1
|
Vaisar T, Hu JH, Airhart N, Fox K, Heinecke J, Nicosia RF, Kohler T, Potter ZE, Simon GM, Dix MM, Cravatt BF, Gharib SA, Dichek DA. Parallel Murine and Human Plaque Proteomics Reveals Pathways of Plaque Rupture. Circ Res 2020; 127:997-1022. [PMID: 32762496 PMCID: PMC7508285 DOI: 10.1161/circresaha.120.317295] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE Plaque rupture is the proximate cause of most myocardial infarctions and many strokes. However, the molecular mechanisms that precipitate plaque rupture are unknown. OBJECTIVE By applying proteomic and bioinformatic approaches in mouse models of protease-induced plaque rupture and in ruptured human plaques, we aimed to illuminate biochemical pathways through which proteolysis causes plaque rupture and identify substrates that are cleaved in ruptured plaques. METHODS AND RESULTS We performed shotgun proteomics analyses of aortas of transgenic mice with macrophage-specific overexpression of urokinase (SR-uPA+/0 mice) and of SR-uPA+/0 bone marrow transplant recipients, and we used bioinformatic tools to evaluate protein abundance and functional category enrichment in these aortas. In parallel, we performed shotgun proteomics and bioinformatics studies on extracts of ruptured and stable areas of freshly harvested human carotid plaques. We also applied a separate protein-analysis method (protein topography and migration analysis platform) to attempt to identify substrates and proteolytic fragments in mouse and human plaque extracts. Approximately 10% of extracted aortic proteins were reproducibly altered in SR-uPA+/0 aortas. Proteases, inflammatory signaling molecules, as well as proteins involved with cell adhesion, the cytoskeleton, and apoptosis, were increased. ECM (Extracellular matrix) proteins, including basement-membrane proteins, were decreased. Approximately 40% of proteins were altered in ruptured versus stable areas of human carotid plaques, including many of the same functional categories that were altered in SR-uPA+/0 aortas. Collagens were minimally altered in SR-uPA+/0 aortas and ruptured human plaques; however, several basement-membrane proteins were reduced in both SR-uPA+/0 aortas and ruptured human plaques. Protein topography and migration analysis platform did not detect robust increases in proteolytic fragments of ECM proteins in either setting. CONCLUSIONS Parallel studies of SR-uPA+/0 mouse aortas and human plaques identify mechanisms that connect proteolysis with plaque rupture, including inflammation, basement-membrane protein loss, and apoptosis. Basement-membrane protein loss is a prominent feature of ruptured human plaques, suggesting a major role for basement-membrane proteins in maintaining plaque stability.
Collapse
Affiliation(s)
- Tomáš Vaisar
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Jie H Hu
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Nathan Airhart
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Kate Fox
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Jay Heinecke
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Roberto F Nicosia
- Departments of Pathology and Laboratory Medicine (D.A.D., R.F.N.), University of Washington, Seattle.,Departments of Pathology and Laboratory Medicine (R.F.N.), VA Puget Sound Health Care System, Seattle, WA
| | - Ted Kohler
- Departments of Surgery (T.K.), University of Washington, Seattle.,Departments of Surgery (T.K.), VA Puget Sound Health Care System, Seattle, WA
| | - Zachary E Potter
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | | | - Melissa M Dix
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | - Sina A Gharib
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - David A Dichek
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle.,Departments of Pathology and Laboratory Medicine (D.A.D., R.F.N.), University of Washington, Seattle
| |
Collapse
|
2
|
Liu X, Ma J, Ma L, Liu F, Zhang C, Zhang Y, Ni M. Overexpression of tissue factor induced atherothrombosis in apolipoprotein E-/- mice via both enhanced plaque thrombogenicity and plaque instability. J Mol Cell Cardiol 2018; 127:1-10. [PMID: 30500376 DOI: 10.1016/j.yjmcc.2018.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/16/2018] [Accepted: 11/26/2018] [Indexed: 01/07/2023]
Abstract
The mechanisms leading to atherothrombosis from "vulnerable plaque" are more complex than initially proposed. We aimed to clarify whether plaque thrombogenicity is critical in atherothrombosis in mice. In a murine model of plaque destabilization, we enhanced plaque thrombogenicity by systemically overexpressing murine tissue factor (TF) by adenovirus-mediated gene transfer. The potential effects and mechanisms of TF on plaque destabilization were examined in cultured human aortic smooth muscle cells (HASMCs), RAW264.7 cells and human umbilical vein endothelial cells (HUVECs). To elucidate the TF noncoagulant effects on plaque destabilization, TF-overexpressed mice were treated with the protease-activated receptor 2 (PAR-2) antagonist ENMD-1068. In TF-overexpressing apolipoprotein (E)-deficient (ApoE-/-) mice, 67% (8 of 12) of carotid plaques exhibited plaque disruption and atherothrombosis. Moreover, 58% (7 of 12) showed plaque hemorrhage, including 1 due to plaque disruption, 4 neovascularization and 2 both. In contrast, only 17% (2 of 12) of control mice showed atherothrombosis, both with plaque hemorrhage but no neovascularization. On PCR, TF overexpression increased the expression of inflammatory factors. In cultured cells, the TF-FVIIa complex enhanced the expression of inflammatory factors and a vicious cycle of inflammation. Also, TF-FVIIa complex induced intra-plaque angiogenesis via PAR-2. ENMD-1068 treatment significantly inhibited the expression of inflammatory factors and neovascularization, and the incidence of intra-plaque hemorrhage decreased in TF-overexpressing mice. In conclusions, TF overexpression enhanced plaque thrombogenicity, which played a pivotal role in atherothrombosis in ApoE-/- mice. In addition, TF promoted plaque instability by activating inflammatory and proangiogenic effects via TF-FVIIa/PAR-2 signaling.
Collapse
Affiliation(s)
- Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lianyue Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fangfang Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Characterization of Atherosclerosis Formation in a Murine Model of Type IIa Human Familial Hypercholesterolemia. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1878964. [PMID: 29977908 PMCID: PMC6011105 DOI: 10.1155/2018/1878964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/27/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
A murine genetic model of LDL-cholesterol- (LDL-C-) driven atherosclerosis, based on complete deficiencies of both the LDL-receptor (Ldlr-/-) and key catalytic component of an apolipoprotein B-edisome complex (Apobec1-/-), which converts apoB-100 to apoB-48, has been extensively characterized. These gene deficiencies allow high levels of apoB-100 to be present and inefficiently cleared, thus leading to very high levels of LDL-C in mice on a normal diet. Many key features of atherosclerotic plaques observed in human familial hypercholesterolemia are found in these mice as they are allowed to age through 72 weeks. The general characteristics include the presence of high levels of LDL-C in plasma and macrophage-related fatty streak formation in the aortic tree, which progressively worsens with age. More specifically, plaque found in the aortic sinuses contains a lipid core with relatively high numbers of macrophages and a smooth muscle cell α-actin- and collagen-containing cap, which thins with age. These critical features of plaque progression suggest that the Ldlr-/-/Apobec1-/- mouse line presents a superior model of LDL-C-driven atherosclerosis.
Collapse
|
4
|
Daemen MJ, Gijsen FJH, Heiden KVD, Hoogendoorn A. Animal models for plaque rupture: a biomechanical assessment. Thromb Haemost 2018; 115:501-8. [DOI: 10.1160/th15-07-0614] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/22/2015] [Indexed: 11/05/2022]
Abstract
SummaryRupture of atherosclerotic plaques is the main cause of acute cardiovascular events. Animal models of plaque rupture are rare but essential for testing new imaging modalities to enable diagnosis of the patient at risk. Moreover, they enable the design of new treatment strategies to prevent plaque rupture. Several animal models for the study of atherosclerosis are available. Plaque rupture in these models only occurs following severe surgical or pharmaceutical intervention. In the process of plaque rupture, composition, biology and mechanics each play a role, but the latter has been disregarded in many animal studies. The biomechanical environment for atherosclerotic plaques is comprised of two parts, the pressure-induced stress distribution, mainly - but not exclusively – influenced by plaque composition, and the strength distribution throughout the plaque, largely determined by the inflammatory state. This environment differs considerably between humans and most animals, resulting in suboptimal conditions for plaque rupture. In this review we describe the role of the biomechanical environment in plaque rupture and assess this environment in animal models that present with plaque rupture.
Collapse
|
5
|
Daugherty A, Tall AR, Daemen MJ, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Circ Res 2017; 121:e53-e79. [DOI: 10.1161/res.0000000000000169] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
6
|
Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2017; 37:e131-e157. [PMID: 28729366 DOI: 10.1161/atv.0000000000000062] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
7
|
Reduced Necrosis and Content of Apoptotic M1 Macrophages in Advanced Atherosclerotic Plaques of Mice With Macrophage-Specific Loss of Trpc3. Sci Rep 2017; 7:42526. [PMID: 28186192 PMCID: PMC5301208 DOI: 10.1038/srep42526] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/09/2017] [Indexed: 01/08/2023] Open
Abstract
In previous work we reported that ApoeKO mice transplanted with bone marrow cells deficient in the Transient Receptor Potential Canonical 3 (TRPC3) channel have reduced necrosis and number of apoptotic macrophages in advanced atherosclerotic plaques. Also, in vitro studies with polarized macrophages derived from mice with macrophage-specific loss of TRPC3 showed that M1, but not M2 macrophages, deficient in Trpc3 are less susceptible to ER stress-induced apoptosis than Trpc3 expressing cells. The questions remained (a) whether the plaque phenotype in transplanted mice resulted from a genuine effect of Trpc3 on macrophages, and (b) whether the reduced necrosis and macrophage apoptosis in plaques of these mice was a manifestation of the selective effect of TRPC3 on apoptosis of M1 macrophages previously observed in vitro. Here, we addressed these questions using Ldlr knockout (Ldlr−/−) mice with macrophage-specific loss of Trpc3 (MacTrpc3−/−/Ldlr−/− → Ldlr−/−). Compared to controls, we observed decreased plaque necrosis and number of apoptotic macrophages in MacTrpc3−/−/Ldlr−/− → Ldlr−/− mice. Immunohistochemical analysis revealed a reduction in apoptotic M1, but not apoptotic M2 macrophages. These findings confirm an effect of TRPC3 on plaque necrosis and support the notion that this is likely a reflection of the reduced susceptibility of Trpc3-deficient M1 macrophages to apoptosis.
Collapse
|
8
|
Daeichin V, Sluimer JC, van der Heiden K, Skachkov I, Kooiman K, Janssen A, Janssen B, Bosch JG, de Jong N, Daemen MJAP, van der Steen AFW. Live Observation of Atherosclerotic Plaque Disruption in Apolipoprotein E-Deficient Mouse. Ultrasound Int Open 2016; 1:E67-71. [PMID: 27689156 DOI: 10.1055/s-0035-1565092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
AIM The actual occurrence of spontaneous plaque rupture in mice has been a matter of debate. We report on an in vivo observation of the actual event of possible plaque disruption in a living ApoE(-/-) mouse. METHODS AND RESULTS During live contrast-enhanced ultrasonography of a 50-week-old ApoE(-/-) male mouse, symptoms suggesting plaque disruption in the brachiocephalic artery were observed. Histological analysis confirmed the presence of advanced atherosclerotic lesions with dissections and intraplaque hemorrhage in the affected brachiocephalic trunk, pointing towards plaque rupture as the cause of the observed event. However, we did not detect a luminal thrombus or cap rupture, which is a key criterion for plaque rupture in human atherosclerosis. CONCLUSION This study reports the real-time occurrence of a possible plaque rupture in a living ApoE(-/-) mouse.
Collapse
Affiliation(s)
- V Daeichin
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands
| | - J C Sluimer
- Department of Pathology, Maastricht University, CARIM, Maastricht, Netherlands
| | - K van der Heiden
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands
| | - I Skachkov
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands
| | - K Kooiman
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands
| | - A Janssen
- Department of Pathology, Maastricht University, CARIM, Maastricht, Netherlands
| | - B Janssen
- Pharmacology & Toxicology, Maastricht University Medical Center, Maastricht, Netherlands
| | - J G Bosch
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands
| | - N de Jong
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands; Lab of Acoustical Wavefield Imaging, Delft University of Technology, Delft, the Netherlands
| | - M J A P Daemen
- Pathology, Amsterdam Medical Center, Amsterdam, Netherlands
| | - A F W van der Steen
- Erasmus Medical Center, Thoraxcenter Biomedical Engineering, Rotterdam, Netherlands; Lab of Acoustical Wavefield Imaging, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
9
|
Geronimo FRB, Barter PJ, Rye KA, Heather AK, Shearston KD, Rodgers KJ. Plaque stabilizing effects of apolipoprotein A-IV. Atherosclerosis 2016; 251:39-46. [PMID: 27240254 DOI: 10.1016/j.atherosclerosis.2016.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 03/20/2016] [Accepted: 04/24/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS Apolipoprotein (apo) A-IV, the third most abundant HDL-associated protein, is atheroprotective and shares similar properties as apoA-I. We have reported previously that apoA-I, the most abundant apolipoprotein in HDL, inhibits plaque disruption in a mouse model. We aimed at examining the effects of apoA-IV on markers of plaque stability in vivo. METHODS Plaques within brachiocephalic arteries of 16-week old apoE-knockout C57BL/6 mice were examined for changes in composition after 10 weeks on a high-fat diet (HFD). The animals received twice-weekly injections of human lipid-free apoA-IV (1 mg/kg, n = 31) or PBS (n = 32) during the 9th and 10th weeks of the HFD. RESULTS In the apoA-IV treated mice, there were significantly fewer hemorrhagic plaque disruptions (9/31 vs. 18/32, p < 0.05), thicker fibrous caps, smaller lipid cores, a lower macrophage:SMC ratio, less MMP-9 protein, more collagen, and fewer proliferating cells. In the plaques of mice given apoA-IV, MCP-1, VCAM-1, and inducible NOS were also significantly lower. Based on the percentage of cleaved PARP-positive and TUNEL-positive plaque nuclei, apoA-IV reduced apoptosis. in HMDMs, apoA-IV reduced MMP-9 mRNA expression by half, doubled mRNA levels of TIMP1 and decreased MMP-9 activity. CONCLUSIONS ApoA-IV treatment is associated with a more stable plaque phenotype and a reduced incidence of acute disruptions in this mouse model.
Collapse
Affiliation(s)
| | - P J Barter
- School of Medical Sciences, University of New South Wales, Australia.
| | - K A Rye
- School of Medical Sciences, University of New South Wales, Australia.
| | - A K Heather
- The Heart Research Institute, Sydney, Australia; School of Medical Sciences, University of Otago, Dunedin, New Zealand.
| | - K D Shearston
- School of Dentistry, University of Western Australia, Australia.
| | - K J Rodgers
- School of Life Sciences, Faculty of Science, University of Technology, Sydney, Australia.
| |
Collapse
|
10
|
Targeting blood thrombogenicity precipitates atherothrombotic events in a mouse model of plaque destabilization. Sci Rep 2015; 5:10225. [PMID: 25959659 PMCID: PMC4426696 DOI: 10.1038/srep10225] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/07/2015] [Indexed: 11/09/2022] Open
Abstract
Although some features of plaque instability can be observed in genetically modified mouse models, atherothrombosis induction in mice has been attested to be difficult. We sought to test the hypothesis that alterations in blood thrombogenicity might have an essential role in the development of atherothrombosis in ApoE-/- mice. In a mouse model of plaque destabilization established in our laboratory, we targeted blood thrombogenicity by systemically overexpressing murine prothrombin via adenovirus-mediated gene transfer. Systemic overexpression of prothrombin increased blood thrombogenicity, and remarkably, precipitated atherothrombotic events in 70% of the animals. The affected plaques displayed features of culprit lesions as seen in human coronary arteries, including fibrous cap disruption, luminal thrombosis, and plaque hemorrhage. Treatment with aspirin and clopidogrel substantially reduced the incidence of atherothrombosis in this model. Mechanistically, increased inflammation, apoptosis and upregulation of metalloproteinases contributed to the development of plaque destabilization and atherothrombosis. As conclusions, targeting blood thrombogenicity in mice can faithfully reproduce the process of atherothrombosis as occurring in human coronary vessels. Our results suggest that blood-plaque interactions are critical in the development of atherothrombosis in mice, substantiating the argument that changes in blood coagulation status may have a determinant role in the onset of acute coronary syndrome.
Collapse
|
11
|
Newby AC. Metalloproteinases promote plaque rupture and myocardial infarction: A persuasive concept waiting for clinical translation. Matrix Biol 2015; 44-46:157-66. [PMID: 25636537 DOI: 10.1016/j.matbio.2015.01.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 02/05/2023]
Abstract
Atherosclerotic plaque rupture provokes most myocardial infarctions. Matrix metalloproteinases (MMPs) have counteracting roles in intimal thickening, which stabilizes plaques, on the one hand and extracellular matrix destruction that leads to plaque rupture on the other. This review briefly summarizes the key points supporting the involvement of individual MMPs in provoking plaque rupture and discusses the barriers that stand in the way of clinical translation, which can be itemised as follows: structural and functional complexity of the MMP family; lack of adequate preclinical models partly owing to different expression patterns of MMPs and TIMPs in mouse and human macrophages; the need to target individual MMPs selectively; the difficulties in establishing causality in human studies; and the requirement for surrogate markers of efficacy. Overcoming these barriers would open the way to new treatments that could have a major impact on cardiovascular mortality worldwide.
Collapse
Affiliation(s)
- Andrew C Newby
- University of Bristol, School of Clinical Sciences and Bristol Heart Institute, Bristol, UK.
| |
Collapse
|
12
|
Abstract
BACKGROUND Anxiety is a common experience among patients with acute coronary syndrome (ACS) that can have a negative impact on health outcomes. Nonetheless, the negative role of anxiety remains underappreciated, as reflected by clinicians' underrecognition and undertreatment of anxious hospitalized and nonhospitalized patients with ACS. Underappreciation of the role of anxiety is possibly related to inadequate understanding of the mechanisms whereby anxiety may adversely affect health outcomes. PURPOSE The aim of this study was to synthesize the evidence about potential mechanisms by which anxiety and adverse health outcomes are related. CONCLUSIONS A biobehavioral model links anxiety to the development of thrombogenic and arrhythmic events in patients with ACS. Biologically, anxiety may interfere with the immune system, lipid profile, automatic nervous system balance, and the coagulation cascade, whereas behaviorally, anxiety may adversely affect adoption of healthy habits and cardiac risk-reducing behaviors. The biological and behavioral pathways complement each other in the production of poor outcomes. CLINICAL IMPLICATIONS Anxiety requires more attention from clinical cardiology. The adverse impact of anxiety on health outcomes could be avoided by efficient assessment and treatment of anxiety.
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW To review progress over the past 5 years in relating extracellular proteinases to plaque rupture, the cause of most myocardial infarctions, and consider the most promising prospects for developing related treatments. RECENT FINDINGS Cysteinyl cathepsins have been implicated in multiple macrophage functions that could promote plaque rupture. Cathepsin K is an attractive target because it is a collagenase and selective inhibitors are already being used in phase III clinical trials. Several serine proteinases clearly influence vascular remodelling and atherogenesis but important, unrelated actions limit their value as therapeutic targets. Among the metalloproteinases, new evidence supports roles for A Disintigrin and Metalloproteinases (ADAMs), including ADAM-10, ADAM-17 and ADAM-33, which suggest that selective inhibitors might be effective treatments. For ADAMs with ThromboSpondin domains (ADAMTSs), there are biological and genome-wide association data linking ADAMTS-7 to incidence of coronary heart disease but not increased risk of myocardial infarctions. In the case of matrix metalloproteinases (MMPs), selective inhibitors of MMP-12 and MMP-13 are available and may be appropriate for development as therapies. Novel targets, including MMP-8, MMP-10, MMP-14, MMP-19, MMP-25 and MMP-28, are also being considered. SUMMARY New opportunities exist to exploit proteinases as therapeutic targets in plaque rupture.
Collapse
Affiliation(s)
- Andrew C Newby
- University of Bristol and Bristol Heart Institute, Bristol, UK
| |
Collapse
|
14
|
Riou LM, Broisat A, Ghezzi C, Finet G, Rioufol G, Gharib AM, Pettigrew RI, Ohayon J. Effects of mechanical properties and atherosclerotic artery size on biomechanical plaque disruption - mouse vs. human. J Biomech 2014; 47:765-72. [PMID: 24491495 DOI: 10.1016/j.jbiomech.2014.01.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2014] [Indexed: 12/16/2022]
Abstract
Mouse models of atherosclerosis are extensively being used to study the mechanisms of atherosclerotic plaque development and the results are frequently extrapolated to humans. However, major differences have been described between murine and human atherosclerotic lesions and the determination of similarities and differences between these species has been largely addressed recently. This study takes over and extends previous studies performed by our group and related to the biomechanical characterization of both mouse and human atherosclerotic lesions. Its main objective was to determine the distribution and amplitude of mechanical stresses including peak cap stress (PCS) in aortic vessels from atherosclerotic apoE(-/-) mice, in order to evaluate whether such biomechanical data would be in accordance with the previously suggested lack of plaque rupture in this model. Successful finite element analysis was performed from the zero-stress configuration of aortic arch sections and mainly indicated (1) the modest role of atherosclerotic lesions in the observed increase in residual parietal stresses in apoE(-/-) mouse vessels and (2) the low amplitude of murine PCS as compared to humans. Overall, the results from the present study support the hypothesis that murine biomechanical properties and artery size confer less propensity to rupture for mouse lesions in comparison with those of humans.
Collapse
Affiliation(s)
- Laurent M Riou
- INSERM, UMR_S 1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, Grenoble, France
| | - Alexis Broisat
- INSERM, UMR_S 1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, Grenoble, France
| | - Catherine Ghezzi
- INSERM, UMR_S 1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, Grenoble, France
| | - Gérard Finet
- Department of Hemodynamics and Interventional Cardiology, Hospices Civils de Lyon and Claude Bernard University Lyon1, INSERM Unit 886, Lyon, France
| | - Gilles Rioufol
- Department of Hemodynamics and Interventional Cardiology, Hospices Civils de Lyon and Claude Bernard University Lyon1, INSERM Unit 886, Lyon, France
| | - Ahmed M Gharib
- Laboratory of Integrative Cardiovascular Imaging Science, National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Roderic I Pettigrew
- Laboratory of Integrative Cardiovascular Imaging Science, National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Jacques Ohayon
- Laboratory TIMC-IMAG/DyCTiM, UJF, CNRS UMR 5525, In(3)S, Grenoble, France; Polytech Annecy-Chambéry, University of Savoie, Le Bourget du Lac, France.
| |
Collapse
|
15
|
Su L, Zhang Q, Bao H, Li W, Miao Y, Yan Z, Chen D. Effect of dalteparin on atherosclerotic lesion formation in apolipoprotein E-deficient mice. Clin Appl Thromb Hemost 2013; 21:266-72. [PMID: 23965336 DOI: 10.1177/1076029613499818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We aimed to investigate whether prolonged treatment with dalteparin could inhibit plaque progression. With C57BL/6J mice as the control, genetically deficient apolipoprotein E (apo E) male mice of C57BL/6J strain (apo E(-/-)) were randomly divided into 3 groups. The model group received no dalteparin, while the other 2 groups received dalteparin at 100 and 200 U/kg d, respectively. The aorta was harvested for hematoxylin and eosin staining to observe plaque formation and for immunohistochemical staining to detect the expression of oxidized low-density lipoprotein receptor 1 (LOX-1). The expression of LOX-1 messenger RNA was detected by reverse transcription polymerase chain reaction, while the expression of LOX-1 protein was detected by Western blotting. Dalteparin decreased aortic plaque volume and inhibited aortic LOX-1 protein expression in apo E(-/-) mice. The effect persisted 4 weeks after dalteparin treatment was discontinued. Dalteparin may inhibit atherosclerotic lesions by downregulating the expression of LOX-1 protein.
Collapse
Affiliation(s)
- Lin Su
- Department of Geriatrics, Peking University People's Hospital, Beijing, China
| | - Qingwen Zhang
- Department of Geriatrics, Peking University People's Hospital, Beijing, China
| | - Hui Bao
- Department of Geriatrics, Peking University People's Hospital, Beijing, China
| | - Wei Li
- Department of Geriatrics, Peking University People's Hospital, Beijing, China
| | - Yide Miao
- Department of Geriatrics, Peking University People's Hospital, Beijing, China
| | - Zheng Yan
- Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Dingbao Chen
- Department of Pathology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
16
|
Meng X, Li W, Yang J, Zhang K, Qin W, An G, Gao F, Wang Y, Zhang C, Zhang Y. Regulatory T cells prevent plaque disruption in apolipoprotein E-knockout mice. Int J Cardiol 2013; 168:2684-92. [PMID: 23566492 DOI: 10.1016/j.ijcard.2013.03.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 01/03/2013] [Accepted: 03/17/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND CD4(+)CD25(+) regulatory T cells (Tregs) have received considerable interest in atherogenesis. We hypothesized that Tregs treatment may dose-dependently stabilize atherosclerotic plaques by inhibiting inflammatory cytokine secretion and matrix metalloproteinases (MMPs) expression and enhancing P4Hα1 expression in atherosclerotic lesions. METHODS AND RESULTS We established a vulnerable carotid plaque model in apolipoprotein E- knockout mice (ApoE-/-). Mice were divided into control, phosphate buffered saline (PBS), small-dose Tregs, moderate-dose Tregs, large-dose Tregs and PC groups. Histopathological analysis showed that the plaque disruption rate was 50%, 50%, 43.8%, 12.5%, 12.5% and 43.8% in the control, PBS, small-dose Tregs, moderate-dose Tregs, large-dose Tregs and PC groups. Tregs treatment resulted in a significant decrease in the relative contents of macrophages and lipids and a substantial increase in those of SMCs and collagen in the carotid plaque, leading to an almost 50% reduction of plaque vulnerability index. Furthermore, Tregs treatment decreased the expression of proinflammatory cytokines, MMP-2 and MMP-9 but increased the expression of P4Hα1 both in vivo and in vitro. Most of these therapeutic effects of Tregs were found to be mediated by transforming growth factor and interleukin-10. CONCLUSION Adoptive transfer of Tregs dose-dependently changed plaque composition to a stable plaque phenotype and lowered the incidence of plaque disruption in ApoE-/- mice. The major mechanisms involved reduced expression of inflammatory cytokines and MMP-2 and MMP-9, and enhanced expression of P4Hα1 in the carotid plaque. Tregs may provide a novel approach to the treatment of vulnerable plaques.
Collapse
Affiliation(s)
- Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Najafi AH, Aghili N, Tilan JU, Andrews JA, Peng X, Lassance-Soares RM, Sood S, Alderman LO, Abe K, Li L, Kolodgie FD, Virmani R, Zukowska Z, Epstein SE, Burnett MS. A new murine model of stress-induced complex atherosclerotic lesions. Dis Model Mech 2013; 6:323-31. [PMID: 23324329 PMCID: PMC3597015 DOI: 10.1242/dmm.009977] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The primary purpose of this investigation was to determine whether ApoE−/− mice, when subjected to chronic stress, exhibit lesions characteristic of human vulnerable plaque and, if so, to determine the time course of such changes. We found that the lesions were remarkably similar to human vulnerable plaque, and that the time course of lesion progression raised interesting insights into the process of plaque development. Lard-fed mixed-background ApoE−/− mice exposed to chronic stress develop lesions with large necrotic core, thin fibrous cap and a high degree of inflammation. Neovascularization and intraplaque hemorrhage are observed in over 80% of stressed animals at 20 weeks of age. Previously described models report a prevalence of only 13% for neovascularization observed at a much later time point, between 36 and 60 weeks of age. Thus, our new stress-induced model of advanced atherosclerotic plaque provides an improvement over what is currently available. This model offers a tool to further investigate progression of plaque phenotype to a more vulnerable phenotype in humans. Our findings also suggest a possible use of this stress-induced model to determine whether therapeutic interventions have effects not only on plaque burden, but also, and importantly, on plaque vulnerability.
Collapse
Affiliation(s)
- Amir H Najafi
- Cardiovascular Research Institute, MedStar Health Research Institute, 108 Irving Street, NW Washington, DC 20010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gijsen F, van der Giessen A, van der Steen A, Wentzel J. Shear stress and advanced atherosclerosis in human coronary arteries. J Biomech 2013; 46:240-7. [DOI: 10.1016/j.jbiomech.2012.11.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 12/15/2022]
|
19
|
de Vries MR, Niessen HWM, Löwik CWGM, Hamming JF, Jukema JW, Quax PHA. Plaque rupture complications in murine atherosclerotic vein grafts can be prevented by TIMP-1 overexpression. PLoS One 2012; 7:e47134. [PMID: 23071737 PMCID: PMC3469549 DOI: 10.1371/journal.pone.0047134] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/11/2012] [Indexed: 01/17/2023] Open
Abstract
The current study describes the incidence and phenotype of plaque rupture complications in murine vein grafts. Since matrix metalloproteinases (MMPs) are highly involved in atherosclerotic plaque vulnerability and plaque rupture, we hypothesized that this model can be validated by overexpression of the MMP inhibitor TIMP-1. First we studied 47 vein grafts in hypercholesterolemic ApoE3*Leiden mice for the incidence of plaque complications. In 79% of these grafts, extensive lesions with plaque rupture complications like dissections, intraplaque hemorrhages or erosions with intramural thrombi were found. Next, in vivo Near-InfraRed-Fluorescence imaging demonstrated that electroporation mediated TIMP-1-overexpression reduced local MMP activity in vein grafts by 73% (p<0.01). This led to a 40% reduction in lesion-size after 28d (p = 0.01) and a more stable lesion phenotype with significant more smooth muscle cells (135%), collagen (47%) and significant less macrophages (44%) and fibrin (55%) than controls. More importantly, lesions in the TIMP-1 group showed a 90% reduction of plaque complications (10/18 of control mice showed plaque complications versus 1/18 in TIMP-1 treated mice). Murine vein grafts are a relevant spontaneous model to study plaque stability and subsequent hemorrhagic complications, resulting in plaque instability. Moreover, inhibition of MMPs by TIMP-1-overexpression resulted in decreased plaque progression, increased stabilization and decreased plaque rupture complications in murine vein grafts.
Collapse
Affiliation(s)
- Margreet R. de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans W. M. Niessen
- Department of Pathology and Cardiac Surgery, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Jaap F. Hamming
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J. Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H. A. Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Liu XL, Zhang PF, Ding SF, Wang Y, Zhang M, Zhao YX, Ni M, Zhang Y. Local gene silencing of monocyte chemoattractant protein-1 prevents vulnerable plaque disruption in apolipoprotein E-knockout mice. PLoS One 2012; 7:e33497. [PMID: 22428064 PMCID: PMC3299803 DOI: 10.1371/journal.pone.0033497] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/11/2012] [Indexed: 02/05/2023] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1), a CC chemokine (CCL2), has been demonstrated to play important roles in atherosclerosis and becoming an important therapeutic target for atherosclerosis. The present study was undertaken to test the hypothesis that local RNAi of MCP-1 by site-specific delivery of adenovirus-mediated small hairpin RNA (shRNA) may enhance plaque stability and prevent plaque disruption in ApoE-/- mice. We designed an adenovirus-mediated shRNA against mouse MCP-1 (rAd5-MCP-1-shRNA). Male apolipoprotein E-knockout (ApoE-/-) mice (n = 120) were fed a high-fat diet and vulnerable plaques were induced by perivascular placement of constrictive collars around the carotid artery, intraperitoneal injection of lipopolysaccharide and stress stimulation. Mice were randomly divided into RNA interference (Ad-MCP-1i) group receiving local treatment of rAd5-MCP-1-shRNA suspension, Ad-EGFP group receiving treatment of rAd5-mediated negative shRNA and mock group receiving treatment of saline. Two weeks after treatment, plaque disruption rates were significantly lower in the Ad-MCP-1i group than in the Ad-EGFP group (13.3% vs. 60.0%, P = 0.01), and local MCP-1 expression was significantly inhibited in the Ad-MCP-1i group confirmed by immunostaining, qRT-PCR and western blot (P<0.001). Compared with the Ad-EGFP group, carotid plaques in the Ad-MCP-1i group showed increased levels of collagen and smooth muscle cells, and decreased levels of lipid and macrophages. The expression of inflammatory cytokines and activities of matrix metalloproteinases (MMPs) were lower in the Ad-MCP-1i group than in the Ad-EGFP group. In conclusion, site-specific delivery of adenoviral-mediated shRNA targeting mouse MCP-1 downregulated MCP-1 expression, turned a vulnerable plaque into a more stable plaque phenotype and prevented plaque disruption. A marked suppression of the local inflammatory cytokine expression may be the central mechanism involved.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Xia Zhao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
- * E-mail: (YXZ); (MN)
| | - Mei Ni
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
- * E-mail: (YXZ); (MN)
| | | |
Collapse
|
21
|
Wen 文颂 S, Liu 柳东芳 DF, Liu 刘振 Z, Harris S, Yao 姚玉宇 YY, Ding 丁琪 Q, Nie 聂芳 F, Lu 卢瞳 T, Chen 陈华俊 HJ, An 安艳丽 YL, Zang 臧凤超 FC, Teng 滕皋军 GJ. OxLDL-targeted iron oxide nanoparticles for in vivo MRI detection of perivascular carotid collar induced atherosclerotic lesions in ApoE-deficient mice. J Lipid Res 2012; 53:829-838. [PMID: 22393161 PMCID: PMC3329382 DOI: 10.1194/jlr.m018895] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atherosclerotic disease is a leading cause of morbidity and mortality in developed countries, and oxidized LDL (OxLDL) plays a key role in the formation, rupture, and subsequent thrombus formation in atherosclerotic plaques. In the current study, anti-mouse OxLDL polyclonal antibody and nonspecific IgG antibody were conjugated to polyethylene glycol-coated ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles, and a carotid perivascular collar model in apolipoprotein E-deficient mice was imaged at 7.0 Tesla MRI before contrast administration and at 8 h and 24 h after injection of 30 mg Fe/kg. The results showed MRI signal loss in the carotid atherosclerotic lesions after administration of targeted anti-OxLDL-USPIO at 8 h and 24 h, which is consistent with the presence of the nanoparticles in the lesions. Immunohistochemistry confirmed the colocalization of the OxLDL/macrophages and iron oxide nanoparticles. The nonspecific IgG-USPIO, unconjugated USPIO nanoparticles, and competitive inhibition groups had limited signal changes (p < 0.05). This report shows that anti-OxLDL-USPIO nanoparticles can be used to directly detect OxLDL and image atherosclerotic lesions within 24 h of nanoparticle administration and suggests a strategy for the therapeutic evaluation of atherosclerotic plaques in vivo.
Collapse
Affiliation(s)
- Song Wen 文颂
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Dong-Fang Liu 柳东芳
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zhen Liu 刘振
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China
| | - Steven Harris
- Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, GA
| | - Yu-Yu Yao 姚玉宇
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qi Ding 丁琪
- Jiangsu Key Laboratory for Biomaterials and Devices, State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fang Nie 聂芳
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Tong Lu 卢瞳
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Hua-Jun Chen 陈华俊
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yan-Li An 安艳丽
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Feng-Chao Zang 臧凤超
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Gao-Jun Teng 滕皋军
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
22
|
Phinikaridou A, Hamilton JA. Application of MRI to detect high-risk atherosclerotic plaque. Expert Rev Cardiovasc Ther 2011; 9:545-50. [PMID: 21615314 DOI: 10.1586/erc.11.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Li L, Najafi AH, Kitlinska JB, Neville R, Laredo J, Epstein SE, Burnett MS, Zukowska Z. Of mice and men: neuropeptide Y and its receptors are associated with atherosclerotic lesion burden and vulnerability. J Cardiovasc Transl Res 2011; 4:351-62. [PMID: 21468772 DOI: 10.1007/s12265-011-9271-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 03/08/2011] [Indexed: 01/13/2023]
Abstract
Neuropeptide Y (NPY), a sympathetic and platelet-derived vascular mitogen and angiogenic factor, has been implicated in atherosclerosis in animal and human genetic studies. Here we evaluate its association with human and murine atherosclerosis, and assess the role of platelet-derived NPY in lesion vulnerability. NPY immunoreactivity (NPY-ir) was measured in the platelet-poor and platelet-rich (PRP) plasmas, and NPY receptors (mitogenic Y1R and angiogenic Y2 and Y5Rs), CD26/DPPIV (a protease forming Y2/Y5-selective agonist), CD31-positive vascularity, and lesion morphology assessed by histo- and immunocyto-chemistry-in patients with peripheral artery disease (PAD) and healthy volunteers, and in lard-fed ApoE-/- mice. NPY and NPY-R immunostaining was greater in lesions from PAD patients compared to normal vessels of healthy volunteers (p < 0.001), and localized to smooth muscle cells, macrophages, and adventitial/neovascular endothelial cells. CD26/DPPIV staining co-localized with CD31-positive endothelial cells only in atherosclerotic lesions. NPY-ir in PRP (but not plasma) and vascular immunostaining was higher (p < 0.05 and 0.001, respectively) in men (not women) with PAD compared to healthy subjects. A similar gender specificity was observed in mice. PRP NPY-ir levels correlated with lesion area (p = 0.03), necrotic core area, and the necrotic core-to-lesion area ratio (p < 0.01) in male, but not female, mice. Also males with neovascularized lesions had higher PRP NPY-ir levels than those lacking lesion microvessels (p < 0.05). NPY and its Rs are up-regulated in human and murine atherosclerotic lesions suggesting pathogenic role. DPPIV expression by microvascular endothelium in atherosclerotic tissue may shift NPY's affinity toward angiogenic Y2/Y5Rs, and thus enhance angiogenesis and lesion vulnerability. Remarkably, plaque neovascularization was associated with increased NPY-ir in PRP in males but not females, suggesting that platelet NPY may be a novel mediator/marker of lesion vulnerability particularly in males, for reasons that remain to be determined. Both animal and human data suggest that NPY is an important contributor to, and platelet NPY-ir a marker of, atherosclerotic lesion burden and vulnerability but only in males, perhaps due to androgen-dependent up-regulation of NPY, previously shown in rats.
Collapse
Affiliation(s)
- Lijun Li
- Department of Physiology and Biophysics, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
The Fat-Fed Apolipoprotein E Knockout Mouse Brachiocephalic Artery in the Study of Atherosclerotic Plaque Rupture. J Biomed Biotechnol 2011; 2011:379069. [PMID: 21076539 PMCID: PMC2975993 DOI: 10.1155/2011/379069] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 10/11/2010] [Accepted: 10/11/2010] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis has been studied in animals for almost a century, yet the events leading up to the rupture of an atherosclerotic plaque (the underlying cause of the majority of fatal thrombosis formation) have only been studied in the past decade, due in part to the development of a mouse model of spontaneous plaque rupture. Apolipoprotein E knockout mice, when fed a high-fat diet, consistently develop lesions in the brachiocephalic artery that rupture at a known time point. It is therefore now possible to observe the development of lesions to elucidate the mechanisms behind the rupture of plaques. Critics argue that the model does not replicate the appearance of human atherosclerotic plaque ruptures. The purpose of this review is to highlight the reasons why we should be looking to the apolipoprotein E knockout mouse to further our understanding of plaque rupture.
Collapse
|
25
|
Hayashi C, Viereck J, Hua N, Phinikaridou A, Madrigal AG, Gibson FC, Hamilton JA, Genco CA. Porphyromonas gingivalis accelerates inflammatory atherosclerosis in the innominate artery of ApoE deficient mice. Atherosclerosis 2010. [PMID: 21251656 DOI: 10.1016/j.atherosclerosis.2010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Studies in humans support a role for the oral pathogen Porphyromonas gingivalis in the development of inflammatory atherosclerosis. The goal of this study was to determine if P. gingivalis infection accelerates inflammation and atherosclerosis in the innominate artery of mice, an artery which has been reported to exhibit many features of human atherosclerotic disease, including plaque rupture. METHODS AND RESULTS Apolipoprotein E-deficient (ApoE-/-) mice were orally infected with P. gingivalis, and magnetic resonance imaging (MRI) was used to monitor the progression of atherosclerosis in live mice. P. gingivalis infected mice exhibited a statistically significant increase in atherosclerotic plaque in the innominate artery as compared to uninfected mice. Polarized light microscopy and immunohistochemistry revealed that the innominate arteries of infected mice had increased lipids, macrophages and T cells as compared to uninfected mice. Increases in plaque, total cholesterol esters and cholesterol monohydrate crystals, macrophages, and T cells were prevented by immunization with heat-killed P. gingivalis prior to pathogen exposure. CONCLUSIONS These are the first studies to demonstrate progression of inflammatory plaque accumulation in the innominate arteries by in vivo MRI analysis following pathogen exposure, and to document protection from plaque progression in the innominate artery via immunization.
Collapse
Affiliation(s)
- Chie Hayashi
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hayashi C, Viereck J, Hua N, Phinikaridou A, Madrigal AG, Gibson FC, Hamilton JA, Genco CA. Porphyromonas gingivalis accelerates inflammatory atherosclerosis in the innominate artery of ApoE deficient mice. Atherosclerosis 2010; 215:52-9. [PMID: 21251656 DOI: 10.1016/j.atherosclerosis.2010.12.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 11/29/2010] [Accepted: 12/10/2010] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Studies in humans support a role for the oral pathogen Porphyromonas gingivalis in the development of inflammatory atherosclerosis. The goal of this study was to determine if P. gingivalis infection accelerates inflammation and atherosclerosis in the innominate artery of mice, an artery which has been reported to exhibit many features of human atherosclerotic disease, including plaque rupture. METHODS AND RESULTS Apolipoprotein E-deficient (ApoE-/-) mice were orally infected with P. gingivalis, and magnetic resonance imaging (MRI) was used to monitor the progression of atherosclerosis in live mice. P. gingivalis infected mice exhibited a statistically significant increase in atherosclerotic plaque in the innominate artery as compared to uninfected mice. Polarized light microscopy and immunohistochemistry revealed that the innominate arteries of infected mice had increased lipids, macrophages and T cells as compared to uninfected mice. Increases in plaque, total cholesterol esters and cholesterol monohydrate crystals, macrophages, and T cells were prevented by immunization with heat-killed P. gingivalis prior to pathogen exposure. CONCLUSIONS These are the first studies to demonstrate progression of inflammatory plaque accumulation in the innominate arteries by in vivo MRI analysis following pathogen exposure, and to document protection from plaque progression in the innominate artery via immunization.
Collapse
Affiliation(s)
- Chie Hayashi
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, United States.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Targeting atherosclerosis by using modular, multifunctional micelles. Proc Natl Acad Sci U S A 2009; 106:9815-9. [PMID: 19487682 DOI: 10.1073/pnas.0903369106] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Subtle clotting that occurs on the luminal surface of atherosclerotic plaques presents a novel target for nanoparticle-based diagnostics and therapeutics. We have developed modular multifunctional micelles that contain a targeting element, a fluorophore, and, when desired, a drug component in the same particle. Targeting atherosclerotic plaques in ApoE-null mice fed a high-fat diet was accomplished with the pentapeptide cysteine-arginine-glutamic acid-lysine-alanine, which binds to clotted plasma proteins. The fluorescent micelles bind to the entire surface of the plaque, and notably, concentrate at the shoulders of the plaque, a location that is prone to rupture. We also show that the targeted micelles deliver an increased concentration of the anticoagulant drug hirulog to the plaque compared with untargeted micelles.
Collapse
|
28
|
Ni M, Wang Y, Zhang M, Zhang PF, Ding SF, Liu CX, Liu XL, Zhao YX, Zhang Y. Atherosclerotic plaque disruption induced by stress and lipopolysaccharide in apolipoprotein E knockout mice. Am J Physiol Heart Circ Physiol 2009; 296:H1598-606. [PMID: 19286965 DOI: 10.1152/ajpheart.01202.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
To establish an animal model with disruptions of atherosclerotic plaques, 96 male apolipoprotein E knockout (apoE(-/-)) mice were randomly divided into stress, lipopolysaccharide (LPS), stress+LPS, and control groups (n = 24 each). All mice were fed a high-fat diet throughout the experiment, and carotid atherosclerotic lesions were induced by placement of a constrictive perivascular collar. Four weeks after surgery, mice in the LPS and stress+LPS groups were intraperitoneally injected with LPS (1 mg/kg twice per week for 8 wk). Eight weeks after surgery, mice in the stress and stress+LPS groups were treated with intermittent physical stress (electric foot shock and noise stimulation) for 4 wk. Morphological analysis revealed a plaque disruption rate of 16.7% in control, 34.8% in LPS, 54.2% in stress, and 60.9% in stress+LPS groups. The disruption rates in stress and stress+LPS groups were both significantly higher than those of controls (P = 0.007 and P = 0.002, respectively). Luminal thrombosis secondary to plaque disruption was observed only in the stress+LPS group. Both stress and LPS stimulation significantly decreased fibrous cap thickness and increased macrophage and lipid contents in plaques. Moreover, the combination of stress and LPS stimulation further lowered cap thickness and enhanced accumulation of macrophages and expression of inflammatory cytokines and matrix metalloproteinases. Stress activated the sympathetic nervous system, as manifested by increased blood pressure and flow velocity. Plasma fibrinogen levels were remarkably elevated in the stress and stress+LPS groups. In conclusion, stress- and LPS-costimulated apoE(-/-) mice provide a useful model for studies of plaque vulnerability and interventions.
Collapse
Affiliation(s)
- Mei Ni
- Shandong Univ. Qilu Hospital, No. 107, Wen Hua Xi Road, Jinan, Shandong 250012, P. R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Shiomi M, Fan J. Unstable coronary plaques and cardiac events in myocardial infarction-prone Watanabe heritable hyperlipidemic rabbits: questions and quandaries. Curr Opin Lipidol 2008; 19:631-6. [PMID: 18957890 DOI: 10.1097/mol.0b013e3283189c18] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Use of suitable animal models is essential for investigation of the mechanisms underlying cardiac events and development of the therapeutic strategies; however, ideal animal models that can recapitulate human coronary atherosclerosis and subsequent acute myocardial infarction are still lacking. In this article, we review the insights learned from myocardial infarction-prone Watanabe heritable hyperlipidemic (designated as WHHLMI) rabbits and discuss the possibility of using this model for the study of human acute coronary syndromes. RECENT FINDINGS The vulnerable plaques of human coronary arteries are histologically characterized by a large lipid core and a thin fibrous cap with inflammatory cells. Recent studies have revealed that inflammatory cells and inflammatory mediators (such as cytokines and matrix metalloproteinases) play an important role in the plaque rupture. SUMMARY We developed the WHHLMI rabbit that shows spontaneous myocardial infarction caused by coronary atherosclerosis. The coronary lesions of WHHLMI rabbits have features of fatty streaks, fibrous plaques, and fibroatheromatous plaques. Some plaques contain a lipid core and a thin fibrous cap similar to human vulnerable plaques. In spite of this, the plaque rupture is not observed in WHHLMI rabbits, suggesting that other additional factors such as mechanical stress are required to trigger the rupture. WHHLMI rabbits may become an important means for elucidating the possible mechanisms of plaque rupture by exposing the plaques to additional risk factors beyond hyperlipidemia.
Collapse
Affiliation(s)
- Masashi Shiomi
- Institute for Experimental Animals, Kobe University School of Medicine, Kobe, Hyogo, Japan.
| | | |
Collapse
|
30
|
Cao RY, St Amand T, Gräbner R, Habenicht AJR, Funk CD. Genetic and pharmacological inhibition of the 5-lipoxygenase/leukotriene pathway in atherosclerotic lesion development in ApoE deficient mice. Atherosclerosis 2008; 203:395-400. [PMID: 18804760 DOI: 10.1016/j.atherosclerosis.2008.07.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 07/25/2008] [Accepted: 07/28/2008] [Indexed: 11/29/2022]
Abstract
The 5-lipoxygenase (5-LO) catalyzed formation of leukotriene (LT) lipid mediators is a pathway contributing to inflammatory events in asthma and more recently has been associated with cardiovascular disease. However, the relative impact of this pathway in atherogenesis has been controversial and a variety of mixed results reported. The goal of these studies was to assess the importance of the 5-LO/LT pathway in mice with either genetic (5-LO(-/-)) or pharmacological (L-739,010) inhibition of the 5-LO pathway on an apolipoprotein E deficient (apoE(-/-)) background when subjected to either an 8-week (Paigen) or 6 months (Western) atherosclerotic diet regimen. Atherosclerotic lesion analysis at the aortic root, brachiocephalic artery and throughout the whole aorta by en face Sudan IV staining was determined, as well as blood lipid levels. Ex vivo calcium ionophore-stimulation of whole blood demonstrated a significant reduction in the capacity to form LTB(4) in 5-LO(-/-) and drug-treated 5-LO(+/+) mice. Quantitative analysis of atherosclerotic lesions did not differ between groups at all three sites. Moreover, the composition of advanced lesions in the brachiocephalic arteries did not indicate altered plaque disruption as a result of 5-LO gene inactivation. These results do not support a role for the 5-LO/LT pathway in intermediate to advanced atherosclerotic lesion development in mice.
Collapse
Affiliation(s)
- Richard Yang Cao
- Department of Physiology, Queen's University, Kingston, ON, Canada
| | | | | | | | | |
Collapse
|
31
|
Single-Dose and Fractionated Irradiation Promote Initiation and Progression of Atherosclerosis and Induce an Inflammatory Plaque Phenotype in ApoE−/− Mice. Int J Radiat Oncol Biol Phys 2008; 71:848-57. [DOI: 10.1016/j.ijrobp.2008.02.031] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 01/18/2008] [Accepted: 01/23/2008] [Indexed: 12/14/2022]
|
32
|
|
33
|
|