1
|
Song M, Dai H, Zhou Q, Meng X. The immunology of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1542208. [PMID: 40260277 PMCID: PMC12009709 DOI: 10.3389/fendo.2025.1542208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Diabetic cardiomyopathy is a notable microvascular complication of diabetes, characterized primarily by myocardial fibrosis and functional abnormalities. Long-term hyperglycemia induces excessive activation and recruitment of immune cells and triggers the cascade of inflammatory responses, resulting in systemic and local cardiac inflammation. Emerging evidence highlights the significant roles of immunology in modulating the pathology of diabetic cardiomyopathy. As the primary effectors of inflammatory reactions, immune cells are consistently present in cardiac tissue and can be recruited under pathological hyperglycemia circumstances. A disproportionate favor to proinflammatory types of immune cells and the increased proinflammatory cytokine levels mediate fibroblast proliferation, phenotypic transformation, and collagen synthesis and ultimately rise to cardiac fibrosis and hypertrophy. Meanwhile, the severity of cardiac fibrosis is also strongly associated with the diverse phenotypes and phenotypic alterations of the immune cells, including macrophages, dendritic cells, mast cells, neutrophils, and natural killer cells in innate immunity and CD4+ T lymphocytes, CD8+ T lymphocytes, and B lymphocytes in adaptive immunity. In this review, we synthesized the current analysis of the critical role played by the immune system and its components in the progression of diabetic cardiomyopathy. Finally, we highlight preclinical and clinical immune targeting strategies and translational implications.
Collapse
Affiliation(s)
| | | | | | - Xiao Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Nappi F. Myocarditis and Inflammatory Cardiomyopathy in Dilated Heart Failure. Viruses 2025; 17:484. [PMID: 40284927 PMCID: PMC12031395 DOI: 10.3390/v17040484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory cardiomyopathy is a condition that is characterised by the presence of inflammatory cells in the myocardium, which can lead to a significant deterioration in cardiac function. The etiology of this condition involves multiple factors, both infectious and non-infectious causes. While it is primarily associated with viral infections, other potential causes include bacterial, protozoal, or fungal infections, as well as a wide variety of toxic substances and drugs, and systemic immune-mediated pathological conditions. In spite of comprehensive investigation, the presence of inflammatory cardiomyopathy accompanied by left ventricular dysfunction, heart failure or arrhythmia is indicative of an unfavourable outcome. The reasons for the occurrence of either favourable outcomes, characterised by the absence of residual myocardial injury, or unfavourable outcomes, marked by the development of dilated cardiomyopathy, in patients afflicted by the condition remain to be elucidated. The relative contributions of pathogenic agents, genomic profiles of the host, and environmental factors in disease progression and resolution remain subjects of ongoing discourse. This includes the determination of which viruses function as active inducers and which merely play a bystander role. It remains unknown which changes in the host immune profile are critical in determining the outcome of myocarditis caused by various viruses, including coxsackievirus B3 (CVB3), adenoviruses, parvoviruses B19 and SARS-CoV-2. The objective of this review is unambiguous: to provide a concise summary and comprehensive assessment of the extant evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy. Its focus is exclusively on virus-induced and virus-associated myocarditis. In addition, the extant lacunae of knowledge in this field are identified and the extant experimental models are evaluated, with the aim of proposing future directions for the research domain. This includes differential gene expression that regulates iron and lipid and metabolic remodelling. Furthermore, the current state of knowledge regarding the cardiovascular implications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is also discussed, along with the open questions that remain to be addressed.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
3
|
Pressley KR, Naseem Y, Nalawade S, Forsthuber TG. The distinct functions of MIF in inflammatory cardiomyopathy. Front Immunol 2025; 16:1544484. [PMID: 40092999 PMCID: PMC11906721 DOI: 10.3389/fimmu.2025.1544484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
The immune system plays a crucial role in cardiac homeostasis and disease, and the innate and adaptive immune systems can be beneficial or detrimental in cardiac injury. The pleiotropic proinflammatory cytokine macrophage migration inhibitory factor (MIF) is involved in the pathogenesis of many human disease conditions, including heart diseases and inflammatory cardiomyopathies. Inflammatory cardiomyopathies are frequently observed after microbial infection but can also be caused by systemic immune-mediated diseases, drugs, and toxic substances. Immune cells and MIF are implicated in many of these conditions and may affect progression of inflammatory cardiomyopathy (ICM) to myocardial remodeling and dilated cardiomyopathy (DCM). The potential for targeting MIF therapeutically in patients with inflammatory diseases is an active area of investigation. Here we review the current literature supporting the role(s) of MIF in ICM and cardiac dysfunction. We posit that future research to further elucidate the underlying functions of MIF in cardiac pathologies is warranted.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Yashfa Naseem
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Saisha Nalawade
- Department of Pre-clinical Immunology, Corner Therapeutics, Watertown, MA, United States
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
4
|
Dandel M. Autoimmunity in Cardiomyopathy-Induced Heart Failure and Cardiac Autoantibody Removal by Immunoadsorption. J Clin Med 2025; 14:947. [PMID: 39941618 PMCID: PMC11818089 DOI: 10.3390/jcm14030947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
There is increasing evidence that β1-adrenoreceptor autoantibody (β1AR-AAb) elimination can break the vicious circle induced by certain pathological conditions associated with alteration of the physiological self-tolerance, followed by generation of such AAbs and activation of cell-mediated immune processes directed against the myocardium. Concerning this, the present narrative review article provides an updated overview of the state of knowledge about the role of auto-immunity in the etiopathogenesis of cardiomyopathies, with a particular focus on immunoadsorption (IA) therapy for β1AR-AAb-positive adult patients with a dilated cardiomyopathy (DCM)-associated refractory heart failure (HF). Among many relevant findings, the increasing prevalence (up to 97%) of β1AR-AAb-positive patients related to the aggravation of HF, the high prevalence (between 84% and 91%) of HF patients in which IA can reduce to a minimum any increased β1AR-AAb level, as well as the high prevalence (about 80%) of responders to the IA-induced normalization of β1AR-AAb levels by long-term improvement in LV ejection fraction with increase in LV stroke volume and cardiac output, are of particular relevance. Given that after the elimination of β1AR-AAbs in potential candidates for heart transplantation (HTx), the post-IA 3- and 5-year HTx-/mechanical support-free survival probability reached 80% and 63-69%, respectively, the good tolerability of IA and the possibility to repeat that therapy also in elderly persons strongly suggest that in appropriately selected patients, this therapy deserves much more attention in the future.
Collapse
Affiliation(s)
- Michael Dandel
- German Centre for Heart and Circulatory Research (DZHK), Potsdamer Str. 58, 10785 Berlin, Germany
| |
Collapse
|
5
|
Wculek SK. Harnessing dendritic cell metabolism for healthy ageing: reducing the risk of cardiovascular disease? Eur Heart J 2024; 45:2355-2357. [PMID: 38842574 DOI: 10.1093/eurheartj/ehae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Affiliation(s)
- Stefanie K Wculek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac, 10, 08028 Barcelona, Spain
| |
Collapse
|
6
|
Costa C, Moniati F. The Epidemiology of COVID-19 Vaccine-Induced Myocarditis. Adv Med 2024; 2024:4470326. [PMID: 38681683 PMCID: PMC11045291 DOI: 10.1155/2024/4470326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/26/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Background In December 2019, the emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) led to the COVID-19 pandemic, with millions of deaths worldwide. Vaccine breakthroughs in late 2020 resulted in the authorization of COVID-19 vaccines. While these vaccines have demonstrated efficacy, evidence from vaccine safety monitoring systems around the globe supported a causal association between COVID-19 vaccines, in particular those using mRNA technology, i.e., Moderna's mRNA-1273 and Pfizer-BioNTech's BNT162b2, and myocarditis. Objective This paper aims to investigate the epidemiology of mRNA COVID-19 vaccine-induced myocarditis, including age, ethnicity, and gender associations with these vaccines. It also discusses the immunopathophysiological mechanisms of mRNA COVID-19 vaccine-associated myocarditis and outlines principles of diagnosis, clinical presentation, and management. Methods A literature review was conducted using PubMed, Embase, and Queen Mary University of London Library Services databases. Search terms included "myocarditis," "coronavirus disease 2019," "SARS-CoV-2," "mRNA Covid-19 vaccines," "Covid vaccine-associated myocarditis," "epidemiology," "potential mechanisms," "myocarditis diagnosis," and "myocarditis management." Results While the definite mechanism of mRNA COVID-19 vaccine-associated myocarditis remains ambiguous, potential mechanisms include molecular mimicry of spike proteins and activation of the adaptive immune response with dysregulated cytokine expression. Male predominance in COVID-19 vaccine-induced myocarditis may be attributed to sex hormones, variations in inflammatory reactions, coagulation states based on gender, and female-specific protective factors. Moreover, an analysis of diagnostic and management strategies reveals a lack of consensus on acute patient presentation management. Conclusion In contrast to viral infections that stand as the predominant etiological factor for myocarditis with more severe consequences, the mRNA COVID-19 vaccination elicits a mild and self-limiting manifestation of the condition. There is currently insufficient evidence to confirm the definite underlying mechanism of COVID-19 vaccine-associated myocarditis. Further research is needed to develop preventive and therapeutic solutions in this context.
Collapse
Affiliation(s)
| | - Foteini Moniati
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
7
|
Petta I, Thorp M, Ciers M, Blancke G, Boon L, Meese T, Van Nieuwerburgh F, Wullaert A, Grencis R, Elewaut D, van Loo G, Vereecke L. Myeloid A20 is critical for alternative macrophage polarization and type-2 immune-mediated helminth resistance. Front Immunol 2024; 15:1373745. [PMID: 38680500 PMCID: PMC11045979 DOI: 10.3389/fimmu.2024.1373745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024] Open
Abstract
Background Protective immunity against intestinal helminths requires induction of robust type-2 immunity orchestrated by various cellular and soluble effectors which promote goblet cell hyperplasia, mucus production, epithelial proliferation, and smooth muscle contractions to expel worms and re-establish immune homeostasis. Conversely, defects in type-2 immunity result in ineffective helminth clearance, persistent infection, and inflammation. Macrophages are highly plastic cells that acquire an alternatively activated state during helminth infection, but they were previously shown to be dispensable for resistance to Trichuris muris infection. Methods We use the in vivo mouse model A20myel-KO, characterized by the deletion of the potent anti-inflammatory factor A20 (TNFAIP3) specifically in the myeloid cells, the excessive type-1 cytokine production, and the development of spontaneous arthritis. We infect A20myel-KO mice with the gastrointestinal helminth Trichuris muris and we analyzed the innate and adaptive responses. We performed RNA sequencing on sorted myeloid cells to investigate the role of A20 on macrophage polarization and type-2 immunity. Moreover, we assess in A20myel-KO mice the pharmacological inhibition of type-1 cytokine pathways on helminth clearance and the infection with Salmonella typhimurium. Results We show that proper macrophage polarization is essential for helminth clearance, and we identify A20 as an essential myeloid factor for the induction of type-2 immune responses against Trichuris muris. A20myel-KO mice are characterized by persistent Trichuris muris infection and intestinal inflammation. Myeloid A20 deficiency induces strong classical macrophage polarization which impedes anti-helminth type-2 immune activation; however, it promotes detrimental Th1/Th17 responses. Antibody-mediated neutralization of the type-1 cytokines IFN-γ, IL-18, and IL-12 prevents myeloid-orchestrated Th1 polarization and re-establishes type-2-mediated protective immunity against T. muris in A20myel-KO mice. In contrast, the strong Th1-biased immunity in A20myel-KO mice offers protection against Salmonella typhimurium infection. Conclusions We hereby identify A20 as a critical myeloid factor for correct macrophage polarization and appropriate adaptive mucosal immunity in response to helminth and enteric bacterial infection.
Collapse
Affiliation(s)
- Ioanna Petta
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Marie Thorp
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Maarten Ciers
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Gillian Blancke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | | | - Tim Meese
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
- NXTGNT, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
- NXTGNT, Ghent University, Ghent, Belgium
| | - Andy Wullaert
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
- Cell Death Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Richard Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Dirk Elewaut
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lars Vereecke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
8
|
Liu K, Han B. Role of immune cells in the pathogenesis of myocarditis. J Leukoc Biol 2024; 115:253-275. [PMID: 37949833 DOI: 10.1093/jleuko/qiad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Myocarditis is an inflammatory heart disease that mostly affects young people. Myocarditis involves a complex immune network; however, its detailed pathogenesis is currently unclear. The diversity and plasticity of immune cells, either in the peripheral blood or in the heart, have been partially revealed in a number of previous studies involving patients and several kinds of animal models with myocarditis. It is the complexity of immune cells, rather than one cell type that is the culprit. Thus, recognizing the individual intricacies within immune cells in the context of myocarditis pathogenesis and finding the key intersection of the immune network may help in the diagnosis and treatment of this condition. With the vast amount of cell data gained on myocarditis and the recent application of single-cell sequencing, we summarize the multiple functions of currently recognized key immune cells in the pathogenesis of myocarditis to provide an immune background for subsequent investigations.
Collapse
Affiliation(s)
- Keyu Liu
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, 250021, Jinan, China
- Shandong Provincial Hospital, Shandong Provincial Clinical Research Center for Children' s Health and Disease office, No. 324 Jingwu Road, 250021, Jinan, China
| |
Collapse
|
9
|
Hayashi T, Lim KRQ, Kovacs A, Mann DL. Recurrent Adrenergic Stress Provokes Persistent Myocarditis in PD-1-Deficient Mice. JACC Basic Transl Sci 2023; 8:1503-1517. [PMID: 38205352 PMCID: PMC10774592 DOI: 10.1016/j.jacbts.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 01/12/2024]
Abstract
It is unclear how the immune system initiates effective tissue repair responses without also simultaneously activating adaptive immune responses to self-antigens released by damaged or necrotic cells. We studied the role of repetitive adrenergic mediated stress on cardiac injury wild-type and programmed death-1-deficient (PD-1-/-) mice treated with 3 intraperitoneal low doses of isoproterenol followed by an intraperitoneal injection of high-dose ISO 7 days later (ISOprimed/ISOinjury). Repetitive adrenergic stress in ISOprimed/ISOinjury PD-1-/- mice resulted in a persistent dysregulated myocardial inflammatory response characterized by the expansion of autoreactive effector CD8+ T cells, increased cardiac hypertrophy, mild left ventricular dysfunction, and increased lethality when compared with ISOprimed/ISOinjury wild-type mice.
Collapse
Affiliation(s)
- Tomohiro Hayashi
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Community Medicine and Career Development, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Rowel Q. Lim
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Attila Kovacs
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Douglas L. Mann
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
He W, Zhou L, Xu K, Li H, Wang JJ, Chen C, Wang D. Immunopathogenesis and immunomodulatory therapy for myocarditis. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2112-2137. [PMID: 37002488 PMCID: PMC10066028 DOI: 10.1007/s11427-022-2273-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/16/2023] [Indexed: 04/03/2023]
Abstract
Myocarditis is an inflammatory cardiac disease characterized by the destruction of myocardial cells, infiltration of interstitial inflammatory cells, and fibrosis, and is becoming a major public health concern. The aetiology of myocarditis continues to broaden as new pathogens and drugs emerge. The relationship between immune checkpoint inhibitors, severe acute respiratory syndrome coronavirus 2, vaccines against coronavirus disease-2019, and myocarditis has attracted increased attention. Immunopathological processes play an important role in the different phases of myocarditis, affecting disease occurrence, development, and prognosis. Excessive immune activation can induce severe myocardial injury and lead to fulminant myocarditis, whereas chronic inflammation can lead to cardiac remodelling and inflammatory dilated cardiomyopathy. The use of immunosuppressive treatments, particularly cytotoxic agents, for myocarditis, remains controversial. While reasonable and effective immunomodulatory therapy is the general trend. This review focuses on the current understanding of the aetiology and immunopathogenesis of myocarditis and offers new perspectives on immunomodulatory therapies.
Collapse
Affiliation(s)
- Wu He
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Ling Zhou
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Ke Xu
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Huihui Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - James Jiqi Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - DaoWen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
11
|
Cohen CD, Rousseau ST, Bermea KC, Bhalodia A, Lovell JP, Dina Zita M, Čiháková D, Adamo L. Myocardial Immune Cells: The Basis of Cardiac Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1198-1207. [PMID: 37068299 PMCID: PMC10111214 DOI: 10.4049/jimmunol.2200924] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/14/2023] [Indexed: 04/19/2023]
Abstract
The mammalian heart is characterized by the presence of striated myocytes, which allow continuous rhythmic contraction from early embryonic development until the last moments of life. However, the myocardium contains a significant contingent of leukocytes from every major class. This leukocyte pool includes both resident and nonresident immune cells. Over recent decades, it has become increasingly apparent that the heart is intimately sensitive to immune signaling and that myocardial leukocytes exhibit an array of critical functions, both in homeostasis and in the context of cardiac adaptation to injury. Here, we systematically review current knowledge of all major leukocyte classes in the heart, discussing their functions in health and disease. We also highlight the connection between the myocardium, immune cells, lymphoid organs, and both local and systemic immune responses.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sylvie T. Rousseau
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kevin C. Bermea
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aashik Bhalodia
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jana P. Lovell
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marcelle Dina Zita
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Luigi Adamo
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
12
|
Zhang Y, Shan L, Li D, Tang Y, Qian W, Dai J, Du M, Sun X, Zhu Y, Wang Q, Zhou L. Identification of key biomarkers associated with immune cells infiltration for myocardial injury in dermatomyositis by integrated bioinformatics analysis. Arthritis Res Ther 2023; 25:69. [PMID: 37118825 PMCID: PMC10142164 DOI: 10.1186/s13075-023-03052-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Dermatomyositis (DM) is an acquired autoimmune disease that can cause damage to various organs, including the heart muscle. However, the mechanisms underlying myocardial injury in DM are not yet fully understood. METHODS In this study, we utilized publicly available datasets from the Gene Expression Omnibus (GEO) database to identify hub-genes that are enriched in the immune system process in DM and myocarditis. Weighted gene co-expression network analysis (WGCNA), differentially expressed genes (DEGs) analysis, protein-protein interaction (PPI), and gene ontology (GO) analysis were employed to identify these hub-genes. We then used the CIBERSORT method to analyze immune cell infiltration in skeletal muscle specimens of DM and myocardium specimens of myocarditis respectively. Correlation analysis was performed to investigate the relationship between key genes and infiltrating immune cells. Finally, we predicted regulatory miRNAs of hub-genes through miRNet and validated their expression in online datasets and clinical samples. RESULTS Using integrated bioinformatics analysis, we identified 10 and 5 hub-genes that were enriched in the immune system process in the database of DM and myocarditis respectively. The subsequent intersections between hub-genes were IFIT3, OAS3, ISG15, and RSAD2. We found M2 macrophages increased in DM and myocarditis compared to the healthy control, associating with the expression of IFIT3, OAS3, ISG15, and RSAD2 in DM and myocarditis positively. Gene function enrichment analysis (GSEA) showed that IFIT3, OAS3, ISG15, and RSAD2 were mainly enriched in type I interferon (IFN) signaling pathway, cellular response to type I interferon, and response to type I interferon. Finally, we verified that the expression of miR-146a-5p was significantly higher in the DM with myocardial injury than those without myocardial injury (p = 0.0009). CONCLUSION Our findings suggest that IFIT3, OAS3, ISG15, and RSAD2 may play crucial roles in the underlying mechanism of myocardial injury in DM. Serum miR-146a-5p could be a potential biomarker for myocardial injury in DM.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Linwei Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongyu Li
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinghong Tang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Dai
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengdi Du
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoxuan Sun
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinsu Zhu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Lei Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Su X, Wang L, Ma N, Yang X, Liu C, Yang F, Li J, Yi X, Xing Y. Immune heterogeneity in cardiovascular diseases from a single-cell perspective. Front Cardiovasc Med 2023; 10:1057870. [PMID: 37180791 PMCID: PMC10167030 DOI: 10.3389/fcvm.2023.1057870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
A variety of immune cell subsets occupy different niches in the cardiovascular system, causing changes in the structure and function of the heart and vascular system, and driving the progress of cardiovascular diseases (CVDs). The immune cells infiltrating the injury site are highly diverse and integrate into a broad dynamic immune network that controls the dynamic changes of CVDs. Due to technical limitations, the effects and molecular mechanisms of these dynamic immune networks on CVDs have not been fully revealed. With recent advances in single-cell technologies such as single-cell RNA sequencing, systematic interrogation of the immune cell subsets is feasible and will provide insights into the way we understand the integrative behavior of immune populations. We no longer lightly ignore the role of individual cells, especially certain highly heterogeneous or rare subpopulations. We summarize the phenotypic diversity of immune cell subsets and their significance in three CVDs of atherosclerosis, myocardial ischemia and heart failure. We believe that such a review could enhance our understanding of how immune heterogeneity drives the progression of CVDs, help to elucidate the regulatory roles of immune cell subsets in disease, and thus guide the development of new immunotherapies.
Collapse
Affiliation(s)
- Xin Su
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Li Wang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, China
| | - Ning Ma
- Department of Breast Surgery, Dezhou Second People’s Hospital, Dezhou, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Can Liu
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Fan Yang
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Jun Li
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Xin Yi
- Department of Cardiology, Beijing Huimin Hospital, Beijing, China
| | - Yanwei Xing
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| |
Collapse
|
14
|
Feng G, Zhu C, Lin CY, Bredemeyer A, Förster I, Kreisel D, Lavine KJ. CCL17 Protects Against Viral Myocarditis by Suppressing the Recruitment of Regulatory T Cells. J Am Heart Assoc 2023; 12:e028442. [PMID: 36752267 PMCID: PMC10111487 DOI: 10.1161/jaha.122.028442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Background Viral myocarditis is characterized by leukocyte infiltration of the heart and cardiomyocyte death. We recently identified C-C chemokine ligand (CCL) 17 as a proinflammatory effector of C-C chemokine receptor 2-positive macrophages and dendritic cells that are recruited to the heart and contribute to adverse left ventricular remodeling following myocardial infarction and pressure overload. Methods and Results Mouse encephalomyocarditis virus was used to investigate the function of CCL17 in a viral myocarditis model. Ccl17Gfp reporter and knockout mice were used to identify the cell types that express CCL17 and delineate the functional importance of CCL17 in encephalomyocarditis virus clearance and myocardial inflammation. Cardiac CCL17 was expressed in C-C chemokine receptor 2-positive macrophages and dendritic cells following encephalomyocarditis virus infection. Colony-stimulating factor 2 (granulocyte-macrophage colony-stimulating factor) signaling was identified as a key regulator of CCL17 expression. Ccl17 deletion resulted in impaired encephalomyocarditis virus clearance, increased cardiomyocyte death, and higher mortality during infection early stage, and aggravated hypertrophy and fibrotic responses in infection long-term stage. An increased abundance of regulatory T cells was detected in the myocardium of injured Ccl17-deficient mice. Depletion of regulatory T cells in Ccl17-deficient mice abrogated the detrimental role of CCL17 deletion by restoring interferon signaling. Conclusions Collectively, these findings identify CCL17 as an important mediator of the host immune response during cardiac viral infection early stage and suggest that CCL17 targeted therapies should be avoided in acute viral myocarditis.
Collapse
Affiliation(s)
- Guoshuai Feng
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO
| | - Cuige Zhu
- Division of Oncology Washington University School of Medicine St. Louis MO
| | - Chieh-Yu Lin
- Department of Pathology and Immunology Washington University St. Louis MO
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO
| | - Irmgard Förster
- Immunology and Environment, LIMES Institute University of Bonn Germany
| | - Daniel Kreisel
- Department of Surgery Washington University St. Louis MO
| | - Kory J Lavine
- Cardiovascular Division, Department of Medicine Washington University School of Medicine St. Louis MO.,Department of Pathology and Immunology Washington University St. Louis MO.,Department of Developmental Biology Washington University St. Louis MO
| |
Collapse
|
15
|
Wienecke LM, Leid JM, Leuschner F, Lavine KJ. Imaging Targets to Visualize the Cardiac Immune Landscape in Heart Failure. Circ Cardiovasc Imaging 2023; 16:e014071. [PMID: 36649453 PMCID: PMC9858350 DOI: 10.1161/circimaging.122.014071] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Heart failure involves a complex interplay between diverse populations of immune cells that dynamically shift across the natural history of disease. Within this context, the character of the immune response is a key determinant of clinical outcomes. Recent technological advances in single-cell transcriptomic, spatial, and proteomic technologies have fueled an explosion of new and clinically relevant insights into distinct immune cell populations that reside within the diseased heart including potential targets for molecular imaging and therapy. In this review, we will discuss the immune cell types and their respective functions with respect to myocardial infarction remodeling, dilated cardiomyopathy, and heart failure with preserved ejection fraction. In addition, we give a brief overview regarding myocarditis and cardiac sarcoidosis as inflammatory heart failure etiologies. We will highlight markers and cell populations as targets for molecular imaging to visualize inflammation and tissue healing and discuss clinical implications including the development and implementation of precision medicine approaches.
Collapse
Affiliation(s)
- Laura M. Wienecke
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Jamison M. Leid
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Florian Leuschner
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Regenerative Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
16
|
Meeting the Challenges of Myocarditis: New Opportunities for Prevention, Detection, and Intervention—A Report from the 2021 National Heart, Lung, and Blood Institute Workshop. J Clin Med 2022; 11:jcm11195721. [PMID: 36233593 PMCID: PMC9571285 DOI: 10.3390/jcm11195721] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/05/2022] Open
Abstract
The National Heart, Lung, and Blood Institute (NHLBI) convened a workshop of international experts to discuss new research opportunities for the prevention, detection, and intervention of myocarditis in May 2021. These experts reviewed the current state of science and identified key gaps and opportunities in basic, diagnostic, translational, and therapeutic frontiers to guide future research in myocarditis. In addition to addressing community-acquired myocarditis, the workshop also focused on emerging causes of myocarditis including immune checkpoint inhibitors and SARS-CoV-2 related myocardial injuries and considered the use of systems biology and artificial intelligence methodologies to define workflows to identify novel mechanisms of disease and new therapeutic targets. A new priority is the investigation of the relationship between social determinants of health (SDoH), including race and economic status, and inflammatory response and outcomes in myocarditis. The result is a proposal for the reclassification of myocarditis that integrates the latest knowledge of immunological pathogenesis to refine estimates of prognosis and target pathway-specific treatments.
Collapse
|
17
|
Resident cardiac macrophages: Heterogeneity and function in health and disease. Immunity 2022; 55:1549-1563. [PMID: 36103852 DOI: 10.1016/j.immuni.2022.08.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/20/2022]
Abstract
Understanding tissue macrophage biology has become challenging in recent years due the ever-increasing complexity in macrophage-subset identification and functional characterization. This is particularly important within the myocardium, as we have come to understand that macrophages play multifaceted roles in cardiac health and disease, and heart disease remains the leading cause of death worldwide. Here, we review recent progress in the field, focusing on resident cardiac macrophage heterogeneity, origins, and functions at steady state and after injury. We stratify resident cardiac macrophage functions by the ability of macrophages to either directly influence cardiac physiology or indirectly influence cardiac physiology through orchestrating multi-cellular communication with cardiomyocytes and stromal and immune populations.
Collapse
|
18
|
A cardioimmunologist's toolkit: genetic tools to dissect immune cells in cardiac disease. Nat Rev Cardiol 2022; 19:395-413. [PMID: 35523863 DOI: 10.1038/s41569-022-00701-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Cardioimmunology is a field that encompasses the immune cells and pathways that modulate cardiac function in homeostasis and regulate the temporal balance between tissue injury and repair in disease. Over the past two decades, genetic fate mapping and high-dimensional sequencing techniques have defined increasing functional heterogeneity of innate and adaptive immune cell populations in the heart and other organs, revealing a complexity not previously appreciated and challenging established frameworks for the immune system. Given these rapid advances, understanding how to use these tools has become crucial. However, cardiovascular biologists without immunological expertise might not be aware of the strengths and caveats of immune-related tools and how they can be applied to examine the pathogenesis of myocardial diseases. In this Review, we guide readers through case-based examples to demonstrate how tool selection can affect data quality and interpretation and we provide critical analysis of the experimental tools that are currently available, focusing on their use in models of ischaemic heart injury and heart failure. The goal is to increase the use of relevant immunological tools and strategies among cardiovascular researchers to improve the precision, translatability and consistency of future studies of immune cells in cardiac disease.
Collapse
|
19
|
Feng G, Bajpai G, Ma P, Koenig A, Bredemeyer A, Lokshina I, Lai L, Förster I, Leuschner F, Kreisel D, Lavine KJ. CCL17 Aggravates Myocardial Injury by Suppressing Recruitment of Regulatory T Cells. Circulation 2022; 145:765-782. [PMID: 35113652 PMCID: PMC8957788 DOI: 10.1161/circulationaha.121.055888] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (β-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.
Collapse
Affiliation(s)
- Guoshuai Feng
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Geetika Bajpai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Pan Ma
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrew Koenig
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Inessa Lokshina
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | - Lulu Lai
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
| | | | - Florian Leuschner
- LIMES Institute, University of Bonn, Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany (F.L.)
| | - Daniel Kreisel
- Department of Surgery, Washington University, Saint Louis, Missouri, USA (D.K.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA (G.F., G.B., P.M., A.K., A.B., I.L., L.L., K.L.)
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA (D.K., K.L.)
- Department of Developmental Biology, Washington University, Saint Louis, Missouri, USA (K.L.)
| |
Collapse
|
20
|
Lee SE, Rudd BD, Smith NL. Fate-mapping mice: new tools and technology for immune discovery. Trends Immunol 2022; 43:195-209. [PMID: 35094945 PMCID: PMC8882138 DOI: 10.1016/j.it.2022.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/20/2022]
Abstract
The fate-mapping mouse has become an essential tool in the immunologist's toolbox. Although traditionally used by developmental biologists to trace the origins of cells, immunologists are turning to fate-mapping to better understand the development and function of immune cells. Thus, an expansion in the variety of fate-mapping mouse models has occurred to answer fundamental questions about the immune system. These models are also being combined with new genetic tools to study cancer, infection, and autoimmunity. In this review, we summarize different types of fate-mapping mice and describe emerging technologies that might allow immunologists to leverage this valuable tool and expand our functional knowledge of the immune system.
Collapse
Affiliation(s)
- Scarlett E Lee
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14850, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14850, USA
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
21
|
Kuna J, Żuber Z, Chmielewski G, Gromadziński L, Krajewska-Włodarczyk M. Role of Distinct Macrophage Populations in the Development of Heart Failure in Macrophage Activation Syndrome. Int J Mol Sci 2022; 23:2433. [PMID: 35269577 PMCID: PMC8910409 DOI: 10.3390/ijms23052433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is one of the few entities in rheumatology with the potential to quickly cause multiple organ failure and loss of life, and as such, requires urgent clinical intervention. It has a broad symptomatology, depending on the organs it affects. One especially dangerous aspect of MAS's course of illness is myocarditis leading to acute heart failure and possibly death. Research in recent years has proved that macrophages settled in different organs are not a homogenous group, with particular populations differing in both structure and function. Within the heart, we can determine two major groups, based on the presence of the C-C 2 chemokine receptor (CCR2): CCR2+ and CCR2-. There are a number of studies describing their function and the changes in the population makeup between normal conditions and different illnesses; however, to our knowledge, there has not been one touching on the matter of changes occurring in the populations of heart macrophages during MAS and their possible consequences. This review summarizes the most recent knowledge on heart macrophages, the influence of select cytokines (those particularly significant in the development of MAS) on their activity, and both the immediate and long-term consequences of changes in the makeup of specific macrophage populations-especially the loss of CCR2- cells that are responsible for regenerative processes, as well as the substitution of tissue macrophages by the highly proinflammatory CCR2+ macrophages originating from circulating monocytes. Understanding the significance of these processes may lead to new discoveries that could improve the therapeutic methods in the treatment of MAS.
Collapse
Affiliation(s)
- Jakub Kuna
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Kraków University, 30-705 Kraków, Poland;
| | - Grzegorz Chmielewski
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| |
Collapse
|
22
|
Characterization of Transverse Aortic Constriction in Mice Based on the Specific Recruitment of Leukocytes to the Hypertrophic Myocardium and the Aorta Ascendens. Mediators Inflamm 2021; 2021:1376859. [PMID: 34776787 PMCID: PMC8580661 DOI: 10.1155/2021/1376859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Transverse aortic constriction (TAC) is a model that mimics pressure overload-induced left ventricular (LV) hypertrophy in mice. Alterations in immune cell functionality can promote cardiac and vascular remodeling. In the present study, we characterized the time course in innate immune cell dynamics in response to TAC in the different tissues of mice. It was determined whether TAC induces a characteristic leukocyte-driven immune response in the myocardium, aorta ascendens and descendens, spleen, blood, and draining lymph nodes supported by cytokine-driven chemotaxis in mice at 3, 6, and 21 days following surgery. We used complex flow cytometry staining combinations to characterize the various innate immune cell subsets and a multiplex array to determine cytokine concentrations in the serum. The results of the current study indicated that leukocytes accumulate in the myocardium and aorta ascendens in response to TAC. The leukocyte dynamics in the myocardium were dominated by the Ly6Clow macrophages with an early accumulation, whereas the response in the aorta ascendens was characterized by a long-lasting proinflammatory phenotype driven by Ly6Chigh macrophages, neutrophils, and activated DCs. In contrast to the high-pressure environment of the aorta ascendens, the tissue of the aorta descendens did not react to TAC with any leukocyte increase. The levels of proinflammatory cytokines in the blood were elevated in response to TAC, indicating a systemic reaction. Moreover, our findings strongly suggest that cardiac macrophages could origin from splenic pools and reach the site of the inflammation via the blood. Based on the current findings, it can be concluded that the high-pressure conditions in the aorta ascendens cause a characteristic immune response, dominated by the accumulation of leukocytes and the activation of DCs that varies in comparison to the immune cell dynamics in the myocardium and the aorta descendens.
Collapse
|
23
|
Bickett TE, Knitz M, Darragh LB, Bhatia S, Van Court B, Gadwa J, Bhuvane S, Piper M, Nguyen D, Tu H, Lenz L, Clambey ET, Barry K, Karam SD. FLT3L Release by Natural Killer Cells Enhances Response to Radioimmunotherapy in Preclinical Models of HNSCC. Clin Cancer Res 2021; 27:6235-6249. [PMID: 34518311 PMCID: PMC8595694 DOI: 10.1158/1078-0432.ccr-21-0971] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/12/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Natural killer (NK) cells are type I innate lymphoid cells that are known for their role in killing virally infected cells or cancer cells through direct cytotoxicity. In addition to direct tumor cell killing, NK cells are known to play fundamental roles in the tumor microenvironment through secretion of key cytokines, such as FMS-like tyrosine kinase 3 ligand (FLT3L). Although radiotherapy is the mainstay treatment in most cancers, the role of radiotherapy on NK cells is not well characterized. EXPERIMENTAL DESIGN This study combines radiation, immunotherapies, genetic mouse models, and antibody depletion experiments to identify the role of NK cells in overcoming resistance to radiotherapy in orthotopic models of head and neck squamous cell carcinoma. RESULTS We have found that NK cells are a crucial component in the development of an antitumor response, as depleting them removes efficacy of the previously successful combination treatment of radiotherapy, anti-CD25, and anti-CD137. However, in the absence of NK cells, the effect can be rescued through treatment with FLT3L. But neither radiotherapy with FLT3L therapy alone nor radiotherapy with anti-NKG2A yields any meaningful tumor growth delay. We also identify a role for IL2 in activating NK cells to secrete FLT3L. This activity, we show, is mediated through CD122, the intermediate affinity IL2 receptor, and can be targeted with anti-CD25 therapy. CONCLUSIONS These findings highlight the complexity of using radio-immunotherapies to activate NK cells within the tumor microenvironment, and the importance of NK cells in activating dendritic cells for increased tumor surveillance.
Collapse
Affiliation(s)
- Thomas E Bickett
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Michael Knitz
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Shiv Bhuvane
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Hua Tu
- Lake Pharma, The Biologics Company, San Francisco, California
| | - Laurel Lenz
- Department of Immunology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Eric T Clambey
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kevin Barry
- Immunotherapy Integrated Research Center, Fred Hutchinson Research Institute, Seattle, Washington
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
24
|
Cardiac hybrid imaging: novel tracers for novel targets. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2021; 18:748-758. [PMID: 34659381 PMCID: PMC8501382 DOI: 10.11909/j.issn.1671-5411.2021.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Non-invasive cardiac imaging has explored enormous advances in the last few decades. In particular, hybrid imaging represents the fusion of information from multiple imaging modalities, allowing to provide a more comprehensive dataset compared to traditional imaging techniques in patients with cardiovascular diseases. The complementary anatomical, functional and molecular information provided by hybrid systems are able to simplify the evaluation procedure of various pathologies in a routine clinical setting. The diagnostic capability of hybrid imaging modalities can be further enhanced by introducing novel and specific imaging biomarkers. The aim of this review is to cover the most recent advancements in radiotracers development for SPECT/CT, PET/CT, and PET/MRI for cardiovascular diseases.
Collapse
|
25
|
Wang T, Yang Y, Wang D. Letter by Wang et al Regarding Article, "Cross-Priming Dendritic Cells Exacerbate Immunopathology After Ischemic Tissue Damage in the Heart". Circulation 2021; 144:e92-e93. [PMID: 34339309 DOI: 10.1161/circulationaha.121.055114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ti Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Jiangsu, China. Yangzhou University Medical College, Jiangsu, China
| | - Yuxue Yang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Jiangsu, China. Yangzhou University Medical College, Jiangsu, China
| | - Daxin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Jiangsu, China. Yangzhou University Medical College, Jiangsu, China
| |
Collapse
|
26
|
Abstract
Conduction disorders and arrhythmias remain difficult to treat and are increasingly prevalent owing to the increasing age and body mass of the general population, because both are risk factors for arrhythmia. Many of the underlying conditions that give rise to arrhythmia - including atrial fibrillation and ventricular arrhythmia, which frequently occur in patients with acute myocardial ischaemia or heart failure - can have an inflammatory component. In the past, inflammation was viewed mostly as an epiphenomenon associated with arrhythmia; however, the recently discovered inflammatory and non-canonical functions of cardiac immune cells indicate that leukocytes can be arrhythmogenic either by altering tissue composition or by interacting with cardiomyocytes; for example, by changing their phenotype or perhaps even by directly interfering with conduction. In this Review, we discuss the electrophysiological properties of leukocytes and how these cells relate to conduction in the heart. Given the thematic parallels, we also summarize the interactions between immune cells and neural systems that influence information transfer, extrapolating findings from the field of neuroscience to the heart and defining common themes. We aim to bridge the knowledge gap between electrophysiology and immunology, to promote conceptual connections between these two fields and to explore promising opportunities for future research.
Collapse
|
27
|
Leuschner F, Nahrendorf M. Novel functions of macrophages in the heart: insights into electrical conduction, stress, and diastolic dysfunction. Eur Heart J 2021; 41:989-994. [PMID: 30945736 DOI: 10.1093/eurheartj/ehz159] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/12/2019] [Accepted: 03/25/2019] [Indexed: 12/24/2022] Open
Abstract
Over a century ago, Élie Metchnikoff described the macrophages' ability to phagocytose. Propelled by advances in technology enabling phenotypic and functional analyses at unpreceded resolution, a recent renaissance in macrophage research has shed new light on these 'big eaters'. We here give an overview of cardiac macrophages' provenance in the contexts of cardiac homeostasis and stress. We highlight the recently identified mechanism by which these cells regulate electrical conduction in the atrioventricular node and discuss why we need a deeper understanding of monocytes and macrophages in systolic and diastolic dysfunctions.
Collapse
Affiliation(s)
- Florian Leuschner
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,Partner site Heidelberg, DZHK (German Centre for Cardiovascular Research), Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA.,Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
28
|
Marín-Sedeño E, de Morentin XM, Pérez-Pomares JM, Gómez-Cabrero D, Ruiz-Villalba A. Understanding the Adult Mammalian Heart at Single-Cell RNA-Seq Resolution. Front Cell Dev Biol 2021; 9:645276. [PMID: 34055776 PMCID: PMC8149764 DOI: 10.3389/fcell.2021.645276] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/09/2021] [Indexed: 12/24/2022] Open
Abstract
During the last decade, extensive efforts have been made to comprehend cardiac cell genetic and functional diversity. Such knowledge allows for the definition of the cardiac cellular interactome as a reasonable strategy to increase our understanding of the normal and pathologic heart. Previous experimental approaches including cell lineage tracing, flow cytometry, and bulk RNA-Seq have often tackled the analysis of cardiac cell diversity as based on the assumption that cell types can be identified by the expression of a single gene. More recently, however, the emergence of single-cell RNA-Seq technology has led us to explore the diversity of individual cells, enabling the cardiovascular research community to redefine cardiac cell subpopulations and identify relevant ones, and even novel cell types, through their cell-specific transcriptomic signatures in an unbiased manner. These findings are changing our understanding of cell composition and in consequence the identification of potential therapeutic targets for different cardiac diseases. In this review, we provide an overview of the continuously changing cardiac cellular landscape, traveling from the pre-single-cell RNA-Seq times to the single cell-RNA-Seq revolution, and discuss the utilities and limitations of this technology.
Collapse
Affiliation(s)
- Ernesto Marín-Sedeño
- Department of Animal Biology, Faculty of Sciences, Instituto Malagueño de Biomedicina, University of Málaga, Málaga, Spain
- BIONAND, Centro Andaluz de Nanomedicina y Biotecnología, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Xabier Martínez de Morentin
- Traslational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad Pública de Navarra, Pamplona, Spain
| | - Jose M. Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, Instituto Malagueño de Biomedicina, University of Málaga, Málaga, Spain
- BIONAND, Centro Andaluz de Nanomedicina y Biotecnología, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - David Gómez-Cabrero
- Traslational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad Pública de Navarra, Pamplona, Spain
- Centre of Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Adrián Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, Instituto Malagueño de Biomedicina, University of Málaga, Málaga, Spain
- BIONAND, Centro Andaluz de Nanomedicina y Biotecnología, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
29
|
Go DM, Lee SH, Lee SH, Woo SH, Kim K, Kim K, Park KS, Park JH, Ha SJ, Kim WH, Choi JH, Kim DY. Programmed Death Ligand 1-Expressing Classical Dendritic Cells MitigateHelicobacter-Induced Gastritis. Cell Mol Gastroenterol Hepatol 2021; 12:715-739. [PMID: 33894424 PMCID: PMC8267570 DOI: 10.1016/j.jcmgh.2021.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Helicobacter pylori has been reported to modulate local immune responses to colonize persistently in gastric mucosa. Although the induced expression of programmed cell death ligand 1 (PD-L1) has been suggested as an immune modulatory mechanism for persistent infection of H pylori, the main immune cells expressing PD-L1 and their functions in Helicobacter-induced gastritis still remain to be elucidated. METHODS The blockades of PD-L1 with antibody or PD-L1-deficient bone marrow transplantation were performed in Helicobacter-infected mice. The main immune cells expressing PD-L1 in Helicobacter-infected stomach were determined by flow cytometry and immunofluorescence staining. Helicobacter felis or H pylori-infected dendritic cell (DC)-deficient mouse models including Flt3-/-, Zbtb46-diphtheria toxin receptor, and BDCA2-diphtheria toxin receptor mice were analyzed for pathologic changes and colonization levels. Finally, the location of PD-L1-expressing DCs and the correlation with H pylori infection were analyzed in human gastric tissues using multiplexed immunohistochemistry. RESULTS Genetic or antibody-mediated blockade of PD-L1 aggravated Helicobacter-induced gastritis with mucosal metaplasia. Gastric classical DCs expressed considerably higher levels of PD-L1 than other immune cells and co-localized with T cells in gastritis lesions from Helicobacter-infected mice and human beings. H felis- or H pylori-infected Flt3-/- or classical DC-depleted mice showed aggravated gastritis with severe T-cell and neutrophil accumulation with low bacterial loads compared with that in control mice. Finally, PD-L1-expressing DCs were co-localized with T cells and showed a positive correlation with H pylori infection in human subjects. CONCLUSIONS The PD-1/PD-L1 pathway may be responsible for the immune modulatory function of gastric DCs that protects the gastric mucosa from Helicobacter-induced inflammation, but allows persistent Helicobacter colonization.
Collapse
Affiliation(s)
- Du-Min Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seung Hyun Lee
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Su-Hyung Lee
- Division of Cancer Biology, Research Institute of National Cancer Center, Gyeonggi-do, Republic of Korea
| | - Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kibyeong Kim
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyeongdae Kim
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyu Seong Park
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Woo Ho Kim
- Department of Pathology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea.
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Sun K, Li YY, Jin J. A double-edged sword of immuno-microenvironment in cardiac homeostasis and injury repair. Signal Transduct Target Ther 2021; 6:79. [PMID: 33612829 PMCID: PMC7897720 DOI: 10.1038/s41392-020-00455-6] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/14/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
The response of immune cells in cardiac injury is divided into three continuous phases: inflammation, proliferation and maturation. The kinetics of the inflammatory and proliferation phases directly influence the tissue repair. In cardiac homeostasis, cardiac tissue resident macrophages (cTMs) phagocytose bacteria and apoptotic cells. Meanwhile, NK cells prevent the maturation and transport of inflammatory cells. After cardiac injury, cTMs phagocytose the dead cardiomyocytes (CMs), regulate the proliferation and angiogenesis of cardiac progenitor cells. NK cells prevent the cardiac fibrosis, and promote vascularization and angiogenesis. Type 1 macrophages trigger the cardioprotective responses and promote tissue fibrosis in the early stage. Reversely, type 2 macrophages promote cardiac remodeling and angiogenesis in the late stage. Circulating macrophages and neutrophils firstly lead to chronic inflammation by secreting proinflammatory cytokines, and then release anti-inflammatory cytokines and growth factors, which regulate cardiac remodeling. In this process, dendritic cells (DCs) mediate the regulation of monocyte and macrophage recruitment. Recruited eosinophils and Mast cells (MCs) release some mediators which contribute to coronary vasoconstriction, leukocyte recruitment, formation of new blood vessels, scar formation. In adaptive immunity, effector T cells, especially Th17 cells, lead to the pathogenesis of cardiac fibrosis, including the distal fibrosis and scar formation. CMs protectors, Treg cells, inhibit reduce the inflammatory response, then directly trigger the regeneration of local progenitor cell via IL-10. B cells reduce myocardial injury by preserving cardiac function during the resolution of inflammation.
Collapse
Affiliation(s)
- Kang Sun
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Yuan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
- Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
31
|
Lafuse WP, Wozniak DJ, Rajaram MVS. Role of Cardiac Macrophages on Cardiac Inflammation, Fibrosis and Tissue Repair. Cells 2020; 10:E51. [PMID: 33396359 PMCID: PMC7824389 DOI: 10.3390/cells10010051] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.
Collapse
Affiliation(s)
- William P. Lafuse
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
- Department of Microbiology, Ohio State University, Columbus, OH 43210, USA
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| |
Collapse
|
32
|
Zaman R, Hamidzada H, Epelman S. Exploring cardiac macrophage heterogeneity in the healthy and diseased myocardium. Curr Opin Immunol 2020; 68:54-63. [PMID: 33128959 DOI: 10.1016/j.coi.2020.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Cardiac macrophages maintain homeostasis and orchestrate response to disease. Utilization of genetic fate-mapping and single-cell RNA sequencing shifted the paradigm of macrophage heterogeneity from the canonical M1/M2 classification in favour of a nuanced approach that reconciles divergent origins, lifecycles, and transcriptional states. Here, we provide a conceptual framework to assess cardiac macrophage complexity that integrates transcriptional and functional heterogeneity that tracks with subset-specific markers (TIMD4 and CCR2). Our goal is to provide a starting point for researchers to dissect the functions of known resident cardiac macrophage subpopulations. We discuss recent advances and limitations in our understanding of cardiac macrophage diversity in ischemic injury, hypertension and myocarditis.
Collapse
Affiliation(s)
- Rysa Zaman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada.
| |
Collapse
|
33
|
Abstract
Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.
Collapse
|
34
|
Steffens S, Van Linthout S, Sluijter JPG, Tocchetti CG, Thum T, Madonna R. Stimulating pro-reparative immune responses to prevent adverse cardiac remodelling: consensus document from the joint 2019 meeting of the ESC Working Groups of cellular biology of the heart and myocardial function. Cardiovasc Res 2020; 116:1850-1862. [DOI: 10.1093/cvr/cvaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Cardiac injury may have multiple causes, including ischaemic, non-ischaemic, autoimmune, and infectious triggers. Independent of the underlying pathophysiology, cardiac tissue damage induces an inflammatory response to initiate repair processes. Immune cells are recruited to the heart to remove dead cardiomyocytes, which is essential for cardiac healing. Insufficient clearance of dying cardiomyocytes after myocardial infarction (MI) has been shown to promote unfavourable cardiac remodelling, which may result in heart failure (HF). Although immune cells are integral key players of cardiac healing, an unbalanced or unresolved immune reaction aggravates tissue damage that triggers maladaptive remodelling and HF. Neutrophils and macrophages are involved in both, inflammatory as well as reparative processes. Stimulating the resolution of cardiac inflammation seems to be an attractive therapeutic strategy to prevent adverse remodelling. Along with numerous experimental studies, the promising outcomes from recent clinical trials testing canakinumab or colchicine in patients with MI are boosting the interest in novel therapies targeting inflammation in cardiovascular disease patients. The aim of this review is to discuss recent experimental studies that provide new insights into the signalling pathways and local regulators within the cardiac microenvironment promoting the resolution of inflammation and tissue regeneration. We will cover ischaemia- and non-ischaemic-induced as well as infection-related cardiac remodelling and address potential targets to prevent adverse cardiac remodelling.
Collapse
Affiliation(s)
- Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Via Paradisa, Pisa 56124, Italy
| |
Collapse
|
35
|
Okyere AD, Tilley DG. Leukocyte-Dependent Regulation of Cardiac Fibrosis. Front Physiol 2020; 11:301. [PMID: 32322219 PMCID: PMC7156539 DOI: 10.3389/fphys.2020.00301] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Cardiac fibrosis begins as an intrinsic response to injury or ageing that functions to preserve the tissue from further damage. Fibrosis results from activated cardiac myofibroblasts, which secrete extracellular matrix (ECM) proteins in an effort to replace damaged tissue; however, excessive ECM deposition leads to pathological fibrotic remodeling. At this extent, fibrosis gravely disturbs myocardial compliance, and ultimately leads to adverse outcomes like heart failure with heightened mortality. As such, understanding the complexity behind fibrotic remodeling has been a focal point of cardiac research in recent years. Resident cardiac fibroblasts and activated myofibroblasts have been proven integral to the fibrotic response; however, several findings point to additional cell types that may contribute to the development of pathological fibrosis. For one, leukocytes expand in number after injury and exhibit high plasticity, thus their distinct role(s) in cardiac fibrosis is an ongoing and controversial field of study. This review summarizes current findings, focusing on both direct and indirect leukocyte-mediated mechanisms of fibrosis, which may provide novel targeted strategies against fibrotic remodeling.
Collapse
Affiliation(s)
- Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Abstract
The prevalence of heart failure (HF), including reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF), has increased significantly worldwide. However, the prognosis and treatment of HF are still not good. Recent studies have demonstrated that high-density lipoprotein (HDL) plays an important role in cardiac repair during HF. The exact role and mechanism of HDL in the regulation of HF remain unexplained. Here, we discuss recent findings regarding HDL in the progression of HF, such as the regulation of excitation-contraction coupling, energy homeostasis, inflammation, neurohormone activation, and microvascular dysfunction. The effects of HDL on the regulation of cardiac-related cells, such as endothelial cells (ECs), cardiomyocytes (CMs), and on cardiac resident immune cell dysfunction in HF are also explained. An in-depth understanding of HDL function in the heart may provide new strategies for the prevention and treatment of HF.
Collapse
|
37
|
Epelman S. Microbes and genes in heart failure. Science 2019; 366:806-807. [PMID: 31727821 DOI: 10.1126/science.aaz4400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Slava Epelman
- Department of Immunology, University of Toronto, Heart & Stroke/Richard Lewar Centre of Excellence, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Rhee AJ, Lavine KJ. New Approaches to Target Inflammation in Heart Failure: Harnessing Insights from Studies of Immune Cell Diversity. Annu Rev Physiol 2019; 82:1-20. [PMID: 31658002 DOI: 10.1146/annurev-physiol-021119-034412] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite mounting evidence implicating inflammation in cardiovascular diseases, attempts at clinical translation have shown mixed results. Recent preclinical studies have reenergized this field and provided new insights into how to favorably modulate cardiac macrophage function in the context of acute myocardial injury and chronic disease. In this review, we discuss the origins and roles of cardiac macrophage populations in the steady-state and diseased heart, focusing on the human heart and mouse models of ischemia, hypertensive heart disease, and aortic stenosis. Specific attention is given to delineating the roles of tissue-resident and recruited monocyte-derived macrophage subsets. We also highlight emerging concepts of monocyte plasticity and heterogeneity among monocyte-derived macrophages, describe possible mechanisms by which infiltrating monocytes acquire unique macrophage fates, and discuss the putative impact of these populations on cardiac remodeling. Finally, we discuss strategies to target inflammatory macrophage populations.
Collapse
Affiliation(s)
- Aaron J Rhee
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; .,Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.,Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
39
|
Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, Rosenthal N, Kovacic JC. The Macrophage in Cardiac Homeostasis and Disease: JACC Macrophage in CVD Series (Part 4). J Am Coll Cardiol 2019; 72:2213-2230. [PMID: 30360829 DOI: 10.1016/j.jacc.2018.08.2149] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [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: 12/24/2022]
Abstract
Macrophages are integral components of cardiac tissue and exert profound effects on the healthy and diseased heart. Paradigm shifting studies using advanced molecular techniques have revealed significant complexity within these macrophage populations that reside in the heart. In this final of a 4-part review series covering the macrophage in cardiovascular disease, the authors review the origins, dynamics, cell surface markers, and respective functions of each cardiac macrophage subset identified to date, including in the specific scenarios of myocarditis and after myocardial infarction. Looking ahead, a deeper understanding of the diverse and often dichotomous functions of cardiac macrophages will be essential for the development of targeted therapies to mitigate injury and orchestrate recovery of the diseased heart. Moreover, as macrophages are critical for cardiac healing, they are an emerging focus for therapeutic strategies aimed at minimizing cardiomyocyte death, ameliorating pathological cardiac remodeling, and for treating heart failure and after myocardial infarction.
Collapse
Affiliation(s)
- Kory J Lavine
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri; Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, Melbourne, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, Toronto, Ontario, Canada
| | - Benjamin J Kopecky
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, Maine; Mt. Desert Island Biological Laboratory, Bar Harbor, Maine
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
40
|
Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol 2019; 18:733-744. [PMID: 30228378 DOI: 10.1038/s41577-018-0065-8] [Citation(s) in RCA: 532] [Impact Index Per Article: 88.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The past few decades have generated growing recognition that the immune system makes an important contribution to cardiac development, composition and function. Immune cells infiltrate the heart at gestation and remain in the myocardium, where they participate in essential housekeeping functions throughout life. After myocardial infarction or in response to infection, large numbers of immune cells are recruited to the heart to remove dying tissue, scavenge pathogens and promote healing. Under some circumstances, immune cells can cause irreversible damage, contributing to heart failure. This Review focuses on the role of the immune system in the heart under both homeostatic and perturbed conditions.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Affiliation(s)
| | - Matthias Nahrendorf
- Center for Systems Biology (S.C., M.N.) .,Cardiovascular Research Center (M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
42
|
Psarras S, Beis D, Nikouli S, Tsikitis M, Capetanaki Y. Three in a Box: Understanding Cardiomyocyte, Fibroblast, and Innate Immune Cell Interactions to Orchestrate Cardiac Repair Processes. Front Cardiovasc Med 2019; 6:32. [PMID: 31001541 PMCID: PMC6454035 DOI: 10.3389/fcvm.2019.00032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Following an insult by both intrinsic and extrinsic pathways, complex cellular, and molecular interactions determine a successful recovery or inadequate repair of damaged tissue. The efficiency of this process is particularly important in the heart, an organ characterized by very limited regenerative and repair capacity in higher adult vertebrates. Cardiac insult is characteristically associated with fibrosis and heart failure, as a result of cardiomyocyte death, myocardial degeneration, and adverse remodeling. Recent evidence implies that resident non-cardiomyocytes, fibroblasts but also macrophages -pillars of the innate immunity- form part of the inflammatory response and decisively affect the repair process following a cardiac insult. Multiple studies in model organisms (mouse, zebrafish) of various developmental stages (adult and neonatal) combined with genetically engineered cell plasticity and differentiation intervention protocols -mainly targeting cardiac fibroblasts or progenitor cells-reveal particular roles of resident and recruited innate immune cells and their secretome in the coordination of cardiac repair. The interplay of innate immune cells with cardiac fibroblasts and cardiomyocytes is emerging as a crucial platform to help our understanding and, importantly, to allow the development of effective interventions sufficient to minimize cardiac damage and dysfunction after injury.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
43
|
Heo GS, Kopecky B, Sultan D, Ou M, Feng G, Bajpai G, Zhang X, Luehmann H, Detering L, Su Y, Leuschner F, Combadière C, Kreisel D, Gropler RJ, Brody SL, Liu Y, Lavine KJ. Molecular Imaging Visualizes Recruitment of Inflammatory Monocytes and Macrophages to the Injured Heart. Circ Res 2019; 124:881-890. [PMID: 30661445 PMCID: PMC6435034 DOI: 10.1161/circresaha.118.314030] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/14/2019] [Indexed: 12/19/2022]
Abstract
RATIONALE Paradigm shifting studies have revealed that the heart contains functionally diverse populations of macrophages derived from distinct embryonic and adult hematopoietic progenitors. Under steady-state conditions, the heart is largely populated by CCR2- (C-C chemokine receptor type 2) macrophages of embryonic descent. After tissue injury, a dramatic shift in macrophage composition occurs whereby CCR2+ monocytes are recruited to the heart and differentiate into inflammatory CCR2+ macrophages that contribute to heart failure progression. Currently, there are no techniques to noninvasively detect CCR2+ monocyte recruitment into the heart and thus identify patients who may be candidates for immunomodulatory therapy. OBJECTIVE To develop a noninvasive molecular imaging strategy with high sensitivity and specificity to visualize inflammatory monocyte and macrophage accumulation in the heart. METHODS AND RESULTS We synthesized and tested the performance of a positron emission tomography radiotracer (68Ga-DOTA [1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid]-ECL1i [extracellular loop 1 inverso]) that allosterically binds to CCR2. In naive mice, the radiotracer was quickly cleared from the blood and displayed minimal retention in major organs. In contrast, biodistribution and positron emission tomography demonstrated strong myocardial tracer uptake in 2 models of cardiac injury (diphtheria toxin induced cardiomyocyte ablation and reperfused myocardial infarction). 68Ga-DOTA-ECL1i signal localized to sites of tissue injury and was independent of blood pool activity as assessed by quantitative positron emission tomography and ex vivo autoradiography. 68Ga-DOTA-ECL1i uptake was associated with CCR2+ monocyte and CCR2+ macrophage infiltration into the heart and was abrogated in CCR2-/- mice, demonstrating target specificity. Autoradiography demonstrated that 68Ga-DOTA-ECL1i specifically binds human heart failure specimens and with signal intensity associated with CCR2+ macrophage abundance. CONCLUSIONS These findings demonstrate the sensitivity and specificity of 68Ga-DOTA-ECL1i in the mouse heart and highlight the translational potential of this agent to noninvasively visualize CCR2+ monocyte recruitment and inflammatory macrophage accumulation in patients.
Collapse
Affiliation(s)
- Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Benjamin Kopecky
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Monica Ou
- Department of Biology, Saint Louis University, St. Louis, MO USA
| | - Guoshuai Feng
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Geetika Bajpai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Xiaohui Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Yi Su
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Florian Leuschner
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Christophe Combadière
- Sorbonne Université, Inserm, CNRS, Centre d’immunologie et des maladies infectieuses, Cimi-Paris, F-75013 Paris, France
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO USA
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
| | - Robert J. Gropler
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Kory J. Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO USA
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
44
|
Anzai A, Mindur JE, Halle L, Sano S, Choi JL, He S, McAlpine CS, Chan CT, Kahles F, Valet C, Fenn AM, Nairz M, Rattik S, Iwamoto Y, Fairweather D, Walsh K, Libby P, Nahrendorf M, Swirski FK. Self-reactive CD4 + IL-3 + T cells amplify autoimmune inflammation in myocarditis by inciting monocyte chemotaxis. J Exp Med 2019; 216:369-383. [PMID: 30670465 PMCID: PMC6363430 DOI: 10.1084/jem.20180722] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 12/07/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
Acquisition of self-reactive effector CD4+ T cells is a major component of the autoimmune response that can occur during myocarditis, an inflammatory form of cardiomyopathy. Although the processes by which self-reactive T cells gain effector function have received considerable attention, how these T cells contribute to effector organ inflammation and damage is less clear. Here, we identified an IL-3-dependent amplification loop that exacerbates autoimmune inflammation. In experimental myocarditis, we show that effector organ-accumulating autoreactive IL-3+ CD4+ T cells stimulate IL-3R+ tissue macrophages to produce monocyte-attracting chemokines. The newly recruited monocytes differentiate into antigen-presenting cells that stimulate local IL-3+ CD4+ T cell proliferation, thereby amplifying organ inflammation. Consequently, Il3 -/- mice resist developing robust autoimmune inflammation and myocardial dysfunction, whereas therapeutic IL-3 targeting ameliorates disease. This study defines a mechanism that orchestrates inflammation in myocarditis, describes a previously unknown function for IL-3, and identifies IL-3 as a potential therapeutic target in patients with myocarditis.
Collapse
Affiliation(s)
- Atsushi Anzai
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - John E Mindur
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lennard Halle
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Jennifer L Choi
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Shun He
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Cameron S McAlpine
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Christopher T Chan
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Florian Kahles
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Colin Valet
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ashley M Fenn
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Manfred Nairz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sara Rattik
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
45
|
Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, Chen J, Kantores C, Hosseinzadeh S, Aronoff L, Wong A, Zaman R, Barbu I, Besla R, Lavine KJ, Razani B, Ginhoux F, Husain M, Cybulsky MI, Robbins CS, Epelman S. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol 2019; 20:29-39. [PMID: 30538339 PMCID: PMC6565365 DOI: 10.1038/s41590-018-0272-2] [Citation(s) in RCA: 596] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/30/2018] [Indexed: 12/14/2022]
Abstract
Macrophages promote both injury and repair after myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping, parabiosis and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2- resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4-LYVE1-MHC-IIhiCCR2- macrophages and fully replaced TIMD4-LYVE1-MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4- resident macrophage abundance, whereas CCR2+ monocyte-derived macrophages adopted multiple cell fates within infarcted tissue, including those nearly indistinguishable from resident macrophages. Recruited macrophages did not express TIMD4, highlighting the ability of TIMD4 to track a subset of resident macrophages in the absence of fate mapping. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, revealing a nonredundant, cardioprotective role of resident cardiac macrophages.
Collapse
Affiliation(s)
- Sarah A Dick
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Ted Rogers Centre for Heart Research, Toronto, Canada
| | - Jillian A Macklin
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Ted Rogers Centre for Heart Research, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Sara Nejat
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
| | - Marwan G Althagafi
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Jinmiao Chen
- Singapore Immunology Network(SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Crystal Kantores
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
| | - Siyavash Hosseinzadeh
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Laura Aronoff
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Rysa Zaman
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Iulia Barbu
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Rickvinder Besla
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kory J Lavine
- Division of Cardiology, Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Division of Cardiology, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Florent Ginhoux
- Singapore Immunology Network(SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Mansoor Husain
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Ted Rogers Centre for Heart Research, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto, Canada
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto, Canada
| | - Clinton S Robbins
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, Canada.
- Ted Rogers Centre for Heart Research, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Immunology, University of Toronto, Toronto, Canada.
- Peter Munk Cardiac Centre, Toronto, Canada.
| |
Collapse
|
46
|
Li H, Zhou Y, Song W, Li J, Xu J. Expression of ADAMTS-1 mRNA in myocardium of viral heart disease mice and its clinical significance. Exp Ther Med 2018; 17:153-158. [PMID: 30651776 PMCID: PMC6307383 DOI: 10.3892/etm.2018.6894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/05/2018] [Indexed: 12/21/2022] Open
Abstract
The expression of ADAMTS-1 mRNA in myocardium of viral heart disease (VHD) mice was investigated to explore its role in myocardial fibrosis. A total of 150 purebred inbred Balb/c mice were used in this study. According to the principle of similar body weight, 50 mice were selected to make an acute viral myocarditis (VMC) animal model (acute VMC group), and 50 mice were selected to make a chronic VMC animal model (chronic VMC group), and the remaining 50 mice were selected as a control group. RT-qPCR was used to detect the relative expression of transforming growth factor-β1 (TGF-β1) mRNA and ADAMTS-1 mRNA in myocardial tissue of three groups of mice, and their relationship in myocardial fibrosis was analyzed. Compared with the control group, the collagen volume fraction (CVF) in the myocardial tissue of the acute VMC group was significantly increased, and the increase of CVF in the myocardial tissue of the chronic VMC group was the most significant (p<0.001). Compared with the control group, the relative expression of TGF-β1 mRNA and ADAMTS-1 mRNA in myocardial tissue of the mice in the acute and chronic VMC group were significantly increased (p<0.001). The relative expression of TGF-β1 mRNA and ADAMTS-1 mRNA in myocardial tissue of chronic VMC group was significantly higher than that of acute VMC group (p<0.001). Pearson's correlation test results showed that ADAMTS-1 mRNA was positively correlated with CVF and TGF-β1 mRNA, and the correlation coefficients were (r=0.351, p<0.01, r=0.401, p<0.01). ADAMTS-1 is involved in the occurrence and development of myocardial fibrosis, and it is positively correlated with CVF and TGF-β1. It may play a role in promoting myocardial fibrosis during the development of VHD. It can be used as a biological index for predicting myocardial fibrosis.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Yanchun Zhou
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Wei Song
- Department of Psychology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Jianxiu Li
- Department of Cardiovascular Medicine, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Jingtao Xu
- Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
47
|
Aronoff L, Epelman S, Clemente-Casares X. Isolation and Identification of Extravascular Immune Cells of the Heart. J Vis Exp 2018. [PMID: 30199044 DOI: 10.3791/58114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The immune system is an essential component of a healthy heart. The myocardium is home to a rich population of different immune cell subsets with functional compartmentalization both during steady state and during different forms of inflammation. Until recently, the study of immune cells in the heart required the use of microscopy or poorly developed digestion protocols, which provided enough sensitivity during severe inflammation but were unable to confidently identify small - but key - populations of cells during steady state. Here, we discuss a simple method combining enzymatic (collagenase, hyaluronidase and DNAse) and mechanical digestion of murine hearts preceded by intravascular administration of fluorescently-labelled antibodies to differentiate small but unavoidable intravascular cell contaminants. This method generates a suspension of isolated viable cells that can be analyzed by flow cytometry for identification, phenotyping and quantification, or further purified with fluorescence-activated cell sorting or magnetic bead separation for transcriptional analysis or in vitro studies. We include an example of a step-by-step flow cytometric analysis to differentiate the key macrophage and dendritic cell populations of the heart. For a medium sized experiment (10 hearts) the completion of the procedure requires 2-3 h.
Collapse
Affiliation(s)
- Laura Aronoff
- Toronto General Hospital Research Institute, University Health Network (UHN); Dept of Laboratory Medicine and Pathobiology, University of Toronto
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network (UHN); Dept of Laboratory Medicine and Pathobiology, University of Toronto; Dept of Immunology, University of Toronto; Peter Munk Cardiac Centre; Ted Rogers Centre for Heart Research;
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network (UHN); Dept of Laboratory Medicine and Pathobiology, University of Toronto;
| |
Collapse
|
48
|
Vereecke L, Elewaut D. Tumour necrosis factor: out of my heart! Ann Rheum Dis 2018; 77:793-794. [PMID: 29615412 DOI: 10.1136/annrheumdis-2018-213118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 11/03/2022]
Affiliation(s)
- Lars Vereecke
- Department of Rheumatology, University Hospital Gent, Gent, Belgium
- VIB Inflammation Research Center, Unit for Molecular Immunology and Inflammation, Ghent University, Ghent, Belgium
| | - Dirk Elewaut
- Department of Rheumatology, University Hospital Gent, Gent, Belgium
- VIB Inflammation Research Center, Unit for Molecular Immunology and Inflammation, Ghent University, Ghent, Belgium
| |
Collapse
|
49
|
Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, Lelios I, Heppner FL, Kipnis J, Merkler D, Greter M, Becher B. High-Dimensional Single-Cell Mapping of Central Nervous System Immune Cells Reveals Distinct Myeloid Subsets in Health, Aging, and Disease. Immunity 2018; 48:380-395.e6. [DOI: 10.1016/j.immuni.2018.01.011] [Citation(s) in RCA: 595] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/05/2017] [Accepted: 01/14/2018] [Indexed: 12/14/2022]
|