1
|
Getz GS, Reardon CA. Insights from Murine Studies on the Site Specificity of Atherosclerosis. Int J Mol Sci 2024; 25:6375. [PMID: 38928086 PMCID: PMC11204064 DOI: 10.3390/ijms25126375] [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: 05/06/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis is an inflammatory reaction that develops at specific regions within the artery wall and at specific sites of the arterial tree over a varying time frame in response to a variety of risk factors. The mechanisms that account for the interaction of systemic factors and atherosclerosis-susceptible regions of the arterial tree to mediate this site-specific development of atherosclerosis are not clear. The dynamics of blood flow has a major influence on where in the arterial tree atherosclerosis develops, priming the site for interactions with atherosclerotic risk factors and inducing cellular and molecular participants in atherogenesis. But how this accounts for lesion development at various locations along the vascular tree across differing time frames still requires additional study. Currently, murine models are favored for the experimental study of atherogenesis and provide the most insight into the mechanisms that may contribute to the development of atherosclerosis. Based largely on these studies, in this review, we discuss the role of hemodynamic shear stress, SR-B1, and other factors that may contribute to the site-specific development of atherosclerosis.
Collapse
Affiliation(s)
- Godfrey S. Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA;
| | | |
Collapse
|
2
|
Zhang Y, Fatima M, Hou S, Bai L, Zhao S, Liu E. Research methods for animal models of atherosclerosis (Review). Mol Med Rep 2021; 24:871. [PMID: 34713295 PMCID: PMC8569513 DOI: 10.3892/mmr.2021.12511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that threatens human health and lives by causing vascular stenosis and plaque rupture. Various animal models have been employed for elucidating the pathogenesis, drug development and treatment validation studies for atherosclerosis. To the best of our knowledge, the species used for atherosclerosis research include mice, rats, hamsters, rabbits, pigs, dogs, non-human primates and birds, among which the most commonly used ones are mice and rabbits. Notably, apolipoprotein E knockout (KO) or low-density lipoprotein receptor KO mice have been the most widely used animal models for atherosclerosis research since the late 20th century. Although the aforementioned animal models can form atherosclerotic lesions, they cannot completely simulate those in humans with respect to lesion location, lesion composition, lipoprotein composition and physiological structure. Hence, an appropriate animal model needs to be selected according to the research purpose. Additionally, it is necessary for atherosclerosis research to include quantitative analysis results of atherosclerotic lesion size and plaque composition. Laboratory animals can provide not only experimental tissues for in vivo studies but also cells needed for in vitro experiments. The present review first summarizes the common animal models and their practical applications, followed by focus on mouse and rabbit models and elucidating the methods to quantify atherosclerotic lesions. Finally, the methods of culturing endothelial cells, macrophages and smooth muscle cells were elucidated in detail and the experiments involved in atherosclerosis research were discussed.
Collapse
Affiliation(s)
- Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Mahreen Fatima
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Siyuan Hou
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
3
|
Zou J, Wang G, Li H, Yu X, Tang C. IgM natural antibody T15/E06 in atherosclerosis. Clin Chim Acta 2020; 504:15-22. [DOI: 10.1016/j.cca.2020.01.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 11/28/2022]
|
4
|
Johnston JM, Francis SE, Kiss-Toth E. Experimental models of murine atherosclerosis: does perception match reality? Cardiovasc Res 2019; 114:1845-1847. [PMID: 29878146 PMCID: PMC6255687 DOI: 10.1093/cvr/cvy140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/30/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jessica M Johnston
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Sheila E Francis
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
5
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
6
|
VanderLaan PA, Reardon CA, Cabana VG, Wang CR, Getz GS. Invariant Natural Killer T-Cells and Total CD1d Restricted Cells Differentially Influence Lipid Metabolism and Atherosclerosis in Low Density Receptor Deficient Mice. Int J Mol Sci 2019; 20:ijms20184566. [PMID: 31540125 PMCID: PMC6770011 DOI: 10.3390/ijms20184566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 11/16/2022] Open
Abstract
Natural killer T (NKT) cells are a distinct subset of lymphocytes that bridge the innate and adaptive immune response and can be divided into type I invariant NKT cells (iNKT) and type II NKT cells. The objective of this study is to examine the effects of NKT cell on lipid metabolism and the initiation and progression of atherosclerosis in LDL receptor deficient (LDLR−/−) mice. Mice were fed an atherogenic diet for 4 or 8 weeks and plasma lipids, lipoproteins, and atherosclerosis were measured. The selective absence of iNKT cells in Jα18−/−LDLR−/− mice led to an increase in plasma cholesterol levels in female mice. Transgenic Vα14tg/LDLR−/− mice with elevated numbers of iNKT cells had increased late atherosclerosis of the innominate artery, though absence of either iNKT cells or all NKT cells and other CD1d expressing cells had varying effects on atherosclerotic lesion burden in the ascending aortic arch and aortic root. These studies not only highlight the potential modulatory role played by NKT cells in atherosclerosis and lipid metabolism, but also raise the possibility that divergent roles may be played by iNKT and CD1d restricted cells such as type II NKT cells or other CD1d expressing cells.
Collapse
Affiliation(s)
- Paul A VanderLaan
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University, 633 Clark St, Evanston, IL 60208, USA.
| | - Godfrey S Getz
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
7
|
Mahdavi Gorabi A, Banach M, Reiner Ž, Pirro M, Hajighasemi S, Johnston TP, Sahebkar A. The Role of Mesenchymal Stem Cells in Atherosclerosis: Prospects for Therapy via the Modulation of Inflammatory Milieu. J Clin Med 2019; 8:E1413. [PMID: 31500373 PMCID: PMC6780166 DOI: 10.3390/jcm8091413] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic, inflammatory disease that mainly affects the arterial intima. The disease is more prevalent in middle-age and older individuals with one or more cardiovascular risk factors, including dyslipidemia, hypertension, diabetes, smoking, obesity, and others. The beginning and development of atherosclerosis has been associated with several immune components, including infiltration of inflammatory cells, monocyte/macrophage-derived foam cells, and inflammatory cytokines and chemokines. Mesenchymal stem cells (MSCs) originate from several tissue sources of the body and have self-renewal and multipotent differentiation characteristics. They also have immunomodulatory and anti-inflammatory properties. Recently, it was shown that MSCs have a regulatory role in plasma lipid levels. In addition, MSCs have shown to have promising potential in terms of treatment strategies for several diseases, including those with an inflammatory component. In this regard, transplantation of MSCs to patients with atherosclerosis has been proposed as a novel strategy in the treatment of this disease. In this review, we summarize the current advancements regarding MSCs for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1411713138, Iran
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), 93-338 Lodz, Poland
| | - Željko Reiner
- Department of Internal medicine, University Hospital Center Zagreb, Kišpatićeva 12, Zagreb 1000, Croatia
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, 06123 Perugia, Italy
| | - Saeideh Hajighasemi
- Department of Medical Biotechnology, Faculty of Paramedicine, Qazvin University of Medical Sciences, Qazvin 1531534199, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran.
| |
Collapse
|
8
|
|
9
|
Qu L, Li D, Gao X, Li Y, Wu J, Zou W. Di'ao Xinxuekang Capsule, a Chinese Medicinal Product, Decreases Serum Lipids Levels in High-Fat Diet-Fed ApoE -/- Mice by Downregulating PCSK9. Front Pharmacol 2018; 9:1170. [PMID: 30443213 PMCID: PMC6221936 DOI: 10.3389/fphar.2018.01170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/26/2018] [Indexed: 11/13/2022] Open
Abstract
Numerous risk factors are responsible for the development of atherosclerosis, for which an increased serum level of low-density lipoprotein cholesterol (LDL-C) is a driving force. By binding to the low-density lipoprotein cholesterol receptor (LDLR) and inducing LDLR degradation, proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a key role in cholesterol homeostasis regulation. The inducement of PCSK9 expression is also an important reason for statin intolerance. The Di'ao Xinxuekang (DXXK) capsule extracted from Dioscorea nipponica Makino is a well-known traditional Chinese herbal medicinal product used in atherosclerotic cardiovascular disease. Although DXXK has been widely used in atherosclerotic cardiovascular treatment for nearly 30 years, studies on the potential mechanisms of the lipid-lowering effect are very limited. The purpose of the present study was to demonstrate the possible involvement of the PCSK9/LDLR signaling pathway in the lipid-lowering and antiatherosclerotic effect of DXXK in high-fat diet-fed ApoE-/- mice. The results showed that DXXK treatment alleviated hyperlipidemia, fat accumulation, and atherosclerosis formation in ApoE-/- mice. Furthermore, changes in the expression of PCSK9 mRNA in liver tissue and the circulating PCSK9 level in ApoE-/- mice were both reversed after DXXK treatment, and upregulation of LDLR in the liver was also detected in the protein level in DXXK-treated mice. Our study is the first to show that DXXK could alleviate lipid disorder and ameliorate atherosclerosis with downregulation of the PCSK9 in high-fat diet-fed ApoE-/- mice, suggesting that DXXK may be a potential novel therapeutic treatment and may support statin action in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Liping Qu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Didi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoping Gao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongwei Li
- Department of New Drug Research and Development, National Engineering Research Center for Natural Medicines, Chengdu, China
| | - Jianming Wu
- Laboratory of Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Mallone A, Stenger C, Von Eckardstein A, Hoerstrup SP, Weber B. Biofabricating atherosclerotic plaques: In vitro engineering of a three-dimensional human fibroatheroma model. Biomaterials 2017; 150:49-59. [PMID: 29032330 DOI: 10.1016/j.biomaterials.2017.09.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/11/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022]
Abstract
Atherosclerotic plaques are cholesterol-induced inflammatory niches accumulating in the vascular sub-endothelial space. Cellular and extracellular composition of human plaques is maneuvered by local inflammation that leads to alterations in the original vascular microenvironment and to the recruitment of an invading fibrous layer (fibroatharoma). In the present study we introduce a bioengineered three-dimensional model of human fibroatheroma (ps-plaque) assembled with a tailored hanging-drop protocol. Using vi-SNE based multidimensional flow cytometry data analysis we compared the myeloid cell-populations in ps-plaques to those in plaques isolated from human carotid arteries. We observed that plasmacytoid and activated dendritic cells are the main myeloid components of human carotid plaques and that both cell types are present in the biofabricated model. We found that low-density lipoproteins affect cell viability and contribute to population polarization in ps-plaques. The current work describes the first human bioengineered in vitro model of late atherosclerotic lesion for the investigation of atherosclerosis aetiopathogenesis.
Collapse
Affiliation(s)
- Anna Mallone
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.
| | - Chantal Stenger
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | | | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Benedikt Weber
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Daugherty A, Tall AR, Daemen MJ, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Circ Res 2017; 121:e53-e79. [DOI: 10.1161/res.0000000000000169] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
12
|
Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2017; 37:e131-e157. [PMID: 28729366 DOI: 10.1161/atv.0000000000000062] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
13
|
Li F, Guo X, Chen SY. Function and Therapeutic Potential of Mesenchymal Stem Cells in Atherosclerosis. Front Cardiovasc Med 2017; 4:32. [PMID: 28589127 PMCID: PMC5438961 DOI: 10.3389/fcvm.2017.00032] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/01/2017] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a complicated disorder and largely attributable to dyslipidaemia and chronic inflammation. Despite therapeutic advances over past decades, atherosclerosis remains the leading cause of mortality worldwide. Due to their capability of immunomodulation and tissue regeneration, mesenchymal stem cells (MSCs) have evolved as an attractive therapeutic agent in various diseases including atherosclerosis. Accumulating evidences support the protective role of MSCs in all stages of atherosclerosis. In this review, we highlight the current understanding of MSCs including their characteristics such as molecular markers, tissue distribution, migratory property, immune-modulatory competence, etc. We also summarize MSC functions in animal models of atherosclerosis. MSC transplantation is able to modulate cytokine and chemokine secretion, reduce endothelial dysfunction, promote regulatory T cell function, decrease dyslipidemia, and stabilize vulnerable plaques during atherosclerosis development. In addition, MSCs may migrate to lesions where they develop into functional cells during atherosclerosis formation. Finally, the perspectives of MSCs in clinical atherosclerosis therapy are discussed.
Collapse
Affiliation(s)
- Feifei Li
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA.,The Department of Cardiovascular Surgery, Union Hospital, Wuhan, China
| | - Xia Guo
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| |
Collapse
|
14
|
Kostogrys RB, Franczyk-Zarow M, Gasior-Glogowska M, Kus E, Jasztal A, Wrobel TP, Baranska M, Czyzynska-Cichon I, Drahun A, Manterys A, Chlopicki S. Anti-atherosclerotic effects of pravastatin in brachiocephalic artery in comparison with en face aorta and aortic roots in ApoE/LDLR -/- mice. Pharmacol Rep 2016; 69:112-118. [PMID: 27915184 DOI: 10.1016/j.pharep.2016.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cholesterol-dependent and independent mechanisms were proposed to explain anti-atherosclerotic action of statins in humans. However, their effects in murine models of atherosclerosis have not been consistently demonstrated. Here, we studied the effects of pravastatin on atherosclerosis in ApoE/LDLR-/- mice fed a control and atherogenic diet. METHODS ApoE/LDLR-/- mice were fed a control (CHOW) or an atherogenic (Low Carbohydrate High Protein, LCHP) diet. Two doses of pravastatin (40mg/kg and 100mg/kg) were used. The anti-atherosclerotic effects of pravastatin in en face aorta, cross-sections of aortic roots and brachiocephalic artery (BCA) were analysed. The lipid profile was determined. Fourier Transform Infrared Spectroscopy followed by Fuzzy C-Means (FCM) clustering was used for the quantitative assessment of plaque composition. RESULTS Treatment with pravastatin (100mg/kg) decreased total and LDL cholesterol only in the LCHP group, but displayed a pronounced anti-atherosclerotic effect in BCA and abdominal aorta. The anti-atherosclerotic effect of pravastatin (100mg/kg) in BCA was associated with significant alterations of the chemical plaque composition, including a fall in cholesterol and cholesterol esters contents independently on total cholesterol and LDL concentration in plasma. CONCLUSIONS Pravastatin at high (100mg/kg), but not low dose displayed a pronounced anti-atherosclerotic effect in ApoE/LDLR-/- mice fed a CHOW or LCHP diet that was remarkable in BCA, visible in en face aorta, whereas it was not observed in aortic roots, suggesting that previous inconsistencies might have been due to the various sites of atherosclerotic plaque analysis.
Collapse
Affiliation(s)
- Renata B Kostogrys
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kopernika 15 A, 31-501 Kraków, Poland.
| | - Magdalena Franczyk-Zarow
- Department of Human Nutrition, Faculty of Food Technology, Agricultural University of Kraków, Balicka 122, 30-149 Kraków, Poland
| | - Marlena Gasior-Glogowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland
| | - Edyta Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland
| | - Tomasz P Wrobel
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland; Faculty of Chemistry, Jagiellonian University, 3 Ingardena Str., 30-060 Kraków, Poland
| | - Izabela Czyzynska-Cichon
- Department of Human Nutrition, Faculty of Food Technology, Agricultural University of Kraków, Balicka 122, 30-149 Kraków, Poland; Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland
| | - Anna Drahun
- Department of Human Nutrition, Faculty of Food Technology, Agricultural University of Kraków, Balicka 122, 30-149 Kraków, Poland
| | - Angelika Manterys
- Department of Human Nutrition, Faculty of Food Technology, Agricultural University of Kraków, Balicka 122, 30-149 Kraków, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Kraków, Poland; Chair of Pharmacology, Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Kraków, Poland
| |
Collapse
|
15
|
Molecular Imaging of Vulnerable Atherosclerotic Plaques in Animal Models. Int J Mol Sci 2016; 17:ijms17091511. [PMID: 27618031 PMCID: PMC5037788 DOI: 10.3390/ijms17091511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is characterized by intimal plaques of the arterial vessels that develop slowly and, in some cases, may undergo spontaneous rupture with subsequent heart attack or stroke. Currently, noninvasive diagnostic tools are inadequate to screen atherosclerotic lesions at high risk of acute complications. Therefore, the attention of the scientific community has been focused on the use of molecular imaging for identifying vulnerable plaques. Genetically engineered murine models such as ApoE−/− and ApoE−/−Fbn1C1039G+/− mice have been shown to be useful for testing new probes targeting biomarkers of relevant molecular processes for the characterization of vulnerable plaques, such as vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, intercellular adhesion molecule (ICAM)-1, P-selectin, and integrins, and for the potential development of translational tools to identify high-risk patients who could benefit from early therapeutic interventions. This review summarizes the main animal models of vulnerable plaques, with an emphasis on genetically altered mice, and the state-of-the-art preclinical molecular imaging strategies.
Collapse
|
16
|
Getz GS, Reardon CA. Do the Apoe-/- and Ldlr-/- Mice Yield the Same Insight on Atherogenesis? Arterioscler Thromb Vasc Biol 2016; 36:1734-41. [PMID: 27386935 DOI: 10.1161/atvbaha.116.306874] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/24/2016] [Indexed: 02/02/2023]
Abstract
Murine models of atherosclerosis are useful for investigating the environmental and genetic influences on lesion formation and composition. Apoe(-/-) and Ldlr(-/-) mice are the 2 most extensively used models. The models differ in important ways with respect to the precise mechanism by which their absence enhances atherosclerosis, including differences in plasma lipoproteins. The majority of the gene function studies have utilized only 1 model, with the results being generalized to atherogenic mechanisms. In only a relatively few cases have studies been conducted in both atherogenic murine models. This review will discuss important differences between the 2 atherogenic models and will point out studies that have been performed in the 2 models where results are comparable and those where different results were obtained.
Collapse
Affiliation(s)
- Godfrey S Getz
- From the Department of Pathology (G.S.G.) and Ben May Institute for Cancer Biology (C.A.R.), University of Chicago, IL.
| | - Catherine A Reardon
- From the Department of Pathology (G.S.G.) and Ben May Institute for Cancer Biology (C.A.R.), University of Chicago, IL
| |
Collapse
|
17
|
Natural killer cells in the innate immunity network of atherosclerosis. Immunol Lett 2015; 168:51-7. [DOI: 10.1016/j.imlet.2015.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 08/20/2015] [Accepted: 09/07/2015] [Indexed: 12/11/2022]
|
18
|
Wang L, Tian F, Arias A, Yang M, Sharifi BG, Shah PK. Comparative Effects of Diet-Induced Lipid Lowering Versus Lipid Lowering Along With Apo A-I Milano Gene Therapy on Regression of Atherosclerosis. J Cardiovasc Pharmacol Ther 2015; 21:320-8. [DOI: 10.1177/1074248415610216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/06/2015] [Indexed: 11/15/2022]
Abstract
Apolipoprotein A-1 (Apo A-I) Milano, a naturally occurring Arg173 to Cys mutant of Apo A-1, has been shown to reduce atherosclerosis in animal models and in a small phase 2 human trial. We have shown the superior atheroprotective effects of Apo A-I Milano (Apo A-IM) gene compared to wild-type Apo A-I gene using transplantation of retrovirally transduced bone marrow in Apo A-I/Apo E null mice. In this study, we compared the effect of dietary lipid lowering versus lipid lowering plus Apo A-IM gene transfer using recombinant adeno-associated virus (rAAV) 8 as vectors on atherosclerosis regression in Apo A-I/Apo E null mice. All mice were fed a high-cholesterol diet from age of 6 weeks until week 20, and at 20 weeks, 10 mice were euthanized to determine the extent of atherosclerosis. After 20 weeks, an additional 20 mice were placed on either a low-cholesterol diet plus empty rAAV (n = 10) to serve as controls or low-cholesterol diet plus 1 single intravenous injection of 1.2 × 1012 vector genomes of adeno-associated virus (AAV) 8 vectors expressing Apo A-IM (n = 10). At the 40 week time point, intravenous AAV8 Apo A-IM recipients showed a significant regression of atherosclerosis in the whole aorta ( P < .01), aortic sinuses ( P < .05), and brachiocephalic arteries ( P < .05) compared to 20-week-old mice, whereas low-cholesterol diet plus empty vector control group showed no significant regression in lesion size. Immunostaining showed that compared to the 20-week-old mice, there was a significantly reduced macrophage content in the brachiocephalic ( P < .05) and aortic sinus plaques ( P < .05) of AAV8 Apo A-IM recipients. These data show that although dietary-mediated cholesterol lowering halts progression of atherosclerosis, it does not induce regression, whereas combination of low-cholesterol diet and AAV8 mediated Apo A-I Milano gene therapy induces rapid and significant regression of atherosclerosis in mice. These data provide support for the potential feasibility of this approach for atherosclerosis regression.
Collapse
Affiliation(s)
- Lai Wang
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Fang Tian
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ana Arias
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mingjie Yang
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Behrooz G. Sharifi
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Prediman K. Shah
- Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
19
|
Ng HP, Zhu X, Harmon EY, Lennartz MR, Nagarajan S. Reduced Atherosclerosis in apoE-inhibitory FcγRIIb-Deficient Mice Is Associated With Increased Anti-Inflammatory Responses by T Cells and Macrophages. Arterioscler Thromb Vasc Biol 2015; 35:1101-12. [PMID: 25792447 PMCID: PMC4409543 DOI: 10.1161/atvbaha.115.305290] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 02/27/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Fcγ receptors (FcγRs) are classified as activating (FcγRI, III, and IV) and inhibitory (FcγRII) receptors. We have reported that deletion of activating FcγRs in apolipoprotein E (apoE) single knockout mice attenuated atherosclerosis. In this report, we investigated the hypothesis that deficiency of inhibitory FcγRIIb exacerbates atherosclerosis. APPROACH AND RESULTS ApoE-FcγRIIb double knockout mice, congenic to the C57BL/6 (apoE-FcγRIIbB6 (-/-)), were generated and atherosclerotic lesions were assessed. In contrary to our hypothesis, when compared with apoE single knockout mice, arterial lesions were significantly decreased in apoE-FcγRIIbB6 (-/-) male and female mice fed chow or high-fat diets. Chimeric mice generated by transplanting apoE-FcγRIIbB6 (-/-) marrow into apoE single knockout mice also developed reduced lesions. CD4(+) T cells from apoE-FcγRIIbB6 (-/-) mice produced higher levels of interleukin-10 and transforming growth factor-β than their apoE single knockout counterparts. As our findings conflict with a previous report using apoE-FcγRIIb129/B6 (-/-) mice on a mixed genetic background, we investigated whether strain differences contributed to the anti-inflammatory response. Macrophages from FcγRIIb129/B6 (-/-) mice on a mixed genetic background produced more interleukin-1β and MCP-1 (monocyte chemoattractant protein-1) in response to immune complexes, whereas congenic FcγRIIbB6 (-/-) mice generated more interleukin-10 and significantly less interleukin-1β. Interestingly, the expression of lupus-associated slam genes, located in proximity to fcgr2b in mouse chromosome 1, is upregulated only in mixed FcγRIIb129/B6 (-/-) mice. CONCLUSIONS Our findings demonstrate a detrimental role for FcγRIIb signaling in atherosclerosis and the contribution of anti-inflammatory cytokine responses in the attenuated lesions observed in apoE-FcγRIIbB6 (-/-) mice. As 129/sv genome-derived lupus-associated genes have been implicated in lupus phenotype in FcγRIIb129/B6 (-/-) mice, our findings suggest possible epistatic mechanism contributing to the decreased lesions.
Collapse
Affiliation(s)
- Hang Pong Ng
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Xinmei Zhu
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Erin Y Harmon
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Michelle R Lennartz
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Shanmugam Nagarajan
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.).
| |
Collapse
|
20
|
Abstract
Cardiovascular disease is the major cause of death in most developed nations and the social and economic burden of this disease is quite high. Atherosclerosis is a major underlying basis for most cardiovascular diseases including myocardial infarction and stroke. Genetically modified mouse models, particularly mice deficient in apoprotein E or the LDL receptor, have been widely used in preclinical atherosclerosis studies to gain insight into the mechanisms underlying this pathology. This chapter reviews several mouse models of atherosclerosis progression and regression as well as the role of immune cells in disease progression and the genetics of murine atherogenesis.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology, University of Chicago, Box MC 1089, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA.
| | - Catherine A Reardon
- Department of Pathology, University of Chicago, Box MC 1089, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
| |
Collapse
|
21
|
Getz GS, Reardon CA. The mutual interplay of lipid metabolism and the cells of the immune system in relation to atherosclerosis. ACTA ACUST UNITED AC 2014; 9:657-671. [PMID: 25705263 DOI: 10.2217/clp.14.50] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is a chronic inflammation in the arterial wall involving cells of the innate and adaptive immune system that is promoted by hyperlipidemia. In addition, the immune system can influence lipids and lipoprotein levels and cellular lipid homeostasis can influence the level and function of the immune cells. We will review the effects of manipulation of adaptive immune cells and immune cell products on lipids and lipoproteins, focusing mainly on studies performed in murine models of atherosclerosis. We also review how lipoproteins and cellular lipid levels, particularly cholesterol levels, influence the function of cells of the innate and adaptive immune systems. The overriding theme is that these interactions are driven by the need to provide the energy and membrane components for cell proliferation and migration, membrane expansion and other functions that are so important in the functioning of the immune cells.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology, University of Chicago, Box MC 1089, 5841 S. Maryland Avenue, Chicago, IL 60637, USA
| | - Catherine A Reardon
- Department of Pathology, University of Chicago, Box MC 1089, 5841 S. Maryland Avenue, Chicago, IL 60637, USA
| |
Collapse
|
22
|
Tsiantoulas D, Sage AP, Mallat Z, Binder CJ. Targeting B cells in atherosclerosis: closing the gap from bench to bedside. Arterioscler Thromb Vasc Biol 2014; 35:296-302. [PMID: 25359862 DOI: 10.1161/atvbaha.114.303569] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Atherosclerotic plaque formation is strongly influenced by different arms of the immune system, including B lymphocytes. B cells are divided into 2 main families: the B1 and the B2 cells. B1 cells are atheroprotective mainly via the production of natural IgM antibodies that bind oxidized low-density lipoprotein and apoptotic cells. B2 cells, which include follicular and marginal zone B cells, are suggested to be proatherogenic. Antibody-mediated depletion of B cells has become a valuable treatment option for certain autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis that are also characterized by the development of premature atherosclerosis. Thus, B cells represent a novel interesting target for therapeutic modulation of the atherosclerotic disease process. Here, we discuss the effect of different of B-cell subsets in experimental atherosclerosis, their mechanism of action as well as potential ways to exploit these findings for the treatment of human disease.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Andrew P Sage
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Ziad Mallat
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Christoph J Binder
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.).
| |
Collapse
|
23
|
Sage AP, Murphy D, Maffia P, Masters LM, Sabir SR, Baker LL, Cambrook H, Finigan AJ, Ait-Oufella H, Grassia G, Harrison JE, Ludewig B, Reith W, Hansson GK, Reizis B, Hugues S, Mallat Z. MHC Class II-restricted antigen presentation by plasmacytoid dendritic cells drives proatherogenic T cell immunity. Circulation 2014; 130:1363-73. [PMID: 25223984 DOI: 10.1161/circulationaha.114.011090] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.
Collapse
Affiliation(s)
- Andrew P Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Deirdre Murphy
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Pasquale Maffia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Leanne M Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Suleman R Sabir
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Lauren L Baker
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Helen Cambrook
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Alison J Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Hafid Ait-Oufella
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Gianluca Grassia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - James E Harrison
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Burkhard Ludewig
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Walter Reith
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Göran K Hansson
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Boris Reizis
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Stéphanie Hugues
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.).
| |
Collapse
|
24
|
Tian F, Wang L, Arias A, Yang M, Sharifi BG, Shah PK. Comparative antiatherogenic effects of intravenous AAV8- and AAV2-mediated ApoA-IMilano gene transfer in hypercholesterolemic mice. J Cardiovasc Pharmacol Ther 2014; 20:66-75. [PMID: 24742767 DOI: 10.1177/1074248414530041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Apolipoprotein A-IMilano (ApoA-IM), a naturally occurring Arg173 to Cys mutant of ApoA-I, has been shown to reduce atherosclerosis in animal models and in a small phase 2 human trial. We have shown superior atheroprotective effects of ApoA-IM gene compared with wild-type ApoA-I gene using transplantation of retrovirally transduced bone marrow in ApoA-I/ApoE null mice. In this study, we compared the antiatherogenic efficacy of ApoA-IM gene transfer using Recombinant adeno-associated virus (rAAV) 2 or rAAV8 as vectors in ApoA-I/ApoE null mice. Mice received a single intravenous injection of 1.2 × 10(12) vector genomes of AAV2 or AAV8 vectors expressing ApoA-IM or control empty vectors (12 mice/group). Circulating levels of ApoA-IM were higher in recipients of AAV8 compared with AAV2 at 4, 12, and 20 weeks postinjection. Qualitative polymerase chain reaction analysis of RNA collected from different tissues showed that the AAV8-mediated gene transfer resulted in a more efficient transgene expression in the heart, brain, liver, lung, spleen, and kidney of the recipient mice compared with AAV2. Intravenous AAV8-ApoA-IM injection reduced atherosclerosis in the whole aorta (P < .01), aortic sinuses (P < .05), and brachiocephalic arteries (P < .05) compared with the vector control, whereas there was no statistically significant reduction in atherosclerosis in mice receiving intravenous AAV2-ApoA-IM. The ApoA-IM gene was expressed in the aortic tissue of mice receiving AAV8 ApoA-IM but not in those receiving AAV2 ApoA-IM. Immunostaining showed that compared with the vector control, there was reduced macrophage content in the brachiocephalic (P < .05) and aortic sinus plaques (P < .05) of AAV8 ApoA-IM recipients but not in the recipients of AAV2 ApoA-IM. Thus, intravenous injection of AAV8 is more effective than intravenous injection of AAV2 in the expression of ApoA-IM gene. These data provide support for the potential feasibility of this approach for atheroprotection in humans.
Collapse
Affiliation(s)
- Fang Tian
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Lai Wang
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Ana Arias
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Mingjie Yang
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Behrooz G Sharifi
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Prediman K Shah
- Division of Cardiology and Oppenheimer Atherosclerosis Research Center, Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
25
|
Legein B, Temmerman L, Biessen EAL, Lutgens E. Inflammation and immune system interactions in atherosclerosis. Cell Mol Life Sci 2013; 70:3847-69. [PMID: 23430000 PMCID: PMC11113412 DOI: 10.1007/s00018-013-1289-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 16.7 million deaths each year. The underlying cause of the majority of CVD is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. Today's picture is far more complex. Atherosclerosis is considered a chronic inflammatory disease that results in the formation of plaques in large and mid-sized arteries. Both cells of the innate and the adaptive immune system play a crucial role in its pathogenesis. By transforming immune cells into pro- and anti-inflammatory chemokine- and cytokine-producing units, and by guiding the interactions between the different immune cells, the immune system decisively influences the propensity of a given plaque to rupture and cause clinical symptoms like myocardial infarction and stroke. In this review, we give an overview on the newest insights in the role of different immune cells and subtypes in atherosclerosis.
Collapse
Affiliation(s)
- Bart Legein
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lieve Temmerman
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Erik A. L. Biessen
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, Pettenkoferstrasse 8a/9, 80336 Munich, Germany
| |
Collapse
|
26
|
Tsimikas S, Miyanohara A, Hartvigsen K, Merki E, Shaw PX, Chou MY, Pattison J, Torzewski M, Sollors J, Friedmann T, Lai NC, Hammond HK, Getz GS, Reardon CA, Li AC, Banka CL, Witztum JL. Human oxidation-specific antibodies reduce foam cell formation and atherosclerosis progression. J Am Coll Cardiol 2013; 58:1715-27. [PMID: 21982317 DOI: 10.1016/j.jacc.2011.07.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 07/11/2011] [Accepted: 07/11/2011] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We sought to assess the in vivo importance of scavenger receptor (SR)-mediated uptake of oxidized low-density lipoprotein (OxLDL) in atherogenesis and to test the efficacy of human antibody IK17-Fab or IK17 single-chain Fv fragment (IK17-scFv), which lacks immunologic properties of intact antibodies other than the ability to inhibit uptake of OxLDL by macrophages, to inhibit atherosclerosis. BACKGROUND The unregulated uptake of OxLDL by macrophage SR contributes to foam cell formation, but the importance of this pathway in vivo is uncertain. METHODS Cholesterol-fed low-density lipoprotein receptor knockout (LDLR(-/-)) mice were treated with intraperitoneal infusion of human IK17-Fab (2.5 mg/kg) 3 times per week for 14 weeks. Because anti-human antibodies developed in these mice, LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice (lacking the ability to make immunoglobulins due to loss of T- and B-cell function) were treated with an adenoviral vector encoding adenovirus expressed (Adv)-IK17-scFv or control adenovirus-enhanced green fluorescent protein vector intravenously every 2 weeks for 16 weeks. RESULTS In LDLR(-/-) mice, infusion of IK17-Fab was able to sustain IK17 plasma levels for the first 8 weeks, but these diminished afterward due to increasing murine anti-IK17 antibody titers. Despite this, after 14 weeks, a 29% decrease in en face atherosclerosis was noted compared with phosphate-buffered saline-treated mice. In LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice, sustained levels of plasma IK17-scFv was achieved by Adv-IK17-scFv-mediated hepatic expression, which led to a 46% reduction (p < 0.001) in en face atherosclerosis compared with adenovirus-enhanced green fluorescent protein vector. Importantly, peritoneal macrophages isolated from Adv-IK17-scFv treated mice had decreased lipid accumulation compared with adenovirus-enhanced green fluorescent protein-treated mice. CONCLUSIONS These data support an important role for SR-mediated uptake of OxLDL in the pathogenesis of atherosclerosis and demonstrate that oxidation-specific antibodies reduce the progression of atherosclerosis, suggesting their potential in treating cardiovascular disease in humans.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Preusch MR, Baeuerle M, Albrecht C, Blessing E, Bischof M, Katus HA, Bea F. GDF-15 protects from macrophage accumulation in a mousemodel of advanced atherosclerosis. Eur J Med Res 2013; 18:19. [PMID: 23800095 PMCID: PMC3701574 DOI: 10.1186/2047-783x-18-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/29/2013] [Indexed: 01/10/2023] Open
Abstract
Background The cytokine growth differentiation factor-15 (GDF-15), a member of the TGF beta superfamily, has recently been discovered to play an important role in cardiovascular diseases. It is mostly expressed in macrophages of atherosclerotic lesions, but its impact on advanced atherosclerosis is still unknown. This study was performed to evaluate the effects of GDF-15 in an established mouse model of advanced atherosclerosis. Methods Thirty-eight LDL receptor deficient mice received a lethal body radiation. Half of the group was transplanted with bone marrow of GDF-15 deficient mice. Nineteen mice were transplanted with bone marrow from wild-type controls. After 24 weeks on an atherogenic diet, animals were euthanized and sections of the aortic sinus were prepared. Lesion size and lesion composition, as well as macrophage content,were evaluated. Results While demonstrating no difference in lesion size, LDL-receptor knockout mice transplanted with bone marrow from GDF-15 deficient mice showed enhanced macrophage accumulation and features of atherosclerotic plaque destabilization, such as thinning of fibrous caps. Immunostaining against intercellular adhesion molecule-1 further revealed an increased expression in mice receiving GDF-15-deficient bone marrow. Conclusions This is the first study that demonstrates a protective role of GDF-15 in advanced atherosclerosis and macrophage accumulation, possibly due to the reduced expression of adhesion molecules.
Collapse
Affiliation(s)
- Michael R Preusch
- Department of Internal Medicine, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
Ait-Oufella H, Herbin O, Lahoute C, Coatrieux C, Loyer X, Joffre J, Laurans L, Ramkhelawon B, Blanc-Brude O, Karabina S, Girard CA, Payré C, Yamamoto K, Binder CJ, Murakami M, Tedgui A, Lambeau G, Mallat Z. Group X Secreted Phospholipase A2 Limits the Development of Atherosclerosis in LDL Receptor–Null Mice. Arterioscler Thromb Vasc Biol 2013; 33:466-73. [DOI: 10.1161/atvbaha.112.300309] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Several secreted phospholipases A2 (sPLA2s), including group IIA, III, V, and X, have been linked to the development of atherosclerosis, which led to the clinical testing of A-002 (varespladib), a broad sPLA2 inhibitor for the treatment of coronary artery disease. Group X sPLA2 (PLA2G10) has the most potent hydrolyzing activity toward phosphatidylcholine and is believed to play a proatherogenic role.
Methods and Results—
Here, we show that
Ldlr
–/–
mice reconstituted with bone marrow from mouse group X–deficient mice (
Pla2g10
–/–
) unexpectedly display a doubling of plaque size compared with
Pla2g10
+/+
chimeric mice. Macrophages of
Pla2g10
–/–
mice are more susceptible to apoptosis in vitro, which is associated with a 4-fold increase of plaque necrotic core in vivo. In addition, chimeric
Pla2g10
–/–
mice show exaggerated T lymphocyte (Th)1 immune response, associated with enhanced T-cell infiltration in atherosclerotic plaques. Interestingly, overexpression of human PLA2G10 in murine bone marrow cells leads to significant reduction of Th1 response and to 50% reduction of lesion size.
Conclusion—
PLA2G10 expression in bone marrow cells controls a proatherogenic Th1 response and limits the development of atherosclerosis. The results may provide an explanation for the recently reported inefficacy of A-002 (varespladib) to treat patients with coronary artery disease. Indeed, A-002 is a nonselective sPLA2 inhibitor that inhibits both proatherogenic (groups IIA and V) and antiatherogenic (group X) sPLA2s. Our results suggest that selective targeting of individual sPLA2 enzymes may be a better strategy to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Olivier Herbin
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Charlotte Lahoute
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christelle Coatrieux
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Xavier Loyer
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Jeremie Joffre
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Ludivine Laurans
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Bhama Ramkhelawon
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Olivier Blanc-Brude
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Sonia Karabina
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christophe A. Girard
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christine Payré
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Kei Yamamoto
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christoph J. Binder
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Makoto Murakami
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Alain Tedgui
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Gérard Lambeau
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Ziad Mallat
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| |
Collapse
|
29
|
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: 1.3] [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
|
30
|
Chellan B, Koroleva EP, Sontag TJ, Tumanov AV, Fu YX, Getz GS, Reardon CA. LIGHT/TNFSR14 can regulate hepatic lipase expression by hepatocytes independent of T cells and Kupffer cells. PLoS One 2013; 8:e54719. [PMID: 23355893 PMCID: PMC3552851 DOI: 10.1371/journal.pone.0054719] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 12/14/2012] [Indexed: 12/17/2022] Open
Abstract
LIGHT/TNFSF14 is a costimulatory molecule expressed on activated T cells for activation and maintenance of T cell homeostasis. LIGHT over expressed in T cells also down regulates hepatic lipase levels in mice through lymphotoxin beta receptor (LTβR) signaling. It is unclear whether LIGHT regulates hepatic lipase directly by interacting with LTβR expressing cells in the liver or indirectly by activation of T cells, and whether Kupffer cells, a major cell populations in the liver that expresses the LTβR, are required. Here we report that LIGHT expression via an adenoviral vector (Ad-LIGHT) is sufficient to down regulate hepatic lipase expression in mice. Depletion of Kupffer cells using clodronate liposomes had no effect on LIGHT-mediated down regulation of hepatic lipase. LIGHT-mediated regulation of hepatic lipase is also independent of LIGHT expression by T cells or activation of T cells. This is demonstrated by the decreased hepatic lipase expression in the liver of Ad-LIGHT infected recombination activating gene deficient mice that lack mature T cells and by the Ad-LIGHT infection of primary hepatocytes. Hepatic lipase expression was not responsive to LIGHT when mice lacking LTβR globally or only on hepatocytes were infected with Ad-LIGHT. Therefore, our data argues that interaction of LIGHT with LTβR on hepatocytes, but not Kupffer cells, is sufficient to down regulate hepatic lipase expression and that this effect can be independent of LIGHT’s costimulatory function.
Collapse
Affiliation(s)
- Bijoy Chellan
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | | | - Timothy J. Sontag
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | | | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (YXF); (CAR)
| | - Godfrey S. Getz
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Catherine A. Reardon
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (YXF); (CAR)
| |
Collapse
|
31
|
Acute myocardial infarctions, strokes and influenza: seasonal and pandemic effects. Epidemiol Infect 2013; 141:735-44. [PMID: 23286343 DOI: 10.1017/s0950268812002890] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The incidence of myocardial infarctions and influenza follow similar seasonal patterns. To determine if acute myocardial infarctions (AMIs) and ischaemic strokes are associated with influenza activity, we built time-series models using data from the Nationwide Inpatient Sample. In these models, we used influenza activity to predict the incidence of AMI and ischaemic stroke. We fitted national models as well as models based on four geographical regions and five age groups. Across all models, we found consistent significant associations between AMIs and influenza activity, but not between ischaemic strokes and influenza. Associations between influenza and AMI increased with age, were greatest in those aged >80 years, and were present in all geographical regions. In addition, the natural experiment provided by the second wave of the influenza pandemic in 2009 provided further evidence of the relationship between influenza and AMI, because both series peaked in the same non-winter month.
Collapse
|
32
|
Abstract
Atherosclerosis is a chronic inflammatory disorder that is the underlying cause of most cardiovascular disease. Both cells of the vessel wall and cells of the immune system participate in atherogenesis. This process is heavily influenced by plasma lipoproteins, genetics, and the hemodynamics of the blood flow in the artery. A variety of small and large animal models have been used to study the atherogenic process. No model is ideal as each has its own advantages and limitations with respect to manipulation of the atherogenic process and modeling human atherosclerosis or lipoprotein profile. Useful large animal models include pigs, rabbits, and nonhuman primates. Due in large part to the relative ease of genetic manipulation and the relatively short time frame for the development of atherosclerosis, murine models are currently the most extensively used. Although not all aspects of murine atherosclerosis are identical to humans, studies using murine models have suggested potential biological processes and interactions that underlie this process. As it becomes clear that different factors may influence different stages of lesion development, the use of mouse models with the ability to turn on or delete proteins or cells in tissue specific and temporal manner will be very valuable.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
33
|
Wang L, Wang W, Shah PK, Song L, Yang M, Sharifi BG. Deletion of tenascin-C gene exacerbates atherosclerosis and induces intraplaque hemorrhage in Apo-E-deficient mice. Cardiovasc Pathol 2012; 21:398-413. [PMID: 22300502 DOI: 10.1016/j.carpath.2011.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/13/2011] [Accepted: 12/17/2011] [Indexed: 12/20/2022] Open
Abstract
AIMS Tenascin-C (TNC), a matricellular protein, is up-regulated in atherosclerotic plaques. We investigated whether the deletion of TNC gene affects the development of atherosclerosis in a murine model. METHODS TNC-/-/apo E-/- mice were generated and used for atherosclerosis studies. We compared these results to those observed in control groups of apo E-/- mice. RESULTS The en face analysis of aortic area showed that the mean aortic lesion area of the double knockout (KO) mice was significantly higher than that of control mice at different times after feeding of atherogenic diet; the accumulation of lesional macrophages and lipids was significantly higher. Analysis of cell adhesion molecules revealed that vascular cell adhesion molecule-1 (VCAM-1), but not intercellular adhesion molecule-1, was up-regulated 1 week after feeding of atherogenic diet in the double KO mouse as compared to apo E-/- mouse. Cell culture studies revealed that the expression of VCAM-1 in endothelial cells isolated from the double KO mouse is more sensitive to the tumor necrosis factor α stimulation than the cells isolated from apo E-/- mice. Cell adhesion studies showed that the adherence of RAW monocytic cells to the endothelial cells was significantly enhanced in the cultured endothelial cells from the TNC gene-deleted cells. Following the prolonged feeding of an atherogenic diet (28-30 weeks), the aortic and carotid atherosclerotic lesions frequently demonstrated large grossly visible areas of intraplaque hemorrhage in the double KO mice compared to control. CONCLUSIONS These data unveil a protective role for TNC in atherosclerosis and suggest that TNC signaling may have the potential to reduce atherosclerosis, in part by modulating VCAM-1 expression.
Collapse
Affiliation(s)
- Lai Wang
- Oppenheimer Atherosclerosis Research Center and the Division of Cardiology, Cedars Sinai Heart Institute, Los Angeles, CA 90048, USA
| | | | | | | | | | | |
Collapse
|
34
|
Gambone JE, Owens AP, Mackman N. Comment on "tissue factor-dependent chemokine production aggravates experimental colitis". Mol Med 2011; 17:1131; author reply 1132. [PMID: 22027905 DOI: 10.2119/molmed.2011.00303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 08/26/2011] [Indexed: 11/06/2022] Open
|
35
|
Getz GS, Vanderlaan PA, Reardon CA. Natural killer T cells in lipoprotein metabolism and atherosclerosis. Thromb Haemost 2011; 106:814-9. [PMID: 21946866 DOI: 10.1160/th11-05-0336] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 08/23/2011] [Indexed: 01/28/2023]
Abstract
Cells of both the innate and adaptive immune system participate in the development of atherosclerosis, a chronic inflammatory disorder of medium and large arteries. Natural killer T (NKT) cells express surface markers characteristic of natural killer cells and conventional T cells and bridge the innate and adaptive immune systems. The development and activation of NKT cells is dependent upon CD1d, a MHC-class I-type molecule that presents lipids, especially glycolipids to the T cell receptors on NKT cells. There are two classes of NKT cells; invariant NKT cells that express a semi-invariant T cell receptor and variant NKT cells. This review summarises studies in murine models in which the effect of the activation, overexpression or deletion of NKT cells or only invariant NKT cells on atherosclerosis has been examined.
Collapse
Affiliation(s)
- G S Getz
- University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
36
|
Taleb S, Herbin O, Ait-Oufella H, Verreth W, Gourdy P, Barateau V, Merval R, Esposito B, Clément K, Holvoet P, Tedgui A, Mallat Z. Potential role of regulatory T cells in reversing obesity-linked insulin resistance and diabetic nephropathy. Arterioscler Thromb Vasc Biol 2011; 27:2691-8. [PMID: 17690315 DOI: 10.1161/atvbaha.107.149567] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To assess the potential role of FoxP3-expressing regulatory T cells (Tregs) in reversing obesity-linked insulin resistance and diabetic nephropathy in rodent models and humans. RESEARCH DESIGN AND METHODS To characterize the role of Tregs in insulin resistance, human visceral adipose tissue was first evaluated for Treg infiltration and second, the db/db mouse model was evaluated. RESULTS Obese patients with insulin resistance displayed significantly decreased natural Tregs but an increase in adaptive Tregs in their visceral adipose tissue as compared with lean control subjects. To further evaluate the pathogenic role of Tregs in insulin resistance, the db/db mouse model was used. Treg depletion using an anti-CD25 monoclonal antibody enhanced insulin resistance as shown by increased fasting blood glucose levels as well as an impaired insulin sensitivity. Moreover, Treg-depleted db/db mice developed increased signs of diabetic nephropathy, such as albuminuria and glomerular hyperfiltration. This was paralleled by a proinflammatory milieu in both murine visceral adipose tissue and the kidney. Conversely, adoptive transfer of CD4(+)FoxP3(+) Tregs significantly improved insulin sensitivity and diabetic nephropathy. Accordingly, there was increased mRNA expression of FoxP3 as well as less abundant proinflammatory CD8(+)CD69(+) T cells in visceral adipose tissue and kidneys of Treg-treated animals. CONCLUSIONS Data suggest a potential therapeutic value of Tregs to improve insulin resistance and end organ damage in type 2 diabetes by limiting the proinflammatory milieu.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- CD28 Antigens/genetics
- CD28 Antigens/metabolism
- Cell Proliferation
- Cells, Cultured
- Dietary Fats/administration & dosage
- Disease Models, Animal
- Female
- Forkhead Transcription Factors/metabolism
- Immunity, Cellular/genetics
- Interferon-gamma/metabolism
- Leptin/deficiency
- Leptin/genetics
- Leptin/metabolism
- Lipids/blood
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Obese
- Obesity/complications
- Obesity/genetics
- Obesity/immunology
- Obesity/metabolism
- Obesity/pathology
- Receptors, Leptin/deficiency
- Receptors, Leptin/genetics
- Receptors, Leptin/metabolism
- Signal Transduction/genetics
- T-Lymphocytes, Regulatory/metabolism
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Soraya Taleb
- Institut National de la Sante et de la Recherche Medicale, U689, 41 Bd de la Chapelle, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hayashi C, Viereck J, Hua N, Phinikaridou A, Madrigal AG, Gibson FC, Hamilton JA, Genco CA. Porphyromonas gingivalis accelerates inflammatory atherosclerosis in the innominate artery of ApoE deficient mice. Atherosclerosis 2010. [PMID: 21251656 DOI: 10.1016/j.atherosclerosis.2010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Studies in humans support a role for the oral pathogen Porphyromonas gingivalis in the development of inflammatory atherosclerosis. The goal of this study was to determine if P. gingivalis infection accelerates inflammation and atherosclerosis in the innominate artery of mice, an artery which has been reported to exhibit many features of human atherosclerotic disease, including plaque rupture. METHODS AND RESULTS Apolipoprotein E-deficient (ApoE-/-) mice were orally infected with P. gingivalis, and magnetic resonance imaging (MRI) was used to monitor the progression of atherosclerosis in live mice. P. gingivalis infected mice exhibited a statistically significant increase in atherosclerotic plaque in the innominate artery as compared to uninfected mice. Polarized light microscopy and immunohistochemistry revealed that the innominate arteries of infected mice had increased lipids, macrophages and T cells as compared to uninfected mice. Increases in plaque, total cholesterol esters and cholesterol monohydrate crystals, macrophages, and T cells were prevented by immunization with heat-killed P. gingivalis prior to pathogen exposure. CONCLUSIONS These are the first studies to demonstrate progression of inflammatory plaque accumulation in the innominate arteries by in vivo MRI analysis following pathogen exposure, and to document protection from plaque progression in the innominate artery via immunization.
Collapse
Affiliation(s)
- Chie Hayashi
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Hayashi C, Viereck J, Hua N, Phinikaridou A, Madrigal AG, Gibson FC, Hamilton JA, Genco CA. Porphyromonas gingivalis accelerates inflammatory atherosclerosis in the innominate artery of ApoE deficient mice. Atherosclerosis 2010; 215:52-9. [PMID: 21251656 DOI: 10.1016/j.atherosclerosis.2010.12.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 11/29/2010] [Accepted: 12/10/2010] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Studies in humans support a role for the oral pathogen Porphyromonas gingivalis in the development of inflammatory atherosclerosis. The goal of this study was to determine if P. gingivalis infection accelerates inflammation and atherosclerosis in the innominate artery of mice, an artery which has been reported to exhibit many features of human atherosclerotic disease, including plaque rupture. METHODS AND RESULTS Apolipoprotein E-deficient (ApoE-/-) mice were orally infected with P. gingivalis, and magnetic resonance imaging (MRI) was used to monitor the progression of atherosclerosis in live mice. P. gingivalis infected mice exhibited a statistically significant increase in atherosclerotic plaque in the innominate artery as compared to uninfected mice. Polarized light microscopy and immunohistochemistry revealed that the innominate arteries of infected mice had increased lipids, macrophages and T cells as compared to uninfected mice. Increases in plaque, total cholesterol esters and cholesterol monohydrate crystals, macrophages, and T cells were prevented by immunization with heat-killed P. gingivalis prior to pathogen exposure. CONCLUSIONS These are the first studies to demonstrate progression of inflammatory plaque accumulation in the innominate arteries by in vivo MRI analysis following pathogen exposure, and to document protection from plaque progression in the innominate artery via immunization.
Collapse
Affiliation(s)
- Chie Hayashi
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang X. T-cell co-stimulators as anti-inflammatory targets for atherosclerotic disease. Future Cardiol 2010; 2:187-95. [PMID: 19804075 DOI: 10.2217/14796678.2.2.187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Currently, most pharmacological therapies for atherosclerosis rely on lowering plasma low-density lipoprotein levels. Several ongoing clinical trials are testing the possibility of reducing atherosclerosis with drugs that raise plasma high-density lipoprotein levels and/or promote high-density lipoprotein-mediated protective functions. Atherosclerosis can also be treated by targeting inflammatory cells. Recent studies have shown that atherosclerosis is primarily an inflammatory disease and that immune cells, particularly T cells, are found in atherosclerotic lesions throughout the early and late stages. Therefore, therapies that modulate T-cell co-stimulators might slow down the atherosclerosis process by inhibiting T-cell-mediated inflammation.
Collapse
Affiliation(s)
- Xiaosong Wang
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| |
Collapse
|
40
|
Bennett BJ, Wang SS, Wang X, Wu X, Lusis AJ. Genetic regulation of atherosclerotic plaque size and morphology in the innominate artery of hyperlipidemic mice. Arterioscler Thromb Vasc Biol 2009; 29:348-55. [PMID: 19122174 DOI: 10.1161/atvbaha.108.176685] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We sought to determine the genetic factors contributing to atherosclerotic plaque size and cellular composition in the innominate artery, a murine model of advanced atherosclerosis. METHODS AND RESULTS We examined genetic contributions to innominate atherosclerotic plaque size and cellular composition in an intercross between C57BL/6J.Apoe(-/-), a strain susceptible to aortic lesions, and C3H/HeJ.Apoe(-/-), a strain resistant to aortic lesions. Surprisingly, total innominate lesion size was similar in the two strains. Genetic analyses identified one novel locus on Chromosome 2 for innominate artery lesion size, a significant locus for fibrous cap thickness on Chromosome 15, and several suggestive loci for cellular composition, all distinct from loci influencing aortic lesions. The Chromosome 2 locus contains a candidate, CD44. We show that CD44 is expressed in the innominate artery and differs strikingly in expression between the parental strains. CONCLUSION Multiple aspects of innominate lesion composition are genetically determined, but in a manner largely independent of the genetic contributions to aortic lesions.
Collapse
Affiliation(s)
- Brian J Bennett
- Department of Medicine, 675 Charles E. Young Dr South, 3730 MRL, School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1679, USA.
| | | | | | | | | |
Collapse
|
41
|
Modes of defining atherosclerosis in mouse models: relative merits and evolving standards. Methods Mol Biol 2009; 573:1-15. [PMID: 19763919 DOI: 10.1007/978-1-60761-247-6_1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mouse models have become the most common model for defining mechanisms of atherosclerotic disease. Many genetic manipulations have enabled the development of atherosclerosis in mice due to either endogenous or diet-induced hypercholesterolemia. This availability of lesion-susceptible mice has facilitated many studies using pharmacological and genetic approaches. Unfortunately, this expansive literature on mouse atherosclerosis has generated many contradictions on the role of specific pathways. A contributor to these inconsistencies may be the multiple modes in which atherosclerosis is evaluated. Also, for each specific technique, there are no consistent standards applied to the measurements. This chapter will discuss the imaging, biochemical, and compositional modes of evaluating atherosclerosis with suggestions for standard execution of these techniques.
Collapse
|
42
|
VanderLaan PA, Reardon CA, Thisted RA, Getz GS. VLDL best predicts aortic root atherosclerosis in LDL receptor deficient mice. J Lipid Res 2008; 50:376-385. [PMID: 18957695 DOI: 10.1194/jlr.m800284-jlr200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hyperlipidemia is a major risk factor for developing atherosclerosis in humans, and epidemiological studies have correlated specific lipoprotein levels with cardiovascular disease risk. Murine models of atherosclerosis rely on the induction of hyperlipidemia for vascular lesions to form, but the pathogenic contributions attributed to different lipoprotein populations are not well defined. To address this issue, we analyzed over 300 LDL receptor (LDLR) deficient mice that have been fed a high-fat diet and for which a full lipoprotein profile and aortic root atherosclerosis values were assessed. Overall, aortic root atherosclerosis is best predicted by plasma VLDL cholesterol levels with less predictive value derived from either LDL or HDL cholesterol. Triglyceride levels are more atherogenic in female mice, especially immune competent females, and depletion of the adaptive immune system leads to a global reduction in plasma lipid levels and aortic root lesion size yet does not appear to alter the atherogenic potential of individual lipoprotein subspecies. In contrast, HDL-cholesterol is a better predictor of aortic root atherosclerosis in apoE-deficient mice. In summary, this large scale analysis of high-fat diet fed LDLR deficient mice highlight the relationship between different plasma lipid components, especially VLDL-cholesterol, and aortic root atherosclerosis.
Collapse
Affiliation(s)
- Paul A VanderLaan
- Departments of Pathology and Health Studies, University of Chicago, Chicago, IL 60637
| | - Catherine A Reardon
- Departments of Pathology and Health Studies, University of Chicago, Chicago, IL 60637
| | - Ronald A Thisted
- Biostatistics, Epidemiology, and Health Services Research, University of Chicago, Chicago, IL 60637
| | - Godfrey S Getz
- Departments of Pathology and Health Studies, University of Chicago, Chicago, IL 60637; Biostatistics, Epidemiology, and Health Services Research, University of Chicago, Chicago, IL 60637.
| |
Collapse
|
43
|
Grönros J, Wikström J, Brandt-Eliasson U, Forsberg GB, Behrendt M, Hansson GI, Gan LM. Effects of rosuvastatin on cardiovascular morphology and function in an ApoE-knockout mouse model of atherosclerosis. Am J Physiol Heart Circ Physiol 2008; 295:H2046-53. [PMID: 18790840 DOI: 10.1152/ajpheart.00133.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated the effects of rosuvastatin on plaque progression and in vivo coronary artery function in apolipoprotein E-knockout (ApoE-KO) mice, using noninvasive high-resolution ultrasound techniques. Eight-week-old male ApoE-KO mice (n = 20) were fed a high-fat diet with or without rosuvastatin (10 micromol.kg(-1).day(-1)) for 16 wk. When compared with control, rosuvastatin reduced total cholesterol levels (P < 0.05) and caused significant retardation of lesion progression in the brachiocephalic artery, as visualized in vivo using an ultrasound biomicroscope (P < 0.05). Histological analysis confirmed the reduction of brachiocephalic atherosclerosis and also revealed an increase in collagen content in the statin-treated group (P < 0.05). Coronary volumetric flow was measured by simultaneous recording of Doppler velocity signals and left coronary artery morphology before and during adenosine infusion. The hyperemic flow in response to adenosine was significantly greater in left coronary artery following 16 wk of rosuvastatin treatment (P < 0.001), whereas the baseline flow was similar in both groups. In conclusion, rosuvastatin reduced brachiocephalic artery atherosclerotic plaques in ApoE-KO mice. Coronary artery function assessed using recently developed in vivo ultrasound-based protocols, also improved.
Collapse
Affiliation(s)
- J Grönros
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Göteborg University, Göteborg, Sweden
| | | | | | | | | | | | | |
Collapse
|
44
|
Peng D, Hiipakka RA, Dai Q, Guo J, Reardon CA, Getz GS, Liao S. Antiatherosclerotic effects of a novel synthetic tissue-selective steroidal liver X receptor agonist in low-density lipoprotein receptor-deficient mice. J Pharmacol Exp Ther 2008; 327:332-42. [PMID: 18723776 DOI: 10.1124/jpet.108.142687] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Liver X receptor (LXR) agonists have the potential to treat atherosclerosis based on their ability to enhance reverse cholesterol transport. However, their side effects, such as induction of liver lipogenesis and triglyceridemia, may limit their pharmaceutical development. In contrast to the nonsteroidal LXR agonist N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide (T0901317), 3alpha, 6alpha, 24-trihydroxy-24, 24-di(trifluoromethyl)-5beta-cholane (ATI-829), a novel potent synthetic steroidal LXR agonist, was a poor inducer of sterol regulatory element-binding protein 1c expression in hepatoma HepG2 cells, whereas both compounds increased ABCA1 expression in macrophage THP-1 cells. In male low-density lipoprotein receptor-deficient mice, ATI-829 selectively activated LXR target gene expression in mouse intestines and macrophages but not in the liver. A significant increase in liver triglyceride and plasma triglyceriderich small very low-density lipoprotein (VLDL) was observed in T0901317 but not ATI-829-treated mice. Compared with vehicle-treated mice, atherosclerosis development was significantly inhibited in the innominate artery after treatment with either compound. However, in the aortic root, inhibition of atherosclerosis was only observed in the right (right coronary artery-associated sinus) but not the left coronary-related sinus (left coronary artery-associated sinus; LC) of mice treated with either compound. Lesions in the innominate artery were less complex after treatment with either compound and contained mostly macrophage foam cells. In contrast, LC lesions were more complex and had a large collagen-positive fibrous cap and less macrophage foam cell area after treatment with either compound. The T0901317-induced hypertriglyceridemia was accompanied by an increase in small triglyceride-rich VLDL that may influence LXR agonist-mediated antiatherosclerotic effects at certain vascular sites. ATI-829, by selectively activating LXR in certain tissues without inducing hypertriglyceridemia, is a good candidate for drug development.
Collapse
Affiliation(s)
- Dacheng Peng
- Ben May Department for Cancer Research, University of Chicago, W334, 929 E. 57th Street, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Differential distribution of vasa vasorum in different vascular beds in humans. Atherosclerosis 2008; 199:47-54. [DOI: 10.1016/j.atherosclerosis.2007.09.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 08/22/2007] [Accepted: 09/04/2007] [Indexed: 11/18/2022]
|
46
|
Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J 2008; 22:3595-606. [PMID: 18559988 DOI: 10.1096/fj.08-112201] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is now recognized as an inflammatory disease involving the vascular wall. Recent results indicate that acute inflammation does not simply passively resolve as previously assumed but is actively terminated by a homeostatic process that is governed by specific lipid-derived mediators initiated by lipoxygenases. Experiments with animals and humans support a proinflammatory role for the 5-lipoxygenase system. In contrast, results from animal experiments show a range of responses with the 12/15-lipoxygenase pathways in atherosclerosis. To date, the only two clinical epidemiology human studies both support an antiatherogenic role for 12/15-lipoxygenase downstream actions. We tested the hypothesis that atherosclerosis results from a failure in the resolution of local inflammation by analyzing apolipoprotein E-deficient mice with 1) global leukocyte 12/15-lipoxygenase deficiency, 2) normal enzyme expression, or 3) macrophage-specific 12/15-lipoxygenase overexpression. Results from these indicate that 12/15-lipoxygenase expression protects mice against atherosclerosis via its role in the local biosynthesis of lipid mediators, including lipoxin A(4), resolvin D1, and protectin D1. These mediators exert potent agonist actions on macrophages and vascular endothelial cells that can control the magnitude of the local inflammatory response. Taken together, these findings suggest that a failure of local endogenous resolution mechanisms may underlie the unremitting inflammation that fuels atherosclerosis.
Collapse
Affiliation(s)
- Aksam J Merched
- Department of Molecular and Cellular Biology, Baylor College of Medicine and St. Luke's Episcopal Hospital, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
47
|
Peng D, Hiipakka RA, Reardon CA, Getz GS, Liao S. Differential anti-atherosclerotic effects in the innominate artery and aortic sinus by the liver X receptor agonist T0901317. Atherosclerosis 2008; 203:59-66. [PMID: 18639878 DOI: 10.1016/j.atherosclerosis.2008.05.058] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 04/24/2008] [Accepted: 05/29/2008] [Indexed: 10/22/2022]
Abstract
Activation of liver X receptors (LXRs) has been reported to reduce atherosclerosis in mouse models. However, this can be associated with enhanced liver de novo lipogenesis and elevation of plasma triglyceride-rich VLDL, which may limit its clinical use. In this study, we administered orally the LXR agonist T0901317 to male LDLR-/- mice fed a Western diet. This induced a persistent enhanced hypertriglyceridemia by largely increasing plasma triglyceride-rich VLDL. T0901317 treatment decreased atherosclerosis with a much more pronounced response and dose dependence in the innominate artery than in the aortic sinus. Lesions in the innominate artery were less complex containing mostly macrophage foam cells in T0901317-treated mice. However, in the aortic root, a significant reduction of atherosclerosis was seen only in the right coronary-related aortic sinus (RC) of T0901317-treated mice. Increasing the dose of T0901317 did not extend atheroprotection to the other sinuses of the aortic root. Lesions in the RC were less complex both in T0901317 and vehicle-treated mice with macrophage foam cells predominating. On the other hand, in T0901317-treated mice, the left coronary-related sinus (LC) lesions while not reduced in size, were more complex with a large fibrous cap and necrotic core, more collagen-positive areas, and variable macrophage foam cell content compared to vehicle-treated mice. These data suggest that activation of LXR by T0901317 had differential anti-atherosclerotic effects in two arterial regions in mice with hypertriglyceridemia.
Collapse
Affiliation(s)
- Dacheng Peng
- Ben May Department for Cancer Research, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA.
| | | | | | | | | |
Collapse
|
48
|
Out R, Hoekstra M, Habets K, Meurs I, de Waard V, Hildebrand RB, Wang Y, Chimini G, Kuiper J, Van Berkel TJC, Van Eck M. Combined deletion of macrophage ABCA1 and ABCG1 leads to massive lipid accumulation in tissue macrophages and distinct atherosclerosis at relatively low plasma cholesterol levels. Arterioscler Thromb Vasc Biol 2007; 28:258-64. [PMID: 18006857 DOI: 10.1161/atvbaha.107.156935] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The purpose of this study was to evaluate the effect of the combined deletion of ABCA1 and ABCG1 expression in macrophages on foam cell formation and atherosclerosis. METHODS AND RESULTS LDL receptor knockout (KO) mice were transplanted with bone marrow from ABCA1/ABCG1 double KO (dKO) mice. Plasma cholesterol levels after 6 weeks of Western-type diet (WTD) feeding were significantly lower in dKO transplanted mice than ABCA1 KO, ABCG1 KO, and control transplanted animals. Extreme foam cell formation was present in macrophages of various tissues and the peritoneal cavity of dKO transplanted animals. Furthermore, severe hypoplasia of the thymus and a significant decrease in CD4-positive T cells in blood was observed. Despite relatively low plasma cholesterol levels dKO transplanted animals developed lesion sizes of 156+/-19x10(3) microm2 after only 6 weeks of WTD feeding. Lesions, however, were smaller than single ABCA1 KO transplanted animals (226+/-30x10(3) microm2; P<0.05) and not significantly different from single ABCG1 KO (117+/-22x10(3) microm2) and WT transplanted mice (112+/-15x10(3) microm2). CONCLUSIONS Macrophage ABCA1 and ABCG1 play a crucial role in the prevention of macrophage foam cell formation, whereas combined deletion only modestly influences atherosclerosis which is associated with an attenuated increase in WTD-induced plasma cholesterol and decreased proinflammatory CD4-positive T cell counts.
Collapse
Affiliation(s)
- Ruud Out
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
MacDonald MLE, Singaraja RR, Bissada N, Ruddle P, Watts R, Karasinska JM, Gibson WT, Fievet C, Vance JE, Staels B, Hayden MR. Absence of stearoyl-CoA desaturase-1 ameliorates features of the metabolic syndrome in LDLR-deficient mice. J Lipid Res 2007; 49:217-29. [PMID: 17960025 DOI: 10.1194/jlr.m700478-jlr200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A combination of the interrelated metabolic risk factors obesity, insulin resistance, dyslipidemia, and hypertension, often described as the "metabolic syndrome," is known to increase the risk of developing cardiovascular disease and diabetes. Stearoyl-coenzyme A desaturase (SCD) activity has been implicated in the metabolic syndrome, but detailed studies of the beneficial metabolic effects of SCD deficiency have been limited. Here, we show that absence of the Scd1 gene product reduces plasma triglycerides and reduces weight gain in severely hyperlipidemic low density lipoprotein receptor (LDLR)-deficient mice challenged with a Western diet. Absence of SCD1 also increases insulin sensitivity, as measured by intraperitoneal glucose and insulin tolerance testing. SCD1 deficiency dramatically reduces hepatic lipid accumulation while causing more modest reductions in plasma apolipoproteins, suggesting that in conditions of sustained hyperlipidemia, SCD1 functions primarily to mediate lipid stores. In addition, absence of SCD1 partially ameliorates the undesirable hypertriglyceridemic effect of antiatherogenic liver X receptor agonists. Our results demonstrate that constitutive reduction of SCD activity improves the metabolic phenotype of LDLR-deficient mice on a Western diet.
Collapse
Affiliation(s)
- Marcia L E MacDonald
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
VanderLaan PA, Reardon CA, Sagiv Y, Blachowicz L, Lukens J, Nissenbaum M, Wang CR, Getz GS. Characterization of the natural killer T-cell response in an adoptive transfer model of atherosclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1100-7. [PMID: 17322392 PMCID: PMC1864866 DOI: 10.2353/ajpath.2007.060188] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Natural killer T (NKT) cells have recently been implicated in atherogenesis, primarily for their ability to recognize and respond to lipid antigens. Because the atherosclerotic lesion is characterized by the retention and modification of lipids in the vascular wall, NKT cells may be involved in promoting the local vascular inflammatory response. Here, we investigate the proatherogenic role of NKT cells in an adoptive transfer model of atherosclerosis, using as recipients immune-deficient, atherosclerosis-susceptible RAG1(-/-)LDLR(-/-) mice. The adoptive transfer of an NKT cell-enriched splenocyte population from Valpha14Jalpha18 T-cell receptor transgenic mice resulted in a 73% increase in aortic root lesion area compared with recipients of NKT cell-deficient splenocytes derived from CD1d(-/-) mice after 12 weeks of Western-type diet feeding. The total serum from hypercholesterolemic mice leads to a small but significant activation of Valpha14Jalpha18 T-cell receptor-expressing hybridoma line by dendritic cells that is CD1d-dependent. Therefore, these studies demonstrate that NKT cells are proatherogenic in the absence of exogenous stimulation, and this activity is likely associated with endogenous lipid antigens carried by lipoproteins in the circulation and perhaps also in the atherosclerotic plaque.
Collapse
Affiliation(s)
- Paul A VanderLaan
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|