101
|
Targeted theranostic photoactivation on atherosclerosis. J Nanobiotechnology 2021; 19:338. [PMID: 34689768 PMCID: PMC8543964 DOI: 10.1186/s12951-021-01084-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
Background Photoactivation targeting macrophages has emerged as a therapeutic strategy for atherosclerosis, but limited targetable ability of photosensitizers to the lesions hinders its applications. Moreover, the molecular mechanistic insight to its phototherapeutic effects on atheroma is still lacking. Herein, we developed a macrophage targetable near-infrared fluorescence (NIRF) emitting phototheranostic agent by conjugating dextran sulfate (DS) to chlorin e6 (Ce6) and estimated its phototherapeutic feasibility in murine atheroma. Also, the phototherapeutic mechanisms of DS-Ce6 on atherosclerosis were investigated. Results The phototheranostic agent DS-Ce6 efficiently internalized into the activated macrophages and foam cells via scavenger receptor-A (SR-A) mediated endocytosis. Customized serial optical imaging-guided photoactivation of DS-Ce6 by light illumination reduced both atheroma burden and inflammation in murine models. Immuno-fluorescence and -histochemical analyses revealed that the photoactivation of DS-Ce6 produced a prominent increase in macrophage-associated apoptotic bodies 1 week after laser irradiation and induced autophagy with Mer tyrosine-protein kinase expression as early as day 1, indicative of an enhanced efferocytosis in atheroma. Conclusion Imaging-guided DS-Ce6 photoactivation was able to in vivo detect inflammatory activity in atheroma as well as to simultaneously reduce both plaque burden and inflammation by harmonic contribution of apoptosis, autophagy, and lesional efferocytosis. These results suggest that macrophage targetable phototheranostic nanoagents will be a promising theranostic strategy for high-risk atheroma. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01084-z.
Collapse
|
102
|
Potor L, Hendrik Z, Patsalos A, Katona É, Méhes G, Póliska S, Csősz É, Kalló G, Komáromi I, Combi Z, Posta N, Sikura KÉ, Pethő D, Oros M, Vereb G, Tóth C, Gergely P, Nagy L, Balla G, Balla J. Oxidation of Hemoglobin Drives a Proatherogenic Polarization of Macrophages in Human Atherosclerosis. Antioxid Redox Signal 2021; 35:917-950. [PMID: 34269613 PMCID: PMC8905252 DOI: 10.1089/ars.2020.8234] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Aim: The aim of our study was to explore the pathophysiologic role of oxidation of hemoglobin (Hb) to ferrylHb in human atherosclerosis. Results: We observed a severe oxidation of Hb to ferrylHb in complicated atherosclerotic lesions of carotid arteries with oxidative changes of the globin moieties, detected previously described oxidation hotspots in Hb (β1Cys93; β1Cys112; β2Cys112) and identified a novel oxidation hotspot (α1Cys104). After producing a monoclonal anti-ferrylHb antibody, ferrylHb was revealed to be localized extracellularly and also internalized by macrophages in the human hemorrhagic complicated lesions. We demonstrated that ferrylHb is taken up via phagocytosis as well as CD163 receptor-mediated endocytosis and then transported to lysosomes involving actin polymerization. Internalization of ferrylHb was accompanied by upregulation of heme oxygenase-1 and H-ferritin and accumulation of iron within lysosomes as a result of heme/iron uptake. Importantly, macrophages exposed to ferrylHb in atherosclerotic plaques exhibited a proinflammatory phenotype, as reflected by elevated levels of IL-1β and TNF-α. To find further signatures of ferrylHb in complicated lesions, we performed RNA-seq analysis on biopsies from patients who underwent endarterectomies. RNA-seq analysis demonstrated that human complicated lesions had a unique transcriptomic profile different from arteries and atheromatous plaques. Pathways affected in complicated lesions included gene changes associated with phosphoinositide 3-kinase (PI3K) signaling, lipid transport, tissue remodeling, and vascularization. Targeted analysis of gene expression associated with calcification, apoptosis, and hemolytic-specific clusters indicated an increase in the severity of complicated lesions compared with atheroma. A 39% overlap in the differential gene expression profiles of human macrophages exposed to ferrylHb and the complicated lesion profiles was uncovered. Among these 547 genes, we found inflammatory, angiogenesis, and iron metabolism gene clusters regulated in macrophages. Innovation and Conclusion: We conclude that oxidation of Hb to ferrylHb contributes to the progression of atherosclerosis via polarizing macrophages into a proatherogenic phenotype. Antioxid. Redox Signal. 35, 917-950.
Collapse
Affiliation(s)
- László Potor
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hendrik
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.,Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andreas Patsalos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Éva Katona
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Komáromi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Combi
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Niké Posta
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Katalin Éva Sikura
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Melinda Oros
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Tóth
- Division of Vascular Surgery, Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - György Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
103
|
Verberk SGS, Kuiper KL, Lauterbach MA, Latz E, Van den Bossche J. The multifaceted therapeutic value of targeting ATP-citrate lyase in atherosclerosis. Trends Mol Med 2021; 27:1095-1105. [PMID: 34635427 DOI: 10.1016/j.molmed.2021.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/10/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
ATP-citrate lyase (Acly) is the target of the new class low-density lipoprotein-cholesterol (LDL-C)-lowering drug bempedoic acid (BA). Acly is a key metabolic enzyme synthesizing acetyl-CoA as the building block of cholesterol and fatty acids. Treatment with BA lowers circulating lipid levels and reduces systemic inflammation, suggesting a dual benefit of this drug for atherosclerosis therapy. Recent studies have shown that targeting Acly in macrophages can attenuate inflammatory responses and decrease atherosclerotic plaque vulnerability. Therefore, it could be beneficial to extend the application of Acly inhibition from solely lipid-lowering by liver-specific inhibition to also targeting macrophages in atherosclerosis. Here, we outline the possibilities of targeting Acly and describe the future needs to translate these findings to the clinic.
Collapse
Affiliation(s)
- Sanne G S Verberk
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kirsten L Kuiper
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mario A Lauterbach
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany
| | - Jan Van den Bossche
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
104
|
Buono MF, Slenders L, Wesseling M, Hartman RJG, Monaco C, den Ruijter HM, Pasterkamp G, Mokry M. The changing landscape of the vulnerable plaque: a call for fine-tuning of preclinical models. Vascul Pharmacol 2021; 141:106924. [PMID: 34607015 DOI: 10.1016/j.vph.2021.106924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022]
Abstract
For decades, the pathological definition of the vulnerable plaque led to invaluable insights into the mechanisms that underlie myocardial infarction and stroke. Beyond plaque rupture, other mechanisms, such as erosion, may elicit thrombotic events underlining the complexity and diversity of the atherosclerotic disease. Novel insights, based on single-cell transcriptomics and other "omics" methods, provide tremendous opportunities in the ongoing search for cell-specific determinants that will fine-tune the description of the thrombosis prone lesion. It coincides with an increasing awareness that knowledge on lesion characteristics, cell plasticity and clinical presentation of ischemic cardiovascular events have shifted over the past decades. This shift correlates with an observed changes of cell composition towards phenotypical stabilizing of human plaques. These stabilization features and mechanisms are directly mediated by the cells present in plaques and can be mimicked in vitro via primary plaque cells derived from human atherosclerotic tissues. In addition, the rapidly evolving of sequencing technologies identify many candidate genes and molecular mechanisms that may influence the risk of developing an atherosclerotic thrombotic event - which bring the next challenge in sharp focus: how to translate these cell-specific insights into tangible functional and translational discoveries?
Collapse
Affiliation(s)
- Michele F Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marian Wesseling
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robin J G Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
105
|
Chaudhary S, Patidar A, Dhiman A, Chaubey GK, Dilawari R, Talukdar S, Modanwal R, Raje M. Exposure of a specific pleioform of multifunctional glyceraldehyde 3-phosphate dehydrogenase initiates CD14-dependent clearance of apoptotic cells. Cell Death Dis 2021; 12:892. [PMID: 34593755 PMCID: PMC8482365 DOI: 10.1038/s41419-021-04168-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Rapid clearance of apoptotic cells by phagocytes is crucial for organogenesis, tissue homeostasis, and resolution of inflammation. This process is initiated by surface exposure of various 'eat me' ligands. Though phosphatidylserine (PS) is the best recognized general recognition ligand till date, recent studies have shown that PS by itself is not sufficient for clearance of apoptotic cells. In this study, we have identified a specific pleioform of GAPDH (Glyceraldehyde 3-phosphate dehydrogenase) that functions as an 'eat me' signal on apoptotic cell surface. This specific form of GAPDH which is exposed on surface of apoptotic cells was found to interact with CD14 present on plasma membrane of phagocytes leading to their engulfment. This is the first study demonstrating the novel interaction between multifunctional GAPDH and the phagocytic receptor CD14 resulting in apoptotic cell clearance (efferocytosis).
Collapse
Affiliation(s)
- Surbhi Chaudhary
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Anil Patidar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Asmita Dhiman
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | | | - Rahul Dilawari
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Sharmila Talukdar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Radheshyam Modanwal
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Manoj Raje
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India.
| |
Collapse
|
106
|
Shimizu-Albergine M, Basu D, Kanter JE, Kramer F, Kothari V, Barnhart S, Thornock C, Mullick AE, Clouet-Foraison N, Vaisar T, Heinecke JW, Hegele RA, Goldberg IJ, Bornfeldt KE. CREBH normalizes dyslipidemia and halts atherosclerosis in diabetes by decreasing circulating remnant lipoproteins. J Clin Invest 2021; 131:e153285. [PMID: 34491909 DOI: 10.1172/jci153285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function mutations in the transcription factor CREB3L3 (CREBH) associate with severe hypertriglyceridemia in humans. CREBH is believed to lower plasma triglycerides by augmenting the action of lipoprotein lipase (LPL). However, by using a mouse model of type 1 diabetes mellitus (T1DM), we found that greater liver expression of active CREBH normalized both elevated plasma triglycerides and cholesterol. Residual triglyceride-rich lipoprotein (TRL) remnants were enriched in apolipoprotein E (APOE) and impoverished in APOC3, an apolipoprotein composition indicative of increased hepatic clearance. The underlying mechanism was independent of LPL as CREBH reduced both triglycerides and cholesterol in LPL-deficient mice. Instead, APOE was critical for CREBH's ability to lower circulating remnant lipoproteins because it failed to reduce TRL cholesterol in Apoe-/- mice. Importantly, humans with CREB3L3 loss-of-function mutations exhibited increased levels of remnant lipoproteins that were deprived of APOE. Recent evidence suggests that impaired clearance of TRL remnants promotes cardiovascular disease in patients with T1DM. Consistently, we found that hepatic expression of CREBH prevented the progression of diabetes-accelerated atherosclerosis. Our results support the proposal that CREBH acts through an APOE-dependent pathway to increase hepatic clearance of remnant lipoproteins. They also implicate elevated levels of remnants in the pathogenesis of atherosclerosis in T1DM.
Collapse
Affiliation(s)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Medical Center, New York, United States of America
| | - Jenny E Kanter
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Farah Kramer
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Vishal Kothari
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Shelley Barnhart
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Carissa Thornock
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Adam E Mullick
- Cardiovascular Disease Research, Ionis Pharmaceuticals, Inc., Carlsbad, United States of America
| | | | - Tomas Vaisar
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, United States of America
| | - Robert A Hegele
- Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, London, Canada
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Medical Center, New York, United States of America
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington, Seattle, United States of America
| |
Collapse
|
107
|
Nakagawa K, Tanaka M, Hahm TH, Nguyen HN, Matsui T, Chen YX, Nakashima Y. Accumulation of Plasma-Derived Lipids in the Lipid Core and Necrotic Core of Human Atheroma: Imaging Mass Spectrometry and Histopathological Analyses. Arterioscler Thromb Vasc Biol 2021; 41:e498-e511. [PMID: 34470476 DOI: 10.1161/atvbaha.121.316154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kazunori Nakagawa
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| | - Mitsuru Tanaka
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Tae-Hun Hahm
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Huu-Nghi Nguyen
- Department of Science and International Collaboration, Institute for Research and Development of Organic Products, Hanoi, Vietnam (H.-N.N.)
| | - Toshiro Matsui
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Yong-Xiang Chen
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan.,Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Alberta, Canada (Y.-X.C.)
| | - Yutaka Nakashima
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| |
Collapse
|
108
|
Recognition of Oxidized Lipids by Macrophages and Its Role in Atherosclerosis Development. Biomedicines 2021; 9:biomedicines9080915. [PMID: 34440119 PMCID: PMC8389651 DOI: 10.3390/biomedicines9080915] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a multifactorial chronic disease that has a prominent inflammatory component. Currently, atherosclerosis is regarded as an active autoimmune process that involves both innate and adaptive immune pathways. One of the drivers of this process is the presence of modified low-density lipoprotein (LDL). For instance, lipoprotein oxidation leads to the formation of oxidation-specific epitopes (OSE) that can be recognized by the immune cells. Macrophage response to OSEs is recognized as a key trigger for initiation and a stimulator of progression of the inflammatory process in the arteries. At the same time, the role of oxidized LDL components is not limited to pro-inflammatory stimulation, but includes immunoregulatory effects that can have protective functions. It is, therefore, important to better understand the complexity of oxidized LDL effects in atherosclerosis in order to develop new therapeutic approaches to correct the inflammatory and metabolic imbalance associated with this disorder. In this review, we discuss the process of oxidized LDL formation, mechanisms of OSE recognition by macrophages and the role of these processes in atherosclerosis.
Collapse
|
109
|
Gautier EL, Askia H, Murcy F, Yvan-Charvet L. Macrophage ontogeny and functional diversity in cardiometabolic diseases. Semin Cell Dev Biol 2021; 119:119-129. [PMID: 34229949 DOI: 10.1016/j.semcdb.2021.06.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/01/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
Macrophages are the dominant immune cell types in the adipose tissue, the liver or the aortic wall and they were originally believed to mainly derived from monocytes to fuel tissue inflammation in cardiometabolic diseases. However, over the last decade the identification of tissue resident macrophages (trMacs) from embryonic origin in these metabolic tissues has provided a breakthrough in the field forcing to better comprehend macrophage diversity during pathological states. Infiltrated monocyte-derived macrophages (moMacs), similar to trMacs, adapt to the local metabolic environment that eventually shapes their functions. In this review, we will summarize the emerging versatility of macrophages in cardiometabolic diseases with a focus in the control of adipose tissue, liver and large vessels homeostasis.
Collapse
Affiliation(s)
- Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMR-S 1166, Sorbonne Université, 75013 Paris, France.
| | - Haoussa Askia
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMR-S 1166, Sorbonne Université, 75013 Paris, France
| | - Florent Murcy
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| |
Collapse
|
110
|
Specialized Pro-Resolving Lipid Mediators in Neonatal Cardiovascular Physiology and Diseases. Antioxidants (Basel) 2021; 10:antiox10060933. [PMID: 34201378 PMCID: PMC8229722 DOI: 10.3390/antiox10060933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease remains a leading cause of mortality worldwide. Unresolved inflammation plays a critical role in cardiovascular diseases development. Specialized Pro-Resolving Mediators (SPMs), derived from long chain polyunsaturated fatty acids (LCPUFAs), enhances the host defense, by resolving the inflammation and tissue repair. In addition, SPMs also have anti-inflammatory properties. These physiological effects depend on the availability of LCPUFAs precursors and cellular metabolic balance. Most of the studies have focused on the impact of SPMs in adult cardiovascular health and diseases. In this review, we discuss LCPUFAs metabolism, SPMs, and their potential effect on cardiovascular health and diseases primarily focusing in neonates. A better understanding of the role of these SPMs in cardiovascular health and diseases in neonates could lead to the development of novel therapeutic approaches in cardiovascular dysfunction.
Collapse
|
111
|
Dead cell and debris clearance in the atherosclerotic plaque: Mechanisms and therapeutic opportunities to promote inflammation resolution. Pharmacol Res 2021; 170:105699. [PMID: 34087352 DOI: 10.1016/j.phrs.2021.105699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023]
Abstract
Phagocytic clearance of dead cells and debris is critical for inflammation resolution and maintenance of tissue homeostasis. Consequently, defective clearance of dead cells and debris is associated with initiation and exacerbation of several autoimmune disorders and chronic inflammatory diseases such as atherosclerosis. The progressive loss of dead cell clearance capacity within the atherosclerotic plaque leads to accumulation of necrotic cells, chronic non-resolving inflammation, and expansion of the necrotic core, which triggers atherosclerotic plaque rupture and clinical manifestation of acute thrombotic cardiovascular adverse events. In this review, we describe the fundamental molecular and cellular mechanisms of dead cell clearance and how it goes awry in atherosclerosis. Finally, we highlight novel therapeutic strategies that enhance dead cell and debris clearance within the atherosclerotic plaque to promote inflammation resolution and atherosclerotic plaque stabilization.
Collapse
|
112
|
Che X, Xiao Q, Song W, Zhang H, Sun B, Geng N, Tao Z, Shao Q, Pu J. Protective Functions of Liver X Receptor α in Established Vulnerable Plaques: Involvement of Regulating Endoplasmic Reticulum-Mediated Macrophage Apoptosis and Efferocytosis. J Am Heart Assoc 2021; 10:e018455. [PMID: 33969692 PMCID: PMC8200716 DOI: 10.1161/jaha.120.018455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Liver X receptor (LXR) belongs to the metabolic nuclear receptor superfamily, which plays a critical regulatory role in vascular physiology/pathology. However, effects of systemic LXR activation on established vulnerable plaques and the potential isotype‐specific role involved remain unclear. Methods and Results The 8‐week‐old male apolipoprotein E−/− mice went through carotid branch ligation and renal artery constriction, combined with a high‐fat diet. Plaques in the left carotid artery acquired vulnerable features 4 weeks later, confirmed by magnetic resonance imaging scans and histological analysis. From that time on, mice were injected intraperitoneally daily with PBS or GW3965 (10 mg/kg per day) for an additional 4 weeks. Treatment with LXR agonists reduced the lesion volume by 52.61%, compared with the vehicle group. More important, a profile of less intraplaque hemorrhage detection and necrotic core formation was found. These actions collectively attenuated the incidence of plaque rupture. Mechanistically, reduced lesional apoptosis, enhanced efferocytosis, and alleviated endoplasmic reticulum stress are involved in the process. Furthermore, genetic ablation of LXRα, but not LXRβ, blunted the protective effects of LXR on the endoplasmic reticulum stress–elicited C/EBP‐homologous protein pathway in peritoneal macrophages. In concert with the LXRα‐predominant role in vitro, activated LXR failed to stabilize vulnerable plaques and correct the acquired cellular anomalies in LXRα−/− apolipoprotein E−/− mice. Conclusions Our results revealed that LXRα mediates the capacity of LXR activation to stabilize vulnerable plaques and prevent plaque rupture via amelioration of macrophage endoplasmic reticulum stress, lesional apoptosis, and defective efferocytosis. These findings might expand the application scenarios of LXR therapeutics for atherosclerosis.
Collapse
Affiliation(s)
- Xinyu Che
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Qingqing Xiao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Wei Song
- Cardiovascular Department of Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Hengyuan Zhang
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Beibei Sun
- Department of Radiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Na Geng
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Zhenyu Tao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Qin Shao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Jun Pu
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
113
|
Shang J, Yang Q, Fan W, Chen Y, Tang D, Guo H, Xiong B, Huang S, Zhang XB. Probing Dynamic Features of Phagosome Maturation in Macrophage using Au@MnO x @SiO 2 Nanoparticles as pH-Sensitive Plasmonic Nanoprobes. Chem Asian J 2021; 16:1150-1156. [PMID: 33724702 DOI: 10.1002/asia.202100031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/13/2021] [Indexed: 11/05/2022]
Abstract
Phagosome maturation in macrophage is essential to the clearance of pathogenic materials in host defence but the dynamic features remain difficult to be measured in real time. Herein, we reported the multilayered Au@MnOx @SiO2 nanoparticle as a robust pH-sensitive plasmonic nanosensor for monitoring the dynamic acidification features over the phagosome maturation process in macrophage under darkfield microscopy. For this multilayered nanosensor, the gold nanoparticle core plays a role of signal reporter, the MnOx shell and the outmost SiO2 act as the sensing layer and the protecting layer, respectively. After subject to the acidic buffer solution, the MnOx layer in the multilayered nanoprobe could be decomposed rapidly, resulting in a remarkable spectral shift and color change under darkfield microscopy. We demonstrated this nanosensor for the investigation of single phagosome acidification dynamics by monitoring the color changes of nanoprobes after phagocytosis over time. The nanoprobes after phagocytosized in macrophage displayed a slight color change within the first hour and then cost several minutes to change from red to green in the next stage, indicating the phagosome undergoes a slow first and then fast acidification feature as well as a slow-to-fast acidification translation over the phagosome maturation process. Moreover, we validated that the slow-to-fast acidification translation was dependent on the activation of V-ATPase from the ATP depletion assay. We believed that this nanosensor is promising for studying the dynamic acidification features as well as disorders in phagosome maturation in phagocytic cells, which might provide valuable information for understanding the disease pathogenesis related to phagosome dysfunctions.
Collapse
Affiliation(s)
- Jinhui Shang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Qian Yang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Wenjun Fan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Yancao Chen
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Decui Tang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Haowei Guo
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Bin Xiong
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Shuangyan Huang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
114
|
Wu JH, Zhang L, Nepliouev I, Brian L, Huang T, Snow KP, Schickling BM, Hauser ER, Miller FJ, Freedman NJ, Stiber JA. Drebrin attenuates atherosclerosis by limiting smooth muscle cell transdifferentiation. Cardiovasc Res 2021; 118:772-784. [PMID: 33914863 DOI: 10.1093/cvr/cvab156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/27/2021] [Indexed: 01/01/2023] Open
Abstract
AIMS The F-actin-binding protein Drebrin inhibits smooth muscle cell (SMC) migration, proliferation and pro-inflammatory signaling. Therefore, we tested the hypothesis that Drebrin constrains atherosclerosis. METHODS AND RESULTS SM22-Cre+/Dbnflox/flox/Ldlr-/- (SMC-Dbn-/-/Ldlr-/-) and control mice (SM22-Cre+/Ldlr-/-, Dbnflox/flox/Ldlr-/-, and Ldlr-/-) were fed a Western diet for 14-20 weeks. Brachiocephalic arteries of SMC-Dbn-/-/Ldlr-/- mice exhibited 1.5- or 1.8-fold greater cross-sectional lesion area than control mice at 14 or 20 wk, respectively. Aortic atherosclerotic lesion surface area was 1.2-fold greater in SMC-Dbn-/-/Ldlr-/- mice. SMC-Dbn-/-/Ldlr-/- lesions comprised necrotic cores that were two-fold greater in size than those of control mice. Consistent with their bigger necrotic core size, lesions in SMC-Dbn-/- arteries also showed more transdifferentiation of SMCs to macrophage-like cells: 1.5- to 2.5-fold greater, assessed with BODIPY or with CD68, respectively. In vitro data were concordant: Dbn-/- SMCs had 1.7-fold higher levels of KLF4 and transdifferentiated to macrophage-like cells more readily than Dbnflox/flox SMCs upon cholesterol loading, as evidenced by greater up-regulation of CD68 and galectin-3. Adenovirally mediated Drebrin rescue produced equivalent levels of macrophage-like transdifferentiation in Dbn-/- and Dbnflox/flox SMCs. During early atherogenesis, SMC-Dbn-/-/Ldlr-/- aortas demonstrated 1.6-fold higher levels of reactive oxygen species than control mouse aortas. The 1.8-fold higher levels of Nox1 in Dbn-/- SMCs was reduced to WT levels with KLF4 silencing. Inhibition of Nox1 chemically or with siRNA produced equivalent levels of macrophage-like transdifferentiation in Dbn-/- and Dbnflox/flox SMCs. CONCLUSIONS We conclude that SMC Drebrin limits atherosclerosis by constraining SMC Nox1 activity and SMC transdifferentiation to macrophage-like cells. TRANSLATIONAL PERSPECTIVE Drebrin is abundantly expressed in vascular smooth muscle cells (SMCs) and is up-regulated in human atherosclerosis. A hallmark of atherosclerosis is the accumulation of foam cells that secrete pro-inflammatory cytokines and contribute to plaque instability. A large proportion of these foam cells in humans derive from SMCs. We found that SMC Drebrin limits atherosclerosis by reducing SMC transdifferentiation to macrophage-like foam cells in a manner dependent on Nox1 and KLF4. For this reason, strategies aimed at augmenting SMC Drebrin expression in atherosclerotic plaques may limit atherosclerosis progression and enhance plaque stability by bridling SMC-to-foam-cell transdifferentiation.
Collapse
Affiliation(s)
- Jiao-Hui Wu
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Lisheng Zhang
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Igor Nepliouev
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Leigh Brian
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Taiqin Huang
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Kamie P Snow
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Brandon M Schickling
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Elizabeth R Hauser
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Francis J Miller
- Department of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Neil J Freedman
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| | - Jonathan A Stiber
- Department of Medicine (Cardiology), Duke University Medical Center, 10 Duke Medicine Circle, Durham, NC 27710, USA
| |
Collapse
|
115
|
Kasikara C, Schilperoort M, Gerlach B, Xue C, Wang X, Zheng Z, Kuriakose G, Dorweiler B, Zhang H, Fredman G, Saleheen D, Reilly MP, Tabas I. Deficiency of macrophage PHACTR1 impairs efferocytosis and promotes atherosclerotic plaque necrosis. J Clin Invest 2021; 131:145275. [PMID: 33630758 DOI: 10.1172/jci145275] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Efferocytosis, the process through which apoptotic cells (ACs) are cleared through actin-mediated engulfment by macrophages, prevents secondary necrosis, suppresses inflammation, and promotes resolution. Impaired efferocytosis drives the formation of clinically dangerous necrotic atherosclerotic plaques, the underlying etiology of coronary artery disease (CAD). An intron of the gene encoding PHACTR1 contains rs9349379 (A>G), a common variant associated with CAD. As PHACTR1 is an actin-binding protein, we reasoned that if the rs9349379 risk allele G causes lower PHACTR1 expression in macrophages, it might link the risk allele to CAD via impaired efferocytosis. We show here that rs9349379-G/G was associated with lower levels of PHACTR1 and impaired efferocytosis in human monocyte-derived macrophages and human atherosclerotic lesional macrophages compared with rs9349379-A/A. Silencing PHACTR1 in human and mouse macrophages compromised AC engulfment, and Western diet-fed Ldlr-/- mice in which hematopoietic Phactr1 was genetically targeted showed impaired lesional efferocytosis, increased plaque necrosis, and thinner fibrous caps - all signs of vulnerable plaques in humans. Mechanistically, PHACTR1 prevented dephosphorylation of myosin light chain (MLC), which was necessary for AC engulfment. In summary, rs9349379-G lowered PHACTR1, which, by lowering phospho-MLC, compromised efferocytosis. Thus, rs9349379-G may contribute to CAD risk, at least in part, by impairing atherosclerotic lesional macrophage efferocytosis.
Collapse
Affiliation(s)
- Canan Kasikara
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Brennan Gerlach
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Chenyi Xue
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Ze Zheng
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Hanrui Zhang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical Center, Albany, New York, USA
| | - Danish Saleheen
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Muredach P Reilly
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.,Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.,Department of Physiology and Cellular Biophysics and.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
116
|
Phagocytic clearance of apoptotic, necrotic, necroptotic and pyroptotic cells. Biochem Soc Trans 2021; 49:793-804. [PMID: 33843978 PMCID: PMC8106503 DOI: 10.1042/bst20200696] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/27/2022]
Abstract
Although millions of cells in the human body will undergo programmed cell death each day, dying cells are rarely detected under homeostatic settings in vivo. The swift removal of dying cells is due to the rapid recruitment of phagocytes to the site of cell death which then recognise and engulf the dying cell. Apoptotic cell clearance - the engulfment of apoptotic cells by phagocytes - is a well-defined process governed by a series of molecular factors including 'find-me', 'eat-me', 'don't eat-me' and 'good-bye' signals. However, in recent years with the rapid expansion of the cell death field, the removal of other necrotic-like cell types has drawn much attention. Depending on the type of death, dying cells employ different mechanisms to facilitate engulfment and elicit varying functional impacts on the phagocyte, from wound healing responses to inflammatory cytokine secretion. Nevertheless, despite the mechanism of death, the clearance of dying cells is a fundamental process required to prevent the uncontrolled release of pro-inflammatory mediators and inflammatory disease. This mini-review summarises the current understandings of: (i) apoptotic, necrotic, necroptotic and pyroptotic cell clearance; (ii) the functional consequences of dying cell engulfment and; (iii) the outstanding questions in the field.
Collapse
|
117
|
Purine-rich element binding protein B attenuates the coactivator function of myocardin by a novel molecular mechanism of smooth muscle gene repression. Mol Cell Biochem 2021; 476:2899-2916. [PMID: 33743134 DOI: 10.1007/s11010-021-04117-1] [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: 07/29/2020] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
Myocardin is a potent transcriptional coactivator protein, which functions as the master regulator of vascular smooth muscle cell differentiation. The cofactor activity of myocardin is mediated by its physical interaction with serum response factor, a ubiquitously expressed transactivator that binds to CArG boxes in genes encoding smooth muscle-restricted proteins. Purine-rich element binding protein B (Purβ) represses the transcription of the smooth muscle α-actin gene (Acta2) in fibroblasts and smooth muscle cells by interacting with single-stranded DNA sequences flanking two 5' CArG boxes in the Acta2 promoter. In this study, the ability of Purβ to modulate the cofactor activity of myocardin was investigated using a combination of cellular and biochemical approaches. Results of smooth muscle gene promoter-reporter assays indicated that Purβ specifically inhibits the coactivator function of myocardin in a manner requiring the presence of all three single-stranded DNA binding domains in the Purβ homodimer. DNA binding analyses demonstrated that Purβ interacts with CArG-containing DNA elements with a much lower affinity compared to other purine-rich target sequences present in the Acta2 promoter. Co-immunoprecipitation and DNA pull-down assays revealed that Purβ associates with myocardin and serum response factor when free or bound to duplex DNA containing one or more CArG boxes. Functional analysis of engineered Purβ point mutants identified several amino acid residues essential for suppression of myocardin activity. Collectively, these findings suggest an inhibitory mechanism involving direct protein-protein interaction between the homodimeric Purβ repressor and the myocardin-serum response factor-CArG complex.
Collapse
|
118
|
Canet-Soulas E, Bessueille L, Mechtouff L, Magne D. The Elusive Origin of Atherosclerotic Plaque Calcification. Front Cell Dev Biol 2021; 9:622736. [PMID: 33768090 PMCID: PMC7985066 DOI: 10.3389/fcell.2021.622736] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
It has been known for decades or even centuries that arteries calcify as they age. Vascular calcification probably affects all adults, since virtually all have atherosclerotic plaques: an accumulation of lipids, inflammatory cells, necrotic debris, and calcium phosphate crystals. A high vascular calcium score is associated with a high cardiovascular mortality risk, and relatively recent data suggest that even microcalcifications that form in early plaques may destabilize plaques and trigger a cardiovascular event. If the cellular and molecular mechanisms of plaque calcification have been relatively well characterized in mice, human plaques appear to calcify through different mechanisms that remain obscure. In this context, we will first review articles reporting the location and features of early calcifications in human plaques and then review the articles that explored the mechanisms though which human and mouse plaques calcify.
Collapse
Affiliation(s)
- Emmanuelle Canet-Soulas
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laurence Bessueille
- ICBMS, CNRS, INSA Lyon, CPE, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laura Mechtouff
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Stroke Department, Hospices Civils de Lyon, Lyon, France
| | - David Magne
- ICBMS, CNRS, INSA Lyon, CPE, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
119
|
Impact of myeloid RIPK1 gene deletion on atherogenesis in ApoE-deficient mice. Atherosclerosis 2021; 322:51-60. [PMID: 33706083 DOI: 10.1016/j.atherosclerosis.2021.02.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/23/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Targeting macrophage death is a promising strategy for stabilizing atherosclerotic plaques. Recently, necroptosis was identified as a form of regulated necrosis in atherosclerosis. Receptor-interacting serine/threonine-protein kinase (RIPK)1 is an upstream regulator of RIPK3, which is a crucial kinase for necroptosis induction. We aimed to investigate the impact of myeloid-specific RIPK1 gene deletion on atherogenesis. METHODS RIPK1F/FLysM-Cre+ApoE-/- and RIPK1+/+LysM-Cre+ApoE-/- mice were fed a western-type diet (WD) for 16 or 24 weeks to induce plaque formation. RESULTS After 16 weeks WD, plaque area and percentage necrosis in RIPK1F/FLysM-Cre+ApoE-/- mice were significantly decreased as compared to plaques of RIPK1+/+LysM-Cre+ApoE-/- mice. Moreover, plaques of RIPK1F/FLysM-Cre+ApoE-/- mice showed more apoptosis and a decreased macrophage content. After 24 weeks WD, plaque size and percentage necrosis were no longer different between the two groups. Free apoptotic cells strongly accumulated in plaques of RIPK1F/FLysM-Cre+ApoE-/- mice. In addition to apoptosis, necroptosis was upregulated in plaques of RIPK1F/FLysM-Cre+ApoE-/- mice. In vitro, TNF-α triggered apoptosis in RIPK1F/FLysM-Cre+ApoE-/-, but not in RIPK1+/+LysM-Cre+ApoE-/- macrophages. Moreover, RIPK1F/FLysM-Cre+ApoE-/- macrophages were not protected against RIPK3-dependent necroptosis. CONCLUSIONS The impact of myeloid RIPK1 gene deletion depends on the stage of atherogenesis. At 16 weeks WD, myeloid RIPK1 gene deletion resulted in increased apoptosis, thereby slowing down plaque progression. However, despite decreased macrophage content, plaque and necrotic core size were no longer reduced after 24 weeks of WD, most likely due to the accumulation of free apoptotic and necroptotic cells.
Collapse
|
120
|
Macrophage apoptosis using alendronate in targeted nanoarchaeosomes. Eur J Pharm Biopharm 2021; 160:42-54. [PMID: 33440242 DOI: 10.1016/j.ejpb.2021.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Nanoarchaeosomes are non-hydrolysable nanovesicles made of archaeolipids, naturally functionalised with ligand for scavenger receptor class 1. We hypothesized that nitrogenate bisphosphonate alendronate (ALN) loaded nanoarchaeosomes (nanoarchaeosomes(ALN)) may constitute more efficient macrophage targeted apoptotic inducers than ALN loaded nanoliposomes (nanoliposomes (ALN)). To that aim, ALN was loaded in cholesterol containing (nanoARC-chol(ALN)) or not (nanoARC(ALN)) nanoarchaeosomes. Nanoarchaeosomes(ALN) (220-320 nm sized, ~ -40 mV ξ potential, 38-50 μg ALN/mg lipid ratio) displayed higher structural stability than nanoliposomes(ALN) of matching size and ξ potential, retaining most of ALN against a 1/200 folds dilution. The cytotoxicity of nanoARC(ALN) on J774A.1 cells, resulted > 30 folds higher than free ALN and nanoliposomes(ALN) and was reduced by cholesterol in nanoARC-chol(ALN). Devoid of ALN, nanoARC-chol was non-cytotoxic, exhibited pronounced anti-inflammatory activity on J774.1 cells, strongly reducing reactive oxygen species (ROS) and IL-6 induced by LPS. Nanoarchaeosomes bilayer extensively interacted with serum proteins but resulted refractory to phospholipases. Upon J774A.1 cells uptake, nanoarchaeosomes induced cytoplasmic acid vesicles, reduced the mitochondrial membrane potential by 20-40 % without consuming ATP neither damaging lysosomes and increasing pERK. Refractory to chemoenzymatic attacks, either void or drug loaded, nanoarchaeosomes induced either anti-inflammation or macrophages apoptosis, constituting promising targeted nanovesicles for multiple therapeutic purposes.
Collapse
|
121
|
Baardman J, Verberk SGS, van der Velden S, Gijbels MJJ, van Roomen CPPA, Sluimer JC, Broos JY, Griffith GR, Prange KHM, van Weeghel M, Lakbir S, Molenaar D, Meinster E, Neele AE, Kooij G, de Vries HE, Lutgens E, Wellen KE, de Winther MPJ, Van den Bossche J. Macrophage ATP citrate lyase deficiency stabilizes atherosclerotic plaques. Nat Commun 2020; 11:6296. [PMID: 33293558 PMCID: PMC7722882 DOI: 10.1038/s41467-020-20141-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/13/2020] [Indexed: 12/24/2022] Open
Abstract
Macrophages represent a major immune cell population in atherosclerotic plaques and play central role in the progression of this lipid-driven chronic inflammatory disease. Targeting immunometabolism is proposed as a strategy to revert aberrant macrophage activation to improve disease outcome. Here, we show ATP citrate lyase (Acly) to be activated in inflammatory macrophages and human atherosclerotic plaques. We demonstrate that myeloid Acly deficiency induces a stable plaque phenotype characterized by increased collagen deposition and fibrous cap thickness, along with a smaller necrotic core. In-depth functional, lipidomic, and transcriptional characterization indicate deregulated fatty acid and cholesterol biosynthesis and reduced liver X receptor activation within the macrophages in vitro. This results in macrophages that are more prone to undergo apoptosis, whilst maintaining their capacity to phagocytose apoptotic cells. Together, our results indicate that targeting macrophage metabolism improves atherosclerosis outcome and we reveal Acly as a promising therapeutic target to stabilize atherosclerotic plaques.
Collapse
Affiliation(s)
- Jeroen Baardman
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sanne G S Verberk
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Saskia van der Velden
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marion J J Gijbels
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Pathology and Molecular Genetics, CARIM, Maastricht University, Maastricht, Netherlands
| | - Cindy P P A van Roomen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Judith C Sluimer
- Department of Pathology and Molecular Genetics, CARIM, Maastricht University, Maastricht, Netherlands.,BHF Centre for Cardiovascular Sciences (CVS), University of Edinburgh, Edinburgh, UK
| | - Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Koen H M Prange
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Cardiovascular sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Soufyan Lakbir
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Systems Bioinformatics, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Douwe Molenaar
- Systems Bioinformatics, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Elisa Meinster
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Annette E Neele
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
| | - Kathryn E Wellen
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Menno P J de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.
| | - Jan Van den Bossche
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands. .,Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
122
|
Logtenberg MEW, Scheeren FA, Schumacher TN. The CD47-SIRPα Immune Checkpoint. Immunity 2020; 52:742-752. [PMID: 32433947 DOI: 10.1016/j.immuni.2020.04.011] [Citation(s) in RCA: 386] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The cytotoxic activity of myeloid cells is regulated by a balance of signals that are transmitted through inhibitory and activating receptors. The Cluster of Differentiation 47 (CD47) protein, expressed on both healthy and cancer cells, plays a pivotal role in this balance by delivering a "don't eat me signal" upon binding to the Signal-regulatory protein alpha (SIRPα) receptor on myeloid cells. Here, we review the current understanding of the role of the CD47-SIRPα axis in physiological tissue homeostasis and as a promising therapeutic target in, among others, oncology, fibrotic diseases, atherosclerosis, and stem cell therapies. We discuss gaps in understanding and highlight where additional insight will be beneficial to allow optimal exploitation of this myeloid cell checkpoint as a target in human disease.
Collapse
Affiliation(s)
- Meike E W Logtenberg
- Division of Molecular Oncology and Immunology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ferenc A Scheeren
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
123
|
Dumont A, Lee M, Barouillet T, Murphy A, Yvan-Charvet L. Mitochondria orchestrate macrophage effector functions in atherosclerosis. Mol Aspects Med 2020; 77:100922. [PMID: 33162108 DOI: 10.1016/j.mam.2020.100922] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
Macrophages are pivotal in the initiation and development of atherosclerotic cardiovascular diseases. Recent studies have reinforced the importance of mitochondria in metabolic and signaling pathways to maintain macrophage effector functions. In this review, we discuss the past and emerging roles of macrophage mitochondria metabolic diversity in atherosclerosis and the potential avenue as biomarker. Beyond metabolic functions, mitochondria are also a signaling platform integrating epigenetic, redox, efferocytic and apoptotic regulations, which are exquisitely linked to their dynamics. Indeed, mitochondria functions depend on their density and shape perpetually controlled by mitochondria fusion/fission and biogenesis/mitophagy balances. Mitochondria can also communicate with other organelles such as the endoplasmic reticulum through mitochondria-associated membrane (MAM) or be secreted for paracrine actions. All these functions are perturbed in macrophages from mouse or human atherosclerotic plaques. A better understanding and integration of how these metabolic and signaling processes are integrated and dictate macrophage effector functions in atherosclerosis may ultimately help the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Adélie Dumont
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204, Nice, France
| | - ManKS Lee
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Immunology, Monash University, Melbourne, Victoria, 3165, Australia
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204, Nice, France
| | - Andrew Murphy
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Immunology, Monash University, Melbourne, Victoria, 3165, Australia
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204, Nice, France.
| |
Collapse
|
124
|
Alsina-Sanchis E, Mülfarth R, Moll I, Mogler C, Rodriguez-Vita J, Fischer A. Intraperitoneal Oil Application Causes Local Inflammation with Depletion of Resident Peritoneal Macrophages. Mol Cancer Res 2020; 19:288-300. [PMID: 33139505 DOI: 10.1158/1541-7786.mcr-20-0650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/08/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Oil is frequently used as a solvent to inject lipophilic substances into the peritoneum of laboratory animals. Although mineral oil causes chronic peritoneal inflammation, little is known whether other oils are better suited. We show that olive, peanut, corn, or mineral oil causes xanthogranulomatous inflammation with depletion of resident peritoneal macrophages. However, there were striking differences in the severity of the inflammatory response. Peanut and mineral oil caused severe chronic inflammation with persistent neutrophil and monocyte recruitment, expansion of the vasculature, and fibrosis. Corn and olive oil provoked no or only mild signs of chronic inflammation. Mechanistically, the vegetal oils were taken up by macrophages leading to foam cell formation and induction of cell death. Olive oil triggered caspase-3 cleavage and apoptosis, which facilitate the resolution of inflammation. Peanut oil and, to a lesser degree, corn oil, triggered caspase-1 activation and macrophage pyroptosis, which impair the resolution of inflammation. As such, intraperitoneal oil administration can interfere with the outcome of subsequent experiments. As a proof of principle, intraperitoneal peanut oil injection was compared with its oral delivery in a thioglycolate-induced peritonitis model. The chronic peritoneal inflammation due to peanut oil injection impeded the proper recruitment of macrophages and the resolution of inflammation in this peritonitis model. In summary, the data indicate that it is advisable to deliver lipophilic substances, like tamoxifen, by oral gavage instead of intraperitoneal injection. IMPLICATIONS: This work contributes to the reproducibility of animal research by helping to understand some of the undesired effects observed in animal experiments.
Collapse
Affiliation(s)
- Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
125
|
Yin C, Vrieze AM, Rosoga M, Akingbasote J, Pawlak EN, Jacob RA, Hu J, Sharma N, Dikeakos JD, Barra L, Nagpal AD, Heit B. Efferocytic Defects in Early Atherosclerosis Are Driven by GATA2 Overexpression in Macrophages. Front Immunol 2020; 11:594136. [PMID: 33193444 PMCID: PMC7644460 DOI: 10.3389/fimmu.2020.594136] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/01/2020] [Indexed: 01/01/2023] Open
Abstract
The loss of efferocytosis-the phagocytic clearance of apoptotic cells-is an initiating event in atherosclerotic plaque formation. While the loss of macrophage efferocytosis is a prerequisite for advanced plaque formation, the transcriptional and cellular events in the pre-lesion site that drive these defects are poorly defined. Transcriptomic analysis of macrophages recovered from early-stage human atherosclerotic lesions identified a 50-fold increase in the expression of GATA2, a transcription factor whose expression is normally restricted to the hematopoietic compartment. GATA2 overexpression in vitro recapitulated many of the functional defects reported in patient macrophages, including deficits at multiple stages in the efferocytic process. These findings included defects in the uptake of apoptotic cells, efferosome maturation, and in phagolysosome function. These efferocytic defects were a product of GATA2-driven alterations in the expression of key regulatory proteins, including Src-family kinases, Rab7 and components of both the vacuolar ATPase and NADPH oxidase complexes. In summary, these data identify a mechanism by which efferocytic capacity is lost in the early stages of plaque formation, thus setting the stage for the accumulation of uncleared apoptotic cells that comprise the bulk of atherosclerotic plaques.
Collapse
Affiliation(s)
- Charles Yin
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Angela M Vrieze
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Mara Rosoga
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - James Akingbasote
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Emily N Pawlak
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Rajesh Abraham Jacob
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Jonathan Hu
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Neha Sharma
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Lillian Barra
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada.,Division of Rheumatology, Department of Medicine, The University of Western Ontario, London, ON, Canada
| | - A Dave Nagpal
- Division of Cardiac Surgery, Department of Surgery, The University of Western Ontario, London, ON, Canada.,Division of Critical Care Medicine, Department of Medicine, The University of Western Ontario, London, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, and The Center for Human Immunology, The University of Western Ontario, London, ON, Canada.,Robarts Research Institute, London, ON, Canada
| |
Collapse
|
126
|
Cai B, Kasikara C. TAM receptors and their ligand-mediated activation: Role in atherosclerosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 357:21-33. [PMID: 33234243 DOI: 10.1016/bs.ircmb.2020.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TAM family tyrosine kinase receptors including Tyro3, Axl, and MerTK are the key efferocytosis receptors presenting on antigen-presenting cell that mediate the clearance of apoptotic cells. They are thought to regulate inflammatory diseases by modulating inflammatory response and efferocytosis. Recent studies have revealed novel roles of TAM receptors in the biosynthesis of specialized pro-resolving mediators (SPMs) and inflammation resolution. In this chapter, we discuss the central roles of TAM signaling in atherosclerosis focusing on their regulation in efferocytosis and inflammation resolution and highlight the unique therapeutic potential of SPMs in blocking the progression of atherosclerosis.
Collapse
Affiliation(s)
- Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Canan Kasikara
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| |
Collapse
|
127
|
Bowman ER, Cameron CM, Richardson B, Kulkarni M, Gabriel J, Cichon MJ, Riedl KM, Mustafa Y, Cartwright M, Snyder B, Raman SV, Zidar DA, Koletar SL, Playford MP, Mehta NN, Sieg SF, Freeman ML, Lederman MM, Cameron MJ, Funderburg NT. Macrophage maturation from blood monocytes is altered in people with HIV, and is linked to serum lipid profiles and activation indices: A model for studying atherogenic mechanisms. PLoS Pathog 2020; 16:e1008869. [PMID: 33002093 PMCID: PMC7553323 DOI: 10.1371/journal.ppat.1008869] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/13/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
People with HIV (PWH) are at increased risk for atherosclerotic cardiovascular disease (ASCVD). Proportions of vascular homing monocytes are enriched in PWH; however, little is known regarding monocyte-derived macrophages (MDMs) that may drive atherosclerosis in this population. We isolated PBMCs from people with and without HIV, and cultured these cells for 5 days in medium containing autologous serum to generate MDMs. Differential gene expression (DGE) analysis of MDMs from PWH identified broad alterations in innate immune signaling (IL-1β, TLR expression, PPAR βδ) and lipid processing (LXR/RXR, ACPP, SREBP1). Transcriptional changes aligned with the functional capabilities of these cells. Expression of activation markers and innate immune receptors (CD163, TLR4, and CD300e) was altered on MDMs from PWH, and these cells produced more TNFα, reactive oxygen species (ROS), and matrix metalloproteinases (MMPs) than did cells from people without HIV. MDMs from PWH also had greater lipid accumulation and uptake of oxidized LDL. PWH had increased serum levels of free fatty acids (FFAs) and ceramides, with enrichment of saturated FAs and a reduction in polyunsaturated FAs. Levels of lipid classes and species that are associated with CVD correlated with unique DGE signatures and altered metabolic pathway activation in MDMs from PWH. Here, we show that MDMs from PWH display a pro-atherogenic phenotype; they readily form foam cells, have altered transcriptional profiles, and produce mediators that likely contribute to accelerated ASCVD. People with HIV (PWH) are at greater risk for developing cardiovascular disease (CVD) than the general public, but the mechanisms underlying this increased risk are poorly understood. Macrophages play key roles in the pathogenesis of atherosclerosis, and are potential targets for therapeutic intervention. Here, we investigate phenotypic and functional abnormalities in monocyte-derived macrophages (MDMs) isolated from PWH that may drive CVD risk in this population. MDMs were differentiated in the presence of autologous serum, enabling us to explore the contributions of serum components (lipids, inflammatory cytokines, microbial products) as drivers of altered MDM function. We link serum levels of inflammatory biomarkers and CVD-associated lipid species to MDM activation. Our study provides new insight into drivers of pro-atherogenic MDM phenotype in PWH, and identifies directions for future study and potential intervention strategies to mitigate CVD risk.
Collapse
Affiliation(s)
- Emily R. Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Cheryl M. Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Manjusha Kulkarni
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Janelle Gabriel
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Morgan J. Cichon
- Department of Food Science & Technology and the Nutrient & Phytochemical Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Kenneth M. Riedl
- Department of Food Science & Technology and the Nutrient & Phytochemical Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Yousef Mustafa
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Michael Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brandon Snyder
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Subha V. Raman
- Department of Internal Medicine, Division of Cardiovascular Medicine, Ohio State University, Columbus, Ohio, United States of America
| | - David A. Zidar
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Susan L. Koletar
- Department of Medicine, Division of Infectious Diseases, Ohio State University, Columbus, Ohio, United States of America
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Scott F. Sieg
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Michael L. Freeman
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Michael M. Lederman
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Mark J. Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nicholas T. Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
128
|
Atkin-Smith GK, Miles MA, Tixeira R, Lay FT, Duan M, Hawkins CJ, Phan TK, Paone S, Mathivanan S, Hulett MD, Chen W, Poon IKH. Plexin B2 Is a Regulator of Monocyte Apoptotic Cell Disassembly. Cell Rep 2020; 29:1821-1831.e3. [PMID: 31722200 DOI: 10.1016/j.celrep.2019.10.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/10/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022] Open
Abstract
Billions of cells undergo apoptosis daily and often fragment into small, membrane-bound extracellular vesicles termed apoptotic bodies (ApoBDs). We demonstrate that apoptotic monocytes undergo a highly coordinated disassembly process and form long, beaded protrusions (coined as beaded apoptopodia), which fragment to release ApoBDs. Here, we find that the protein plexin B2 (PlexB2), a transmembrane receptor that regulates axonal guidance in neurons, is enriched in the ApoBDs of THP1 monocytes and is a caspase 3/7 substrate. To determine whether PlexB2 is involved in the disassembly of apoptotic monocytes, we generate PlexB2-deficient THP1 monocytes and demonstrate that lack of PlexB2 impairs the formation of beaded apoptopodia and ApoBDs. Consequently, the loss of PlexB2 in apoptotic THP1 monocytes impairs their uptake by both professional and non-professional phagocytes. Altogether, these data identify PlexB2 as a positive regulator of apoptotic monocyte disassembly and demonstrate the importance of this process in apoptotic cell clearance.
Collapse
Affiliation(s)
- Georgia K Atkin-Smith
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Mark A Miles
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rochelle Tixeira
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Fung T Lay
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Mubing Duan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Christine J Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Stephanie Paone
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
129
|
Lantz C, Radmanesh B, Liu E, Thorp EB, Lin J. Single-cell RNA sequencing uncovers heterogenous transcriptional signatures in macrophages during efferocytosis. Sci Rep 2020; 10:14333. [PMID: 32868786 PMCID: PMC7459098 DOI: 10.1038/s41598-020-70353-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022] Open
Abstract
Efferocytosis triggers cellular reprogramming, including the induction of mRNA transcripts which encode anti-inflammatory cytokines that promote inflammation resolution. Our current understanding of this transcriptional response is largely informed from analysis of bulk phagocyte populations; however, this precludes the resolution of heterogeneity between individual macrophages and macrophage subsets. Moreover, phagocytes may contain so called "passenger" transcripts that originate from engulfed apoptotic bodies, thus obscuring the true transcriptional reprogramming of the phagocyte. To define the transcriptional diversity during efferocytosis, we utilized single-cell mRNA sequencing after co-cultivating macrophages with apoptotic cells. Importantly, transcriptomic analyses were performed after validating the disappearance of apoptotic cell-derived RNA sequences. Our findings reveal new heterogeneity of the efferocytic response at a single-cell resolution, particularly evident between F4/80+ MHCIILO and F4/80- MHCIIHI macrophage sub-populations. After exposure to apoptotic cells, the F4/80+ MHCIILO subset significantly induced pathways associated with tissue and cellular homeostasis, while the F4/80- MHCIIHI subset downregulated these putative signaling axes. Ablation of a canonical efferocytosis receptor, MerTK, blunted efferocytic signatures and led to the escalation of cell death-associated transcriptional signatures in F4/80+ MHCIILO macrophages. Taken together, our results newly elucidate the heterogenous transcriptional response of single-cell peritoneal macrophages after exposure to apoptotic cells.
Collapse
Affiliation(s)
- Connor Lantz
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA.
| | - Behram Radmanesh
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Esther Liu
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Edward B Thorp
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Jennie Lin
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60611, USA
| |
Collapse
|
130
|
Frégeau G, Sarduy R, Elimam H, Esposito CL, Mellal K, Ménard L, Leitão da Graça SD, Proulx C, Zhang J, Febbraio M, Soto Y, Lubell WD, Ong H, Marleau S. Atheroprotective and atheroregressive potential of azapeptide derivatives of GHRP-6 as selective CD36 ligands in apolipoprotein E-deficient mice. Atherosclerosis 2020; 307:52-62. [PMID: 32721647 DOI: 10.1016/j.atherosclerosis.2020.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 06/08/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Scavenger receptor class B member 3, also known as cluster of differentiation-36 (CD36) receptor, is involved in the uptake and accumulation of modified lipoprotein in macrophages, driving atherosclerosis progression. Azapeptide analogs of growth hormone-releasing peptide-6 (GHRP-6) have been developed as selective CD36 ligands and evaluated for their anti-atherosclerotic properties in apoe-/- mice. METHODS From 4 to 19 weeks of age, male apoe-/- mice were fed a high fat high cholesterol (HFHC) diet, then switched to normal chow and treated daily with 300 nmol/kg of MPE-001 ([aza-Tyr4]-GHRP-6) or MPE-003 ([aza-(N,N-diallylaminobut-2-ynyl)Gly4]-GHRP-6) for 9 weeks. In another protocol, mice were fed a HFHC diet throughout the study. RESULTS Azapeptides decreased lesion progression in the aortic arch and reduced aortic sinus lesion areas below pre-existing lesions levels in apoe-/- mice which were switched to chow diet. In mice fed a HFHC throughout the study, azapeptides reduced lesion progression in the aortic vessel and sinus. The anti-atherosclerotic effect of azapeptides was associated with a reduced ratio of iNOS+/CD206+ macrophages within lesions, and lowered plasma inflammatory cytokine levels. Monocytes from azapeptide-treated mice showed altered mitochondrial oxygen consumption rates, consistent with an M2-like phenotype. These effects were dependent on CD36, and not observed in apoe-/-cd36-/- mice. CONCLUSIONS Azapeptides MPE-001 and MPE-003 diminished aortic lesion progression and reduced, below pre-existing levels, lesions in the aortic sinus of atherosclerotic mice. A relative increase of M2-like macrophages was observed in lesions, associated with reduced systemic inflammation. Development of CD36-selective azapeptide ligands merits consideration for treating atherosclerotic disease.
Collapse
Affiliation(s)
- Geneviève Frégeau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Roger Sarduy
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Hanan Elimam
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada; Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Cloé L Esposito
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Katia Mellal
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Liliane Ménard
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | - Caroline Proulx
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Jinqiang Zhang
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yosdel Soto
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
131
|
Song P, Zhao Q, Zou MH. Targeting senescent cells to attenuate cardiovascular disease progression. Ageing Res Rev 2020; 60:101072. [PMID: 32298812 DOI: 10.1016/j.arr.2020.101072] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the most common disease to increase as life expectancy increases. Most high-profile pharmacological treatments for age-related CVD have led to inefficacious results, implying that novel approaches to treating these pathologies are needed. Emerging data have demonstrated that senescent cardiovascular cells, which are characterized by irreversible cell cycle arrest and a distinct senescence-associated secretory phenotype, accumulate in aged or diseased cardiovascular systems, suggesting that they may impair cardiovascular function. This review discusses the evidence implicating senescent cells in cardiovascular ageing, the onset and progression of CVD, and the molecular mechanisms underlying cardiovascular cell senescence. We also review eradication of senescent cardiovascular cells by small-molecule-drug-mediated apoptosis and immune cell-mediated efferocytosis and toxicity as promising and precisely targeted therapeutics for CVD prevention and treatment.
Collapse
|
132
|
Boada-Romero E, Martinez J, Heckmann BL, Green DR. The clearance of dead cells by efferocytosis. Nat Rev Mol Cell Biol 2020; 21:398-414. [PMID: 32251387 DOI: 10.1038/s41580-020-0232-1] [Citation(s) in RCA: 450] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
Multiple modes of cell death have been identified, each with a unique function and each induced in a setting-dependent manner. As billions of cells die during mammalian embryogenesis and daily in adult organisms, clearing dead cells and associated cellular debris is important in physiology. In this Review, we present an overview of the phagocytosis of dead and dying cells, a process known as efferocytosis. Efferocytosis is performed by macrophages and to a lesser extent by other 'professional' phagocytes (such as monocytes and dendritic cells) and 'non-professional' phagocytes, such as epithelial cells. Recent discoveries have shed light on this process and how it functions to maintain tissue homeostasis, tissue repair and organismal health. Here, we outline the mechanisms of efferocytosis, from the recognition of dying cells through to phagocytic engulfment and homeostatic resolution, and highlight the pathophysiological consequences that can arise when this process is abrogated.
Collapse
Affiliation(s)
- Emilio Boada-Romero
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer Martinez
- Inflammation & Autoimmunity Group, National Institute for Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
133
|
Jiang L, Poon IKH. Methods for monitoring the progression of cell death, cell disassembly and cell clearance. Apoptosis 2020; 24:208-220. [PMID: 30684146 DOI: 10.1007/s10495-018-01511-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell death through apoptosis, necrosis, necroptosis and pyroptosis, as well as the clearance of dead cells are crucial biological processes in the human body. Likewise, disassembly of dying cells during apoptosis to generate cell fragments known as apoptotic bodies may also play important roles in regulating cell clearance and intercellular communication. Recent advances in the field have led to the development of new experimental systems to identify cells at different stages of cell death, measure the levels of apoptotic cell disassembly, and monitor the cell clearance process using a range of in vitro, ex vivo and in vivo models. In this article, we will provide a comprehensive review of the methods for monitoring the progression of cell death, cell disassembly and cell clearance.
Collapse
Affiliation(s)
- Lanzhou Jiang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
134
|
Ghobadi H, Hosseini N, Aslani MR. Correlations Between Serum Decoy Receptor 3 and Airflow Limitation and Quality of Life in Male Patients with Stable Stage and Acute Exacerbation of COPD. Lung 2020; 198:515-523. [PMID: 32211977 DOI: 10.1007/s00408-020-00348-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/16/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Serum decoy receptor 3 (DcR3) level increases in chronic inflammatory diseases. The present study aimed to examine serum DcR3 and IL-6 levels in male patients with stable chronic obstructive pulmonary disease (COPD) and acute exacerbation of the disease and correlations between these markers and airflow limitation. METHODS We measured serum DcR3 and IL-6 levels in 60 COPD patients [30 stable COPD (SCOPD), and 30 acute exacerbation of COPD (AECOPD)], and 30 control subjects and assessed their correlations with airflow limitation according to the COPD stage indicated by the global initiative for chronic obstructive pulmonary disease (GOLD) criteria, peripheral O2 saturation (SpO2), and COPD assessment test (CAT) score. We also tested associations between serum DcR3 levels and COPD patients' clinical parameters. RESULTS Both serum DcR3 and IL-6 levels increased with increasing severity of airflow limitation in SCOPD and AECOPD groups (P < 0.01 to 0.001). These markers also increased in patients with AECOPD compared with subjects in SCOPD group in GOLD stages III-IV (P < 0.05 to 0.001). In addition, there was a significant positive correlation between serum DcR3 level and IL-6, CAT score and smoking history (per year). CONCLUSION The study revealed that serum DcR3 level elevated with increasing severity of airflow limitation in male COPD patients, particularly in acute exacerbation phase. This increase was associated with a reduced quality of life and increased severity of hypoxia. These results suggest that DcR3 may be associated with the underlying pathophysiology of COPD in male patients.
Collapse
Affiliation(s)
- Hassan Ghobadi
- Internal Medicine Department (Pulmonary Division), Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Hosseini
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Inflammatory Diseases Research Center, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran. .,Neurogenetic Inflammation Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
135
|
Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020; 9:cells9030671. [PMID: 32164335 PMCID: PMC7140645 DOI: 10.3390/cells9030671] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
Senescent cells are generally characterized by permanent cell cycle arrest, metabolic alteration and activation, and apoptotic resistance in multiple organs due to various stressors. Excessive accumulation of senescent cells in numerous tissues leads to multiple chronic diseases, tissue dysfunction, age-related diseases and organ ageing. Immune cells can remove senescent cells. Immunaging or impaired innate and adaptive immune responses by senescent cells result in persistent accumulation of various senescent cells. Although senolytics-drugs that selectively remove senescent cells by inducing their apoptosis-are recent hot topics and are making significant research progress, senescence immunotherapies using immune cell-mediated clearance of senescent cells are emerging and promising strategies to fight ageing and multiple chronic diseases. This short review provides an overview of the research progress to date concerning senescent cell-caused chronic diseases and tissue ageing, as well as the regulation of senescence by small-molecule drugs in clinical trials and different roles and regulation of immune cells in the elimination of senescent cells. Mounting evidence indicates that immunotherapy targeting senescent cells combats ageing and chronic diseases and subsequently extends the healthy lifespan.
Collapse
|
136
|
Liao J, Xie Y, Lin Q, Yang X, An X, Xia Y, Du J, Wang F, Li HH. Immunoproteasome subunit β5i regulates diet-induced atherosclerosis through altering MERTK-mediated efferocytosis in Apoe knockout mice. J Pathol 2020; 250:275-287. [PMID: 31758542 DOI: 10.1002/path.5368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/01/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022]
Abstract
The immunoproteasome contains three catalytic subunits (β1i, β2i and β5i) that are important modulators of immune cell homeostasis. A previous study showed a correlation between β5i and human atherosclerotic plaque instability; however, the causative role of β5i in atherosclerosis and the underlying mechanisms remain unknown. Here we explored this issue in apolipoprotein E (Apoe) knockout (eKO) mice with genetic deletion or pharmacological inhibition of β5i. We found that β5i expression was upregulated in lesional macrophages after an atherogenic diet (ATD). β5i/Apoe double KO (dKO) mice fed on the ATD had a significant decrease in both lesion area and necrotic core area, compared with eKO controls. Moreover, dKO mice had less caspase-3+ apoptotic cell accumulation but enhanced efferocytosis of apoptotic cells and increased expression of Mer receptor tyrosine kinase (MERTK). Consistently, similar phenotypes were observed in eKO mice transplanted with dKO bone marrow or treated with β5i-specific inhibitor PR-957. Mechanistic studies in vitro revealed that β5i deletion reduced IκBα degradation and inhibited NF-κB activation, promoting Mertk transcription and efferocytosis, thereby attenuating apoptotic cell accumulation. In conclusion, we demonstrate that β5i plays an important role in diet-induced atherosclerosis by altering MERTK-mediated efferocytosis. β5i might be a potential pharmaceutical target against atherosclerosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jiawei Liao
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yunpeng Xie
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Qiuyue Lin
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Xiaolei Yang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yunlong Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Jie Du
- Beijing AnZhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Feng Wang
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
137
|
Tajbakhsh A, Kovanen PT, Rezaee M, Banach M, Moallem SA, Sahebkar A. Regulation of efferocytosis by caspase-dependent apoptotic cell death in atherosclerosis. Int J Biochem Cell Biol 2020; 120:105684. [PMID: 31911118 DOI: 10.1016/j.biocel.2020.105684] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/02/2019] [Accepted: 01/02/2020] [Indexed: 01/05/2023]
Abstract
During the growing process of the atherosclerotic lesions, lipid-filled macrophage foam cells form, accumulate, and ultimately undergo apoptotic death. If the apoptotic foam cells are not timely removed, they may undergo secondary necrosis, and form a necrotic lipid core which renders the plaque unstable and susceptible to rupture. Therefore, the non-lipid-filled fellow macrophages, as the main phagocytic cells in atherosclerotic lesions, need to effectively remove the apoptotic foam cells. In general, in apoptotic macrophages, caspases are the central regulators of several key processes required for their efficient efferocytosis. The processes include the generation of "Find-Me" signals (such as adenosine triphosphate/uridine triphosphate, fractalkine, lysophosphatidylcholine, and sphingosine-1-phosphate) for the recruitment of viable macrophages, generation of the "Eat-Me" signals (for example, phosphatidylserine) for the engulfment process, and, finally, release of anti-inflammatory mediators (including transforming factor β and interleukin-10) as a tolerance-enhancing and an anti-inflammatory response, and for the motile behavior of the apoptotic cell. The caspase-dependent mechanisms are operative also in apoptotic macrophages driving the atherogenesis. In this review, we explore the role of the molecular pathways related to the caspase-dependent events in efferocytosis in the context of atherosclerosis. Understanding of the molecular mechanisms of apoptotic cell death in atherosclerotic lesions is essential when searching for new leads to treat atherosclerosis.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Halal Research Center of IRI, FDA, Tehran, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahdi Rezaee
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Seyed Adel Moallem
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Al-Zahraa University, Karbala, Iraq
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
138
|
Flores AM, Hosseini-Nassab N, Jarr KU, Ye J, Zhu X, Wirka R, Koh AL, Tsantilas P, Wang Y, Nanda V, Kojima Y, Zeng Y, Lotfi M, Sinclair R, Weissman IL, Ingelsson E, Smith BR, Leeper NJ. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. NATURE NANOTECHNOLOGY 2020; 15:154-161. [PMID: 31988506 PMCID: PMC7254969 DOI: 10.1038/s41565-019-0619-3] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 12/09/2019] [Indexed: 05/18/2023]
Abstract
Atherosclerosis is the process that underlies heart attack and stroke. A characteristic feature of the atherosclerotic plaque is the accumulation of apoptotic cells in the necrotic core. Prophagocytic antibody-based therapies are currently being explored to stimulate the phagocytic clearance of apoptotic cells; however, these therapies can cause off-target clearance of healthy tissues, which leads to toxicities such as anaemia. Here we developed a macrophage-specific nanotherapy based on single-walled carbon nanotubes loaded with a chemical inhibitor of the antiphagocytic CD47-SIRPα signalling axis. We demonstrate that these single-walled carbon nanotubes accumulate within the atherosclerotic plaque, reactivate lesional phagocytosis and reduce the plaque burden in atheroprone apolipoprotein-E-deficient mice without compromising safety, and thereby overcome a key translational barrier for this class of drugs. Single-cell RNA sequencing analysis reveals that prophagocytic single-walled carbon nanotubes decrease the expression of inflammatory genes linked to cytokine and chemokine pathways in lesional macrophages, which demonstrates the potential of 'Trojan horse' nanoparticles to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Alyssa M Flores
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xingjun Zhu
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Wirka
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ai Leen Koh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Pavlos Tsantilas
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying Wang
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Vivek Nanda
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yitian Zeng
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Mozhgan Lotfi
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Sinclair
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Irving L Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Bryan Ronain Smith
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, East Lansing, MI, USA.
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
139
|
Wang Z, Sequeira RC, Zabalawi M, Madenspacher J, Boudyguina E, Ou T, Nelson JM, Nie Y, Zhao Q, Fessler MB, Zhu X. Myeloid atg5 deletion impairs n-3 PUFA-mediated atheroprotection. Atherosclerosis 2020; 295:8-17. [PMID: 31978760 DOI: 10.1016/j.atherosclerosis.2020.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/13/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Dietary long-chain (≥20 carbons) n-3 polyunsaturated fatty acids (PUFAs) reduce atherosclerosis and enhance macrophage autophagy activation. How macrophage autophagy impacts atherosclerotic progression, particularly when comparing dietary n-3 PUFA supplementation vs. saturated fat feeding, is unknown. METHODS We generated myeloid-specific autophagy-deficient and control mice in the Ldlr-/- background by transplanting bone marrow from myeloid-specific autophagy-related (atg) 5 knockout mice and wild type controls into irradiated Ldlr-/- recipients. After 7 weeks for recovery from radiation, mice were fed an atherogenic diet containing 0.2% cholesterol and 20% calories as palm oil (PO diet), or 10% calories as PO plus 10% calories as fish oil (FO diet) for 16 weeks. RESULTS Compared to PO, FO significantly reduced plasma cholesterol, triglyceride, hepatic neutral lipid, and aortic caspase-1 cleavage, but increased aortic efferocytosis, leading to attenuated atherosclerosis in Ldlr-/- mice receiving wild type bone marrow. Myeloid atg5 deletion had little impact on plasma lipid concentrations and hepatic neutral lipid content, regardless of diet. Myeloid atg5 deletion increased aortic caspase-1 cleavage, decreased aortic efferocytosis and worsened atherosclerosis only in the FO-fed Ldlr-/- mice. CONCLUSIONS Deficient myeloid autophagy significantly attenuated FO-induced atheroprotection, suggesting that dietary n-3 PUFAs reduce atherosclerosis, in part, by activation of macrophage autophagy.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Russel C Sequeira
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Manal Zabalawi
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jennifer Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709, USA
| | - Elena Boudyguina
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Tiantong Ou
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jonathan M Nelson
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yan Nie
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Qingxia Zhao
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709, USA
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
140
|
Wofford KL, Singh BS, Cullen DK, Spiller KL. Biomaterial-mediated reprogramming of monocytes via microparticle phagocytosis for sustained modulation of macrophage phenotype. Acta Biomater 2020; 101:237-248. [PMID: 31731024 PMCID: PMC6960335 DOI: 10.1016/j.actbio.2019.11.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/14/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022]
Abstract
Monocyte-derived macrophages orchestrate tissue regeneration by homing to sites of injury, phagocytosing pathological debris, and stimulating other cell types to repair the tissue. Accordingly, monocytes have been investigated as a translational and potent source for cell therapy, but their utility has been hampered by their rapid acquisition of a pro-inflammatory phenotype in response to the inflammatory injury microenvironment. To overcome this problem, we designed a cell therapy strategy where monocytes are exogenously reprogrammed by intracellularly loading the cells with biodegradable microparticles containing an anti-inflammatory drug in order to modulate and maintain an anti-inflammatory phenotype over time. To test this concept, poly(lactic-co-glycolic) acid microparticles were loaded with the anti-inflammatory drug dexamethasone (Dex) and administered to primary human monocytes for four hours to facilitate phagocytic uptake. After removal of non-phagocytosed microparticles, microparticle-loaded monocytes differentiated into macrophages and stored the microparticles intracellularly for several weeks in vitro, releasing drug into the extracellular environment over time. Cells loaded with intracellular Dex microparticles showed decreased expression and secretion of inflammatory factors even in the presence of pro-inflammatory stimuli up to 7 days after microparticle uptake compared to untreated cells or cells loaded with blank microparticles, without interfering with phagocytosis of tissue debris. This study represents a new strategy for long-term maintenance of anti-inflammatory macrophage phenotype using a translational monocyte-based cell therapy strategy without the use of genetic modification. Because of the ubiquitous nature of monocyte-derived macrophage involvement in pathology and regeneration, this strategy holds potential as a treatment for a vast number of diseases and disorders. STATEMENT OF SIGNIFICANCE: We report a unique and translational strategy to overcome the challenges associated with monocyte- and macrophage-based cell therapies, in which the cells rapidly take on inflammatory phenotypes when administered to sites of injury. By intracellularly loading monocytes with drug-loaded microparticles prior to administration via phagocytosis, we were able to inhibit inflammation while preserving functional behaviors of human primary macrophages derived from those monocytes up to seven days later. To our knowledge, this study represents the first report of reprogramming macrophages to an anti-inflammatory phenotype without the use of genetic modification.
Collapse
Affiliation(s)
- Kathryn L Wofford
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Departments of Neurosurgery & Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Bhavani S Singh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - D Kacy Cullen
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Departments of Neurosurgery & Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Kara L Spiller
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
141
|
Abstract
The clearance of apoptotic cells by professional and non-professional phagocytes - a process termed 'efferocytosis' - is essential for the maintenance of tissue homeostasis. Accordingly, defective efferocytosis underlies a growing list of chronic inflammatory diseases. Although much has been learnt about the mechanisms of apoptotic cell recognition and uptake, several key areas remain incompletely understood. This Review focuses on new discoveries related to how phagocytes process the metabolic cargo they receive during apoptotic cell uptake; the links between efferocytosis and the resolution of inflammation in health and disease; and the roles of efferocytosis in host defence. Understanding these aspects of efferocytosis sheds light on key physiological and pathophysiological processes and suggests novel therapeutic strategies for diseases driven by defective efferocytosis and impaired inflammation resolution.
Collapse
|
142
|
Hamers AAJ, Dinh HQ, Thomas GD, Marcovecchio P, Blatchley A, Nakao CS, Kim C, McSkimming C, Taylor AM, Nguyen AT, McNamara CA, Hedrick CC. Human Monocyte Heterogeneity as Revealed by High-Dimensional Mass Cytometry. Arterioscler Thromb Vasc Biol 2019; 39:25-36. [PMID: 30580568 DOI: 10.1161/atvbaha.118.311022] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective- Three distinct human monocyte subsets have been identified based on the surface marker expression of CD14 and CD16. We hypothesized that monocytes were likely more heterogeneous in composition. Approach and Results- We used the high dimensionality of mass cytometry together with the FlowSOM clustering algorithm to accurately identify and define monocyte subsets in blood of healthy human subjects and those with coronary artery disease (CAD). To study the behavior and functionality of the newly defined monocyte subsets, we performed RNA sequencing, transwell migration, and efferocytosis assays. Here, we identify 8 human monocyte subsets based on their surface marker phenotype. We found that 3 of these subsets fall within the CD16+ nonclassical monocyte population and 4 subsets belong to the CD14+ classical monocytes, illustrating significant monocyte heterogeneity in humans. As nonclassical monocytes are important in modulating atherosclerosis in mice, we studied the functions of our 3 newly identified nonclassical monocytes in subjects with CAD. We found a marked expansion of a Slan+CXCR6+ nonclassical monocyte subset in CAD subjects, which was positively correlated with CAD severity. This nonclassical subset can migrate towards CXCL16 and shows an increased efferocytosis capacity, indicating it may play an atheroprotective role. Conclusions- Our data demonstrate that human nonclassical monocytes are a heterogeneous population, existing of several subsets with functional differences. These subsets have changed frequencies in the setting of severe CAD. Understanding how these newly identified subsets modulate CAD will be important for CAD-based therapies that target myeloid cells.
Collapse
Affiliation(s)
- Anouk A J Hamers
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Huy Q Dinh
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Graham D Thomas
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Paola Marcovecchio
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Amy Blatchley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Catherine S Nakao
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| | - Cheryl Kim
- Flow Cytometry Core Facility, La Jolla Institute for Allergy and Immunology, CA (C.K.)
| | - Chantel McSkimming
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (C.M., A.M.T., A.T.N., C.A.M.)
| | - Angela M Taylor
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (C.M., A.M.T., A.T.N., C.A.M.)
| | - Anh T Nguyen
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (C.M., A.M.T., A.T.N., C.A.M.)
| | - Coleen A McNamara
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (C.M., A.M.T., A.T.N., C.A.M.)
| | - Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA (A.A.J.H., H.Q.D., G.D.T., P.M., A.B., C.S.N., C.C.H.)
| |
Collapse
|
143
|
Xu H, Jiang J, Chen W, Li W, Chen Z. Vascular Macrophages in Atherosclerosis. J Immunol Res 2019; 2019:4354786. [PMID: 31886303 PMCID: PMC6914912 DOI: 10.1155/2019/4354786] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is the main pathological basis for the occurrence of most cardiovascular diseases, the leading global health threat, and a great burden for society. It has been well established that atherosclerosis is not only a metabolic disorder but also a chronic, sterile, and maladaptive inflammatory process encompassing both innate and adaptive immunity. Macrophages, the major immune cell population in atherosclerotic lesions, have been shown to play critical roles in all stages of atherosclerosis, including the initiation and progression of advanced atherosclerosis. Macrophages have emerged as a novel potential target for antiatherosclerosis therapy. In addition, the macrophage phenotype is greatly influenced by microenvironmental stimuli in the plaques and presents complex heterogeneity. This article reviews the functions of macrophages in different stages of atherosclerosis, as well as the phenotypes and functions of macrophage subsets. New treatment strategies based on macrophage-related inflammation are also discussed.
Collapse
Affiliation(s)
- Hailin Xu
- Department of General Surgery, The First People's Hospital of Jiande, Hangzhou, China
| | - Jingxin Jiang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhigang Chen
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
144
|
Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol 2019; 16:727-744. [PMID: 31243391 DOI: 10.1038/s41569-019-0227-9] [Citation(s) in RCA: 677] [Impact Index Per Article: 112.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type present at all stages of an atherosclerotic plaque. According to the 'response to injury' and 'vulnerable plaque' hypotheses, contractile VSMCs recruited from the media undergo phenotypic conversion to proliferative synthetic cells that generate extracellular matrix to form the fibrous cap and hence stabilize plaques. However, lineage-tracing studies have highlighted flaws in the interpretation of former studies, revealing that these studies had underestimated both the content and functions of VSMCs in plaques and have thus challenged our view on the role of VSMCs in atherosclerosis. VSMCs are more plastic than previously recognized and can adopt alternative phenotypes, including phenotypes resembling foam cells, macrophages, mesenchymal stem cells and osteochondrogenic cells, which could contribute both positively and negatively to disease progression. In this Review, we present the evidence for VSMC plasticity and summarize the roles of VSMCs and VSMC-derived cells in atherosclerotic plaque development and progression. Correct attribution and spatiotemporal resolution of clinically beneficial and detrimental processes will underpin the success of any therapeutic intervention aimed at VSMCs and their derivatives.
Collapse
Affiliation(s)
- Gemma L Basatemur
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Helle F Jørgensen
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Murray C H Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.
- INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
145
|
Kubátová H, Poledne R, Piťha J. Immune cells in carotid artery plaques: what can we learn from endarterectomy specimens? INT ANGIOL 2019; 39:37-49. [PMID: 31782285 DOI: 10.23736/s0392-9590.19.04250-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Endarterectomy specimens represent a unique opportunity to study atherosclerosis. This review aims to summarize the recent knowledge of atherogenesis from studies characterizing a cellular composition of carotid endarterectomy specimens. EVIDENCE ACQUISITION A non-systematic literature review was carried out to summarize recent knowledge regarding ex vivo analysis of carotid artery plaque composition. Upon evaluation of their relevance, and elaborate forward and backward search, 95 articles were included in the review. EVIDENCE SYNTHESIS Despite the significant advancement of in vivo imaging techniques, the stroke prediction based on carotid artery plaque morphology is not reliable. Besides analyses of plaque morphology, present studies focus on precise characterization of the different immune cell types and elucidation of their role in plaque development. Plaque content analyses revealed the presence of various immune cells in carotid artery plaques. Presence of different immune cells subpopulations can be connected to some undesirable changes in plaque stability. CONCLUSIONS Since the destabilization of the atherosclerotic plaque is a multifactorial process, a combination of various methods should be used to characterize the unstable plaques more accurately. In this context, studies characterizing plaque content from a cellular point of view could elucidate some processes underlying the plaque progression. Together with morphological evaluation, these analyses could enable more precise assessment of plaque stability.
Collapse
Affiliation(s)
- Hana Kubátová
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic - .,Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic -
| | - Rudolf Poledne
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Piťha
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,Department of Internal Medicine, Second Medical Faculty, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
146
|
Catan A, Turpin C, Diotel N, Patche J, Guerin-Dubourg A, Debussche X, Bourdon E, Ah-You N, Le Moullec N, Besnard M, Veerapen R, Rondeau P, Meilhac O. Aging and glycation promote erythrocyte phagocytosis by human endothelial cells: Potential impact in atherothrombosis under diabetic conditions. Atherosclerosis 2019; 291:87-98. [PMID: 31704555 DOI: 10.1016/j.atherosclerosis.2019.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 10/16/2019] [Accepted: 10/25/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS Atherothrombotic plaques of type 2 diabetic (T2D) patients are characterized by an increased neovascularization and intraplaque hemorrhage. The clearance of erythrocytes may be carried out by vascular cells. We explored the potential of human endothelial cells to bind and phagocyte in vitro aged and/or glycated erythrocytes as well as erythrocytes obtained from diabetic patients. METHODS Fresh, aged and glycated-aged erythrocytes from healthy volunteers and T2D patients were tested for their binding and phagocytosis capacity as well as the potential functional consequences on endothelial cells (viability, proliferation and wound healing capacity). Immunohistochemistry was also performed in human carotid atherothrombotic samples (from patients with or without T2D). RESULTS Aging and glycation of erythrocytes induced phosphatidylserine (PS) exposure and oxidative stress leading to enhanced endothelial cell binding and engulfment. Phagocytosis by endothelial cells was more pronounced with aged and glycated erythrocytes than with fresh ones. Phagocytosis was enhanced with T2D versus healthy erythrocytes. Furthermore, endothelial wound healing potential was significantly blunted after exposure to glycated-aged versus fresh erythrocytes. Finally, we show that interactions between erythrocytes and endothelial cells and their potential phagocytosis may occur in vivo, in atherothrombotic conditions, in neovessels and in the luminal endothelial lining. CONCLUSIONS Endothelial cells may play an important role in erythrocyte clearance in an atherothrombotic environment. Under diabetic conditions, erythrocyte glycation favors their engulfment by endothelial cells and may participate in endothelial dysfunction, thereby promoting vulnerable atherothrombotic plaques to rupture.
Collapse
Affiliation(s)
- Aurélie Catan
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | - Chloé Turpin
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | - Nicolas Diotel
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | - Jessica Patche
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | | | | | - Emmanuel Bourdon
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | | | | | | | - Reuben Veerapen
- Clinique Sainte Clotilde, Groupe de santé Clinifutur, Saint Denis, France
| | - Philippe Rondeau
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France
| | - Olivier Meilhac
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Sainte-Clotilde, F-97490, France; CHU de La Réunion, Saint-Denis, Saint-Pierre, France.
| |
Collapse
|
147
|
Okwan-Duodu D, Weiss D, Peng Z, Veiras LC, Cao DY, Saito S, Khan Z, Bernstein EA, Giani JF, Taylor WR, Bernstein KE. Overexpression of myeloid angiotensin-converting enzyme (ACE) reduces atherosclerosis. Biochem Biophys Res Commun 2019; 520:573-579. [PMID: 31615657 DOI: 10.1016/j.bbrc.2019.10.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Macrophages are ubiquitous in all stages of atherosclerosis, exerting tremendous impact on lesion progression and plaque stability. Because macrophages in atherosclerotic plaques express angiotensin-converting enzyme (ACE), current dogma posits that local myeloid-mediated effects worsen the disease. In contrast, we previously reported that myeloid ACE overexpression augments macrophage resistance to various immune challenges, including tumors, bacterial infection and Alzheimer's plaque deposition. Here, we sought to assess the impact of myeloid ACE on atherosclerosis. METHODS A mouse model in which ACE is overexpressed in myelomonocytic lineage cells, called ACE10, was generated and sequentially crossed with ApoE-deficient mice to create ACE10/10ApoE-/- (ACE10/ApoE). Control mice were ACEWT/WTApoE-/- (WT/ApoE). Atherosclerosis was induced using an atherogenic diet alone, or in combination with unilateral nephrectomy plus deoxycorticosterone acetate (DOCA) salt for eight weeks. RESULTS With an atherogenic diet alone or in combination with DOCA, the ACE10/ApoE mice showed significantly less atherosclerotic plaques compared to their WT/ApoE counterparts (p < 0.01). When recipient ApoE-/- mice were reconstituted with ACE10/10 bone marrow, these mice showed significantly reduced lesion areas compared to recipients reconstituted with wild type bone marrow. Furthermore, transfer of ACE-deficient bone marrow had no impact on lesion area. CONCLUSION Our data indicate that while myeloid ACE may not be required for atherosclerosis, enhanced ACE expression paradoxically reduced disease progression.
Collapse
Affiliation(s)
- Derick Okwan-Duodu
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Daiana Weiss
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhenzi Peng
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Luciana C Veiras
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Duo-Yao Cao
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Suguru Saito
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zakir Khan
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jorge F Giani
- Department of Biomedical Sciences Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA; Division of Cardiology, Atlanta VA Medical Center, Decatur, GA, USA
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
148
|
Johnston JM, Angyal A, Bauer RC, Hamby S, Suvarna SK, Baidžajevas K, Hegedus Z, Dear TN, Turner M, Wilson HL, Goodall AH, Rader DJ, Shoulders CC, Francis SE, Kiss-Toth E. Myeloid Tribbles 1 induces early atherosclerosis via enhanced foam cell expansion. SCIENCE ADVANCES 2019; 5:eaax9183. [PMID: 31692955 PMCID: PMC6821468 DOI: 10.1126/sciadv.aax9183] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/14/2019] [Indexed: 05/16/2023]
Abstract
Macrophages drive atherosclerotic plaque progression and rupture; hence, attenuating their atherosclerosis-inducing properties holds promise for reducing coronary heart disease (CHD). Recent studies in mouse models have demonstrated that Tribbles 1 (Trib1) regulates macrophage phenotype and shows that Trib1 deficiency increases plasma cholesterol and triglyceride levels, suggesting that reduced TRIB1 expression mediates the strong genetic association between the TRIB1 locus and increased CHD risk in man. However, we report here that myeloid-specific Trib1 (mTrib1) deficiency reduces early atheroma formation and that mTrib1 transgene expression increases atherogenesis. Mechanistically, mTrib1 increased macrophage lipid accumulation and the expression of a critical receptor (OLR1), promoting oxidized low-density lipoprotein uptake and the formation of lipid-laden foam cells. As TRIB1 and OLR1 RNA levels were also strongly correlated in human macrophages, we suggest that a conserved, TRIB1-mediated mechanism drives foam cell formation in atherosclerotic plaque and that inhibiting mTRIB1 could be used therapeutically to reduce CHD.
Collapse
Affiliation(s)
- Jessica M. Johnston
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Robert C. Bauer
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- Perelman School of Medicine at the University of Pennsylvania and Children’s Hospital of Philadelphia, Philadelphia, PA 19104-5158, USA
| | - Stephen Hamby
- Department of Cardiovascular Sciences, University of Leicester and NIHR Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - S. Kim Suvarna
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Kajus Baidžajevas
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Zoltan Hegedus
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvari korut 62, Szeged H-6726, Hungary
- Departments of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Szigeti ut 12, Pecs H-7624, Hungary
| | - T. Neil Dear
- Division of Biomedical Services, University of Leicester, Leicester, UK
| | - Martin Turner
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Heather L. Wilson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Alison H. Goodall
- Department of Cardiovascular Sciences, University of Leicester and NIHR Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Daniel J. Rader
- Perelman School of Medicine at the University of Pennsylvania and Children’s Hospital of Philadelphia, Philadelphia, PA 19104-5158, USA
| | - Carol C. Shoulders
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London and the Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Sheila E. Francis
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
149
|
Zhang Y, Wang Y, Zhou D, Zhang LS, Deng FX, Shu S, Wang LJ, Wu Y, Guo N, Zhou J, Yuan ZY. Angiotensin II deteriorates advanced atherosclerosis by promoting MerTK cleavage and impairing efferocytosis through the AT1R/ROS/p38 MAPK/ADAM17 pathway. Am J Physiol Cell Physiol 2019; 317:C776-C787. [PMID: 31390228 DOI: 10.1152/ajpcell.00145.2019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Vulnerable plaques in advanced atherosclerosis have defective efferocytosis. The role of ANG II in the progression of atherosclerosis is not fully understood. Herein, we investigated the effects and the underlying mechanisms of ANG II on macrophage efferocytosis in advanced atherosclerosis. ANG II decreased the surface expression of Mer tyrosine kinase (MerTK) in macrophages through a disintegrin and metalloproteinase17 (ADAM17)-mediated shedding of the soluble form of MerTK (sMer) in the medium, which led to efferocytosis suppression. ANG II-activated ADAM17 required reactive oxygen species (ROS) and p38 MAPK phosphorylation. Selective angiotensin II type 1 receptor (AT1R) blocker losartan suppressed ROS production, and ROS scavenger N-acetyl-l-cysteine (NAC) prevented p38 MAPK phosphorylation. In addition, mutant MERTKΔ483-488was resistant to ANG II-induced MerTK shedding and efferocytosis suppression. The advanced atherosclerosis model that is characterized by larger necrotic cores, and less collagen content was established by feeding apolipoprotein E knockout (ApoE−/−) mice with a high-fat diet for 16 wk. NAC and losartan oral administration prevented atherosclerotic lesion progression. Meanwhile, the inefficient efferocytosis represented by decreased macrophage-associated apoptotic cells and decreased MerTK+CD68+double-positive macrophages in advanced atherosclerosis were prevented by losartan and NAC. Additionally, the serum levels of sMer were increased and positively correlated with the upregulated levels of ANG II in acute coronary syndrome (ACS) patients. In conclusion, ANG II promotes MerTK shedding via AT1R/ROS/p38 MAPK/ADAM17 pathway in macrophages, which led to defective efferocytosis and atherosclerosis progression. Defining the molecular mechanisms of defective efferocytosis may provide a promising prognosis and therapy for ACS patients.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Ying Wang
- Department of Critical Care Medicine, Xi’an No. 4 Hospital, Xi’an, China
| | - Dong Zhou
- Department of Cardiovascular Medicine, Hanzhong 3201 Hospital, Hanzhong, China
| | - Li-Sha Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Fu-Xue Deng
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Shan Shu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Li-Jun Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Yue Wu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Ning Guo
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
| | - Juan Zhou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shannxi Province, Xi’an, China
| | - Zu-Yi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shannxi Province, Xi’an, China
| |
Collapse
|
150
|
Moore KJ, Koplev S, Fisher EA, Tabas I, Björkegren JLM, Doran AC, Kovacic JC. Macrophage Trafficking, Inflammatory Resolution, and Genomics in Atherosclerosis: JACC Macrophage in CVD Series (Part 2). J Am Coll Cardiol 2019; 72:2181-2197. [PMID: 30360827 DOI: 10.1016/j.jacc.2018.08.2147] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is characterized by the retention of modified lipoproteins in the arterial wall. These modified lipoproteins activate resident macrophages and the recruitment of monocyte-derived cells, which differentiate into mononuclear phagocytes that ingest the deposited lipoproteins to become "foam cells": a hallmark of this disease. In this Part 2 of a 4-part review series covering the macrophage in cardiovascular disease, we critically review the contributions and relevant pathobiology of monocytes, macrophages, and foam cells as relevant to atherosclerosis. We also review evidence that via various pathways, a failure of the resolution of inflammation is an additional key aspect of this disease process. Finally, we consider the likely role played by genomics and biological networks in controlling the macrophage phenotype in atherosclerosis. Collectively, these data provide substantial insights on the atherosclerotic process, while concurrently offering numerous molecular and genomic candidates that appear to hold great promise for selective targeting as clinical therapies.
Collapse
Affiliation(s)
- Kathryn J Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, Marc and Ruti Bell Vascular Biology and Disease Program, New York University School of Medicine, New York, New York
| | - Simon Koplev
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Marc and Ruti Bell Vascular Biology and Disease Program, New York University School of Medicine, New York, New York
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, New York; Department of Pathology and Cell Biology, Columbia University, New York, New York; Department of Physiology, Columbia University, New York, New York
| | - Johan L M Björkegren
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York; Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, New York
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|