heraklion, Greece
| | - Jan Albert Kuivenhoven
- §Department of Pediatrics, Section Molecular Genetics, Groningen, University of Groningen, University Medical Center Groningen, 9700 Groningen, The Netherlands
| | - Eugene Dafnis
- ∥Department of Nephrology, University of Crete Medical School, GR-74100 Heraklion, Greece
| | - Dimitris Kardassis
- ‡Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, GR-74100 Heraklion, Greece
| | - Vassilis I Zannis
- †Molecular Genetics, Boston University School of Medicine, 700 Albany Street, W509, Boston, Massachusetts 02118-2394, United States
| |
Collapse
|
15
|
Kootte RS, Smits LP, van der Valk FM, Dasseux JL, Keyserling CH, Barbaras R, Paolini JF, Santos RD, van Dijk TH, Dallinga-van Thie GM, Nederveen AJ, Mulder WM, Hovingh GK, Kastelein JP, Groen AK, Stroes E. Effect of open-label infusion of an apoA-I-containing particle (CER-001) on RCT and artery wall thickness in patients with FHA. J Lipid Res 2015; 56:703-712. [PMID: 25561459 DOI: 10.1194/jlr.m055665] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reverse cholesterol transport (RCT) contributes to the anti-atherogenic effects of HDL. Patients with the orphan disease, familial hypoalphalipoproteinemia (FHA), are characterized by decreased tissue cholesterol removal and an increased atherogenic burden. We performed an open-label uncontrolled proof-of-concept study to evaluate the effect of infusions with a human apoA-I-containing HDL-mimetic particle (CER-001) on RCT and the arterial vessel wall in FHA. Subjects received 20 infusions of CER-001 (8 mg/kg) during 6 months. Efficacy was assessed by measuring (apo)lipoproteins, plasma-mediated cellular cholesterol efflux, fecal sterol excretion (FSE), and carotid artery wall dimension by MRI and artery wall inflammation by (18)F-fluorodeoxyglucose-positron emission tomography/computed tomography scans. We included seven FHA patients: HDL-cholesterol (HDL-c), 13.8 [1.8-29.1] mg/dl; apoA-I, 28.7 [7.9-59.1] mg/dl. Following nine infusions in 1 month, apoA-I and HDL-c increased directly after infusion by 27.0 and 16.1 mg/dl (P = 0.018). CER-001 induced a 44% relative increase (P = 0.018) in in vitro cellular cholesterol efflux with a trend toward increased FSE (P = 0.068). After nine infusions of CER-001, carotid mean vessel wall area decreased compared with baseline from 25.0 to 22.8 mm(2) (P = 0.043) and target-to-background ratio from 2.04 to 1.81 (P = 0.046). In FHA-subjects, CER-001 stimulates cholesterol mobilization and reduces artery wall dimension and inflammation, supporting further evaluation of CER-001 in FHA patients.
Collapse
Affiliation(s)
- Ruud S Kootte
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | - Loek P Smits
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | - Fleur M van der Valk
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | - Raul D Santos
- Heart Institute (Incor), University of Sao Paolo Medical School Hospital, Sao Paulo, Brazil
| | - Theo H van Dijk
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Geesje M Dallinga-van Thie
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | | | - WillemJ M Mulder
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | - G Kees Hovingh
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | - JohnJ P Kastelein
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - ErikS Stroes
- Departments of Vascular Medicine and Experimental Vascular Medicine Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Sips FLP, Tiemann CA, Oosterveer MH, Groen AK, Hilbers PAJ, van Riel NAW. A computational model for the analysis of lipoprotein distributions in the mouse: translating FPLC profiles to lipoprotein metabolism. PLoS Comput Biol 2014; 10:e1003579. [PMID: 24784354 PMCID: PMC4006703 DOI: 10.1371/journal.pcbi.1003579] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 03/11/2014] [Indexed: 12/27/2022] Open
Abstract
Disturbances of lipoprotein metabolism are recognized as indicators of cardiometabolic disease risk. Lipoprotein size and composition, measured in a lipoprotein profile, are considered to be disease risk markers. However, the measured profile is a collective result of complex metabolic interactions, which complicates the identification of changes in metabolism. In this study we aim to develop a method which quantitatively relates murine lipoprotein size, composition and concentration to the molecular mechanisms underlying lipoprotein metabolism. We introduce a computational framework which incorporates a novel kinetic model of murine lipoprotein metabolism. The model is applied to compute a distribution of plasma lipoproteins, which is then related to experimental lipoprotein profiles through the generation of an in silico lipoprotein profile. The model was first applied to profiles obtained from wild-type C57Bl/6J mice. The results provided insight into the interplay of lipoprotein production, remodelling and catabolism. Moreover, the concentration and metabolism of unmeasured lipoprotein components could be determined. The model was validated through the prediction of lipoprotein profiles of several transgenic mouse models commonly used in cardiovascular research. Finally, the framework was employed for longitudinal analysis of the profiles of C57Bl/6J mice following a pharmaceutical intervention with a liver X receptor (LXR) agonist. The multifaceted regulatory response to the administration of the compound is incompletely understood. The results explain the characteristic changes of the observed lipoprotein profile in terms of the underlying metabolic perturbation and resultant modifications of lipid fluxes in the body. The Murine Lipoprotein Profiler (MuLiP) presented here is thus a valuable tool to assess the metabolic origin of altered murine lipoprotein profiles and can be applied in preclinical research performed in mice for analysis of lipid fluxes and lipoprotein composition.
Collapse
Affiliation(s)
- Fianne L P Sips
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Netherlands Consortium for Systems Biology, University of Amsterdam, Amsterdam, The Netherlands
| | - Christian A Tiemann
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Netherlands Consortium for Systems Biology, University of Amsterdam, Amsterdam, The Netherlands
| | - Maaike H Oosterveer
- Department of Pediatrics, University Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Netherlands Consortium for Systems Biology, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatrics, University Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter A J Hilbers
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Netherlands Consortium for Systems Biology, University of Amsterdam, Amsterdam, The Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Netherlands Consortium for Systems Biology, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Pedrelli M, Davoodpour P, Degirolamo C, Gomaraschi M, Graham M, Ossoli A, Larsson L, Calabresi L, Gustafsson JÅ, Steffensen KR, Eriksson M, Parini P. Hepatic ACAT2 knock down increases ABCA1 and modifies HDL metabolism in mice. PLoS One 2014; 9:e93552. [PMID: 24695360 PMCID: PMC3973598 DOI: 10.1371/journal.pone.0093552] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/06/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES ACAT2 is the exclusive cholesterol-esterifying enzyme in hepatocytes and enterocytes. Hepatic ABCA1 transfers unesterified cholesterol (UC) to apoAI, thus generating HDL. By changing the hepatic UC pool available for ABCA1, ACAT2 may affect HDL metabolism. The aim of this study was to reveal whether hepatic ACAT2 influences HDL metabolism. DESIGN WT and LXRα/β double knockout (DOKO) mice were fed a western-type diet for 8 weeks. Animals were i.p. injected with an antisense oligonucleotide targeted to hepatic ACAT2 (ASO6), or with an ASO control. Injections started 4 weeks after, or concomitantly with, the beginning of the diet. RESULTS ASO6 reduced liver cholesteryl esters, while not inducing UC accumulation. ASO6 increased hepatic ABCA1 protein independently of the diet conditions. ASO6 affected HDL lipids (increased UC) only in DOKO, while it increased apoE-containing HDL in both genotypes. In WT mice ASO6 led to the appearance of large HDL enriched in apoAI and apoE. CONCLUSIONS The use of ASO6 revealed a new pathway by which the liver may contribute to HDL metabolism in mice. ACAT2 seems to be a hepatic player affecting the cholesterol fluxes fated to VLDL or to HDL, the latter via up-regulation of ABCA1.
Collapse
Affiliation(s)
- Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Padideh Davoodpour
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Degirolamo
- Division of Lipid Science, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Monica Gomaraschi
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Mark Graham
- Cardiovascular Group, Department of Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Alice Ossoli
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Lilian Larsson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laura Calabresi
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Jan-Åke Gustafsson
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, United States of America
| | - Knut R. Steffensen
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mats Eriksson
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Paolo Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Spradley FT, De Miguel C, Hobbs J, Pollock DM, Pollock JS. Mycophenolate mofetil prevents high-fat diet-induced hypertension and renal glomerular injury in Dahl SS rats. Physiol Rep 2013; 1:e00137. [PMID: 24400139 PMCID: PMC3871452 DOI: 10.1002/phy2.137] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/23/2013] [Accepted: 10/02/2013] [Indexed: 01/12/2023] Open
Abstract
We designed experiments to test the hypothesis that Dahl salt-sensitive (SS) rats are sensitive to high-fat diet (HFD)–induced hypertension and renal injury via an inflammatory mechanism. Twelve-week-old Dahl SS rats were maintained on a normal diet (ND; 14% fat), HFD (59% fat), or HFD supplemented with the lymphocyte immunosuppressive agent, mycophenolate mofetil (HFD + MMF; 30 mg/kg/day orally in diet), for a period of 4 weeks. Mean arterial pressure (MAP), metabolic parameters, T lymphocyte (CD3+) localization, and renal structural damage were assessed during the studies. Four weeks of HFD significantly elevated MAP and visceral adiposity without changing circulating levels of lipids or adipokines. Immunohistochemical analysis demonstrated that SS rats on HFD had significantly greater numbers of CD3+ cells in renal glomerular and medullary areas compared to ND SS rats. Additionally, HFD led to increased glomerular injury, but did not alter renal medullary injury. Chronic MMF treatment in HFD-fed Dahl SS rats reduced MAP, visceral adiposity, infiltration of CD3+ cells in the glomerulus, as well as glomerular injury. However, MMF treatment did not alter HFD-induced infiltration of CD3+ cells in the renal medulla. In conclusion, Dahl SS rats are sensitized to HFD-induced hypertension and renal glomerular injury via infiltration of T lymphocytes.
Collapse
Affiliation(s)
- Frank T Spradley
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University Augusta, 30912, Georgia
| | - Carmen De Miguel
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University Augusta, 30912, Georgia
| | - Janet Hobbs
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University Augusta, 30912, Georgia
| | - David M Pollock
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University Augusta, 30912, Georgia
| | - Jennifer S Pollock
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University Augusta, 30912, Georgia
| |
Collapse
|
19
|
Food restriction by intermittent fasting induces diabetes and obesity and aggravates spontaneous atherosclerosis development in hypercholesterolaemic mice. Br J Nutr 2013; 111:979-86. [DOI: 10.1017/s0007114513003383] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Different regimens of food restriction have been associated with protection against obesity, diabetes and CVD. In the present study, we hypothesised that food restriction would bring benefits to atherosclerosis- and diabetes-prone hypercholesterolaemic LDL-receptor knockout mice. For this purpose, 2-month-old mice were submitted to an intermittent fasting (IF) regimen (fasting every other day) over a 3-month period, which resulted in an overall 20 % reduction in food intake. Contrary to our expectation, epididymal and carcass fat depots and adipocyte size were significantly enlarged by 15, 72 and 68 %, respectively, in the IF mice compared with the ad libitum-fed mice. Accordingly, plasma levels of leptin were 50 % higher in the IF mice than in the ad libitum-fed mice. In addition, the IF mice showed increased plasma levels of total cholesterol (37 %), VLDL-cholesterol (195 %) and LDL-cholesterol (50 %). As expected, in wild-type mice, the IF regimen decreased plasma cholesterol levels and epididymal fat mass. Glucose homeostasis was also disturbed by the IF regimen in LDL-receptor knockout mice. Elevated levels of glycaemia (40 %), insulinaemia (50 %), glucose intolerance and insulin resistance were observed in the IF mice. Systemic inflammatory markers, TNF-α and C-reactive protein, were significantly increased and spontaneous atherosclerosis development were markedly increased (3-fold) in the IF mice. In conclusion, the IF regimen induced obesity and diabetes and worsened the development of spontaneous atherosclerosis in LDL-receptor knockout mice. Although being efficient in a wild-type background, this type of food restriction is not beneficial in the context of genetic hypercholesterolaemia.
Collapse
|
20
|
Swindell WR. Dietary restriction in rats and mice: a meta-analysis and review of the evidence for genotype-dependent effects on lifespan. Ageing Res Rev 2012; 11:254-70. [PMID: 22210149 PMCID: PMC3299887 DOI: 10.1016/j.arr.2011.12.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/06/2011] [Indexed: 01/20/2023]
Abstract
Laboratory survival experiments have shown that dietary restriction (DR) can increase median and maximum lifespan. This paper provides a meta-analysis of laboratory experiments that have evaluated the effects of DR on lifespan in rats and mice (1934-present). In rats, DR increased median lifespan by 14-45% in half of all experiments, but in mice the effects of DR have been much weaker (4-27%). The least favorable effects of DR on lifespan have been observed among inbred rather than non-inbred mouse strains. In fact, some inbred mouse strains do not necessarily live longer with DR, including DBA/2 male mice and several strains from the ILSXISS recombinant inbred panel. Shortening of lifespan with DR has also been observed and confirmed for ILSXISS strain 114. Importantly, all rodent studies may be biased by the effects of laboratory breeding, since one study has shown that median lifespan is not improved by DR in wild-derived mice. These findings suggest that the set of genetic backgrounds studied in rodent DR experiments should be diversified. This will broaden the scope of genotypes studied in aging research, but may also be critical for translation of findings from rodents to historically outbred and genetically heterogeneous primate species.
Collapse
Affiliation(s)
- William R Swindell
- Department of Genetics, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
21
|
The apolipoprotein C-I content of very-low-density lipoproteins is associated with fasting triglycerides, postprandial lipemia, and carotid atherosclerosis. J Lipids 2011; 2011:271062. [PMID: 21776394 PMCID: PMC3139117 DOI: 10.1155/2011/271062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Accepted: 05/02/2011] [Indexed: 11/26/2022] Open
Abstract
Background. Experimental studies in animals suggest that apolipoprotein (apo) C-I is an important regulator of triglycerides in fasting and postprandial conditions and associated with carotid atherosclerosis.
Methods. A cross-sectional study was conducted with 81 subjects, aged 56–80 years recruited from a population health survey. The participants underwent a fat tolerance test (1 g fat per Kg body weight) and carotid atherosclerosis was determined by ultrasound examination. VLDL particles, Sf 20–400, were isolated and their lipid composition and apoC-I content determined.
Results. The carotid plaque area increased linearly with the number of apoC-I molecules per VLDL particles (P = 0.048) under fasting conditions. Fasting triglycerides increased across tertiles of apoC-I per VLDL particle in analyses adjusted for apoC-II and -C-III, apoE genotype and traditional cardiovascular risk factors (P = 0.011). The relation between apoC-I in VLDL and serum triglycerides was conveyed by triglyceride enrichment of VLDL particles (P for trend <0.001. The amount of apoC-I molecules per VLDL was correlated with the total (r = 0.41, P < 0.0001) and incremental (r = 0.35, P < 0.001) area under the postprandial triglyceride curve.
Conclusions. Our findings support the concept that the content of apoC-I per VLDL particle is an important regulator of triglyceride metabolism in the fasting and postprandial state and associated with carotid athrosclerosis.
Collapse
|
22
|
Wang Y, Berbée JFP, Stroes ES, Smit JWA, Havekes LM, Romijn JA, Rensen PCN. CETP expression reverses the reconstituted HDL-induced increase in VLDL. J Lipid Res 2011; 52:1533-41. [PMID: 21606464 DOI: 10.1194/jlr.m016659] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Human data suggest that reconstituted HDL (rHDL) infusion can induce atherosclerosis regression. Studies in mice indicated that rHDL infusion adversely affects VLDL levels, but this effect is less apparent in humans. This discrepancy may be explained by the fact that humans, in contrast to mice, express cholesteryl ester transfer protein (CETP). The aim of this study was to investigate the role of CETP in the effects of rHDL on VLDL metabolism by using APOE*3-Leiden (E3L) mice, a well-established model for human-like lipoprotein metabolism. At 1 h after injection, rHDL increased plasma VLDL-C and TG in E3L mice, but not in E3L mice cross-bred onto a human CETP background (E3L.CETP mice). This initial raise in VLDL, caused by competition between rHDL and VLDL for LPL-mediated TG hydrolysis, was thus prevented by CETP. At 24 h after injection, rHDL caused a second increase in VLDL-C and TG in E3L mice, whereas rHDL had even decreased VLDL in E3L.CETP mice. This secondary raise in VLDL was due to increased hepatic VLDL-TG production. Collectively, we conclude that CETP protects against the rHDL-induced increase in VLDL. We anticipate that studies evaluating the anti-atherosclerotic efficacy of rHDL in mice that are naturally deficient for CETP should be interpreted with caution, and that treatment of atherogenic dyslipidemia by rHDL should not be combined with agents that aggressively reduce CETP activity.
Collapse
Affiliation(s)
- Yanan Wang
- Department of General Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
23
|
Koornneef A, Maczuga P, van Logtenstein R, Borel F, Blits B, Ritsema T, van Deventer S, Petry H, Konstantinova P. Apolipoprotein B knockdown by AAV-delivered shRNA lowers plasma cholesterol in mice. Mol Ther 2011; 19:731-40. [PMID: 21304496 PMCID: PMC3070114 DOI: 10.1038/mt.2011.6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 01/10/2011] [Indexed: 12/22/2022] Open
Abstract
Serum low-density lipoprotein cholesterol (LDL-C) levels are proportionate to the risk of atherosclerotic cardiovascular disease. In order to reduce serum total cholesterol and LDL-C levels in mice, RNA interference (RNAi) was used to inhibit expression of the structural protein of LDL-C, apolipoprotein B100 (ApoB). We developed and screened 19 short hairpin RNAs (shRNAs) targeting conserved sequences in human, mouse, and macaque ApoB mRNAs (shApoB) and subsequently narrowed our focus to one candidate for in vivo testing. Self-complementary adeno-associated virus serotype 8 (scAAV8) was used for long-term transduction of murine liver with shApoB. A strong dose-dependent knockdown of ApoB mRNA and protein was observed, which correlated with a reduction in total cholesterol levels, without obvious signs of toxicity. Furthermore, shApoB was found to specifically reduce LDL-C in diet-induced dyslipidemic mice, whereas high-density lipoprotein cholesterol (HDL-C) remained unaffected. Finally, elevated lipid accumulation was shown in murine liver transduced with shApoB, a known phenotypic side effect of lowering ApoB levels. These results demonstrate a robust dose-dependent knockdown of ApoB by AAV-delivered shRNA in murine liver, thus providing an excellent candidate for development of RNAi-based gene therapy for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Annemart Koornneef
- Department of Research and Development, Amsterdam Molecular Therapeutics, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bighetti EJB, Patrício PR, Casquero AC, Berti JA, Oliveira HCF. Ciprofibrate increases cholesteryl ester transfer protein gene expression and the indirect reverse cholesterol transport to the liver. Lipids Health Dis 2009; 8:50. [PMID: 19930639 PMCID: PMC2784759 DOI: 10.1186/1476-511x-8-50] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 11/23/2009] [Indexed: 11/26/2022] Open
Abstract
Background CETP is a plasma protein that modulates atherosclerosis risk through its HDL-cholesterol reducing action. The aim of this work was to examine the effect of the PPARα agonist, ciprofibrate, on the CETP gene expression, in the presence and absence of apolipoprotein (apo) CIII induced hypertriglyceridemia, and its impact on the HDL metabolism. Results Mice expressing apo CIII and/or CETP and non-transgenic littermates (CIII, CIII/CETP, CETP, non-Tg) were treated with ciprofibrate during 3 weeks. Drug treatment reduced plasma triglycerides (30-43%) and non-esterified fatty acids (19-47%) levels. Cholesterol (chol) distribution in plasma lipoprotein responses to ciprofibrate treatment was dependent on the genotypes. Treated CIII expressing mice presented elevation in VLDL-chol and reduction in HDL-chol. Treated CETP expressing mice responded with reduction in LDL-chol whereas in non-Tg mice the LDL-chol increased. In addition, ciprofibrate increased plasma post heparin lipoprotein lipase activity (1.3-2.1 fold) in all groups but hepatic lipase activity decreased in treated CETP and non-Tg mice. Plasma CETP activity and liver CETP mRNA levels were significantly increased in treated CIII/CETP and CETP mice (30-100%). Kinetic studies with 3H-cholesteryl ether (CEt) labelled HDL showed a 50% reduction in the 3H-CEt found in the LDL fraction in ciprofibrate treated compared to non-treated CETP mice. This means that 3H-CEt transferred from HDL to LDL was more efficiently removed from the plasma in the fibrate treated mice. Accordingly, the amount of 3H-CEt recovered in the liver 6 hours after HDL injection was increased by 35%. Conclusion Together these data showed that the PPARα agonist ciprofibrate stimulates CETP gene expression and changes the cholesterol flow through the reverse cholesterol transport, increasing plasma cholesterol removal through LDL.
Collapse
Affiliation(s)
- Eliete J B Bighetti
- Physiology and Biophysics Division, Biology Institute, State University of Campinas, Campinas, SP, Brazil.
| | | | | | | | | |
Collapse
|
25
|
Wool GD, Vaisar T, Reardon CA, Getz GS. An apoA-I mimetic peptide containing a proline residue has greater in vivo HDL binding and anti-inflammatory ability than the 4F peptide. J Lipid Res 2009; 50:1889-900. [PMID: 19433476 DOI: 10.1194/jlr.m900151-jlr200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Modifying apolipoprotein (apo) A-I mimetic peptides to include a proline-punctuated alpha-helical repeat increases their anti-inflammatory properties as well as allows better mimicry of full-length apoA-I function. This study compares the following mimetics, either acetylated or biotinylated (b): 4F (18mer) and 4F-proline-4F (37mer, Pro). b4F interacts with both mouse HDL (moHDL) and LDL in vitro. b4F in vivo plasma clearance kinetics are not affected by mouse HDL level. Administration of biotinylated peptides to mice demonstrates that b4F does not associate with lipoproteins smaller than LDL in vivo, though it does associate with fractions containing free hemoglobin (Hb). In contrast, bPro specifically interacts with HDL. b4F and bPro show opposite binding responses to HDL by surface plasmon resonance. Administration of acetylated Pro to apoE(-/-) mice significantly decreases plasma serum amyloid A levels, while acetylated 4F does not have this ability. In contrast to previous reports that inferred that 4F associates with HDL in vivo, we systematically examined this potential interaction and demonstrated that b4F does not interact with HDL in vivo but rather elutes with Hb-containing plasma fractions. bPro, however, specifically binds to moHDL in vivo. In addition, the number of amphipathic alpha-helices and their linker influences the anti-inflammatory effects of apoA-I mimetic peptides in vivo.
Collapse
|
26
|
Human cholesteryl ester transfer protein expression enhances the mouse survival rate in an experimental systemic inflammation model: a novel role for CETP. Shock 2009; 30:590-5. [PMID: 18391856 DOI: 10.1097/shk.0b013e31816e30fd] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mice expressing human cholesteryl ester transfer protein (huCETP) are more resistant to Escherichia coli bacterial wall LPS because death rates 5 days after intraperitoneal inoculation of LPS were higher in wild-type than in huCETP+/+ mice, whereas all huCETP+/+ mice remained alive. After LPS inoculation, plasma concentrations of TNF-alpha and IL-6 increased less in huCETP+/+ than in wild-type mice. LPS in vitro elicited lower TNF-alpha production by CETP expressing than by wild-type macrophages. In addition, TNF-alpha production by RAW 264.7 murine macrophages increased on incubation with LPS but decreased in a dose-dependent manner when human CETP was added to the medium. Human CETP in vitro enhanced the LPS binding to plasma high-density lipoprotein/low-density lipoprotein. The liver uptake of intravenous infused 14C-LPS from Salmonella typhimurium was greater in huCETP+/+ than in wild-type mice. Present data indicate for the first time that CETP is an endogenous component involved in the first line of defense against an exacerbated production of proinflammatory mediators.
Collapse
|
27
|
Overactive endocannabinoid signaling impairs apolipoprotein E-mediated clearance of triglyceride-rich lipoproteins. Proc Natl Acad Sci U S A 2008; 105:14561-6. [PMID: 18794527 DOI: 10.1073/pnas.0807232105] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The endocannabinoid (EC) system regulates food intake and energy metabolism. Cannabinoid receptor type 1 (CB1) antagonists show promise in the treatment of obesity and its metabolic consequences. Although the reduction in adiposity resulting from therapy with CB1 antagonists may not account fully for the concomitant improvements in dyslipidemia, direct effects of overactive EC signaling on plasma lipoprotein metabolism have not been documented. The present study used a chemical approach to evaluate the direct effects of increased EC signaling in mice by inducing acute elevations of endogenously produced cannabinoids through pharmacological inhibition of their enzymatic hydrolysis by isopropyl dodecylfluorophosphonate (IDFP). Acute IDFP treatment increased plasma levels of triglyceride (TG) (2.0- to 3.1-fold) and cholesterol (1.3- to 1.4-fold) in conjunction with an accumulation in plasma of apolipoprotein (apo)E-depleted TG-rich lipoproteins. These changes did not occur in either CB1-null or apoE-null mice, were prevented by pretreatment with CB1 antagonists, and were not associated with reduced hepatic apoE gene expression. Although IDFP treatment increased hepatic mRNA levels of lipogenic genes (Srebp1 and Fas), there was no effect on TG secretion into plasma. Instead, IDFP treatment impaired clearance of an intravenously administered TG emulsion, despite increased postheparin lipoprotein lipase activity. Therefore, overactive EC signaling elicits an increase in plasma triglyceride levels associated with reduced plasma TG clearance and an accumulation in plasma of apoE-depleted TG-rich lipoproteins. These findings suggest a role of CB1 activation in the pathogenesis of obesity-related hypertriglyceridemia and underscore the potential efficacy of CB1 antagonists in treating metabolic disease.
Collapse
|
28
|
Barbosa CR, Albuquerque EMV, Faria EC, Oliveira HCF, Castilho LN. Opposite lipemic response of Wistar rats and C57BL/6 mice to dietary glucose or fructose supplementation. Braz J Med Biol Res 2008; 40:323-31. [PMID: 17334529 DOI: 10.1590/s0100-879x2007000300007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 12/18/2006] [Indexed: 11/22/2022] Open
Abstract
The metabolic effects of carbohydrate supplementation in mice have not been extensively studied. In rats, glucose- and fructose-rich diets induce hypertriacylglycerolemia. In the present study, we compared the metabolic responses to two monosaccharide supplementations in two murine models. Adult male Wistar rats (N = 80) and C57BL/6 mice (N = 60), after 3 weeks on a standardized diet, were submitted to dietary supplementation by gavage with glucose (G) or fructose (F) solutions (500 g/L), 8 g/kg body weight for 21 days. Glycemia was significantly higher in rats after fructose treatment (F: 7.9 vs 9.3 mM) and in mice (G: 6.5 vs 10 and F: 6.6 vs 8.9 mM) after both carbohydrate treatments. Triacylglycerolemia increased significantly 1.5 times in rats after G or F supplementation. Total cholesterol did not change with G treatment in rats, but did decrease after F supplementation (1.5 vs 1.4 mM, P < 0.05). Both supplementations in rats induced insulin resistance, as suggested by the higher Homeostasis Model Assessment Index. In contrast, mice showed significant decreases in triacylglycerol (G: 1.8 vs 1.4 and F: 1.9 vs 1.4 mM, P < 0.01) and total cholesterol levels (G and F: 2.7 vs 2.5 mM, P < 0.05) after both monosaccharide supplementations. Wistar rats and C57BL/6 mice, although belonging to the same family (Muridae), presented opposite responses to glucose and fructose supplementation regarding serum triacylglycerol, free fatty acids, and insulin levels after monosaccharide treatment. Thus, while Wistar rats developed features of plurimetabolic syndrome, C57BL/6 mice presented changes in serum biochemical profile considered to be healthier for the cardiovascular system.
Collapse
Affiliation(s)
- C R Barbosa
- Departamento de Patologia Clínica e Núcleo de Medicina e Cirurgia Experimental, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, 13084-971 Campinas, SP, Brazil
| | | | | | | | | |
Collapse
|
29
|
van der Hoogt CC, de Haan W, Westerterp M, Hoekstra M, Dallinga-Thie GM, Romijn JA, Princen HMG, Jukema JW, Havekes LM, Rensen PCN. Fenofibrate increases HDL-cholesterol by reducing cholesteryl ester transfer protein expression. J Lipid Res 2007; 48:1763-71. [PMID: 17525476 DOI: 10.1194/jlr.m700108-jlr200] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In addition to efficiently decreasing VLDL-triglycerides (TGs), fenofibrate increases HDL-cholesterol levels in humans. We investigated whether the fenofibrate-induced increase in HDL-cholesterol is dependent on the expression of the cholesteryl ester transfer protein (CETP). To this end, APOE*3-Leiden (E3L) transgenic mice without and with the human CETP transgene, under the control of its natural regulatory flanking regions, were fed a Western-type diet with or without fenofibrate. Fenofibrate (0.04% in the diet) decreased plasma TG in E3L and E3L.CETP mice (-59% and -60%; P < 0.001), caused by a strong reduction in VLDL. Whereas fenofibrate did not affect HDL-cholesterol in E3L mice, fenofibrate dose-dependently increased HDL-cholesterol in E3L.CETP mice (up to +91%). Fenofibrate did not affect the turnover of HDL-cholesteryl ester (CE), indicating that fenofibrate causes a higher steady-state HDL-cholesterol level without altering the HDL-cholesterol flux through plasma. Analysis of the hepatic gene expression profile showed that fenofibrate did not differentially affect the main players in HDL metabolism in E3L.CETP mice compared with E3L mice. However, in E3L.CETP mice, fenofibrate reduced hepatic CETP mRNA (-72%; P < 0.01) as well as the CE transfer activity in plasma (-73%; P < 0.01). We conclude that fenofibrate increases HDL-cholesterol by reducing the CETP-dependent transfer of cholesterol from HDL to (V)LDL, as related to lower hepatic CETP expression and a reduced plasma (V)LDL pool.
Collapse
Affiliation(s)
- Caroline C van der Hoogt
- Netherlands Organization for Applied Scientific Research-Quality of Life, Gaubius Laboratory, 2301 CE Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Westerterp M, van der Hoogt CC, de Haan W, Offerman EH, Dallinga-Thie GM, Jukema JW, Havekes LM, Rensen PCN. Cholesteryl Ester Transfer Protein Decreases High-Density Lipoprotein and Severely Aggravates Atherosclerosis in
APOE*3-Leiden
Mice. Arterioscler Thromb Vasc Biol 2006; 26:2552-9. [PMID: 16946130 DOI: 10.1161/01.atv.0000243925.65265.3c] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Objective—
The role of cholesteryl ester transfer protein (CETP) in the development of atherosclerosis is still undergoing debate. Therefore, we evaluated the effect of human CETP expression on atherosclerosis in
APOE*3-Leiden
(
E3L
) mice with a humanized lipoprotein profile.
Methods and Results—
E3L
mice were crossbred with human
CETP
transgenic mice. On a chow diet, CETP expression increased plasma total cholesterol (TC) (+43%;
P
<0.05). To evaluate the effects of CETP on the development of atherosclerosis, mice were fed a Western-type diet containing 0.25% cholesterol, leading to 4.3-fold elevated TC levels in both
E3L
and
CETP.E3L
mice (
P
<0.01). On both diets, CETP expression shifted the distribution of cholesterol from high-density lipoprotein (HDL) toward very-low-density lipoprotein (VLDL)/low-density lipoprotein (LDL). Moreover, plasma of
CETP.E3L
mice had reduced capacity (−39%;
P
<0.05) to induce SR-BI–mediated cholesterol efflux from Fu5AH cells than plasma of
E3L
mice. After 19 weeks on the Western-type diet,
CETP.E3L
mice showed a 7.0-fold increased atherosclerotic lesion area in the aortic root compared with
E3L
mice (
P
<0.0001).
Conclusions—
CETP expression in
E3L
mice shifts the distribution of cholesterol from HDL to VLDL/LDL, reduces plasma-mediated SR-BI–dependent cholesterol efflux, and represents a clear pro-atherogenic factor in
E3L
mice. We anticipate that the
CETP.E3L
mouse will be a valuable model for the preclinical evaluation of HDL-raising interventions on atherosclerosis development.
Collapse
Affiliation(s)
- Marit Westerterp
- Department of Biomedical Research, Gaubius Laboratory, CE Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Casquero AC, Berti JA, Salerno AG, Bighetti EJB, Cazita PM, Ketelhuth DFJ, Gidlund M, Oliveira HCF. Atherosclerosis is enhanced by testosterone deficiency and attenuated by CETP expression in transgenic mice. J Lipid Res 2006; 47:1526-34. [PMID: 16603720 DOI: 10.1194/jlr.m600135-jlr200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this work, we investigated the impact of testosterone deficiency and cholesteryl ester transfer protein (CETP) expression on lipoprotein metabolism and diet-induced atherosclerosis. CETP transgenic mice and nontransgenic (nTg) littermates were studied 4 weeks after bilateral orchidectomy or sham operation. Castrated mice had an increase in the LDL fraction (+36% for CETP and +79% for nTg mice), whereas the HDL fraction was reduced (-30% for CETP and -11% for nTg mice). Castrated mice presented 1.7-fold higher titers of anti-oxidized LDL (Ox-LDL) antibodies than sham-operated controls. Plasma levels of CETP, lipoprotein lipase, and hepatic lipase were not changed by castration. Kinetic studies showed no differences in VLDL secretion rate, VLDL-LDL conversion rate, or number of LDL and HDL receptors. Competition experiments showed lower affinity of LDL from castrated mice for tissue receptors. Diet-induced atherosclerosis studies showed that testosterone deficiency increased by 100%, and CETP expression reduced by 44%, the size of aortic lesion area in castrated mice. In summary, testosterone deficiency increased plasma levels of apolipoprotein B-containing lipoproteins (apoB-LPs) and anti-OxLDL antibodies, decreased LDL receptor affinity, and doubled the size of diet-induced atherosclerotic lesions. The expression of CETP led to a milder increase of apoB-LPs and reduced atherosclerotic lesion size in testosterone-deficient mice.
Collapse
Affiliation(s)
- A C Casquero
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas, Sao Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Alberts P, Klingström G, Arrhenius‐Nyberg V, Larsson C, Sakariassen KS. A micro‐method for lipoprotein cholesterol profiles: Impact of CETP in KKA
y
mice. EUR J LIPID SCI TECH 2006. [DOI: 10.1002/ejlt.200500261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Pe¯teris Alberts
- Pharmacology, Department of Biology, Biovitrum, Stockholm, Sweden
| | | | | | - Catarina Larsson
- Pharmacology, Department of Biology, Biovitrum, Stockholm, Sweden
| | - Kjell S. Sakariassen
- Pharmacology, Department of Biology, Biovitrum, Stockholm, Sweden
- Present address: KellSa s.a.s., Str. Campo e Zampe 12, I‐13900 Biella, BI, Italy
| |
Collapse
|
33
|
Krieger MH, Santos KFR, Shishido SM, Wanschel ACBA, Estrela HFG, Santos L, De Oliveira MG, Franchini KG, Spadari-Bratfisch RC, Laurindo FRM. Antiatherogenic effects of S-nitroso-N-acetylcysteine in hypercholesterolemic LDL receptor knockout mice. Nitric Oxide 2005; 14:12-20. [PMID: 16198610 DOI: 10.1016/j.niox.2005.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2004] [Revised: 07/08/2005] [Accepted: 07/29/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND The pathophysiology of the NO/NO synthase system and dysfunctional changes in the endothelium in the early phases of the atherogenic process are incompletely understood. In this study, we investigated the effects of the nitrosothiol NO donor S-nitroso-N-acetylcysteine (SNAC) in the early prevention of plaque development in the hypercholesterolemic LDLr-/- mice as well as the changes in endothelium-dependent relaxation and NO synthase expression. METHODS AND RESULTS LDLr-/- mice were fed a 1.25% cholesterol-enriched diet for 15 days. Plasma cholesterol/triglyceride levels increased and this increase was accompanied by the development of aortic root lesions. Aortic vasorelaxation to acetylcholine was increased, although endothelium-independent relaxation in response to sodium nitroprusside did not change, which suggest stimulated NO release enhanced. This dysfunction was associated with enhanced aortic superoxide production and with increased levels of constitutive NOS isoform expression, particularly neuronal NOS. SNAC (S-nitroso-N-acetylcysteine) administration (0.51 micromol/kg/day i.p. for 15 days) decreased the extent of the plaque by 55% in hypercholesterolemic mice, but had no effects on vasomotor changes. It did, however, lead to a decrease in constitutive NOS expression. The SNAC induced only minor changes in plasma lipid profile. CONCLUSION The present study has shown that, in early stages of plaque development in LDLr-/- mice, specific changes in NO/NO synthase system develop, that are characterized by increased endothelium-dependent vasorelaxation and increased constitutive NOS expression. Since the development of plaque and the indicator of endothelial cell dysfunction were prevented by SNAC, such treatment may constitute a novel strategy for the halting of progression of early plaque.
Collapse
Affiliation(s)
- M H Krieger
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), SP, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Berbée JFP, van der Hoogt CC, Sundararaman D, Havekes LM, Rensen PCN. Severe hypertriglyceridemia in human APOC1 transgenic mice is caused by apoC-I-induced inhibition of LPL. J Lipid Res 2005; 46:297-306. [PMID: 15576844 DOI: 10.1194/jlr.m400301-jlr200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies in humans and mice have shown that increased expression of apolipoprotein C-I (apoC-I) results in combined hyperlipidemia with a more pronounced effect on triglycerides (TGs) compared with total cholesterol (TC). The aim of this study was to elucidate the main reason for this effect using human apoC-I-expressing (APOC1) mice. Moderate plasma human apoC-I levels (i.e., 4-fold higher than human levels) caused a 12-fold increase in TG, along with a 2-fold increase in TC, mainly confined to VLDL. Cross-breeding of APOC1 mice on an apoE-deficient background resulted in a marked 55-fold increase in TG, confirming that the apoC-I-induced hyperlipidemia cannot merely be attributed to blockade of apoE-recognizing hepatic lipoprotein receptors. The plasma half-life of [3H]TG-VLDL-mimicking particles was 2-fold increased in APOC1 mice, suggesting that apoC-I reduces the lipolytic conversion of VLDL. Although total postheparin plasma LPL activity was not lower in APOC1 mice compared with controls, apoC-I was able to dose-dependently inhibit the LPL-mediated lipolysis of [3H]TG-VLDL-mimicking particles in vitro with a 60% efficiency compared with the main endogenous LPL inhibitor apoC-III. Finally, purified apoC-I impaired the clearance of [3H]TG-VLDL-mimicking particles independent of apoE-mediated hepatic uptake in lactoferrin-treated mice. Therefore, we conclude that apoC-I is a potent inhibitor of LPL-mediated TG-lipolysis.
Collapse
Affiliation(s)
- Jimmy F P Berbée
- Netherlands Organization for Applied Scientific Research-Prevention and Health, Gaubius Laboratory, 2301 CE Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
35
|
Berti JA, Salerno AG, Bighetti EJB, Casquero AC, Boschero AC, Oliveira HCF. Effects of diabetes and CETP expression on diet-induced atherosclerosis in LDL receptor-deficient mice. APMIS 2005; 113:37-44. [PMID: 15676013 DOI: 10.1111/j.1600-0463.2005.apm1130106.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The role of CETP expression and diabetes in atherogenesis was investigated in mice with heterozygous disruption of the LDL receptor gene (LDLR1). LDLR1 mice with and without CETP expression were treated with streptozotocin (STZ) and maintained on a standard diet for one month before switching to an atherogenic diet for an additional month. STZ-sensitive mice had approximately 2.5-fold higher glycemia and 7.5- to 8.0-fold higher cholesterolemia. Factorial analysis of variance showed no significant effect of diabetes, CETP or diabetes-CETP interaction on the size of the atherosclerotic lesions. CETP expression in non-diabetic mice resulted in a 50% reduction in the area of the atherosclerotic lesions. Multiple regression analysis showed a positive and independent atherogenic effect of triglyceridemia in LDLR1 mice and of cholesterolemia in diabetic mice. Logistic analysis showed that elevated plasma cholesterol level significantly increased the risk of developing large lesion size (>75th percentile). In conclusion, CETP expression did not alter the lesion formation in response to diabetes, although it may be protective in the euglycemic state; the triglyceride level was an independent risk factor for LDL receptor-deficient mice but not for CETP-expressing mice; and elevated plasma cholesterol levels increased the risk of developing large atherosclerotic lesions, independently of CETP and diabetes.
Collapse
Affiliation(s)
- Jairo A Berti
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
36
|
Berti JA, Casquero AC, Patricio PR, Bighetti EJB, Carneiro EM, Boschero AC, Oliveira HCF. Cholesteryl ester transfer protein expression is down-regulated in hyperinsulinemic transgenic mice. J Lipid Res 2003; 44:1870-6. [PMID: 12867535 DOI: 10.1194/jlr.m300036-jlr200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) mediates cholesteryl ester (CE) and triglyceride redistribution among plasma lipoproteins. In this work, we investigated whether varying levels of insulin regulate the CETP expression in vivo. Insulin deficiency [streptozotocin (STZ) injection], and hyperinsulinemia (insulin injections, 14 days) were induced in transgenic mice expressing a human CETP minigene flanked by its natural regulatory sequences. Glucose supplementation was provided to the hyperinsulinemic group (INS+GLUC) and to an extra group of mice (GLUC). In the STZ group, endogenous CE transfer rate, plasma CETP, and hepatic CETP mRNA levels were enhanced 3.0-, 1.5-, and 2.5-fold, respectively, as compared with controls. Insulin replacement in STZ mice normalized their glycemia and liver mRNA levels. Higher plasma CETP levels were observed in GLUC mice, which were decreased in INS+GLUC mice. Hepatic CETP mRNA was not altered in GLUC mice and was reduced by one-third in INS+GLUC mice. These results show that: 1) STZ treatment increases CETP plasma levels and liver mRNA expression; 2) diet glucose supplementation increases plasma CETP levels but does not change liver mRNA abundance; and 3) daily insulin injections blunt the glucose-stimulated CETP expression by reducing its liver mRNA levels. These data suggest that insulin down-regulates CETP gene expression.
Collapse
Affiliation(s)
- J A Berti
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, 13083-970, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
37
|
Hammad SM, Powell-Braxton L, Otvos JD, Eldridge L, Won W, Lyons TJ. Lipoprotein subclass profiles of hyperlipidemic diabetic mice measured by nuclear magnetic resonance spectroscopy. Metabolism 2003; 52:916-21. [PMID: 12870170 DOI: 10.1016/s0026-0495(03)00058-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dyslipidemia accelerates vascular complications of diabetes. Nuclear magnetic resonance (NMR) analysis of lipoprotein subclasses is used to evaluate a mouse model of human familial hypercholesterolemia +/- streptozotocin (STZ)-induced diabetes. A double knockout (DKO) mouse (low-density lipoprotein receptor [LDLr] -/-; apolipoprotein B [apoB] mRNA editing catalytic polypeptide-1 [Apobec1] -/-) was studied. Wild-type (WT) and DKO mice received sham or STZ injections at age 7 weeks, yielding control (WT-C, DKO-C) and diabetic (WT-D, DKO-D) groups. Fasting serum was collected when the mice were killed (age 40 weeks) for Cholestech analysis (Cholestech Corp, Hayward, CA) and NMR lipoprotein subclass profile. By Cholestech, fasting triglyceride and total cholesterol increased in DKO-C versus WT-C. Diabetes further increased total cholesterol in DKO. High-density lipoprotein cholesterol (HDL-C) was similar among all groups. NMR revealed that LDL in all groups was present in a subclass the size of large human LDL and was increased 48-fold in DKO-C versus WT-C animals, but was unaffected by diabetes. HDL was found in a subclass equivalent to large human HDL, and was similar among groups. In conclusion, NMR analysis reveals lipoprotein subclass distributions and the effects of genetic modification and diabetes in mice, but lack of particles the size of human small LDL and small HDL may limit the relevance of the present animal model to human disease.
Collapse
Affiliation(s)
- Samar M Hammad
- Division of Endocrinology Diabetes and Medical Genetics, Medical University of South Carolina, Charleston 29425, USA
| | | | | | | | | | | |
Collapse
|
38
|
Ishida T, Choi S, Kundu RK, Hirata KI, Rubin EM, Cooper AD, Quertermous T. Endothelial lipase is a major determinant of HDL level. J Clin Invest 2003; 111:347-55. [PMID: 12569160 PMCID: PMC151857 DOI: 10.1172/jci16306] [Citation(s) in RCA: 234] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A new member of the lipase gene family, initially termed endothelial lipase (gene nomenclature, LIPG; protein, EL), is expressed in a variety of different tissues, suggesting a general role in lipid metabolism. To assess the hypothesis that EL plays a physiological role in lipoprotein metabolism in vivo, we have used gene targeting of the native murine locus and transgenic introduction of the human LIPG locus in mice to modulate the level of EL expression. Evaluation of these alleles in a C57Bl/6 background revealed an inverse relationship between HDL cholesterol level and EL expression. Fasting plasma HDL cholesterol was increased by 57% in LIPG(-/-) mice and 25% in LIPG(+/-) mice and was decreased by 19% in LIPG transgenic mice as compared with syngeneic controls. Detailed analysis of lipoprotein particle composition indicated that this increase was due primarily to an increased number of HDL particles. Phospholipase assays indicated that EL is a primary contributor to phospholipase activity in mouse. These data indicate that expression levels of this novel lipase have a significant effect on lipoprotein metabolism.
Collapse
MESH Headings
- Alleles
- Animals
- Blotting, Southern
- Cholesterol/metabolism
- Cholesterol, HDL/metabolism
- Chromatography, High Pressure Liquid
- DNA, Complementary/metabolism
- Electrophoresis, Polyacrylamide Gel
- Exons
- Heterozygote
- Lipase/physiology
- Lipid Metabolism
- Lipoproteins, HDL/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Genetic
- Phospholipases/metabolism
- Plasma/metabolism
- RNA/metabolism
Collapse
Affiliation(s)
- Tatsuro Ishida
- Division of Cardiovascular Medicine and Donald W. Reynolds Cardiovascular Clinical Research Center, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Cianflone K, Xia Z, Chen LY. Critical review of acylation-stimulating protein physiology in humans and rodents. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1609:127-43. [PMID: 12543373 DOI: 10.1016/s0005-2736(02)00686-7] [Citation(s) in RCA: 263] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the last few years, there has been increasing interest in the physiological role of acylation-stimulating protein (ASP). Recent studies in rats and mice, in particular in C3 (-/-) mice that are ASP deficient, have advanced our understanding of the role of ASP. Of note, the background strain of the mice influences the phenotype of delayed postprandial triglyceride clearance in ASP-deficient mice. Administration of ASP in all types of lean and obese mice studied to date, however, enhances postprandial triglyceride clearance. On the other hand, regardless of the background strain, ASP-deficient mice demonstrate reduced body weight, reduced leptin and reduced adipose tissue mass, suggesting that ASP deficiency results in protection against development of obesity. In humans, a number of studies have examined the relationship between ASP, obesity, diabetes and dyslipidemia as well as the influence of diet, exercise and pharmacological therapy. While many of these studies have small subject numbers, interesting observations may help us to better understand the parameters that may influence ASP production and ASP action. The aim of the present review is to provide a comprehensive overview of the recent literature on ASP, with particular emphasis on those studies carried out in rodents and humans.
Collapse
Affiliation(s)
- Katherine Cianflone
- McGill University, Cardiology, H7.30, Royal Victoria Hospital, 687 Pine Ave West, Montreal, Quebec, Canada H3A 1A1.
| | | | | |
Collapse
|
40
|
Cazita PM, Berti JA, Aoki C, Gidlund M, Harada LM, Nunes VS, Quintão ECR, Oliveira HCF. Cholesteryl ester transfer protein expression attenuates atherosclerosis in ovariectomized mice. J Lipid Res 2003; 44:33-40. [PMID: 12518020 DOI: 10.1194/jlr.m100440-jlr200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reduced estrogen levels result in loss of protection from coronary heart disease in postmenopausal women. Enhanced and diminished atherosclerosis have been associated with plasma levels of cholesteryl ester transfer protein (CETP); however, little is known about the role of CETP-ovarian hormone interactions in atherogenesis. We assessed the severity of diet-induced atherosclerosis in ovariectomized (OV) CETP transgenic mice crossbred with LDL receptor knockout mice. Compared with OV CETP expressing ((+)), OV CETP non-expressing ((-)) mice had higher plasma levels of total, VLDL-, LDL-, and HDL-cholesterol, as well as higher antibodies titers against oxidized LDL. The mean aortic lesion area was 2-fold larger in OV CETP(-) than in OV CETP(+) mice (147 +/- 90 vs. 73 +/- 42 x 10(3) micro m(2), respectively). Estrogen therapy in OV mice blunted the CETP dependent differences in plasma lipoproteins, oxLDL antibodies, and atherosclerosis severity. Macrophages from OV CETP(+) mice took up less labeled cholesteryl ether (CEt) from acetyl-LDL than macrophages from OV CETP(-) mice. Estrogen replacement induced a further reduction in CEt uptake and an elevation in HDL mediated cholesterol efflux from pre-loaded OV CETP(+) as compared with OV CETP(-) macrophages. These findings support the proposed anti-atherogenic role of CETP in specific metabolic settings.
Collapse
Affiliation(s)
- Patrícia M Cazita
- Laboratório de Lípides, Faculdade de Medicina da Universidade de São Paulo, 01246-903, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zak Z, Lagrost L, Gautier T, Masson D, Deckert V, Duverneuil L, De Barros JPP, Le Guern N, Dumont L, Schneider M, Risson V, Moulin P, Autran D, Brooker G, Sassard J, Bataillard A. Expression of simian CETP in normolipidemic Fisher rats has a profound effect on large sized apoE-containing HDL. J Lipid Res 2002; 43:2164-71. [PMID: 12454279 DOI: 10.1194/jlr.m200253-jlr200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In order to investigate the direct effect of cholesteryl ester transfer protein (CETP) on the structure and composition of HDL in vivo, simian CETP was expressed in Fisher rat that spontaneously displays high plasma levels of HDL1. In the new CETPTg rat line, the production of active CETP by the liver induced a significant 48% decrease in plasma HDL cholesterol, resulting in a 34% decrease in total cholesterol level (P < 0.01 in both cases). Among the various plasma HDL subpopulations, the largest HDL were those mostly affected by CETP, with a 74% decrease in HDL1 versus a significantly weaker 38% decrease in smaller HDL2 (P < 0.0001). Apolipoprotein E (apoE)-containing HDL1 were selectively affected by CETP expression, whereas apoA content of HDL remained unmodified. The reduction in the apoE content of serum HDL observed in CETPTg rats compared to controls (53%, P < 0.02) suggests that apoE in HDL may constitute in vivo a major determinant of their ability to interact with CETP. These results bring new insight into the lack of HDL1 in plasma from CETP-deficient heterozygotes despite their substantial 50% decrease in CETP activity. In addition, they indicate that HDL1 constitute reliable and practicable sensors of very low plasma CETP activity in vivo.
Collapse
Affiliation(s)
- Zoulika Zak
- Département de Physiologie et de Pharmacologie Clinique, CNRS UMR 5014, IFR 39, Université Claude Bernard Lyon I, 69008 Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hofmann SM, Dong HJ, Li Z, Cai W, Altomonte J, Thung SN, Zeng F, Fisher EA, Vlassara H. Improved insulin sensitivity is associated with restricted intake of dietary glycoxidation products in the db/db mouse. Diabetes 2002; 51:2082-9. [PMID: 12086936 DOI: 10.2337/diabetes.51.7.2082] [Citation(s) in RCA: 230] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Advanced glycation end products (AGEs), known promoters of diabetic complications, form abundantly in heated foods and are ingested in bioreactive forms. To test whether dietary AGEs play a role in the progression of insulin resistance, C57/BL/KsJ db/db mice were randomly placed for 20 weeks on a diet with either a low AGE content (LAD) or a 3.4-fold higher content of AGE (high AGE diet [HAD]), including (epsilon)N-carboxymethyllysine (CML) and methylglyoxal (MG). LAD-fed mice showed lower fasting plasma insulin levels throughout the study (P = 0.01). Body weight was reduced by approximately 13% compared with HAD-fed mice (P = 0.04) despite equal food intake. LAD-fed mice exhibited significantly improved responses to both glucose (at 40 min, P = 0.003) and insulin (at 60 min, P = 0.007) tolerance tests, which correlated with a twofold higher glucose uptake by adipose tissue (P = 0.02). Compared with the severe hypertrophy and morphological disorganization of islets from HAD-fed mice, LAD-fed mice presented a better-preserved structure of the islets. LAD-fed mice demonstrated significantly increased plasma HDL concentrations (P < 0.0001). Consistent with these observations, LAD-fed mice exhibited twofold lower serum CML and MG concentrations compared with HAD-fed mice (P = 0.02). These results demonstrate that reduced AGE intake leads to lower levels of circulating AGE and to improved insulin sensitivity in db/db mice.
Collapse
Affiliation(s)
- Susanna M Hofmann
- Brookdale Department of Genetics, Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6574, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Fuchs M, Hafer A, Münch C, Kannenberg F, Teichmann S, Scheibner J, Stange EF, Seedorf U. Disruption of the sterol carrier protein 2 gene in mice impairs biliary lipid and hepatic cholesterol metabolism. J Biol Chem 2001; 276:48058-65. [PMID: 11673458 DOI: 10.1074/jbc.m106732200] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hepatic up-regulation of sterol carrier protein 2 (Scp2) in mice promotes hypersecretion of cholesterol into bile and gallstone formation in response to a lithogenic diet. We hypothesized that Scp2 deficiency may alter biliary lipid secretion and hepatic cholesterol metabolism. Male gallstone-susceptible C57BL/6 and C57BL/6(Scp2(-/-)) knockout mice were fed a standard chow or lithogenic diet. Hepatic biles were collected to determine biliary lipid secretion rates, bile flow, and bile salt pool size. Plasma lipoprotein distribution was investigated, and gene expression of cytosolic lipid-binding proteins, lipoprotein receptors, hepatic regulatory enzymes, and intestinal cholesterol absorption was measured. Compared with chow-fed wild-type animals, C57BL/6(Scp2(-/-)) mice had higher bile flow and lower bile salt secretion rates, decreased hepatic apolipoprotein expression, increased hepatic cholesterol synthesis, and up-regulation of liver fatty acid-binding protein. In addition, the bile salt pool size was reduced and intestinal cholesterol absorption was unaltered in C57BL/6(Scp2(-/-)) mice. When C57BL/6(Scp2(-/-)) mice were challenged with a lithogenic diet, a smaller increase of hepatic free cholesterol failed to suppress cholesterol synthesis and biliary cholesterol secretion increased to a much smaller extent than phospholipid and bile salt secretion. Scp2 deficiency did not prevent gallstone formation and may be compensated in part by hepatic up-regulation of liver fatty acid-binding protein. These results support a role of Scp2 in hepatic cholesterol metabolism, biliary lipid secretion, and intracellular cholesterol distribution.
Collapse
Affiliation(s)
- M Fuchs
- Division of Gastroenterology, Department of Medicine I, Medical University of Lübeck, D-23538 Lübeck, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Monks J, Huey PU, Hanson L, Eckel RH, Neville MC, Gavigan S. A lipoprotein-containing particle is transferred from the serum across the mammary epithelium into the milk of lactating mice. J Lipid Res 2001. [DOI: 10.1016/s0022-2275(20)31630-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
45
|
Srivastava N, Chowdhury PR, Averna M, Srivastava RA. Estrogen increases hepatic lipase levels in inbred strains of mice: a possible mechanism for estrogen-dependent lowering of high density lipoprotein. Mol Cell Biochem 2001; 220:87-93. [PMID: 11451387 DOI: 10.1023/a:1010845032399] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We have shown mouse to be an useful animal model for studies on the estrogen-mediated synthesis and secretion of lipoproteins since, unlike in rats, low density lipoprotein receptors are not upregulated in mice. This results into the elevation of plasma levels of apolipoprotein (apo) B and apoE, and lowering of apoA-1-containing particles. The mechanisms of apoB and apoE elevation by estrogen have been elucidated, but the mechanism of lowering of plasma levels of HDL is still not known. Among other factors, apoA-I, cholesterol ester transfer protein (CETP), scavenger receptor B1 (SR-B1), and hepatic lipase are potential candidates that modulate plasma levels of HDL. Since estrogen treatment increased hepatic apoA-I mRNA and apoA-I synthesis, and mouse express undetectable levels of CETP, we tested the hypothesis that estradiol-mediated lowering of HDL in mice may occur through modulation of hepatic lipase (HL). Four mouse strains (C57L, C57BL, BALB, C3H) were administered supraphysiological doses of estradiol, and plasma levels of HDL as well as HL mRNA were quantitated. In all 4 strains estradiol decreased plasma levels of HDL by 30%, and increased HL mRNA 2-3 fold. In a separate experiment groups of male C57BL mouse were castrated or sham-operated, and low and high doses of estradiol administered. We found 1.4-2.5 fold elevation of HL mRNA with concomitant lowering of HDL levels. Ten other mouse strains examined also showed estradiol-induced elevation of HL mRNA, but the extent of elevation was found to be strain-specific. Based on these studies, we conclude that hepatic lipase is an important determinant of plasma levels of HDL and that HL mRNA is modulated by estrogen which in turn may participate in the lowering of plasma levels of HDL.
Collapse
Affiliation(s)
- N Srivastava
- Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | | | | | | |
Collapse
|
46
|
Lyons MA, Brown AJ. 7-Ketocholesterol delivered to mice in chylomicron remnant-like particles is rapidly metabolised, excreted and does not accumulate in aorta. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1530:209-18. [PMID: 11239823 DOI: 10.1016/s1388-1981(01)00083-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cholesterol oxidation products (oxysterols) have been implicated in atherogenesis due to their presence in atherosclerotic tissue and their potent effects in vitro. One of the major oxysterols currently of interest is 7-ketocholesterol (7K) and it has been suggested that the diet is an important source of this oxysterol. This investigation tested the hypothesis that 7K, delivered in a physiologically relevant vehicle, chylomicron remnant-like emulsion (CMR), would be metabolised and excreted by mice in a similar manner and to a similar extent as previously observed in rats when delivered in a chemically modified lipoprotein, acetylated low-density lipoprotein (acLDL). Indeed, the metabolism of 14C-7K delivered in CMR mirrored that of acLDL and was much more rapid than (3)H-cholesterol delivered simultaneously. The 7K-derived (14)C was cleared from the liver, appeared in the intestine and was excreted in the faeces. A substantial proportion of the 7K-derived (14)C in the intestine and faeces was aqueous-soluble, indicating metabolism to polar products, presumably bile acids. Moreover, while cholesterol-derived (3)H increased in the aorta, (14)C appeared transiently and there was no observable accumulation within 24 h. The data confirm our previous findings of rapid hepatic metabolism of 7K when delivered in acLDL and demonstrate that 7K delivered in a vehicle of dietary significance is similarly metabolised and excreted. Indeed, the data encourage further investigation into the contribution that dietary oxysterols may or may not make to atherogenesis.
Collapse
Affiliation(s)
- M A Lyons
- Cell Biology Group, Heart Research Institute, 145 Missenden Road, Camperdown, 2050, Sydney, N.S.W., Australia
| | | |
Collapse
|
47
|
Nunes VS, Quintão ECR, Cazita PM, Harada LM, de Faria EC, Oliveira HCF. Plasma lipases and lipid transfer proteins increase phospholipid but not free cholesterol transfer from lipid emulsion to high density lipoproteins. BMC BIOCHEMISTRY 2001; 2:1. [PMID: 11242564 PMCID: PMC31329 DOI: 10.1186/1471-2091-2-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2000] [Accepted: 02/20/2001] [Indexed: 11/10/2022]
Abstract
BACKGROUND Plasma lipases and lipid transfer proteins are involved in the generation and speciation of high density lipoproteins. In this study we have examined the influence of plasma lipases and lipid transfer protein activities on the transfer of free cholesterol (FC) and phospholipids (PL) from lipid emulsion to human, rat and mouse lipoproteins. The effect of the lipases was verified by incubation of labeled (3H-FC,14C-PL) triglyceride rich emulsion with human plasma (control, post-heparin and post-heparin plus lipase inhibitor), rat plasma (control and post-heparin) and by the injection of the labeled lipid emulsion into control and heparinized functionally hepatectomized rats. RESULTS In vitro, the lipase enriched plasma stimulated significantly the transfer of 14C-PL from emulsion to high density lipoprotein (p<0.001) but did not modify the transfer of 3H-FC. In hepatectomized rats, heparin stimulation of intravascular lipolysis increased the plasma removal of 14C-PL and the amount of 14C-PL found in the low density lipoprotein density fraction but not in the high density lipoprotein density fraction. The in vitro and in vivo experiments showed that free cholesterol and phospholipids were transferred from lipid emulsion to plasma lipoproteins independently from each other. The incubation of human plasma, control and control plus monoclonal antibody anti-cholesteryl ester transfer protein (CETP), with 14C-PL emulsion showed that CETP increases 14C-PL transfer to human HDL, since its partial inhibition by the anti-CETP antibody reduced significantly the 14C-PL transfer (p<0.05). However, comparing the nontransgenic (no CETP activity) with the CETP transgenic mouse plasma, no effect of CETP on the 14C-PL distribution in mice lipoproteins was observed. CONCLUSIONS It is concluded that: 1-intravascular lipases stimulate phospholipid transfer protein mediated phospholipid transfer, but not free cholesterol, from triglyceride rich particles to human high density lipoproteins and rat low density lipoproteins and high density lipoproteins; 2-free cholesterol and phospholipids are transferred from triglyceride rich particles to plasma lipoproteins by distinct mechanisms, and 3 - CETP also contributes to phospholipid transfer activity in human plasma but not in transgenic mice plasma, a species which has high levels of the specific phospholipid transfer protein activity.
Collapse
Affiliation(s)
- Valéria S Nunes
- Lipid Laboratory, University of São Paulo Medical School, São Paulo, Brazil
| | - Eder CR Quintão
- Lipid Laboratory, University of São Paulo Medical School, São Paulo, Brazil
| | - Patrícia M Cazita
- Lipid Laboratory, University of São Paulo Medical School, São Paulo, Brazil
| | - Lila M Harada
- Lipid Laboratory, University of São Paulo Medical School, São Paulo, Brazil
| | - Eliana C de Faria
- Dept. Clinical Pathology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Helena CF Oliveira
- Dept. Physiology and Biophysics, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
48
|
Garber DW, Kulkarni KR, Anantharamaiah G. A sensitive and convenient method for lipoprotein profile analysis of individual mouse plasma samples. J Lipid Res 2000. [DOI: 10.1016/s0022-2275(20)32045-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
49
|
Brousseau ME, Kauffman RD, Herderick EE, Demosky SJ, Evans W, Marcovina S, Santamarina-Fojo S, Brewer HB, Hoeg JM. LCAT modulates atherogenic plasma lipoproteins and the extent of atherosclerosis only in the presence of normal LDL receptors in transgenic rabbits. Arterioscler Thromb Vasc Biol 2000; 20:450-8. [PMID: 10669643 DOI: 10.1161/01.atv.20.2.450] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elevated low density lipoprotein cholesterol (LDL-C) and reduced high density lipoprotein cholesterol (HDL-C) concentrations are independent risk factors for coronary heart disease. We have previously demonstrated that overexpression of an enzyme with a well established role in HDL metabolism, lecithin:cholesterol acyltransferase (LCAT), in New Zealand White rabbits not only raises HDL-C concentrations but reduces those of LDL-C as well, ultimately preventing diet-induced atherosclerosis. In the present study, the human LCAT gene (hLCAT) was introduced into LDL receptor (LDLr)-deficient (Watanabe heritable hyperlipidemic) rabbits to (1) investigate the role of the LDLr pathway in the hLCAT-mediated reductions of LDL-C and (2) determine the influence of hLCAT overexpression on atherosclerosis susceptibility in an animal model of familial hypercholesterolemia. Heterozygosity or homozygosity for the LDLr defect was determined by polymerase chain reaction, and 3 groups of hLCAT-transgenic (hLCAT+) rabbits that differed in LDLr status were established: (1) LDLr wild-type (LDLr+/+), (2) LDLr heterozygotes (LDLr+/-), and (3) LDLr homozygotes (LDLr-/-). Data for hLCAT+ rabbits were compared with those of nontransgenic (hLCAT-) rabbits of the same LDLr status. Plasma HDL-C concentrations were significantly elevated in the hLCAT+ animals of each LDLr status. However, LDL-C levels were significantly reduced only in hLCAT+/LDLr+/+ and hLCAT+/LDLr+/- rabbits but not in hLCAT+/LDLr-/- rabbits (405+/-14 versus 392+/-31 mg/dL). Metabolic studies revealed that the fractional catabolic rate (FCR, d(-1)) of LDL apolipoprotein (apo) B-100 was increased in hLCAT+/LDLr+/+ (26+/-4 versus 5+/-0) and hLCAT+/LDLr+/- (4+/-1 versus 1+/-0) rabbits, whereas the FCR of LDL apoB-100 in both groups of LDLr-/- rabbits was nearly identical (0.16+/-0.02 versus 0.15+/-0.02). Consistently, neither aortic lipid concentrations nor the extent of aortic atherosclerosis was significantly different between hLCAT+/LDLr-/- and hLCAT-/LDLr-/- rabbits. Significant correlations were observed between the percent of aortic atherosclerosis and both LDL-C (r=0.985) and LDL apoB-100 FCR (-0.745), as well as between LDL-C and LDL apoB-100 FCR (-0.866). These data are the first to establish that LCAT modulates LDL metabolism via the LDLr pathway, ultimately influencing atherosclerosis susceptibility. Moreover, LCAT's antiatherogenic effect requires only a single functional LDLr allele, identifying LCAT as an attractive gene therapy candidate for the majority of dyslipoproteinemic patients.
Collapse
Affiliation(s)
- M E Brousseau
- Molecular Disease Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Murray I, Sniderman AD, Havel PJ, Cianflone K. Acylation stimulating protein (ASP) deficiency alters postprandial and adipose tissue metabolism in male mice. J Biol Chem 1999; 274:36219-25. [PMID: 10593909 DOI: 10.1074/jbc.274.51.36219] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Acylation stimulating protein (ASP) is a potent stimulator of triglyceride synthesis in adipocytes. In the present study, we have examined the effect of an ASP functional knockout (ASP(-/-)) on lipid metabolism in male mice. In both young (14 weeks) and older (26 weeks) mice there were marked delays in postprandial triglyceride clearance (80% increase at 14 weeks and 120% increase at 26 weeks versus wild type (+/+)). Postprandial nonesterified fatty acids were also increased in ASP(-/-) mice versus ASP(+/+) mice by 37% (low fat 10% Kcal) and by 73% (high fat 40% Kcal) diets, although there were no differences in fasting lipid levels. The ASP(-/-) mice had moderately increased energy intake (16% +/- 2% p < 0.0001) and reduced feed efficiency (33% increase in calories/g of body weight gained on low fat diet) versus wild type. The ASP(-/-) mice also had modest changes in insulin/glucose metabolism (30% to 40% decrease in insulin.glucose product), implying increased insulin sensitivity. As well, there were decreases in leptin (29% shift in leptin to body weight ratio) and up to a 26% decrease in specific adipose tissue depots versus the wild type mice on both low fat and high fat diets. These results demonstrate that ASP plays an important role in adipose tissue metabolism and fat partitioning.
Collapse
Affiliation(s)
- I Murray
- Mike Rosenbloom Laboratory for Cardiovascular Research, McGill University Health Centre, Montreal, Quebec, Canada, H3A 1A1 Canada
| | | | | | | |
Collapse
|