1
|
Ye ZC, Liu JY, Chen CJ, Chen YY, Li WC, Liu MY. Investigating the binding products between human apolipoproteins and oxidized 1-palmitoyl-2-arachidonyl-sn-glycerol-3-phosphocholine by micellar electrokinetic chromatography. J Chromatogr A 2025; 1751:465898. [PMID: 40220603 DOI: 10.1016/j.chroma.2025.465898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/16/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
A micellar electrokinetic chromatography (MEKC) method has been developed to investigate the binding products between human apolipoproteins (Apos) and oxidized 1-palmitoyl-2-arachidonoyl-sn‑glycero-3-phosphocholine (ox-PAPC) products. The optimal MEKC separation buffer was composed of a solution mixture of 10 mM sodium phosphate, 50 mM bile salts (50 % sodium cholate and 50 % sodium deoxycholate), 30 % (v/v)1-propanol and 70 % (v/v) water, pH 7.4. The optimal MEKC sample buffer was composed of 70 % (v/v) PBS buffer and 30 % (v/v) MeOH. The selected separation voltage was 20 kV, and the capillary temperature was 25℃. The MEKC profiles of ox-PAPC products showed good separations and repeatability. The MEKC profiles of apos and their binding products also showed good repeatability. For the analysis of native PAPC (n-PAPC), the method is linear in the range of 0.00-6.00 mg/mL with a correlation coefficient 0.9984. The concentration limit of detection (LOD) is 0.29 mg/mL. The concentration limit of quantitation (LOQ) is 0.98 mg/mL. The binding reactions between several important human apolipoproteins (Apos A-I, A-II, C-I, C-II, C-III and E) and native PAPC, ox-PAPC products have been investigated. The concentrations of Apos and ox-PAPC products for binding reactions have been examined. The optimal binding buffer selected was 70 % (v/v) PBS buffer and 30 % (v/v) MeOH. The binding reaction was performed at 37 ℃ for 3 hr. The results indicated that ox-PAPC products bound to Apos A-I, A-II, C-I, and E more strongly than n-PAPC. However, both ox-PAPC products and n-PAPC did not bind to Apos C-II and C-III strongly. The results suggested pro-inflammatory properties of Apos A-I, A-II, C-I, and E, and implied one of the molecular mechanisms resulting in dysfunctional HDL particles. This study also demonstrated the feasibility of investigating the binding reactions between human apolipoproteins and ox-PAPC products by MEKC.
Collapse
Affiliation(s)
- Zhi-Cheng Ye
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Jia-Yuan Liu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yen-Yi Chen
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Wen-Chun Li
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Mine-Yine Liu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan.
| |
Collapse
|
2
|
Kothari V, Ho TW, Cabodevilla AG, He Y, Kramer F, Shimizu-Albergine M, Kanter JE, Snell-Bergeon J, Fisher EA, Shao B, Heinecke JW, Wobbrock JO, Lee WL, Goldberg IJ, Vaisar T, Bornfeldt KE. Imbalance of APOB Lipoproteins and Large HDL in Type 1 Diabetes Drives Atherosclerosis. Circ Res 2024; 135:335-349. [PMID: 38828596 PMCID: PMC11223987 DOI: 10.1161/circresaha.123.323100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 04/25/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Individuals with type 1 diabetes (T1D) generally have normal or even higher HDL (high-density lipoprotein)-cholesterol levels than people without diabetes yet are at increased risk for atherosclerotic cardiovascular disease (CVD). Human HDL is a complex mixture of particles that can vary in cholesterol content by >2-fold. To investigate if specific HDL subspecies contribute to the increased atherosclerosis associated with T1D, we created mouse models of T1D that exhibit human-like HDL subspecies. We also measured HDL subspecies and their association with incident CVD in a cohort of people with T1D. METHODS We generated LDL receptor-deficient (Ldlr-/-) mouse models of T1D expressing human APOA1 (apolipoprotein A1). Ldlr-/-APOA1Tg mice exhibited the main human HDL subspecies. We also generated Ldlr-/-APOA1Tg T1D mice expressing CETP (cholesteryl ester transfer protein), which had lower concentrations of large HDL subspecies versus mice not expressing CETP. HDL particle concentrations and sizes and proteins involved in lipoprotein metabolism were measured by calibrated differential ion mobility analysis and targeted mass spectrometry in the mouse models of T1D and in a cohort of individuals with T1D. Endothelial transcytosis was analyzed by total internal reflection fluorescence microscopy. RESULTS Diabetic Ldlr-/-APOA1Tg mice were severely hyperglycemic and hyperlipidemic and had markedly elevated plasma APOB levels versus nondiabetic littermates but were protected from the proatherogenic effects of diabetes. Diabetic Ldlr-/-APOA1Tg mice expressing CETP lost the atheroprotective effect and had increased lesion necrotic core areas and APOB accumulation, despite having lower plasma APOB levels. The detrimental effects of low concentrations of larger HDL particles in diabetic mice expressing CETP were not explained by reduced cholesterol efflux. Instead, large HDL was more effective than small HDL in preventing endothelial transcytosis of LDL mediated by scavenger receptor class B type 1. Finally, in humans with T1D, increased concentrations of larger HDL particles relative to APOB100 negatively predicted incident CVD independently of HDL-cholesterol levels. CONCLUSIONS Our results suggest that the balance between APOB lipoproteins and the larger HDL subspecies contributes to atherosclerosis progression and incident CVD in the setting of T1D and that larger HDLs exert atheroprotective effects on endothelial cells rather than by promoting macrophage cholesterol efflux.
Collapse
MESH Headings
- Adult
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Apolipoprotein A-I/blood
- Apolipoprotein A-I/metabolism
- Apolipoprotein B-100/metabolism
- Apolipoprotein B-100/genetics
- Apolipoprotein B-100/blood
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/blood
- Atherosclerosis/pathology
- Cholesterol Ester Transfer Proteins/genetics
- Cholesterol Ester Transfer Proteins/metabolism
- Cholesterol Ester Transfer Proteins/blood
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/blood
- Disease Models, Animal
- Lipoproteins, HDL/blood
- Lipoproteins, HDL/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/metabolism
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Tse W.W. Ho
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, Canada (T.W.W.H., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (T.W.W.H., W.L.L.)
| | | | - Yi He
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Masami Shimizu-Albergine
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Jenny E. Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Janet Snell-Bergeon
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora (J.S.-B.)
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (E.A.F.)
| | - Baohai Shao
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Jay W. Heinecke
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | | | - Warren L. Lee
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, Canada (T.W.W.H., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (T.W.W.H., W.L.L.)
- Interdepartmental Division of Critical Care and the Department of Biochemistry, University of Toronto, Canada (W.L.L.)
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism (A.G.C., I.J.G.)
| | - Tomas Vaisar
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Karin E. Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle (K.E.B.)
| |
Collapse
|
3
|
Bruter AV, Varlamova EA, Okulova YD, Tatarskiy VV, Silaeva YY, Filatov MA. Genetically modified mice as a tool for the study of human diseases. Mol Biol Rep 2024; 51:135. [PMID: 38236499 DOI: 10.1007/s11033-023-09066-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/23/2023] [Indexed: 01/19/2024]
Abstract
Modeling a human disease is an essential part of biomedical research. The recent advances in the field of molecular genetics made it possible to obtain genetically modified animals for the study of various diseases. Not only monogenic disorders but also chromosomal and multifactorial disorders can be mimicked in lab animals due to genetic modification. Even human infectious diseases can be studied in genetically modified animals. An animal model of a disease enables the tracking of its pathogenesis and, more importantly, to test new therapies. In the first part of this paper, we review the most common DNA modification technologies and provide key ideas on specific technology choices according to the task at hand. In the second part, we focus on the application of genetically modified mice in studying human diseases.
Collapse
Affiliation(s)
- Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334
- Federal State Budgetary Institution "National Medical Research Center of Oncology Named After N.N. Blokhin" of the Ministry of Health of the Russian Federation, Research Institute of Carcinogenesis, Moscow, Russia, 115478
| | - Ekaterina A Varlamova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334
- Federal State Budgetary Institution "National Medical Research Center of Oncology Named After N.N. Blokhin" of the Ministry of Health of the Russian Federation, Research Institute of Carcinogenesis, Moscow, Russia, 115478
| | - Yulia D Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334
| | - Victor V Tatarskiy
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334
| | - Yulia Y Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334
| | - Maxim A Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia, 119334.
| |
Collapse
|
4
|
Lund J, Lähteenmäki E, Eklund T, Bakke HG, Thoresen GH, Pirinen E, Jauhiainen M, Rustan AC, Lehti M. Human HDL subclasses modulate energy metabolism in skeletal muscle cells. J Lipid Res 2024; 65:100481. [PMID: 38008260 PMCID: PMC10770614 DOI: 10.1016/j.jlr.2023.100481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023] Open
Abstract
In addition to its antiatherogenic role, HDL reportedly modulates energy metabolism at the whole-body level. HDL functionality is associated with its structure and composition, and functional activities can differ between HDL subclasses. Therefore, we studied if HDL2 and HDL3, the two major HDL subclasses, are able to modulate energy metabolism of skeletal muscle cells. Differentiated mouse and primary human skeletal muscle myotubes were used to investigate the influences of human HDL2 and HDL3 on glucose and fatty uptake and oxidation. HDL-induced changes in lipid distribution and mRNA expression of genes related to energy substrate metabolism, mitochondrial function, and HDL receptors were studied with human myotubes. Additionally, we examined the effects of apoA-I and discoidal, reconstituted HDL particles on substrate metabolism. In mouse myotubes, HDL subclasses strongly enhanced glycolysis upon high and low glucose concentrations. HDL3 caused a minor increase in ATP-linked respiration upon glucose conditioning but HDL2 improved complex I-mediated mitochondrial respiration upon fatty acid treatment. In human myotubes, glucose metabolism was attenuated but fatty acid uptake and oxidation were markedly increased by both HDL subclasses, which also increased mRNA expression of genes related to fatty acid metabolism and HDL receptors. Finally, both HDL subclasses induced incorporation of oleic acid into different lipid classes. These results, demonstrating that HDL subclasses enhance fatty acid oxidation in human myotubes but improve anaerobic metabolism in mouse myotubes, support the role of HDL as a circulating modulator of energy metabolism. Exact mechanisms and components of HDL causing the change, require further investigation.
Collapse
Affiliation(s)
- Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Emilia Lähteenmäki
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Tiia Eklund
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Hege G Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Research Unit for Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Matti Jauhiainen
- Department of Public Health and Welfare, Minerva Foundation Institute for Medical Research and Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Maarit Lehti
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
5
|
Knapp M, Łukaszuk B, Lisowska A, Hirnle T, Górski J, Chabowski A, Mikłosz A. Multivessel Coronary Artery Disease Complicated by Diabetes Mellitus Has a Relatively Small Effect on Endothelial and Lipoprotein Lipases Expression in the Human Atrial Myocardium and Coronary Perivascular Adipose Tissue. Int J Mol Sci 2023; 24:13552. [PMID: 37686357 PMCID: PMC10487606 DOI: 10.3390/ijms241713552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Endothelial (EL) and lipoprotein (LPL) lipases are enzymes involved in lipoproteins metabolism and formation of atherosclerosis, a pathological feature of coronary artery disease (CAD). This paper examines the role of the lipases in the right atrial appendage (RAA) and coronary perivascular adipose tissue (PVAT) of patients with CAD alone or with accompanying diabetes. Additionally, correlation analysis for plasma concentration of the lipases, apolipoproteins (ApoA-ApoJ) and blood lipids (Chol, HDL-C, LDL-C, TAG) was performed. We observed that CAD had little effect on the lipases gene/protein levels in the RAA, while their transcript content was elevated in the PVAT of diabetic CAD patients. Interestingly, the RAA was characterized by higher expression of EL/LPL (EL: +1-fold for mRNA, +5-fold for protein; LPL: +2.8-fold for mRNA, +12-fold for protein) compared to PVAT. Furthermore, ApoA1 plasma concentration was decreased, whereas ApoC1 and ApoH were increased in the patients with CAD and/or diabetes. The concentrations of ApoC3 and ApoD were strongly positively correlated with TAG content in the blood, and the same was true for ApoB with respect to LDL-C and total cholesterol. Although plasma concentrations of EL/LPL were elevated in the patients with diabetes, CAD alone had little effect on blood, myocardial and perivascular fat expression of the lipases.
Collapse
Affiliation(s)
- Małgorzata Knapp
- Department of Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (M.K.); (A.L.); (T.H.)
| | - Bartłomiej Łukaszuk
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C Street, 15-222 Bialystok, Poland; (B.L.); (A.C.)
| | - Anna Lisowska
- Department of Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (M.K.); (A.L.); (T.H.)
| | - Tomasz Hirnle
- Department of Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (M.K.); (A.L.); (T.H.)
| | - Jan Górski
- Faculty of Health Sciences, University of Lomza, 18-400 Lomza, Poland;
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C Street, 15-222 Bialystok, Poland; (B.L.); (A.C.)
| | - Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C Street, 15-222 Bialystok, Poland; (B.L.); (A.C.)
| |
Collapse
|
6
|
Busnelli M, Manzini S, Colombo A, Franchi E, Bonacina F, Chiara M, Arnaboldi F, Donetti E, Ambrogi F, Oleari R, Lettieri A, Horner D, Scanziani E, Norata GD, Chiesa G. Lack of ApoA-I in ApoEKO Mice Causes Skin Xanthomas, Worsening of Inflammation, and Increased Coronary Atherosclerosis in the Absence of Hyperlipidemia. Arterioscler Thromb Vasc Biol 2022; 42:839-856. [PMID: 35587694 PMCID: PMC9205301 DOI: 10.1161/atvbaha.122.317790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background: HDL (high-density lipoprotein) and its major protein component, apoA-I (apolipoprotein A-I), play a unique role in cholesterol homeostasis and immunity. ApoA-I deficiency in hyperlipidemic, atheroprone mice was shown to drive cholesterol accumulation and inflammatory cell activation/proliferation. The present study was aimed at investigating the impact of apoA-I deficiency on lipid deposition and local/systemic inflammation in normolipidemic conditions. Methods: ApoE deficient mice, apoE/apoA-I double deficient (DKO) mice, DKO mice overexpressing human apoA-I, and C57Bl/6J control mice were fed normal laboratory diet until 30 weeks of age. Plasma lipids were quantified, atherosclerosis development at the aortic sinus and coronary arteries was measured, skin ultrastructure was evaluated by electron microscopy. Blood and lymphoid organs were characterized through histological, immunocytofluorimetric, and whole transcriptome analyses. Results: DKO were characterized by almost complete HDL deficiency and by plasma total cholesterol levels comparable to control mice. Only DKO showed xanthoma formation and severe inflammation in the skin-draining lymph nodes, whose transcriptome analysis revealed a dramatic impairment in energy metabolism and fatty acid oxidation pathways. An increased presence of CD4+ T effector memory cells was detected in blood, spleen, and skin-draining lymph nodes of DKO. A worsening of atherosclerosis at the aortic sinus and coronary arteries was also observed in DKO versus apoE deficient. Human apoA-I overexpression in the DKO background was able to rescue the skin phenotype and halt atherosclerosis development. Conclusions: HDL deficiency, in the absence of hyperlipidemia, is associated with severe alterations of skin morphology, aortic and coronary atherosclerosis, local and systemic inflammation.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Matteo Chiara
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health (F. Arnaboldi, E.D.), Università degli Studi di Milano, Italy
| | - Elena Donetti
- Department of Biomedical Sciences for Health (F. Arnaboldi, E.D.), Università degli Studi di Milano, Italy
| | - Federico Ambrogi
- Department of Clinical Sciences and Community Health (F. Ambrogi), Università degli Studi di Milano, Italy
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - David Horner
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Eugenio Scanziani
- Department of Veterinary Medicine (E.S.), Università degli Studi di Milano, Italy.,Mouse and Animal Pathology Laboratory (MAPLab), Fondazione UniMi, Milan, Italy (E.S.)
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy.,Centro per lo Studio dell'Aterosclerosi, Bassini Hospital, Cinisello B, Milan, Italy (G.D.N.)
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| |
Collapse
|
7
|
Consumption of Low Dose Fucoxanthin Does Not Prevent Hepatic and Adipose Inflammation and Fibrosis in Mouse Models of Diet-Induced Obesity. Nutrients 2022; 14:nu14112280. [PMID: 35684079 PMCID: PMC9183127 DOI: 10.3390/nu14112280] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 11/23/2022] Open
Abstract
Fucoxanthin (FCX) is a xanthophyll carotenoid present in brown seaweed. The goal of this study was to examine whether FCX supplementation could attenuate obesity-associated metabolic abnormalities, fibrosis, and inflammation in two diet-induced obesity (DIO) mouse models. C57BL/6J mice were fed either a high-fat/high-sucrose/high-cholesterol (HFC) diet or a high-fat/high-sucrose (HFS) diet. The former induces more severe liver injury than the latter model. In the first study, male C57BL/6J mice were fed an HFC diet, or an HFC diet containing 0.015% or 0.03% (w/w) FCX powder for 12 weeks to develop obesity-induced nonalcoholic steatohepatitis (NASH). In the second study, mice were fed an HFS diet or an HFS diet containing 0.01% FCX powder for 8 weeks. FCX did not change body weight gain and serum lipid profiles compared to the HFC or HFS controls. No significant differences were present in liver triglyceride and total cholesterol, hepatic fat accumulation, and serum alanine aminotransferase levels between control and FCX-fed mice regardless of whether they were on an HFC or HFS diet. FCX did not mitigate mRNA abundance of genes involved in lipid synthesis, cholesterol metabolism, inflammation, and fibrosis in the liver and white adipose tissue, while hepatic fatty acid β-oxidation genes were significantly elevated by FCX in both HFC and HFS feeding studies. Additionally, in the soleus muscle, FCX supplementation significantly elevated genes that regulate mitochondrial biogenesis and fatty acid β-oxidation, concomitantly increasing mitochondrial DNA copy number, compared with HFC. In summary, FCX supplementation had minor effects on hepatic and white adipose inflammation and fibrosis in two different DIO mouse models.
Collapse
|
8
|
Opoku E, Berisha S, Brubaker G, Robinet P, Smith JD. Oxidant resistant human apolipoprotein A-I functions similarly to the unmodified human isoform in delaying atherosclerosis progression and promoting atherosclerosis regression in hyperlipidemic mice. PLoS One 2022; 17:e0259751. [PMID: 35120132 PMCID: PMC8815868 DOI: 10.1371/journal.pone.0259751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Transgenic overexpression of apolipoprotein A-I (apoA1) has been shown to delay atherosclerosis lesion progression and promote lesion regression in mouse models; however, apoA1 is subject to oxidation by myeloperoxidase (MPO) and loss of function. The activity of oxidant resistant human apoA1 was compared to unmodified human apoA1 in mouse models of atherosclerosis progression and regression. METHODS AND RESULTS Human apoA1 and the MPO oxidant resistant 4WF isoform transgenic mice were bred to LDL receptor deficient (LDLr KO) mice and fed a western-type diet. High level expression of these human apoA1 isoforms did not lead to increased HDL-cholesterol levels on the LDLr KO background. In males and females, lesion progression was studied over time, and both apoA1 and 4WF transgenic mice vs. LDLr KO mice had significant and similar delayed lesion progression and reduced non-HDL cholesterol. Using time points with equivalent lesion areas, lesion regression was initiated by feeding the mice a low-fat control diet containing a microsomal triglyceride transfer protein inhibitor for 7 weeks. Lesions regressed more in the male apoA1 and 4WF transgenics vs. the LDLr KO, but the 4WF isoform was not superior to the unmodified isoform in promoting lesion regression. CONCLUSIONS Both human apoA1 and the 4WF MPO oxidant resistant apoA1 isoform delayed lesion progression and promoted lesion regression in LDLr KO mice, with more pronounced effects in males than females; moreover, the 4WF isoform functioned similarly to the unmodified human apoA1 isoform.
Collapse
Affiliation(s)
- Emmanuel Opoku
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Stela Berisha
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Gregory Brubaker
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Peggy Robinet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jonathan D. Smith
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
9
|
The effect of cranberry consumption on lipid metabolism and inflammation in human apo A-I transgenic mice fed a high-fat and high-cholesterol diet. Br J Nutr 2021; 126:183-190. [PMID: 33059793 DOI: 10.1017/s0007114520004080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lipid metabolism and inflammation contribute to CVD development. This study investigated whether the consumption of cranberries (CR; Vaccinium macrocarpon) can alter HDL metabolism and prevent inflammation in mice expressing human apo A-I transgene (hApoAITg), which have similar HDL profiles to those of humans. Male hApoAITg mice were fed a modified American Institute of Nutrition-93M high-fat/high-cholesterol diet (16 % fat, 0·25 % cholesterol, w/w; n 15) or the high-fat/high-cholesterol diet containing CR (5 % dried CR powder, w/w, n 16) for 8 weeks. There were no significant differences in body weight between the groups. Serum total cholesterol, non-HDL-cholesterol and TAG concentrations were significantly lower in the control than CR group with no significant differences in serum HDL-cholesterol and apoA-I. Mice fed CR showed significantly lower serum lecithin-cholesterol acyltransferase activity than the control. Liver weight and steatosis were not significantly different between the groups, but hepatic expression of genes involved in cholesterol metabolism was significantly lower in the CR group. In the epididymal white adipose tissue (eWAT), the CR group showed higher weights with decreased expression of genes for lipogenesis and fatty acid oxidation. The mRNA abundance of F4/80, a macrophage marker and the numbers of crown-like structures were less in the CR group. In the soleus muscle, the CR group also demonstrated higher expression of genes for fatty acid β-oxidation and mitochondrial biogenesis than those of the control. In conclusion, although CR consumption elicited minor effects on HDL metabolism, it prevented obesity-induced inflammation in eWAT with concomitant alterations in soleus muscle energy metabolism.
Collapse
|
10
|
Kothari V, Tang J, He Y, Kramer F, Kanter JE, Bornfeldt KE. ADAM17 Boosts Cholesterol Efflux and Downstream Effects of High-Density Lipoprotein on Inflammatory Pathways in Macrophages. Arterioscler Thromb Vasc Biol 2021; 41:1854-1873. [PMID: 33882688 PMCID: PMC8159900 DOI: 10.1161/atvbaha.121.315145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
| | - Jingjing Tang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
| | - Yi He
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
| | - Jenny E. Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
| | - Karin E. Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98109
| |
Collapse
|
11
|
Yiu JHC, Chan KS, Cheung J, Li J, Liu Y, Wang Y, Fung WWL, Cai J, Cheung SWM, Dorweiler B, Wan EYF, Tso P, Xu A, Woo CW. Gut Microbiota-Associated Activation of TLR5 Induces Apolipoprotein A1 Production in the Liver. Circ Res 2020; 127:1236-1252. [PMID: 32820707 PMCID: PMC7580858 DOI: 10.1161/circresaha.120.317362] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Dysbiosis of gut microbiota plays an important role in cardiovascular diseases but the molecular mechanisms are complex. An association between gut microbiome and the variance in HDL-C (high-density lipoprotein-cholesterol) level was suggested in a human study. Besides, dietary fat was shown to increase both HDL-C and LDL-C (low-density lipoprotein-cholesterol) levels. We speculate that certain types of gut bacteria responding to dietary fat may help to regulate HDL-C level and potentially affect atherosclerotic development. Objective: We aimed to investigate whether and how high-fat diet (HFD)-associated gut microbiota regulated HDL-C level. Methods and Results: We found that HFD increased gut flagellated bacteria population in mice. The increase in HDL-C level was adopted by mice receiving fecal microbiome transplantation from HFD-fed mouse donors. HFD led to increased hepatic but not circulating flagellin, and deletion of TLR5 (Toll-like receptor 5), a receptor sensing flagellin, suppressed HFD-stimulated HDL-C and ApoA1 (apolipoprotein A1) levels. Overexpression of TLR5 in the liver of TLR5-knockout mice was able to partially restore the production of ApoA1 and HDL-C levels. Mechanistically, TLR5 activation by flagellin in primary hepatocytes stimulated ApoA1 production through the transcriptional activation responding to the binding of NF-κB (nuclear factor-κB) on Apoa1 promoter region. Furthermore, oral supplementation of flagellin was able to stimulate hepatic ApoA1 production and HDL-C level and decrease atherosclerotic lesion size in apolipoprotein E-deficient (Apoe−/−) mice without triggering hepatic and systemic inflammation. The stimulation of ApoA1 production was also seen in human ApoA1-transgenic mice treated with oral flagellin. Conclusions: Our finding suggests that commensal flagellated bacteria in gut can facilitate ApoA1 and HDL-C productions in liver through activation of TLR5 in hepatocytes. Hepatic TLR5 may be a potential drug target to increase ApoA1.
Collapse
Affiliation(s)
- Jensen H C Yiu
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Kam-Suen Chan
- Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Jamie Cheung
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Jin Li
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Medicine (J.L., Y.L., A.X.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Endocrinology, Second Affiliated Hospital, Shanxi Medical University, China (J.L.)
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Medicine (J.L., Y.L., A.X.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Yao Wang
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - William W L Fung
- Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Jieling Cai
- Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Samson W M Cheung
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Bernhard Dorweiler
- Department of Vascular and Endovascular Surgery, University Hospital Cologne, Germany (B.D.)
| | - Eric Y F Wan
- Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Family Medicine and Primary Care (E.Y.F.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Patrick Tso
- Metabolic Phenotyping Center, Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, OH (P.T.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Medicine (J.L., Y.L., A.X.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Connie W Woo
- State Key Laboratory of Pharmaceutical Biotechnology (J.H.C.Y., J. Cheung, J.L., Y.L., Y.W., J.Cai, S.W.M.C., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China.,Department of Pharmacology and Pharmacy (J.H.C.Y., K.-S.C., J. Cheung, Y.W., W.W.L.F., J. Cai, S.W.M.C., E.Y.F.W., A.X., C.W.W.), Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| |
Collapse
|
12
|
Kluck GEG, Durham KK, Yoo JA, Trigatti BL. High Density Lipoprotein and Its Precursor Protein Apolipoprotein A1 as Potential Therapeutics to Prevent Anthracycline Associated Cardiotoxicity. Front Cardiovasc Med 2020; 7:65. [PMID: 32411725 PMCID: PMC7198830 DOI: 10.3389/fcvm.2020.00065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/06/2020] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular disease and cancer are the leading causes of death in developed societies. Despite their effectiveness, many cancer therapies exhibit deleterious cardiovascular side effects such as cardiotoxicity and heart failure. The cardiotoxic effects of anthracyclines such as doxorubicin are the most well-characterized of cardiotoxic anti-cancer therapies. While other anti-neoplastic drugs also induce cardiotoxicity, often leading to heart failure, they are beyond the scope of this review. This review first summarizes the mechanisms of doxorubicin-induced cardiotoxicity. It then reviews emerging preclinical evidence that high density lipoprotein and its precursor protein apolipoprotein A1, which are known for their protective effects against ischemic cardiovascular disease, may also protect against doxorubicin-induced cardiotoxicity both directly and indirectly, when used therapeutically.
Collapse
Affiliation(s)
- George E. G. Kluck
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Kristina K. Durham
- Faculty of Health Sciences, Institute of Applied Health Sciences, School of Rehabilitation Sciences, McMaster University, Hamilton, ON, Canada
| | - Jeong-Ah Yoo
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Bernardo L. Trigatti
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| |
Collapse
|
13
|
van der Vorst EPC, Biessen EAL, Donners MMPC. Letter by van der Vorst et al Regarding Article, "Anti-Inflammatory Effects of HDL (High-Density Lipoprotein) in Macrophages Predominate Over Proinflammatory Effects in Atherosclerotic Plaques". Arterioscler Thromb Vasc Biol 2020; 40:e31-e32. [PMID: 31967904 DOI: 10.1161/atvbaha.119.313725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Emiel P C van der Vorst
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands (E.P.C.v.d.V., E.A.L.B., M.M.P.C.D.), RWTH Aachen University, Germany.,Institute for Molecular Cardiovascular Research (IMCAR) (E.P.C.v.d.V., E.A.L.B.), RWTH Aachen University, Germany.,Interdisciplinary Center for Clinical Research (IZKF) (E.P.C.v.d.V.), RWTH Aachen University, Germany.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Germany (E.P.C.v.d.V.).,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (E.P.C.v.d.V)
| | - Erik A L Biessen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands (E.P.C.v.d.V., E.A.L.B., M.M.P.C.D.), RWTH Aachen University, Germany.,Institute for Molecular Cardiovascular Research (IMCAR) (E.P.C.v.d.V., E.A.L.B.), RWTH Aachen University, Germany
| | - Marjo M P C Donners
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands (E.P.C.v.d.V., E.A.L.B., M.M.P.C.D.), RWTH Aachen University, Germany
| |
Collapse
|
14
|
Fotakis P, Kothari V, Thomas DG, Westerterp M, Molusky MM, Altin E, Abramowicz S, Wang N, He Y, Heinecke JW, Bornfeldt KE, Tall AR. Anti-Inflammatory Effects of HDL (High-Density Lipoprotein) in Macrophages Predominate Over Proinflammatory Effects in Atherosclerotic Plaques. Arterioscler Thromb Vasc Biol 2019; 39:e253-e272. [PMID: 31578081 DOI: 10.1161/atvbaha.119.313253] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE HDL (high-density lipoprotein) infusion reduces atherosclerosis in animal models and is being evaluated as a treatment in humans. Studies have shown either anti- or proinflammatory effects of HDL in macrophages, and there is no consensus on the underlying mechanisms. Here, we interrogate the effects of HDL on inflammatory gene expression in macrophages. Approach and Results: We cultured bone marrow-derived macrophages, treated them with reconstituted HDL or HDL isolated from APOA1Tg;Ldlr-/- mice, and challenged them with lipopolysaccharide. Transcriptional profiling showed that HDL exerts a broad anti-inflammatory effect on lipopolysaccharide-induced genes and proinflammatory effect in a subset of genes enriched for chemokines. Cholesterol removal by POPC (1-palmitoyl-2-oleoyl-glycero-3-phosphocholine) liposomes or β-methylcyclodextrin mimicked both pro- and anti-inflammatory effects of HDL, whereas cholesterol loading by POPC/cholesterol-liposomes or acetylated LDL (low-density lipoprotein) before HDL attenuated these effects, indicating that these responses are mediated by cholesterol efflux. While early anti-inflammatory effects reflect reduced TLR (Toll-like receptor) 4 levels, late anti-inflammatory effects are due to reduced IFN (interferon) receptor signaling. Proinflammatory effects occur late and represent a modified endoplasmic reticulum stress response, mediated by IRE1a (inositol-requiring enzyme 1a)/ASK1 (apoptosis signal-regulating kinase 1)/p38 MAPK (p38 mitogen-activated protein kinase) signaling, that occurs under conditions of extreme cholesterol depletion. To investigate the effects of HDL on inflammatory gene expression in myeloid cells in atherosclerotic lesions, we injected reconstituted HDL into Apoe-/- or Ldlr-/- mice fed a Western-type diet. Reconstituted HDL infusions produced anti-inflammatory effects in lesion macrophages without any evidence of proinflammatory effects. CONCLUSIONS Reconstituted HDL infusions in hypercholesterolemic atherosclerotic mice produced anti-inflammatory effects in lesion macrophages suggesting a beneficial therapeutic effect of HDL in vivo.
Collapse
Affiliation(s)
- Panagiotis Fotakis
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle (V.K., Y.H., J.W.H., K.E.B.)
| | - David G Thomas
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Marit Westerterp
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.).,Department of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands (M.W.)
| | - Matthew M Molusky
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Elissa Altin
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Sandra Abramowicz
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Nan Wang
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| | - Yi He
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle (V.K., Y.H., J.W.H., K.E.B.)
| | - Jay W Heinecke
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle (V.K., Y.H., J.W.H., K.E.B.)
| | - Karin E Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle (V.K., Y.H., J.W.H., K.E.B.).,Department of Pathology, University of Washington, Seattle (K.E.B.)
| | - Alan R Tall
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York (P.F., D.G.T., M.W., M.M.M., E.A., S.A., N.W., A.R.T.)
| |
Collapse
|
15
|
Barrett TJ, Distel E, Murphy AJ, Hu J, Garshick MS, Ogando Y, Liu J, Vaisar T, Heinecke JW, Berger JS, Goldberg IJ, Fisher EA. Apolipoprotein AI) Promotes Atherosclerosis Regression in Diabetic Mice by Suppressing Myelopoiesis and Plaque Inflammation. Circulation 2019; 140:1170-1184. [PMID: 31567014 PMCID: PMC6777860 DOI: 10.1161/circulationaha.119.039476] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Despite robust cholesterol lowering, cardiovascular disease risk remains increased in patients with diabetes mellitus. Consistent with this, diabetes mellitus impairs atherosclerosis regression after cholesterol lowering in humans and mice. In mice, this is attributed in part to hyperglycemia-induced monocytosis, which increases monocyte entry into plaques despite cholesterol lowering. In addition, diabetes mellitus skews plaque macrophages toward an atherogenic inflammatory M1 phenotype instead of toward the atherosclerosis-resolving M2 state typical with cholesterol lowering. Functional high-density lipoprotein (HDL), typically low in patients with diabetes mellitus, reduces monocyte precursor proliferation in murine bone marrow and has anti-inflammatory effects on human and murine macrophages. Our study aimed to test whether raising functional HDL levels in diabetic mice prevents monocytosis, reduces the quantity and inflammation of plaque macrophages, and enhances atherosclerosis regression after cholesterol lowering. METHODS Aortic arches containing plaques developed in Ldlr-/- mice were transplanted into either wild-type, diabetic wild-type, or diabetic mice transgenic for human apolipoprotein AI, which have elevated functional HDL. Recipient mice all had low levels of low-density lipoprotein cholesterol to promote plaque regression. After 2 weeks, plaques in recipient mouse aortic grafts were examined. RESULTS Diabetic wild-type mice had impaired atherosclerosis regression, which was normalized by raising HDL levels. This benefit was linked to suppressed hyperglycemia-driven myelopoiesis, monocytosis, and neutrophilia. Increased HDL improved cholesterol efflux from bone marrow progenitors, suppressing their proliferation and monocyte and neutrophil production capacity. In addition to reducing circulating monocytes available for recruitment into plaques, in the diabetic milieu, HDL suppressed the general recruitability of monocytes to inflammatory sites and promoted plaque macrophage polarization to the M2, atherosclerosis-resolving state. There was also a decrease in plaque neutrophil extracellular traps, which are atherogenic and increased by diabetes mellitus. CONCLUSIONS Raising apolipoprotein AI and functional levels of HDL promotes multiple favorable changes in the production of monocytes and neutrophils and in the inflammatory environment of atherosclerotic plaques of diabetic mice after cholesterol lowering and may represent a novel approach to reduce cardiovascular disease risk in people with diabetes mellitus.
Collapse
Affiliation(s)
- Tessa J. Barrett
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Emilie Distel
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Andrew J. Murphy
- Haematopoiesis and Leukocyte Biology, Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, NY 10016, USA
| | - Michael S. Garshick
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Yoscar Ogando
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Jianhua Liu
- Department of Surgery, Mount Sinai School of Medicine, New York, NY, USA
| | - Tomas Vaisar
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle
| | - Jay W. Heinecke
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle
| | - Jeffrey S. Berger
- Department of Medicine, Divisions of Cardiology and Hematology, Department of Surgery, Division of Vascular Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Ira J. Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY 10016, USA
| | - Edward A. Fisher
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
- Department of Microbiology and Immunology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
16
|
Neufeld EB, Sato M, Gordon SM, Durbhakula V, Francone N, Aponte A, Yilmaz G, Sviridov D, Sampson M, Tang J, Pryor M, Remaley AT. ApoA-I-Mediated Lipoprotein Remodeling Monitored with a Fluorescent Phospholipid. BIOLOGY 2019; 8:E53. [PMID: 31336888 PMCID: PMC6784057 DOI: 10.3390/biology8030053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
We describe simple, sensitive and robust methods to monitor lipoprotein remodeling and cholesterol and apolipoprotein exchange, using fluorescent Lissamine Rhodamine B head-group tagged phosphatidylethanolamine (*PE) as a lipoprotein reference marker. Fluorescent Bodipy cholesterol (*Chol) and *PE directly incorporated into whole plasma lipoproteins in proportion to lipoprotein cholesterol and phospholipid mass, respectively. *Chol, but not *PE, passively exchanged between isolated plasma lipoproteins. Fluorescent apoA-I (*apoA-I) specifically bound to high-density lipoprotein (HDL) and remodeled *PE- and *Chol-labeled synthetic lipoprotein-X multilamellar vesicles (MLV) into a pre-β HDL-like particle containing *PE, *Chol, and *apoA-I. Fluorescent MLV-derived *PE specifically incorporated into plasma HDL, whereas MLV-derived *Chol incorporation into plasma lipoproteins was similar to direct *Chol incorporation, consistent with apoA-I-mediated remodeling of fluorescent MLV to HDL with concomitant exchange of *Chol between lipoproteins. Based on these findings, we developed a model system to study lipid transfer by depositing fluorescent *PE and *Chol-labeled on calcium silicate hydrate crystals, forming dense lipid-coated donor particles that are readily separated from acceptor lipoprotein particles by low-speed centrifugation. Transfer of *PE from donor particles to mouse plasma lipoproteins was shown to be HDL-specific and apoA-I-dependent. Transfer of donor particle *PE and *Chol to HDL in whole human plasma was highly correlated. Taken together, these studies suggest that cell-free *PE efflux monitors apoA-I functionality.
Collapse
Affiliation(s)
- Edward B Neufeld
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Masaki Sato
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott M Gordon
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vinay Durbhakula
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Francone
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angel Aponte
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gizem Yilmaz
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Denis Sviridov
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maureen Sampson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jingrong Tang
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Milton Pryor
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Méndez-Lara KA, Farré N, Santos D, Rivas-Urbina A, Metso J, Sánchez-Quesada JL, Llorente-Cortes V, Errico TL, Lerma E, Jauhiainen M, Martín-Campos JM, Alonso N, Escolà-Gil JC, Blanco-Vaca F, Julve J. Human ApoA-I Overexpression Enhances Macrophage-Specific Reverse Cholesterol Transport but Fails to Prevent Inherited Diabesity in Mice. Int J Mol Sci 2019; 20:E655. [PMID: 30717414 PMCID: PMC6387412 DOI: 10.3390/ijms20030655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 12/18/2022] Open
Abstract
Human apolipoprotein A-I (hApoA-I) overexpression improves high-density lipoprotein (HDL) function and the metabolic complications of obesity. We used a mouse model of diabesity, the db/db mouse, to examine the effects of hApoA-I on the two main functional properties of HDL, i.e., macrophage-specific reverse cholesterol transport (m-RCT) in vivo and the antioxidant potential, as well as the phenotypic features of obesity. HApoA-I transgenic (hA-I) mice were bred with nonobese control (db/+) mice to generate hApoA-I-overexpressing db/+ offspring, which were subsequently bred to obtain hA-I-db/db mice. Overexpression of hApoA-I significantly increased weight gain and the incidence of fatty liver in db/db mice. Weight gain was mainly explained by the increased caloric intake of hA-I-db/db mice (>1.2-fold). Overexpression of hApoA-I also produced a mixed type of dyslipidemia in db/db mice. Despite these deleterious effects, the overexpression of hApoA-I partially restored m-RCT in db/db mice to levels similar to nonobese control mice. Moreover, HDL from hA-I-db/db mice also enhanced the protection against low-density lipoprotein (LDL) oxidation compared with HDL from db/db mice. In conclusion, overexpression of hApoA-I in db/db mice enhanced two main anti-atherogenic HDL properties while exacerbating weight gain and the fatty liver phenotype. These adverse metabolic side-effects were also observed in obese mice subjected to long-term HDL-based therapies in independent studies and might raise concerns regarding the use of hApoA-I-mediated therapy in obese humans.
Collapse
Affiliation(s)
- Karen Alejandra Méndez-Lara
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| | - Núria Farré
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
| | - David Santos
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| | - Andrea Rivas-Urbina
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| | - Jari Metso
- Minerva Foundation Institute for Medical Research, Biomedicum 2U and National Institute for Health and Welfare, Genomics and Biomarkers Unit, FIN-00290 Helsinki, Finland.
| | - José Luis Sánchez-Quesada
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| | - Vicenta Llorente-Cortes
- CSIC-ICCC-IIB-Sant Pau i CSIC-Institut d'Investigacions Biomèdiques de Barcelona (IIBB), 08025 Barcelona, Spain.
- CIBER de Enfermedades Cardiovasculares, CIBERCV, 28029 Madrid, Spain.
| | - Teresa L Errico
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| | - Enrique Lerma
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain.
- Departament de Patologia, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain.
- Departament de Ciències Morfològiques, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U and National Institute for Health and Welfare, Genomics and Biomarkers Unit, FIN-00290 Helsinki, Finland.
| | - Jesús M Martín-Campos
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| | - Núria Alonso
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
- Servei d'Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain.
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| | - Francisco Blanco-Vaca
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain.
| | - Josep Julve
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain.
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain.
| |
Collapse
|
18
|
Aboumsallem JP, Mishra M, Amin R, Muthuramu I, Kempen H, De Geest B. Successful treatment of established heart failure in mice with recombinant HDL (Milano). Br J Pharmacol 2018; 175:4167-4182. [PMID: 30079544 PMCID: PMC6177616 DOI: 10.1111/bph.14463] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The pleiotropic properties of HDL may exert beneficial effects on the myocardium. The effect of recombinant HDLMilano on established heart failure was evaluated in C57BL/6 mice. EXPERIMENTAL APPROACH Mice were subjected to transverse aortic constriction (TAC) or sham operation at the age of 14 weeks. Eight weeks later, TAC and sham mice were each randomized into three different groups. Reference groups were killed at day 56 after the operation for baseline analysis. Five i.p. injections of recombinant HDLMilano (MDCO-216), 100 mg·kg-1 , or an equivalent volume of control buffer were administered with a 48 h interval starting at day 56. Endpoint analyses in the control buffer groups and in the MDCO-216 groups were executed at day 65. KEY RESULTS Lung weight in MDCO-216 TAC mice was 25.3% lower than in reference TAC mice and 27.9% lower than in control buffer TAC mice and was similar in MDCO-216 sham mice. MDCO-216 significantly decreased interstitial fibrosis and increased relative vascularity compared to reference TAC mice and control buffer TAC mice. The peak rate of isovolumetric relaxation in MDCO-216 TAC mice was 30.4 and 36.3% higher than in reference TAC mice and control buffer TAC mice respectively. Nitro-oxidative stress and myocardial apoptosis were significantly reduced in MDCO-216 TAC mice compared to control buffer TAC mice. CONCLUSIONS AND IMPLICATIONS MDCO-216 improves diastolic function, induces regression of interstitial fibrosis and normalizes lung weight in mice with established heart failure. Recombinant HDL may emerge as a treatment modality in heart failure.
Collapse
Affiliation(s)
- Joseph Pierre Aboumsallem
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Mudit Mishra
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Ruhul Amin
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Ilayaraja Muthuramu
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Herman Kempen
- The Medicines Company (Schweiz) GmbHZürichSwitzerland
| | - Bart De Geest
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| |
Collapse
|
19
|
Wacker BK, Dronadula N, Bi L, Stamatikos A, Dichek DA. Apo A-I (Apolipoprotein A-I) Vascular Gene Therapy Provides Durable Protection Against Atherosclerosis in Hyperlipidemic Rabbits. Arterioscler Thromb Vasc Biol 2018; 38:206-217. [PMID: 29122817 PMCID: PMC5746433 DOI: 10.1161/atvbaha.117.309565] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 10/30/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Gene therapy that expresses apo A-I (apolipoprotein A-I) from vascular wall cells has promise for preventing and reversing atherosclerosis. Previously, we reported that transduction of carotid artery endothelial cells with a helper-dependent adenoviral (HDAd) vector expressing apo A-I reduced early (4 weeks) fatty streak development in fat-fed rabbits. Here, we tested whether the same HDAd could provide long-term protection against development of more complex lesions. APPROACH AND RESULTS Fat-fed rabbits (n=25) underwent bilateral carotid artery gene transfer, with their left and right common carotids randomized to receive either a control vector (HDAdNull) or an apo A-I-expressing vector (HDAdApoAI). Twenty-four additional weeks of high-fat diet yielded complex intimal lesions containing lipid-rich macrophages as well as smooth muscle cells, often in a lesion cap. Twenty-four weeks after gene transfer, high levels of apo A-I mRNA (median ≥250-fold above background) were present in all HDAdApoAI-treated arteries. Compared with paired control HDAdNull-treated arteries in the same rabbit, HDAdApoAI-treated arteries had 30% less median intimal lesion volume (P=0.03), with concomitant reductions (23%-32%) in intimal lipid, macrophage, and smooth muscle cell content (P≤0.05 for all). HDAdApoAI-treated arteries also had decreased intimal inflammatory markers. VCAM-1 (vascular cell adhesion molecule-1)-stained area was reduced by 36% (P=0.03), with trends toward lower expression of ICAM-1 (intercellular adhesion molecule-1), MCP-1 (monocyte chemoattractant protein 1), and TNF-α (tumor necrosis factor-α; 13%-39% less; P=0.06-0.1). CONCLUSIONS In rabbits with severe hyperlipidemia, transduction of vascular endothelial cells with an apo A-I-expressing HDAd yields at least 24 weeks of local apo A-I expression that durably reduces atherosclerotic lesion growth and intimal inflammation.
Collapse
Affiliation(s)
- Bradley K Wacker
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Nagadhara Dronadula
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Lianxiang Bi
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Alexis Stamatikos
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - David A Dichek
- From the Department of Medicine, University of Washington School of Medicine, Seattle.
| |
Collapse
|
20
|
Durham KK, Chathely KM, Mak KC, Momen A, Thomas CT, Zhao YY, MacDonald ME, Curtis JM, Husain M, Trigatti BL. HDL protects against doxorubicin-induced cardiotoxicity in a scavenger receptor class B type 1-, PI3K-, and Akt-dependent manner. Am J Physiol Heart Circ Physiol 2017; 314:H31-H44. [PMID: 28986362 DOI: 10.1152/ajpheart.00521.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Doxorubicin is a widely used chemotherapeutic with deleterious cardiotoxic side effects. HDL has been shown to protect cardiomyocytes in vitro against doxorubicin-induced apoptosis. Scavenger receptor class B type 1 (SR-B1), a high-affinity HDL receptor, mediates cytoprotective signaling by HDL through Akt. Here, we assessed whether increased HDL levels protect against doxorubicin-induced cardiotoxicity in vivo and in cardiomyocytes in culture and explored the intracellular signaling mechanisms involved, particularly the role of SR-B1. Transgenic mice with increased HDL levels through overexpression of human apolipoprotein A1 (apoA1Tg/Tg) and wild-type mice (apoA1+/+) with normal HDL levels were treated repeatedly with doxorubicin. After treatment, apoA1+/+ mice displayed cardiac dysfunction, as evidenced by reduced left ventricular end-systolic pressure and +dP/d t, and histological analysis revealed cardiomyocyte atrophy and increased cardiomyocyte apoptosis after doxorubicin treatment. In contrast, apoA1Tg/Tg mice were protected against doxorubicin-induced cardiac dysfunction and cardiomyocyte atrophy and apoptosis. When SR-B1 was knocked out, however, overexpression of apoA1 did not protect against doxorubicin-induced cardiotoxicity. Using primary neonatal mouse cardiomyocytes and human immortalized ventricular cardiomyocytes in combination with genetic knockout, inhibitors, or siRNA-mediated knockdown, we demonstrated that SR-B1 is required for HDL-mediated protection of cardiomyocytes against doxorubicin-induced apoptosis in vitro via a pathway involving phosphatidylinositol 3-kinase and Akt1/2. Our findings provide proof of concept that raising apoA1 to supraphysiological levels can dramatically protect against doxorubicin-induced cardiotoxicity via a pathway that is mediated by SR-B1 and involves Akt1/2 activation in cardiomyocytes. NEW & NOTEWORTHY We have identified an important role for the scavenger receptor class B type 1 in facilitating high-density lipoprotein-mediated protection of cardiomyocytes against stress-induced apoptosis and shown that increasing plasma high-density lipoprotein protects against the deleterious side effects of the chemotherapeutic and cardiotoxic drug doxorubicin.
Collapse
Affiliation(s)
- Kristina K Durham
- Medical Sciences Graduate Program, McMaster University , Hamilton, Ontario , Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, Ontario , Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| | - Kevin M Chathely
- Medical Sciences Graduate Program, McMaster University , Hamilton, Ontario , Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, Ontario , Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| | - Kei Cheng Mak
- Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, Ontario , Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| | - Abdul Momen
- Division of Cell and Molecular Biology, Toronto General Hospital Research Institute and Heart and Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto , Toronto, Ontario , Canada
| | - Cyrus T Thomas
- Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| | - Yuan-Yuan Zhao
- Lipid Chemistry Group, Department of Agricultural, Food, and Nutritional Sciences, University of Alberta , Edmonton, Alberta , Canada
| | - Melissa E MacDonald
- Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| | - Jonathan M Curtis
- Lipid Chemistry Group, Department of Agricultural, Food, and Nutritional Sciences, University of Alberta , Edmonton, Alberta , Canada
| | - Mansoor Husain
- Division of Cell and Molecular Biology, Toronto General Hospital Research Institute and Heart and Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto , Toronto, Ontario , Canada.,Department of Medicine, University of Toronto , Toronto, Ontario , Canada
| | - Bernardo L Trigatti
- Medical Sciences Graduate Program, McMaster University , Hamilton, Ontario , Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, Ontario , Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
21
|
Selective HDL-Raising Human Apo A-I Gene Therapy Counteracts Cardiac Hypertrophy, Reduces Myocardial Fibrosis, and Improves Cardiac Function in Mice with Chronic Pressure Overload. Int J Mol Sci 2017; 18:ijms18092012. [PMID: 28930153 PMCID: PMC5618660 DOI: 10.3390/ijms18092012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/14/2022] Open
Abstract
Epidemiological studies support an independent inverse association between high-density lipoprotein (HDL) cholesterol levels and heart failure incidence. The effect of selective HDL-raising adeno-associated viral serotype 8-human apolipoprotein (apo) A-I (AAV8-A-I) gene transfer on cardiac remodeling induced by transverse aortic constriction (TAC) was evaluated in C57BL/6 low-density lipoprotein receptor-deficient mice. Septal wall thickness and cardiomyocyte cross-sectional area were reduced by 16.5% (p < 0.001) and by 13.8% (p < 0.01), respectively, eight weeks after TAC in AAV8-A-I mice (n = 24) compared to control mice (n = 39). Myocardial capillary density was 1.11-fold (p < 0.05) higher and interstitial cardiac fibrosis was 45.3% (p < 0.001) lower in AAV8-A-I TAC mice than in control TAC mice. Lung weight and atrial weight were significantly increased in control TAC mice compared to control sham mice, but were not increased in AAV8-A-I TAC mice. The peak rate of isovolumetric contraction was 1.19-fold (p < 0.01) higher in AAV8-A-I TAC mice (n = 17) than in control TAC mice (n = 29). Diastolic function was also significantly enhanced in AAV8-A-I TAC mice compared to control TAC mice. Nitro-oxidative stress and apoptosis were significantly reduced in the myocardium of AAV8-A-I TAC mice compared to control TAC mice. In conclusion, selective HDL-raising human apo A-I gene transfer potently counteracts the development of pressure overload-induced cardiomyopathy.
Collapse
|
22
|
Mao R, Meng S, Gu Q, Araujo-Gutierrez R, Kumar S, Yan Q, Almazan F, Youker KA, Fu Y, Pownall HJ, Cooke JP, Miller YI, Fang L. AIBP Limits Angiogenesis Through γ-Secretase-Mediated Upregulation of Notch Signaling. Circ Res 2017; 120:1727-1739. [PMID: 28325782 DOI: 10.1161/circresaha.116.309754] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/22/2022]
Abstract
RATIONALE Angiogenesis improves perfusion to the ischemic tissue after acute vascular obstruction. Angiogenesis in pathophysiological settings reactivates signaling pathways involved in developmental angiogenesis. We showed previously that AIBP (apolipoprotein A-I [apoA-I]-binding protein)-regulated cholesterol efflux in endothelial cells controls zebra fish embryonic angiogenesis. OBJECTIVE This study is to determine whether loss of AIBP affects angiogenesis in mice during development and under pathological conditions and to explore the underlying molecular mechanism. METHODS AND RESULTS In this article, we report the generation of AIBP knockout (Apoa1bp-/-) mice, which are characterized of accelerated postnatal retinal angiogenesis. Mechanistically, AIBP triggered relocalization of γ-secretase from lipid rafts to nonlipid rafts where it cleaved Notch. Consistently, AIBP treatment enhanced DLL4 (delta-like ligand 4)-stimulated Notch activation in human retinal endothelial cells. Increasing high-density lipoprotein levels in Apoa1bp-/- mice by crossing them with apoA-I transgenic mice rescued Notch activation and corrected dysregulated retinal angiogenesis. Notably, the retinal vessels in Apoa1bp-/- mice manifested normal pericyte coverage and vascular integrity. Similarly, in the subcutaneous Matrigel plug assay, which mimics ischemic/inflammatory neovascularization, angiogenesis was dramatically upregulated in Apoa1bp-/- mice and associated with a profound inhibition of Notch activation and reduced expression of downstream targets. Furthermore, loss of AIBP increased vascular density and facilitated the recovery of blood vessel perfusion function in a murine hindlimb ischemia model. In addition, AIBP expression was significantly increased in human patients with ischemic cardiomyopathy. CONCLUSIONS Our data reveal a novel mechanistic connection between AIBP-mediated cholesterol metabolism and Notch signaling, implicating AIBP as a possible druggable target to modulate angiogenesis under pathological conditions.
Collapse
Affiliation(s)
- Renfang Mao
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Shu Meng
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Qilin Gu
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Raquel Araujo-Gutierrez
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Sandeep Kumar
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Qing Yan
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Felicidad Almazan
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Keith A Youker
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Yingbin Fu
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Henry J Pownall
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - John P Cooke
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Yury I Miller
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.)
| | - Longhou Fang
- From the Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences (R.M., S.M., Q.G., R.A.-G., Q.Y., J.P.C., L.F.), Houston Methodist DeBakey Heart and Vascular Center, Department of Cardiology (R.A.-G., K.A.Y.), Department of Bioenergetics (H.J.P.), Houston Methodist Research Institute, TX; Department of Ophthalmology, Baylor College of Medicine, Houston, TX (S.K., Y.F.); and Department of Medicine, University of California, San Diego, La Jolla (F.A., Y.I.M.).
| |
Collapse
|
23
|
Wacker BK, Dronadula N, Zhang J, Dichek DA. Local Vascular Gene Therapy With Apolipoprotein A-I to Promote Regression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 37:316-327. [PMID: 27932352 DOI: 10.1161/atvbaha.116.308258] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/28/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Gene therapy, delivered directly to the blood vessel wall, could potentially prevent atherosclerotic lesion growth and promote atherosclerosis regression. Previously, we reported that a helper-dependent adenoviral (HDAd) vector expressing apolipoprotein A-I (apoA-I) in carotid endothelium of fat-fed rabbits reduced early (4 weeks) atherosclerotic lesion growth. Here, we tested whether the same HDAd-delivered to the existing carotid atherosclerotic lesions-could promote regression. APPROACH AND RESULTS Rabbits (n=26) were fed a high-fat diet for 7 months, then treated with bilateral carotid gene transfer. One carotid was infused with an HDAd expressing apoA-I (HDAdApoAI) and the other with a control nonexpressing HDAd (HDAdNull). The side with HDAdApoAI was randomized. Rabbits were then switched to regular chow, lowering their plasma cholesterols by over 70%. ApoA-I mRNA and protein were detected in HDAdApoAI-transduced arteries. After 7 weeks of gene therapy, compared with HDAdNull-treated arteries in the same rabbits, HDAdApoAI-treated arteries had significantly less vascular cell adhesion molecule-1 expression (28%; P=0.04) along with modest but statistically insignificant trends toward decreased intimal lesion volume, lipid and macrophage content, and intercellular adhesion molecule-1 expression (9%-21%; P=0.1-0.4). Post hoc subgroup analysis of rabbits with small-to-moderate-sized lesions (n=20) showed that HDAdApoAI caused large reductions in lesion volume, lipid content, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 expression (30%-50%; P≤0.04 for all). Macrophage content was reduced by 30% (P=0.06). There was a significant interaction (P=0.02) between lesion size and treatment efficacy. CONCLUSIONS Even when administered on a background of aggressive lowering of plasma cholesterol, local HDAdApoAI vascular gene therapy may promote rapid regression of small-to-moderate-sized atherosclerotic lesions.
Collapse
Affiliation(s)
- Bradley K Wacker
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Nagadhara Dronadula
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Jingwan Zhang
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - David A Dichek
- From the Department of Medicine, University of Washington School of Medicine, Seattle.
| |
Collapse
|
24
|
Iqbal AJ, Barrett TJ, Taylor L, McNeill E, Manmadhan A, Recio C, Carmineri A, Brodermann MH, White GE, Cooper D, DiDonato JA, Zamanian-Daryoush M, Hazen SL, Channon KM, Greaves DR, Fisher EA. Acute exposure to apolipoprotein A1 inhibits macrophage chemotaxis in vitro and monocyte recruitment in vivo. eLife 2016; 5. [PMID: 27572261 PMCID: PMC5030090 DOI: 10.7554/elife.15190] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
Apolipoprotein A1 (apoA1) is the major protein component of high-density lipoprotein (HDL) and has well documented anti-inflammatory properties. To better understand the cellular and molecular basis of the anti-inflammatory actions of apoA1, we explored the effect of acute human apoA1 exposure on the migratory capacity of monocyte-derived cells in vitro and in vivo. Acute (20-60 min) apoA1 treatment induced a substantial (50-90%) reduction in macrophage chemotaxis to a range of chemoattractants. This acute treatment was anti-inflammatory in vivo as shown by pre-treatment of monocytes prior to adoptive transfer into an on-going murine peritonitis model. We find that apoA1 rapidly disrupts membrane lipid rafts, and as a consequence, dampens the PI3K/Akt signalling pathway that coordinates reorganization of the actin cytoskeleton and cell migration. Our data strengthen the evidence base for therapeutic apoA1 infusions in situations where reduced monocyte recruitment to sites of inflammation could have beneficial outcomes.
Collapse
Affiliation(s)
- Asif J Iqbal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Tessa J Barrett
- Division of Cardiology, NYU School of Medicine, New York, United States.,Department of Medicine, NYU School of Medicine, New York, United States.,Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, United States
| | - Lewis Taylor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Eileen McNeill
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom.,John Radcliffe Hospital, Oxford, United Kingdom.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Arun Manmadhan
- Division of Cardiology, NYU School of Medicine, New York, United States.,Department of Medicine, NYU School of Medicine, New York, United States.,Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, United States
| | - Carlota Recio
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alfredo Carmineri
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | - Gemma E White
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Dianne Cooper
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Joseph A DiDonato
- Department of Cellular and Molecular Medicine, Lerner Research Institute of the Cleveland Clinic, Cleavland, United States
| | - Maryam Zamanian-Daryoush
- Department of Cellular and Molecular Medicine, Lerner Research Institute of the Cleveland Clinic, Cleavland, United States
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute of the Cleveland Clinic, Cleavland, United States
| | - Keith M Channon
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom.,John Radcliffe Hospital, Oxford, United Kingdom.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Edward A Fisher
- Division of Cardiology, NYU School of Medicine, New York, United States.,Department of Medicine, NYU School of Medicine, New York, United States.,Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, United States
| |
Collapse
|
25
|
Sakata N, Hoshii Y, Nakamura T, Kiyama M, Arai H, Omoto M, Morimatsu M, Ishihara T. Colocalization of Apolipoprotein AI in Various Kinds of Systemic Amyloidosis. J Histochem Cytochem 2016; 53:237-42. [PMID: 15684336 DOI: 10.1369/jhc.4a6387.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Apolipoprotein AI (apoAI), a major component of high-density lipoproteins, is one of the major amyloid fibril proteins and a minor constituent of the senile plaques observed in Alzheimer's disease. We examined colocalization of apoAI in various kinds of systemic amyloidosis in this study. Forty-three of 48 formalin-fixed paraffin-embedded heart specimens with various forms of systemic amyloidosis reacted immunohistochemically with anti-human apoAI antibody. ApoAI was also detected in water-extracted amyloid material by immunoblotting. In addition, we observed colocalization of apoAI and murine amyloid A (AA) amyloidosis in human apoAI transgenic mice. This is the first report of colocalization of apoAI with amyloid deposits in various forms of human systemic amyloidosis and murine AA amyloidosis in human apoAI transgenic mice. ApoAI may not always be a major component of amyloid fibrils, even when it is present in systemic amyloid deposits.
Collapse
Affiliation(s)
- Naohiro Sakata
- Department of Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Canfrán-Duque A, Lin CS, Goedeke L, Suárez Y, Fernández-Hernando C. Micro-RNAs and High-Density Lipoprotein Metabolism. Arterioscler Thromb Vasc Biol 2016; 36:1076-84. [PMID: 27079881 DOI: 10.1161/atvbaha.116.307028] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
Improved prevention and treatment of cardiovascular diseases is one of the challenges in Western societies, where ischemic heart disease and stroke are the leading cause of death. Early epidemiological studies have shown an inverse correlation between circulating high-density lipoprotein-cholesterol (HDL-C) and cardiovascular diseases. The cardioprotective effect of HDL is because of its ability to remove cholesterol from plaques in the artery wall to the liver for excretion by a process known as reverse cholesterol transport. Numerous studies have reported the role that micro-RNAs (miRNA) play in the regulation of the different steps in reverse cholesterol transport, including HDL biogenesis, cholesterol efflux, and cholesterol uptake in the liver and bile acid synthesis and secretion. Because of their ability to control different aspects of HDL metabolism and function, miRNAs have emerged as potential therapeutic targets to combat cardiovascular diseases. In this review, we summarize the recent advances in the miRNA-mediated control of HDL metabolism. We also discuss how HDL particles serve as carriers of miRNAs and the potential use of HDL-containing miRNAs as cardiovascular diseases biomarkers.
Collapse
Affiliation(s)
- Alberto Canfrán-Duque
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Chin-Sheng Lin
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Leigh Goedeke
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Yajaira Suárez
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.).
| |
Collapse
|
27
|
Baldán Á, de Aguiar Vallim TQ. miRNAs and High-Density Lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2053-2061. [PMID: 26869447 DOI: 10.1016/j.bbalip.2016.01.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
Altered lipoprotein metabolism plays a key role during atherogenesis. For over 50years, epidemiological data have fueled the proposal that HDL-cholesterol (HDL-c) in circulation is inversely correlated to cardiovascular risk. However, the atheroprotective role of HDL is currently the focus of much debate and remains an active field of research. The emerging picture from research in the past decade suggests that HDL function, rather than HDL-c content, is important in disease. Recent developments demonstrate that miRNAs play an important role in fine-tuning the expression of key genes involved in HDL biogenesis, lipidation, and clearance, as well as in determining the amounts of HDL-c in circulation. Thus, it has been proposed that miRNAs that affect HDL metabolism might be exploited therapeutically in patients. Whether HDL-based therapies, alone or in combination with LDL-based treatments (e.g. statins), provide superior outcomes in patients has been recently questioned by human genetics studies and clinical trials. The switch in focus from "HDL-cholesterol" to "HDL function" opens a new paradigm to understand the physiology and therapeutic potential of HDL, and to find novel modulators of cardiovascular risk. In this review we summarize the current knowledge on the regulation of HDL metabolism and function by miRNAs. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Ángel Baldán
- Edward A. Doisy Department of Biochemistry & Molecular Biology, Center for Cardiovascular Research, and Liver Center, 1100 S. Grand Blvd., Saint Louis University, Saint Louis, MO 63104, United States.
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, 650 Charles E. Young Drive S, A2-237 CHS, UCLA Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
28
|
Black LL, Srivastava R, Schoeb TR, Moore RD, Barnes S, Kabarowski JH. Cholesterol-Independent Suppression of Lymphocyte Activation, Autoimmunity, and Glomerulonephritis by Apolipoprotein A-I in Normocholesterolemic Lupus-Prone Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:4685-98. [PMID: 26466956 DOI: 10.4049/jimmunol.1500806] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/21/2015] [Indexed: 11/19/2022]
Abstract
Apolipoprotein (Apo)A-I, the major lipid-binding protein of high-density lipoprotein, can prevent autoimmunity and suppress inflammation in hypercholesterolemic mice by attenuating lymphocyte cholesterol accumulation and removing tissue-oxidized lipids. However, whether ApoA-I mediates immune-suppressive or anti-inflammatory effects under normocholesterolemic conditions and the mechanisms involved remain unresolved. We transferred bone marrow from systemic lupus erythematosus (SLE)-prone Sle123 mice into normal, ApoA-I-knockout (ApoA-I(-/-)) and ApoA-I-transgenic (ApoA-I(tg)) mice. Increased ApoA-I in ApoA-I(tg) mice suppressed CD4(+) T and B cell activation without changing lymphocyte cholesterol levels or reducing major ApoA-I-binding oxidized fatty acids. Unexpectedly, oxidized fatty acid peroxisome proliferator-activated receptor γ ligands 13- and 9-hydroxyoctadecadienoic acid were increased in lymphocytes of autoimmune ApoA-I(tg) mice. ApoA-I reduced Th1 cells independently of changes in CD4(+)Foxp3(+) regulatory T cells or CD11c(+) dendritic cell activation and migration. Follicular helper T cells, germinal center B cells, and autoantibodies were also lower in ApoA-I(tg) mice. Transgenic ApoA-I also improved SLE-mediated glomerulonephritis. However, ApoA-I deficiency did not have the opposite effects on autoimmunity or glomerulonephritis, possibly as the result of compensatory increases in ApoE on high-density lipoprotein. We conclude that, although compensatory mechanisms prevent the proinflammatory effects of ApoA-I deficiency in normocholesterolemic mice, increasing ApoA-I can attenuate lymphocyte activation and autoimmunity in SLE independently of cholesterol transport, possibly through oxidized fatty acid peroxisome proliferator-activated receptor γ ligands, and it can reduce renal inflammation in glomerulonephritis.
Collapse
Affiliation(s)
- Leland L Black
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Roshni Srivastava
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Trenton R Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Ray D Moore
- Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Janusz H Kabarowski
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
29
|
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: 84] [Impact Index Per Article: 8.4] [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
|
30
|
Berisha SZ, Brubaker G, Kasumov T, Hung KT, DiBello PM, Huang Y, Li L, Willard B, Pollard KA, Nagy LE, Hazen SL, Smith JD. HDL from apoA1 transgenic mice expressing the 4WF isoform is resistant to oxidative loss of function. J Lipid Res 2015; 56:653-664. [PMID: 25561462 DOI: 10.1194/jlr.m056754] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HDL functions are impaired by myeloperoxidase (MPO), which selectively targets and oxidizes human apoA1. We previously found that the 4WF isoform of human apoA1, in which the four tryptophan residues are substituted with phenylalanine, is resistant to MPO-mediated loss of function. The purpose of this study was to generate 4WF apoA1 transgenic mice and compare functional properties of the 4WF and wild-type human apoA1 isoforms in vivo. Male mice had significantly higher plasma apoA1 levels than females for both isoforms of human apoA1, attributed to different production rates. With matched plasma apoA1 levels, 4WF transgenics had a trend for slightly less HDL-cholesterol versus human apoA1 transgenics. While 4WF transgenics had 31% less reverse cholesterol transport (RCT) to the plasma compartment, equivalent RCT to the liver and feces was observed. Plasma from both strains had similar ability to accept cholesterol and facilitate ex vivo cholesterol efflux from macrophages. Furthermore, we observed that 4WF transgenic HDL was partially (∼50%) protected from MPO-mediated loss of function while human apoA1 transgenic HDL lost all ABCA1-dependent cholesterol acceptor activity. In conclusion, the structure and function of HDL from 4WF transgenic mice was not different than HDL derived from human apoA1 transgenic mice.
Collapse
Affiliation(s)
- Stela Z Berisha
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Greg Brubaker
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Takhar Kasumov
- Department of Gastroenterology and Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Kimberly T Hung
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Patricia M DiBello
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ying Huang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ling Li
- Department of Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Belinda Willard
- Department of Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Katherine A Pollard
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Laura E Nagy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Jonathan D Smith
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195.
| |
Collapse
|
31
|
Kardassis D, Gafencu A, Zannis VI, Davalos A. Regulation of HDL genes: transcriptional, posttranscriptional, and posttranslational. Handb Exp Pharmacol 2015; 224:113-179. [PMID: 25522987 DOI: 10.1007/978-3-319-09665-0_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
HDL regulation is exerted at multiple levels including regulation at the level of transcription initiation by transcription factors and signal transduction cascades; regulation at the posttranscriptional level by microRNAs and other noncoding RNAs which bind to the coding or noncoding regions of HDL genes regulating mRNA stability and translation; as well as regulation at the posttranslational level by protein modifications, intracellular trafficking, and degradation. The above mechanisms have drastic effects on several HDL-mediated processes including HDL biogenesis, remodeling, cholesterol efflux and uptake, as well as atheroprotective functions on the cells of the arterial wall. The emphasis is on mechanisms that operate in physiologically relevant tissues such as the liver (which accounts for 80% of the total HDL-C levels in the plasma), the macrophages, the adrenals, and the endothelium. Transcription factors that have a significant impact on HDL regulation such as hormone nuclear receptors and hepatocyte nuclear factors are extensively discussed both in terms of gene promoter recognition and regulation but also in terms of their impact on plasma HDL levels as was revealed by knockout studies. Understanding the different modes of regulation of this complex lipoprotein may provide useful insights for the development of novel HDL-raising therapies that could be used to fight against atherosclerosis which is the underlying cause of coronary heart disease.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, Heraklion, Crete, 71110, Greece,
| | | | | | | |
Collapse
|
32
|
Stukas S, Robert J, Lee M, Kulic I, Carr M, Tourigny K, Fan J, Namjoshi D, Lemke K, DeValle N, Chan J, Wilson T, Wilkinson A, Chapanian R, Kizhakkedathu JN, Cirrito JR, Oda MN, Wellington CL. Intravenously injected human apolipoprotein A-I rapidly enters the central nervous system via the choroid plexus. J Am Heart Assoc 2014; 3:e001156. [PMID: 25392541 PMCID: PMC4338702 DOI: 10.1161/jaha.114.001156] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Background Brain lipoprotein metabolism is dependent on lipoprotein particles that resemble plasma high‐density lipoproteins but that contain apolipoprotein (apo) E rather than apoA‐I as their primary protein component. Astrocytes and microglia secrete apoE but not apoA‐I; however, apoA‐I is detectable in both cerebrospinal fluid and brain tissue lysates. The route by which plasma apoA‐I enters the central nervous system is unknown. Methods and Results Steady‐state levels of murine apoA‐I in cerebrospinal fluid and interstitial fluid are 0.664 and 0.120 μg/mL, respectively, whereas brain tissue apoA‐I is ≈10% to 15% of its levels in liver. Recombinant, fluorescently tagged human apoA‐I injected intravenously into mice localizes to the choroid plexus within 30 minutes and accumulates in a saturable, dose‐dependent manner in the brain. Recombinant, fluorescently tagged human apoA‐I accumulates in the brain for 2 hours, after which it is eliminated with a half‐life of 10.3 hours. In vitro, human apoA‐I is specifically bound, internalized, and transported across confluent monolayers of primary human choroid plexus epithelial cells and brain microvascular endothelial cells. Conclusions Following intravenous injection, recombinant human apoA‐I rapidly localizes predominantly to the choroid plexus. Because apoA‐I mRNA is undetectable in murine brain, our results suggest that plasma apoA‐I, which is secreted from the liver and intestine, gains access to the central nervous system primarily by crossing the blood–cerebrospinal fluid barrier via specific cellular mediated transport, although transport across the blood–brain barrier may also contribute to a lesser extent.
Collapse
Affiliation(s)
- Sophie Stukas
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Jerome Robert
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Michael Lee
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Iva Kulic
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Michael Carr
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Katherine Tourigny
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Dhananjay Namjoshi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Kalistyne Lemke
- Children's Hospital Oakland Research Institute, Oakland, CA (K.L., N.D.V., M.N.O.)
| | - Nicole DeValle
- Children's Hospital Oakland Research Institute, Oakland, CA (K.L., N.D.V., M.N.O.)
| | - Jeniffer Chan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Tammy Wilson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Anna Wilkinson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| | - Rafi Chapanian
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.) Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada (R.C., J.N.K.)
| | - Jayachandran N Kizhakkedathu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.) Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada (R.C., J.N.K.)
| | - John R Cirrito
- Department of Neurology, Washington University, St. Louis, MO (J.R.C.)
| | - Michael N Oda
- Children's Hospital Oakland Research Institute, Oakland, CA (K.L., N.D.V., M.N.O.)
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (S.S., J.R., M.L., I.K., M.C., K.T., J.F., D.N., J.C., T.W., A.W., R.C., J.N.K., C.L.W.)
| |
Collapse
|
33
|
Meyers L, Groover CJ, Douglas J, Lee S, Brand D, Levin MC, Gardner LA. A role for Apolipoprotein A-I in the pathogenesis of multiple sclerosis. J Neuroimmunol 2014; 277:176-85. [PMID: 25468275 DOI: 10.1016/j.jneuroim.2014.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/13/2023]
Abstract
Apolipoprotein A1 (Apo A-I), the most abundant component of high-density lipoprotein (HDL), is an anti-inflammatory molecule, yet its potential role in the pathogenesis of multiple sclerosis (MS) has not been fully investigated. In this study, Western blot analyses of human plasma showed differential Apo A-I expression in healthy controls compared to MS patients. Further, primary progressive MS patients had less plasma Apo A-I than other forms of MS. Using experimental allergic encephalomyelitis (EAE) as a model for MS, Apo A-I deficient mice exhibited worse clinical disease and more neurodegeneration concurrent with increased levels of pro-inflammatory cytokines compared to wild-type animals. These data suggest that Apo A-I plays a role in the pathogenesis of EAE, a model for MS, creating the possibility for agents that increase Apo A-I levels as potential therapies for MS.
Collapse
Affiliation(s)
| | | | | | - Sangmin Lee
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - David Brand
- Research Service VAMC, Memphis, TN 38104, United States
| | - Michael C Levin
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Lidia A Gardner
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
34
|
Fischer NO, Weilhammer DR, Dunkle A, Thomas C, Hwang M, Corzett M, Lychak C, Mayer W, Urbin S, Collette N, Chiun Chang J, Loots GG, Rasley A, Blanchette CD. Evaluation of nanolipoprotein particles (NLPs) as an in vivo delivery platform. PLoS One 2014; 9:e93342. [PMID: 24675794 PMCID: PMC3968139 DOI: 10.1371/journal.pone.0093342] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 03/05/2014] [Indexed: 12/05/2022] Open
Abstract
Nanoparticles hold great promise for the delivery of therapeutics, yet limitations remain with regards to the use of these nanosystems for efficient long-lasting targeted delivery of therapeutics, including imparting functionality to the platform, in vivo stability, drug entrapment efficiency and toxicity. To begin to address these limitations, we evaluated the functionality, stability, cytotoxicity, toxicity, immunogenicity and in vivo biodistribution of nanolipoprotein particles (NLPs), which are mimetics of naturally occurring high-density lipoproteins (HDLs). We found that a wide range of molecules could be reliably conjugated to the NLP, including proteins, single-stranded DNA, and small molecules. The NLP was also found to be relatively stable in complex biological fluids and displayed no cytotoxicity in vitro at doses as high as 320 µg/ml. In addition, we observed that in vivo administration of the NLP daily for 14 consecutive days did not induce significant weight loss or result in lesions on excised organs. Furthermore, the NLPs did not display overt immunogenicity with respect to antibody generation. Finally, the biodistribution of the NLP in vivo was found to be highly dependent on the route of administration, where intranasal administration resulted in prolonged retention in the lung tissue. Although only a select number of NLP compositions were evaluated, the findings of this study suggest that the NLP platform holds promise for use as both a targeted and non-targeted in vivo delivery vehicle for a range of therapeutics.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/genetics
- Antigens, Bacterial/metabolism
- Apolipoprotein E4/chemistry
- Apolipoprotein E4/genetics
- Apolipoprotein E4/metabolism
- Biomimetic Materials/chemical synthesis
- Biomimetic Materials/pharmacokinetics
- DNA, Bacterial/chemistry
- DNA, Bacterial/metabolism
- DNA, Single-Stranded/metabolism
- Dimyristoylphosphatidylcholine/chemistry
- Dimyristoylphosphatidylcholine/metabolism
- Drug Carriers
- Drug Stability
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Female
- Fluorescent Dyes
- Lipoproteins, HDL/chemical synthesis
- Lipoproteins, HDL/pharmacokinetics
- Male
- Mice
- Mice, Inbred BALB C
- Nanoparticles/chemistry
- Nanoparticles/toxicity
- Particle Size
- Phosphatidylcholines/chemistry
- Phosphatidylcholines/metabolism
- Pore Forming Cytotoxic Proteins/chemistry
- Pore Forming Cytotoxic Proteins/genetics
- Pore Forming Cytotoxic Proteins/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- Nicholas O. Fischer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Dina R. Weilhammer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Alexis Dunkle
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Cynthia Thomas
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Mona Hwang
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Michele Corzett
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Cheri Lychak
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Wasima Mayer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Salustra Urbin
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Nicole Collette
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Jiun Chiun Chang
- School of Natural Sciences, University of California Merced, Merced, California, United States of America
| | - Gabriela G. Loots
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- School of Natural Sciences, University of California Merced, Merced, California, United States of America
| | - Amy Rasley
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- * E-mail: (AR); (CB)
| | - Craig D. Blanchette
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- * E-mail: (AR); (CB)
| |
Collapse
|
35
|
Modulation of adipose tissue lipolysis and body weight by high-density lipoproteins in mice. Nutr Diabetes 2014; 4:e108. [PMID: 24567123 PMCID: PMC3940828 DOI: 10.1038/nutd.2014.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/05/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Obesity is associated with reduced levels of circulating high-density lipoproteins (HDLs) and its major protein, apolipoprotein (apo) A-I. As a result of the role of HDL and apoA-I in cellular lipid transport, low HDL and apoA-I may contribute directly to establishing or maintaining the obese condition. METHODS To test this, male C57BL/6 wild-type (WT), apoA-I deficient (apoA-I(-/-)) and apoA-I transgenic (apoA-I(tg/tg)) mice were fed obesogenic diets (ODs) and monitored for several clinical parameters. We also performed cell culture studies. RESULTS ApoA-I(-/-) mice gained significantly more body weight and body fat than WT mice over 20 weeks despite their reduced food intake. During a caloric restriction regime imposed on OD-fed mice, apoA-I deficiency significantly inhibited the loss of body fat as compared with WT mice. Reduced body fat loss with caloric restriction in apoA-I(-/-) mice was associated with blunted stimulated adipose tissue lipolysis as verified by decreased levels of phosphorylated hormone-sensitive lipase (p-HSL) and lipolytic enzyme mRNA. In contrast to apoA-I(-/-) mice, apoA-I(tg/tg) mice gained relatively less weight than WT mice, consistent with other reports. ApoA-I(tg/tg) mice showed increased adipose tissue lipolysis, verified by increased levels of p-HSL and lipolytic enzyme mRNA. In cell culture studies, HDL and apoA-I specifically increased catecholamine-induced lipolysis possibly through modulating the adipocyte plasma membrane cholesterol content. CONCLUSIONS Thus, apoA-I and HDL contribute to modulating body fat content by controlling the extent of lipolysis. ApoA-I and HDL are key components of lipid metabolism in adipose tissue and constitute new therapeutic targets in obesity.
Collapse
|
36
|
Liu M, Seo J, Allegood J, Bi X, Zhu X, Boudyguina E, Gebre AK, Avni D, Shah D, Sorci-Thomas MG, Thomas MJ, Shelness GS, Spiegel S, Parks JS. Hepatic apolipoprotein M (apoM) overexpression stimulates formation of larger apoM/sphingosine 1-phosphate-enriched plasma high density lipoprotein. J Biol Chem 2013; 289:2801-14. [PMID: 24318881 DOI: 10.1074/jbc.m113.499913] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Apolipoprotein M (apoM), a lipocalin family member, preferentially associates with plasma HDL and binds plasma sphingosine 1-phosphate (S1P), a signaling molecule active in immune homeostasis and endothelial barrier function. ApoM overexpression in ABCA1-expressing HEK293 cells stimulated larger nascent HDL formation, compared with cells that did not express apoM; however, the in vivo role of apoM in HDL metabolism remains poorly understood. To test whether hepatic apoM overexpression increases plasma HDL size, we generated hepatocyte-specific apoM transgenic (APOM Tg) mice, which had an ∼3-5-fold increase in plasma apoM levels compared with wild-type mice. Although HDL cholesterol concentrations were similar to wild-type mice, APOM Tg mice had larger plasma HDLs enriched in apoM, cholesteryl ester, lecithin:cholesterol acyltransferase, and S1P. Despite the presence of larger plasma HDLs in APOM Tg mice, in vivo macrophage reverse cholesterol transport capacity was similar to that in wild-type mice. APOM Tg mice had an ∼5-fold increase in plasma S1P, which was predominantly associated with larger plasma HDLs. Primary hepatocytes from APOM Tg mice generated larger nascent HDLs and displayed increased sphingolipid synthesis and S1P secretion. Inhibition of ceramide synthases in hepatocytes increased cellular S1P levels but not S1P secretion, suggesting that apoM is rate-limiting in the export of hepatocyte S1P. Our data indicate that hepatocyte-specific apoM overexpression generates larger nascent HDLs and larger plasma HDLs, which preferentially bind apoM and S1P, and stimulates S1P biosynthesis for secretion. The unique apoM/S1P-enriched plasma HDL may serve to deliver S1P to extrahepatic tissues for atheroprotection and may have other as yet unidentified functions.
Collapse
Affiliation(s)
- Mingxia Liu
- From the Departments of Pathology-Lipid Sciences and
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kempen HJ, Gomaraschi M, Bellibas SE, Plassmann S, Zerler B, Collins HL, Adelman SJ, Calabresi L, Wijngaard PLJ. Effect of repeated apoA-IMilano/POPC infusion on lipids, (apo)lipoproteins, and serum cholesterol efflux capacity in cynomolgus monkeys. J Lipid Res 2013; 54:2341-53. [PMID: 23828780 DOI: 10.1194/jlr.m033779] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MDCO-216, a complex of dimeric recombinant apoA-IMilano (apoA-IM) and palmitoyl-oleoyl-phosphatidylcholine (POPC), was administered to cynomolgus monkeys at 30, 100, and 300 mg/kg every other day for a total of 21 infusions, and effects on lipids, (apo)lipoproteins, and ex-vivo cholesterol efflux capacity were monitored. After 7 or 20 infusions, free cholesterol (FC) and phospholipids (PL) were strongly increased, and HDL-cholesterol (HDL-C), apoA-I, and apoA-II were strongly decreased. We then measured short-term effects on apoA-IM, lipids, and (apo)lipoproteins after the first or the last infusion. After the first infusion, PL and FC went up in the HDL region and also in the LDL and VLDL regions. ApoE shifted from HDL to LDL and VLDL regions, while ApoA-IM remained located in the HDL region. On day 41, ApoE levels were 8-fold higher than on day 1, and FC, PL, and apoE resided mostly in LDL and VLDL regions. Drug infusion quickly decreased the endogenous cholesterol esterification rate. ABCA1-mediated cholesterol efflux on day 41 was markedly increased, whereas scavenger receptor type B1 (SRB1) and ABCG1-mediated effluxes were only weakly increased. Strong increase of FC is due to sustained stimulation of ABCA1-mediated efflux, and drop in HDL and formation of large apoE-rich particles are due to lack of LCAT activation.
Collapse
|
38
|
Kasumov T, Willard B, Li L, Li M, Conger H, Buffa JA, Previs S, McCullough A, Hazen SL, Smith JD. 2H2O-based high-density lipoprotein turnover method for the assessment of dynamic high-density lipoprotein function in mice. Arterioscler Thromb Vasc Biol 2013; 33:1994-2003. [PMID: 23766259 DOI: 10.1161/atvbaha.113.301700] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE High-density lipoprotein (HDL) promotes reverse cholesterol transport from peripheral tissues to the liver for clearance. Reduced HDL-cholesterol (HDLc) is associated with atherosclerosis; however, as a predictor of cardiovascular disease, HDLc has limitations because it is not a direct marker of HDL functionality. Our objective was to develop a mass spectrometry-based method for the simultaneous measurement of HDLc and ApoAI kinetics in mice, using a single (2)H2O tracer, and use it to examine genetic and drug perturbations on HDL turnover in vivo. APPROACH AND RESULTS Mice were given (2)H2O in the drinking water, and serial blood samples were collected at different time points. HDLc and ApoAI gradually incorporated (2)H, allowing experimental measurement of fractional catabolic rates and production rates for HDLc and ApoAI. ApoE(-/-) mice displayed increased fractional catabolic rates (P<0.01) and reduced production rates of both HDLc and ApoAI (P<0.05) compared with controls. In human ApoAI transgenic mice, levels and production rates of HDLc and human ApoAI were strikingly higher than in wild-type mice. Myriocin, an inhibitor of sphingolipid synthesis, significantly increased both HDL flux and macrophage-to-feces reverse cholesterol transport, indicating compatibility of this HDL turnover method with the macrophage-specific reverse cholesterol transport assay. CONCLUSIONS (2)H2O-labeling can be used to measure HDLc and ApoAI flux in vivo, and to assess the role of genetic and pharmacological interventions on HDL turnover in mice. Safety, simplicity, and low cost of the (2)H2O-based HDL turnover approach suggest that this assay can be scaled for human use to study effects of HDL targeted therapies on dynamic HDL function.
Collapse
Affiliation(s)
- Takhar Kasumov
- Department of Gastroenterology and Hepatology, Case Western Reserve University School of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zamanian-Daryoush M, Lindner D, Tallant TC, Wang Z, Buffa J, Klipfell E, Parker Y, Hatala D, Parsons-Wingerter P, Rayman P, Yusufishaq MSS, Fisher EA, Smith JD, Finke J, DiDonato JA, Hazen SL. The cardioprotective protein apolipoprotein A1 promotes potent anti-tumorigenic effects. J Biol Chem 2013; 288:21237-21252. [PMID: 23720750 DOI: 10.1074/jbc.m113.468967] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Here, we show that apolipoprotein A1 (apoA1), the major protein component of high density lipoprotein (HDL), through both innate and adaptive immune processes, potently suppresses tumor growth and metastasis in multiple animal tumor models, including the aggressive B16F10L murine malignant melanoma model. Mice expressing the human apoA1 transgene (A1Tg) exhibited increased infiltration of CD11b(+) F4/80(+) macrophages with M1, anti-tumor phenotype, reduced tumor burden and metastasis, and enhanced survival. In contrast, apoA1-deficient (A1KO) mice showed markedly heightened tumor growth and reduced survival. Injection of human apoA1 into A1KO mice inoculated with tumor cells remarkably reduced both tumor growth and metastasis, enhanced survival, and promoted regression of both tumor and metastasis burden when administered following palpable tumor formation and metastasis development. Studies with apolipoprotein A2 revealed the anti-cancer therapeutic effect was specific to apoA1. In vitro studies ruled out substantial direct suppressive effects by apoA1 or HDL on tumor cells. Animal models defective in different aspects of immunity revealed both innate and adaptive arms of immunity contribute to complete apoA1 anti-tumor activity. This study reveals a potent immunomodulatory role for apoA1 in the tumor microenvironment, altering tumor-associated macrophages from a pro-tumor M2 to an anti-tumor M1 phenotype. Use of apoA1 to redirect in vivo elicited tumor-infiltrating macrophages toward tumor rejection may hold benefit as a potential cancer therapeutic.
Collapse
Affiliation(s)
- Maryam Zamanian-Daryoush
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | | | - Thomas C Tallant
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | - Zeneng Wang
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | - Jennifer Buffa
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | - Elizabeth Klipfell
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | | | | | - Patricia Parsons-Wingerter
- the John H. Glenn Research Center, National Aeronautics and Space Administration, Cleveland, Ohio 44135, and
| | | | | | - Edward A Fisher
- the Department of Cell Biology and the Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York 10016
| | - Jonathan D Smith
- From the Department of Cellular and Molecular Medicine,; Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195
| | | | - Joseph A DiDonato
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention
| | - Stanley L Hazen
- From the Department of Cellular and Molecular Medicine,; Center for Cardiovascular Diagnostics and Prevention,; Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195,.
| |
Collapse
|
40
|
Guo L, Ai J, Zheng Z, Howatt DA, Daugherty A, Huang B, Li XA. High density lipoprotein protects against polymicrobe-induced sepsis in mice. J Biol Chem 2013; 288:17947-53. [PMID: 23658016 DOI: 10.1074/jbc.m112.442699] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
HDL has been considered to be a protective factor in sepsis; however, most contributing studies were conducted using the endotoxic animal model, and evidence from clinically relevant septic animal models remains limited and controversial. Furthermore, little is known about the roles of HDL in sepsis other than LPS neutralization. In this study, we employed cecal ligation and puncture (CLP), a clinically relevant septic animal model, and utilized apoA-I knock-out (KO) and transgenic mice to elucidate the roles of HDL in sepsis. ApoA-I-KO mice were more susceptible to CLP-induced septic death as shown by the 47.1% survival of apoA-I-KO mice versus the 76.7% survival of C57BL/6J (B6) mice (p = 0.038). ApoA-I-KO mice had exacerbated inflammatory cytokine production during sepsis compared with B6 mice. Further study indicated that serum from apoA-I-KO mice displayed less capacity for LPS neutralization compared with serum from B6 mice. In addition, apoA-I-KO mice had less LPS clearance, reduced corticosterone generation, and impaired leukocyte recruitment in sepsis. In contrast to apoA-I-KO mice, apoA-I transgenic mice were moderately resistant to CLP-induced septic death compared with B6 mice. In conclusion, our findings reveal multiple protective roles of HDL in CLP-induced sepsis. In addition to its well established role in neutralization of LPS, HDL exerts its protection against sepsis through promoting LPS clearance and modulating corticosterone production and leukocyte recruitment. Our study supports efforts to raise HDL levels as a therapeutic approach for sepsis.
Collapse
Affiliation(s)
- Ling Guo
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Wang JB, Zhang YJ, Zhang Y, Guan J, Chen LY, Fu CH, Du HJ, Sheng Y, Zhou L, Si YF, Zhang Y. Negative correlation between serum syndecan-1 and apolipoprotein A1 in patients with type 2 diabetes mellitus. Acta Diabetol 2013; 50:111-5. [PMID: 20683626 DOI: 10.1007/s00592-010-0216-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 07/26/2010] [Indexed: 10/19/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) are involved in the regulation of cell growth, apoptosis and lipid metabolism in vitro. Syndecans are the primary form of HSPGs. Syndecan-1 is involved in the processes of cell growth, differentiation, adhesion, wound healing and inflammation. Additionally, as a sinusoidal transmembrane HSPG facing the plasma compartment, syndecan-1 is a promising target to be involved in lipoprotein physiology. We aimed to examine the possible correlation of syndecan-1 and lipid profile in type 2 diabetes mellitus. In this study, serum syndecan-1 was detected by ELISA, and potential correlations between syndecan-1 and triglyceride, cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, lipoprotein a, apolipoprotein (apo) B, apoA1 and apoB/apoA1 were analyzed. Forty-one patients with type 2 diabetes and 31 age-matched, non-diabetic healthy subjects (controls) were enrolled. Syndecan-1 in patients with diabetes (26.15 ± 2.42 ng/ml) was significantly higher than that of the controls (16.85 ± 1.98 ng/ml, t = -2.98, P = 0.005). Serum syndecan-1 level correlated negatively with apoA1 (r = -0.46, P = 0.003). Multiple regression analysis showed that apoA1 (b = -0.43, P = 0.003) was a predictor of serum syndecan-1 levels in subjects with type 2 diabetes.
Collapse
Affiliation(s)
- Jing-Bo Wang
- Department of Ophthalmology, The 309th Hospital of Chinese People's Liberation Army, Jia 17, Heishanhu Road, Haidian District, Beijing, 100091, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lyssenko NN, Nickel M, Tang C, Phillips MC. Factors controlling nascent high-density lipoprotein particle heterogeneity: ATP-binding cassette transporter A1 activity and cell lipid and apolipoprotein AI availability. FASEB J 2013; 27:2880-92. [PMID: 23543682 PMCID: PMC3688743 DOI: 10.1096/fj.12-216564] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nascent high-density lipoprotein (HDL) particles arise in different sizes. We have sought to uncover factors that control this size heterogeneity. Gel filtration, native PAGE, and protein cross-linking were used to analyze the size heterogeneity of nascent HDL produced by BHK-ABCA1, RAW 264.7, J774, and HepG2 cells under different levels of two factors considered as a ratio, the availability of apolipoprotein AI (apoAI) -accessible cell lipid, and concentration of extracellular lipid-free apoAI. Increases in the available cell lipid:apoAI ratio due to either elevated ATP-binding cassette transporter A1 (ABCA1) expression and activity or raised cell density (i.e., increasing numerator) shifted the production of nascent HDL from smaller particles with fewer apoAI molecules per particle and fewer molecules of choline-phospholipid and cholesterol per apoAI molecule to larger particles that contained more apoAI and more lipid per molecule of apoAI. A further shift to larger particles was observed in BHK-ABCA1 cells when the available cell lipid:apoAI ratio was raised still higher by decreasing the apoAI concentration (i.e., the denominator). These changes in nascent HDL biogenesis were reminiscent of the transition that occurs in the size composition of reconstituted HDL in response to an increasing initial lipid:apoAI molar ratio. Thus, the ratio of available cell lipid:apoAI is a fundamental cause of nascent HDL size heterogeneity, and rHDL formation is a good model of nascent HDL biogenesis.—Lyssenko, N. N., Nickel, M., Tang, C., Phillips, M. C. Factors controlling nascent high-density lipoprotein particle heterogeneity: ATP-binding cassette transporter A1 activity and cell lipid and apolipoprotein AI availability.
Collapse
Affiliation(s)
- Nicholas N Lyssenko
- Lipid Research Group, Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
43
|
Umemoto T, Han CY, Mitra P, Averill MM, Tang C, Goodspeed L, Omer M, Subramanian S, Wang S, Den Hartigh LJ, Wei H, Kim EJ, Kim J, O'Brien KD, Chait A. Apolipoprotein AI and high-density lipoprotein have anti-inflammatory effects on adipocytes via cholesterol transporters: ATP-binding cassette A-1, ATP-binding cassette G-1, and scavenger receptor B-1. Circ Res 2013; 112:1345-54. [PMID: 23501697 DOI: 10.1161/circresaha.111.300581] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
RATIONALE Macrophage accumulation in adipose tissue associates with insulin resistance and increased cardiovascular disease risk. We previously have shown that generation of reactive oxygen species and monocyte chemotactic factors after exposure of adipocytes to saturated fatty acids, such as palmitate, occurs via translocation of NADPH oxidase 4 into lipid rafts (LRs). The anti-inflammatory effects of apolipoprotein AI (apoAI) and high-density lipoprotein (HDL) on macrophages and endothelial cells seem to occur via cholesterol depletion of LRs. However, little is known concerning anti-inflammatory effects of HDL and apoAI on adipocytes. OBJECTIVE To determine whether apoAI and HDL inhibit inflammation in adipocytes and adipose tissue, and whether this is dependent on LRs. METHODS AND RESULTS In 3T3L-1 adipocytes, apoAI, HDL, and methyl-β-cyclodextrin inhibited chemotactic factor expression. ApoAI and HDL also disrupted LRs, reduced plasma membrane cholesterol content, inhibited NADPH oxidase 4 translocation into LRs, and reduced palmitate-induced reactive oxygen species generation and monocyte chemotactic factor expression. Silencing ATP-binding cassette A-1 abrogated the effect of apoAI, but not HDL, whereas silencing ATP-binding cassette G-1 or scavenger receptor B-1 abrogated the effect of HDL but not apoAI. In vivo, apoAI transgenic mice fed a high-fat, high-sucrose, cholesterol-containing diet showed reduced chemotactic factor and proinflammatory cytokine expression and reduced macrophage accumulation in adipose tissue. CONCLUSIONS ApoAI and HDL have anti-inflammatory effects in adipocytes and adipose tissue similar to their effects in other cell types. These effects are consistent with disruption and removal of cholesterol from LRs, which are regulated by cholesterol transporters, such as ATP-binding cassette A-1, ATP-binding cassette G-1, and scavenger receptor B-1.
Collapse
Affiliation(s)
- Tomio Umemoto
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mukhopadhyay R. Mouse models of atherosclerosis: explaining critical roles of lipid metabolism and inflammation. J Appl Genet 2013; 54:185-92. [PMID: 23361320 DOI: 10.1007/s13353-013-0134-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 10/27/2022]
Abstract
Atherosclerosis is the most common cause of death globally. It is a complex disease involving morphological and cellular changes in vascular walls. Studying molecular mechanism of the disease is hindered by disease complexity and lack of robust noninvasive diagnostics in human. Mouse models are the most popular animal models that allow researchers to study the mechanism of disease progression. In this review we discuss the advantage and development of mouse as a model for atherosclerotic research. Along with commonly used models, this review discusses strains that are used to study the role of two critical processes associated with the disease-lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, 784 028, India.
| |
Collapse
|
45
|
Stennett D, Oladeinde F, Wheatley A, Bryant J, Dilworth L, Asemota H. Effects of Dioscorea Polygonoides
(Jamaican Bitter Yam) Supplementation in Normocholesterolemic and Genetically Modified Hypercholesterolemic Mice Species. J Food Biochem 2013. [DOI: 10.1111/jfbc.12022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Dewayne Stennett
- Basic Medical Sciences Department; Biochemistry Section; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
| | - Frederick Oladeinde
- Chemistry Department; Morgan State University; Baltimore MD
- Center for Complementary and Alternative Medicine; School of Public Health & Policy; Morgan State University; Baltimore MD
| | - Andrew Wheatley
- Basic Medical Sciences Department; Biochemistry Section; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
- Biotechnology Centre; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
| | - Joseph Bryant
- Animal Core Facility Department; University of Maryland Biotechnology Institute; Baltimore MD
| | - Lowell Dilworth
- Basic Medical Sciences Department; Biochemistry Section; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
| | - Helen Asemota
- Basic Medical Sciences Department; Biochemistry Section; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
- Biotechnology Centre; University of the West Indies; Mona Campus Kingston 7 Jamaica W.I
- Shaw University Nanotechnology Initiative; Raleigh NC
| |
Collapse
|
46
|
Hung KT, Berisha SZ, Ritchey BM, Santore J, Smith JD. Red blood cells play a role in reverse cholesterol transport. Arterioscler Thromb Vasc Biol 2012; 32:1460-5. [PMID: 22499994 PMCID: PMC3360517 DOI: 10.1161/atvbaha.112.248971] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Reverse cholesterol transport (RCT) involves the removal of cholesterol from peripheral tissue for excretion in the feces. Here, we determined whether red blood cells (RBCs) can contribute to RCT. METHODS AND RESULTS We performed a series of studies in apolipoprotein AI-deficient mice where the high-density lipoprotein-mediated pathway of RCT is greatly diminished. RBCs carried a higher fraction of whole blood cholesterol than plasma in apolipoprotein AI-deficient mice, and as least as much of the labeled cholesterol derived from injected foam cells appeared in RBCs compared with plasma. To determine whether RBCs mediate RCT to the fecal compartment, we measured RCT in anemic and control apolipoprotein AI-deficient mice and found that anemia decreased RCT to the feces by over 35% after correcting for fecal mass. Transfusion of [(3)H]cholesterol-labeled RBCs led to robust delivery of the labeled cholesterol to the feces in apolipoprotein AI-deficient hosts. In wild-type mice, the majority of the blood cholesterol mass, as well as [(3)H]cholesterol derived from the injected foam cells, was found in plasma, and anemia did not significantly alter RCT to the feces after correction for fecal mass. CONCLUSIONS The RBC cholesterol pool is dynamic and facilitates RCT of peripheral cholesterol to the feces, particularly in the low high-density lipoprotein state.
Collapse
Affiliation(s)
- Kimberly T Hung
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
47
|
Marchesi M, Parolini C, Caligari S, Gilio D, Manzini S, Busnelli M, Cinquanta P, Camera M, Brambilla M, Sirtori CR, Chiesa G. Rosuvastatin does not affect human apolipoprotein A-I expression in genetically modified mice: a clue to the disputed effect of statins on HDL. Br J Pharmacol 2012; 164:1460-8. [PMID: 21486287 DOI: 10.1111/j.1476-5381.2011.01429.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Besides a significant reduction of low-density lipoprotein (LDL) cholesterol, statins moderately increase high-density lipoprotein (HDL) levels. In vitro studies have indicated that this effect may be the result of an increased expression of apolipoprotein (apo)A-I, the main protein component of HDL. The aim of the present study was to investigate in vivo the effect of rosuvastatin on apoA-I expression and secretion in a transgenic mouse model for human apoA-I. EXPERIMENTAL APPROACH Human apoA-I transgenic mice were treated for 28 days with 5, 10 or 20 mg·kg(-1) ·day(-1) of rosuvastatin, the most effective statin in raising HDL levels. Possible changes of apoA-I expression by treatment were investigated by quantitative real-time RT-PCR on RNA extracted from mouse livers. The human apoA-I secretion rate was determined in primary hepatocytes isolated from transgenic mice from each group after treatment. KEY RESULTS Rosuvastatin treatment with 5 and 10 mg·kg(-1) ·day(-1) did not affect apoA-I plasma levels, whereas a significant decrease was observed in mice treated with 20 mg·kg(-1) ·day(-1) of rosuvastatin (-16%, P < 0.01). Neither relative hepatic mRNA concentrations of apoA-I nor apoA-I secretion rates from primary hepatocytes were influenced by rosuvastatin treatment at each tested dose. CONCLUSIONS AND IMPLICATIONS In human apoA-I transgenic mice, rosuvastatin treatment does not increase either apoA-I transcription and hepatic secretion, or apoA-I plasma levels. These results support the hypothesis that other mechanisms may account for the observed HDL increase induced by statin therapy in humans.
Collapse
Affiliation(s)
- Marta Marchesi
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yasuda T, Johnston TP, Shinohara M, Inoue M, Ishida T. The effect of poloxamer 407 on the functional properties of HDL in mice. J Pharm Pharmacol 2012; 64:677-87. [DOI: 10.1111/j.2042-7158.2011.01444.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Abstract
Objectives
There is an inverse relationship between high-density lipoprotein (HDL) and heart disease. HDL possesses not only both antioxidant and anti-inflammatory properties, but also anti-thrombotic and endothelial function-promoting qualities. However, it is not only the serum concentration of HDL that is important, but also the ‘functional’ quality of the HDL. The objective was to determine the functional status of HDL in a well-established mouse model of dyslipidaemia and atherosclerosis induced by the administration of a block copolymer (poloxamer 407; P-407).
Methods
C57BL/6 mice were administered a single intraperitoneal dose of P-407 (0.5 g/kg) and blood was collected at 24 h post-dosing. HDL was isolated from controls (control HDL) and P-407-treated (P-407 HDL) mice and used to test its anti-inflammatory properties in vitro. Additionally, antioxidant enzymes associated with HDL, namely, platelet activating factor-acetylhydrolase (PAF-AH) and paraoxonase (PON), were evaluated for any potential reduction in their biological activity.
Key findings
A single injection of P-407 in C57BL/6 mice resulted in a marked decrease in the levels of HDL-cholesterol and phospholipids. HDL particle size significantly increased, primarily due to remodelling of HDL with triglyceride. It was demonstrated that (i) long-chain saturated fatty acids were higher and the n-3/n-6 fatty acid ratio was significantly lower for P-407 HDL compared with control HDL, and (ii) P-407 HDL lost its capacity to inhibit tumour necrosis factor-α (TNF-α)-induced vascular cell adhesion molecule-1 (VCAM-1) expression compared with control HDL. Additionally, P-407 HDL was not able to neutralize lipopolysaccharide and inhibit subsequent TNF-α production compared with control HDL. The biological activity of platelet-activating factor acetylhydrolase (PAF-AH) and paraoxonase (PON) decreased in direct proportion to the circulating levels of both HDL-cholesterol and apolipoprotein (apoA-1).
Conclusions
Combination of previously reported findings in P-407-treated mice, such as (i) production of both oxidized LDL and malondialdehyde, and (ii) profound elevations in the soluble forms of intercellular adhesion molecule-1 (ICAM-1), VCAM-1, and E-selectin, with the present results, would strongly suggest that HDL in P-407-treated mice is rendered dysfunctional. Thus, these findings help to explain why P-407-treated mice begin to form aortic atherosclerotic lesions about one month after initiating P-407 treatment.
Collapse
Affiliation(s)
- Tomoyuki Yasuda
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | | | | | | | | |
Collapse
|
49
|
Cao G, Beyer TP, Zhang Y, Schmidt RJ, Chen YQ, Cockerham SL, Zimmerman KM, Karathanasis SK, Cannady EA, Fields T, Mantlo NB. Evacetrapib is a novel, potent, and selective inhibitor of cholesteryl ester transfer protein that elevates HDL cholesterol without inducing aldosterone or increasing blood pressure. J Lipid Res 2011; 52:2169-2176. [PMID: 21957197 DOI: 10.1194/jlr.m018069] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) catalyses the exchange of cholesteryl ester and triglyceride between HDL and apoB containing lipoprotein particles. The role of CETP in modulating plasma HDL cholesterol levels in humans is well established and there have been significant efforts to develop CETP inhibitors to increase HDL cholesterol for the treatment of coronary artery disease. These efforts, however, have been hampered by the fact that most CETP inhibitors either have low potency or have undesirable side effects. In this study, we describe a novel benzazepine compound evacetrapib (LY2484595), which is a potent and selective inhibitor of CETP both in vitro and in vivo. Evacetrapib inhibited human recombinant CETP protein (5.5 nM IC(50)) and CETP activity in human plasma (36 nM IC(50)) in vitro. In double transgenic mice expressing human CETP and apoAI, evacetrapib exhibited an ex vivo CETP inhibition ED(50) of less than 5 mg/kg at 8 h post oral dose and significantly elevated HDL cholesterol. Importantly, no blood pressure elevation was observed in rats dosed with evacetrapib at high exposure multiples compared with the positive control, torcetrapib. In addition, in a human adrenal cortical carcinoma cell line (H295R cells), evacetrapib did not induce aldosterone or cortisol biosynthesis whereas torcetrapib dramatically induced aldosterone and cortisol biosynthesis. Our data indicate that evacetrapib is a potent and selective CETP inhibitor without torcetrapib-like off-target liabilities. Evacetrapib is currently in phase II clinical development.
Collapse
Affiliation(s)
- Guoqing Cao
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285.
| | - Thomas P Beyer
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Youyan Zhang
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Robert J Schmidt
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Yan Q Chen
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Sandra L Cockerham
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Karen M Zimmerman
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | | | - Ellen A Cannady
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Todd Fields
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| | - Nathan B Mantlo
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, IN 46285
| |
Collapse
|
50
|
Ruan X, Li Z, Zhang Y, Yang L, Pan Y, Wang Z, Feng GS, Chen Y. Apolipoprotein A-I possesses an anti-obesity effect associated with increase of energy expenditure and up-regulation of UCP1 in brown fat. J Cell Mol Med 2011; 15:763-72. [PMID: 20193037 PMCID: PMC3922665 DOI: 10.1111/j.1582-4934.2010.01045.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Apolipoprotein A-I (ApoA-I) is the most abundant protein constituent of high-density lipoprotein (HDL). Reduced plasma HDL and ApoA-I levels have been found to be associated with obesity and metabolic syndrome in human beings. However, whether or not ApoA-I has a direct effect on obesity is largely unknown. Here we analysed the anti-obesity effect of ApoA-I using two mouse models, a transgenic mouse with overexpression of ApoA-I and the mice administered with an ApoA-I mimetic peptide D-4F. The mice were induced to develop obesity by feeding with high fat diet. Both ApoA-I overexpression and D-4F treatment could significantly reduce white fat mass and slightly improve insulin sensitivity in the mice. Metabolic analyses revealed that ApoA-I overexpression and D-4F treatment enhanced energy expenditure in the mice. The mRNA level of uncoupling protein (UCP)1 in brown fat tissue was elevated by ApoA-I transgenic mice. ApoA-I and D-4F treatment was able to increase UCP1 mRNA and protein levels as well as to stimulate AMP-activated protein kinase (AMPK) phosphorylation in brown adipocytes in culture. Taken together, our results reveal that ApoA-I has an anti-obesity effect in the mouse and such effect is associated with increases in energy expenditure and UCP1 expression in the brown fat tissue.
Collapse
Affiliation(s)
- Xiangbo Ruan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|