Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Cardioimmunology: the immune system in cardiac homeostasis and disease

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.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Cardiac anatomy with respect to immune cells.
Fig. 2: Participation of macrophages in normal electrical conduction.
Fig. 3: Immune cells in coronary heart disease.
Fig. 4: Potential immune mechanisms in viral myocarditis.

Similar content being viewed by others

References

  1. Ramos, G. C. et al. Myocardial aging as a T cell-mediated phenomenon. Proc. Natl Acad. Sci. USA 114, E2420–E2429 (2017).This manuscript provides a cardiac immune cell atlas during ageing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Geissmann, F. et al. Development of monocytes, macrophages, and dendritic cells. Science 327, 656–661 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38, 792–804 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Hoeffel, G. et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42, 665–678 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. van Furth, R. & Cohn, Z. A. The origin and kinetics of mononuclear phagocytes. J. Exp. Med. 128, 415–435 (1968).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Nag, A. C. Study of non-muscle cells of the adult mammalian heart: a fine structural analysis and distribution. Cytobios 28, 41–61 (1980).

    CAS  PubMed  Google Scholar 

  10. Pinto, A. R. et al. An abundant tissue macrophage population in the adult murine heart with a distinct alternatively-activated macrophage profile. PLOS ONE 7, e36814 (2012).This study uses a macrophage-specific promoter to focus on resident macrophages in the normal mouse heart.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Epelman, S. et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 40, 91–104 (2014).This work introduces cardiac macrophage subsets in the mouse and describes their origins.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Epelman, S., Lavine, K. J. & Randolph, G. J. Origin and functions of tissue macrophages. Immunity 41, 21–35 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Epelman, S., Liu, P. P. & Mann, D. L. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat. Rev. Immunol. 15, 117–129 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Heidt, T. et al. Differential contribution of monocytes to heart macrophages in steady-state and after myocardial infarction. Circ. Res. 115, 284–295 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ensan, S. et al. Self-renewing resident arterial macrophages arise from embryonic CX3CR1(+) precursors and circulating monocytes immediately after birth. Nat. Immunol. 17, 159–168 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Molawi, K. et al. Progressive replacement of embryo-derived cardiac macrophages with age. J. Exp. Med. 211, 2151–2158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bajpai, G. et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat. Med. 24, 1234–1245 (2018).This work confirms that most human cardiac macrophages, as in the mouse, do not derive from circulating monocytes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Choi, J. H. et al. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J. Exp. Med. 206, 497–505 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Frangogiannis, N. G. et al. Resident cardiac mast cells degranulate and release preformed TNF-alpha, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation 98, 699–710 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Ito, B. R., Engler, R. L. & del Balzo, U. Role of cardiac mast cells in complement C5a-induced myocardial ischemia. Am. J. Physiol. 264, H1346–H1354 (1993).

    CAS  PubMed  Google Scholar 

  21. Ngkelo, A. et al. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J. Exp. Med. 213, 1353–1374 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Patella, V. et al. Human heart mast cells. Isolation, purification, ultrastructure, and immunologic characterization. J. Immunol. 154, 2855–2865 (1995).

    CAS  PubMed  Google Scholar 

  23. Leid, J. et al. Primitive embryonic macrophages are required for coronary development and maturation. Circ. Res. 118, 1498–1511 (2016).This study suggests that macrophages are involved in cardiac development.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hulsmans, M. et al. Macrophages facilitate electrical conduction in the heart. Cell 169, 510–522 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Butts, B. et al. Increased inflammation in pericardial fluid persists 48 hours after cardiac surgery. Circulation 136, 2284–2286 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Horckmans, M. et al. Pericardial adipose tissue regulates granulopoiesis, fibrosis, and cardiac function after myocardial infarction. Circulation 137, 948–960 (2018).

    Article  PubMed  Google Scholar 

  27. Wang, J. & Kubes, P. A reservoir of mature cavity macrophages that can rapidly invade visceral organs to affect tissue repair. Cell 165, 668–678 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Weber, G. F. et al. Pleural innate response activator B cells protect against pneumonia via a GM-CSF-IgM axis. J. Exp. Med. 211, 1243–1256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gwechenberger, M. et al. Cardiac myocytes produce interleukin-6 in culture and in viable border zone of reperfused infarctions. Circulation 99, 546–551 (1999).

    Article  CAS  PubMed  Google Scholar 

  30. Wu, L. et al. Cardiac fibroblasts mediate IL-17A-driven inflammatory dilated cardiomyopathy. J. Exp. Med. 211, 1449–1464 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Anzai, A. et al. The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes. J. Exp. Med. 214, 3293–3310 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hilgendorf, I. et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ. Res. 114, 1611–1622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nahrendorf, M. et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J. Exp. Med. 204, 3037–3047 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bach, L. A. Endothelial cells and the IGF system. J. Mol. Endocrinol. 54, R1–R13 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Li, Q. & Barres, B. A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18, 225–242 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111–121 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Palmer, D. B. The effect of age on thymic function. Front. Immunol. 4, 316 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Rea, I. M. et al. Age and age-related diseases: role of inflammation triggers and cytokines. Front. Immunol. 9, 586 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Sano, S. et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1beta/NLRP3 inflammasome. J. Am. Coll. Cardiol. 71, 875–886 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Swirski, F. K. Inflammation and CVD in 2017: from clonal haematopoiesis to the CANTOS trial. Nat. Rev. Cardiol. 15, 79–80 (2018).

    Article  PubMed  Google Scholar 

  41. Chen, W. & Frangogiannis, N. G. The role of inflammatory and fibrogenic pathways in heart failure associated with aging. Heart Fail Rev. 15, 415–422 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Loffredo, F. S. et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 153, 828–839 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Smith, S. C. et al. GDF11 does not rescue aging-related pathological hypertrophy. Circ. Res. 117, 926–932 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Frangogiannis, N. G. Chemokines in the ischemic myocardium: from inflammation to fibrosis. Inflamm Res. 53, 585–595 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Frangogiannis, N. G. et al. Critical role of monocyte chemoattractant protein-1/CC chemokine ligand 2 in the pathogenesis of ischemic cardiomyopathy. Circulation 115, 584–592 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Swirski, F. K. & Nahrendorf, M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science 339, 161–166 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Swirski, F. K. et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 325, 612–616 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dewald, O. et al. CCL2/monocyte chemoattractant protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ. Res. 96, 881–889 (2005).This work describes the importance of the chemokine CCL2 in the response to myocardial infarction.

    Article  CAS  PubMed  Google Scholar 

  49. Shi, C. et al. Bone marrow mesenchymal stem and progenitor cells induce monocyte emigration in response to circulating toll-like receptor ligands. Immunity 34, 590–601 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zouggari, Y. et al. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat. Med. 19, 1273–1280 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Berg, K. E. et al. Elevated CD14++CD16- monocytes predict cardiovascular events. Circ. Cardiovasc. Genet. 5, 122–131 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Coller, B. S. Leukocytosis and ischemic vascular disease morbidity and mortality: is it time to intervene? Arterioscler Thromb. Vasc. Biol. 25, 658–670 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Rogacev, K. S. et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J. Am. Coll. Cardiol. 60, 1512–1520 (2012).

    Article  CAS  PubMed  Google Scholar 

  54. Wan, E. et al. Enhanced efferocytosis of apoptotic cardiomyocytes through myeloid-epithelial-reproductive tyrosine kinase links acute inflammation resolution to cardiac repair after infarction. Circ. Res. 113, 1004–1012 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. King, K. R. et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Frangogiannis, N. G., Smith, C. W. & Entman, M. L. The inflammatory response in myocardial infarction. Cardiovasc. Res. 53, 31–47 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Horckmans, M. et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur. Heart J. 38, 187–197 (2017).

    CAS  PubMed  Google Scholar 

  58. Leuschner, F. et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J. Exp. Med. 209, 123–137 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Krishnamurthy, P. et al. IL-10 inhibits inflammation and attenuates left ventricular remodeling after myocardial infarction via activation of STAT3 and suppression of HuR. Circ. Res. 104, e9–e18 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Hofmann, U. et al. Activation of CD4+ T lymphocytes improves wound healing and survival after experimental myocardial infarction in mice. Circulation 125, 1652–1663 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Saxena, A. et al. Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function. Am. J. Physiol. Heart Circ. Physiol. 307, H1233–H1242 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Weirather, J. et al. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ. Res. 115, 55–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Korf-Klingebiel, M. et al. Myeloid-derived growth factor (C19orf10) mediates cardiac repair following myocardial infarction. Nat. Med. 21, 140–149 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Panizzi, P. et al. Impaired infarct healing in atherosclerotic mice with Ly-6Chi monocytosis. J. Am. Coll. Cardiol. 55, 1629–1638 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  65. van Amerongen, M. J., Harmsen, M. C., van Rooijen, N., Petersen, A. H. & van Luyn, M. J. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am. J. Pathol. 170, 818–829 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Tsujioka, H. et al. Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction. J. Am. Coll. Cardiol. 54, 130–138 (2009).

    Article  PubMed  Google Scholar 

  67. van der Laan, A. M. et al. Monocyte subset accumulation in the human heart following acute myocardial infarction and the role of the spleen as monocyte reservoir. Eur. Heart J. 35, 376–385 (2014).

    Article  PubMed  CAS  Google Scholar 

  68. Lee, W. W. et al. PET/MRI of inflammation in myocardial infarction. J. Am. Coll. Cardiol. 59, 153–163 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ismahil, M. A. et al. Remodeling of the mononuclear phagocyte network underlies chronic inflammation and disease progression in heart failure: critical importance of the cardiosplenic axis. Circ. Res. 114, 266–282 (2014).This manuscript elucidates the role of the spleen in the progression of heart failure in mice.

    Article  CAS  PubMed  Google Scholar 

  70. Sager, H. B. et al. Proliferation and recruitment contribute to myocardial macrophage expansion in chronic heart failure. Circ. Res. 119, 853–864 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Bansal, S. S. et al. Activated T lymphocytes are essential drivers of pathological remodeling in ischemic heart failure. Circ. Heart Fail. 10, e003688 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Engstrom, G., Melander, O. & Hedblad, B. Leukocyte count and incidence of hospitalizations due to heart failure. Circ. Heart Fail. 2, 217–222 (2009).

    Article  PubMed  Google Scholar 

  73. Moslehi, J. J., Salem, J. E., Sosman, J. A., Lebrun-Vignes, B. & Johnson, D. B. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 391, 933 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Braunwald, E. Cardiomyopathies: an overview. Circ. Res. 121, 711–721 (2017).

    Article  CAS  PubMed  Google Scholar 

  75. Cooper, L. T. J. Myocarditis. N. Engl. J. Med. 360, 1526–1538 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Cihakova, D. & Rose, N. R. Pathogenesis of myocarditis and dilated cardiomyopathy. Adv. Immunol. 99, 95–114 (2008).

    Article  CAS  PubMed  Google Scholar 

  77. Coyne, C. B. & Bergelson, J. M. Virus-induced Abl and Fyn kinase signals permit coxsackievirus entry through epithelial tight junctions. Cell 124, 119–131 (2006).

    Article  CAS  PubMed  Google Scholar 

  78. Fairweather, D. et al. Mast cells and innate cytokines are associated with susceptibility to autoimmune heart disease following coxsackievirus B3 infection. Autoimmunity 37, 131–145 (2004).

    Article  CAS  PubMed  Google Scholar 

  79. Fairweather, D. et al. IL-12 protects against coxsackievirus B3-induced myocarditis by increasing IFN-gamma and macrophage and neutrophil populations in the heart. J. Immunol. 174, 261–269 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Fuse, K. et al. Myeloid differentiation factor-88 plays a crucial role in the pathogenesis of Coxsackievirus B3-induced myocarditis and influences type I interferon production. Circulation 112, 2276–2285 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Holm, G. H. et al. Interferon regulatory factor 3 attenuates reovirus myocarditis and contributes to viral clearance. J. Virol. 84, 6900–6908 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Riad, A. et al. Myeloid differentiation factor-88 contributes to TLR9-mediated modulation of acute coxsackievirus B3-induced myocarditis in vivo. Am. J. Physiol. Heart Circ. Physiol. 298, H2024–H2031 (2010).

    Article  CAS  PubMed  Google Scholar 

  83. Valaperti, A. et al. Innate immune interleukin-1 receptor-associated kinase 4 exacerbates viral myocarditis by reducing CCR5+ CD11b+ monocyte migration and impairing interferon production. Circulation 128, 1542–1554 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Clemente-Casares, X. et al. A CD103+ conventional dendritic cell surveillance system prevents development of overt heart failure during subclinical viral myocarditis. Immunity 47, 974–989 (2017).

    Article  CAS  PubMed  Google Scholar 

  85. Liu, P. et al. The tyrosine kinase p56lck is essential in coxsackievirus B3-mediated heart disease. Nat. Med. 6, 429–434 (2000).

    Article  CAS  PubMed  Google Scholar 

  86. Baldeviano, G. C. et al. Interleukin-17A is dispensable for myocarditis but essential for the progression to dilated cardiomyopathy. Circ. Res. 106, 1646–1655 (2010).

    Article  CAS  PubMed  Google Scholar 

  87. Nindl, V. et al. Cooperation of Th1 and Th17 cells determines transition from autoimmune myocarditis to dilated cardiomyopathy. Eur. J. Immunol. 42, 2311–2321 (2012).

    Article  CAS  PubMed  Google Scholar 

  88. Shi, Y. et al. Regulatory T cells protect mice against coxsackievirus-induced myocarditis through the transforming growth factor beta-coxsackie-adenovirus receptor pathway. Circulation 121, 2624–2634 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. Frisancho-Kiss, S. et al. Cutting edge: cross-regulation by TLR4 and T cell Ig mucin-3 determines sex differences in inflammatory heart disease. J. Immunol. 178, 6710–6714 (2007).

    Article  CAS  PubMed  Google Scholar 

  90. Li, Y., Heuser, J. S., Cunningham, L. C., Kosanke, S. D. & Cunningham, M. W. Mimicry and antibody-mediated cell signaling in autoimmune myocarditis. J. Immunol. 177, 8234–8240 (2006).

    Article  CAS  PubMed  Google Scholar 

  91. Eriksson, U. et al. Dendritic cell-induced autoimmune heart failure requires cooperation between adaptive and innate immunity. Nat. Med. 9, 1484–1490 (2003).

    Article  CAS  PubMed  Google Scholar 

  92. Kaya, Z., Leib, C. & Katus, H. A. Autoantibodies in heart failure and cardiac dysfunction. Circ. Res. 110, 145–158 (2012).

    Article  CAS  PubMed  Google Scholar 

  93. Diny, N. L. et al. Eosinophil-derived IL-4 drives progression of myocarditis to inflammatory dilated cardiomyopathy. J. Exp. Med. 214, 943–957 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Paulus, W. J. et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the heart failure and echocardiography associations of the european society of cardiology. Eur. Heart J. 28, 2539–2550 (2007).

    Article  PubMed  Google Scholar 

  95. Sharma, K. & Kass, D. A. Heart failure with preserved ejection fraction: mechanisms, clinical features, and therapies. Circ. Res. 115, 79–96 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Laroumanie, F. et al. CD4+ T cells promote the transition from hypertrophy to heart failure during chronic pressure overload. Circulation 129, 2111–2124 (2014).

    Article  CAS  PubMed  Google Scholar 

  97. Nevers, T. et al. Left ventricular T cell recruitment contributes to the pathogenesis of heart failure. Circ. Heart Fail. 8, 776–787 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Nevers, T. et al. Th1 effector T cells selectively orchestrate cardiac fibrosis in nonischemic heart failure. J. Exp. Med. 214, 3311–3329 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Glezeva, N. et al. Exaggerated inflammation and monocytosis associate with diastolic dysfunction in heart failure with preserved ejection fraction: evidence of M2 macrophage activation in disease pathogenesis. J. Card Fail. 21, 167–177 (2015).

    Article  CAS  PubMed  Google Scholar 

  100. Hulsmans, M. et al. Cardiac macrophages promote diastolic dysfunction. J. Exp. Med. 215, 423–440 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Pinto, A. R., Godwin, J. W. & Rosenthal, N. A. Macrophages in cardiac homeostasis, injury responses and progenitor cell mobilisation. Stem Cell Res. 13, 705–714 (2014).

    Article  CAS  PubMed  Google Scholar 

  102. Holland, T. L. et al. Infective endocarditis. Nat. Rev. Dis. Primers. 2, 16059 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Thammavongsa, V., Kim, H. K., Missiakas, D. & Schneewind, O. Staphylococcal manipulation of host immune responses. Nat. Rev. Microbiol. 13, 529–543 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Feuerstein, R., Seidl, M., Prinz, M. & Henneke, P. MyD88 in macrophages is critical for abscess resolution in staphylococcal skin infection. J. Immunol. 194, 2735–2745 (2015).

    Article  CAS  PubMed  Google Scholar 

  105. Veltrop, M. H., Bancsi, M. J., Bertina, R. M. & Thompson, J. Role of monocytes in experimental Staphylococcus aureus endocarditis. Infect. Immun. 68, 4818–4821 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Friedrich, R. et al. Staphylocoagulase is a prototype for the mechanism of cofactor-induced zymogen activation. Nature 425, 535–539 (2003).

    Article  CAS  PubMed  Google Scholar 

  107. Shahreyar, M. et al. Severe sepsis and cardiac arrhythmias. Ann. Transl Med. 6, 6 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Moss, T. J. et al. New-onset atrial fibrillation in the critically ill. Crit. Care Med. 45, 790–797 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Lazzerini, P. E., Capecchi, P. L. & Laghi-Pasini, F. Systemic inflammation and arrhythmic risk: lessons from rheumatoid arthritis. Eur. Heart J. 38, 1717–1727 (2017).

    CAS  PubMed  Google Scholar 

  110. Abdelhadi, R. H., Gurm, H. S., Van Wagoner, D. R. & Chung, M. K. Relation of an exaggerated rise in white blood cells after coronary bypass or cardiac valve surgery to development of atrial fibrillation postoperatively. Am. J. Cardiol. 93, 1176–1178 (2004).

    Article  PubMed  Google Scholar 

  111. Guo, Y., Lip, G. Y. & Apostolakis, S. Inflammation in atrial fibrillation. J. Am. Coll. Cardiol. 60, 2263–2270 (2012).

    Article  CAS  PubMed  Google Scholar 

  112. Chen, M. C. et al. Increased inflammatory cell infiltration in the atrial myocardium of patients with atrial fibrillation. Am. J. Cardiol. 102, 861–865 (2008).

    Article  PubMed  Google Scholar 

  113. Smorodinova, N. et al. Analysis of immune cell populations in atrial myocardium of patients with atrial fibrillation or sinus rhythm. PLOS ONE 12, e0172691 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Yue, L., Xie, J. & Nattel, S. Molecular determinants of cardiac fibroblast electrical function and therapeutic implications for atrial fibrillation. Cardiovasc. Res. 89, 744–753 (2011).

    Article  CAS  PubMed  Google Scholar 

  115. Sun, Z. et al. Cross-talk between macrophages and atrial myocytes in atrial fibrillation. Bas. Res. Cardiol. 111, 63 (2016).

    Article  CAS  Google Scholar 

  116. Riley, G., Syeda, F., Kirchhof, P. & Fabritz, L. An introduction to murine models of atrial fibrillation. Front. Physiol. 3, 296 (2012).

    PubMed  PubMed Central  Google Scholar 

  117. De Jesus, N. M. et al. Atherosclerosis exacerbates arrhythmia following myocardial infarction: role of myocardial inflammation. Heart Rhythm. 12, 169–178 (2015).

    Article  PubMed  Google Scholar 

  118. De Jesus, N. M. et al. Antiarrhythmic effects of interleukin 1 inhibition after myocardial infarction. Heart Rhythm. 14, 727–736 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Monnerat, G. et al. Macrophage-dependent IL-1beta production induces cardiac arrhythmias in diabetic mice. Nat. Commun. 7, 13344 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mann, D. L. Inflammatory mediators and the failing heart: past, present, and the foreseeable future. Circ. Res. 91, 988–998 (2002).

    Article  CAS  PubMed  Google Scholar 

  121. Meier, L. A. et al. CD301b/MGL2+ mononuclear phagocytes orchestrate autoimmune cardiac valve inflammation and fibrosis. Circulation 137, 2478–2493 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ridker, P. M. et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

  123. Johnson, D. B. et al. Fulminant myocarditis with combination immune checkpoint blockade. N. Engl. J. Med. 375, 1749–1755 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Okazaki, T. et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat. Med. 9, 1477–1483 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Rudolph, V. et al. Myeloperoxidase acts as a profibrotic mediator of atrial fibrillation. Nat. Med. 16, 470–474 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Stanek, G., Wormser, G. P., Gray, J. & Strle, F. Lyme borreliosis. Lancet 379, 461–473 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge K. Joyes for editing the manuscript. This work was funded in part by federal funds from the US National Heart, Lung, and Blood Institute (NHLBI) (HL135752 and HL139598) and the Massachusetts General Hospital Research Scholar Program.

Reviewer information

Nature Reviews Immunology thanks D. Ciháková, S. Epelman and J.-S. Silvestre for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Both authors researched data for the article, discussed its content and wrote, reviewed and edited the article.

Corresponding authors

Correspondence to Filip K. Swirski or Matthias Nahrendorf.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Cardiomyocytes

Large muscle cells specific to the heart.

Definitive haematopoiesis

The production of mature blood cells from haematopoietic progenitor cells in the bone marrow that occurs throughout life.

Atrioventricular node

The node that controls the ventricular heart rate and is a major relay station for electrical conduction from atria to ventricles in the heart.

Pericardium

The membrane layer enclosing the heart.

Serosal fluid

Fluid in body cavities such as the pericardium.

Primitive coronary plexus

A developmental stage of the coronary vasculature.

Gap junctions

Intercellular connections formed by connexin proteins that directly connect the cytoplasm of two cells and enable exchange of ions, which electrically couples cardiomyocytes to each other and to macrophages.

Source–sink relationship

An interaction that involves the exchange of charges between excitable cardiomyocytes and passively depolarized macrophages.

Optical tissue clearance

A method by which biological specimens are treated to enable whole organ microscopy.

Sinus node

The pacemaker of the heart, consisting of a cluster of spontaneously depolarizing cells in which electrical impulses are generated.

CD11b DTR mice

Transgenic mice expressing the diphtheria toxin receptor under control of the Cd11b (also known as Itgam) gene promoter, which enables macrophage depletion after injection of non-toxic doses of diphtheria toxin.

Granulation tissue

Newly forming tissue during wound healing that contains leukocytes, stromal cells, extracellular matrix and blood vessels.

Non-reperfused myocardial infarction

An event that occurs when a coronary artery is closed off and not re-opened for blood flow, leading to the ischaemic death of a large number of cardiac cells.

Remote myocardium

The part of the heart that is not directly affected by ischaemia during myocardial infarction.

Ejection fraction

The volume of blood ejected with each heartbeat, which is normalized to the size of the left ventricular cavity.

Sympathetic tone

Innervation by a branch of the autonomous nervous system that increases the output of the heart but also has many other functions, including in the haematopoietic and immune systems.

Dilated cardiomyopathy

Enlargement of the left ventricle leading to heart failure.

Re-entry arrhythmias

Conduction disorders in which the depolarizing wave front circles, often leading to rapid and uncoordinated cardiomyocyte contractions.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Swirski, F.K., Nahrendorf, M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol 18, 733–744 (2018). https://doi.org/10.1038/s41577-018-0065-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-018-0065-8

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing