Short abstract
Following cardiac injury, IL‐33 produced by fibroblast promoted ILCs and macrophages showing the type II immune phenotype to ameliorate inflammation; in pericardial fluid, IL‐5 and IL‐13 secreted by activated ILC2s led to cardiac fibroblasts to produce a large number of eotaxin; furthermore, the production of IL‐1β by cardiac macrophages may promote ILCs differentiation into ILC3s.
Keywords: cardiovascular disease, cardiovascular system, innate lymphoid cells
Summary
As a distinctive population of leucocytes, innate lymphoid cells (ILCs) participate in immune‐mediated diseases and play crucial roles in tissue remodelling after injury. ILC lineages can be divided into helper ILCs and cytotoxic ILCs. Most helper ILCs are integrated into the fabric of tissues and produce different types of cytokines involving in the pathogenesis of many kinds of cardiovascular disease and form intricate response circuits with adaptive immune cells. However, the specific phenotype and function of helper ILC subsets in cardiovascular diseases are still poorly understood. In this review, we firstly highlight the distribution of helper ILCs in cardiovascular system and further discuss the potential contribution of helper ILCs in inflammation‐associated cardiovascular disease.
Abbreviations
- AT
adipose tissue
- CVD
cardiovascular diseases
- EGPA
eosinophilic granulomatosis with polyangiitis
- ILCs
innate lymphoid cells
- LTi
lymphoid tissue‐induced cells
- MI
myocardial infarction
INTRODUCTION
Cardiovascular diseases (CVD) are the leading cause of global mortality and incidence rate in non‐communicable diseases. Many factors can lead to the occurrence of CVD, although its pathogenesis remains ulcear. 1 The role of inflammation in CVD has been widely accepted in the initial and progression. B and T cells are involved in promoting the production of antibodies and inflammatory factors, such as IFN‐γ, TNF‐α, and IL‐17A, leading to sustain damage to the cardiovascular system. 2 IFN‐γ and TNF‐α have been reported as proatherogenic cytokines in vascular diseases, and IL‐17A aggravates cardiac damage, promoting fibrosis. 3 , 4 Therefore, whether in clinical or experimental data, the modulation of adaptive immune cells has already been thoroughly studied, and it is of great help for the prevention and treatment of CVD. However, it is worth noting that more and more researchers have discovered that the innate immune plays an important role in the pathogenesis of CVD. Previous studies have found that the primary innate immune cells within cardiac tissue are mainly macrophages, including tissue‐resident macrophages and monocyte‐derived macrophages, contributing to cardiac tissue injury and repair. 5 Other innate immune cells such as mast cells and dendritic cells also found sparsely distributed in cardiac tissue. 6 , 7 Beyond innate immune cells given here, innate lymphoid cells (ILCs) are newly discovered innate immune cell subset in recent years; however, their specific role in CVD is still unclear.
ILCs mainly include cytotoxic ILCs and helper ILCs. ILCs mostly located on the barrier surface of the body, respond to environmental stress signals and participate in the protection of pathogens, tissue remodelling and maintenance of internal balance and promote the formation of secondary lymphoid organs with the interaction of mesenchymal cells during embryonic development. 8 , 9 , 10 , 11 This review will discuss the distribution of ILCs in the cardiovascular system, the regulation of its response and its roles in physiological and pathophysiological processes.
INNATE LYMPHOID CELLS
According to whether there is the inhibitor of DNA binding 2 (ID2) expression, innate lymphoid progenitor, the progeny of common lymphoid progenitor which could differentiate into almost all types of lymphocytes, mainly divided into two ILC lineages: helper ILCs and cytotoxic ILCs. 12 , 13 , 14 Moreover, common helper ILC progenitor whether with the existence of PLZF could give rise to ILC precursors, and lymphoid tissue inducer progenitors, which differentiate into lymphoid tissue‐induced cells (LTi). 13 , 15 NKs are cytotoxic ILCs, which are analogous to CD8+ T cells. NKs, circulating in the bloodstream, with the ability to secrete pro‐inflammatory cytokines like TNF‐α and IFN‐γ, are the first line of eliminating virus‐infected cells through two major pathway, including target cell death pathway and death receptor pathways (FasL and TRAIL) induced by the production of perforin and granzyme. 16 , 17 , 18 , 19 Helper ILCs are categorized into several subsets, which resemble in CD4+ T cells subsets (Th1, Th2, Th17), including LTi, ILC1, ILC2 and ILC3, which is based on the ranks of categorization of signature cytokines, transcription factors and so on. 20 Like adaptive lymphocytes, ILCs respond specifically to different immune responses through secreting distinct cytokines. The development of ILCs relies on the IL‐2Rγc and IL‐7Rα, whereas lack of recombinant activating gene and as a result, they do not exhibit antigen specificity. 8 , 21
ILC1s are characterized by their ability to produce IFN‐γ and TNF‐α, which depend on T‐bet and not on eomesodermin (Eomes). 22 Similar to NKs, ILC1s that respond to IL‐12, IL‐15 and IL‐18 can participate in the first line of defence against viral and certain parasite infections such as T. gondii 23 or C. difficile 24 . ILC1s were originally distinguished from NKs because of their lower cytotoxic activity, whereas they are also capable of recognizing and killing target cells, although less efficiently. 25 , 26 ILC1s are heterogeneous and have different phenotypic markers, which express CD127 and CD200R in humans, 27 and express NKp46 in both mice and humans, depending on the microenvironment of the tissue. ILC2s are tissue‐resident and express GATA3. 28 , 29 , 30 , 31 ILC2s can produce Th2‐associated cytokines, such as IL‐4, IL‐5, IL‐9, IL‐13, the epidermal growth factor amphiregulin with the responses to IL‐25, IL‐33 and thymic stromal lymphopoietin. 32 , 33 , 34 ILC3s depend on RORγt, encoded by RORC related to retinoic acid receptor for their development and function, which is characterized by the production of IL‐17, IL‐22 and GM‐CSF. 35 ILC3s have at least two subsets and every subset is quite different in transcription, development and function, and locate in different tissue microenvironments. ILC3 subsets are distinguished according to the expression of natural cytotoxic receptor NKp46 (also known as NCR1 in mice or NCR2 in humans). 36 , 37 IL‐22 predominantly produced by NCR+ ILC3, whereas IL‐17 mainly derived from NCR− ILC3. 38 Additionally, Th17 cells can also be regulated by ILC3s. 39 LTi cells are a population identified earlier than other ILC subsets. 40 Although LTi cells can secrete IL‐17, IL‐22 and lymphotoxin with expressing RORγt, they are different from ILC3s in the development of precursor cells. 41 , 42 Lymph nodes and Peyer's patches require LTi to express surface lymphotoxin α1β2, which can bind to its receptor LTβR on stromal cells during the fetal stage to promote their formation and development, 43 , 44 , 45 whereas adult LTi‐like cells protect the gastrointestinal tract from pathogens by producing IL‐17 and IL‐22, 46 , 47 , 48 and can also express OX40L and CD30L, which both are T‐cell survival molecules, indicating that they may affect T‐cell function. 49 , 50
Recently, ILCs have already been studied in various tissues and organs, especially in mucosal tissues. 51 , 52 They can effectively participate in the regulation of tissue microenvironment in both homeostasis and inflammation conditions. Although the helper ILCs have been extensively reported in barrier organs, there are few studies on their roles in cardiovascular system. 53 NKs, as a protector, prevent cardiac inflammation and injury in most CVDs; however, more and more studies suggested that they promote inflammation in atherogenesis. 54 , 55 , 56 The characteristics of NKs in CVD have been well reviewed elsewhere 54 , 57 and therefore not be discussed here.
ILCs IN CARDIOVASCULAR SYSTEM
The heart is not homogeneous with the unique histological characteristics so that the distribution of immune cells in the heart is not uniform, which is quite different from the tissue previously studied by ILCs. In healthy adult mice, it contains almost all major white blood cell types, including monocyte phagocytic cells, neutrophils, T and B cells. 58 Recent data have shown that high proportion of cell population characterized by lineagenegCD45+CD127+RORγt−T‐bet−CRTH2− and lineagenegCD45+CD90+RORγt−T‐bet−CRTH2− are present in the heart of human and naïve mouse, respectively 59 (lineage‐negative cell is the rest of cell in which mature cell lineage markers were not expressed, including T, B cells, monocytes/macrophages, granulocytes, NK cells and erythrocytes markers). These cell populations do not have specific markers of ILC subsets, so they are considered as undifferentiated ILCs in the normal heart. 59 , 60 The heart lacks typical epithelial cells, mainly muscle tissue. The surface of the heart is covered with a membranous structure called the pericardium, which is mainly composed of the epicardium, pericardial cavity, parietal layer of serous and fibrous layer. 61 Under steady state, serous cavities with the protective serosal fluid harbour large numbers of immune cells, such as macrophages, B cells and so on, and as a result, it may be one of a source of tissue‐infiltrating leucocytes during pathological conditions. 62 , 63 Pericardial adipose tissue (AT) is an abundant source of leucocytes, such as B, T, dendritic cells and myeloid cells. 64 ILCs may also exist in AT under normal conditions. It has been reported that ILC2s reside in the visceral AT‐secreting IL‐5 and IL‐13 following IL‐33 stimulation, and promote the accumulation of alternatively activated macrophages and eosinophils. Besides IL‐4 secreted by eosinophils co‐operate with ILC2s‐derived IL‐13 to activate IL‐4 receptor signalling in PDGFRα+ adipocyte precursors to control beige fat biogenesis. 65 , 66 , 67 Moreover, ILC1s, which indicated as a stable population in AT to produce IFN‐γ, can suppress ILC2s activation, thereby restraining the accumulation of Treg cells. 68 ILC1s also contribute to promote obesity‐associated insulin resistance and polarize macrophages towards M1 macrophages by expressing IFN‐γ. 69 , 70 There are initial indications that ILCs with progenitor‐like features have cardiac specific feature, 59 which is worth further investigation.
The intima of the artery is the layer between the arterial endothelium and the first elastic layer of the artery, such that the intima is on top of the medial layer that is rich in smooth muscle cells and outer layers of arteries, known as the adventitia. Anatomically and histologically, the intimal layer is the main area of chronic inflammatory development, which is the characteristic of atherosclerosis, and eventually caused the corresponding tissue of the vascular supply to ischaemia or necrosis. 71 The pathological lesion in the arterial intima continuously accumulates and increases the size of the intima under the influence of inflammatory factors, forming an inflammatory structure called plaque where contains a lot of inflammatory cells. In human plaques, T cells account for about 10%, of which 70% are regarded as CD4+ T cells, and the others largely CD8+ T cells. 72 Most CD4+ T cells in the vessel wall are Th1 cells, which promote the development of atherosclerosis by producing TNF‐α and IFN‐γ. Other T‐cell subsets also have been addressed in plaques, such as Th2, regulatory Th17 cells, NKT cells and so on. 73 In the ‘pre‐ILC‐era’, many studies’ primary aim is to study T‐cell responses in vascular diseases. However, it is worth noting that the lesion size cannot be significantly affected in the Ldlr−/−Rag1−/− mice, which are depleted the ILCs globally by anti‐CD90.2, whereas each subset of ILCs has divergent effects on atherosclerosis 74 (see below).
HELPER ILCs IN CARDIOVASCULAR DISEASE
Helper ILCs in cardiac inflammation
Myocarditis is caused by infectious agents including viruses, bacteria, protozoa and non‐infectious agents, which have the characteristics of leucocyte infiltration, myocardial oedema and fibrosis. 75 The viral infection is the most common cause of myocarditis. The development of viral myocarditis is related to autoimmune diseases. 76 If the disease cannot be recovered, it will convert into dilated cardiomyopathy. 58 The cardiac ILC is a static, phenotypic undifferentiated population; in physiologic conditions, ILCs do not have specific subset and immunophenotypes, although flow cytometry revealed that they are all characterized as CD45+Lin−CD127+ cells, and activated cardiac ILCs are characterized by transient co‐expression of Ki67 and PLZF in vitro. As described for ILC progenitors, while the inflammatory processes, the cardiac ILCs will develop ILC type 2 characteristics as a pathogenic role in promoting cardiac inflammation 14 , 59 , 77 (Figure 1).
Figure 1.
The role of the helper ILCs in inflammatory cardiac disease. Cardiac ILCs lack of specific markers of helper ILCs. In the inflammatory phase, CD29+ Sca‐1+ cardiac fibroblasts secrete IL‐33, which makes the cardiac ILCs transform into the cytokines produced nILC2s, and macrophages transform into anti‐inflammatory M2 type, to alleviate the cardiac inflammation. In pericardial fluid, activated ILC2s produce IL‐5 and IL‐13, which leads to cardiac fibroblasts to produce a large number of eotaxin, as well as the accumulation of eosinophils in heart. The production of IL‐1β by cardiac macrophages may promote the transformation of ILCs into ILC3s.
It has been demonstrated that there are two subsets of ILC2s, one of which is the ‘homeostatic or natural ILC2 cells’ (nILC2 cells, Lin−IL‐7Rα+Thy‐1hiST2+KLRG1int) stimulated by IL‐33, and the other is the IL‐25‐responsive ILC2 cells defined as ‘inflammatory ILC2 cells’ (iILC2 cells, Lin− IL‐7Rα+ Thy‐1loST2−KLRG1hi). 78 , 79 Not all organs and tissue have two types of ILC2s, and in experimental autoimmune myocarditis, cardiac ILCs cannot be differentiated into iILC2 for lacking IL‐25 receptor. 59 The activation and amplification for ILC2s through the IL‐33 have been demonstrated. 62 , 80 , 81 IL‐33 is involved in relieving cardiac hypertrophy induced by the transverse aortic constriction in mouse and has antihypertrophic and antifibrotic effects in myocardial infarction (MI) mouse model, which is a kind of myocardial necrosis caused by myocardial ischaemia and anoxia after coronary artery occlusion thrombosis. 82 , 83 , 84 , 85 IL‐33 can induce cardiac ST2L+CD4+T cells and ST2L+ macrophages to express IL‐4. IL‐4 offers the opportunity to induce macrophages into M2 macrophages and to promote the proliferation and accumulation of ILC2s, which play a protective role in viral myocarditis. 86 , 87 Additionally, Th2 differentiation relies on IL‐4‐mediated STAT6 signalling, providing the possibility that ILC2‐derived IL‐4 induced by IL‐33 might contribute as an initial source of IL‐4 for CD4+ T‐cell differentiation into Th2. 88 , 89 , 90 , 91 In contrast, other studies have found an inverse function of IL‐33 in the heart diseases. For example, pericarditis induced by IL‐33 in naïve and coxsackievirus B3‐infected mice results in increasing the proportion of eosinophils and ventricular dilation. 92 Also, it is found that the expression of IL‐33, promoting the transformation of ILC2 in heart (Figure 1), is mainly derived from CD29+Sca‐1+ cardiac fibroblasts both in experimental autoimmune myocarditis and MI models. 59 The inflammation of IL‐33‐induced pericarditis in Rag2−/−IL‐2Rγ−/− mice is relieved for the deficiency of lymphocytes including ILC2s; however, adoptive transfer of ILC2s into Rag2−/−IL‐2Rγ−/− mice restores the susceptibility of eosinophil infiltration; up‐regulated expression of eotaxin‐1 by cardiac fibroblasts dependent on IL‐5 secreted by ILC2s, and as a result, it promotes eosinophils to traffic into the heart directly. 93 Moreover, ILC2s can be stimulated to release IL‐5 through vasoactive intestinal peptide receptor type 2, which indicated that central circadian rhythms also involve in mediating basal eosinophilopoiesis and tissue eosinophil infiltration and accumulation. 94 Recently, many data have demonstrated that neuropeptides produced by neuronal, including positive regulators, such as neuromedin U and negative regulators, such as calca‐encoding calcitonin gene‐related peptide and β2‐adrenergic receptor, serve as mediators in ILC2 proliferation and effector cytokine production. 95 , 96 , 97 , 98 , 99 , 100 , 101 For example, ILC2 expresses neuromedin U receptor 1 involving in promoting ILC2‐mediated type 2 inflammation by increasing ILC2 frequency, 96 , 97 , 98 whereas neuropeptide calca‐encoding calcitonin gene‐related peptide co‐operates with neuromedin U and IL‐33 increasing IL‐5 expression and constraining ILC2s accumulation and IL‐13 production in lung inflammation. 99 , 100
ILC3 mainly expresses IL‐22 and IL‐17. IL‐17 has a great influence on the injury after MI, especially on ventricular remodelling. It has been indicated that IL‐23/IL‐17 axis obviously up‐regulated in myocardium following post‐MI. 102 , 103 IL‐17 as a contributor in promoting the development of cardiac diseases to dilated cardiomyopathy through their effects on cardiac fibroblasts. 104 , 105 IL‐17 results in cardiomyocyte apoptosis and participates in post‐MI ventricular remodelling through MAPK‐p53‐Bax signalling pathway. 106 Other data show that the size of MI decreases and the cardiac function improves in IL‐17‐deficient MI model. 107 The status of IL‐22 in cardiac diseases is controversial. Some data show that IL‐22 has a protective effect; for example, IL‐22 inhibits myocardial fibrosis in viral myocarditis and alleviates the pathological changes of rat heart tissue by reducing the stimulation of IL‐1. 108 , 109 However, the deficiency of IL‐17 or IL‐22 will not protect the heart from the inflammation of viral myocarditis. 110 Moreover, the expression of ILC3‐derived IL‐22 is decreased without neurotrophic factors RET which as a mediator influence ILC3s responsiveness to the external environment and signals. 111 There are no data addressing the role of ILC3s in heart disease, but ILC1s and ILC3s exist in myocarditis and MI 59 ; the number is less than ILC2s, so the value of ILC1s and ILC3s still needs to be studied.
HELPER ILCs IN VASCULAR DISEASE
A lot of studies have shown that plaque burden decreased in the atherosclerosis mice model lacking T‐bet, IFN‐γ and IL‐12, indicating the relationship between Th1 and atherosclerosis. 112 , 113 , 114 As a mirror cell of Th1, ILC1s sorted from the spleen of ApoE−/− TLR4+/+ mice are transferred into ApoE−/− Rag1−/− mice with anti‐NK1.1 mAbs for depleting ILC1 cells and aggravate atherosclerosis, whereas ILC1s from ApoE−/− TLR4−/− mice block the progression of atherosclerosis, and the size of atherosclerotic plaque in ApoE−/− Rag1−/− mice with the knockdown of ILC1s is smaller than that in NKs knockdown mice. 115 ILC1s in atherosclerosis express CD127, IFN‐γ, T‐bet, IL‐6 and IL‐1β, but lack c‐Kit, CD34, GATA‐3 and RORγt, distinct with NK cells. 116 IL‐12 plus IL‐18 could stimulate ILC1s from the spleen to secret IFN‐γ in vitro, whereas IL‐23 and IL‐1β cannot. The increase of IFN‐γ produced by ILC1 in ApoE−/− mice induced by oxLDL is related to the decrease of Bach2 expression. ILC1s respond to atherosclerosis danger signals may convey by the signalling pathway of TLR4/BACH2, which has significant effects on atherosclerosis in the early stage. 115
ILC2s are mainly distributed in lung and AT and can spontaneously secrete IL‐5. Similarly, the aortic CD90+ ST2+ Sca‐1+ CD117+ ILC2s can also produce IL‐5 to promote the proliferation of IgM‐secreting B cells through the regulation of transcription factor Id3. 117 Th2 has a protective effect as its production of IL‐4 and IL‐5 decelerate disease progression; however, IL‐4 produced by ILC2s benefits Th2 differentiation. 91 , 118 , 119 ILC2s have been authenticated in the atherosclerotic aorta of Ldlr−/−Rag1−/− mice, and CD25+ ILCs expanded by IL‐2/anti‐IL‐2 complexes can ameliorate atherosclerosis. Notably, the administration of IL‐2/anti‐IL‐2 also causes early signs of fibrosis in liver; however, it remains unclear whether the impact on lipid and cholesterol levels is due to the physiological correlation of ILC2s or just due to the pathological impact of chronic ILC2s activation on liver injury and scar formation. 74 IL‐33 and IL‐25 derived from epithelial cells are key activators of ILC2s and have an important role in injury or inflammation. IL‐33 decreases atherosclerosis in mice, while injecting the soluble ST2 promotes atherosclerosis progression. 118 Similarly, IL‐25 administration reduced atherosclerosis with increasing the number of ILC2s and their cytokines levels in serum (Figure 2). 119 Moreover, the lesional collagen content is increased in mice with IL‐13 treatment, whereas reduces monocyte recruitment; and lack of IL‐13 results in the aggravation of atherosclerosis. 120 IL‐4 from activated ILC2s functionally antagonizes the production of allergen‐specific Treg, while atherosclerosis is exacerbated significantly for Treg depletion. 121 , 122 Another study demonstrates that thymic stromal lymphopoietin plays a protective role in atherosclerosis that is mainly associated with reducing the macrophage infiltration and increasing lesional Treg. 123 , 124
Figure 2.
The role of the helper ILCs in vascular disease. In atherosclerotic plaque, ILC1s express transcription factor T‐bet induced by IL‐12 plus IL‐18 to produce a high level of IFN‐γ and aggravate atherosclerosis. CD25+ ILC2s can be induced to produce cytokines IL‐4, IL‐5, IL‐13 and promote the increase of M2 and Th2 cells to change lipid metabolism to reduce atherosclerosis. Circulating ILCs in blood vessels enter atherosclerotic plaques and participate in the formation of early atherosclerotic plaques. Macrophage phagocytosis of oxLDL forms foam cells, releasing IL‐1β, and promotes ILC3s to release IL‐17.
In the past decade, there are almost no published data of ILC3s in human or mice atherosclerosis. Mucosal tissue and skin are the common region of T cells and ILC3s secreting IL‐22 and IL‐17 to maintain barrier homeostasis. The treatment of ApoE−/−IL17A−/− mice with IL‐17 aggravates the pathological changes, involving in the decrease of the smooth muscle cells and collagen, and plaque instability. 107 , 125 Conversely, other studies have indicate that IL‐17 reduces atherosclerotic lesion formation on atherosclerosis in the mice of IL‐17 or IL‐17RA gene knockout. 126 Higher expression of RORγt and T‐bet was observed in ApoE−/− mouse atherosclerotic plaques, which results in higher levels of IFN‐γ and IL‐17. Furthermore, IL‐17 and IFN‐γ double production cells can be seen in the vascular wall, which is related to the size of the lesion. Although the authors only detect Th1 and Th17, ILC1s and ILC3s may also play critical roles in the pathogenesis of atherosclerosis. 107
Anti‐neutrophil cytoplasm antibody‐associated vasculitis is a severe multi‐system autoimmune diseases affecting the microvasculature, including granulomatosis with polyangiitis, microscopic polyangiitis and eosinophilic granulomatosis with polyangiitis (EGPA), which is classified according to the presence of affected small blood vessels and granuloma or asthma. 127 Despite the strong Th2 preponderance of EGPA, ILC2 has been proved to exist in the eosinophilic inflammatory site of humans. 128 , 129 , 130 , 131 Recent advances suggest that the increase of peripheral ILC2s accompaning with the higher IL‐33 in serum is related to disease activity in EGPA and the presence of active vasculitis shows the increase of serum IL‐33 concentration. 132 Other studies have shown that the total ILCs are decreased significantly containing the frequencies of ILC1s, while both ILC2s and ILC3s decrease during acute phase in granulomatosis with polyangiitis and microscopic polyangiitis in contrast with healthy controls. 133 However, these studies do not fully explain the reason for ILCs changes in each subgroup, and the specific mechanism needs further study.
CONCLUSIONS AND FUTURE DIRECTIONS
ILCs are one of the important cell subset in maintaining homeostasis and involving in innate immune response. Recently, it has been proved that most ILC subsets, as the first line against external infection, settle in specific organs, but the crosstalk between ILCs and tissues is still not clear. No comprehensive studies are addressing ILCs properties in cardiovascular system, and most of the reported data were obtained from mouse models or descriptive results of a small number of human patients. Many studies on the role of ILCs in diseases are carried out in the context of the lack of T and B cells, which leads us to neglect the interaction between the adaptive immune and ILCs. It is worth noting that knockdown of ILCs in atherosclerosis model does not alleviate the lesions, while with drugs that can stimulate the expansion of ILC2 in vivo, the lesions are relieved. In addition, ILCs in the adjacent tissues may release some corresponding molecules to stimulate the corresponding inflammatory cells to act on the cardiovascular system. Future work needs to address the different subsets of ILCs in the inflammatory environment, which would be helpful to give a novel perspective of ILCs in CVD. The efforts will help to design preventive strategies for innate and adaptive immune responses in CVD.
CONFLICTS OF INTEREST
None.
ACKNOWLEDGEMENTS
This work was supported by National Natural Science Foundation of China (Grant No. 81671567, 81871244), Jiangsu Province ‘333’ project (BRA2018016), Primary Research & Development Plan of Jiangsu Province (BE2019700) and Six talent peaks project in Jiangsu Province (2019‐WSN‐122).
Xiangmei Gong and Lin Xia contributed equally to this article.
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
REFERENCES
- 1. Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K, et al. Reducing the global burden of cardiovascular disease, Part 1: The epidemiology and risk factors. Circ Res. 2017; 121:677–94. [DOI] [PubMed] [Google Scholar]
- 2. Ketelhuth DFJ, Hansson GK. Adaptive response of T and B cells in atherosclerosis. Circ Res. 2016; 118:668–78. [DOI] [PubMed] [Google Scholar]
- 3. Ait‐Oufella H, Taleb S, Mallat Z, Tedgui A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011; 31:969–79. [DOI] [PubMed] [Google Scholar]
- 4. Liu Y, Zhu H, Su Z, Sun C, Yin J, Yuan H, et al. IL‐17 contributes to cardiac fibrosis following experimental autoimmune myocarditis by a PKCbeta/Erk1/2/NF‐kappaB‐dependent signaling pathway. Int Immunol. 2012; 24:605–12. [DOI] [PubMed] [Google Scholar]
- 5. Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente‐Casares X, Godwin J, et al. The macrophage in cardiac homeostasis and disease: JACC macrophage in CVD series (Part 4). J Am Coll Cardiol. 2018; 72:2213–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Epelman S, Liu PP, Mann DL. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol. 2015; 15:117–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Frangogiannis NG, Lindsey ML, Michael LH, Youker KA, Bressler RB, Mendoza LH, et al. Resident cardiac mast cells degranulate and release preformed TNF‐alpha, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation. 1998; 98:699–710. [DOI] [PubMed] [Google Scholar]
- 8. Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate lymphoid cells–a proposal for uniform nomenclature. Nat Rev Immunol. 2013; 13:145–9. [DOI] [PubMed] [Google Scholar]
- 9. Eberl G, Littman DR. The role of the nuclear hormone receptor RORgammat in the development of lymph nodes and Peyer's patches. Immunol Rev. 2003; 195:81–90. [DOI] [PubMed] [Google Scholar]
- 10. Scandella E, Bolinger B, Lattmann E, Miller S, Favre S, Littman DR, et al. Restoration of lymphoid organ integrity through the interaction of lymphoid tissue‐inducer cells with stroma of the T cell zone. Nat Immunol. 2008; 9:667–75. [DOI] [PubMed] [Google Scholar]
- 11. van de Pavert SA, Mebius RE. New insights into the development of lymphoid tissues. Nat Rev Immunol. 2010; 10:664–74. [DOI] [PubMed] [Google Scholar]
- 12. Diefenbach A, Colonna M, Koyasu S. Development, differentiation, and diversity of innate lymphoid cells. Immunity. 2014; 41:354–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Constantinides MG, McDonald BD, Verhoef PA, Bendelac A. A committed precursor to innate lymphoid cells. Nature. 2014; 508:397–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Harly C, Cam M, Kaye J, Bhandoola A. Development and differentiation of early innate lymphoid progenitors. J Exp Med. 2018; 215:249–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Ishizuka IE, Chea S, Gudjonson H, Constantinides MG, Dinner AR, Bendelac A, et al. Single‐cell analysis defines the divergence between the innate lymphoid cell lineage and lymphoid tissue‐inducer cell lineage. Nat Immunol. 2016; 17:269–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011; 331:44–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Seillet C, Belz GT, Huntington ND. Development, Homeostasis, and Heterogeneity of NK Cells and ILC1. Curr Top Microbiol Immunol. 2016; 395:37–61. [DOI] [PubMed] [Google Scholar]
- 18. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, et al. Activation of NK cell cytotoxicity. Mol Immunol. 2005; 42:501–10. [DOI] [PubMed] [Google Scholar]
- 19. Wallin RP, Screpanti V, Michaelsson J, Grandien A, Ljunggren HG. Regulation of perforin‐independent NK cell‐mediated cytotoxicity. Eur J Immunol. 2003; 33:2727–35. [DOI] [PubMed] [Google Scholar]
- 20. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate lymphoid cells: 10 years on. Cell. 2018; 174:1054–66. [DOI] [PubMed] [Google Scholar]
- 21. Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells–how did we miss them? Nat Rev Immunol. 2013; 13:75–87. [DOI] [PubMed] [Google Scholar]
- 22. Spits H, Bernink JH, Lanier L. NK cells and type 1 innate lymphoid cells: partners in host defense. Nat Immunol. 2016; 17:758–64. [DOI] [PubMed] [Google Scholar]
- 23. Klose CSN, Flach M, Mohle L, Rogell L, Hoyler T, Ebert K, et al. Differentiation of type 1 ILCs from a common progenitor to all helper‐like innate lymphoid cell lineages. Cell. 2014; 157:340–56. [DOI] [PubMed] [Google Scholar]
- 24. Abt MC, Lewis BB, Caballero S, Xiong H, Carter RA, Susac B, et al. Innate immune defenses mediated by two ILC subsets are critical for protection against acute clostridium difficile infection. Cell Host Microbe. 2015; 18:27–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Vosshenrich CA, Garcia‐Ojeda ME, Samson‐Villeger SI, Pasqualetto V, Enault L, Richard‐Le Goff O, et al. A thymic pathway of mouse natural killer cell development characterized by expression of GATA‐3 and CD127. Nat Immunol. 2006; 7:1217–24. [DOI] [PubMed] [Google Scholar]
- 26. Paust S, Gill HS, Wang BZ, Flynn MP, Moseman EA, Senman B, et al. Critical role for the chemokine receptor CXCR6 in NK cell‐mediated antigen‐specific memory of haptens and viruses. Nat Immunol. 2010; 11:1127‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Weizman OE, Adams NM, Schuster IS, Krishna C, Pritykin Y, Lau C, et al. ILC1 confer early host protection at initial sites of viral infection. Cell. 2017; 171:795–808.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015; 517:293–301. [DOI] [PubMed] [Google Scholar]
- 29. Hoyler T, Klose CS, Souabni A, Turqueti‐Neves A, Pfeifer D, Rawlins EL, et al. The transcription factor GATA‐3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity. 2012; 37:634–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Klein Wolterink RG, Serafini N, van Nimwegen M, Vosshenrich CA, de Bruijn MJ, Fonseca Pereira D, et al. Essential, dose‐dependent role for the transcription factor Gata3 in the development of IL‐5+ and IL‐13+ type 2 innate lymphoid cells. Proc Natl Acad Sci USA. 2013; 110:10240–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Moro K, Kabata H, Tanabe M, Koga S, Takeno N, Mochizuki M, et al. Interferon and IL‐27 antagonize the function of group 2 innate lymphoid cells and type 2 innate immune responses. Nat Immunol. 2016; 17:76–86. [DOI] [PubMed] [Google Scholar]
- 32. Price AE, Liang HE, Sullivan BM, Reinhardt RL, Eisley CJ, Erle DJ, et al. Systemically dispersed innate IL‐13‐expressing cells in type 2 immunity. Proc Natl Acad Sci USA. 2010; 107:11489–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Neill DR, Wong SH, Bellosi A, Flynn RJ, Daly M, Langford TK, et al. Nuocytes represent a new innate effector leukocyte that mediates type‐2 immunity. Nature. 2010; 464:1367–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Turner JE, Morrison PJ, Wilhelm C, Wilson M, Ahlfors H, Renauld JC, et al. IL‐9‐mediated survival of type 2 innate lymphoid cells promotes damage control in helminth‐induced lung inflammation. J Exp Med. 2013; 210:2951–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Lim AI, Li Y, Lopez‐Lastra S, Stadhouders R, Paul F, Casrouge A, et al. Systemic human ILC precursors provide a substrate for tissue ILC differentiation. Cell. 2017; 168:1086–100.e10. [DOI] [PubMed] [Google Scholar]
- 36. Melo‐Gonzalez F, Hepworth MR. Functional and phenotypic heterogeneity of group 3 innate lymphoid cells. Immunology. 2017; 150:265–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Withers DR, Hepworth MR. Group 3 innate lymphoid cells: communications hubs of the intestinal immune system. Front Immunol. 2017; 8:1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Hoorweg K, Peters CP, Cornelissen F, Aparicio‐Domingo P, Papazian N, Kazemier G, et al. Functional differences between human NKp44‐ and NKp44+RORC+ innate lymphoid cells. Front Immunol. 2012; 3:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Hepworth MR, Monticelli LA, Fung TC, Ziegler CG, Grunberg S, Sinha R, et al. Innate lymphoid cells regulate CD4+T‐cell responses to intestinal commensal bacteria. Nature. 2013; 498:113–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Mebius RE, Rennert P, Weissman IL. Developing lymph nodes collect CD4+CD3‐ LTβ+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity. 1997; 7:493–504. [DOI] [PubMed] [Google Scholar]
- 41. Cherrier M, Eberl G. The development of LTi cells. Curr Opin Immunol. 2012; 24:178–83. [DOI] [PubMed] [Google Scholar]
- 42. Scoville SD, Freud AG, Caligiuri MA. Cellular pathways in the development of human and murine innate lymphoid cells. Curr Opin Immunol. 2019; 56:100–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Eberl G, Marmon S, Sunshine MJ, Rennert PD, Choi Y, Littman DR. An essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer cells. Nat Immunol. 2004; 5:64–73. [DOI] [PubMed] [Google Scholar]
- 44. Yoshida H, Naito A, Inoue J‐I, Satoh M, Santee‐Cooper SM, Ware CF, et al. Different cytokines induce surface lymphotoxin‐αβ on IL‐7 receptor‐α cells that differentially engender lymph nodes and Peyer's Patches. Immunity. 2002; 17:823–33. [DOI] [PubMed] [Google Scholar]
- 45. Cupedo T, Vondenhoff MF, Heeregrave EJ, De Weerd AE, Jansen W, Jackson DG, et al. Presumptive lymph node organizers are differentially represented in developing mesenteric and peripheral nodes. J Immunol. 2004; 173:2968–75. [DOI] [PubMed] [Google Scholar]
- 46. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, et al. Lymphoid tissue inducer‐like cells are an innate source of IL‐17 and IL‐22. J Exp Med. 2009; 206:35–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Ota N, Wong K, Valdez PA, Zheng Y, Crellin NK, Diehl L, et al. IL‐22 bridges the lymphotoxin pathway with the maintenance of colonic lymphoid structures during infection with Citrobacter rodentium . Nat Immunol. 2011; 12:941–8. [DOI] [PubMed] [Google Scholar]
- 48. Ciccia F, Guggino G, Rizzo A, Saieva L, Peralta S, Giardina A, et al. Type 3 innate lymphoid cells producing IL‐17 and IL‐22 are expanded in the gut, in the peripheral blood, synovial fluid and bone marrow of patients with ankylosing spondylitis. Ann Rheum Dis. 2015; 74:1739–47. [DOI] [PubMed] [Google Scholar]
- 49. Kim M‐Y, Gaspal FMC, Wiggett HE, McConnell FM, Gulbranson‐Judge A, Raykundalia C, et al. CD4+CD3− accessory cells costimulate primed CD4 T cells through OX40 and CD30 at sites where T cells collaborate with B cells. Immunity. 2003; 18:643–54. [DOI] [PubMed] [Google Scholar]
- 50. Kim MY, Anderson G, White A, Jenkinson E, Arlt W, Martensson IL, et al. OX40 ligand and CD30 ligand are expressed on adult but not neonatal CD4+CD3‐ inducer cells: evidence that IL‐7 signals regulate CD30 ligand but not OX40 ligand expression. J Immunol. 2005; 174:6686–91. [DOI] [PubMed] [Google Scholar]
- 51. Ebbo M, Crinier A, Vely F, Vivier E. Innate lymphoid cells: major players in inflammatory diseases. Nat Rev Immunol. 2017; 17:665–78. [DOI] [PubMed] [Google Scholar]
- 52. Klose CS, Artis D. Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat Immunol. 2016; 17:765–74. [DOI] [PubMed] [Google Scholar]
- 53. Sonnenberg GF, Hepworth MR. Functional interactions between innate lymphoid cells and adaptive immunity. Nat Rev Immunol. 2019; 19:599–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Ong S, Ligons DL, Barin JG, Wu L, Talor MV, Diny N, et al. Natural killer cells limit cardiac inflammation and fibrosis by halting eosinophil infiltration. Am J Pathol. 2015; 185:847–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Selathurai A, Deswaerte V, Kanellakis P, Tipping P, Toh BH, Bobik A, et al. Natural killer (NK) cells augment atherosclerosis by cytotoxic‐dependent mechanisms. Cardiovasc Res. 2014; 102:128–37. [DOI] [PubMed] [Google Scholar]
- 56. Bonaccorsi I, Spinelli D, Cantoni C, Barilla C, Pipito N, De Pasquale C, et al. Symptomatic carotid atherosclerotic plaques are associated with increased infiltration of Natural Killer (NK) cells and higher serum levels of NK activating receptor ligands. Front Immunol. 2019; 10:1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Ong S, Rose NR, Cihakova D. Natural killer cells in inflammatory heart disease. Clin Immunol. 2017; 175:26–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol. 2018; 18:733–44. [DOI] [PubMed] [Google Scholar]
- 59. Bracamonte‐Baran W, Chen G, Hou X, Talor MV, Choi HS, Davogustto G, et al. Non‐cytotoxic cardiac innate lymphoid cells are a resident and quiescent Type 2‐commited population. Front Immunol. 2019; 10:634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Mjosberg J, Spits H. Human innate lymphoid cells. J Allergy Clin Immunol. 2016; 138:1265–76. [DOI] [PubMed] [Google Scholar]
- 61. Meilhac SM, Lescroart F, Blanpain C, Buckingham ME. Cardiac cell lineages that form the heart. Cold Spring Harb Perspect Med. 2014; 4:a013888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of T(H)2 cytokines by adipose tissue‐associated c‐Kit+Sca‐1+ lymphoid cells. Nature. 2010; 463:540–4. [DOI] [PubMed] [Google Scholar]
- 63. Butts B, Goeddel LA, George DJ, Steele C, Davies JE, Wei CC, et al. Increased inflammation in pericardial fluid persists 48 hours after cardiac surgery. Circulation. 2017; 136:2284–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Horckmans M, Bianchini M, Santovito D, Megens RTA, Springael JY, Negri I, et al. Pericardial adipose tissue regulates granulopoiesis, fibrosis, and cardiac function after myocardial infarction. Circulation. 2018; 137:948–60. [DOI] [PubMed] [Google Scholar]
- 65. Molofsky AB, Nussbaum JC, Liang HE, Van Dyken SJ, Cheng LE, Mohapatra A, et al. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J Exp Med. 2013; 210:535–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Lee MW, Odegaard JI, Mukundan L, Qiu Y, Molofsky AB, Nussbaum JC, et al. Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell. 2015; 160:74–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Brestoff JR, Kim BS, Saenz SA, Stine RR, Monticelli LA, Sonnenberg GF, et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 2015; 519:242–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Molofsky AB, Van Gool F, Liang HE, Van Dyken SJ, Nussbaum JC, Lee J, et al. Interleukin‐33 and Interferon‐gamma Counter‐Regulate Group 2 innate lymphoid cell activation during immune perturbation. Immunity. 2015; 43:161–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. O'Sullivan TE, Rapp M, Fan X, Weizman OE, Bhardwaj P, Adams NM, et al. Adipose‐resident Group 1 innate lymphoid cells promote obesity‐associated insulin resistance. Immunity. 2016; 45:428–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Boulenouar S, Michelet X, Duquette D, Alvarez D, Hogan AE, Dold C, et al. Adipose type one innate lymphoid cells regulate macrophage homeostasis through targeted cytotoxicity. Immunity. 2017; 46:273–86. [DOI] [PubMed] [Google Scholar]
- 71. Williams JW, Huang LH, Randolph GJ. Cytokine circuits in cardiovascular disease. Immunity. 2019; 50:941–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. McGuire PJ. Chemical individuality in T cells: a Garrodian view of immunometabolism. Immunol Rev. 2020; 295:82–100. [DOI] [PubMed] [Google Scholar]
- 73. Ketelhuth DF, Hansson GK. Cellular immunity, low‐density lipoprotein and atherosclerosis: break of tolerance in the artery wall. Thromb Haemost. 2011; 106:779–86. [DOI] [PubMed] [Google Scholar]
- 74. Engelbertsen D, Foks AC, Alberts‐Grill N, Kuperwaser F, Chen T, Lederer JA, et al. Expansion of CD25+ innate lymphoid cells reduces atherosclerosis. Arterioscler Thromb Vasc Biol. 2015; 35:2526–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Caforio AL, Marcolongo R, Basso C, Iliceto S. Clinical presentation and diagnosis of myocarditis. Heart. 2015; 101:1332–44. [DOI] [PubMed] [Google Scholar]
- 76. Rose NR. Viral myocarditis. Curr Opin Rheumatol. 2016; 28:383–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Xu W, Cherrier DE, Chea S, Vosshenrich C, Serafini N, Petit M, et al. An Id2(RFP)‐reporter mouse redefines innate lymphoid cell precursor potentials. Immunity 2019; 50:1054–68.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Jiang S, Yan W. T‐cell immunometabolism against cancer. Cancer Lett. 2016; 382:255–8. [DOI] [PubMed] [Google Scholar]
- 79. Huang Y, Guo L, Qiu J, Chen X, Hu‐Li J, Siebenlist U, et al. IL‐25‐responsive, lineage‐negative KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. Nat Immunol. 2015; 16:161–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. von Moltke J, Locksley RM. I‐L‐C‐2 it: type 2 immunity and group 2 innate lymphoid cells in homeostasis. Curr Opin Immunol. 2014; 31:58–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Kim BS, Artis D. Group 2 innate lymphoid cells in health and disease. Cold Spring Harb Perspect Biol. 2015; 7:a016337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Chen WY, Hong J, Gannon J, Kakkar R, Lee RT. Myocardial pressure overload induces systemic inflammation through endothelial cell IL‐33. Proc Natl Acad Sci USA. 2015; 112:7249–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Sanada S, Hakuno D, Higgins LJ, Schreiter ER, McKenzie AN, Lee RT. IL‐33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest. 2007; 117:1538–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Palasubramaniam J, Wang X, Peter K. Myocardial infarction‐from atherosclerosis to thrombosis. Arterioscler Thromb Vasc Biol. 2019; 39:e176–85. [DOI] [PubMed] [Google Scholar]
- 85. Musher DM, Abers MS, Corrales‐Medina VF. Acute infection and myocardial infarction. N Engl J Med. 2019; 380:171–6. [DOI] [PubMed] [Google Scholar]
- 86. Wang C, Dong C, Xiong S. IL‐33 enhances macrophage M2 polarization and protects mice from CVB3‐induced viral myocarditis. J Mol Cell Cardiol. 2017; 103:22–30. [DOI] [PubMed] [Google Scholar]
- 87. Symowski C, Voehringer D. Th2 cell‐derived IL‐4/IL‐13 promote ILC2 accumulation in the lung by ILC2‐intrinsic STAT6 signaling in mice. Eur J Immunol. 2019; 49:1421–32. [DOI] [PubMed] [Google Scholar]
- 88. Shimoda K, van Deursen J, Sangster MY, Sarawar SR, Carson RT, Tripp RA, et al. Lack of IL‐4‐induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature. 1996; 380:630–3. [DOI] [PubMed] [Google Scholar]
- 89. Kaplan MH, Schindler U, Smiley ST, Grusby MJ. Stat6 is required for mediating responses to IL‐4 and for the development of Th2 cells. Immunity. 1996; 4:313–9. [DOI] [PubMed] [Google Scholar]
- 90. Oliphant CJ, Hwang YY, Walker JA, Salimi M, Wong SH, Brewer JM, et al. MHCII‐mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity. 2014; 41:283–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Pelly VS, Kannan Y, Coomes SM, Entwistle LJ, Ruckerl D, Seddon B, et al. IL‐4‐producing ILC2s are required for the differentiation of TH2 cells following Heligmosomoides polygyrus infection. Mucosal Immunol. 2016; 9:1407–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Abston ED, Barin JG, Cihakova D, Bucek A, Coronado MJ, Brandt JE, et al. IL‐33 independently induces eosinophilic pericarditis and cardiac dilation: ST2 improves cardiac function. Circ Heart Fail. 2012; 5:366–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Choi HS, Won T, Hou X, Chen G, Bracamonte‐Baran W, Talor MV, et al. Innate lymphoid cells play a pathogenic role in pericarditis. Cell Rep 2020; 30:2989–3003.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature. 2013; 502:245–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Moriyama S, Brestoff JR, Flamar AL, Moeller JB, Klose CSN, Rankin LC, et al. beta2‐adrenergic receptor‐mediated negative regulation of group 2 innate lymphoid cell responses. Science. 2018; 359:1056–61. [DOI] [PubMed] [Google Scholar]
- 96. Cardoso V, Chesne J, Ribeiro H, Garcia‐Cassani B, Carvalho T, Bouchery T, et al. Neuronal regulation of type 2 innate lymphoid cells via neuromedin U. Nature. 2017; 549:277–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Klose CSN, Mahlakoiv T, Moeller JB, Rankin LC, Flamar AL, Kabata H, et al. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature. 2017; 549:282–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour RE, Nyman J, Dionne D, et al. The neuropeptide NMU amplifies ILC2‐driven allergic lung inflammation. Nature. 2017; 549:351–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Nagashima H, Mahlakoiv T, Shih HY, Davis FP, Meylan F, Huang Y, et al. Neuropeptide CGRP limits Group 2 innate lymphoid cell responses and constrains Type 2 inflammation. Immunity. 2019; 51:682–95.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Wallrapp A, Burkett PR, Riesenfeld SJ, Kim SJ, Christian E, Abdulnour RE, et al. Calcitonin gene‐related peptide negatively regulates alarmin‐driven Type 2 innate lymphoid cell responses. Immunity 2019; 51:709–23.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Veiga‐Fernandes H, Pachnis V. Neuroimmune regulation during intestinal development and homeostasis. Nat Immunol 2017; 18:116–22. [DOI] [PubMed] [Google Scholar]
- 102. Ávalos AM. IL‐17A levels increase in the infarcted region of the left ventricle in a rat model of myocardial infarction. Biol Res. 2012; 45:193–200. [DOI] [PubMed] [Google Scholar]
- 103. Yan X, Shichita T, Katsumata Y, Matsuhashi T, Ito H, Ito K, et al. Deleterious effect of the IL‐23/IL‐17A axis and gammadeltaT cells on left ventricular remodeling after myocardial infarction. J Am Heart Assoc. 2012; 1:e004408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Nindl V, Maier R, Ratering D, De Giuli R, Zust R, Thiel V, et al. Cooperation of Th1 and Th17 cells determines transition from autoimmune myocarditis to dilated cardiomyopathy. Eur J Immunol. 2012; 42:2311–21. [DOI] [PubMed] [Google Scholar]
- 105. Baldeviano GC, Barin JG, Talor MV, Srinivasan S, Bedja D, Zheng D, et al. Interleukin‐17A is dispensable for myocarditis but essential for the progression to dilated cardiomyopathy. Circ Res. 2010; 106:1646–55. [DOI] [PubMed] [Google Scholar]
- 106. Zhou SF, Yuan J, Liao MY, Xia N, Tang TT, Li JJ, et al. IL‐17A promotes ventricular remodeling after myocardial infarction. J Mol Med (Berl). 2014; 92:1105–16. [DOI] [PubMed] [Google Scholar]
- 107. Gao Q, Jiang Y, Ma T, Zhu F, Gao F, Zhang P, et al. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J Immunol. 2010; 185:5820–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Chang H, Hanawa H, Liu H, Yoshida T, Hayashi M, Watanabe R, et al. Hydrodynamic‐based delivery of an interleukin‐22‐Ig fusion gene ameliorates experimental autoimmune myocarditis in rats. J Immunol. 2006; 177:3635–43. [DOI] [PubMed] [Google Scholar]
- 109. Guo Y, Wu W, Cen Z, Li X, Kong Q, Zhou Q. IL‐22‐producing Th22 cells play a protective role in CVB3‐induced chronic myocarditis and dilated cardiomyopathy by inhibiting myocardial fibrosis. Virol J. 2014; 11:230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Kong Q, Xue Y, Wu W, Yang F, Liu Y, Gao M, et al. IL‐22 exacerbates the severity of CVB3‐induced acute viral myocarditis in IL‐17A‐deficient mice. Mol Med Rep. 2013; 7:1329–35. [DOI] [PubMed] [Google Scholar]
- 111. Ibiza S, Garcia‐Cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, et al. Glial‐cell‐derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature. 2016; 535:440–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH. Influence of interferon‐gamma on the extent and phenotype of diet‐induced atherosclerosis in the LDLR‐deficient mouse. Arterioscler Thromb Vasc Biol. 2003; 23:454–60. [DOI] [PubMed] [Google Scholar]
- 113. Davenport P, Tipping PG. The role of Interleukin‐4 and Interleukin‐12 in the progression of atherosclerosis in apolipoprotein E‐deficient mice. Am J Pathol. 2003; 163:1117–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Buono C, Binder CJ, Stavrakis G, Witztum JL, Glimcher LH, Lichtman AH. T‐bet deficiency reduces atherosclerosis and alters plaque antigen‐specific immune responses. Proc Natl Acad Sci USA. 2005; 102:1596–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Wu C, He S, Liu J, Wang B, Lin J, Duan Y, et al. Type 1 innate lymphoid cell aggravation of atherosclerosis is mediated through TLR4. Scand J Immunol. 2018; 87:e12661. [DOI] [PubMed] [Google Scholar]
- 116. Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, et al. Evidence for discrete stages of human natural killer cell differentiation in vivo. J Exp Med. 2006; 203:1033–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Perry HM, Oldham SN, Fahl SP, Que X, Gonen A, Harmon DB, et al. Helix‐loop‐helix factor inhibitor of differentiation 3 regulates interleukin‐5 expression and B‐1a B cell proliferation. Arterioscler Thromb Vasc Biol. 2013; 33:2771–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Liew FY, McInnes IB, Baker AH, Sattar N, Li Y, Denby L, et al. IL‐33 reduces the development of atherosclerosis. J Exp Med. 2008; 205:339–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Mantani PT, Duner P, Bengtsson E, Alm R, Ljungcrantz I, Soderberg I, et al. IL‐25 inhibits atherosclerosis development in apolipoprotein E deficient mice. PLoS One. 2015; 10:e0117255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Cardilo‐Reis L, Gruber S, Schreier SM, Drechsler M, Papac‐Milicevic N, Weber C, et al. Interleukin‐13 protects from atherosclerosis and modulates plaque composition by skewing the macrophage phenotype. EMBO Mol Med. 2012; 4:1072–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Noval Rivas M, Burton OT, Oettgen HC, Chatila T. IL‐4 production by group 2 innate lymphoid cells promotes food allergy by blocking regulatory T‐cell function. J Allergy Clin Immunol. 2016; 138:801–11.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Ait‐Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, Zoll J, et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nat Med. 2006; 12:178–80. [DOI] [PubMed] [Google Scholar]
- 123. Yu K, Dong Q, Mao X, Meng K, Zhao X, Ji Q, et al. Disruption of the TSLP‐TSLPR‐LAP signaling between epithelial and dendritic cells through hyperlipidemia contributes to regulatory T‐Cell defects in atherosclerotic mice. Atherosclerosis. 2015; 238:278–88. [DOI] [PubMed] [Google Scholar]
- 124. Steinmetz M, Laurans L, Nordsiek S, Weiss L, van der Veken B, Ponnuswamy P, et al. Thymic stromal lymphopoietin is a key cytokine for the immunomodulation of atherogenesis with Freund's adjuvant. J Cell Mol Med. 2020; 24:5731–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Danzaki K, Matsui Y, Ikesue M, Ohta D, Ito K, Kanayama M, et al. Interleukin‐17A deficiency accelerates unstable atherosclerotic plaque formation in apolipoprotein E‐deficient mice. Arterioscler Thromb Vasc Biol. 2012; 32:273–80. [DOI] [PubMed] [Google Scholar]
- 126. Butcher MJ, Gjurich BN, Phillips T, Galkina EV. The IL‐17A/IL‐17RA axis plays a proatherogenic role via the regulation of aortic myeloid cell recruitment. Circ Res. 2012; 110:675–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of vasculitides. Arthritis Rheum. 2013; 65:1–11. [DOI] [PubMed] [Google Scholar]
- 128. Kim BS, Siracusa MC, Saenz SA, Noti M, Monticelli LA, Sonnenberg GF, et al. TSLP Elicits IL‐33–Independent innate lymphoid cell responses to promote skin inflammation. Sci Transl Med. 2013; 5:170ra16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kwon BI, Hong S, Shin K, Choi EH, Hwang JJ, Lee SH. Innate type 2 immunity is associated with eosinophilic pleural effusion in primary spontaneous pneumothorax. Am J Respir Crit Care Med. 2013; 188:577–85. [DOI] [PubMed] [Google Scholar]
- 130. Shaw JL, Fakhri S, Citardi MJ, Porter PC, Corry DB, Kheradmand F, et al. IL‐33‐responsive innate lymphoid cells are an important source of IL‐13 in chronic rhinosinusitis with nasal polyps. Am J Respir Crit Care Med. 2013; 188:432–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Bartemes KR, Kephart GM, Fox SJ, Kita H. Enhanced innate type 2 immune response in peripheral blood from patients with asthma. J Allergy Clin Immunol 2014; 134:671–8.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Tsurikisawa N, Oshikata C, Watanabe M, Tsuburai T, Kaneko T, Saito H. Innate immune response reflects disease activity in eosinophilic granulomatosis with polyangiitis. Clin Exp Allergy. 2018; 48:1305–16. [DOI] [PubMed] [Google Scholar]
- 133. Braudeau C, Amouriaux K, Neel A, Herbreteau G, Salabert N, Rimbert M, et al. Persistent deficiency of circulating mucosal‐associated invariant T (MAIT) cells in ANCA‐associated vasculitis. J Autoimmun. 2016; 70:73–9. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.