1
|
Tomas L, Prica F, Schulz C. Trafficking of Mononuclear Phagocytes in Healthy Arteries and Atherosclerosis. Front Immunol 2021; 12:718432. [PMID: 34759917 PMCID: PMC8573388 DOI: 10.3389/fimmu.2021.718432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Monocytes and macrophages play essential roles in all stages of atherosclerosis – from early precursor lesions to advanced stages of the disease. Intima-resident macrophages are among the first cells to be confronted with the influx and retention of apolipoprotein B-containing lipoproteins at the onset of hypercholesterolemia and atherosclerosis development. In this review, we outline the trafficking of monocytes and macrophages in and out of the healthy aorta, as well as the adaptation of their migratory behaviour during hypercholesterolemia. Furthermore, we discuss the functional and ontogenetic composition of the aortic pool of mononuclear phagocytes and its link to the atherosclerotic disease process. The development of mouse models of atherosclerosis regression in recent years, has enabled scientists to investigate the behaviour of monocytes and macrophages during the resolution of atherosclerosis. Herein, we describe the dynamics of these mononuclear phagocytes upon cessation of hypercholesterolemia and how they contribute to the restoration of tissue homeostasis. The aim of this review is to provide an insight into the trafficking, fate and disease-relevant dynamics of monocytes and macrophages during atherosclerosis, and to highlight remaining questions. We focus on the results of rodent studies, as analysis of cellular fates requires experimental manipulations that cannot be performed in humans but point out findings that could be replicated in human tissues. Understanding of the biology of macrophages in atherosclerosis provides an important basis for the development of therapeutic strategies to limit lesion formation and promote plaque regression.
Collapse
Affiliation(s)
- Lukas Tomas
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Filip Prica
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
2
|
Yang Y, Li Z, Liu Q, Guo Y, Mei Y, Lyu J, Zhao M, Feng Y, Xie G. Carotid arterial wall MRI of apolipoprotein e-deficient mouse at 7 T using DANTE-prepared variable-flip-angle rapid acquisition with relaxation enhancement. Magn Reson Imaging 2021; 86:1-9. [PMID: 34688846 DOI: 10.1016/j.mri.2021.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Accepted: 10/17/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE To optimize a sequence combining the delay alternating with nutation for tailored excitation (DANTE) preparative module with the variable-flip-angle rapid acquisition with relaxation enhancement (VF-RARE) sequence (DANTE-VF-RARE) and to investigate its feasibility for vessel wall imaging in Apolipoprotein E-Deficient (ApoE-/-) mouse at 7 Tesla (T). MATERIALS AND METHODS Specific T1/T2 values were used for producing a sharper vessel wall in the variable-flip-angle optimization scheme. The DANTE RF pulse flip angle and pulse train length were optimized for maximizing the wall-lumen contrast. ApoE-/- (fed high fat diet for 20/40/ 60 weeks, n = 9/4/4) and wild-type mice (controls, n = 3) were imaged at 7 T using VF-RARE, DANTE-VF-RARE, time-of-flight (TOF) angiography, and multi-slice T1-weighted 2D RARE coupled with inflow outflow saturation bands (IOSB-RARE). Wall-lumen contrast-to-noise-ratio efficiency (CNReff), lumen area (LA), and wall area (WA) were compared between DANTE-VF-RARE and 2D IOSB-RARE sequences. Additionally, linear regression analysis was conducted between MR measurements and histomorphometric planimetry results. RESULTS Residual blood signal was observed in the four out of eighteen carotids on VF-RARE images, whereas it was significantly suppressed on DANTE-VF-RARE images. Compared with IOSB-RARE, DANTE-VF-RARE offered significantly improved CNReff (P < 0.001). The LA and WA were both comparable (P = 0.085 and 0.112, respectively) and showed excellent agreement between DANTE-VF-RARE and IOSB-RARE (ICC = 0.96 and 0.95, respectively). The luminal stenosis identified by DANTE-VF-RARE was in consistent with the results of TOF. Strong correlations were found between MR measurements and histopathological analysis for both WA (DANTE-VF-RARE: r = 0.92, slope = 0.94, P < 0.001; IOSB-RARE: r = 0.93, slope = 0.94, P < 0.001) and LA (DANTE-VF-RARE: r = 0.82, slope = 0.54, P < 0.001; IOSB-RARE: r = 0.78, slope = 0.50, P < 0.001). CONCLUSION DANTE-VF-RARE achieves effective blood signal suppression and is a feasible approach for the 3D carotid arterial wall imaging of ApoE-/- mouse at 7 T.
Collapse
Affiliation(s)
- Yuanbo Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhonghao Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiang Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Yihao Guo
- MR Collaboration, Siemens Healthcare Ltd., Guangzhou, China
| | - Yingjie Mei
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China; Philips Healthcare, Guangzhou, China
| | - Jian Lyu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China.
| | - Guoxi Xie
- Department of Biomedical Engineering, The Sixth Affiliated Hospital, School of Basic Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
3
|
PEG-modified gadolinium nanoparticles as contrast agents for in vivo micro-CT. Sci Rep 2021; 11:16603. [PMID: 34400681 PMCID: PMC8367985 DOI: 10.1038/s41598-021-95716-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Vascular research is largely performed in rodents with the goal of developing treatments for human disease. Micro-computed tomography (micro-CT) provides non-destructive three-dimensional imaging that can be used to study the vasculature of rodents. However, to distinguish vasculature from other soft tissues, long-circulating contrast agents are required. In this study, we demonstrated that poly(ethylene glycol) (PEG)-coated gadolinium nanoparticles can be used as a vascular contrast agent in micro-CT. The coated particles could be lyophilized and then redispersed in an aqueous solution to achieve 100 mg/mL of gadolinium. After an intravenous injection of the contrast agent into mice, micro-CT scans showed blood pool contrast enhancements of at least 200 HU for 30 min. Imaging and quantitative analysis of gadolinium in tissues showed the presence of contrast agent in clearance organs including the liver and spleen and very low amounts in other organs. In vitro cell culture experiments, subcutaneous injections, and analysis of mouse body weight suggested that the agents exhibited low toxicity. Histological analysis of tissues 5 days after injection of the contrast agent showed cytotoxicity in the spleen, but no abnormalities were observed in the liver, lungs, kidneys, and bladder.
Collapse
|
4
|
Josefs T, Basu D, Vaisar T, Arets B, Kanter JE, Huggins LA, Hu Y, Liu J, Clouet-Foraison N, Heinecke JW, Bornfeldt KE, Goldberg IJ, Fisher EA. Atherosclerosis Regression and Cholesterol Efflux in Hypertriglyceridemic Mice. Circ Res 2021; 128:690-705. [PMID: 33530703 DOI: 10.1161/circresaha.120.317458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Tatjana Josefs
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Tomas Vaisar
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | | | - Jenny E Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Jianhua Liu
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| | - Noemie Clouet-Foraison
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Jay W Heinecke
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Karin E Bornfeldt
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Edward A Fisher
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| |
Collapse
|
5
|
Jinnouchi H, Guo L, Sakamoto A, Torii S, Sato Y, Cornelissen A, Kuntz S, Paek KH, Fernandez R, Fuller D, Gadhoke N, Surve D, Romero M, Kolodgie FD, Virmani R, Finn AV. Diversity of macrophage phenotypes and responses in atherosclerosis. Cell Mol Life Sci 2020; 77:1919-1932. [PMID: 31720740 PMCID: PMC11104939 DOI: 10.1007/s00018-019-03371-3] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
The presence of macrophages within the plaque is a defining hallmark of atherosclerosis. Macrophages are exposed to various microenvironments such as oxidized lipids and cytokines which effect their phenotypic differentiation and activation. Classically, macrophages have been divided into two groups: M1 and M2 macrophages induced by T-helper 1 and T-helper 2 cytokines, respectively. However, for a decade, greater phenotypic heterogeneity and plasticity of these cells have since been reported in various models. In addition to M1 and M2 macrophage phenotypes, the concept of additional macrophage phenotypes such as M (Hb), Mox, and M4 has emerged. Understanding the mechanisms and functions of distinct phenotype of macrophages can lead to determination of their potential role in atherosclerotic plaque pathogenesis. However, there are still many unresolved controversies regarding their phenotype and function with respect to atherosclerosis. Here, we summarize and focus on the differential subtypes of macrophages in atherosclerotic plaques and their differing functional roles based upon microenvironments such as lipid, intraplaque hemorrhage, and plaque regression.
Collapse
Affiliation(s)
| | - Liang Guo
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Atsushi Sakamoto
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Sho Torii
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Yu Sato
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Anne Cornelissen
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Salome Kuntz
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Ka Hyun Paek
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Raquel Fernandez
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Daniela Fuller
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Neel Gadhoke
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Dipti Surve
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Maria Romero
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Frank D Kolodgie
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Renu Virmani
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Aloke V Finn
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
6
|
Chang HR, Josefs T, Scerbo D, Gumaste N, Hu Y, Huggins LA, Barett T, Chiang S, Grossman J, Bagdasarov S, Fisher EA, Goldberg IJ. Role of LpL (Lipoprotein Lipase) in Macrophage Polarization In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2019; 39:1967-1985. [PMID: 31434492 PMCID: PMC6761022 DOI: 10.1161/atvbaha.119.312389] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.
Collapse
Affiliation(s)
- Hye Rim Chang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tatjana Josefs
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Diego Scerbo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tessa Barett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York; Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| | - Stephanie Chiang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Jennifer Grossman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Svetlana Bagdasarov
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Edward A. Fisher
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
7
|
Barrett TJ, Distel E, Murphy AJ, Hu J, Garshick MS, Ogando Y, Liu J, Vaisar T, Heinecke JW, Berger JS, Goldberg IJ, Fisher EA. Apolipoprotein AI) Promotes Atherosclerosis Regression in Diabetic Mice by Suppressing Myelopoiesis and Plaque Inflammation. Circulation 2019; 140:1170-1184. [PMID: 31567014 PMCID: PMC6777860 DOI: 10.1161/circulationaha.119.039476] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Despite robust cholesterol lowering, cardiovascular disease risk remains increased in patients with diabetes mellitus. Consistent with this, diabetes mellitus impairs atherosclerosis regression after cholesterol lowering in humans and mice. In mice, this is attributed in part to hyperglycemia-induced monocytosis, which increases monocyte entry into plaques despite cholesterol lowering. In addition, diabetes mellitus skews plaque macrophages toward an atherogenic inflammatory M1 phenotype instead of toward the atherosclerosis-resolving M2 state typical with cholesterol lowering. Functional high-density lipoprotein (HDL), typically low in patients with diabetes mellitus, reduces monocyte precursor proliferation in murine bone marrow and has anti-inflammatory effects on human and murine macrophages. Our study aimed to test whether raising functional HDL levels in diabetic mice prevents monocytosis, reduces the quantity and inflammation of plaque macrophages, and enhances atherosclerosis regression after cholesterol lowering. METHODS Aortic arches containing plaques developed in Ldlr-/- mice were transplanted into either wild-type, diabetic wild-type, or diabetic mice transgenic for human apolipoprotein AI, which have elevated functional HDL. Recipient mice all had low levels of low-density lipoprotein cholesterol to promote plaque regression. After 2 weeks, plaques in recipient mouse aortic grafts were examined. RESULTS Diabetic wild-type mice had impaired atherosclerosis regression, which was normalized by raising HDL levels. This benefit was linked to suppressed hyperglycemia-driven myelopoiesis, monocytosis, and neutrophilia. Increased HDL improved cholesterol efflux from bone marrow progenitors, suppressing their proliferation and monocyte and neutrophil production capacity. In addition to reducing circulating monocytes available for recruitment into plaques, in the diabetic milieu, HDL suppressed the general recruitability of monocytes to inflammatory sites and promoted plaque macrophage polarization to the M2, atherosclerosis-resolving state. There was also a decrease in plaque neutrophil extracellular traps, which are atherogenic and increased by diabetes mellitus. CONCLUSIONS Raising apolipoprotein AI and functional levels of HDL promotes multiple favorable changes in the production of monocytes and neutrophils and in the inflammatory environment of atherosclerotic plaques of diabetic mice after cholesterol lowering and may represent a novel approach to reduce cardiovascular disease risk in people with diabetes mellitus.
Collapse
Affiliation(s)
- Tessa J. Barrett
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Emilie Distel
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Andrew J. Murphy
- Haematopoiesis and Leukocyte Biology, Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, NY 10016, USA
| | - Michael S. Garshick
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Yoscar Ogando
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Jianhua Liu
- Department of Surgery, Mount Sinai School of Medicine, New York, NY, USA
| | - Tomas Vaisar
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle
| | - Jay W. Heinecke
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle
| | - Jeffrey S. Berger
- Department of Medicine, Divisions of Cardiology and Hematology, Department of Surgery, Division of Vascular Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Ira J. Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY 10016, USA
| | - Edward A. Fisher
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
- Department of Microbiology and Immunology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
8
|
Vulnerable Plaque, Characteristics, Detection, and Potential Therapies. J Cardiovasc Dev Dis 2019; 6:jcdd6030026. [PMID: 31357630 PMCID: PMC6787609 DOI: 10.3390/jcdd6030026] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/21/2019] [Accepted: 07/24/2019] [Indexed: 12/16/2022] Open
Abstract
Plaque development and rupture are hallmarks of atherosclerotic vascular disease. Despite current therapeutic developments, there is an unmet necessity in the prevention of atherosclerotic vascular disease. It remains a challenge to determine at an early stage if atherosclerotic plaque will become unstable and vulnerable. The arrival of molecular imaging is receiving more attention, considering it allows for a better understanding of the biology of human plaque and vulnerabilities. Various plaque therapies with common goals have been tested in high-risk patients with cardiovascular disease. In this work, the process of plaque instability, along with current technologies for sensing and predicting high-risk plaques, is debated. Updates on potential novel therapeutic approaches are also summarized.
Collapse
|
9
|
Allahverdian S, Chaabane C, Boukais K, Francis GA, Bochaton-Piallat ML. Smooth muscle cell fate and plasticity in atherosclerosis. Cardiovasc Res 2019; 114:540-550. [PMID: 29385543 DOI: 10.1093/cvr/cvy022] [Citation(s) in RCA: 347] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Current knowledge suggests that intimal smooth muscle cells (SMCs) in native atherosclerotic plaque derive mainly from the medial arterial layer. During this process, SMCs undergo complex structural and functional changes giving rise to a broad spectrum of phenotypes. Classically, intimal SMCs are described as dedifferentiated/synthetic SMCs, a phenotype characterized by reduced expression of contractile proteins. Intimal SMCs are considered to have a beneficial role by contributing to the fibrous cap and thereby stabilizing atherosclerotic plaque. However, intimal SMCs can lose their properties to such an extent that they become hard to identify, contribute significantly to the foam cell population, and acquire inflammatory-like cell features. This review highlights mechanisms of SMC plasticity in different stages of native atherosclerotic plaque formation, their potential for monoclonal or oligoclonal expansion, as well as recent findings demonstrating the underestimated deleterious role of SMCs in this disease.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Chiraz Chaabane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel Servet-1, 1211 Geneva 4, Switzerland
| | - Kamel Boukais
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Gordon A Francis
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel Servet-1, 1211 Geneva 4, Switzerland
| |
Collapse
|
10
|
Effects of Pharmacological Thermogenic Adipocyte Activation on Metabolism and Atherosclerotic Plaque Regression. Nutrients 2019; 11:nu11020463. [PMID: 30813320 PMCID: PMC6412269 DOI: 10.3390/nu11020463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 11/16/2022] Open
Abstract
Thermogenic adipocytes burn nutrients in order to produce heat. Upon activation, brown adipose tissue (BAT) clears vast amounts of lipids and glucose from the circulation and thus substantially lowers plasma lipid levels. As a consequence, BAT activation protects from the development of atherosclerosis. However, it is unclear if pharmacologic activation of BAT can be exploited therapeutically to reduce plaque burden in established atherosclerotic disease. Here we study the impact of thermogenic adipose tissues on plaque regression in a mouse model of atherosclerosis. Thermogenic adipocytes in atherosclerotic low-density lipoprotein (LDL) receptor (LDLR)-deficient mice were pharmacologically activated by dietary CL316,243 (CL) treatment for 4 weeks and the outcomes on metabolically active tissues, plasma lipids and atherosclerosis were analyzed. While the chronic activation of thermogenic adipocytes reduced adiposity, increased browning of white adipose tissue (WAT), altered liver gene expression, and reduced plasma triglyceride levels, atherosclerotic plaque burden remained unchanged. Our findings suggest that despite improving adiposity and plasma triglycerides, pharmacologic activation of thermogenic adipocytes is not able to reverse atherosclerosis in LDLR-deficient mice.
Collapse
|
11
|
Brito V, Mellal K, Zoccal KF, Soto Y, Ménard L, Sarduy R, Faccioli LH, Ong H, Vázquez AM, Marleau S. Atheroregressive Potential of the Treatment with a Chimeric Monoclonal Antibody against Sulfated Glycosaminoglycans on Pre-existing Lesions in Apolipoprotein E-Deficient Mice. Front Pharmacol 2017; 8:782. [PMID: 29163168 PMCID: PMC5672559 DOI: 10.3389/fphar.2017.00782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023] Open
Abstract
The retention of lipoprotein particles in the intima, in particular to glycosaminoglycan side chains of proteoglycans, is a critical step in atherosclerosis initiation. Administration of chP3R99, a chimeric mouse/human monoclonal antibody inducing an anti-idiotypic network response against glycosaminoglycans was previously shown to prevent atherosclerotic lesion progression, yet its effect in the late-stage progression of lesions remains unknown. This study investigated the effect of chP3R99 at a late stage of disease development in apolipoprotein E-deficient mice and the vascular mechanisms involved. Male apolipoprotein E-deficient mice were fed a high-fat high-cholesterol diet from 4 to 19 weeks old, at which time mice were fed normal chow and 5 doses of chP3R99 (50 μg) or isotype-matched IgG (hR3) were administered subcutaneously weekly for the first 3 administrations, then at weeks 24 and 26 before sacrifice (week 28). Lesions progression was reduced by 88% in treated mice with no change in total plasma cholesterol levels, yet with increased sera reactivity to chP3R99 idiotype and heparin, suggesting the induction of an anti-idiotype antibody cascade against glycosaminoglycans, which was likely related with the atheroprotective effect. chP3R99 treatment initiated regression in a significant number of mice. Circulating levels of interleukin-6 were reduced along with a striking diminution of inflammatory cell accumulation in the vessel wall, and of VCAM-1 labeling in vivo. The ratio of IL-10/iNOS gene expression in aortas increased in chP3R99-treated mice. In conclusion, our results show that treatment with chP3R99 reduces vascular inflammatory burden and halts lesion progression with potential for regression in the late phase of the disease in atherosclerotic mice, and support the therapeutic intervention against glycosaminoglycans as a novel strategy to reverse atherosclerosis.
Collapse
Affiliation(s)
- Victor Brito
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Katia Mellal
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Karina F Zoccal
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Department of Clinical Analysis, Toxicology and Bromatology, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Yosdel Soto
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Liliane Ménard
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Roger Sarduy
- Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Lucia H Faccioli
- Department of Clinical Analysis, Toxicology and Bromatology, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Huy Ong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Ana M Vázquez
- Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
12
|
Abstract
The introduction of molecularly targeted therapies with tyrosine kinase inhibitors has revolutionized cancer therapy and has contributed to a steady decline in cancer-related mortality since the late 1990s. However, not only cardiac but also vascular toxicity has been reported for these agents, some as expected on-target effects (e.g., VEGF receptor inhibitors) and others as unanticipated events (e.g., BCR-Abl inhibitors). A sound understanding of these cardiovascular toxic effects is critical to advance mechanistic insight into vascular disease and clinical care. From a conceptual standpoint, there might be value in defining type I (permanent) and type II (transient) vascular toxicity. This review will focus on the tyrosine kinase inhibitors in current clinical use and their associated vascular side effects.
Collapse
|
13
|
Jung C, Christiansen S, Kaul MG, Koziolek E, Reimer R, Heeren J, Adam G, Heine M, Ittrich H. Quantitative and qualitative estimation of atherosclerotic plaque burden in vivo at 7T MRI using Gadospin F in comparison to en face preparation evaluated in ApoE KO mice. PLoS One 2017; 12:e0180407. [PMID: 28771481 PMCID: PMC5542445 DOI: 10.1371/journal.pone.0180407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Background The aim of the study was to quantify atherosclerotic plaque burden by volumetric assessment and T1 relaxivity measurement at 7T MRI using Gadospin F (GDF) in comparison to en face based measurements. Methods and results 9-weeks old ApoE-/- (n = 5 for each group) and wildtype mice (n = 5) were set on high fat diet (HFD). Progression group received MRI at 9, 13, 17 and 21 weeks after HFD initiation. Regression group was reswitched to chow diet (CD) after 13 weeks HFD and monitored with MRI for 12 weeks. MRI was performed before and two hours after iv injection of GDF (100 μmol/kg) at 7T (Clinscan, Bruker) acquiring a 3D inversion recovery gradient echo sequence and T1 Mapping using Saturation Recovery sequences. Subsequently, aortas were prepared for en face analysis using confocal microscopy. Total plaque volume (TPV) and T1 relaxivity were estimated using ImageJ (V. 1.44p, NIH, USA). 2D and 3D en face analysis showed a strong and exponential increase of plaque burden over time, while plaque burden in regression group was less pronounced. Correspondent in vivo MRI measurements revealed a more linear increase of TPV and T1 relaxivity for regression group. A significant correlation was observed between 2D and 3D en face analysis (r = 0.79; p<0.001) as well as between 2D / 3D en face analysis and MRI (r = 0.79; p<0.001; r = 0.85; p<0.001) and delta R1 (r = 0.79; p<0.001; r = 0.69; p<0.01). Conclusion GDF-enhanced in vivo MRI is a powerful non-invasive imaging technique in mice allowing for reliable estimation of atherosclerotic plaque burden, monitoring of disease progression and regression in preclinical studies.
Collapse
Affiliation(s)
- Caroline Jung
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Sabine Christiansen
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Gerhard Kaul
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Koziolek
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Nuclear Medicine, Berlin Experimental Radionuclide Imaging Center (BERIC), University Medical Center Charité, Berlin, Germany
| | - Rudolph Reimer
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Jörg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harald Ittrich
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Abstract
On the basis of studies that extend back to the early 1900s, regression and stabilization of atherosclerosis in humans has progressed from being a concept to one that is achievable. Successful attempts at regression generally applied robust measures to improve plasma lipoprotein profiles. Possible mechanisms responsible for lesion shrinkage include decreased retention of atherogenic apolipoprotein B within the arterial wall, efflux of cholesterol and other toxic lipids from plaques, emigration of lesional foam cells out of the arterial wall, and influx of healthy phagocytes that remove necrotic debris as well as other components of the plaque. Currently available clinical agents, however, still fail to stop most cardiovascular events. For years, HDL has been considered the 'good cholesterol.' Clinical intervention studies to causally link plasma HDL-C levels to decreased progression or to the regression of atherosclerotic plaques are relatively few because of the lack of therapeutic agents that can selectively and potently increase HDL-C. The negative results of studies that were carried out have led to uncertainty as to the role that HDL plays in atherosclerosis. It is becoming clearer, however, that HDL function rather than quantity is most crucial and, therefore, discovery of agents that enhance the quality of HDL should be the goal.
Collapse
|
15
|
Chistiakov DA, Myasoedova VA, Revin VV, Orekhov AN, Bobryshev YV. The phenomenon of atherosclerosis reversal and regression: Lessons from animal models. Exp Mol Pathol 2017; 102:138-145. [PMID: 28108216 DOI: 10.1016/j.yexmp.2017.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/19/2016] [Accepted: 01/14/2017] [Indexed: 12/18/2022]
Abstract
Studies in non-rodent and murine models showed that atherosclerosis can be reversed. Atherosclerosis progression induced by high-fat or cholesterol-rich diet can be reduced and reversed to plaque regression after switching to a normal diet or through administration of lipid-lowering agents. The similar process should exist in humans after implementation of lipid-lowering therapy and as a result of targeting of small rupture-prone plaques that are major contributors for acute atherosclerotic complications. Lowering of low density lipoprotein (LDL) cholesterol and the activation of reverse cholesterol transport lead to a decline in foam cell content, to the depletion of plaque lipid reservoirs, a decrease in lesional macrophage numbers through the activation of macrophage emigration and, probably, apoptosis, dampening plaque inflammation, and the induction of anti-inflammatory macrophages involved in clearance of the necrotic core and plaque healing. By contrast, plaque regression is characterized by opposite events, leading to the retention of atherogenic LDL and oxidized LDL particles in the plaque, an increased flux of monocytes, the immobilization of macrophages in the intimal vascular tissues, and the propagation of intraplaque inflammation. Transfer of various apolipoprotein (apo) genes to spontaneously hypercholesterolemic mice deficient for either apoE or LDL receptor and, especially, the implementation of the transplantation murine model allowed studying molecular mechanisms of atherosclerotic regression, associated with the depletion of atherogenic lipids in the plaque, egress of macrophages and phenotypic switch of macrophages from the proinflammatory M1 to the anti-inflammatory M2.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical University, Moscow, Russia; Department of Molecular Genetic Diagnostics and Cell Biology, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Veronika A Myasoedova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Victor V Revin
- N.P. Ogaryov Mordovian State University, Republic of Mordovia, Saransk 430005, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia; Department of Biophysics, Moscow State University, Moscow, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia; School of Medical Sciences, University of New South Wales, NSW, Sydney, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.
| |
Collapse
|
16
|
Haj Ahmed S, Kharroubi W, Zarrouk A, Brahmi F, Nury T, Lizard G, Hammami M. Protective effects of bezafibrate against elaidic acid-induced accumulation of lipid droplets in monocytic cells. Curr Res Transl Med 2016; 65:20-30. [PMID: 28340693 DOI: 10.1016/j.retram.2016.08.001] [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] [Received: 04/01/2016] [Revised: 07/24/2016] [Accepted: 08/05/2016] [Indexed: 01/15/2023]
Abstract
Some factors related to diet, such as trans fatty acids (TFA), are known to be involved in the progression of atherosclerosis in humans. Thus, the aim of our study was (i) to evaluate the effects of three dietary free fatty acids (FFA) (elaidic (EA), oleic (OA) and palmitic acid (PA)) on U937 human monocytes, and (ii) to study the eventual benefits of bezafibrate (BZF), a pan-agonist for PPAR isoforms (α, γ and δ) in U937 cells treated with FFA. Morphologic and functional changes were investigated by microscopic and flow cytometric methods. Cellular lipid content, lipid droplets and FA composition were identified and studied. All analyses were also realized in association with or without BZF. Contrary to OA and PA, EA slightly induced both propidium iodide-positive cells and mitochondrial depolarization. In addition, in contrast to OA and PA, EA induced only a slight increase in superoxide anion production. However, EA and OA promoted cytoplasmic lipid droplets accumulation. Only EA and OA significantly increased CD36 expression. It is noteworthy that BZF had a more or less pronounced protective effect against EA-, OA- and PA-induced side effects: BZF attenuated the impaired cell viability and inflammatory response, decreased superoxide anion production and prevented the accumulation of neutral and polar lipids. The effects were less pronounced with OA and PA than with EA. Altogether, our data revealed a benefit of BZF on the side effects induced especially with EA. It may thus be of interest in preventing the early stages of atherosclerotic plaque formation.
Collapse
Affiliation(s)
- S Haj Ahmed
- Laboratoire 'nutrition, aliments fonctionnels et santé vasculaire', UR12ES05 faculté de médecine, université de Monastir, Monastir, Tunisia.
| | - W Kharroubi
- Laboratoire 'nutrition, aliments fonctionnels et santé vasculaire', UR12ES05 faculté de médecine, université de Monastir, Monastir, Tunisia
| | - A Zarrouk
- Laboratoire 'nutrition, aliments fonctionnels et santé vasculaire', UR12ES05 faculté de médecine, université de Monastir, Monastir, Tunisia; Équipe 'biochimie du peroxysome, inflammation et métabolisme lipidique' EA7270/université de Bourgogne Franche Comté/Inserm, 21000 Dijon, France
| | - F Brahmi
- Laboratoire de biophysique, biochimie, biomathématique et scientométrie (3BS), département des sciences alimentaires, faculté des Sciences de la Nature et de la Vie, université Abderrahmane Mira, Béjaia, Algeria
| | - T Nury
- Équipe 'biochimie du peroxysome, inflammation et métabolisme lipidique' EA7270/université de Bourgogne Franche Comté/Inserm, 21000 Dijon, France
| | - G Lizard
- Équipe 'biochimie du peroxysome, inflammation et métabolisme lipidique' EA7270/université de Bourgogne Franche Comté/Inserm, 21000 Dijon, France
| | - M Hammami
- Laboratoire 'nutrition, aliments fonctionnels et santé vasculaire', UR12ES05 faculté de médecine, université de Monastir, Monastir, Tunisia
| |
Collapse
|
17
|
Bartels ED, Christoffersen C, Lindholm MW, Nielsen LB. Altered metabolism of LDL in the arterial wall precedes atherosclerosis regression. Circ Res 2015; 117:933-42. [PMID: 26358193 DOI: 10.1161/circresaha.115.307182] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/10/2015] [Indexed: 12/25/2022]
Abstract
RATIONALE Plasma cholesterol lowering is beneficial in patients with atherosclerosis. However, it is unknown how it affects entry and degradation of low-density lipoprotein (LDL) particles in the lesioned arterial wall. OBJECTIVE We studied the effect of lipid-lowering therapy on LDL permeability and degradation of LDL particles in atherosclerotic aortas of mice by measuring the accumulation of iodinated LDL particles in the arterial wall. METHODS AND RESULTS Cholesterol-fed, LDL receptor-deficient mice were treated with either an anti-Apob antisense oligonucleotide or a mismatch control antisense oligonucleotide once a week for 1 or 4 weeks before injection with preparations of iodinated LDL particles. The anti-Apob antisense oligonucleotide reduced plasma cholesterol by ≈90%. The aortic LDL permeability and degradation rates of newly entered LDL particles were reduced by ≈50% and ≈85% already after 1 week of treatment despite an unchanged pool size of aortic iodinated LDL particles. In contrast, the size, foam cell content, and aortic pool size of iodinated LDL particles of aortic atherosclerotic plaques were not reduced until after 4 weeks of treatment with the anti-Apob antisense oligonucleotide. CONCLUSIONS Improved endothelial barrier function toward the entry of plasma LDL particles and diminished aortic degradation of the newly entered LDL particles precede plaque regression.
Collapse
Affiliation(s)
- Emil D Bartels
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (E.D.B., C.C., L.B.N.); Roche Innovation Center Copenhagen, Hoersholm, Denmark (M.W.L.); and Departments of Biomedical Sciences (C.C., L.B.N.) and Clinical Medicine (L.B.N.), University of Copenhagen, Copenhagen, Denmark.
| | - Christina Christoffersen
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (E.D.B., C.C., L.B.N.); Roche Innovation Center Copenhagen, Hoersholm, Denmark (M.W.L.); and Departments of Biomedical Sciences (C.C., L.B.N.) and Clinical Medicine (L.B.N.), University of Copenhagen, Copenhagen, Denmark
| | - Marie W Lindholm
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (E.D.B., C.C., L.B.N.); Roche Innovation Center Copenhagen, Hoersholm, Denmark (M.W.L.); and Departments of Biomedical Sciences (C.C., L.B.N.) and Clinical Medicine (L.B.N.), University of Copenhagen, Copenhagen, Denmark
| | - Lars B Nielsen
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (E.D.B., C.C., L.B.N.); Roche Innovation Center Copenhagen, Hoersholm, Denmark (M.W.L.); and Departments of Biomedical Sciences (C.C., L.B.N.) and Clinical Medicine (L.B.N.), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Bouchareychas L, Pirault J, Saint-Charles F, Deswaerte V, Le Roy T, Jessup W, Giral P, Le Goff W, Huby T, Gautier EL, Lesnik P. Promoting macrophage survival delays progression of pre-existing atherosclerotic lesions through macrophage-derived apoE. Cardiovasc Res 2015; 108:111-23. [PMID: 26092098 DOI: 10.1093/cvr/cvv177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/11/2015] [Indexed: 12/14/2022] Open
Abstract
AIMS Macrophage apoptosis is a prominent feature of atherosclerosis, yet whether cell death-protected macrophages would favour the resolution of already established atherosclerotic lesions, and thus hold therapeutic potential, remains unknown. METHODS AND RESULTS We irradiated then transplanted into Apoe(-/-) or LDLr(-/-) recipient mice harbouring established atherosclerotic lesions, bone marrow cells from mice displaying enhanced macrophage survival through overexpression of the antiapoptotic gene hBcl-2 (Mø-hBcl2 Apoe(-/-) or Mø-hBcl2 Apoe(+/+) LDLr(-/-)). Both recipient mice exhibited decreased lesional apoptotic cell content and reduced necrotic areas when repopulated with Mø-hBcl2 mouse-derived bone marrow cells. In contrast, only LDLr(-/-) recipients showed a reduction in plasma cholesterol levels and in atherosclerotic lesions. The absence of significant reduction of plasma cholesterol levels in the context of apoE deficiency highlighted macrophage-derived apoE as key in both the regulation of plasma and tissue cholesterol levels and the progression of pre-existing lesion. Accordingly, hBcl2 expression in macrophages was associated with larger pools of Kupffer cells and Ly-6C(low) monocytes, both high producers of apoE. Additionally, increased Kupffer cells population was associated with improved clearance of apoptotic cells and modified lipoproteins. CONCLUSION Collectively, these data show that promoting macrophage survival provides a supplemental source of apoE, which hinders pre-existing plaque progression.
Collapse
Affiliation(s)
- Laura Bouchareychas
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France
| | - John Pirault
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France
| | - Flora Saint-Charles
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France
| | - Virginie Deswaerte
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France
| | - Tiphaine Le Roy
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Wendy Jessup
- Atherosclerosis Group, ANZAC Research Institute, University of Sydney and Concord Hospital, Sydney, Australia
| | - Philippe Giral
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Wilfried Le Goff
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Thierry Huby
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Emmanuel L Gautier
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Philippe Lesnik
- INSERM, UMR_S U1166, Integrative Biology of Atherosclerosis Team, Hôpital de la Pitié, Pavillon Benjamin Delessert, 83 Boulevard de L'hôpital, Paris F-75013, France Sorbonne Universités, UPMC Université Paris 06, UMR_S 1166, ICAN, Integrative Biology of Atherosclerosis Team, Paris F-75005, France Institute of Cardiometabolism and Nutrition, ICAN, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| |
Collapse
|
19
|
Feig JE, Feig JL, Kini AS. Statins, atherosclerosis regression and HDL: Insights from within the plaque. Int J Cardiol 2015; 189:168-71. [DOI: 10.1016/j.ijcard.2015.04.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 04/05/2015] [Indexed: 12/18/2022]
|
20
|
|
21
|
Vengrenyuk Y, Nishi H, Long X, Ouimet M, Savji N, Martinez FO, Cassella CP, Moore KJ, Ramsey SA, Miano JM, Fisher EA. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler Thromb Vasc Biol 2015; 35:535-46. [PMID: 25573853 PMCID: PMC4344402 DOI: 10.1161/atvbaha.114.304029] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We previously showed that cholesterol loading in vitro converts mouse aortic vascular smooth muscle cells (VSMC) from a contractile state to one resembling macrophages. In human and mouse atherosclerotic plaques, it has become appreciated that ≈40% of cells classified as macrophages by histological markers may be of VSMC origin. Therefore, we sought to gain insight into the molecular regulation of this clinically relevant process. APPROACH AND RESULTS VSMC of mouse (or human) origin were incubated with cyclodextrin-cholesterol complexes for 72 hours, at which time the expression at the protein and mRNA levels of contractile-related proteins was reduced and of macrophage markers increased. Concurrent was downregulation of miR-143/145, which positively regulate the master VSMC differentiation transcription factor myocardin. Mechanisms were further probed in mouse VSMC. Maintaining the expression of myocardin or miR-143/145 prevented and reversed phenotypic changes caused by cholesterol loading. Reversal was also seen when cholesterol efflux was stimulated after loading. Notably, despite expression of macrophage markers, bioinformatic analyses showed that cholesterol-loaded cells remained closer to the VSMC state, consistent with impairment in classical macrophage functions of phagocytosis and efferocytosis. In apoE-deficient atherosclerotic plaques, cells positive for VSMC and macrophage markers were found lining the cholesterol-rich necrotic core. CONCLUSIONS Cholesterol loading of VSMC converts them to a macrophage-appearing state by downregulating the miR-143/145-myocardin axis. Although these cells would be classified by immunohistochemistry as macrophages in human and mouse plaques, their transcriptome and functional properties imply that their contributions to atherogenesis would not be those of classical macrophages.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Binding Sites
- Cell Lineage
- Cell Transdifferentiation
- Cholesterol/metabolism
- Cholesterol, HDL/metabolism
- Coculture Techniques
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Humans
- Jurkat Cells
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Phagocytosis
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Sterol Regulatory Element Binding Protein 2/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
Collapse
Affiliation(s)
- Yuliya Vengrenyuk
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Hitoo Nishi
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Xiaochun Long
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Mireille Ouimet
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nazir Savji
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Fernando O Martinez
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Courtney P Cassella
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Stephen A Ramsey
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|
22
|
Mateo J, Benito M, España S, Sanz J, Jiménez-Borreguero J, Fuster V, Ruiz-Cabello J. Magnetic Resonance Imaging of the Atherosclerotic Mouse Aorta. Methods Mol Biol 2015; 1339:387-394. [PMID: 26445806 DOI: 10.1007/978-1-4939-2929-0_29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Plaque development has been extensively studied using magnetic resonance imaging (MRI) in animal models of rapidly progressing atherosclerosis, such as apolipoprotein E-knockout (apoE-KO) mice. Preclinical MRI plays a significant role in the study of experimental atherosclerosis. Currently, MRI is capable of detecting luminal narrowing, plaque size, and morphology with high accuracy and reproducibility, providing reliable measurements of plaque burden. Therefore, MRI offers a noninvasive approach to serially monitor the progression of the disease. Compared with other imaging modalities, MRI appears to have the greatest potential for plaque characterization, through the use of multiple contrast weightings (e.g., T1, T2, and proton density). Here, we illustrate a standard procedure to image the aorta of atherosclerotic mice using noninvasive MRI.
Collapse
Affiliation(s)
- Jesús Mateo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Marina Benito
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Samuel España
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Javier Sanz
- The Zena and Michael A. Wiener Cardiovascular Institute/Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine, New York, NY, USA
| | - Jesús Jiménez-Borreguero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Hospital Universitario de La Princesa, Madrid, Spain
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- The Zena and Michael A. Wiener Cardiovascular Institute/Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine, New York, NY, USA
| | - Jesús Ruiz-Cabello
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
23
|
Angsana J, Chen J, Smith S, Xiao J, Wen J, Liu L, Haller CA, Chaikof EL. Syndecan-1 modulates the motility and resolution responses of macrophages. Arterioscler Thromb Vasc Biol 2014; 35:332-40. [PMID: 25550207 DOI: 10.1161/atvbaha.114.304720] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Syndecan-1 (Sdc-1) is a member of a family of cell surface proteoglycans, which has been reported to participate in the regulation of events relevant to tissue repair and chronic injury responses, including cell-substrate interactions, matrix remodeling, and cell migration. In this study, we report the functional significance of Sdc-1 in polarized macrophage populations and its role in adhesion and motility events relevant to resolution of the inflammatory program. APPROACH AND RESULTS Macrophage Sdc-1 expression is associated with differentiated M2 macrophages with high intrinsic motility, and Sdc-1 deficiency is characterized by impaired migration and enhanced adhesion. Leukocyte infiltration and emigration were examined in a thioglycollate-induced model of peritonitis in Sdc-1(+/+) and Sdc-1(-/-) mice. Although the infiltration of inflammatory cells was similar in both cohorts, a significant delay in the lymphatic clearance of Sdc-1(-/-) macrophages was observed. Moreover, we observed enhanced inflammation and greater burden of atherosclerotic plaques in ApoE(-/-)Sdc-1(-/-) mice maintained on a Western diet. CONCLUSIONS These results demonstrate that defective motility in Sdc-1(-/-) macrophages promotes a persistent inflammatory state with relevance to the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Julianty Angsana
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Jiaxuan Chen
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Sumona Smith
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Jiantao Xiao
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Jing Wen
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Liying Liu
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.)
| | - Carolyn A Haller
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.).
| | - Elliot L Chaikof
- From the Department of Bioengineering, Georgia Institute of Technology, Atlanta (J.A.); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.C., L.L., C.A.H., E.L.C.); Department of Surgery, Emory University, Atlanta, GA (S.S., J.X., J.W.); and Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA (E.L.C.).
| |
Collapse
|
24
|
Ramsey SA, Vengrenyuk Y, Menon P, Podolsky I, Feig JE, Aderem A, Fisher EA, Gold ES. Epigenome-guided analysis of the transcriptome of plaque macrophages during atherosclerosis regression reveals activation of the Wnt signaling pathway. PLoS Genet 2014; 10:e1004828. [PMID: 25474352 PMCID: PMC4256277 DOI: 10.1371/journal.pgen.1004828] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 10/15/2014] [Indexed: 11/19/2022] Open
Abstract
We report the first systems biology investigation of regulators controlling arterial plaque macrophage transcriptional changes in response to lipid lowering in vivo in two distinct mouse models of atherosclerosis regression. Transcriptome measurements from plaque macrophages from the Reversa mouse were integrated with measurements from an aortic transplant-based mouse model of plaque regression. Functional relevance of the genes detected as differentially expressed in plaque macrophages in response to lipid lowering in vivo was assessed through analysis of gene functional annotations, overlap with in vitro foam cell studies, and overlap of associated eQTLs with human atherosclerosis/CAD risk SNPs. To identify transcription factors that control plaque macrophage responses to lipid lowering in vivo, we used an integrative strategy – leveraging macrophage epigenomic measurements – to detect enrichment of transcription factor binding sites upstream of genes that are differentially expressed in plaque macrophages during regression. The integrated analysis uncovered eight transcription factor binding site elements that were statistically overrepresented within the 5′ regulatory regions of genes that were upregulated in plaque macrophages in the Reversa model under maximal regression conditions and within the 5′ regulatory regions of genes that were upregulated in the aortic transplant model during regression. Of these, the TCF/LEF binding site was present in promoters of upregulated genes related to cell motility, suggesting that the canonical Wnt signaling pathway may be activated in plaque macrophages during regression. We validated this network-based prediction by demonstrating that β-catenin expression is higher in regressing (vs. control group) plaques in both regression models, and we further demonstrated that stimulation of canonical Wnt signaling increases macrophage migration in vitro. These results suggest involvement of canonical Wnt signaling in macrophage emigration from the plaque during lipid lowering-induced regression, and they illustrate the discovery potential of an epigenome-guided, systems approach to understanding atherosclerosis regression. Atherosclerosis, a progressive accumulation of lipid-rich plaque within arteries, is an inflammatory disease in which the response of macrophages (a key cell type of the innate immune system) to plasma lipoproteins plays a central role. In humans, the goal of significantly reducing already-established plaque through drug treatments, including statins, remains elusive. In mice, atherosclerosis can be reversed by experimental manipulations that lower circulating lipid levels. A common feature of many regression models is that macrophages transition to a less inflammatory state and emigrate from the plaque. While the molecular regulators that control these responses are largely unknown, we hypothesized that by integrating global measurements of macrophage gene expression in regressing plaques with measurements of the macrophage chromatin landscape, we could identify key molecules that control macrophage responses to the lowering of circulating lipid levels. Our systems biology analysis of plaque macrophages yielded a network in which the Wnt signaling pathway emerged as a candidate upstream regulator. Wnt signaling is known to affect both inflammation and the ability of macrophages to migrate from one location to another, and our targeted validation studies provide evidence that Wnt signaling is increased in plaque macrophages during regression. Our findings both demonstrate the power of a systems approach to uncover candidate regulators of regression and to identify a potential new therapeutic target.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Epigenesis, Genetic/drug effects
- Epigenesis, Genetic/physiology
- Female
- Gene Expression Profiling
- Genome/drug effects
- Hypolipidemic Agents/pharmacology
- Hypolipidemic Agents/therapeutic use
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microarray Analysis
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Receptors, LDL/genetics
- Remission Induction
- Transcriptome/drug effects
- Wnt Signaling Pathway/drug effects
- Wnt Signaling Pathway/genetics
Collapse
Affiliation(s)
- Stephen A. Ramsey
- Department of Biomedical Sciences and School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, United States of America
| | - Yuliya Vengrenyuk
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Prashanthi Menon
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Irina Podolsky
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Jonathan E. Feig
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Alan Aderem
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Edward A. Fisher
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
- * E-mail: (EAF); (ESG)
| | - Elizabeth S. Gold
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- * E-mail: (EAF); (ESG)
| |
Collapse
|
25
|
Quantification of plaque lipids in the aortic root of ApoE-deficient mice by 3D DIXON magnetic resonance imaging in an ex vivo model. Eur Radiol 2014; 25:736-44. [DOI: 10.1007/s00330-014-3456-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 09/03/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022]
|
26
|
Genetic experimental preparations for studying atherosclerosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014. [PMID: 24751424 DOI: 10.1016/b978-0-12-386930-2.00001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Atherosclerosis is a pathological process with several inputs (biological, chemical, physiological, and others) interacting slowly over a lifetime leading to coronary artery disease, significant morbidity, and a limited lifespan. Over the past two decades, biologists have used experimental preparations from cells, animals, and man to understand the biology of atherosclerosis. Much has been discovered but our use of the standard gene-targeted experimental preparations is now nearing its limit. Better preparations to answer the remaining questions in the field of atherosclerosis biology are needed.
Collapse
|
27
|
Björkegren JLM, Hägg S, Talukdar HA, Foroughi Asl H, Jain RK, Cedergren C, Shang MM, Rossignoli A, Takolander R, Melander O, Hamsten A, Michoel T, Skogsberg J. Plasma cholesterol-induced lesion networks activated before regression of early, mature, and advanced atherosclerosis. PLoS Genet 2014; 10:e1004201. [PMID: 24586211 PMCID: PMC3937269 DOI: 10.1371/journal.pgen.1004201] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/09/2014] [Indexed: 12/21/2022] Open
Abstract
Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr−/−Apob100/100Mttpflox/floxMx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions. The main underlying cause of heart attacks and strokes is atherosclerosis. One strategy to prevent these often deadly clinical events is therefore either to slow atherosclerosis progression or better, induce regression of atherosclerotic plaques making them more stable. Plasma cholesterol lowering (PCL) is the most efficient way to induce atherosclerosis regression but sometimes fails to do so. In our study, we used a mouse model with elevated LDL cholesterol levels, similar to humans who develop early atherosclerosis, and a genetic switch to lower plasma cholesterol at any time during atherosclerosis progression. In this model, we examined atherosclerosis gene expression and regression in response to PCL at three different stages of atherosclerosis progression. PCL led to complete regression in mice with early lesions but was incomplete in mice with mature and advanced lesions, indicating that early prevention with PCL in individuals with increased risk for heart attack or stroke would be particularly useful. In addition, by inferring PCL-responsive gene networks in early, mature and advanced atherosclerotic lesions, we identified key drivers specific for regression of early (PPARG), mature (MLL5) and advanced (SRSF10/XRN2) atherosclerosis. These key drivers should be interesting therapeutic targets to enhance PCL-mediated regression of atherosclerosis.
Collapse
Affiliation(s)
- Johan L. M. Björkegren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Cardiovascular Genomics Group, Department of Pathological Anatomy and Forensic Medicine, University of Tartu, Tartu, Estonia
- Institute for Genomics and Multi-scale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Husain A. Talukdar
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rajeev K. Jain
- Cardiovascular Genomics Group, Department of Pathological Anatomy and Forensic Medicine, University of Tartu, Tartu, Estonia
| | - Cecilia Cedergren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ming-Mei Shang
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Aránzazu Rossignoli
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rabbe Takolander
- Department of Surgery, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Hypertension & Cardiovascular Disease, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Anders Hamsten
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tom Michoel
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Josefin Skogsberg
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
28
|
Feig JE, Hewing B, Smith JD, Hazen SL, Fisher EA. High-density lipoprotein and atherosclerosis regression: evidence from preclinical and clinical studies. Circ Res 2014; 114:205-13. [PMID: 24385513 DOI: 10.1161/circresaha.114.300760] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
High-density lipoprotein (HDL) particles transport (among other molecules) cholesterol (HDL-C). In epidemiological studies, plasma HDL-C levels have an inverse relationship to the risk of atherosclerotic cardiovascular disease. It has been assumed that this reflects the protective functions of HDL, which include their ability to promote cholesterol efflux. Yet, several recent pharmacological and genetic studies have failed to demonstrate that increased plasma levels of HDL-C resulted in decreased cardiovascular disease risk, giving rise to a controversy regarding whether plasma levels of HDL-C reflect HDL function, or that HDL is even as protective as assumed. The evidence from preclinical and (limited) clinical studies shows that HDL can promote the regression of atherosclerosis when the levels of functional particles are increased from endogenous or exogenous sources. The data show that regression results from a combination of reduced plaque lipid and macrophage contents, as well as from a reduction in its inflammatory state. Although more research will be needed regarding basic mechanisms and to establish that these changes translate clinically to reduced cardiovascular disease events, that HDL can regress plaques suggests that the recent trial failures do not eliminate HDL from consideration as an atheroprotective agent but rather emphasizes the important distinction between HDL function and plasma levels of HDL-C.
Collapse
Affiliation(s)
- Jonathan E Feig
- From the Departments of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Vascular Biology Program, New York University School of Medicine, New York, NY (J.E.F., B.H., E.A.F.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH (J.D.S., S.L.H.). J.E.F. is currently affiliated with Department of Medicine (Cardiology), Mount Sinai School of Medicine, New York, NY. B.H. is currently affiliated with Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
29
|
Feig JE. Regression of atherosclerosis: insights from animal and clinical studies. Ann Glob Health 2013; 80:13-23. [PMID: 24751561 DOI: 10.1016/j.aogh.2013.12.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/25/2014] [Accepted: 03/15/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Based on studies that date back to the 1920s, regression and stabilization of atherosclerosis in humans has gone from just a dream to one that is achievable. Review of the literature indicates that the successful attempts at regression generally applied robust measures to improve plasma lipoprotein profiles. Examples include extensive lowering of plasma concentrations of atherogenic apolipoprotein B and enhancement of reverse cholesterol transport from atheromata to the liver. FINDINGS Possible mechanisms responsible for lesion shrinkage include decreased retention of atherogenic apolipoprotein B within the arterial wall, efflux of cholesterol and other toxic lipids from plaques, emigration of lesional foam cells out of the arterial wall, and influx of healthy phagocytes that remove necrotic debris as well as other components of the plaque. This review will highlight the role key players such as LXR, HDL and CCR7 have in mediating regression. CONCLUSION Although much progress has been made, there are many unanswered questions. There is, therefore, a clear need for preclinical and clinical testing of new agents expected to facilitate atherosclerosis regression with the hope that additional mechanistic insights will allow further progress.
Collapse
Affiliation(s)
- Jonathan E Feig
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Medical Center, New York, NY.
| |
Collapse
|
30
|
Zeng HT, Fu YC, Yu W, Lin JM, Zhou L, Liu L, Wang W. SIRT1 prevents atherosclerosis via liver‑X‑receptor and NF‑κB signaling in a U937 cell model. Mol Med Rep 2013; 8:23-8. [PMID: 23652462 DOI: 10.3892/mmr.2013.1460] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/08/2013] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis is a chronic immunoinflammatory disease associated with blood lipid disorders. Previous studies in mice have demonstrated that liver X receptor (LXR)‑ATP‑binding cassette (ABC) A1/ABCG1/C‑C chemokine receptor type 7 (CCR7) and nuclear factor κB (NF‑κB) signaling pathways are important for atherosclerotic plaque formation. In addition, Sirtuin 1 (SIRT1) has been reported as a key regulator in the protection from risk of atherosclerosis. However, the exact mechanism by which SIRT1 prevents atherosclerosis remains largely unknown. To explore the possible mechanisms, the expression of SIRT1 and the association between SIRT1, LXR and NF‑κB in the process of foam cell formation was investigated in an in vitro human mononuclear U937 cell line. Monocyte‑derived foam cells were induced by palmitate and Ox‑LDL treatment. Oil Red O staining revealed an accumulation of a large number of lipid droplets in foam cells. Results of reverse transcription polymerase chain reaction (RT-PCR) revealed that SIRT1 expression was downregulated during foam cell formation. In addition, the expression of LXRα and its targets, ABCA1, ABCG1 and CCR7, were downregulated. However, NF‑κB and its targets, tumor necrosis factor α (TNFα) and interleukin (IL)‑1β, were upregulated in foam cells. Following activation of SIRT1 by SRT1720, the expression of LXRα and its targets increased, whereas expression of NF‑κB and its targets decreased. Furthermore, the formation of foam cells was blocked. The SIRT1 inhibitor, nicotinamide, was found to eliminate the effects of SRT1720. Results of the present study indicate that SIRT1 may prevent the formation and progression of atherosclerosis by enhancing the LXR‑ABCA1/ABCG1/CCR7 and inhibiting the NF‑κB pathways.
Collapse
Affiliation(s)
- Hai-Tao Zeng
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | | | | | | | | | | | | |
Collapse
|
31
|
Barazza A, Blachford C, Even-Or O, Joaquin VA, Briley-Saebo KC, Chen W, Jiang XC, Mulder WJM, Cormode DP, Fayad ZA, Fisher EA. The complex fate in plasma of gadolinium incorporated into high-density lipoproteins used for magnetic imaging of atherosclerotic plaques. Bioconjug Chem 2013; 24:1039-48. [PMID: 23617731 DOI: 10.1021/bc400105j] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We have previously reported enhancing the imaging of atherosclerotic plaques in mice using reconstituted high density lipoproteins (HDL) as nanocarriers for the MRI contrast agent gadolinium (Gd). This study focuses on the underlying mechanisms of Gd delivery to atherosclerotic plaques. HDL, LDL, and VLDL particles containing Gd chelated to phosphatidyl ethanolamine (DTPA-DMPE) and a lipidic fluorophore were used to demonstrate the transfer of Gd-phospholipids among plasma lipoproteins in vitro and in vivo. To determine the basis of this transfer, the roles of phospholipid transfer protein (PLTP) and lipoprotein lipase (LpL) in mediating the migration of Gd-DTPA-DMPE among lipoproteins were investigated. The results indicated that neither was an important factor, suggesting that spontaneous transfer of Gd-DTPA-DMPE was the most probable mechanism. Finally, two independent mouse models were used to quantify the relative contributions of HDL and LDL reconstituted with Gd-DTPA-DMPE to plaque imaging enhancement by MR. Both sets of results suggested that Gd-DTPA-DMPE originally associated with LDL was about twice as effective as that injected in the form of Gd-HDL, and that some of Gd-HDL's effectiveness in vivo is indirect through transfer of the imaging agent to LDL. In conclusion, the fate of Gd-DTPA-DMPE associated with a particular type of lipoprotein is complex, and includes its transfer to other lipoprotein species that are then cleared from the plasma into tissues.
Collapse
Affiliation(s)
- Alessandra Barazza
- Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, Smilow 7, 522 First Avenue, New York, New York 10016, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chen W, Cormode DP, Vengrenyuk Y, Herranz B, Feig JE, Klink A, Mulder WJM, Fisher EA, Fayad ZA. Collagen-specific peptide conjugated HDL nanoparticles as MRI contrast agent to evaluate compositional changes in atherosclerotic plaque regression. JACC Cardiovasc Imaging 2013; 6:373-84. [PMID: 23433925 PMCID: PMC3653172 DOI: 10.1016/j.jcmg.2012.06.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 05/14/2012] [Accepted: 06/29/2012] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This study sought to develop magnetic resonance contrast agents based on high-density lipoprotein (HDL) nanoparticles to noninvasively visualize intraplaque macrophages and collagen content in mouse atherosclerotic plaques. BACKGROUND Macrophages and collagen are important intraplaque components that play central roles in plaque progression and/or regression. In a Reversa mouse model, plaque regression with compositional changes (from high macrophage, low collagen to low macrophage, high collagen) can be induced. METHODS This study labeled HDL nanoparticles with amphiphilic gadolinium chelates to enable target-specific imaging of intraplaque macrophages. To render HDL nanoparticles specific for the extracellular matrix, labeled HDL nanoparticles were functionalized with collagen-specific EP3533 peptides (EP3533-HDL) via poly(ethylene glycol) spacers embedded in the HDL lipid layers. The association of nanoparticles with collagen was examined in vitro by optical methods. The in vivo magnetic resonance efficacy of these nanoparticles was evaluated in a Reversa mouse model of atherosclerosis regression. Ex vivo confocal microscopy was applied to corroborate the in vivo findings and to evaluate the fate of the different HDL nanoparticles. RESULTS All nanoparticles had similar sizes (10 ± 2 nm) and longitudinal relaxivity r1 (9 ± 1 s(-1) mmol/l(-1)). EP3533-HDL showed strong association with collagen in vitro. After 28 days of plaque regression in Reversa mice, EP3533-HDL showed significantly increased (p < 0.05) in vivo magnetic resonance signal in aortic vessel walls (normalized enhancement ratio [NERw] = 85 ± 25%; change of contrast-to-noise ratio [ΔCNRw] = 17 ± 5) compared with HDL (NERw = -7 ± 23%; ΔCNRw = -2 ± 4) and nonspecific control EP3612-HDL (NERw = 4 ± 24%; ΔCNRw = 1 ± 6) at 24 h after injection. Ex vivo confocal images revealed the colocalization of EP3533-HDL with collagen. Immunohistostaining analysis confirmed the changes of collagen and macrophage contents in the aortic vessel walls after regression. CONCLUSIONS This study shows that the HDL nanoparticle platform can be modified to monitor in vivo plaque compositional changes in a regression environment, which will facilitate understanding plaque regression and the search for therapeutic interventions.
Collapse
Affiliation(s)
- Wei Chen
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
| | - David P. Cormode
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
| | - Yuliya Vengrenyuk
- Department of Medicine, Leon H. Charney Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York, USA
| | - Beatriz Herranz
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
- Department of Epidemiology, Atherothrombosis and Imaging. Fundacion Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Jonathan E Feig
- Department of Medicine, Leon H. Charney Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York, USA
| | - Ahmed Klink
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
- Paris Cardiovascular Research Center, INSERM Assistance Publique-Hopitaux de Paris, Hopital Europeen Georges Pompidou, Paris, France
| | - Willem J. M. Mulder
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | - Edward A. Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York, USA
| | - Zahi A. Fayad
- Translational and Molecular Imaging Institute, Departments of Radiology and Medicine, Mount Sinai School of Medicine, New York, New York USA
| |
Collapse
|
33
|
Wanschel A, Seibert T, Hewing B, Ramkhelawon B, Ray TD, van Gils JM, Rayner KJ, Feig JE, O'Brien ER, Fisher EA, Moore KJ. Neuroimmune guidance cue Semaphorin 3E is expressed in atherosclerotic plaques and regulates macrophage retention. Arterioscler Thromb Vasc Biol 2013; 33:886-93. [PMID: 23430613 DOI: 10.1161/atvbaha.112.300941] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The persistence of myeloid-derived cells in the artery wall is a characteristic of advanced atherosclerotic plaques. However, the mechanisms by which these cells are retained are poorly understood. Semaphorins, a class of neuronal guidance molecules, play a critical role in vascular patterning and development, and recent studies suggest that they may also have immunomodulatory functions. The present study evaluates the expression of Semaphorin 3E (Sema3E) in settings relevant to atherosclerosis and its contribution to macrophage accumulation in plaques. APPROACH AND RESULTS Immunofluorescence staining of Sema3E, and its receptor PlexinD1, demonstrated their expression in macrophages of advanced atherosclerotic lesions of Apoe(-/-) mice. Notably, in 2 different mouse models of atherosclerosis regression, Sema3E mRNA was highly downregulated in plaque macrophages, coincident with a reduction in plaque macrophage content and an enrichment in markers of reparative M2 macrophages. In vitro, Sema3E mRNA was highly expressed in inflammatory M1 macrophages and in macrophages treated with physiological drivers of plaque progression and inflammation, such as oxidized low-density lipoprotein and hypoxia. To explore mechanistically how Sema3E affects macrophage behavior, we treated macrophages with recombinant protein in the presence/absence of chemokines, including CCL19, a chemokine implicated in the egress of macrophages from atherosclerotic plaques. Sema3E blocked actin polymerization and macrophage migration stimulated by the chemokines, suggesting that it may immobilize these cells in the plaque. CONCLUSIONS Sema3E is upregulated in macrophages of advanced plaques, is dynamically regulated by multiple atherosclerosis-relevant factors, and acts as a negative regulator of macrophage migration, which may promote macrophage retention and chronic inflammation in vivo.
Collapse
Affiliation(s)
- Amarylis Wanschel
- Marc and Ruti Bell Vascular Biology and Disease Program, Department of Medicine, Leon H. Charney Division of Cardiology, New YorkUniversity School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Feig JE, Feig JL. Macrophages, dendritic cells, and regression of atherosclerosis. Front Physiol 2012; 3:286. [PMID: 22934038 PMCID: PMC3429058 DOI: 10.3389/fphys.2012.00286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/29/2012] [Indexed: 01/15/2023] Open
Abstract
Atherosclerosis is the number one cause of death in the Western world. It results from the interaction between modified lipoproteins and cells such as macrophages, dendritic cells (DCs), T cells, and other cellular elements present in the arterial wall. This inflammatory process can ultimately lead to the development of complex lesions, or plaques, that protrude into the arterial lumen. Ultimately, plaque rupture and thrombosis can occur leading to the clinical complications of myocardial infarction or stroke. Although each of the cell types plays roles in the pathogenesis of atherosclerosis, the focus of this review will be primarily on the macrophages and DCs. The role of these two cell types in atherosclerosis is discussed, with a particular emphasis on their involvement in atherosclerosis regression.
Collapse
Affiliation(s)
- Jonathan E Feig
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Medical Center NY, USA
| | | |
Collapse
|
35
|
Regression of atherosclerosis is characterized by broad changes in the plaque macrophage transcriptome. PLoS One 2012; 7:e39790. [PMID: 22761902 PMCID: PMC3384622 DOI: 10.1371/journal.pone.0039790] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 05/29/2012] [Indexed: 01/19/2023] Open
Abstract
We have developed a mouse model of atherosclerotic plaque regression in which an atherosclerotic aortic arch from a hyperlipidemic donor is transplanted into a normolipidemic recipient, resulting in rapid elimination of cholesterol and monocyte-derived macrophage cells (CD68+) from transplanted vessel walls. To gain a comprehensive view of the differences in gene expression patterns in macrophages associated with regressing compared with progressing atherosclerotic plaque, we compared mRNA expression patterns in CD68+ macrophages extracted from plaque in aortic aches transplanted into normolipidemic or into hyperlipidemic recipients. In CD68+ cells from regressing plaque we observed that genes associated with the contractile apparatus responsible for cellular movement (e.g. actin and myosin) were up-regulated whereas genes related to cell adhesion (e.g. cadherins, vinculin) were down-regulated. In addition, CD68+ cells from regressing plaque were characterized by enhanced expression of genes associated with an anti-inflammatory M2 macrophage phenotype, including arginase I, CD163 and the C-lectin receptor. Our analysis suggests that in regressing plaque CD68+ cells preferentially express genes that reduce cellular adhesion, enhance cellular motility, and overall act to suppress inflammation.
Collapse
|
36
|
Jaffer FA. Shining light and illuminating murine atherosclerosis via optical coherence tomography. Arterioscler Thromb Vasc Biol 2012; 32:1068-9. [PMID: 22517362 DOI: 10.1161/atvbaha.112.246439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Molecular Imaging of Macrophages in Atherosclerosis. CURRENT CARDIOVASCULAR IMAGING REPORTS 2012. [DOI: 10.1007/s12410-011-9118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
38
|
Feig JE, Shang Y, Rotllan N, Vengrenyuk Y, Wu C, Shamir R, Torra IP, Fernandez-Hernando C, Fisher EA, Garabedian MJ. Statins promote the regression of atherosclerosis via activation of the CCR7-dependent emigration pathway in macrophages. PLoS One 2011; 6:e28534. [PMID: 22163030 PMCID: PMC3232231 DOI: 10.1371/journal.pone.0028534] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 11/10/2011] [Indexed: 12/16/2022] Open
Abstract
HMG-CoA reductase inhibitors (statins) decrease atherosclerosis by lowering low-density-lipoprotein cholesterol. Statins are also thought to have additional anti-atherogenic properties, yet defining these non-conventional modes of statin action remains incomplete. We have previously developed a novel mouse transplant model of atherosclerosis regression in which aortic segments from diseased donors are placed into normolipidemic recipients. With this model, we demonstrated the rapid loss of CD68+ cells (mainly macrophages) in plaques through the induction of a chemokine receptor CCR7-dependent emigration process. Because the human and mouse CCR7 promoter contain Sterol Response Elements (SREs), we hypothesized that Sterol Regulatory Element Binding Proteins (SREBPs) are involved in increasing CCR7 expression and through this mechanism, statins would promote CD68+ cell emigration from plaques. We examined whether statin activation of the SREBP pathway in vivo would induce CCR7 expression and promote macrophage emigration from plaques. We found that western diet-fed apoE-/- mice treated with either atorvastatin or rosuvastatin led to a substantial reduction in the CD68+ cell content in the plaques despite continued hyperlipidemia. We also observed a significant increase in CCR7 mRNA in CD68+ cells from both the atorvastatin and rosuvastatin treated mice associated with emigration of CD68+ cells from plaques. Importantly, CCR7-/-/apoE-/- double knockout mice failed to display a reduction in CD68+ cell content upon statin treatment. Statins also affected the recruitment of transcriptional regulatory proteins and the organization of the chromatin at the CCR7 promoter to increase the transcriptional activity. Statins promote the beneficial remodeling of plaques in diseased mouse arteries through the stimulation of the CCR7 emigration pathway in macrophages. Therefore, statins may exhibit some of their clinical benefits by not only retarding the progression of atherosclerosis, but also accelerating its regression.
Collapse
Affiliation(s)
- Jonathan E. Feig
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Yueting Shang
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Noemi Rotllan
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Yuliya Vengrenyuk
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Chaowei Wu
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Raanan Shamir
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Ines Pineda Torra
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Carlos Fernandez-Hernando
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Edward A. Fisher
- Division of Cardiology (Marc and Ruti Bell Program in Vascular Biology), Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (EAF); (MJG)
| | - Michael J. Garabedian
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (EAF); (MJG)
| |
Collapse
|
39
|
Dietrich T, Hucko T, Schneemann C, Neumann M, Menrad A, Willuda J, Atrott K, Stibenz D, Fleck E, Graf K, Menssen HD. Local delivery of IL-2 reduces atherosclerosis via expansion of regulatory T cells. Atherosclerosis 2011; 220:329-36. [PMID: 22062588 DOI: 10.1016/j.atherosclerosis.2011.09.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 09/20/2011] [Accepted: 09/27/2011] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Recent studies indicate that regulatory T cells (Tregs) attenuate murine atherosclerosis. Since interleukin (IL)-2 induces Tregs proliferation, we tested the impact of L19-IL2, a fusion antibody specific to extra-domain B of fibronectin (ED-B) containing an active human IL-2 molecule, in experimental atherosclerosis. METHODS AND RESULTS L19-IL2 or appropriate controls were given intravenously to 6 month old Western diet-fed apoE(-/-) mice on day 1, 3, and 5. Human IL-2 was detected on day 7 within atherosclerotic plaques of L19-IL2-treated mice, and magnetic resonance imaging of the plaques showed a significant adventitial gadolinium enhancement on day 7 and 13, suggesting microvascular leakage as a result of the pharmacodynamic activity of L19-IL2. Treatment with L19-IL2 significantly reduced the size of pre-established atherosclerotic plaques at the thoracic aorta (Sudan III stained area) and in the aortic root area (microscopic, morphometric analysis) on day 7 as compared to controls (L19, D1.3-IL2, NaCl) as well as compared to baseline (day 0). Tregs markers Foxp3 and CTLA4 were highly increased in plaques after L19-IL2 treatment compared to controls (p<0.01), whereas the macrophage marker Mac3 was significantly reduced (p<0.03). Co-treatment with IL-2-receptor blocking antibody PC61 abrogated L19-IL2-induced plaque reduction compared with IgG control (p<0.03). CONCLUSION L19-IL2 delivers functional IL-2 to pre-established atherosclerotic plaques of WD-fed apoE(-/-) mice resulting in significant plaque size reduction mediated by local Tregs.
Collapse
Affiliation(s)
- Thore Dietrich
- Department of Medicine-Cardiology, Deutsches Herzzentrum, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pourcet B, Feig JE, Vengrenyuk Y, Hobbs AJ, Kepka-Lenhart D, Garabedian MJ, Morris SM, Fisher EA, Pineda-Torra I. LXRα regulates macrophage arginase 1 through PU.1 and interferon regulatory factor 8. Circ Res 2011; 109:492-501. [PMID: 21757649 DOI: 10.1161/circresaha.111.241810] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Activation of liver X receptors (LXRs) inhibits the progression of atherosclerosis and promotes regression of existing lesions. In addition, LXRα levels are high in regressive plaques. Macrophage arginase 1 (Arg1) expression is inversely correlated with atherosclerosis progression and is markedly decreased in foam cells within the lesion. OBJECTIVE To investigate LXRα regulation of Arg1 expression in cultured macrophages and atherosclerotic regressive lesions. METHODS AND RESULTS We found that Arg1 expression is enhanced in CD68+ cells from regressive versus progressive lesions in a murine aortic arch transplant model. In cultured macrophages, ligand-activated LXRα markedly enhances basal and interleukin-4-induced Arg1 mRNA and protein expression as well as promoter activity. This LXRα-enhanced Arg1 expression correlates with a reduction in nitric oxide levels. Moreover, Arg1 expression within regressive atherosclerotic plaques is LXRα-dependent, as enhanced expression of Arg1 in regressive lesions is impaired in LXRα-deficient CD68+ cells. LXRα does not bind to the Arg1 promoter but instead promotes the interaction between PU.1 and interferon regulatory factor (IRF)8 transcription factors and induces their binding of a novel composite element. Accordingly, knockdown of either IRF8 or PU.1 strongly impairs LXRα regulation of Arg1 expression in macrophage cells. Finally, we demonstrate that LXRα binds the IRF8 locus and its activation increases IRF8 mRNA and protein levels in these cells. CONCLUSIONS This work implicates Arg1 in atherosclerosis regression and identifies LXRα as a novel regulator of Arg1 and IRF8 in macrophages. Furthermore, it provides a unique molecular mechanism by which LXRα regulates macrophage target gene expression through PU.1 and IRF8.
Collapse
Affiliation(s)
- Benoit Pourcet
- Centre for Clinical Pharmacology, Division of Medicine, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
HDL promotes rapid atherosclerosis regression in mice and alters inflammatory properties of plaque monocyte-derived cells. Proc Natl Acad Sci U S A 2011; 108:7166-71. [PMID: 21482781 DOI: 10.1073/pnas.1016086108] [Citation(s) in RCA: 248] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
HDL cholesterol (HDL-C) plasma levels are inversely related to cardiovascular disease risk. Previous studies have shown in animals and humans that HDL promotes regression of atherosclerosis. We hypothesized that this was related to an ability to promote the loss of monocyte-derived cells (CD68(+), primarily macrophages and macrophage foam cells) from plaques. To test this hypothesis, we used an established model of atherosclerosis regression in which plaque-bearing aortic arches from apolipoprotein E-deficient (apoE(-/-)) mice (low HDL-C, high non-HDL-C) were transplanted into recipient mice with differing levels of HDL-C and non-HDL-C: C57BL6 mice (normal HDL-C, low non-HDL-C), apoAI(-/-) mice (low HDL-C, low non-HDL-C), or apoE(-/-) mice transgenic for human apoAI (hAI/apoE(-/-); normal HDL-C, high non-HDL-C). Remarkably, despite persistent elevated non-HDL-C in hAI/apoE(-/-) recipients, plaque CD68(+) cell content decreased by >50% by 1 wk after transplantation, whereas there was little change in apoAI(-/-) recipient mice despite hypolipidemia. The decreased content of plaque CD68(+) cells after HDL-C normalization was associated with their emigration and induction of their chemokine receptor CCR7. Furthermore, in CD68(+) cells laser-captured from the plaques, normalization of HDL-C led to decreased expression of inflammatory factors and enrichment of markers of the M2 (tissue repair) macrophage state. Again, none of these beneficial changes were observed in the apoAI(-/-) recipients, suggesting a major requirement for reverse cholesterol transport for the beneficial effects of HDL. Overall, these results establish HDL as a regulator in vivo of the migratory and inflammatory properties of monocyte-derived cells in mouse atherosclerotic plaques, and highlight the phenotypic plasticity of these cells.
Collapse
|
42
|
Feig JE, Parathath S, Rong JX, Mick SL, Vengrenyuk Y, Grauer L, Young SG, Fisher EA. Reversal of hyperlipidemia with a genetic switch favorably affects the content and inflammatory state of macrophages in atherosclerotic plaques. Circulation 2011; 123:989-98. [PMID: 21339485 PMCID: PMC3131163 DOI: 10.1161/circulationaha.110.984146] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND We previously showed that the progression of atherosclerosis in the Reversa mouse (Ldlr(-/-Apob100/100Mttpfl/fl) Mx1Cre(+/+)) was arrested when the hyperlipidemia was normalized by inactivating the gene for microsomal triglyceride transfer protein. Here, we tested whether atherosclerosis would regress if the lipid levels were reduced after advanced plaques formed. METHODS AND RESULTS Reversa mice were fed an atherogenic diet for 16 weeks. Plasma lipid levels were then reduced. Within 2 weeks, this reduction led to decreased monocyte-derived (CD68(+)) cells in atherosclerotic plaques and was associated with emigration of these cells out of plaques. In addition, the fall in lipid levels was accompanied by lower plaque lipid content and by reduced expression in plaque CD68(+) cells of inflammatory genes and higher expression of genes for markers of antiinflammatory M2 macrophages. Plaque composition was affected more than plaque size, with the decreased content of lipid and CD68(+) cells balanced by a higher content of collagen. When the reduced lipid level was combined with the administration of pioglitazone to simulate the clinical aggressive lipid management and proliferator-activated receptor-γ agonist treatment, the rate of depletion of plaque CD68(+) cells was unaffected, but there was a further increase in their expression of antiinflammatory macrophage markers. CONCLUSION The Reversa mouse is a new model of atherosclerosis regression. After lipid lowering, favorable changes in plaque composition were independent of changes in size. In addition, plaque CD68(+) cells became less inflammatory, an effect enhanced by treatment with pioglitazone.
Collapse
Affiliation(s)
- Jonathan E. Feig
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - Sajesh Parathath
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - James X. Rong
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - Stephanie L. Mick
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - Yuliya Vengrenyuk
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - Lisa Grauer
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
| | - Stephen G. Young
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California Los Angeles
| | - Edward A. Fisher
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine
- Department of Cardiovascular Medicine, University of Oxford
| |
Collapse
|
43
|
Feig JE, Pineda-Torra I, Sanson M, Bradley MN, Vengrenyuk Y, Bogunovic D, Gautier EL, Rubinstein D, Hong C, Liu J, Wu C, van Rooijen N, Bhardwaj N, Garabedian M, Tontonoz P, Fisher EA. LXR promotes the maximal egress of monocyte-derived cells from mouse aortic plaques during atherosclerosis regression. J Clin Invest 2011; 120:4415-24. [PMID: 21041949 DOI: 10.1172/jci38911] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 09/15/2010] [Indexed: 12/20/2022] Open
Abstract
We have previously shown that mouse atherosclerosis regression involves monocyte-derived (CD68+) cell emigration from plaques and is dependent on the chemokine receptor CCR7. Concurrent with regression, mRNA levels of the gene encoding LXRalpha are increased in plaque CD68+ cells, suggestive of a functional relationship between LXR and CCR7. To extend these results, atherosclerotic Apoe-/- mice sufficient or deficient in CCR7 were treated with an LXR agonist, resulting in a CCR7-dependent decrease in plaque CD68+ cells. To test the requirement for LXR for CCR7-dependent regression, we transplanted aortic arches from atherosclerotic Apoe-/- mice, or from Apoe-/- mice with BM deficiency of LXRalpha or LXRbeta, into WT recipients. Plaques from both LXRalpha and LXRbeta-deficient Apoe-/- mice exhibited impaired regression. In addition, the CD68+ cells displayed reduced emigration and CCR7 expression. Using an immature DC line, we found that LXR agonist treatment increased Ccr7 mRNA levels. This increase was blunted when LXRalpha and LXRbeta levels were reduced by siRNAs. Moreover, LXR agonist treatment of primary human immature DCs resulted in functionally significant upregulation of CCR7. We conclude that LXR is required for maximal effects on plaque CD68+ cell expression of CCR7 and monocyte-derived cell egress during atherosclerosis regression in mice.
Collapse
Affiliation(s)
- Jonathan E Feig
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of atherosclerotic cardiovascular disease and loss of renal parenchyma accelerates atherosclerosis in animal models. Macrophages are central to atherogenesis because they regulate cholesterol traffic and inflammation in the arterial wall. CKD influences macrophage behavior at multiple levels, rendering them proatherogenic. Even at normal creatinine levels, macrophages from uninephrectomized Apoe(-/-) mice are enriched in cholesterol owing to downregulation of cholesterol transporter ATP-binding cassette subfamily A member 1 levels and activation of nuclear factor κB, which leads to impaired cholesterol efflux. Interestingly, treatment with an angiotensin-II-receptor blocker (ARB) improves these effects. Moreover, atherosclerotic aortas from Apoe(-/-) mice transplanted into renal-ablated normocholesterolemic recipients show plaque progression and increased macrophage content instead of the substantial regression seen in recipient mice with intact kidneys. ARBs reduce atherosclerosis development in mice with partial renal ablation. These results, combined with the clinical benefits of angiotensin-converting-enzyme (ACE) inhibitors and ARBs in patients with CKD, suggest an important role for the angiotensin system in the enhanced susceptibility to atherosclerosis seen across the spectrum of CKD. The role of macrophages could explain why these therapies may be effective in end-stage renal disease, one of the few conditions in which statins show no clinical benefit.
Collapse
Affiliation(s)
- Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, 383 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37332-6300, USA
| | | | | |
Collapse
|
45
|
Accurate assessment of carotid artery stenosis in atherosclerotic mice using accelerated high-resolution 3D magnetic resonance angiography. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2010; 24:9-18. [PMID: 20862514 DOI: 10.1007/s10334-010-0227-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/04/2010] [Accepted: 09/06/2010] [Indexed: 10/19/2022]
Abstract
OBJECT High-resolution magnetic resonance angiography (MRA) enables non-invasive detection and longitudinal monitoring of atherosclerosis in mouse models of human disease. However, MRA is hampered by long acquisition times putting high demands on the physiological stability of the animal. Therefore, we evaluated the feasibility of accelerated MRA using the parallel imaging technique SENSE with regard to both lesion detection and quantification. MATERIALS AND METHODS MRA acquisitions of supra-aortic vessels were performed in ApoE (-/-) mice that have been shown to develop atherosclerotic plaques. Findings obtained from accelerated data sets were compared to fully sampled reference data sets and histology. RESULTS Our results revealed only minor differences in detecting vascular lesions for data collections accelerated by factors of up to 3.3 using a four-element coil array. For vessels with a mean lumen diameter of 500 μm, morphometry of stenotic lesions revealed no substantial deviations from reference (fully sampled) data for all investigated acceleration factors. For the highest acceleration factor of 3.3, an average deviation of the degree of stenosis of 4.9 ± 3.6% was found. Common carotid stenoses assessed by in vivo MRA displayed a good correlation with histological analyses (slope of linear regression = 0.97, R (2) = 0.98). CONCLUSION According to the results of this work, we have demonstrated the feasibility and accuracy of accelerated high-resolution 3D ToF MRA in mice suitable for detailed depiction of mouse supra-aortic vessels and amenable to non-invasive quantification of small atherosclerotic lesions.
Collapse
|
46
|
Aorta transplantation in young apolipoprotein E-deficient mice: Possible model for studies on regression of atherosclerotic lesions? Open Med (Wars) 2010. [DOI: 10.2478/s11536-010-0006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractSyngeneic transplantation of murine aorta segments with advanced atherosclerotic lesions in defined recipients is a valuable model for regression studies. To date, this model has not been used to study the regression of initial atherosclerotic lesions. The aim of this study was to evaluate a microsurgical technique of syngeneic heterotopic transplantation of the thoracic aorta of young apolipoprotein E-deficient (ApoE-/-) mice to the abdominal aorta of wild-type recipients. Stereological quantification methods were tested in order to assess changes in structure and volume of the aortic wall including the involvement of immune cells in changes of the atherosclerotic lesions. The animals were euthanised one month after surgery and histological analysis including stereological quantification of changes in both the grafts and adjacent aorta segments was performed. The overall survival rate of the recipients was 62.5%. No regression of initial atherosclerotic lesion was achieved and neointima formation and elastin degradation prevailed in all transplanted specimens. The volume of the arteriosclerotic lesions was higher (p<0.001) and elastin length density was lower (p<0.001) in transplanted ApoE-/- samples as compared to adjacent segments. In transplanted grafts, T- and B-lymphocytes, macrophages and neutrophilic granulocytes formed non-random clusters within the vessel wall and they were colocalised with the sutures. The reproducibility of the promising regression model was derogated in young mice by the striking dependence of the results upon the operation technique. Stereological assessment has proven to be accurate, correct and reproducible; it has provided us with robust quantitative estimates, which can be achieved with a reasonable effort.
Collapse
|
47
|
Inflammatory cell recruitment in cardiovascular disease: murine models and potential clinical applications. Clin Sci (Lond) 2010; 118:641-55. [PMID: 20210786 DOI: 10.1042/cs20090488] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is the pathological process that underlies the development of cardiovascular disease, a leading cause of mortality. Atherosclerotic plaque formation is driven by the recruitment of inflammatory monocytes into the artery wall, their differentiation into macrophages and the subsequent transformation of macrophages into cholesterol-laden foam cells. Models of hypercholesterolaemia such as the ApoE (apolipoprotein E)-/- mouse and the application of transgenic technologies have allowed us to undertake a thorough dissection of the cellular and molecular biology of the atherosclerotic disease process. Murine models have emphasized the central role of inflammation in atherogenesis and have been instrumental in the identification of adhesion molecules that support monocyte recruitment, scavenger receptors that facilitate cholesterol uptake by macrophages and other macrophage activation receptors. The study of mice deficient in multiple members of the chemokine family, and their receptors, has shown that chemokines play a critical role in promoting atherosclerotic plaque formation. In the present review, we will discuss novel therapeutic avenues for the treatment of cardiovascular disease that derive directly from our current understanding of atherogenesis gained in experimental animal models.
Collapse
|
48
|
Moderate kidney disease inhibits atherosclerosis regression. Atherosclerosis 2009; 210:57-62. [PMID: 19931862 DOI: 10.1016/j.atherosclerosis.2009.10.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2009] [Revised: 10/10/2009] [Accepted: 10/16/2009] [Indexed: 12/29/2022]
Abstract
UNLABELLED Chronic kidney disease (CKD) accelerates cardiovascular disease. The mechanisms that explain this independent, excess risk associated with CKD have not been fully elucidated. OBJECTIVES We propose that impaired regression of atherosclerosis in renal disease represents a novel risk factor for the heightened morbidity and mortality and resistance to treatment observed in patients with CKD. METHODS AND RESULTS Using a transplant model to study atherosclerosis regression, we transplanted atheromatous aortic segments generated in Apolipoprotein E knock-out (ApoE(-/-)) mice, into either control or moderately uremic, normolipidemic, wild-type mice. In non-uremic mice, lesions regressed 55%, whereas lesions in uremic mice increased in size by 17% (p<0.01 for control vs. uremic). The lesions in uremic mice were also characterized by a greater presence of macrophages (36,300 microm(2) vs. 12,600 microm(2), p<0.01). This finding was despite upregulation of chemokine receptor 7 (CCR7), normally a migration factor, in uremic lesion macrophages. Gene expression analysis of lesion macrophages showed relative down-regulation of serum response factor (SRF) target genes in the uremic group, consistent with impaired CCR7 signaling. CONCLUSION Moderate kidney disease inhibits regression of atherosclerosis in a mouse transplant model. This inhibition may be a result of impaired CCR7 signaling.
Collapse
|
49
|
Deguchi JO, Yamazaki H, Aikawa E, Aikawa M. Chronic Hypoxia Activates the Akt and β-Catenin Pathways in Human Macrophages. Arterioscler Thromb Vasc Biol 2009; 29:1664-70. [DOI: 10.1161/atvbaha.109.194043] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Macrophage activation contributes importantly to the pathogenesis of inflammatory diseases including atherosclerosis. Macrophages exist chronically under moderate hypoxia (2% to 5% O
2
) in inflamed tissues such as atherosclerotic plaques. However, macrophage phenotypes in such environments remain incompletely understood. This study tested the hypothesis that chronic moderate hypoxia induces macrophage activation and explored the underlying mechanisms.
Methods and Results—
We cultured primary human macrophages derived from peripheral blood monocytes in moderate hypoxia (2% O
2
tension) or normoxia (21% O
2
) for 10 days. Moderate hypoxia did not affect macrophage differentiation assessed via expression levels of scavenger receptor A. Chronic moderate hypoxia, but not normoxia, activated Akt and inactivated GSK-3β, a negative effector of Akt, thus allowing nuclear translocation of β-catenin. 2% O
2
tension increased accumulation of hypoxia-inducible factors 1α (HIF-1α) transiently at 3 to 5 days. Hypoxia induced mRNA expression of the β-catenin-associated genes: MMP-7, CD44, and c-Myc. RNAi of TCF7L2, a cofactor of β-catenin, suppressed MMP-7 expression induced by hypoxia. Inhibition of Akt phosphorylation with LY294002 abolished hypoxia-induced GSK-3β inactivation, β-catenin activation, and MMP-7 expression. Macrophages under hypoxia were more resistant for oxLDL-induced apoptosis. Moreover, phospho-Akt colocalized with MMP-7 and CD44 expression in macrophages of human atherosclerotic plaques.
Conclusions—
Chronic moderate hypoxia induces macrophage activation via the Akt and β-catenin pathways, providing new insight into the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Jun-o Deguchi
- From the Cardiovascular Division, Department of Medicine (J.D., H.Y., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; and the Department of Radiology (E.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass
| | - Hiroyuki Yamazaki
- From the Cardiovascular Division, Department of Medicine (J.D., H.Y., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; and the Department of Radiology (E.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass
| | - Elena Aikawa
- From the Cardiovascular Division, Department of Medicine (J.D., H.Y., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; and the Department of Radiology (E.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass
| | - Masanori Aikawa
- From the Cardiovascular Division, Department of Medicine (J.D., H.Y., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; and the Department of Radiology (E.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass
| |
Collapse
|
50
|
Dietrich T, Hucko T, Bourayou R, Jahnke C, Paetsch I, Atrott K, Stawowy P, Gräfe M, Klein C, Schnackenburg B, Fleck E, Graf K. High resolution magnetic resonance imaging in atherosclerotic mice treated with ezetimibe. Int J Cardiovasc Imaging 2009; 25:827-36. [DOI: 10.1007/s10554-009-9487-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 07/27/2009] [Indexed: 11/24/2022]
|