351
|
Getz GS, Reardon CA. ApoE knockout and knockin mice: the history of their contribution to the understanding of atherogenesis. J Lipid Res 2016; 57:758-66. [PMID: 27015743 DOI: 10.1194/jlr.r067249] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Indexed: 12/16/2022] Open
Abstract
ApoE is a multifunctional protein that is expressed by many cell types that influences many aspects of cardiovascular physiology. In humans, there are three major allelic variants that differentially influence lipoprotein metabolism and risk for the development of atherosclerosis. Apoe-deficient mice and human apoE isoform knockin mice, as well as hypomorphic Apoe mice, have significantly contributed to our understanding of the role of apoE in lipoprotein metabolism, monocyte/macrophage biology, and atherosclerosis. This brief history of these mouse models will highlight their contribution to the understanding of the role of apoE in these processes. These Apoe(-/-) mice have also been extensively utilized as an atherosensitive platform upon which to assess the impact of modulator genes on the development and regression of atherosclerosis.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology University of Chicago, Chicago, IL
| | | |
Collapse
|
352
|
Wagner A, Cohen N, Kelder T, Amit U, Liebman E, Steinberg DM, Radonjic M, Ruppin E. Drugs that reverse disease transcriptomic signatures are more effective in a mouse model of dyslipidemia. Mol Syst Biol 2016; 11:791. [PMID: 26148350 PMCID: PMC4380926 DOI: 10.15252/msb.20145486] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High-throughput omics have proven invaluable in studying human disease, and yet day-to-day clinical practice still relies on physiological, non-omic markers. The metabolic syndrome, for example, is diagnosed and monitored by blood and urine indices such as blood cholesterol levels. Nevertheless, the association between the molecular and the physiological manifestations of the disease, especially in response to treatment, has not been investigated in a systematic manner. To this end, we studied a mouse model of diet-induced dyslipidemia and atherosclerosis that was subject to various drug treatments relevant to the disease in question. Both physiological data and gene expression data (from the liver and white adipose) were analyzed and compared. We find that treatments that restore gene expression patterns to their norm are associated with the successful restoration of physiological markers to their baselines. This holds in a tissue-specific manner—treatments that reverse the transcriptomic signatures of the disease in a particular tissue are associated with positive physiological effects in that tissue. Further, treatments that introduce large non-restorative gene expression alterations are associated with unfavorable physiological outcomes. These results provide a sound basis to in silico methods that rely on omic metrics for drug repurposing and drug discovery by searching for compounds that reverse a disease's omic signatures. Moreover, they highlight the need to develop drugs that restore the global cellular state to its healthy norm rather than rectify particular disease phenotypes.
Collapse
Affiliation(s)
- Allon Wagner
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- Department of Electrical Engineering and Computer Science, University of CaliforniaBerkeley, CA, USA
- * Corresponding author. Tel. +972 3 640 5378; E-mail:
| | - Noa Cohen
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
| | - Thomas Kelder
- Microbiology and Systems Biology, TNOZeist, the Netherlands
| | - Uri Amit
- Neufeld Cardiac Research Institute, Tel Aviv UniversityTel Aviv, Israel
- Regenerative Medicine Stem Cells and Tissue Engineering Center, Sheba Medical CenterTel Hashomer, Israel
| | - Elad Liebman
- Department of Computer Science, University of Texas at AustinAustin, TX, USA
| | - David M Steinberg
- Department of Statistics and Operations Research, Tel Aviv UniversityTel Aviv, Israel
| | | | - Eytan Ruppin
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- The Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
- Department of Computer Science, Institute of Advanced Computer Sciences (UMIACS) & the Center for Bioinformatics and Computational Biology, University of MarylandCollege Park, MD, USA
- ** Corresponding author. Tel. +972 3 640 6528; E-mail:
| |
Collapse
|
353
|
Lee-Rueckert M, Escola-Gil JC, Kovanen PT. HDL functionality in reverse cholesterol transport--Challenges in translating data emerging from mouse models to human disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:566-83. [PMID: 26968096 DOI: 10.1016/j.bbalip.2016.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Whereas LDL-derived cholesterol accumulates in atherosclerotic lesions, HDL particles are thought to facilitate removal of cholesterol from the lesions back to the liver thereby promoting its fecal excretion from the body. Because generation of cholesterol-loaded macrophages is inherent to atherogenesis, studies on the mechanisms stimulating the release of cholesterol from these cells and its ultimate excretion into feces are crucial to learn how to prevent lesion development or even induce lesion regression. Modulation of this key anti-atherogenic pathway, known as the macrophage-specific reverse cholesterol transport, has been extensively studied in several mouse models with the ultimate aim of applying the emerging knowledge to humans. The present review provides a detailed comparison and critical analysis of the various steps of reverse cholesterol transport in mouse and man. We attempt to translate this in vivo complex scenario into practical concepts, which could serve as valuable tools when developing novel HDL-targeted therapies.
Collapse
|
354
|
Uwiera RR, Mangat R, Kelly S, Uwiera TC, Proctor SD. Long-Term Catheterization of the Intestinal Lymph Trunk and Collection of Lymph in Neonatal Pigs. J Vis Exp 2016. [PMID: 27023826 DOI: 10.3791/53457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Catheterization of the intestinal lymph trunk in neonatal pigs is a technique allowing for the long-term collection of large quantities of intestinal (central) efferent lymph. Importantly, the collection of central lymph from the intestine enables researchers to study both the mechanisms and lipid constitutes associated with lipid metabolism, intestinal inflammation and cancer metastasis, as well as cells involved in immune function and immunosurveillance. A ventral mid-line surgical approach permits excellent surgical exposure to the cranial abdomen and relatively easy access to the intestinal lymph trunk vessel that lies near the pancreas and the right ventral segment of the portal vein underneath the visceral aspect of the right liver lobe. The vessel is meticulously dissected and released from the surrounding fascia and then dilated with sutures allowing for insertion and subsequent securing of the catheter into the vessel. The catheter is exteriorized and approximately 1 L/24 hr of lymph is collected over a 7 day period. While this technique enables the collection of large quantities of central lymph over an extended period of time, the success depends on careful surgical dissection, tissue handling and close attention to proper surgical technique. This is particularly important with surgeries in young animals as the lymph vessels can easily tear, potentially leading to surgical and experimental failure. The video demonstrates an excellent surgical technique for the collection of intestinal lymph.
Collapse
Affiliation(s)
- Richard R Uwiera
- Department of Agricultural, Food and Nutritional Science, University of Alberta;
| | - Rabban Mangat
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| | - Sandra Kelly
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| | | | - Spencer D Proctor
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| |
Collapse
|
355
|
Kuehn C, Tauchi M, Stumpf C, Daniel C, Bäuerle T, Schwarz M, Kerek F, Steinkasserer A, Zinser E, Achenbach S, Dietel B. Suppression of proatherogenic leukocyte interactions by MCS-18 – Impact on advanced atherosclerosis in ApoE-deficient mice. Atherosclerosis 2016; 245:101-10. [DOI: 10.1016/j.atherosclerosis.2015.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/20/2015] [Accepted: 12/01/2015] [Indexed: 12/11/2022]
|
356
|
McKittrick CM, Kennedy S, Oldroyd KG, McGinty S, McCormick C. Modelling the Impact of Atherosclerosis on Drug Release and Distribution from Coronary Stents. Ann Biomed Eng 2016; 44:477-87. [PMID: 26384667 PMCID: PMC4764635 DOI: 10.1007/s10439-015-1456-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/09/2015] [Indexed: 11/24/2022]
Abstract
Although drug-eluting stents (DES) are now widely used for the treatment of coronary heart disease, there remains considerable scope for the development of enhanced designs which address some of the limitations of existing devices. The drug release profile is a key element governing the overall performance of DES. The use of in vitro, in vivo, ex vivo, in silico and mathematical models has enhanced understanding of the factors which govern drug uptake and distribution from DES. Such work has identified the physical phenomena determining the transport of drug from the stent and through tissue, and has highlighted the importance of stent coatings and drug physical properties to this process. However, there is limited information regarding the precise role that the atherosclerotic lesion has in determining the uptake and distribution of drug. In this review, we start by discussing the various models that have been used in this research area, highlighting the different types of information they can provide. We then go on to describe more recent methods that incorporate the impact of atherosclerotic lesions.
Collapse
Affiliation(s)
- C M McKittrick
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | - S Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - K G Oldroyd
- West of Scotland Region Heart and Lung Centre, Golden Jubilee National Hospital, Dunbartonshire, UK
| | - S McGinty
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - C McCormick
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
357
|
Yeang C, Cotter B, Tsimikas S. Experimental Animal Models Evaluating the Causal Role of Lipoprotein(a) in Atherosclerosis and Aortic Stenosis. Cardiovasc Drugs Ther 2016; 30:75-85. [DOI: 10.1007/s10557-015-6634-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
358
|
|
359
|
Bagalkot V, Badgeley MA, Kampfrath T, Deiuliis JA, Rajagopalan S, Maiseyeu A. Hybrid nanoparticles improve targeting to inflammatory macrophages through phagocytic signals. J Control Release 2015; 217:243-55. [PMID: 26386437 PMCID: PMC4874242 DOI: 10.1016/j.jconrel.2015.09.027] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/27/2015] [Accepted: 09/14/2015] [Indexed: 12/18/2022]
Abstract
Macrophages are innate immune cells with great phenotypic plasticity, which allows them to regulate an array of physiological processes such as host defense, tissue repair, and lipid/lipoprotein metabolism. In this proof-of-principle study, we report that macrophages of the M1 inflammatory phenotype can be selectively targeted by model hybrid lipid-latex (LiLa) nanoparticles bearing phagocytic signals. We demonstrate a simple and robust route to fabricate nanoparticles and then show their efficacy through imaging and drug delivery in inflammatory disease models of atherosclerosis and obesity. Self-assembled LiLa nanoparticles can be modified with a variety of hydrophobic entities such as drug cargos, signaling lipids, and imaging reporters resulting in sub-100nm nanoparticles with low polydispersities. The optimized theranostic LiLa formulation with gadolinium, fluorescein and "eat-me" phagocytic signals (Gd-FITC-LiLa) a) demonstrates high relaxivity that improves magnetic resonance imaging (MRI) sensitivity, b) encapsulates hydrophobic drugs at up to 60% by weight, and c) selectively targets inflammatory M1 macrophages concomitant with controlled release of the payload of anti-inflammatory drug. The mechanism and kinetics of the payload discharge appeared to be phospholipase A2 activity-dependent, as determined by means of intracellular Förster resonance energy transfer (FRET). In vivo, LiLa targets M1 macrophages in a mouse model of atherosclerosis, allowing noninvasive imaging of atherosclerotic plaque by MRI. In the context of obesity, LiLa particles were selectively deposited to M1 macrophages within inflamed adipose tissue, as demonstrated by single-photon intravital imaging in mice. Collectively, our results suggest that phagocytic signals can preferentially target inflammatory macrophages in experimental models of atherosclerosis and obesity, thus opening the possibility of future clinical applications that diagnose/treat these conditions. Tunable LiLa nanoparticles reported here can serve as a model theranostic platform with application in various types of imaging of the diseases such as cardiovascular disorders, obesity, and cancer where macrophages play a pathogenic role.
Collapse
Affiliation(s)
- Vaishali Bagalkot
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Marcus A Badgeley
- Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210, United States
| | - Thomas Kampfrath
- Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210, United States
| | - Jeffrey A Deiuliis
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, MD 21201, United States; Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210, United States
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, MD 21201, United States; Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210, United States
| | - Andrei Maiseyeu
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, MD 21201, United States; Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
360
|
Lagraauw HM, Kuiper J, Bot I. Acute and chronic psychological stress as risk factors for cardiovascular disease: Insights gained from epidemiological, clinical and experimental studies. Brain Behav Immun 2015; 50:18-30. [PMID: 26256574 DOI: 10.1016/j.bbi.2015.08.007] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of death worldwide and identification and therapeutic modulation of all its risk factors is necessary to ensure a lower burden on the patient and on society. The physiological response to acute and chronic stress exposure has long been recognized as a potent modulator of immune, endocrine and metabolic pathways, however its direct implications for cardiovascular disease development, progression and as a therapeutic target are not completely understood. More and more attention is given to the bidirectional interaction between psychological and physical health in relation to cardiovascular disease. With atherosclerosis being a chronic disease starting already at an early age the contribution of adverse early life events in affecting adult health risk behavior, health status and disease development is receiving increased attention. In addition, experimental research into the biological pathways involved in stress-induced cardiovascular complications show important roles for metabolic and immunologic maladaptation, resulting in increased disease development and progression. Here we provide a concise overview of human and experimental animal data linking chronic and acute stress to CVD risk and increased progression of the underlying disease atherosclerosis.
Collapse
Affiliation(s)
- H Maxime Lagraauw
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
361
|
Porras AM, Shanmuganayagam D, Meudt JJ, Krueger CG, Hacker TA, Rahko PS, Reed JD, Masters KS. Development of Aortic Valve Disease in Familial Hypercholesterolemic Swine: Implications for Elucidating Disease Etiology. J Am Heart Assoc 2015; 4:e002254. [PMID: 26508741 PMCID: PMC4845146 DOI: 10.1161/jaha.115.002254] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background Familial hypercholesterolemia (FH) is a prevalent hereditary disease associated with increased atherosclerosis and calcific aortic valve disease (CAVD). However, in both FH and non‐FH individuals, the role of hypercholesterolemia in the development of CAVD is poorly understood. This study used Rapacz FH (RFH) swine, an established model of human FH, to investigate the role of hypercholesterolemia alone in the initiation and progression of CAVD. The valves of RFH swine have not previously been examined. Methods and Results Aortic valve leaflets were isolated from wild‐type (0.25‐ and 1‐year‐old) and RFH (0.25‐, 1‐, 2‐, and 3‐year‐old) swine. Adult RFH animals exhibited numerous hallmarks of early CAVD. Significant leaflet thickening was found in adult RFH swine, accompanied by extensive extracellular matrix remodeling, including proteoglycan enrichment, collagen disorganization, and elastin fragmentation. Increased lipid oxidation and infiltration of macrophages were also evident in adult RFH swine. Intracardiac echocardiography revealed mild aortic valve sclerosis in some of the adult RFH animals, but unimpaired valve function. Microarray analysis of valves from adult versus juvenile RFH animals revealed significant upregulation of inflammation‐related genes, as well as several commonalities with atherosclerosis and overlap with human CAVD. Conclusions Adult RFH swine exhibited several hallmarks of early human CAVD, suggesting potential for these animals to help elucidate CAVD etiology in both FH and non‐FH individuals. The development of advanced atherosclerotic lesions, but only early‐stage CAVD, in RFH swine supports the hypothesis of an initial shared disease process, with additional stimulation necessary for further progression of CAVD.
Collapse
Affiliation(s)
- Ana M. Porras
- Department of Biomedical EngineeringUniversity of Wisconsin–MadisonMadisonWI
| | | | - Jennifer J. Meudt
- Department of Animal SciencesUniversity of Wisconsin–MadisonMadisonWI
| | | | - Timothy A. Hacker
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of Wisconsin–MadisonMadisonWI
| | - Peter S. Rahko
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of Wisconsin–MadisonMadisonWI
| | - Jess D. Reed
- Department of Animal SciencesUniversity of Wisconsin–MadisonMadisonWI
| | - Kristyn S. Masters
- Department of Biomedical EngineeringUniversity of Wisconsin–MadisonMadisonWI
| |
Collapse
|
362
|
Shim J, Al-Mashhadi RH, Sørensen CB, Bentzon JF. Large animal models of atherosclerosis - new tools for persistent problems in cardiovascular medicine. J Pathol 2015; 238:257-66. [DOI: 10.1002/path.4646] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 11/06/2022]
Affiliation(s)
- J Shim
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - RH Al-Mashhadi
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - CB Sørensen
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
| | - JF Bentzon
- Department of Clinical Medicine; Aarhus University, and Department of Cardiology, Aarhus University Hospital; Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III; Madrid Spain
| |
Collapse
|
363
|
Apolipoprotein E-deficient rats develop atherosclerotic plaques in partially ligated carotid arteries. Atherosclerosis 2015; 243:589-92. [PMID: 26545012 DOI: 10.1016/j.atherosclerosis.2015.10.093] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND The goal of this study was to establish an apolipoprotein E-deficient (ApoE(-/-)) rat model. METHODS The ApoE(-/-) rat was created by TALEN-mediated gene targeting in the genetic background of Sprague Dawley rat. Six-to eight-week-old male rats were used in the experiments (n = 10 in each group). RESULTS After fed with high-cholesterol diet (HCD) for 12 weeks, the ApoE(-/-) rats displayed typical dyslipidemia. In contrast, HCD failed to induce hypercholesterolemia in wild-type rats. However, there was no obvious atherosclerotic lesion in oil red O-stained en face aortas and the aortic root sections in both genetic types of rats. Interestingly, partial ligation caused the formation of plaques consist of lipid and macrophages in carotid arteries from ApoE(-/-) rats, but induced the neointimal hyperplasia in wild-type rats. Additionally, we found that HCD slightly increased the expression of adhesion molecules, while partial ligation dramatically upregulated these molecules. CONCLUSIONS The ApoE(-/-) rat is a novel model for dyslipidemia and could also be used in the research of atherosclerosis.
Collapse
|
364
|
Prueitt RL, Cohen JM, Goodman JE. Evaluation of atherosclerosis as a potential mode of action for cardiovascular effects of particulate matter. Regul Toxicol Pharmacol 2015; 73:S1-15. [PMID: 26474868 DOI: 10.1016/j.yrtph.2015.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/30/2015] [Indexed: 01/10/2023]
Abstract
Epidemiology studies have consistently reported associations between PM2.5 exposure and cardiovascular (CV) morbidity and mortality, but the epidemiology evidence for associations between PM2.5 and subclinical measures of atherosclerosis is unclear. We critically reviewed the experimental studies of PM2.5 and effects associated with acceleration and exacerbation of atherosclerosis and evaluated whether they support a biologically plausible, human-relevant mode of action (MoA) for the associations between PM2.5 exposure and adverse CV outcomes reported in epidemiology studies. We focused on outcomes related to atherosclerotic plaque development, thrombosis, and coagulation, and we examined whether these outcomes were correlated with measures of oxidative stress and systemic or pulmonary inflammation, to evaluate whether these processes are likely to be key early events for atherogenic effects of PM. While the current experimental evidence indicates that the acceleration and exacerbation of atherosclerosis is a biologically plausible MoA in experimental animal models, we found that the human relevance of the key events in the proposed MoA is unclear and not well supported by the existing data. Further studies are needed to fill several important data gaps before the human relevance of this MoA can be established.
Collapse
Affiliation(s)
- Robyn L Prueitt
- Gradient, 600 Stewart Street, Suite 1900, Seattle, WA, 98101, USA.
| | - Joel M Cohen
- Gradient, 600 Stewart Street, Suite 1900, Seattle, WA, 98101, USA.
| | - Julie E Goodman
- Gradient, 20 University Road, Suite 5, Cambridge, MA, 02138, USA.
| |
Collapse
|
365
|
|
366
|
Liu S, Bennett DC, Tun HM, Kim JE, Cheng KM, Zhang H, Leung FC. The effect of diet and host genotype on ceca microbiota of Japanese quail fed a cholesterol enriched diet. Front Microbiol 2015; 6:1092. [PMID: 26500632 PMCID: PMC4595795 DOI: 10.3389/fmicb.2015.01092] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/22/2015] [Indexed: 11/23/2022] Open
Abstract
Two Japanese quail strains, respectively atherosclerosis-susceptible (SUS) and –resistant (RES), have been shown to be good models to study cholesterol metabolism and transportation associated with atherosclerosis. Our objective was to examine possible difference in cecal microbiota between these strains when fed a control diet and a cholesterol enriched diet, to determine how host genotype and diet could affect the cecal microbiome that may play a part in cholesterol metabolism. A factorial study with both strains and two diets (control, cholesterol) was carried out. Cecal content was collected from 12 week old quail that have been on their respective diets for 6 weeks. DNA was extracted from the samples and the variable region 3–5 of the bacterial 16S rRNA gene was amplified. The amplicon libraries were subjected to pyrosequencing. Principal Component Analysis (PCA) of β-diversity showed four distinct microbiota communities that can be assigned to the 4 treatment groups (RES/control, RES/cholesterol, SUS/control, SUS/cholesterol). At the Phylum level, the 4 treatment groups has distinct Firmicutes community characteristics but no significant difference in Bacteroidetes. Eubacterium dolichum was rare in RES/control but became overabundant in RES/cholesterol. An unclassified species of Lactobacillaceae was found in abundance in SUS/control but the same species was rare in RES/cholesterol. On the other hand, two Lactobacillus species were only found in RES/control and an unclassified Lachnospiraceae species was abundant in RES/cholesterol but rare in SUS/control. The abundance of four species of Lachnospiraceae, three species of Ruminococcaceae and one species of Coprobacillaceae was positively correlated with plasma Total Cholesterol, plasma LDL, and LDL/HDL ratio. Our study of cecal microbiota in these quail has demonstrated that selection for susceptibility/resistance to diet induced atherosclerosis has also affected the quail's cecal environment to host distinctly different cecal microbiome.
Collapse
Affiliation(s)
- Shasha Liu
- The State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences Beijing, China ; Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Darin C Bennett
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Hein M Tun
- School of Biological Sciences, The University of Hong Kong Hong Kong, Hong Kong
| | - Ji-Eun Kim
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Kimberly M Cheng
- Faculty of Land and Food Systems, Avian Research Centre, The University of British Columbia Vancouver, BC, Canada
| | - Hongfu Zhang
- The State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences Beijing, China
| | - Frederick C Leung
- School of Biological Sciences, The University of Hong Kong Hong Kong, Hong Kong
| |
Collapse
|
367
|
Abstract
PURPOSE OF REVIEW Macrophage foam cells are important cells in the vascular wall that contribute to the inflammation associated with atherosclerotic lesions. Recent studies have demonstrated the heterogeneity of macrophages in lesions. In this review, advances in our understanding of the formation of foam cells by macrophage subsets in atherosclerotic plaques will be discussed. RECENT FINDINGS Macrophage subsets develop in response to the microenvironment in the arterial wall. The uptake of lipoproteins, particularly oxidized LDL, has been considered the major mechanism of foam cell formation. However, native and aggregated LDL can also be taken up by macrophages and M2 macrophages have been shown to be efficient in the uptake of apoptotic cells that can contribute lipids to the cells. The ability of the macrophage subsets to respond to bioactive lipids in the artery wall to either promote macrophage subset polarization and/or to promote foam cell formation is only beginning to be understood. SUMMARY Although we are beginning to appreciate the heterogeneity of macrophages present in atherosclerotic plaques, further work is required to fully understand the molecular basis for the differential ability of macrophage subsets to form foam cells and to respond to bioactive lipids.
Collapse
Affiliation(s)
- Godfrey S Getz
- The University of Chicago, Department of Pathology, Chicago, Illinois, USA
| | | |
Collapse
|
368
|
Hsu CC, Paik J, Brabb TL, O'Brien KD, Kim J, Sullivan BG, Hudkins KL, Seamons A, Finley JC, Meeker SM, Maggio-Price L. Murine Norovirus Infection Variably Alters Atherosclerosis in Mice Lacking Apolipoprotein E. Comp Med 2015; 65:369-381. [PMID: 26473341 PMCID: PMC4617328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/05/2015] [Accepted: 04/21/2015] [Indexed: 06/05/2023]
Abstract
Macrophages play a key role in the development of atherosclerosis. Murine noroviruses (MNV) are highly prevalent in research mouse colonies and infect macrophages and dendritic cells. Our laboratory found that MNV4 infection in mice lacking the LDL receptor alters the development of atherosclerosis, potentially confounding research outcomes. Therefore, we investigated whether MNV4 likewise altered atherosclerosis in ApoE(-/-) mice. In the presence of oxidized LDL, MNV4 infection of ApoE(-/-) bone marrow-derived macrophages increased the gene expression of the inflammatory markers inducible nitric oxide synthase, monocyte chemoattractant protein 1, and IL6. In addition, proteins involved in cholesterol transport were altered in MNV4-infected ApoE -/- bone marrow-derived macrophages and consisted of increased CD36 and decreased ATP-binding cassette transporter A1. MNV4 infection of ApoE(-/-) mice at 12 wk of age (during the development of atherosclerosis) had a variable effect on atherosclerotic lesion size. In one study, MNV4 significantly increased atherosclerotic plaque area whereas in a second study, no effect was observed. Compared with controls, MNV4-infected mice had higher circulating Ly6C-positive monocytes, and viral RNA was detected in the aortas of some mice, suggesting potential mechanisms by which MNV4 alters disease progression. Plaque size did not differ when ApoE -/- mice were infected at 4 wk of age (early during disease development) or in ApoE -/- mice maintained on a high-fat, high-cholesterol diet. Therefore, these data show that MNV4 has the potential to exert a variable and unpredictable effect on atherosclerosis in ApoE(-/-) mice. We therefore propose that performing experiments in MNV-free mouse colonies is warranted.
Collapse
Affiliation(s)
- Charlie C Hsu
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA.
| | - Jisun Paik
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Thea L Brabb
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kevin D O'Brien
- Department of Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jinkyu Kim
- Department of Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Brittany G Sullivan
- Department of Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kelly L Hudkins
- Department of Pathology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Audrey Seamons
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jennifer C Finley
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Stacey M Meeker
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lillian Maggio-Price
- Department of Comparative Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
369
|
Parton A, McGilligan V, O’Kane M, Baldrick FR, Watterson S. Computational modelling of atherosclerosis. Brief Bioinform 2015; 17:562-75. [DOI: 10.1093/bib/bbv081] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Indexed: 12/24/2022] Open
|
370
|
Wei Y, Chen F, Zhang T, Chen D, Jia X, Wang J, Guo W, Chen J. A Tubing-Free Microfluidic Wound Healing Assay Enabling the Quantification of Vascular Smooth Muscle Cell Migration. Sci Rep 2015; 5:14049. [PMID: 26365412 PMCID: PMC4568460 DOI: 10.1038/srep14049] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/17/2015] [Indexed: 01/09/2023] Open
Abstract
This paper presents a tubing-free microfluidic wound healing assay to quantify the migration of vascular smooth muscle cells (VSMCs), where gravity was used to generate a laminar flow within microfluidic channels, enabling cell seeding, culture, and wound generation. As the first systemic study to quantify the migration of VSMCs within microfluidic environments, the effects of channel geometries, surface modifications and chemokines on cellular migration were investigated, revealing that 1) height of the micro channels had a significant impact on cell migration; 2) the surface coating of collagen induced more migration of VSMCs than fibronectin coated surfaces and 3) platelet derived growth factor resulted in maximal cell migration compared to tumor necrosis factor alpha and fetal bovine serum. Furthermore, migrations of five types of VSMCs (e.g., the human vascular smooth muscle cell line, two types of primary vascular smooth cells, and VSMCs isolated from two human samples) were quantified, finding that VSMCs from the cell line and human samples demonstrated comparable migration distances, which were significantly lower than the migration distances of two primary cell types. As a platform technology, this wound healing assay may function as a new model to study migration of VSMCs within microfluidic environments.
Collapse
Affiliation(s)
- Yuanchen Wei
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, P.R. China, 100190
| | - Feng Chen
- Department of Vascular Surgery, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing, P.R. China, 100853
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People’s Hospital, Beijing, P.R. China, 100044
| | - Deyong Chen
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, P.R. China, 100190
| | - Xin Jia
- Department of Vascular Surgery, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing, P.R. China, 100853
| | - Junbo Wang
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, P.R. China, 100190
| | - Wei Guo
- Department of Vascular Surgery, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing, P.R. China, 100853
| | - Jian Chen
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, P.R. China, 100190
| |
Collapse
|
371
|
Kivelä AM, Huusko J, Ylä-Herttuala S. Prospect and progress of gene therapy in treating atherosclerosis. Expert Opin Biol Ther 2015; 15:1699-712. [PMID: 26328616 DOI: 10.1517/14712598.2015.1084282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Despite considerable improvements in therapies, atherosclerotic cardiovascular diseases remain the leading cause of death worldwide. Therefore, in addition to current treatment options, new therapeutic approaches are still needed. AREAS COVERED In this review, novel gene and RNA interference-based therapy approaches and promising target genes for treating atherosclerosis are addressed. In addition, relevant animal models for the demonstration of the efficacy of different gene therapy applications, and current progress toward more efficient, targeted and safer gene transfer vectors are reviewed. EXPERT OPINION Atherosclerosis represents a complex multifactorial disease that is dependent on the interplay between lipoprotein metabolism, cellular reactions and inflammation. Recent advances and novel targets, especially in the field of RNA interference-based therapies, are very promising. However, it should be noted that the modulation of a particular gene is not as clearly associated with a complex polygenic disease as it is in the case of monogenic diseases. A deeper understanding of molecular mechanisms of atherosclerosis, further progress in vector development and the demonstration of treatment efficacy in relevant animal models will be required before gene therapy of atherosclerosis meets its clinical reality.
Collapse
Affiliation(s)
- Annukka M Kivelä
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Jenni Huusko
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Seppo Ylä-Herttuala
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ; .,b 2 Science Service Center , Kuopio, Finland.,c 3 Kuopio University Hospital, Gene Therapy Unit , Kuopio, Finland
| |
Collapse
|
372
|
Lee SM, Nguyen D, Hu Z, Abbott GW. Kcne2 deletion promotes atherosclerosis and diet-dependent sudden death. J Mol Cell Cardiol 2015; 87:148-51. [PMID: 26307149 DOI: 10.1016/j.yjmcc.2015.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 08/12/2015] [Accepted: 08/12/2015] [Indexed: 02/05/2023]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. An estimated half of cases involve genetic predisposition. Sequence variants in human KCNE2, which encodes a cardiac and epithelial K(+) channel β subunit, cause inherited cardiac arrhythmias. Unexpectedly, human KCNE2 polymorphisms also associate with predisposition to atherosclerosis, with unestablished causality or mechanisms. Here, we report that germline Kcne2 deletion promotes atherosclerosis in mice, overcoming the relative resistance of this species to plaque deposition. In female western diet-fed mice, Kcne2 deletion increased plaque deposition >6-fold and also caused premature ventricular complexes and sudden death. The data establish causality for the first example of ion channel-linked atherosclerosis, and demonstrate that the severity of Kcne2-linked cardiac arrhythmias is strongly diet-dependent.
Collapse
Affiliation(s)
- Soo Min Lee
- Bioelectricity Laboratory, Dept. of Pharmacology, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Dara Nguyen
- Bioelectricity Laboratory, Dept. of Pharmacology, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Zhaoyang Hu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Pharmacology, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
373
|
Abstract
Rationale:
The role of circulating antibodies in addition to traditional cardiovascular risk factors in the development of accelerated arteriosclerosis and their long-term clinical consequences have not been demonstrated.
Objective:
We investigated the role of circulating antibodies in accelerated arteriosclerosis and the role of immune-associated arteriosclerosis in graft and patient survival and the occurrence of major adverse cardiovascular events.
Methods and Results:
This was an observational prospective cohort study that included 1065 kidney transplant patients (principal cohort, n=744; validation cohort, n=321) between 2004 and 2010. Participants were assessed for traditional cardiovascular risk factors and circulating anti–human leukocyte antigen (HLA) antibodies. All patients underwent allograft biopsies to assess arteriosclerotic lesions and endothelial activation, endarteritis, and complement deposition. In the principal cohort, 250 (33.6%) patients had severe arteriosclerosis (luminal narrowing >25% via fibrointimal arterial thickening). Circulating donor-specific anti-HLA antibodies were significantly associated with severe allograft arteriosclerosis (hazard ratio, 2.9;
P
<0.0001), independently of traditional risk factors. Patients with severe arteriosclerosis and anti-HLA antibodies (n=91, 12.2%) demonstrated allograft endothelial activation, endarteritis, and complement deposition. High levels of anti-HLA antibodies and their complement binding capacity were associated with increased severity of arteriosclerosis. Patients with antibody-associated severe arteriosclerosis had decreased allograft survival and increased mortality (
P
<0.0001); they exhibited a 2.5- and 4.1-fold increased risk of major adverse cardiovascular events compared with patients who had severe arteriosclerosis without antibodies and patients with minimal arteriosclerosis, respectively (
P
<0.0005). Circulating donor-specific anti-HLA antibodies were significantly associated with occurrence of major adverse cardiovascular events (hazard ratio, 2.4;
P
=0.0004), independently of traditional risk factors.
Conclusions:
Circulating antibodies are major determinants of severe arteriosclerosis and major adverse cardiovascular events, independent of traditional cardiovascular risk factors.
Collapse
|
374
|
Laakkonen JP, Ylä-Herttuala S. Recent Advancements in Cardiovascular Gene Therapy and Vascular Biology. Hum Gene Ther 2015; 26:518-24. [DOI: 10.1089/hum.2015.095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Johanna P. Laakkonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, University of Eastern Finland, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
375
|
Hypercholesterolemia Induced Immune Response and Inflammation on Progression of Atherosclerosis in Apob(tm2Sgy) Ldlr(tm1Her)/J Mice. Lipids 2015; 50:785-97. [PMID: 26178198 DOI: 10.1007/s11745-015-4046-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 06/22/2015] [Indexed: 12/20/2022]
Abstract
The effect of hypercholesterolemia induced immune response and inflammation on progression of atherosclerosis in ApoB(tm25gy) LDLr(tm1Her) mice, expressing only ApoB100 and deficient in the low density lipoprotein (LDL) receptor, thus closely resembling human cholesterol transport is not well defined. Atherosclerosis was induced by a high cholesterol diet and its progression was studied at 8, 14 and 20 weeks. Antibody response was determined by ELISA. Lymphocytes in spleen and aortic expression of inflammatory markers were studied by flow cytometry, and immunohistochemistry respectively. A rapid increase in plasma LDL levels in the first 8 weeks was followed by the exponential development of atherosclerosis between 8 and 14 weeks. Progression of the disease was accompanied by an accumulation of macrophages and increased expression of IL17 and IFN-γ in the aorta. Hypercholesterolemia resulted in increased immune response to modified lipids and aortic inflammation, with an expansion of Th17 cells in the spleen. Progression of atherosclerosis showed a positive correlation (r = 0.84, P < 0.001) with Th17 cells and a negative correlation with Treg cells (r = 0.83, P < 0.001). IgM antibodies to Ox-LDL and Th17 cells in spleen showed greatest association with disease development. Our results suggest that anti Ox-LDL IgM antibodies, Th17 cells could be developed as a potential marker to study disease progression and to study the effect of therapeutic regulation of inflammation.
Collapse
|
376
|
Ultra-high-resolution 3D imaging of atherosclerosis in mice with synchrotron differential phase contrast: a proof of concept study. Sci Rep 2015; 5:11980. [PMID: 26165698 PMCID: PMC4499839 DOI: 10.1038/srep11980] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/12/2015] [Indexed: 11/13/2022] Open
Abstract
The goal of this study was to investigate the performance of 3D synchrotron differential phase contrast (DPC) imaging for the visualization of both macroscopic and microscopic aspects of atherosclerosis in the mouse vasculature ex vivo. The hearts and aortas of 2 atherosclerotic and 2 wild-type control mice were scanned with DPC imaging with an isotropic resolution of 15 μm. The coronary artery vessel walls were segmented in the DPC datasets to assess their thickness, and histological staining was performed at the level of atherosclerotic plaques. The DPC imaging allowed for the visualization of complex structures such as the coronary arteries and their branches, the thin fibrous cap of atherosclerotic plaques as well as the chordae tendineae. The coronary vessel wall thickness ranged from 37.4 ± 5.6 μm in proximal coronary arteries to 13.6 ± 3.3 μm in distal branches. No consistent differences in coronary vessel wall thickness were detected between the wild-type and atherosclerotic hearts in this proof-of-concept study, although the standard deviation in the atherosclerotic mice was higher in most segments, consistent with the observation of occasional focal vessel wall thickening. Overall, DPC imaging of the cardiovascular system of the mice allowed for a simultaneous detailed 3D morphological assessment of both large structures and microscopic details.
Collapse
|
377
|
Priyadharsini RP. Animal models to evaluate anti-atherosclerotic drugs. Fundam Clin Pharmacol 2015; 29:329-40. [PMID: 26095240 DOI: 10.1111/fcp.12130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 06/17/2015] [Accepted: 06/05/2015] [Indexed: 01/08/2023]
Abstract
Atherosclerosis is a multifactorial condition characterized by endothelial injury, fatty streak deposition, and stiffening of the blood vessels. The pathogenesis is complex and mediated by adhesion molecules, inflammatory cells, and smooth muscle cells. Statins have been the major drugs in treating hypercholesterolemia for the past two decades despite little efficacy. There is an urgent need for new drugs that can replace statins or combined with statins. The preclinical studies evaluating atherosclerosis require an ideal animal model which resembles the disease condition, but there is no single animal model which mimics the disease. The animal models used are rabbits, rats, mice, hamsters, mini pigs, etc. Each animal model has its own advantages and disadvantages. The method of induction of atherosclerosis includes diet, chemical induction, mechanically induced injuries, and genetically manipulated animal models. This review mainly focuses on the various animal models, method of induction, the advantages, disadvantages, and the current perspectives with regard to preclinical studies on atherosclerosis.
Collapse
|
378
|
Link JC, Chen X, Prien C, Borja MS, Hammerson B, Oda MN, Arnold AP, Reue K. Increased high-density lipoprotein cholesterol levels in mice with XX versus XY sex chromosomes. Arterioscler Thromb Vasc Biol 2015; 35:1778-86. [PMID: 26112012 DOI: 10.1161/atvbaha.115.305460] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/08/2015] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The molecular mechanisms underlying sex differences in dyslipidemia are poorly understood. We aimed to distinguish genetic and hormonal regulators of sex differences in plasma lipid levels. APPROACH AND RESULTS We assessed the role of gonadal hormones and sex chromosome complement on lipid levels using the four core genotypes mouse model (XX females, XX males, XY females, and XY males). In gonadally intact mice fed a chow diet, lipid levels were influenced by both male-female gonadal sex and XX-XY chromosome complement. Gonadectomy of adult mice revealed that the male-female differences are dependent on acute effects of gonadal hormones. In both intact and gonadectomized animals, XX mice had higher HDL cholesterol (HDL-C) levels than XY mice, regardless of male-female sex. Feeding a cholesterol-enriched diet produced distinct patterns of sex differences in lipid levels compared with a chow diet, revealing the interaction of gonadal and chromosomal sex with diet. Notably, under all dietary and gonadal conditions, HDL-C levels were higher in mice with 2 X chromosomes compared with mice with an X and Y chromosome. By generating mice with XX, XY, and XXY chromosome complements, we determined that the presence of 2 X chromosomes, and not the absence of the Y chromosome, influences HDL-C concentration. CONCLUSIONS We demonstrate that having 2 X chromosomes versus an X and Y chromosome complement drives sex differences in HDL-C. It is conceivable that increased expression of genes escaping X-inactivation in XX mice regulates downstream processes to establish sexual dimorphism in plasma lipid levels.
Collapse
Affiliation(s)
- Jenny C Link
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Xuqi Chen
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Christopher Prien
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Mark S Borja
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Bradley Hammerson
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Michael N Oda
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Arthur P Arnold
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.)
| | - Karen Reue
- From the Molecular Biology Institute (J.C.L., K.R.), Department of Integrative Biology and Physiology (X.C., A.P.A.), Laboratory of Neuroendocrinology of the Brain Research Institute (X.C., A.P.A.), Department of Human Genetics, David Geffen School of Medicine (C.P., K.R.), and Department of Medicine, David Geffen School of Medicine (K.R.), University of California, Los Angeles; and Children's Hospital Oakland Research Institute, California (M.S.B., B.H., M.N.O.).
| |
Collapse
|
379
|
Szostak J, Ansari S, Madan S, Fluck J, Talikka M, Iskandar A, De Leon H, Hofmann-Apitius M, Peitsch MC, Hoeng J. Construction of biological networks from unstructured information based on a semi-automated curation workflow. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015. [PMID: 26200752 PMCID: PMC5630939 DOI: 10.1093/database/bav057] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Capture and representation of scientific knowledge in a structured format are essential to improve the understanding of biological mechanisms involved in complex diseases. Biological knowledge and knowledge about standardized terminologies are difficult to capture from literature in a usable form. A semi-automated knowledge extraction workflow is presented that was developed to allow users to extract causal and correlative relationships from scientific literature and to transcribe them into the computable and human readable Biological Expression Language (BEL). The workflow combines state-of-the-art linguistic tools for recognition of various entities and extraction of knowledge from literature sources. Unlike most other approaches, the workflow outputs the results to a curation interface for manual curation and converts them into BEL documents that can be compiled to form biological networks. We developed a new semi-automated knowledge extraction workflow that was designed to capture and organize scientific knowledge and reduce the required curation skills and effort for this task. The workflow was used to build a network that represents the cellular and molecular mechanisms implicated in atherosclerotic plaque destabilization in an apolipoprotein-E-deficient (ApoE(-/-)) mouse model. The network was generated using knowledge extracted from the primary literature. The resultant atherosclerotic plaque destabilization network contains 304 nodes and 743 edges supported by 33 PubMed referenced articles. A comparison between the semi-automated and conventional curation processes showed similar results, but significantly reduced curation effort for the semi-automated process. Creating structured knowledge from unstructured text is an important step for the mechanistic interpretation and reusability of knowledge. Our new semi-automated knowledge extraction workflow reduced the curation skills and effort required to capture and organize scientific knowledge. The atherosclerotic plaque destabilization network that was generated is a causal network model for vascular disease demonstrating the usefulness of the workflow for knowledge extraction and construction of mechanistically meaningful biological networks.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Sam Ansari
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Sumit Madan
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, Sankt Augustin, Germany
| | - Juliane Fluck
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, Sankt Augustin, Germany
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Anita Iskandar
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Hector De Leon
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, Sankt Augustin, Germany
| | - Manuel C Peitsch
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland and
| |
Collapse
|
380
|
Development of Antiatherosclerotic Drugs on the basis of Natural Products Using Cell Model Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:463797. [PMID: 26347804 PMCID: PMC4549547 DOI: 10.1155/2015/463797] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/07/2015] [Accepted: 01/14/2015] [Indexed: 11/18/2022]
Abstract
Atherosclerosis including its subclinical form is one of the key medical and social problems. At present, there is no therapy available for widespread use against subclinical atherosclerosis. The use of synthetic drugs for the prevention of arteriosclerosis in its early stages is not sufficient because of the limited indications for severe side effects and high cost of treatment. Obviously, effective antiatherosclerotic drugs based on natural products would be a preferred alternative. Simple cell-based models for testing different natural products have been developed and the ability of natural products to prevent intracellular lipid accumulation in primary cell culture was evaluated. This approach utilizing cell models allowed to test effects of such direct antiatherosclerotic therapy, analyzing the effects mimicking those which can occur "at the level" of arterial wall via the inhibition of intracellular lipid deposition. The data from the carried out clinical trials support a point of view that the identification of antiatherosclerotic activity of natural products might offer a great opportunity for the prevention and treatment of atherosclerotic disease, reducing cardiovascular morbidity and mortality.
Collapse
|
381
|
Gordon SM, Li H, Zhu X, Shah AS, Lu LJ, Davidson WS. A comparison of the mouse and human lipoproteome: suitability of the mouse model for studies of human lipoproteins. J Proteome Res 2015; 14:2686-95. [PMID: 25894274 PMCID: PMC4712022 DOI: 10.1021/acs.jproteome.5b00213] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Plasma levels of low density lipoproteins (LDL) and high density lipoproteins (HDL) exhibit opposing associations with cardiovascular disease in human populations and mouse models have been heavily used to derive a mechanistic understanding of these relationships. In humans, recent mass spectrometry studies have revealed that the plasma lipoproteome is significantly more complex than originally appreciated. This is particularly true for HDL which contains some 90 distinct proteins, a majority of which play functional roles that go beyond those expected for simple lipid transport. Unfortunately, the mouse lipoproteome remains largely uncharacterized-a significant gap given the heavy reliance on the model. Using a gel filtration chromatography and mass spectrometry analysis that targets phospholipid-bound plasma proteins, we compared the mouse lipoproteome and its size distribution to a previous, identical human analysis. We identified 113 lipid associated proteins in the mouse. In general, the protein diversity in the LDL and HDL size ranges was similar in mice versus humans, though some distinct differences were noted. For the majority of proteins, the size distributions, that is, whether a given protein was associated with large versus small HDL particles, for example, were also similar between species. Again, however, there were clear differences exhibited by a minority of proteins that may reflect metabolic differences between species. Finally, by correlating the lipid and protein size profiles, we identified five proteins that closely track with the major HDL protein, apolipoprotein A-I across both species. Thus, mice have most of the minor proteins identified in human lipoproteins that play key roles in inflammation, innate immunity, proteolysis and its inhibition, and vitamin transport. This provides support for the continued use of the mouse as a model for many aspects of human lipoprotein metabolism.
Collapse
Affiliation(s)
- Scott M. Gordon
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, Ohio 45237-0507, United States
| | - Hailong Li
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - Amy S. Shah
- Department of Pediatrics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7012, Cincinnati, Ohio 45229-3039, United States
| | - L. Jason Lu
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation, 3333 Burnet Avenue, MLC 7024, Cincinnati, Ohio 45229-3039, United States
| | - W. Sean Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, 2120 East Galbraith Road, Cincinnati, Ohio 45237-0507, United States
| |
Collapse
|
382
|
Hovell CM, Sei YJ, Kim Y. Microengineered vascular systems for drug development. JOURNAL OF LABORATORY AUTOMATION 2015; 20:251-8. [PMID: 25424383 PMCID: PMC5663643 DOI: 10.1177/2211068214560767] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Indexed: 11/15/2022]
Abstract
Recent advances in microfabrication technologies and advanced biomaterials have allowed for the development of in vitro platforms that recapitulate more physiologically relevant cellular components and function. Microengineered vascular systems are of particular importance for the efficient assessment of drug candidates to physiological barriers lining microvessels. This review highlights advances in the development of microengineered vascular structures with an emphasis on the potential impact on drug delivery studies. Specifically, this article examines the development of models for the study of drug delivery to the central nervous system and cardiovascular system. We also discuss current challenges and future prospects of the development of microengineered vascular systems.
Collapse
Affiliation(s)
- Candice M Hovell
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA
| | - Yoshitaka J Sei
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA
| | - YongTae Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
383
|
Abstract
Atherosclerosis is a progressive disease characterized by the accumulation of lipids in medium to large sized arteries. Atherothrombosis is a term used to describe formation of a thrombus after rupture of an atherosclerotic plaque. Thrombosis can lead to myocardial infarction and stroke. Risk factors for atherosclerosis include hyperlipidemia, diabetes, smoking and hypertension all of which increase tissue factor (TF) expression. High levels of TF are present in atherosclerotic plaques due to expression by macrophages and vascular smooth muscle cells and the presence of cell-derived TF-positive microvesicles (MVs). In addition, hyperlipidemia leads to the formation of oxidized LDL, which induces TF expression in circulating monocytes and the release of TF-positive MVs. The major source of TF that drives thrombosis after plaque rupture is TF within the plaque. However, TF in the blood on monocytes and MVs may also contribute the thrombosis. Inhibition of the TF/factor VIIa complex is unlikely to be an effective strategy to reduce atherothrombosis due the essential role of the complex in hemostasis. However, selective blockade of pathologic TF without affecting protective TF may be effective in reducing atherothrombosis. For instance, statins have been shown to reduce TF expression in the plaque and in circulating monocytes, which would be expected to reduce thrombosis. Further studies are needed to determine safe strategies to reduce pathologic TF expression and atherothrombosis.
Collapse
Affiliation(s)
- Kohei Tatsumi
- Department of Medicine, Division of Hematology and Oncology, McAllister Heart Institute, Thrombosis and Hemostasis Group,University of North Carolina at Chapel Hill
| | | |
Collapse
|
384
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 327] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
385
|
Langbein H, Hofmann A, Brunssen C, Goettsch W, Morawietz H. Impact of high-fat diet and voluntary running on body weight and endothelial function in LDL receptor knockout mice. ATHEROSCLEROSIS SUPP 2015; 18:59-66. [DOI: 10.1016/j.atherosclerosissup.2015.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
386
|
Buckley ML, Ramji DP. The influence of dysfunctional signaling and lipid homeostasis in mediating the inflammatory responses during atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1498-510. [PMID: 25887161 DOI: 10.1016/j.bbadis.2015.04.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/25/2015] [Accepted: 04/08/2015] [Indexed: 12/14/2022]
Abstract
Atherosclerosis, the underlying cause of myocardial infarction and thrombotic cerebrovascular events, is responsible for the majority of deaths in westernized societies. Mortality from this disease is also increasing at a marked rate in developing countries due to the acquisition of a westernized lifestyle accompanied with elevated rates of obesity and diabetes. Atherosclerosis is recognized as a chronic inflammatory disorder associated with lipid accumulation and the development of fibrotic plaques within the walls of medium and large arteries. A range of immune cells, such as macrophages and T-lymphocytes, through the action of various cytokines, such as interleukins-1 and -33, transforming growth factor-β and interferon-γ, orchestrates the inflammatory response in this disease. The disease is also characterized by marked dysfunction in lipid homeostasis and signaling pathways that control the inflammatory response. This review will discuss the molecular basis of atherosclerosis with particular emphasis on the roles of the immune cells and cytokines along with the dysfunctional lipid homeostasis and cell signaling associated with this disease.
Collapse
Affiliation(s)
- Melanie L Buckley
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK.
| |
Collapse
|
387
|
Reschen ME, Gaulton KJ, Lin D, Soilleux EJ, Morris AJ, Smyth SS, O'Callaghan CA. Lipid-induced epigenomic changes in human macrophages identify a coronary artery disease-associated variant that regulates PPAP2B Expression through Altered C/EBP-beta binding. PLoS Genet 2015; 11:e1005061. [PMID: 25835000 PMCID: PMC4383549 DOI: 10.1371/journal.pgen.1005061] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 02/09/2015] [Indexed: 01/17/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified over 40 loci that affect risk of coronary artery disease (CAD) and the causal mechanisms at the majority of loci are unknown. Recent studies have suggested that many causal GWAS variants influence disease through altered transcriptional regulation in disease-relevant cell types. We explored changes in transcriptional regulation during a key pathophysiological event in CAD, the environmental lipid-induced transformation of macrophages to lipid-laden foam cells. We used a combination of open chromatin mapping with formaldehyde-assisted isolation of regulatory elements (FAIRE-seq) and enhancer and transcription factor mapping using chromatin immuno-precipitation (ChIP-seq) in primary human macrophages before and after exposure to atherogenic oxidized low-density lipoprotein (oxLDL), with resultant foam cell formation. OxLDL-induced foam cell formation was associated with changes in a subset of open chromatin and active enhancer sites that strongly correlated with expression changes of nearby genes. OxLDL-regulated enhancers were enriched for several transcription factors including C/EBP-beta, which has no previously documented role in foam cell formation. OxLDL exposure up-regulated C/EBP-beta expression and increased genomic binding events, most prominently around genes involved in inflammatory response pathways. Variants at CAD-associated loci were significantly and specifically enriched in the subset of chromatin sites altered by oxLDL exposure, including rs72664324 in an oxLDL-induced enhancer at the PPAP2B locus. OxLDL increased C/EBP beta binding to this site and C/EBP beta binding and enhancer activity were stronger with the protective A allele of rs72664324. In addition, expression of the PPAP2B protein product LPP3 was present in foam cells in human atherosclerotic plaques and oxLDL exposure up-regulated LPP3 in macrophages resulting in increased degradation of pro-inflammatory mediators. Our results demonstrate a genetic mechanism contributing to CAD risk at the PPAP2B locus and highlight the value of studying epigenetic changes in disease processes involving pathogenic environmental stimuli. Coronary artery disease is a complex disease where over 40 genomic loci contributing to genetic risk have been identified. However, identifying the precise variants, genomic elements and genes that mediate this risk at each locus has proved challenging. We hypothesized that some genetic risk variants may influence a key step in development of coronary artery disease, which occurs when macrophages encounter environmentally-derived lipid. These cells take up lipid and accumulate in atherosclerotic plaques in the walls of blood vessels where they contribute to the inflammatory atherosclerotic disease process. Therefore, we studied the effects of this lipid exposure on the genomic activity of these cells. Environmental lipid exposure triggered changes in transcriptional regulation and gene expression. Variants at coronary artery disease risk loci were enriched for genomic regions altered by lipid exposure. We studied one such risk variant rs72664324 in detail and found that it altered binding of the C/EBP-beta transcription factor and altered expression of the PPAP2B gene. PPAP2B encodes an enzyme that degrades pro-inflammatory substances. Our study demonstrates a hitherto unknown genetic mechanism underlying atherosclerotic heart disease and demonstrates the value of studying changes in transcriptional regulation in key disease processes involving environmental influences.
Collapse
Affiliation(s)
- Michael E. Reschen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kyle J. Gaulton
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Da Lin
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Elizabeth J. Soilleux
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford and Department of Cellular Pathology, John Radcliffe Hospital, Oxford, United Kingdom
| | - Andrew J. Morris
- Division of Cardiovascular Medicine, The Gill Heart Institute, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Veterans Affairs Medical Center, Lexington, Kentucky, United States of America
| | - Susan S. Smyth
- Division of Cardiovascular Medicine, The Gill Heart Institute, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Veterans Affairs Medical Center, Lexington, Kentucky, United States of America
| | | |
Collapse
|
388
|
Robinet P, Smith JD. Development and Use of Mouse Models of Atherosclerosis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
389
|
Abstract
All aspects of the pathogenesis of atherosclerosis are critically influenced by the inflammatory response in vascular plaques. Research in the field of innate immunity from the past 2 decades has uncovered many novel mechanisms elucidating how immune cells sense microbes, tissue damage, and metabolic derangements. Here, we summarize which triggers of innate immunity appear during atherogenesis and by which pathways they can contribute to inflammation in atherosclerotic plaques. The increased understanding gained from studies assessing how immune activation is associated with the pathogenesis of atherosclerosis has provided many novel targets for potential therapeutic intervention. Excitingly, the concept that inflammation may be the core of cardiovascular disease is currently being clinically evaluated and will probably encourage further studies in this area.
Collapse
Affiliation(s)
- Sebastian Zimmer
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Alena Grebe
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Eicke Latz
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.).
| |
Collapse
|
390
|
Mechanisms of action for arsenic in cardiovascular toxicity and implications for risk assessment. Toxicology 2015; 331:78-99. [PMID: 25771173 DOI: 10.1016/j.tox.2015.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/09/2015] [Accepted: 02/27/2015] [Indexed: 11/20/2022]
Abstract
The possibility of an association between inorganic arsenic (iAs) exposure and cardiovascular outcomes has received increasing attention in the literature over the past decade. The United States Environmental Protection Agency (US EPA) is currently revising its Integrated Risk Assessment System (IRIS) review of iAs, and one of the non-cancer endpoints of interest is cardiovascular disease (CVD). Despite the increased interest in this area, substantial gaps remain in the available information, particularly regarding the mechanism of action (MOA) by which iAs could cause or exacerbate CVD. Few studies specifically address the plausibility of an association between iAs and CVD at the low exposure levels which are typical in the United States (i.e., below 100 μg As/L in drinking water). We have conducted a review and evaluation of the animal, mechanistic, and human data relevant to the potential MOAs of iAs and CVD. Specifically, we evaluated the most common proposed MOAs, which include disturbance of endothelial function and hepatic dysfunction. Our analysis of the available evidence indicates that there is not a well-established MOA for iAs in the development or progression of CVD. Few human studies of the potential MOAs have addressed plausibility at low doses and the applicability of extrapolation from animal studies to humans is questionable. However, the available evidence indicates that regardless of the specific MOA, the effects of iAs on physiological processes at the cellular level appear to operate via a threshold mechanism. This finding is consistent with the lack of association of CVD with iAs exposure in humans at levels below 100 μg/L, particularly when considering important exposure and risk modifiers such as nutrition and genetics. Based on this analysis, we conclude that there are no data supporting a linear dose-response relationship between iAs and CVD, indicating this relationship has a threshold.
Collapse
|
391
|
Ammirati E, Moroni F, Magnoni M, Camici PG. The role of T and B cells in human atherosclerosis and atherothrombosis. Clin Exp Immunol 2015; 179:173-87. [PMID: 25352024 DOI: 10.1111/cei.12477] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 01/05/2023] Open
Abstract
Far from being merely a passive cholesterol accumulation within the arterial wall, the development of atherosclerosis is currently known to imply both inflammation and immune effector mechanisms. Adaptive immunity has been implicated in the process of disease initiation and progression interwined with traditional cardiovascular risk factors. Although the body of knowledge regarding the correlation between atherosclerosis and immunity in humans is growing rapidly, a relevant proportion of it derives from studies carried out in animal models of cardiovascular disease (CVD). However, while the mouse is a well-suited model, the results obtained therein are not fully transferrable to the human setting due to intrinsic genomic and environmental differences. In the present review, we will discuss mainly human findings, obtained either by examination of post-mortem and surgical atherosclerotic material or through the analysis of the immunological profile of peripheral blood cells. In particular, we will discuss the findings supporting a pro-atherogenic role of T cell subsets, such as effector memory T cells or the potential protective function of regulatory T cells. Recent studies suggest that traditional T cell-driven B2 cell responses appear to be atherogenic, while innate B1 cells appear to exert a protective action through the secretion of naturally occurring antibodies. The insights into the immune pathogenesis of atherosclerosis can provide new targets in the quest for novel therapeutic targets to abate CVD morbidity and mortality.
Collapse
Affiliation(s)
- E Ammirati
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy; Cardiovascular and Thoracic Department, AO Niguarda Ca' Granda, Milan, Italy
| | | | | | | |
Collapse
|
392
|
Vindis C. Autophagy: an emerging therapeutic target in vascular diseases. Br J Pharmacol 2015; 172:2167-78. [PMID: 25537552 DOI: 10.1111/bph.13052] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/27/2014] [Accepted: 12/07/2014] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a cellular catabolic process responsible for the destruction of long-lived proteins and organelles via lysosome-dependent pathway. This process is of great importance in maintaining cellular homeostasis, and deregulated autophagy has been implicated in the pathogenesis of a wide range of diseases. A growing body of evidence suggests that autophagy can be activated in vascular disorders such as atherosclerosis. Autophagy occurs under basal conditions and mediates homeostatic functions in cells but in the setting of pathological states up-regulated autophagy can exert both protective and detrimental functions. Therefore, the precise role of autophagy and its relationship with the progression of the disease need to be clarified. This review highlights recent findings regarding autophagy activity in vascular cells and its potential contribution to vascular disorders with a focus on atherogenesis. Finally, whether the manipulation of autophagy represents a new therapeutic approach to treat or prevent vascular diseases is also discussed.
Collapse
Affiliation(s)
- Cécile Vindis
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse III, Toulouse, France
| |
Collapse
|
393
|
Daugherty A, Tabas I, Rader DJ. Accelerating the pace of atherosclerosis research. Arterioscler Thromb Vasc Biol 2015; 35:11-2. [PMID: 25520521 DOI: 10.1161/atvbaha.114.304833] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Departments of Medicine and Genetics, Institute for Translational Medicine and Therapeutics, and Cardiovascular Institute, Perelman School of Medicine of the University of Pennsylvania, Philadelphia (D.J.R.).
| | - Ira Tabas
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Departments of Medicine and Genetics, Institute for Translational Medicine and Therapeutics, and Cardiovascular Institute, Perelman School of Medicine of the University of Pennsylvania, Philadelphia (D.J.R.)
| | - Daniel J Rader
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Departments of Medicine and Genetics, Institute for Translational Medicine and Therapeutics, and Cardiovascular Institute, Perelman School of Medicine of the University of Pennsylvania, Philadelphia (D.J.R.)
| |
Collapse
|
394
|
Henne KR, Ason B, Howard M, Wang W, Sun J, Higbee J, Tang J, Matsuda KC, Xu R, Zhou L, Chan JCY, King C, Piper DE, Ketchem RR, Michaels ML, Jackson SM, Retter MW. Anti-PCSK9 antibody pharmacokinetics and low-density lipoprotein-cholesterol pharmacodynamics in nonhuman primates are antigen affinity-dependent and exhibit limited sensitivity to neonatal Fc receptor-binding enhancement. J Pharmacol Exp Ther 2015; 353:119-31. [PMID: 25653417 DOI: 10.1124/jpet.114.221242] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as an attractive therapeutic target for cardiovascular disease. Monoclonal antibodies (mAbs) that bind PCSK9 and prevent PCSK9:low-density lipoprotein receptor complex formation reduce serum low-density lipoprotein-cholesterol (LDL-C) in vivo. PCSK9-mediated lysosomal degradation of bound mAb, however, dramatically reduces mAb exposure and limits duration of effect. Administration of high-affinity mAb1:PCSK9 complex (1:2) to mice resulted in significantly lower mAb1 exposure compared with mAb1 dosed alone in normal mice or in PCSK9 knockout mice lacking antigen. To identify mAb-binding characteristics that minimize lysosomal disposition, the pharmacokinetic behavior of four mAbs representing a diverse range of PCSK9-binding affinities at neutral (serum) and acidic (endosomal) pH was evaluated in cynomolgus monkeys. Results revealed an inverse correlation between affinity and both mAb exposure and duration of LDL-C lowering. High-affinity mAb1 exhibited the lowest exposure and shortest duration of action (6 days), whereas mAb2 displayed prolonged exposure and LDL-C reduction (51 days) as a consequence of lower affinity and pH-sensitive PCSK9 binding. mAbs with shorter endosomal PCSK9:mAb complex dissociation half-lives (<20 seconds) produced optimal exposure-response profiles. Interestingly, incorporation of previously reported Fc-region amino acid substitutions or novel loop-insertion peptides that enhance in vitro neonatal Fc receptor binding, led to only modest pharmacokinetic improvements for mAbs with pH-dependent PCSK9 binding, with only limited augmentation of pharmacodynamic activity relative to native mAbs. A pivotal role for PCSK9 in mAb clearance was demonstrated, more broadly suggesting that therapeutic mAb-binding characteristics require optimization based on target pharmacology.
Collapse
Affiliation(s)
- Kirk R Henne
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Brandon Ason
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Monique Howard
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Wei Wang
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jeonghoon Sun
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jared Higbee
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jie Tang
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Katherine C Matsuda
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Ren Xu
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Lei Zhou
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Joyce C Y Chan
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Chadwick King
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Derek E Piper
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Randal R Ketchem
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Mark Leo Michaels
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Simon M Jackson
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Marc W Retter
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| |
Collapse
|
395
|
Fan J, Kitajima S, Watanabe T, Xu J, Zhang J, Liu E, Chen YE. Rabbit models for the study of human atherosclerosis: from pathophysiological mechanisms to translational medicine. Pharmacol Ther 2015; 146:104-19. [PMID: 25277507 PMCID: PMC4304984 DOI: 10.1016/j.pharmthera.2014.09.009] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 01/09/2023]
Abstract
Laboratory animal models play an important role in the study of human diseases. Using appropriate animals is critical not only for basic research but also for the development of therapeutics and diagnostic tools. Rabbits are widely used for the study of human atherosclerosis. Because rabbits have a unique feature of lipoprotein metabolism (like humans but unlike rodents) and are sensitive to a cholesterol diet, rabbit models have not only provided many insights into the pathogenesis and development of human atherosclerosis but also made a great contribution to translational research. In fact, rabbit was the first animal model used for studying human atherosclerosis, more than a century ago. Currently, three types of rabbit model are commonly used for the study of human atherosclerosis and lipid metabolism: (1) cholesterol-fed rabbits, (2) Watanabe heritable hyperlipidemic rabbits, analogous to human familial hypercholesterolemia due to genetic deficiency of LDL receptors, and (3) genetically modified (transgenic and knock-out) rabbits. Despite their importance, compared with the mouse, the most widely used laboratory animal model nowadays, the use of rabbit models is still limited. In this review, we focus on the features of rabbit lipoprotein metabolism and pathology of atherosclerotic lesions that make it the optimal model for human atherosclerotic disease, especially for the translational medicine. For the sake of clarity, the review is not an attempt to be completely inclusive, but instead attempts to summarize substantial information concisely and provide a guideline for experiments using rabbits.
Collapse
Affiliation(s)
- Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Shimokato 1110, Chuo-City 409-3898, Japan.
| | - Shuji Kitajima
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Teruo Watanabe
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease and Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|
396
|
Gjurich BN, Taghavie-Moghadam PL, Galkina EV. Flow Cytometric Analysis of Immune Cells Within Murine Aorta. Methods Mol Biol 2015; 1339:161-175. [PMID: 26445788 PMCID: PMC4638413 DOI: 10.1007/978-1-4939-2929-0_11] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The immune system plays a critical role in the modulation of atherogenesis at all stages of the disease. However, there are many technical difficulties when studying the immune system within murine aortas. Common techniques such as PCR and immunohistochemistry have answered many questions about the presence of immune cells and mediators of inflammation within the aorta yet many questions remain unanswered due to the limitations of these techniques. On the other hand, cumulatively the flow cytometry approach has propelled the immunology field forward but it has been challenging to apply this technique to aortic tissues. Here, we describe the methodology to isolate and characterize the immune cells within the murine aorta and provide examples of functional assays for aortic leukocytes using flow cytometry. The method involves the harvesting and enzymatic digestion of the aorta, extracellular and intracellular protein staining, and a subsequent flow cytometric analysis.
Collapse
Affiliation(s)
- Breanne N Gjurich
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA, 23507-1696, USA
| | - Parésa L Taghavie-Moghadam
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA, 23507-1696, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA, 23507-1696, USA.
| |
Collapse
|
397
|
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
|
398
|
Zhang X, Ha S, Wei W, Duan S, Shi Y, Yang Y. Noninvasive imaging of aortic atherosclerosis by ultrasound biomicroscopy in a mouse model. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2015; 34:111-116. [PMID: 25542946 DOI: 10.7863/ultra.34.1.111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
OBJECTIVES The noninvasive and accurate evaluation of vessel characteristics in mouse models has become an intensive focus of vascular medicine. This study aimed to apply ultrasound biomicroscopy to evaluate aortic atherosclerotic progression in a low-density lipoprotein receptor (LDL-R) knockout mouse model of atherosclerosis. METHODS Ten male LDL-R(-/-)C57BL/6 mice aged 16 and 24 weeks and 8 male wild-type C57BL/6 mice aged 16 and 24 weeks were used as experimental and control groups, respectively. Ultrasound biomicroscopy was applied to detect the morphologic characteristics of the aortic root, ascending aorta, aortic arch, and carotid artery and to measure the aortic root intima-media thickness and carotid artery bifurcation. RESULTS Ultrasound biomicroscopy showed a significant increase in the aortic root intima-media thickness from 0.10 ± 0.03 mm in 16-week-old mice to 0.16 ± 0.04 mm in 24-week-old mice (P < .01). The ultrasound biomicroscopically measured intima-media thickness was highly correlated with the histologic measurement (r = 0.81). CONCLUSIONS Ultrasound biomicroscopy could be used for a noninvasive, accurate, and dynamic analysis of aortic atherosclerosis in LDL-R knockout mice.
Collapse
Affiliation(s)
- Xiaoshan Zhang
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.)
| | - Si Ha
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.)
| | - Wei Wei
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.)
| | - Shasha Duan
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.)
| | - Yilu Shi
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.)
| | - Ya Yang
- Department of Ultrasound, Affiliated Hospital of Innermongolia Medical University, Huhhot, China (X.Z., S.H., W.W., S.D., Y.S.); and Department of Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.Y.).
| |
Collapse
|
399
|
Rivera-Torres J. Analysis of Gene and Protein Expression in Atherosclerotic Mouse Aorta by Western Blot and Quantitative Real-Time PCR. Methods Mol Biol 2015; 1339:309-22. [PMID: 26445798 DOI: 10.1007/978-1-4939-2929-0_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Atherosclerosis involves changes in gene and protein expression patterns in affected arteries. Quantification of these alterations is essential for understanding the molecular mechanisms underlying this pathology. Western blot and real-time PCR-used to quantify protein and messenger RNA levels, respectively-are invaluable molecular biology tools, particularly when material is limited. The availability of many genetically modified mouse models of atherosclerosis makes the mouse aorta an ideal tissue in which to carry out these expression pattern analyses. In this chapter, protocols are presented for mRNA and protein extraction from mouse aorta and for the accurate quantification of mRNA expression by RT-PCR and of proteins by western blot.
Collapse
Affiliation(s)
- José Rivera-Torres
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Vascular Biology Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
400
|
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
|