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.

1. Introduction

Myocarditis, an inflammatory heart disease (i.e. mainly observed in young people), can cause sudden death, secondary dilated cardiomyopathy (DCM), and heart failure.1 Although myocarditis has a clear definition, its diagnosis is far from simple. Clinical symptoms, laboratory tests, electrocardiography, echocardiography, cardiac magnetic resonance imaging (cMRI), and endomyocardial biopsy (EMB) are all used to diagnose myocarditis.2,3 Several previous studies4–7 have revealed susceptible strains of mice and genetically susceptible individuals8–11 that progress to DCM after myocarditis.

Even though the detailed pathophysiology of myocarditis is still unknown, immune cells have attracted much interest as playing key roles. The pathogenesis of myocarditis is thought to primarily involve neutrophils, eosinophils, mast cells, innate lymphoid cells (ILCs), monocytes, macrophages, dendritic cells (DCs), T lymphocytes, and B lymphocytes.12–15 In the steady state, the heart contains a population of intrinsic immune cells. During illness, these cells act as the body's first line of defense,12 and along with subsequent activation of adaptive immunity, they form a complex immune network that plays a dual role comprising both protection and destruction. In addition to immune system dysregulation, myocarditis may also involve an imbalance within immune cells.15

Single-cell sequencing techniques16 have been used to gain deeper insights at the single-cell level. Several studies17–20 based on single-cell RNA sequencing (scRNA-seq) have suggested that scRNA-seq has been regarded as the more suitable tool to find out some molecules specifically expressed within certain cell classes because these molecules may be easily overlooked because they are likely to be expressed sightly on bulk RNA-seq.21

In this review, we comprehensively describe the diversity and plasticity in the function of major immune cells and their associated cellular contents in the context of myocarditis, laying the foundation for subsequent in-depth studies based on the vast amount of immunological data.

2. Myocarditis

2.1 Definition, prevalence, diagnosis, and etiology

Myocarditis is an inflammatory disease of the myocardium that appears without an ischemic event. Myocarditis is a frequent contributor to sudden death, secondary DCM, and heart failure in the young population.1

In 2021, myocarditis was estimated to be the cause of 33,600 deaths (95% uncertainty interval [UI] 27,100–38,000), and the number of disability-adjusted life-years was 962,000 (95% UI 810,000–1,090,000).22 Males make up a larger proportion of adult myocarditis patients than females, and mortality follows a similar trend.22,23

With the increased use of immune checkpoint inhibitors (ICIs), which target programmed cell death 1 (PD-1), CTLA-4 (cytotoxic T lymphocyte-associated protein 4), and programmed cell death ligand 1 (PD-L1) in oncology therapy, ICI-associated myocarditis (ICI-MC) is becoming more common. The incidence of ICI-MC ranges from 0.04% to 1.14%, but the associated mortality rate is up to 25% to 50%, which is much greater than that of other immune-related adverse events.24

With the recent appearance of COVID-19, myocarditis after COVID-19 and messenger RNA (mRNA) COVID-19 vaccination has attracted more attention. Research based on the Premier Healthcare Database Special COVID-19 Release found that patients with COVID-19 had an approximately 16 times greater risk of myocarditis than patients without.25 According to many previous reports, myocarditis is regarded as a rare complication of COVID-19 vaccination.26–30 Based on a self-controlled case series study of individuals 16 years of age or older who received COVID-19 vaccination in England between December 1, 2020, and August 24, 2021, researchers31 found that increased risks of myocarditis within 1 to 28 d postvaccination were related to the first dose of adenovirus ChAdOx1 vaccines (incidence rate ratio [IRR] 1.29, 95% confidence interval [CI] 1.05–1.58) and mRNA-based BNT162b2 vaccines (IRR 1.31, 95% CI 1.03–1.66), as well as the first (IRR 2.97; 95% CI 1.34–6.58) and second (IRR 9.84, 95% CI 2.69–36.03) doses of the mRNA-1273 vaccine. In the meantime, the risk of myocarditis was increased within 1 to 28 d after COVID-19 (IRR 9.76, 95% CI 7.51–12.69).

Medical history, clinical manifestation, electrocardiography, echocardiography, and laboratory tests are now used to support the clinical diagnosis of myocarditis.3 Nowadays, histological or immunohistological evidence of inflammatory cell infiltration, with or without myocardial cell damage in the myocardium as revealed by EMB, is thought to be the gold-standard diagnostic method for myocarditis.32 When EMB is performed by a skilled technician, complications are quite rare.33 However, EMB can only be conducted locally, is intrusive, and has risks that are correlated with the operator's degree of competence. As a result, its performance is constrained by the patient's preference and the operator's skill.

cMRI is gradually being used as a noninvasive imaging test for the diagnosis and dynamic detection of myocarditis. Only symptomatic patients, patients with significant clinical evidence of myocarditis, and patients whose cMRI results are likely to have an impact on clinical therapy should undergo cMRI examination.34 The initial Lake Louise Criteria34 for myocarditis were published in 2009, which identified 3 signs of myocardial inflammation and their corresponding markers on cMRI, including (1) hyperemia, which presents as an intense signal on early gadolinium enhancement images; (2) tissue edema, which manifests as an increased myocardial T2 relaxation time or an elevated signal intensity on T2-weighted images; and (3) necrosis/fibrosis, which appears as a late gadolinium enhancement image. The diagnosis of acute myocarditis is possible if 2 of these 3 criteria are fulfilled. In 2018, the updated Lake Louise Criteria2 were published, including (1) T2-based markers, which include a regional high T2 signal intensity, a global T2 signal intensity ratio of ≥2.0 on T2-weighted images, or a regional or global increase in myocardial T2 relaxation time; and (2) T1-based markers, which include a regional or global increase in the native myocardial T1 relaxation time or extracellular volume mapping, or areas with a high signal intensity in a nonischemic distribution pattern on late gadolinium enhancement images. The specificity for identifying acute myocarditis may increase if both a positive T2-based marker and a T1-based marker are present.2 In 2020, the Society for Cardiovascular Magnetic Resonance included myocarditis as a primary indication for cMRI, revealing the importance of this imaging modality in the evaluation of myocarditis.35

An additional noninvasive imaging test is echocardiography, which is often useful for ruling out heart failure not due to myocarditis and in detecting ventricular thrombi. Echocardiography may reveal ventricular enlargement, ventricular wall thickening, decreased myocardial segmental mobility, and lower ejection fraction.3

The main way to identify changes in the electrical activity of the heart in patients with myocarditis is electrocardiography. Nonspecific T-wave alterations are the most frequent modifications; however, ST-segment dynamic modifications and newly developed pathogenic Q waves resembling myocardial infarction may also be present. Many arrhythmias, such as ventricular tachycardia, ventricular fibrillation, premature contraction, atrioventricular block, and (in severe cases) cardiac arrest, are commonly present in patients with myocarditis.3,36

Laboratory tests in patients with myocarditis may show elevated myocardial injury markers, such as cardiac troponin T, cardiac troponin I, creatine kinase isoenzyme, and N-terminal pro–brain natriuretic peptide, as well as nonspecific serum markers associated with inflammation, such as elevated white blood cell counts, C-reactive protein, and erythrocyte sedimentation rate.1,3,32 However, it is worth noting that the troponins are not often increased in mild cases of myocarditis.

Patients with myocarditis may have a variety of clinical manifestations, including chest pain, dyspnea, syncope, palpitations, and prodromal respiratory and gastrointestinal infections.3

The etiology of myocarditis is mainly divided into infectious and noninfectious factors. Infectious factors include a variety of pathogens, such as viruses, bacteria, chlamydia, Rickettsia, fungi, and parasites, with viral infections predominating in developed countries.37 Noninfectious factors include systemic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis; drugs, such as aminophylline, amphetamines, anthracyclines, catecholamines, and ICIs; hypersensitivity reactions; and tumors.1

2.2 Pathogenesis of myocarditis

Previous studies have examined the pathogenesis of myocarditis, summarizing the classic 3-phase model,38,39 comprising the acute infection phase, the subacute immune response phase, and the chronic myopathy phase, using viral myocarditis as an example.

2.2.1 Acute infection phase

The acute infection phase is mainly caused by the first invasion of the pathogen40,41 or reactivation of the pathogen in a dormant state.40,42 When the virus reaches the cardiomyocyte, it uses cell membrane surface-specific receptors or receptor complexes to enter the cell. For example, coxsackievirus (CVB3) uses the coxsackievirus-adenovirus receptor (CAR) to enter the cell and then proliferates, leading to direct cellular damage.41,43,44

2.2.2 Subacute immune response phase

An appropriate immune response contributes to pathogen clearance, whereas an excessive immune response can cause tissue damage.40 Pattern recognition receptors (PRRs), such as Toll-like receptors and nucleotide oligomerization domain-like receptors,45 are present on the surface or in the cytoplasm of most cells in the cardiovascular system.46 PRRs recognize and bind to pathogen-associated molecular patterns, such as lipopolysaccharides, lipoproteins, and nucleic acids, as well as to damage-associated molecular patterns, activating47 a series of downstream signaling pathways, such as nuclear factor (NF-κB) signaling, to produce proinflammatory cytokines.47 The combination can also induce NLRP3 inflammasomes, subsequently causing pyroptosis.48

Complement (C), which is one of the key components of innate immunity, can be activated by bound antibodies through either the alternative pathway or the classical pathway.49 In response to activation, C split products are produced, which have strong proinflammatory effects and enhance both the innate and adaptive immune responses by binding to C receptors (CRs).49 Upregulation of CD11b/CR3 in the majority of infiltrating immune cells has been reported, both in male and female BALB/c mice using the new model of CVB3 myocarditis.50–53 The depletion of C3 or the blockade of CR1/2 can prevent experimental autoimmune myocarditis (EAM) in A/J mice by impairing the ability to form the membrane attack complex.5,54 The lack of CR1/2 promotes the severity of CVB3 myocarditis in male C57BL/6 backcrossed to A/J mice by increasing macrophage, interleukin (IL)-1β, and immune complex deposition in the myocardium.51

Subsequently, T lymphocytes, otherwise known as T cells, are activated by the binding of antigen peptide–major histocompatibility complexes (MHCs) to T cell receptors,55 which subsequently participate in B-lymphocyte (B cell) activation.56 The activation of adaptive immunity and the production of cytokines and antibodies lead to pathogen elimination and/or tissue damage.56

2.2.3 Chronic myopathy phase

When there is persistent overreaction of the immune response, myocardial tissue undergoes matrix remodeling and fibrosis, which can lead to heart failure and secondary DCM. Several susceptible strains of mice4–7 and genetically susceptible individuals8–11 progress to this phase. It has been reported that more males develop DCM and have a worse outcome after acute myocarditis than females.57,58 Although the presence of enteroviral genomic RNA has been examined in EMB tissues obtained from patients with DCM,59–61 non-DCM populations have parvovirus B19 DNA detected in heart tissues.62 Not only viruses, but also overreactive immune responses are necessities of the progress to DCM.58,63 Matrix remodeling and fibrosis are mainly triggered by the dysregulation of collagen production and degradation. In male BALB/c mice with CVB3 myocarditis, the imbalance in the matrix-degrading system, which refers to increased expression of matrix metalloproteinases (MMPs) and decreased expression of tissue inhibitors of MMPs, causes a pathologic collagen turnover that compromises left ventricular function and causes the structural integrity of the heart to deteriorate.64–67 Some proinflammatory cytokines and enzymes have been reported to induce this imbalance.58,67–69

2.3 Sex differences in myocarditis

According to previous reports,70–74 the incidence of myocarditis and the cardiac expression of proteins related to apoptosis and fibrosis are higher in men, which reveals that there may be sex differences in the pathogenesis of myocarditis. Male rodents, such as BALB/c mice and Lewis rats, show more acute and chronic inflammation in CVB3-induced myocarditis52,75 and EAM,53,76 and these differences are likely due to sex hormones.72 During CVB3 myocarditis, testosterone contributes to the predominant T helper 1 (Th1) immune response in the myocardium and more viral replication, but estradiol tends to develop the Th2 immune response and promotes the generation of regulatory T (Treg) cells and anti-inflammatory M2a macrophages.77–82 Greater testosterone concentrations are linked to a number of cardiac pathologies,58,74 which encourage higher collagen synthesis, fibrosis development, and extracellular matrix (ECM) remodeling.

In terms of the immune response, 17β-estradiol (E2) enhances the expression of TLR4 in rodent peritoneal macrophages83 and upregulates TLR7 and TLR9 signaling in human monocytes exposed to granulocyte-macrophage colony-stimulating factor (GM-CSF).84 In contrast, testosterone tends to deregulate the expression of TLR4 in rodent macrophages.85 E2 exhibits bimodal effects on human monocytes/macrophages, boosting the production of proinflammatory cytokines (including IL-1, IL-6, and tumor necrosis factor α [TNF-α]) at low concentrations, while decreasing the production of these cytokines at high concentrations.86 Increased levels of inflammatory cytokines, such as IL-1, IL-2, and TNF-α, have been observed in men with androgen deficiencies.87–89

3. Immune cells in myocarditis

3.1 Neutrophils

Neutrophils are the specialized phagocytic cells of innate immunity and are the first line of defense against invading pathogens. Neutrophils perform antipathogen functions, including phagocytosis, degranulation, production of neutrophil extracellular traps (NETs), and immunomodulation.90 Neutrophils express a variety of surface receptors, including PRRs, cytokine receptors (TNF-α, IL-1, and IL-6 receptors), and chemokine receptors (CXCR2 [C-X-C motif chemokine receptor 2]).91 In autopsies of patients with severe COVID-19, researchers found extensive neutrophil infiltration in the myocardium of patients who died of active myocarditis.92 Advanced depletion of neutrophils with anti-Ly6G antibodies in male C57BL/6 mice of CVB3-induced myocarditis led to a reduction in myocardial tissue inflammation and viral load.93 These studies suggest a possible role for neutrophils in the pathogenesis of myocarditis during the immune response phase.

In myocarditis, neutrophils engulf invading pathogens, forming phagocytic vesicles. The phagocytic vesicles come into contact with intracytoplasmic toxic particles, and the pathogens within the vesicles can be eliminated. Reactive oxygen species generation mediated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation can also enter the phagocytic vesicles containing pathogens and participate in their elimination. Neutrophil infiltration and microabscesses were found in the myocardium of a patient with nonrheumatic streptococcal fulminant myocarditis, in which neutrophils that had phagocytosed streptococci were observed.94 Phagocytosis of pathogens or cellular debris activates neutrophils, which in turn release proinflammatory cytokines and chemokines to amplify inflammation and recruit more proinflammatory cells.93 In male C57BL/6 mice with CVB3-induced myocarditis, neutrophils recognize CVB3 mainly through intracellular TLR8,93 which triggers certain signaling pathways, such as the NF-κB pathway, further mediating the release of proinflammatory factors, such as IL-6, IL-1β, TNF-α, and IL-12, as well as chemokines. After phagocytosis of CVB3, CD11b expression in neutrophils is upregulated, enhancing their adhesion to the ECM and contributing to their residence at the site of inflammation. Neutrophils infected with CVB3 have an extended lifespan, but CVB3 does not replicate massively in neutrophils; thus, whether neutrophils can act as vectors for viral transmission needs further investigation.93 The phagocytosis of neutrophils plays both a protective role in the elimination of pathogens and a pathogenic role through the production of proinflammatory cytokines, chemokines, and adhesion factors in response to pathogens or dead cells.12

The cytoplasm of neutrophils contains a variety of granules that can be classified as primary, secondary, and tertiary granules containing myeloperoxidase and elastase, which are involved in killing pathogens and digestion, and containing lactoferrin and gelatinase proteins that degrade the ECM.95 The excreted granules also contain preformed cytokines.95 The degranulation process is dependent on adhesion signals and receptor–ligand binding signals, such as the binding of formylated peptides to formylated peptide receptor 1.91,96 The dual signals are activated to avoid random degranulation of neutrophils,95 while the gradual release of granules contributes to the control of inflammation.97 CVB3-infected neutrophils are stimulated by TNF-α to release myeloperoxidase and trigger NET formation.93 Neutrophil granules are essentially highly cytotoxic, so when the contents of these granules leak or are excreted excessively, they can damage the normal tissues that surround the inflammation and drive neutrophil recruitment.95 However, the released particles are not always detrimental. It has been suggested that human CAR may be a target for neutrophil elastase shedding and that the elastase released by neutrophils may protect cardiomyocytes from viral infection by cleaving CAR from the surface98 during the acute infection phase.

In recent years, the role of NETs, which are extracellular reticulated complexes that consist of DNA, histones, and neutrophil granule proteins, has been increasingly recognized.99,100 This unique cell death program in which neutrophils abandon their integrity and release DNA is called NETosis.101 Not only do NETs help to fix and remove pathogens, but also they cause direct or indirect damage to surrounding tissues due to the combination of various proteases and cytotoxic proteins.102 NETs are found in myocardial tissues from patients with myocarditis, as well as in male mice with CVB3-induced myocarditis and EAM, and they exert their pathogenic effects mainly in the acute phase.103,104 Inhibition of NET formation during the acute phase helps to reduce the inflammatory response in the heart.103,104 Cytokine intermediate factor (MK) can induce neutrophil infiltration and NET formation, a process that may be achieved through interaction with low-density lipoprotein receptor–related protein 1.104 IL-37 attenuates CVB3-induced myocardial injury by inhibiting NET formation.105 In the lung, NETs induce activation of lung fibroblasts to the myofibroblast phenotype and exhibit increased production of connective tissue growth factor and collagen.106 In male BALB/c mice with EAM, NET formation can be reduced by inhibiting MK,104 which also reduces fibrosis and improves cardiac contractile function. Thus, NET formation in the myocardium may contribute to fibrosis. Moreover, NETs activate CD4+ T cells by lowering their activation threshold and promoting the adaptive immune response phase.107

Lipocalin-2 (LCN2), also known as neutrophil gelatinase-B-associated lipid transport protein, is involved in the antimicrobial response, iron transport, apoptosis regulation, and resistance to oxidative stress.108,109 Recently, Sun's team110 showed that LCN2 exacerbates lupus nephritis by promoting Th1 cell differentiation. Previously, Ding et al.111 found that neutrophils in the hearts of patients with myocarditis and male Lewis rats with EAM express LCN2, while its role in myocarditis has not been revealed.

Neutrophils are also involved in the regulation of other immune cells. On the one hand, neutrophils recruit themselves and monocytes by secreting several proinflammatory mediators mentioned previously, which promotes the formation of inflammatory macrophages and the differentiation of Th1 and Th17 cells.103 On the other hand, neutrophils contribute to the maintenance of the cardiac CD8+ T cell inflammatory response.112 Moreover, neutrophils produce the cytokines B cell activating factor and proliferation-inducing ligand, both of which are required for B cell survival and activation.113 Further studies are needed to determine whether it is necessary to modulate B cell function in myocarditis.

3.2 Eosinophils

Eosinophils are a special type of leukocyte that are usually considered as cytotoxic effector cells because they contain a variety of cytotoxic granules. They contain major basic protein (MBP), eosinophil cationic granule protein (ECP), and eosinophil peroxidase, which can be toxic to pathogens and cells, either directly or by promoting the production of reactive oxygen species. Eosinophils also contain cytokines (IL-2, IL-4, IL-5, IL-6, IL-13, and TNF-α), chemokines (C-C motif chemokine ligand 5 [CCL5] and CCL11), transforming growth factor (TGF), and lipid mediators, which are selectively released in response to different stimuli.114–116 Recently, it has been shown that eosinophils are involved in the regulation of immunity and homeostasis.117

Myocarditis with predominant eosinophil infiltration on EMB is known as eosinophilic myocarditis. According to a previous summary, eosinophilic myocarditis is mainly associated with hypersensitivity reactions, infections (parasites, fungi), malignancies, drugs, hypereosinophilic syndrome, eosinophilic granulomatous polyangiitis, systemic lupus erythematosus, and Churg-Strauss syndrome, and is usually treated with strong immunosuppressive agents.118–120 Recently, in autopsies of deceased individuals who developed fulminant myocarditis after the COVID-19 mRNA vaccination, cardiac tissue demonstrated eosinophil infiltration.121

The pathogenesis of eosinophilic myocarditis is commonly described as follows. First, eosinophils infiltrate into the heart and recognize invading parasites, fungi, certain bacteria, and damage-associated molecular patterns signals in vivo through PRRs during the immune response phase. Then, eosinophils enter an activated state, achieve degranulation, and kill pathogens, mainly through piecemeal degranulation, exocytosis, and cytolysis, causing myocardial injury and dysfunction.122 This can be confirmed by the detection of ECP, MBP, and eosinophil peroxidase deposition on EMB.123

Subsequently, the thrombogenic phase occurs, in which the formation of the thrombomodulin-thrombin complex is blocked due to the binding of ECP to the anionic site on thrombomodulin, resulting in an elevated level of circulating thrombin and putting the body in a hypercoagulable state with the release of tissue factor stored in specific granules by eosinophils. In addition, MCP activates platelets. These processes contribute to the formation of cardiac thrombosis.124

The final stage is dominated by fibrosis. In male BALB/c mice with EAM, eosinophil depletion prevents the transition from myocarditis to DCM, while high eosinophil counts promote the development of more severe DCM.125 This may be due to endothelial cell proliferation stimulated by ECP and MBP, fibroblast activation, and proliferation promoted by secreted TGF-β and IL-1, and the unknown function of eosinophil-derived IL-4.116,118,125

IL-5 is a key biological factor (i.e. involved in the activation, survival, and migration of eosinophils in tissues), and it is the most potent activator of eosinophils, produced by both Th2 cells and type 2 ILCs.126 Researchers have found that hypereosinophilic mice with IL-5 transgene expression have spontaneous eosinophilic myocarditis, and that more than 60% of cardiac infiltrating cells are eosinophils in IL-5 transgene male BALB/c mice with EAM.115,125 IL-33, which is a cytokine from the IL-1β family,127 significantly induces eosinophilic myocarditis during CVB3 infection.128 The recruitment of eosinophils is influenced by Th2 cells, macrophages, fibroblasts, and eosinophils themselves, by producing chemokine ligands in the myocardium.116,129

Similar to neutrophils, eosinophils produce extracellular traps called eosinophil extracellular traps (EETs), and the form of eosinophil death is called EETosis. The nuclear and plasma membranes of eosinophils rupture under the mediation of reactive oxygen species generated by NADPH oxidase, releasing nuclear or mitochondrial DNA and intact eosinophil granules, mediating the killing of pathogens and tissue damage.130,131 EETs are more stable than NETs.122 In eosinophilic airway inflammation, activated platelets and their products induce EET formation, which enhances their inflammatory effects.132 Therefore, we hypothesized that MCP activates platelets, which induces EET formation and MCP release, and it may enter a vicious MCP–platelet–EET–MCP cycle.

In addition, ECP and MBP activate human cardiac mast cells. Then, mast cells release histamine, trypsin, and prostaglandin D2.133 Whether this process plays a definite role in the pathogenesis of myocarditis remains to be explored.

3.3 Mast cells

Mast cells are immunomodulatory cells that play an important role in both innate and adaptive immunity. Depending on their contents, mast cells can be divided into 3 subpopulations in humans: tryptase-positive mast cells, chymase-positive mast cells, and tryptase/chymase-positive mast cells.134 Mast cells are mainly found in contact with the environment and are thought to be closely associated with allergic reactions.135

A previous study showed that in encephalomyocarditis virus myocarditis, mast cell–deficient male mice had a higher survival rate, significantly less myocardial necrosis, and less inflammatory infiltration than control mice.136 This suggests that mast cells play a pathogenic role. In 2022, Luo et al.137 found that in male C57BL/6 mice with CVB3 myocarditis, fibroblasts triggered mast cell degranulation and CCL2 and TNF-α release by producing early stem cell factor, and mast cell–derived TNF-α in turn stimulated fibroblasts to increase CCL2, α-smooth muscle actin, collagen, and TGF-β, thereby exacerbating cardiac fibrosis. In addition, mast cell–derived CCL2 recruits monocytes, which differentiate into inflammatory macrophages, promoting inflammation. The stem cell factor/mast cell/CCL2/monocyte/macrophage axis was first described in CVB3-induced myocarditis137; however, mast cells are not simple. The subpopulations and enzymatic profiles of mast cells are related to the tissue.138 Under different stimuli in vivo or in vitro, mast cells produce a variety of ILs (IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13, among others), interferon-γ (IFN-γ), TNF, TGF-β, vascular endothelial growth factor (VEGF), and chemokines.139 All of these molecules exert proinflammatory, anti-inflammatory, and regulatory fibrosis, as well as angiogenic effects.139 Mast cells, like macrophages, have considerable plasticity and may play unique roles in different local conditions through different mechanisms.138

A detailed summary of cardiac mast cells (CMCs) was previously presented by Varricchi et al.140 Mast cells are found in both human and animal hearts, mainly between the myocardium, in the epicardium, and near blood vessels.141 Varricchi et al.140 concluded that activated human CMCs can release preformed histamine, trypsin, and chymotrypsin. Histamine can lower blood pressure. In addition, Stasiak et al.142 found that inhibition of histamine receptor 4 (H4R) in Lewis male rats with EAM inhibited cardiac remodeling, improved cardiac systolic function, and increased survival. However, blocking histamine receptor 1 inhibited cardiac dilation, but it did not prolong life. Chymotrypsin has different effects in different models of heart disease. Chymotrypsin activates the renin–angiotensin system and promotes the release of local angiotensin II and norepinephrine, which are closely associated with arrhythmia.140

Varricchi et al.140 concluded that activated CMCs also synthesize secreted renin, which is involved in RAS activation; release cysteine leukotrienes to promote inflammation and proangiogenesis; possibly secrete PGD2 to reduce allergic reactions and fibrosis; synthesize VEGF-A to promote angiogenesis and VEGF-C to promote lymphatic growth; and reduce tissue edema and fibrosis.

Human mast cells express PD-L1 and PD-L2. Varricchi et al.140 postulated that ICIs may cause myocardial damage by killing peripheral CMCs, which releases cytotoxic factors and recruits inflammatory cells.

Mast cells actively form mast cell extracellular traps in response to specific stimuli that surround and clear pathogens, possibly limiting the inflammatory response.143 However, it is unclear whether CMCs form mast cell extracellular traps to function in myocarditis.

Varricchi et al.140 analyzed previous studies on mast cells and cardiac fibrosis. They summarized the possible reasons for the conflicting results among these studies, including the different phenotypes between mouse CMCs and human CMCs, the existence of models that eliminate mast cells while causing a lack of other cell types or incomplete elimination of mast cells, and the fact that the mast cell degranulation inhibitors used for the experiments were not specific. In 2021, Bruno et al.144 found that during CVB3-induced myocarditis, feeding female BALB/c mice in plastic cages or with water containing bisphenol A increased the number of degranulated mast cells in the myocardium and pericardium, and an increase in myocardial fibrosis was observed. However, these manifestations were not present in male BALB/c mice under the same conditions.144 We speculate that the effect of mast cells on myocardial fibrosis is affected by sex; however, this requires further experimental investigation.

3.4 Innate lymphoid cells

ILCs can be divided into natural killer (NK) cells, ILC1, ILC2, and ILC3. NK cells are the only cytotoxic cell ones that originate from the bone marrow and migrate continuously through the circulation to peripheral tissues.145 Human NK cells can be divided into 2 subpopulations based on the differential expression of CD56 and CD16.146 CD56loCD16hi NK cells account for approximately 90% of human peripheral blood NK cells and exhibit classic cytotoxic effects. The remaining 10% are CD56hiCD16−/lo NK cells, which are involved in the secretion of cytokines for immune regulation.147,148

NK cells are cytotoxic and directly lyse pathogen-infected cells, autoreactive T cells, overactivated inflammatory cells, and tumor cells, and the choice of target is related to abnormalities in MHC class I molecules in target cells.149 Activated NK cells express the Fas ligand or TNF-related apoptosis-inducing ligand, and binding of these ligands to the corresponding receptors induces target cell apoptosis.150 Moreover, NK cells release cytotoxic proteins (perforin, granzyme, etc.).151,152 Perforins undergo oligomerization in the target cell membrane and form transmembrane pores with the transmembrane structural domain–MACPF structural domain. Granzymes and proapoptotic serine proteases are released through these pores and initiate target cell apoptosis, leading to cell death.153 This suggests a role for NK cells in pathogen clearance, but uncontrolled perforin expression can also lead to autoimmune diseases, including cerebral myocarditis.153 Pores induced by perforins on the surface of virus-infected cardiomyocytes have been found in both patients with myocarditis and animal models of viral myocarditis,154 and attacking virus-infected cardiomyocytes also inevitably leads to the release of autoantigens and subsequent induction of the autoimmune response.14

In addition to cytotoxicity, activated NK cells secrete a variety of cytokines (IFN-α, IFN-β, IFN-γ, TNF-α, and ILs, among others)155 that function in antiviral responses, induction of the inflammatory response, and activation or suppression of other immune cells.156 Estrogen downregulates the expression of Th1 response–specific T box transcription factor, a key transcription factor for IFN-γ production, in CVB3-stimulated NK cells, thereby reducing IFN-γ-secreting NK cell infiltration and myocardial injury in myocardial tissues.157 Loebel et al.158 found that in viral myocarditis, the forkhead transcription factor negatively regulates IFN-γ production and cytotoxic degranulation in NK cells, which is detrimental to the control of early viral infection, but it has the potential to play a protective role in the subsequent chronic inflammatory process.

According to previous studies, NK cells control the number of eosinophils to play a protective role in humans with myocarditis, EAM models, and cellular assays in vitro.159–161 This protective effect appeared to be dependent on the presence of eosinophils, as depletion of NK cells in eosinophil-deficient mice did not increase the severity of myocarditis.159 The mechanism may be related to the increase in reactive oxygen species and apoptosis in eosinophils induced by NK cells.161 Moreover, NK cells can reduce cardiac eosinophil recruitment by producing IFN-γ, which downregulates the release of Th2-related cytokines in ILC2.160,162

The finding that NK cells exert a protective effect by inhibiting fibrosis has been demonstrated mainly in liver disease,163 mainly by directly lysing activated fibroblasts or promoting the inactivation of hepatic stellate cells, which reduces collagen production and deposition.164,165 However, the functions of the different human NK cell subpopulations in different tissues are not identical.146 In Ong et al.'s study,159 NK cells reduced fibrosis during myocarditis, but when NK cells were cocultured with activated primary cardiac fibroblasts in vitro, cardiac fibroblasts did not show cytolysis similar to that of liver fibroblasts. Further study159 in male BALB/c mice with EAM revealed that NK cells, by directly limiting collagen formation in cardiac fibroblasts, may protect against fibrosis and DCM development by preventing the accumulation of certain inflammatory cells in the heart and negatively regulating profibrotic factors. However, the exact mechanism has not been well revealed.

In addition to NK cells, the remaining noncytotoxic ILCs include ILC1, ILC2, and ILC3. ILC1 is stimulated by IL-12 to express IFN-γ and TNF-α, which assist in the antiviral response in a noncytotoxic manner and promote the Th1 response.166,167 ILC2 participates in the Th2 response mainly by secreting IL-5 and IL-13, which contribute to eosinophil maturation and recruitment.168 ILC3 responds to IL-23 and IL-1β and promotes the Th17 response by producing IL-17A and GM-CSF, and is associated with antipathogen and autoimmune diseases.169 Under normal conditions, the major cardiac ILC population is in an undifferentiated state with the potential to differentiate toward ILC2. Studying samples from myocarditis patients and BALB/c mice with EAM has revealed that cardiac ILCs differentiate into conventional ILC2, but they do not differentiate into ILC1 and ILC3 during myocarditis. Their differentiation is positively associated with increased IL-33 production by cardiac fibroblasts. Unlike NK cells, circulating noncytotoxic ILCs are unable to enter the heart during adulthood. Thus, cardiac noncytotoxic ILCs are strictly cardiac-resident cells.170 However, the findings may need further verification because of the limitations of cardiac tissue sampling. Subsequent tissue-specific single-cell spatial transcriptome sequencing may provide a better understanding.

3.5 Monocytes

Monocytes are circulating leukocytes that are released into the circulatory system from the bone marrow or extramedullary hematopoietic tissues (spleen). They are then transported through the circulatory system to peripheral tissues. During inflammation, circulating monocytes are recruited by monocyte chemoattractant protein 1, leave the bloodstream to migrate to inflamed tissues, kill a variety of pathogens, promote or inhibit inflammation by secreting mediators (e.g. nitric oxide, TNF, ILs, reactive nitrogen intermediates, and reactive oxygen intermediates, and phagocytic lysosomal enzymes, among others), and differentiate into macrophages or DCs that remain in the tissues and continue to function.171

In general, monocytes are divided into 3 major categories, the first being classical or inflammatory monocytes,172 which in mice are characterized by high expression of Ly6C and C-C chemokine receptor 2 (CCR2), and low expression of C-X3-C chemokine receptor 1, named Ly6chi monocytes. In humans, they are named CD14++CD16 monocytes based on high expression of CD14 and lack of CD16.173 Although their markers vary by species, they also exhibit higher peroxidase activity and can produce more chemokine receptors, suggesting that they are associated with phagocytosis to destroy pathogens and migrate more easily to sites of inflammation or injury. It has been shown that in human myocarditis and male BALB/c mice with EAM, inflammatory monocytes are the main precursors of cardiac monocyte-derived macrophages.174 The second category, nonclassical monocytes, whose function differs from that of classical monocytes in that they play a patrolling role along the vessel wall, promote healing and respond to viral infection. Nonclassical monocytes include Ly6clo monocytes in mice and CD14 + CD16++ monocytes in humans.175 For the question of whether nonclassical monocytes can be transformed into macrophages in tissues, Hou et al.174 demonstrated that in male BALB/c mice with EAM and patients with myocarditis, nonclassical monocytes have a limited ability to convert into macrophages, possibly due to IL-17A signaling through cardiac fibroblasts to inhibit conversion. In the myocardium of EAM mice lacking IL-17A, approximately 6% of nonclassical monocytes converted into macrophages and exhibited different molecular features that may have a unique role compared with inflammatory monocyte–derived macrophages in the pathogenesis of myocarditis.174 The third category is intermediate monocytes, including Ly6cint monocytes in mice and CD14++CD16+ monocytes in humans, which are intermediate between classical and nonclassical monocytes. They display inflammatory properties and share low peroxidative activity with nonclassical monocytes, and they also secrete TNF and IL-1β.176

Unlike other innate immune cells, monocytes can differentiate into macrophages or DCs in the periphery and continue to function in the proinflammatory and anti-inflammatory responses, as well as in antigen presentation,177 as detailed subsequently. In addition, monocytes are directly involved in regulating T cell differentiation, including differentiation of Th1, Th2, Th17, and Treg cells, among others, and they promote or inhibit T cell differentiation through antigen presentation, secretion of cytokines or killing mediators, regulation of cysteine metabolism, and expression of multifunctional transmembrane receptors.178,179 By inhibiting myocardial inflammatory monocyte recruitment in A/J mice with EAM using silencing RNA targeting CCR2, myocardial inflammation and fibrosis were attenuated.180 In male and female C57/BL6 mice with CVB3-induced myocarditis, myocardial monocyte/macrophage infiltration was reduced, and the Th1 response was increased in the absence of stabilin-1 (STAB1). The authors considered that STAB1 deletion leads to a block in fibronectin-mediated recruitment of STAB1+ monocytes, which play a role in suppressing the T cell response.181 We hypothesized that monocytes themselves can exert opposite regulatory effects through different mechanisms, and that the final outcome depends on the type of disease and the strength of different triggers or maintenance factors.

We realize that the definition of monocytes is by no means monotonous. When monocytes enter tissues, they exhibit changes in their transcriptional profile, especially when they enter inflammatory tissues and begin to synthesize some proinflammatory or anti-inflammatory factors. To a large extent, the diversity of monocytes depends on the local microenvironment.171,177 Using scRNA-seq technology, researchers have shown that in patients who develop myocarditis after BNT162b2 (Pfizer COVID-19 vaccine) vaccination, the transcriptome profile of classical monocytes showed upregulation of genes related to fatty acid metabolism and downregulation of transcription factor AP-1.182 These findings revealed the importance of classical monocytes in the pathogenesis of myocarditis after BNT162b2 vaccination, as well as the role of vaccination on monocytes leading to myocarditis. In addition, Jimenez et al.183 concluded that at least 7 types of monocytes with different morphological and transcriptional characteristics were identified in the infarcted myocardium and may play their own unique roles. Therefore, further studies are needed to demonstrate whether the inflamed myocardium may also have a variety of monocytes with different characteristics that play as yet unspecified roles.

3.6 Macrophages

Macrophages were first recognized as phagocytes, which represents their most basic role of pathogen removal. In CVB3-induced myocarditis, macrophage-depleted male C3H/HeJ mice had higher viral loads, suggesting that reduced macrophages may lead to reduced pathogen clearance.184 However, macrophage-depleted mice exhibited milder cardiac inflammation and fibrosis. Thus, the function of macrophages is never simply limited to phagocytosis. With further studies, macrophages began to be recognized as immune cells.

Traditionally, macrophages are divided into 2 types according to their activating factors. The first type is classical (M1) macrophages activated by TLR ligands or IFN-γ, which mainly mediate oxidative stress, inflammasome formation, and proinflammatory factor secretion (TNF-α, IL-1β, IL-6, and IL-12, among others) to produce costimulating factors and chemokines that promote inflammatory cell infiltration and proliferation. They subsequently cause cardiac injury. The other type are nonclassical (M2) macrophages, which form under the stimulation of anti-inflammatory signals, such as IL-4. M2 macrophages secrete anti-inflammatory cytokines, chemokines, and growth factors, mainly playing an anti-inflammatory role.185 In BALB/c male mice with CVB3-induced myocarditis, the myocardial macrophage phenotype changes from the classical M1 phenotype to the nonclassical M2 phenotype at approximately days 7 to 10 when the disease enters a phase characterized by fibrotic repair186 and conditional knockdown of CAPN4 could reduce M1 macrophage transformation, and moderately increase M2 transformation via inhibiting the C/EBP homologous protein-STAT1/STAT3 signaling pathway and could ameliorate myocardial inflammation and injury.187 Besides, macrophage-specific CAR depletion and IL-13 knockout have been reported to increase M1 transformation and induce T cell activation in CVB3-infected mice188 or EAM mice,189 along with enhanced heart inflammation and injury. In contrary, treating with CD73 inhibitor prevented the premature transformation from M1 to M2 and decreased the parasite burden in heart during the acute phase in BALB/c mice infected with Trypanosoma cruzi, leading to a good outcome, because the persistence of parasites is a necessary condition for the development of Chagas cardiomyopathy.190 It seems that dysregulated transformation may be the key point in myocarditis. Macrophages also secrete several types of MMP.185 For example, Amoah et al.191 concluded that the release of MMPs not only hydrolyzes ECM proteins and non-ECM proteins to amplify the inflammatory response, but also plays a protective role in viral myocarditis by cleaving chemokines and reducing inflammatory cell infiltration.

In addition to traditional M1/M2 macrophage polarization, the understanding of cardiac macrophages has been further improved. There are native tissue-resident macrophages in the heart, which are derived from embryonic (yolk sac or fetal liver) precursor cells. Tissue-resident macrophages in the heart lack CCR2 on their surface and rely on replication in the tissue for renewal.192 Circulating monocyte–derived macrophages are also present in the heart and are distinguished from the former type by their expression of CCR2. The roles of these 2 types of macrophages are different. It is currently believed that cardiac-resident macrophages play an important role in tissue developmental remodeling and maintenance of homeostasis by removing abnormal cells or dysregulated mitochondria,193 secreting anti-inflammatory factors, regulating lymphangiogenesis, and avoiding the excessive inflammatory response.183 Circulating monocyte–derived macrophages are thought to play a potent inflammatory role. Upon activation, they generate an inflammatory cascade by secreting proinflammatory factors and chemokines, and further recruiting neutrophils and monocytes.137,183 Their abundance may be associated with a poor prognosis in cardiac diseases.183 Researchers found that reduction of CCR2+ macrophage infiltration in the heart by CCR2-siRNA attenuated myocardial inflammation and fibrosis in EAM mice.180 However, impeding CCR2-macrophage development and aggregation by knocking down CX3CR1 exacerbated cardiac inflammation and fibrosis in CVB3-infected mice.194

It appears that macrophages respond differently to different signals, and the macrophage phenotype can be modulated by external signals. Mosser et al.195 proposed a new concept in which macrophages act as cellular transducers, receiving different input signals from the environment and translating them into a corresponding response. Hulsmans et al.196 revealed that cardiac macrophages expressing connexin 43 facilitated electrical conduction in the distal atrioventricular node by altering the membrane potential, and that connexin 43 macrophage deficiency delays atrioventricular conduction. Whether connexin 43 macrophages are associated with the development of atrioventricular block in myocarditis needs to be clarified. Matheis et al.197 showed that intestinal myocardial macrophages upregulate neuroprotective programs via β2-adrenergic receptor signaling in response to intestinal infection and complete neuronal protection via the arginase 1–polyamine axis. Whether similarly acting macrophages reside in the heart to exert neuronal protection against myocarditis needs to be explored in future studies.

3.7 Dendritic cells

DCs, specialized antigen-presenting cells with a complex subpopulation, are considered to be the body's outpost cells, linking innate and adaptive immunity.198,199 According to the current understanding of DCs, they are classified into 3 categories. Conventional DCs (cDCs) recognize signals from intracellular and extracellular antigens in an inflammatory environment through PRRs, upregulate costimulatory molecules, present antigens to naïve T cells and activate them200 in lymph nodes. In the steady state, cDCs also present innocuous antigens and mediate T cell incompetence to complete the body's immune tolerance to autoantigens, inhaled antigens, and food antigens.201,202 cDCs include type 1 cDCs, characterized by their strong ability to activate cytotoxic CD8+ cells through cross-antigen presentation, and type 2 cDCs, which express a wide variety of TLRs and NLRs in response to different danger signals, and they may be more inclined to present MHC-II-antigenic peptides to CD4+ T cells.203 Second, plasmacytoid DCs (pDCs) are important for host antiviral immune response and autoimmunity as efficient producers of IFN-α following stimulation of the nucleic acid-sensitive receptors TLR7 and TLR9,203 not as potent in antigen presentation as cDCs. Finally, monocyte-derived DCs are a DC subpopulation that differentiate from tissue-infiltrating monocytes in response to inflammatory stimuli.203 Monocyte-derived/myeloid DCs are mainly considered to be a subpopulation of inflammatory DCs that can elicit local Th1 or Th2 responses.201,202

Following the viral infection in the myocardium, the number of myocardial DCs increased rapidly relying on local proliferation and recruitment from circulation.204,205 During subclinical myocarditis in mice infected with encephalomyocarditis variant-D virus, CD103+ resident type 1 cDCs facilitated the development and infiltration of encephalomyocarditis variant-D virus–specific CD8+ T cells, alleviated cardiac damage, and prevented the progression to heart failure.206 Abnormalities in the maturation/activation and MHC class I antigen processing of DCs were proved to be responsible for persistent chronic CVB3-induced myocarditis in A.BY/SnJ mice caused by a failure to remove the virus.207 The activation of antiviral CD8+ T cells by DCs in the early stage is conducive to viral clearance and prevent cardiac remodeling from sustained immune activation by hidden viruses. In addition, it has been reported that the number of circulating myeloid DCs and pDCs increased in the patients with acute myocarditis, and increased myeloid DCs exhibited a proinflammatory phenotype with higher costimulatory molecules and lower inhibitory receptor expression linked to the worser clinical outcome.208 Besides, increased pDCs could be the source of peripheral cytokine storm in myocarditis. In CVB3-infected mice, myeloid DCs accumulated following the increased level of CCL4 and CCL19 in the heart.209 Overaccumulation of DCs might mean loss of the body's immune tolerance to myocardial tissue. Activated by IL-1210 and GM-CSF211 in the proinflammatory environment, DCs, especially type 2 cDCs,212 have the potential to take up self-antigens released from damaged cardiomyocytes or newly generated ECM, activate self-reactive CD4+ T cells, leading to cardiac injury.202,213 TN-C, a nonstructural ECM glycoprotein (i.e. strongly expressed during acute inflammation in mice with EAM) can activate DCs to generate pathogenic autoreactive T cells that attack cardiac tissue.214

The body responds to the inflammatory cascade by generating a corresponding anti-inflammatory response to maintain homeostasis.215 In an anti-inflammatory environment, DCs show phenotypic changes. IL-10–induced human regulatory DCs express a series of inhibitory receptors (PD-L1 and PD-L2, among others) that suppress harmful T cell responses.216 PD-L2–deficient DCs enhance the proliferation of self-reactive CD4+ T cells in the presence of T cell receptors and CD28 transduction signals,217 which exacerbated EAM. This also indirectly suggests that reprograms of DCs induced by IL-10 express PD-L2 and prevent self-reactive CD4+ T cell expansion which alleviates the severity of myocarditis, indicating the multiple roles of DCs due to different environments.

Furthermore, with the emergence of immune-targeted therapies, investigators have recognized a new DC type, namely tolerogenic DCs (tDCs). The mechanism of tDCs formation in vivo is currently unclear, but it is associated with stimulation of immunosuppressive factors, including anti-inflammatory factors, such as IL-10 and TGF-β, as well as activation of vitamin A receptor ligands.218,219 In addition, knockdown of NEAT1 induced the formation of tDCs by inhibiting activation of the NLRP3 inflammasome.220 Several studies221,222 have shown that injecting IL-10 gene–modified tDCs into EAM mice could regulate Th1/Th2 unbalance and cardiac inflammation has been alleviated. Antigen-specific tDCs generated by exposing immature DCs to cardiac myosin with TNF-α223 or protosappanin A224 were found to significantly reduce myocardial inflammation in treated EAM mice by enhancing the induction of myosin-specific Treg cells and anti-inflammatory cytokine secretion. These tDCs had switched phenotype with lower levels of surface DC–related markers and higher levels of indoleamine 2,3-dioxygenase compared with mature DCs, or reprogrammed glycolytic phenotype. Therefore, tDCs may be a novel therapy direction in myocarditis and exert immunosuppressive effects by secreting anti-inflammatory factors, suppressing effector T cells, regulating the Th1/Th2 response, and inducing Treg cell activation.

In summary, the various roles of DCs are closely related to their phenotypes and surroundings. We recognize that their phenotypes also go far beyond the species identified. Villar et al.225 combined single-cell sequencing to provide new insights into the classification of human DC subpopulations, suggesting that single-cell transcriptome sequencing could theoretically provide specific and unique molecules for the identification of each DCs subpopulation. This is expected to make it possible to isolate specific DCs for functional characterization beyond transcriptome profiling in healthy and disease states. This also provides some ideas for subsequent studies on myocarditis.

3.8 T lymphocytes

In the bone marrow, pluripotent hematopoietic stem cells differentiate into lymph-like progenitor cells, which follow the blood into the thymus and undergo positive and negative selection before differentiating into mature T cells. Depending on the expression of CD4 and CD8, T cells can be divided into CD4+ T cells and CD8+ T cells. CD4+ naïve T cells (Th0 cells) migrate to peripheral immune organs and are activated into Th cells upon stimulation by antigenic peptide–MHC class II complexes and co-stimulatory signals. They can differentiate into Th cell subsets with different functions (Th1, Th2, Th9, Th17, Th22, follicular helper T, and Treg cells) under different cytokine environments. In contrast, CD8+ T cells are activated into cytotoxic T cells or CD8+ Treg cells upon stimulation by antigenic peptide–MHC class I complexes. The roles of T cells and their secreted cytokines in myocarditis have been summarized in more detail in previous reviews.15,191,226 Here, we make a brief summary and add details on the regulation or crosstalk. We also briefly summarize other cell subsets that have not yet been mentioned to provide a comprehensive understanding.

3.8.1 CD4+ T cell subsets in myocarditis

Th1 cells could be a double-edged sword in the pathogenesis of myocarditis by secreting IL-12, IFN-γ, IL-1β, and TNF-α. IL-12 has been reported to be proinflammatory, and the knockout of IL-12R showed the resistance to EAM, while exogenous usage of IL-12 leads to aggravated heart inflammation. IFN-γ shows strong abilities of antivirus, activating immune response, and balance the excessive inflammation.227 In the acute phase, knockout of IFN-γ receptor in T cell receptor transgenic mice with EAM,228 depleting IFN-γ in viral myocarditis,229 or lowering IFN-γ production in T. cruzi–related myocarditis230 reduced myocarditis. However, IFN-γ–deficient mice showed more severe chronic myocarditis after CVB3 infection50 or high levels of T. cruzi hidden in heart and reactivation,231 leading to bad prognosis. Besides, knockout of IFN-γ receptor in cardiac α-myosin heavy chain–immunized EAM mice leaded to enhanced CD4+ T cell response and persistent myocarditis.227

Th2 cells secrete IL-4, IL-10 and IL-13, and also exhibit reverse function in myocarditis. Th2-biased inflammatory phenotype was observed in vitamin D receptor knockout mice with spontaneous myocarditis232 and A/J mice with EAM.233 While anti-IL-4 treatment could reduce inflammation.233 Besides, the amount of Th2 cells and related cytokines were higher in the heart from patients with myocarditis than them with DCM.234 In contrary, hepatocyte growth factor has been reported to promote IL-4 and IL-10 production by Th2 cells, inhibit cardiomyocyte apoptosis, and attenuate the severity in Lewis rats with EAM.235

Th17 cells are novel proinflammatory subset characterized by the production of IL-17. In the early phase of CVB3-induced myocarditis, Th17 cell differentiation was activated directly by CVB3236 and regulated positively by the IL-17-NLPR3 positive feedback.237 The inhibition of Th17 differentiation and IL-17 production has been reported to ameliorate viral myocarditis.238,239 In addition, Gergely et al.240 found that anti-PD-1 treatment led to cardiac insufficiency and left ventricular dilation in male C57BL/6J mice and resulted in a significant increase in thymic inflammatory IL-17A signaling, and IL-17A inhibition prevented anti-PD-1-induced cardiac dysfunction in male C57BL/6J mice. This study revealed that IL-17A plays a promoting role in ICI-MC, but the source of IL-17A needed to be verified. In terms of EAM, neutralization of IL-17241 or inhibiting myocardial-tropic Th17 responses242 could alleviate the production of antimyocardial antibodies and cardiac inflammation. Interestingly, IL-17 exhibited strong association with myocardial fibrosis, cardiac remodeling, and DCM242 in the later stages of myocarditis. Both inducing EAM in IL-17–deficient mice and treatment EAM mice with anti-IL-17A monoclonal antibodies reduced cardiac fibrosis and improved cardiac function, indicating the essential role of IL-17 in the progression to DCM.243 A recent study244 used single-cell sequencing analysis to show that PGK1 in mice with EAM promotes Th17 cell differentiation by increasing glycolytic activity and promoting pyruvate dehydrogenase kinase 1 phosphorylation, which reduces mitochondrial reactive oxygen species production, leading to increased myocardial inflammation. This reveals the dysregulated metabolism in Th17 cells in myocarditis and provides a new direction for further exploration.

Treg cells, characterized by CD4, CD25, and FOXP3 (forkhead box protein 3), have been reported to execute immunosuppressive function via anti-inflammatory cytokines IL-10 and TGF-β, inhibitory receptors. Patients with myocarditis/DCM showed the lower percentage of Treg cells in peripheral blood.245 Depletion of GITRhighFOXP3+ natural Treg cells led to fatal autoimmune myocarditis in nude mice.246 Before CVB3 infection, adoptive transfer of Treg cells protected from the excessive inflammatory response and promote virus clearance via TGF-β-CAR pathway.247 Besides, adoptive transfer after CVB3 infection alleviated fibrosis via secreting more IL-10.248 Heart stromal/progenitor cell–derived extracellular vesicles promoted Treg cell differentiation and the production of IL-10, resulting in the reduced myocardial inflammation and functional failure in mice with EAM.249 However, TGF-β produced by Treg cells in the chronic phase exhibited profibrotic effect.250 Pre-exposure to particulate matter 2.5 activated Treg cells in spleen and aggravated CVB3-induced myocarditis,251 indicating that the premature or long-term activation of Treg cells had a bad influence in myocarditis.

The latest finding revealed that the previous Th cell differentiation could be regulated by autonomic nerves. Li et al.252 found that vagotomy in BALB/c male mice with CVB3-induced myocarditis increased the percentage of Th1 and Th17 cells, but it decreased the percentage of Th2 and Treg cells in the spleen. It also increased the expression of proinflammatory mediators and amplified inflammatory infiltration, as well as myocardial lesions in the acute and subacute phases. The authors hypothesized that this might be related to the fact that vagotomy inhibited Janus kinase 2/signal transducer and activator of transcription 3 activation and enhanced NF-κB activation of splenic CD4+ T cells. We realize that there is complicated communication between the autonomic nerves and CD4+ T cells. Vagus nerves may have a potential protective effect on inflammation, which requires more rigorous experiments in the future.

In addition to the common Th cells, there is increasing interest in the role of cytotoxic CD4+ T cells in different pathologies. With granzyme B and perforin, CD4+ T cells exert specific cytotoxicity on cells presenting pathogenic antigens and express immunomodulatory or exhaustion-related molecules, which is controlled through intrinsic IL-18R/MyD88 signaling.13,253,254 A previous study253 found that cytotoxic CD4+ T cells predominated among CD4+ T cells in the cardiac tissue of T. cruzi–infected mice and that the number correlated with the severity of myocardial inflammation.

3.8.2 CD8+ T cell subsets in myocarditis

CD8+ T cells, recognized primarily as cytotoxic T cells, usually kill infected cells via granzymes, perforins, and FAS-induced apoptosis.255 CD8+ T cells also take part in viral clearance by producing IFN-γ256 and TNF-α.257 Deficiency of CD8+ T cell with reduced IFN-γ impaired early CVB3 clearance and promoted the progression to chronic myocarditis.258 While type I IFN DNA therapy relieved chronic myocarditis by reducing CD8+ T cell aggregation in the myocardium after cytomegalovirus infection.259 These results indicated that CD8+ T cells are conducive to early viral clearance while appropriate reduced aggregation prevent the progression to chronic myocarditis. CD8+ T cells are also involved in autoimmunity. One previous study260 sequenced single-cell RNA and T cell receptors in the context of cardiac immune infiltration in Pdcd1Ctla4 mice (an ICI-MC animal model), and the investigator identified CD8+ T cells with clonal effects as the predominant cell population. Treatment with anti-CD8–depleting antibodies improved survival in Pdcd1Ctla4 mice. In both mice and patients with ICI-MC, this study suggested that α-myosin may be an important autoantigen in ICI-MC and highlights the key pathogenic role of cytotoxic CD8+ T cells. Similar immune intolerance also happened in A/J mice with EAM.261 Cytotoxic CD8+ T cells attack tissues that have lost their immune tolerance, just as they attack allografts. This process may be regulated by the surrounding tissues that express PD-L1. In cardiac transplantation,262 deletion of PD-L1 expression in graft endothelial cells may play a role in regulating CD8+ T cell infiltration. Results from mouse models suggest that loss of PD-L1 in graft endothelial cells may promote the infiltration of CD8+ T cells, thereby promoting allogeneic responses and rejection.

3.8.3 Other phenotypic T cells in myocarditis

Komarowska et al.263 described that T cells expressing c-mesenchymal epithelial transition factor (c-Met) mediated T cell cardiophilia during lymph node initiation by interacting with cardiac-produced hepatocyte growth factor. c-Met signaling facilitated T cell recruitment to the heart by inducing the release of autocrine CCL5. The authors found that autoantigen-specific c-Met T cells were increased in the circulation and myocardium of patients with inflammatory cardiomyopathy, representing loss of cardiac immune tolerance. Unlike c-Met T cells, c-Met+ T cells preferentially proliferate in the myocardium and produce multiple cytokines (IL-4, IL-17, and IL-22) to exert pathogenic effects.264 During the immune response of myocarditis, is it possible to protect the myocardium from excessive inflammatory damage by temporarily targeting c-Met inhibition, thereby reducing excessive T cell migration to the myocardium. This provides a new idea for the treatment of myocarditis.

In addition, existing studies on the role of T cells in myocarditis are mainly directed at αβT cells. Here, we briefly summarize the role of γδT cells, which are named according to the T cell receptor type. These cells mainly recognize CD1-presenting molecules and have anti-infective and antitumor effects, but they also secrete a variety of cytokines, such as IFN-γ, ILs, and TNF, upon activation to exert immunomodulatory effects.265 Several studies266–268 in viral myocarditis models have shown that γδ T cells promote the Th1 cell inflammatory response or memory function through Fas-Fas ligand interaction, restriction of Treg cell activation, and IFN-γ secretion, which also subsequently promotes the effects of self-reactive CD8+ cells. The effects of limiting Treg cells may be through the killing of Treg cells by Vγ4+γδ T cells via recognition of CD1d expressed on Treg cells using caspase-dependent apoptosis. However, these studies are far from sufficient to reveal the veil of γδT cells.

3.9 B lymphocytes

B lymphocytes, otherwise known as B cells, can be divided into 2 groups depending on their origin: B1 cells, which originate mainly from the yolk sac or fetal liver, and B2 cells, which originate mainly from the bone marrow. Both B1 and B2 cells participate in adaptive immunity, differentiating into memory B cells or secreting plasma cells that produce specific antibodies, provided that BCR and costimulatory signals are activated.269 A recent study270 used the term “regulatory B” (Breg) cell to describe the ability of B cells to suppress the immune response, and this ability to regulate immunity was present in almost all B cell subsets.

Another study showed that B cells act as a dissemination vector271 in the early stages of CVB3-induced myocarditis, and that CVB3 can directly invade and replicate in mouse B cells. Using CVB3-infected B cells as a vector for CVB3 transfer to various tissues of the body, CVB3-infected B cells were detected in the hearts of mice with CVB3-mediated myocarditis. When the virus expands substantially, leading to host cell death, CVB3 is released into local tissues and continues to infect other cells.

The main function of B cells, as an important part of humoral immunity, is antibody production. The roles of these antibodies are to neutralize toxins through antigen binding by the Fab segment, activate complement, exert regulatory effects through the Fc segment, and exert antibody-dependent cell-mediated cytotoxic effects in concert with NK cells and macrophages, among others. However, antibodies are a double-edged sword, as some low-neutralizing activity or non-neutralizing virus-specific antibodies promote viral entry into, and massive replication in, immune cells through the antibody-dependent enhancement effect, an effect that has been demonstrated in CVB3272 and COVID-19.273 Thus, the pathogenic role of antibody-dependent enhancement cannot be ignored in the pathogenesis of this virus-associated myocarditis. Anticardiac antibody–mediated myocardial injury also plays a pivotal role in the pathogenesis of myocarditis. A variety of anti-heart antibodies (AHAs) have been identified in patients with myocarditis and in animal models of CVB3-induced myocarditis and EAM, including antimyocardial contractile protein antibodies, antimyocardial membrane protein receptor antibodies, antimyocardial mitochondrial antibodies, and antimyocardial structural protein antibodies.274,275 Part of the production of these antibodies may be related to the presence of highly homologous sequences between certain viral proteins and myocardial proteins that can be mimicked by antigenic molecules to produce anticardiac antibodies. Alternatively, autoantigens that leak from cardiomyocytes after damage can cause B cells to produce AHA-mediated myocardial injury. Recently, researchers214 also identified autoreactive T cells that target TN-C, a component of the cardiac ECM, so further studies are needed to determine whether anticardiac ECM antibodies play a role in the pathogenesis of myocarditis.

B cells internalize antigens bound to B cell receptors and process them toward the presentation of T cell antigenic peptide-MHC class II molecular complexes to activate T cells. By targeting disruption of the IgM transmembrane structural domain or using anti-IgM antibodies from birth to establish a mouse model of B cell deficiency, a previous study observed comparable myocardial inflammation in B cell–deficient mice immunized simultaneously with cardiac myosin vs in wild-type mice. They inferred that macrophages or DCs, but not B cells, were the key antigen-presenting cells in the pathogenesis in EAM.276 In addition, Cen et al.277 found that myocardial pathology scores and cardiac troponin T levels in B cell–deficient mice and severe combined immunodeficient mice were lower than in wild-type mice and B cell–reconstituted severe combined immunodeficient mice, respectively, in CVB3-induced myocarditis. B cells demonstrate more powerful antigen presentation and the ability to function independent of T cells. In addition, trehalose278 alleviates myocardial inflammatory injury in CVB3-induced myocarditis by inducing B cell autophagy mediated by the AMP-activated protein kinase/ULK1 signaling pathway.

B cells secrete a variety of cytokines, including IL-2, IFN-γ, IL-12, IL-6, IL-4, TNF-α, IL-10, and IL-17, which play antipathogenic, proinflammatory, profibrotic, anti-inflammatory, and immunomodulatory roles. Here, we focused on the role of B cells in regulating the activity of other immune cells. Cen et al.277 observed a decrease in the number of Th1 and Th17 cells, no difference in Th22 cells, and an increase in Th2 cells in the spleen of B cell knockout mice compared with wild-type mice in CVB3-induced myocarditis. This trend was also demonstrated in the quantitative polymerase chain reaction results for Th1, Th2, Th17, and Th22 cell–specific transcription factors, and was also verified in in vitro cellular assays. This may be related to the fact that B cells overexpressing CD80 activate CD4+ T cells, which in turn promotes Th1 and Th17 cell differentiation.279 In contrast, Wei et al.280 found that IL-10–producing B cells are T cell dependent in the pathogenesis of CVB3-induced myocarditis. They mainly downregulate T-bet and RORγt mRNA early in the pathogenesis, and they reduce the proportion of Th1 and Th17 cells, thereby suppressing inflammation and injury in the myocardium.

In conjunction with the proposed concept of Breg cells, we should not focus only on the pathogenic role of B cells; rather, we should consider their potential protective role. In addition to secreting IL-10 to exert immunosuppressive effects, Breg cells also act by affecting Treg cells. Lu et al.281 found that B cell knockout, although alleviating the severity of myocardial inflammation in CVB3-induced myocarditis, significantly reduced the number and function of Treg cells in the spleen, blood, and heart of infected mice. This situation was reversed by B cell transmigration. Furthermore, coculture of purified B cells from wild-type mice with effector T cells in vitro allowed their conversion to Treg cells. It can be concluded that B cells contribute to the homeostasis of Treg cells in myocarditis.

In addition to interacting with T cells, B cells can increase myocardial inflammation in CVB3 mice by inhibiting M2 macrophage polarization. Li et al.282 found that in CVB3-induced myocarditis, the number of M2 macrophages was increased in the B cell knockout group compared with the wild-type group, whereas after B cell reconstitution, the number of M2 macrophages tended to decrease, and myocardial pathology scores were significantly higher. This may be related to the increased production of certain cytokines, such as IFN-γ, which promote the polarization of M1 macrophages, and the relatively low production of other cytokines, such as IL-4, which promote the polarization of M2 macrophages.277

4. Single-cell sequencing in myocarditis

scRNA-seq is a high-resolution transcriptomics sequencing technology that maps gene expression profiles and differential profiles at the single-cell level through novel technologies and probes tissue heterogeneity issues, cell development issues, and the discovery of new cell populations through in-depth and detailed analysis of complex cell populations. In addition, there is single-cell T cell receptor sequencing and single-cell B cell receptor sequencing based on the expansion of scRNA-seq. Single-cell sequencing studies in the context of myocarditis have led to a deeper understanding of these major immune cells. Hua et al.20 used scRNA-seq on the hearts from BALB/c male mice with EAM to show that macrophages were the predominant immune cell population at all disease stages and identified an inflammation-associated population of macrophages that highly expressed hypoxia-inducible factor 1-α (HIF-1α). In terms of neutrophils, they found that neutrophils increased and released IL-1 to participate in early pathogenesis. Moreover, Th17 cells with upregulated HIF-1α expression constituted the major T cell population in the acute inflammatory phase, Treg cells were the major T cell population in the subacute inflammatory phase, and γδT cells releasing IL-17 were the major T cell population in the chronic cardiomyopathy phase. They proposed for the first time that the expression of HIF-1α correlates with the degree of inflammation.

In addition, Yoon et al.19 described for the first time the peripheral immune landscape of patients with myocarditis after BNT162b2 vaccination based on the analysis of peripheral blood mononuclear cells. They found the greatest change in the transcriptome profile of monocytes and detected highly increased mRNA expression of IL-7R in a T cell subpopulation, but it was not increased in terms of protein expression. This may give us a warning that scRNA-seq detects changes only at the transcriptional level. However, this may not necessarily reflect a change at the translational level (i.e. protein expression). The different results between transcription and translation remind us that something may happen during posttranscriptional modification that prevents the protein from being expressed, and noncoding RNAs may contribute to this outcome.15

Zhu et al.18 found a significant increase in clonal cytotoxic Temra CD8+ T cells in the blood of patients with ICI-MC, while a similar phenotype was found in the blood and myocardium of relevant animal models. This study described unique transcriptional changes in Temra CD8+ T cells, including chemokine upregulation (CCL5/CCL4/CCL4L2). This subpopulation of T cells has been described in myocarditis for the first time, which may provide a new interpretation in terms of myocarditis pathogenesis.

Lasrado et al.17 studied the hearts of male A/J mice with CVB3 myocarditis and found that M2 phenotype myeloid cells and T cells exhibited increased expression of unique transcription factors that regulate cardiac remodeling, while fibroblasts exerted immunomodulatory effects due to their distinct transcriptomic profiles and subtypes. In addition, this study revealed that the composition of cardiac ILCs includes NK cells, ILC2, and ILC3, confirming our suspicions in the previous section on ILCs regarding the composition of the ILC population in myocarditis. All of the studies described previously provide an immune and transcriptome landscape of the circulation and the heart for further exploration.

5. Conclusions and expectations

Different types of innate immune cells (Fig. 1) and adaptive immune cells (Fig. 2) are involved in myocarditis development and they form a complex immune network. They play different roles in the pathogenesis of myocarditis independently or cooperatively during the acute infection, subacute immune response, and cardiac remodeling. The diversity of immune cells imply that a single pathological classification based on the major infiltrating cell types in myocardial tissue is gradually failing to meet the needs of diagnosis and treatment in clinical practice. Cooper283 suggested the identification of the immunophenotype in patients considering a combination of clinical features and high-dimensional immunological data, could be an important milestone on the road to improving the treatment and prognosis of patients with acute myocarditis. To better elaborate, select high-dimensional immunological data, and find the key intersection of the immune network, the application of single-cell sequencing technology provides a viable approach to extensive analysis of cell signaling and immunomodulatory effects in the internal transcriptome of various immune cells and their subsequent immune products.

Innate immune cells in myocarditis. Innate immune cells in myocarditis include neutrophils, eosinophils, mast cells, ILCs, monocytes, macrophages, and DCs. When suffering from myocarditis, activated neutrophils kill pathogens through phagocytosis, degranulation, extracellular traps, and cytokines, while activating the myocardial inflammatory response to produce myocardial injuries. Activated eosinophils harm the myocardium and promote fibrosis while killing pathogens through degranulation and extracellular traps. Activated mast cells produce a variety of enzymes and cytokines involved in the pathogenesis of myocarditis, with both pro- and antifibrotic effects. Activated NK cells clear infected target cells while they inhibit myocardial fibrosis. Monocytes migrate into the heart and differentiate into macrophages and DCs. Macrophages can phagocytose pathogens, and they also produce pro- and anti-inflammatory cytokines. DCs can both activate T cells through alloantigen or self-antigen presentation, while they can secrete a variety of cytokines involved in myocarditis. Figure created using FigDraw.
Fig. 1.

Innate immune cells in myocarditis. Innate immune cells in myocarditis include neutrophils, eosinophils, mast cells, ILCs, monocytes, macrophages, and DCs. When suffering from myocarditis, activated neutrophils kill pathogens through phagocytosis, degranulation, extracellular traps, and cytokines, while activating the myocardial inflammatory response to produce myocardial injuries. Activated eosinophils harm the myocardium and promote fibrosis while killing pathogens through degranulation and extracellular traps. Activated mast cells produce a variety of enzymes and cytokines involved in the pathogenesis of myocarditis, with both pro- and antifibrotic effects. Activated NK cells clear infected target cells while they inhibit myocardial fibrosis. Monocytes migrate into the heart and differentiate into macrophages and DCs. Macrophages can phagocytose pathogens, and they also produce pro- and anti-inflammatory cytokines. DCs can both activate T cells through alloantigen or self-antigen presentation, while they can secrete a variety of cytokines involved in myocarditis. Figure created using FigDraw.

Adaptive immune cells in myocarditis. Adaptive immune cells in myocarditis include CD4 T cells (Th1, Th2, Th17, and Treg cells) and CD8 T cells. When experiencing myocarditis, Th1 cells secrete IL-12 and IFN-γ. IL-12 and IFN-γ promote inflammation and myocardium injuries, while IFN-γ is conducive to pathogen clearance. Th2 cells secrete IL-4 and IL-10, while IL-4 is proinflammatory and IL-10 is anti-inflammatory, antifibrotic, and can alleviate myocardium injuries. Th17 cells secrete IL-17, which promotes myocardium injuries and fibrosis. Treg cells produce IL-10 and TGF-β, and TGF-β can promote pathogen clearance in the acute phase, while it promotes myocardium fibrosis in the chronic phase. CD8 T cells can remove pathogens via granzyme and perforin, as well as produce IFN-γ. Autoreactive CD8 T cells in myocarditis can cause myocardium injuries. B cells secret antibodies to remove pathogens or promote viral entrance via an antibody-dependent enhancement effect. AHAs produced by B cells damage the myocardium. B cells in myocarditis promote Th1 and Th17 differentiation, while they suppress Th2 and Treg differentiation. Figure created using FigDraw.
Fig. 2.

Adaptive immune cells in myocarditis. Adaptive immune cells in myocarditis include CD4 T cells (Th1, Th2, Th17, and Treg cells) and CD8 T cells. When experiencing myocarditis, Th1 cells secrete IL-12 and IFN-γ. IL-12 and IFN-γ promote inflammation and myocardium injuries, while IFN-γ is conducive to pathogen clearance. Th2 cells secrete IL-4 and IL-10, while IL-4 is proinflammatory and IL-10 is anti-inflammatory, antifibrotic, and can alleviate myocardium injuries. Th17 cells secrete IL-17, which promotes myocardium injuries and fibrosis. Treg cells produce IL-10 and TGF-β, and TGF-β can promote pathogen clearance in the acute phase, while it promotes myocardium fibrosis in the chronic phase. CD8 T cells can remove pathogens via granzyme and perforin, as well as produce IFN-γ. Autoreactive CD8 T cells in myocarditis can cause myocardium injuries. B cells secret antibodies to remove pathogens or promote viral entrance via an antibody-dependent enhancement effect. AHAs produced by B cells damage the myocardium. B cells in myocarditis promote Th1 and Th17 differentiation, while they suppress Th2 and Treg differentiation. Figure created using FigDraw.

Author contributions

K.L. reviewed the literature and wrote the manuscript. B.H. revised the manuscript. Both authors read and approved the final manuscript.

Funding

This study was supported by the Jinan Science and Technology Bureau (after subsidy) (no. 202134015) and the Taishan Scholar Foundation of Shandong Province (no. 201511099).

References

1

Schultheiss
HP
,
Kühl
U
,
Cooper
LT
.
The management of myocarditis
.
Eur Heart J
.
2011
:
32
(
21
):
2616
2625
. https://doi.org/10.1093/eurheartj/ehr165

2

Ferreira
VM
,
Schulz-Menger
J
,
Holmvang
G
,
Kramer
CM
,
Carbone
I
,
Sechtem
U
,
Kindermann
I
,
Gutberlet
M
,
Cooper
LT
,
Liu
P
, et al.
Cardiovascular magnetic resonance in nonischemic myocardial inflammation: expert recommendations
.
J Am Coll Cardiol
.
2018
:
72
(
24
):
3158
3176
. https://doi.org/10.1016/j.jacc.2018.09.072

3

Ammirati
E
,
Cipriani
M
,
Lilliu
M
,
Sormani
P
,
Varrenti
M
,
Raineri
C
,
Petrella
D
,
Garascia
A
,
Pedrotti
P
,
Roghi
A
, et al.
Survival and left ventricular function changes in fulminant versus nonfulminant acute myocarditis
.
Circulation
.
2017
:
136
(
6
):
529
545
. https://doi.org/10.1161/CIRCULATIONAHA.117.026386

4

Rose
NR
,
Wolfgram
LJ
,
Herskowitz
A
,
Beisel
KW
.
Postinfectious autoimmunity: two distinct phases of coxsackievirus B3-induced myocarditis
.
Ann N Y Acad Sci
.
1986
:
475
(
1
):
146
156
. https://doi.org/10.1111/j.1749-6632.1986.tb20864.x

5

Fairweather
D
,
Kaya
Z
,
Shellam
GR
,
Lawson
CM
,
Rose
NR
.
From infection to autoimmunity
.
J Autoimmun
.
2001
:
16
(
3
):
175
186
. https://doi.org/10.1006/jaut.2000.0492

6

Fairweather
D
,
Frisancho-Kiss
S
,
Rose
NR
.
Viruses as adjuvants for autoimmunity: evidence from coxsackievirus-induced myocarditis
.
Rev Med Virol
.
2005
:
15
(
1
):
17
27
. https://doi.org/10.1002/rmv.445

7

Neu
N
,
Rose
NR
,
Beisel
KW
,
Herskowitz
A
,
Gurri-Glass
G
,
Craig
SW
.
Cardiac myosin induces myocarditis in genetically predisposed mice
.
J Immunol
.
1987
:
139
(
11
):
3630
3636
. https://doi.org/10.4049/jimmunol.139.11.3630

8

Bachmaier
K
,
Neu
N
,
Yeung
RS
,
Mak
TW
,
Liu
P
,
Penninger
JM
.
Generation of humanized mice susceptible to peptide-induced inflammatory heart disease
.
Circulation
.
1999
:
99
(
14
):
1885
1891
. https://doi.org/10.1161/01.CIR.99.14.1885

9

Limas
CJ
,
Limas
C
,
Goldenberg
IF
,
Blair
R
.
Possible involvement of the HLA-DQB1 gene in susceptibility and resistance to human dilated cardiomyopathy
.
Am Heart J
.
1995
:
129
(
6
):
1141
1144
. https://doi.org/10.1016/0002-8703(95)90395-X

10

Vyse
TJ
,
Todd
JA
.
Genetic analysis of autoimmune disease
.
Cell
.
1996
:
85
(
3
):
311
318
. https://doi.org/10.1016/S0092-8674(00)81110-1

11

Lasrado
N
,
Reddy
J
.
An overview of the immune mechanisms of viral myocarditis
.
Rev Med Virol
.
2020
:
30
(
6
):
1
14
. https://doi.org/10.1002/rmv.2131

12

Khawaja
A
,
Bromage
DI
.
The innate immune response in myocarditis
.
Int J Biochem Cell Biol
.
2021
:
134
:
105973
. https://doi.org/10.1016/j.biocel.2021.105973

13

Han
LN
,
Li
TL
,
Zhang
YJ
,
Yang
TS
,
Ding
Y
,
Guo
SL
.
[Immune state of Th1, Th2 and Th17 subpopulation in experimental autoimmune myocarditis]
.
Sichuan Da Xue Xue Bao Yi Xue Ban
.
2011
:
42
(
6
):
751
756
.

14

Ong
S
,
Rose
NR
,
Čiháková
D
.
Natural killer cells in inflammatory heart disease
.
Clin Immunol
.
2017
:
175
:
26
33
. https://doi.org/10.1016/j.clim.2016.11.010

15

Wang
J
,
Han
B
.
Dysregulated CD4+ T cells and microRNAs in myocarditis
.
Front Immunol
.
2020
:
11
:
539
. https://doi.org/10.3389/fimmu.2020.00539

16

Huang
YV
,
Waliany
S
,
Lee
D
,
Galdos
FX
,
Witteles
RM
,
Neal
JW
,
Fan
AC
,
Maecker
HT
,
Nguyen
PK
,
Wu
SM
, et al.
The role of single-cell profiling and deep immunophenotyping in understanding immune therapy cardiotoxicity
.
J Am Coll Cardiol CardioOnc
.
2022
:
4
(
5
):
629
634
. https://doi.org/10.1016/j.jaccao.2022.08.012

17

Lasrado
N
,
Borcherding
N
,
Arumugam
R
,
Starr
TK
,
Reddy
J
.
Dissecting the cellular landscape and transcriptome network in viral myocarditis by single-cell RNA sequencing
.
iScience
.
2022
:
25
(
3
):
103865
. https://doi.org/10.1016/j.isci.2022.103865

18

Zhu
H
,
Galdos
FX
,
Lee
D
,
Waliany
S
,
Huang
YV
,
Ryan
J
,
Dang
K
,
Neal
JW
,
Wakelee
HA
,
Reddy
SA
, et al.
Identification of pathogenic immune cell subsets associated with checkpoint inhibitor-induced myocarditis
.
Circulation
.
2022
:
146
(
4
):
316
335
. https://doi.org/10.1161/CIRCULATIONAHA.121.056730

19

Yoon
BK
,
Oh
TG
,
Bu
S
,
Seo
KJ
,
Kwon
SH
,
Lee
JY
,
Kim
Y
,
Kim
J-w
,
Ahn
H-S
,
Fang
S
, et al.
The peripheral immune landscape in a patient with myocarditis after the administration of BNT162b2 mRNA vaccine
.
Mol Cells
.
2022
:
45
(
10
):
738
748
. https://doi.org/10.14348/molcells.2022.0031

20

Hua
X
,
Hu
G
,
Hu
Q
,
Chang
Y
,
Hu
Y
,
Gao
L
,
Chen
X
,
Yang
P-C
,
Zhang
Y
,
Li
M
, et al.
Single-cell RNA sequencing to dissect the immunological network of autoimmune myocarditis
.
Circulation
.
2020
:
142
(
4
):
384
400
. https://doi.org/10.1161/CIRCULATIONAHA.119.043545

21

Tang
F
,
Barbacioru
C
,
Wang
Y
,
Nordman
E
,
Lee
C
,
Xu
N
,
Wang
X
,
Bodeau
J
,
Tuch
BB
,
Siddiqui
A
, et al.
mRNA-Seq whole-transcriptome analysis of a single cell
.
Nat Methods
.
2009
:
6
(
5
):
377
382
. https://doi.org/10.1038/nmeth.1315

22

Vaduganathan
M
,
Mensah
GA
,
Turco
JV
,
Fuster
V
,
Roth
GA
.
The global burden of cardiovascular diseases and risk: a compass for future health
.
J Am Coll Cardiol
.
2022
:
80
(
25
):
2361
2371
. https://doi.org/10.1016/j.jacc.2022.11.005

23

Roth
GA
,
Mensah
GA
,
Johnson
CO
,
Addolorato
G
,
Ammirati
E
,
Baddour
LM
,
Barengo
NC
,
Beaton
AZ
,
Benjamin
EJ
,
Benziger
CP
, et al.
Global burden of cardiovascular diseases and risk factors, 1990-2019: update from the GBD 2019 study
.
J Am Coll Cardiol
.
2020
:
76
(
25
):
2982
3021
. https://doi.org/10.1016/j.jacc.2020.11.010

24

Jiménez-Alejandre
R
,
Ruiz-Fernández
I
,
Martín
P
.
Pathophysiology of immune checkpoint inhibitor-induced myocarditis
.
Cancers (Basel)
.
2022
:
14
(
18
):
4494
. https://doi.org/10.3390/cancers14184494

25

Boehmer
TK
,
Kompaniyets
L
,
Lavery
AM
,
Hsu
J
,
Ko
JY
,
Yusuf
H
,
Romano
SD
,
Gundlapalli
AV
,
Oster
ME
,
Harris
AM
.
Association between COVID-19 and myocarditis using hospital-based administrative data—united States, march 2020-January 2021
.
MMWR Morb Mortal Wkly Rep
.
2021
:
70
(
35
):
1228
1232
. https://doi.org/10.15585/mmwr.mm7035e5

26

Barda
N
,
Dagan
N
,
Ben-Shlomo
Y
,
Kepten
E
,
Waxman
J
,
Ohana
R
,
Hernán
MA
,
Lipsitch
M
,
Kohane
I
,
Netzer
D
, et al.
Safety of the BNT162b2 mRNA Covid-19 vaccine in a nationwide setting
.
N Engl J Med
.
2021
:
385
(
12
):
1078
1090
. https://doi.org/10.1056/NEJMoa2110475

27

Bozkurt
B
,
Kamat
I
,
Hotez
PJ
.
Myocarditis with COVID-19 mRNA vaccines
.
Circulation
.
2021
:
144
(
6
):
471
484
. https://doi.org/10.1161/CIRCULATIONAHA.121.056135

28

Diaz
GA
,
Parsons
GT
,
Gering
SK
,
Meier
AR
,
Hutchinson
IV
,
Robicsek
A
.
Myocarditis and pericarditis after vaccination for COVID-19
.
JAMA
.
2021
:
326
(
12
):
1210
1212
. https://doi.org/10.1001/jama.2021.13443

29

Montgomery
J
,
Ryan
M
,
Engler
R
,
Hoffman
D
,
McClenathan
B
,
Collins
L
,
Loran
D
,
Hrncir
D
,
Herring
K
,
Platzer
M
, et al.
Myocarditis following immunization with mRNA COVID-19 vaccines in members of the US military
.
JAMA Cardiol
.
2021
:
6
(
10
):
1202
1206
. https://doi.org/10.1001/jamacardio.2021.2833

30

Kim
HW
,
Jenista
ER
,
Wendell
DC
,
Azevedo
CF
,
Campbell
MJ
,
Darty
SN
,
Parker
MA
,
Kim
RJ
.
Patients with acute myocarditis following mRNA COVID-19 vaccination
.
JAMA Cardiol
.
2021
:
6
(
10
):
1196
1201
. https://doi.org/10.1001/jamacardio.2021.2828

31

Patone
M
,
Mei
XW
,
Handunnetthi
L
,
Dixon
S
,
Zaccardi
F
,
Shankar-Hari
M
,
Watkinson
P
,
Khunti
K
,
Harnden
A
, et al.
Risks of myocarditis, pericarditis, and cardiac arrhythmias associated with COVID-19 vaccination or SARS-CoV-2 infection
.
Nat Med
.
2022
:
28
(
2
):
410
422
. https://doi.org/10.1038/s41591-021-01630-0

32

Caforio
AL
,
Pankuweit
S
,
Arbustini
E
,
Basso
C
,
Gimeno-Blanes
J
,
Felix
SB
,
Fu
M
,
Helio
T
,
Heymans
S
,
Jahns
R
, et al.
Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European society of cardiology working group on myocardial and pericardial diseases
.
Eur Heart J
.
2013
:
34
(
33
):
2636
2648
, 26

33

Holzmann
M
,
Nicko
A
,
Kühl
U
,
Noutsias
M
,
Poller
W
,
Hoffmann
W
,
Morguet
A
,
Witzenbichler
B
,
Tschöpe
C
,
Schultheiss
H-P
, et al.
Complication rate of right ventricular endomyocardial biopsy via the femoral approach: a retrospective and prospective study analyzing 3048 diagnostic procedures over an 11-year period
.
Circulation
.
2008
:
118
(
17
):
1722
1728
. https://doi.org/10.1161/CIRCULATIONAHA.107.743427

34

Friedrich
MG
,
Sechtem
U
,
Schulz-Menger
J
,
Holmvang
G
,
Alakija
P
,
Cooper
LT
,
White
JA
,
Abdel-Aty
H
,
Gutberlet
M
,
Prasad
S
, et al.
Cardiovascular magnetic resonance in myocarditis: a JACC white paper
.
J Am Coll Cardiol
.
2009
:
53
(
17
):
1475
1487
. https://doi.org/10.1016/j.jacc.2009.02.007

35

Leiner
T
,
Bogaert
J
,
Friedrich
MG
,
Mohiaddin
R
,
Muthurangu
V
,
Myerson
S
,
Powell
AJ
,
Raman
SV
,
Pennell
DJ
.
SCMR Position paper (2020) on clinical indications for cardiovascular magnetic resonance
.
J Cardiovasc Magn Reson
.
2020
:
22
(
1
):
76
. https://doi.org/10.1186/s12968-020-00682-4

36

Othman
HF
,
Byrnes
J
,
Elsamny
E
,
Hamzah
M
.
Impact of ventricular arrhythmias on outcomes in children with myocarditis
.
Eur J Pediatr
.
2020
:
179
(
11
):
1779
1786
. https://doi.org/10.1007/s00431-020-03687-4

37

Tschöpe
C
,
Ammirati
E
,
Bozkurt
B
,
Caforio
ALP
,
Cooper
LT
,
Felix
SB
,
Hare
JM
,
Heidecker
B
,
Heymans
S
,
Hübner
N
, et al.
Myocarditis and inflammatory cardiomyopathy: current evidence and future directions
.
Nat Rev Cardiol
.
2021
:
18
(
3
):
169
193
. https://doi.org/10.1038/s41569-020-00435-x

38

Heymans
S
,
Eriksson
U
,
Lehtonen
J
,
Cooper
LT
Jr
.
The quest for new approaches in myocarditis and inflammatory cardiomyopathy
.
J Am Coll Cardiol
.
2016
:
68
(
21
):
2348
2364
. https://doi.org/10.1016/j.jacc.2016.09.937

39

Sagar
S
,
Liu
PP
,
Cooper
LT
Jr
.
Myocarditis
.
Lancet
.
2012
:
379
(
9817
):
738
747
. https://doi.org/10.1016/S0140-6736(11)60648-X

40

Pollack
A
,
Kontorovich
AR
,
Fuster
V
,
Dec
GW
.
Viral myocarditis–diagnosis, treatment options, and current controversies
.
Nat Rev Cardiol
.
2015
:
12
(
11
):
670
680
. https://doi.org/10.1038/nrcardio.2015.108

41

Noutsias
M
,
Fechner
H
,
de Jonge
H
,
Wang
X
,
Dekkers
D
,
Houtsmuller
AB
,
Pauschinger
M
,
Bergelson
J
,
Warraich
R
,
Yacoub
M
, et al.
Human coxsackie-adenovirus receptor is colocalized with integrins alpha(v)beta(3) and alpha(v)beta(5) on the cardiomyocyte sarcolemma and upregulated in dilated cardiomyopathy: implications for cardiotropic viral infections
.
Circulation
.
2001
:
104
(
3
):
275
280
. https://doi.org/10.1161/01.CIR.104.3.275

42

Bock
CT
,
Klingel
K
,
Kandolf
R
.
Human parvovirus B19-associated myocarditis
.
N Engl J Med
.
2010
:
362
(
13
):
1248
1249
. https://doi.org/10.1056/NEJMc0911362

43

Coyne
CB
,
Bergelson
JM
.
Virus-induced Abl and Fyn kinase signals permit coxsackievirus entry through epithelial tight junctions
.
Cell
.
2006
:
124
(
1
):
119
131
. https://doi.org/10.1016/j.cell.2005.10.035

44

Liu
PP
,
Opavsky
MA
.
Viral myocarditis: receptors that bridge the cardiovascular with the immune system?
Circ Res
.
2000
:
86
(
3
):
253
254
. https://doi.org/10.1161/01.RES.86.3.253

45

Takeuchi
O
,
Akira
S
.
Pattern recognition receptors and inflammation
.
Cell
.
2010
:
140
(
6
):
805
820
. https://doi.org/10.1016/j.cell.2010.01.022

46

Frantz
S
,
Falcao-Pires
I
,
Balligand
JL
,
Bauersachs
J
,
Brutsaert
D
,
Ciccarelli
M
,
Dawson
D
,
de Windt
LJ
,
Giacca
M
,
Hamdani
N
, et al.
The innate immune system in chronic cardiomyopathy: a European society of cardiology (ESC) scientific statement from the working group on myocardial function of the ESC
.
Eur J Heart Fail
.
2018
:
20
(
3
):
445
459
. https://doi.org/10.1002/ejhf.1138

47

Mann
DL
,
Topkara
VK
,
Evans
S
,
Barger
PM
.
Innate immunity in the adult mammalian heart: for whom the cell tolls
.
Trans Am Clin Climatol Assoc
.
2010
:
121
:
34
50
.
discussion 50–51
.

48

Mezzaroma
E
,
Toldo
S
,
Farkas
D
,
Seropian
IM
,
Van Tassell
BW
,
Salloum
FN
,
Kannan
HR
,
Menna
AC
,
Voelkel
NF
,
Abbate
A
, et al.
The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse
.
Proc Natl Acad Sci U S A
.
2011
:
108
(
49
):
19725
19730
. https://doi.org/10.1073/pnas.1108586108

49

Carroll
MC
.
The role of complement and complement receptors in induction and regulation of immunity
.
Annu Rev Immunol
.
1998
:
16
(
1
):
545
568
. https://doi.org/10.1146/annurev.immunol.16.1.545

50

Fairweather
D
,
Frisancho-Kiss
S
,
Yusung
SA
,
Barrett
MA
,
Davis
SE
,
Gatewood
SJ
,
Njoku
DB
,
Rose
NR
.
Interferon-gamma protects against chronic viral myocarditis by reducing mast cell degranulation, fibrosis, and the profibrotic cytokines transforming growth factor-beta 1, interleukin-1 beta, and interleukin-4 in the heart
.
Am J Pathol
.
2004
:
165
(
6
):
1883
1894
. https://doi.org/10.1016/S0002-9440(10)63241-5

51

Fairweather
D
,
Frisancho-Kiss
S
,
Njoku
DB
,
Nyland
JF
,
Kaya
Z
,
Yusung
SA
,
Davis
SE
,
Frisancho
JA
,
Barrett
MA
,
Rose
NR
.
Complement receptor 1 and 2 deficiency increases coxsackievirus B3-induced myocarditis, dilated cardiomyopathy, and heart failure by increasing macrophages, IL-1beta, and immune complex deposition in the heart
.
J Immunol
.
2006
:
176
(
6
):
3516
3524
. https://doi.org/10.4049/jimmunol.176.6.3516

52

Frisancho-Kiss
S
,
Davis
SE
,
Nyland
JF
,
Frisancho
JA
,
Cihakova
D
,
Barrett
MA
,
Rose
NR
,
Fairweather
D
.
Cutting edge: cross-regulation by TLR4 and T cell ig mucin-3 determines sex differences in inflammatory heart disease
.
J Immunol
.
2007
:
178
(
11
):
6710
6714
. https://doi.org/10.4049/jimmunol.178.11.6710

53

Ciháková
D
,
Sharma
RB
,
Fairweather
D
,
Afanasyeva
M
,
Rose
NR
.
Animal models for autoimmune myocarditis and autoimmune thyroiditis
.
Methods Mol Med
.
2004
:
102
:
175
193
. https://doi.org/10.1385/1-59259-805-6:175

54

Kaya
Z
,
Afanasyeva
M
,
Wang
Y
,
Dohmen
KM
,
Schlichting
J
,
Tretter
T
,
Fairweather
D
,
Holers
VM
,
Rose
NR
.
Contribution of the innate immune system to autoimmune myocarditis: a role for complement
.
Nat Immunol
.
2001
:
2
(
8
):
739
745
. https://doi.org/10.1038/90686

55

Park
S
,
Krshnan
L
,
Call
MJ
,
Call
ME
,
Im
W
.
Structural conservation and effects of alterations in T cell receptor transmembrane interfaces
.
Biophys J
.
2018
:
114
(
5
):
1030
1035
. https://doi.org/10.1016/j.bpj.2018.01.004

56

Maisch
B
.
Cardio-immunology of myocarditis: focus on immune mechanisms and treatment options
.
Front Cardiovasc Med
.
2019
:
6
:
48
. https://doi.org/10.3389/fcvm.2019.00048

57

McNamara
DM
,
Starling
RC
,
Cooper
LT
,
Boehmer
JP
,
Mather
PJ
,
Janosko
KM
,
Gorcsan
J
,
Kip
KE
,
Dec
GW
.
Clinical and demographic predictors of outcomes in recent onset dilated cardiomyopathy: results of the IMAC (intervention in myocarditis and acute cardiomyopathy)-2 study
.
J Am Coll Cardiol
.
2011
:
58
(
11
):
1112
1118
. https://doi.org/10.1016/j.jacc.2011.05.033

58

Coronado
MJ
,
Brandt
JE
,
Kim
E
,
Bucek
A
,
Bedja
D
,
Abston
ED
,
Shin
J
,
Gabrielson
KL
,
Mitzner
W
,
Fairweather
D
.
Testosterone and interleukin-1β increase cardiac remodeling during coxsackievirus B3 myocarditis via serpin A 3n
.
Am J Physiol Heart Circ Physiol
.
2012
:
302
(
8
):
H1726
H1736
. https://doi.org/10.1152/ajpheart.00783.2011

59

Satoh
M
,
Tamura
G
,
Segawa
I
,
Tashiro
A
,
Hiramori
K
,
Satodate
R
.
Expression of cytokine genes and presence of enteroviral genomic RNA in endomyocardial biopsy tissues of myocarditis and dilated cardiomyopathy
.
Virchows Arch
.
1996
:
427
(
5
):
503
509
. https://doi.org/10.1007/BF00199511

60

Pauschinger
M
,
Kühl
U
,
Dörner
A
,
Schieferecke
K
,
Petschauer
S
,
Rauch
U
,
Schwimmbeck
PL
,
Kandolf
R
,
Schultheiss
HP
.
[Detection of enteroviral RNA in endomyocardial biopsies in inflammatory cardiomyopathy and idiopathic dilated cardiomyopathy]
.
Z Kardiol
.
1998
:
87
(
6
):
443
452
. https://doi.org/10.1007/s003920050199

61

Kühl
U
,
Pauschinger
M
,
Schwimmbeck
PL
,
Seeberg
B
,
Lober
C
,
Noutsias
M
,
Poller
W
,
Schultheiss
H-P
.
Interferon-beta treatment eliminates cardiotropic viruses and improves left ventricular function in patients with myocardial persistence of viral genomes and left ventricular dysfunction
.
Circulation
.
2003
:
107
(
22
):
2793
2798
. https://doi.org/10.1161/01.CIR.0000072766.67150.51

62

Kuethe
F
,
Lindner
J
,
Matschke
K
,
Wenzel
JJ
,
Norja
P
,
Ploetze
K
,
Schaal
S
,
Kamvissi
V
,
Bornstein
SR
,
Schwanebeck
U
, et al.
Prevalence of parvovirus B19 and human bocavirus DNA in the heart of patients with no evidence of dilated cardiomyopathy or myocarditis
.
Clin Infect Dis
.
2009
:
49
(
11
):
1660
1666
. https://doi.org/10.1086/648074

63

Wang
Z
,
Liao
Y
,
Dong
J
,
Li
S
,
Wang
J
,
Fu
ML
.
Clinical significance and pathogenic role of anti-cardiac myosin autoantibody in dilated cardiomyopathy
.
Chin Med J (Engl)
.
2003
:
116
(
4
):
499
502
.

64

Woodiwiss
AJ
,
Tsotetsi
OJ
,
Sprott
S
,
Lancaster
EJ
,
Mela
T
,
Chung
ES
,
Meyer
TE
,
Norton
GR
.
Reduction in myocardial collagen cross-linking parallels left ventricular dilatation in rat models of systolic chamber dysfunction
.
Circulation
.
2001
:
103
(
1
):
155
160
. https://doi.org/10.1161/01.CIR.103.1.155

65

Li
J
,
Schwimmbeck
PL
,
Tschope
C
,
Leschka
S
,
Husmann
L
,
Rutschow
S
,
Reichenbach
F
,
Noutsias
M
,
Kobalz
U
,
Poller
W
.
Collagen degradation in a murine myocarditis model: relevance of matrix metalloproteinase in association with inflammatory induction
.
Cardiovasc Res
.
2002
:
56
(
2
):
235
247
. https://doi.org/10.1016/S0008-6363(02)00546-1

66

Pauschinger
M
,
Knopf
D
,
Petschauer
S
,
Doerner
A
,
Poller
W
,
Schwimmbeck
PL
,
Kühl
U
,
Schultheiss
H-P
.
Dilated cardiomyopathy is associated with significant changes in collagen type I/III ratio
.
Circulation
.
1999
:
99
(
21
):
2750
2756
. https://doi.org/10.1161/01.CIR.99.21.2750

67

Deswal
A
,
Petersen
NJ
,
Feldman
AM
,
Young
JB
,
White
BG
,
Mann
DL
.
Cytokines and cytokine receptors in advanced heart failure: an analysis of the cytokine database from the vesnarinone trial (VEST)
.
Circulation
.
2001
:
103
(
16
):
2055
2059
. https://doi.org/10.1161/01.CIR.103.16.2055

68

Siwik
DA
,
Chang
DL
,
Colucci
WS
.
Interleukin-1beta and tumor necrosis factor-alpha decrease collagen synthesis and increase matrix metalloproteinase activity in cardiac fibroblasts in vitro
.
Circ Res
.
2000
:
86
(
12
):
1259
1265
. https://doi.org/10.1161/01.RES.86.12.1259

69

Sivasubramanian
N
,
Coker
ML
,
Kurrelmeyer
KM
,
MacLellan
WR
,
DeMayo
FJ
,
Spinale
FG
,
Mann
DL
.
Left ventricular remodeling in transgenic mice with cardiac restricted overexpression of tumor necrosis factor
.
Circulation
.
2001
:
104
(
7
):
826
831
. https://doi.org/10.1161/hc3401.093154

70

Sainani
GS
,
Krompotic
E
,
Slodki
SJ
.
Adult heart disease due to the coxsackie virus B infection
.
Medicine (Baltimore)
.
1968
:
47
(
2
):
133
147
. https://doi.org/10.1097/00005792-196803000-00003

71

Woodruff
JF
.
Viral myocarditis. A review
.
Am J Pathol
.
1980
:
101
(
2
):
425
484
.

72

Fairweather
D
,
Cooper
LT
Jr
,
Blauwet
LA
.
Sex and gender differences in myocarditis and dilated cardiomyopathy
.
Curr Probl Cardiol
.
2013
:
38
(
1
):
7
46
. https://doi.org/10.1016/j.cpcardiol.2012.07.003

73

Sheppard
R
,
Bedi
M
,
Kubota
T
,
Semigran
MJ
,
Dec
W
,
Holubkov
R
,
Feldman
AM
,
Rosenblum
WD
,
McTiernan
CF
,
McNamara
DM
.
Myocardial expression of fas and recovery of left ventricular function in patients with recent-onset cardiomyopathy
.
J Am Coll Cardiol
.
2005
:
46
(
6
):
1036
1042
. https://doi.org/10.1016/j.jacc.2005.05.067

74

Cocker
MS
,
Abdel-Aty
H
,
Strohm
O
,
Friedrich
MG
.
Age and gender effects on the extent of myocardial involvement in acute myocarditis: a cardiovascular magnetic resonance study
.
Heart
.
2009
:
95
(
23
):
1925
1930
. https://doi.org/10.1136/hrt.2008.164061

75

Frisancho-Kiss
S
,
Nyland
JF
,
Davis
SE
,
Frisancho
JA
,
Barrett
MA
,
Rose
NR
,
Fairweather
D
.
Sex differences in coxsackievirus B3-induced myocarditis: IL-12Rbeta1 signaling and IFN-gamma increase inflammation in males independent from STAT4
.
Brain Res
.
2006
:
1126
(
1
):
139
147
. https://doi.org/10.1016/j.brainres.2006.08.003

76

Barcena
ML
,
Jeuthe
S
,
Niehues
MH
,
Pozdniakova
S
,
Haritonow
N
,
Kühl
AA
,
Messroghli
DR
,
Regitz-Zagrosek
V
.
Sex-Specific differences of the inflammatory state in experimental autoimmune myocarditis
.
Front Immunol
.
2021
:
12
:
686384
. https://doi.org/10.3389/fimmu.2021.686384

77

Huber
SA
,
Pfaeffle
B
.
Differential th1 and th2 cell responses in male and female BALB/c mice infected with coxsackievirus group B type 3
.
J Virol
.
1994
:
68
(
8
):
5126
5132
. https://doi.org/10.1128/jvi.68.8.5126-5132.1994

78

Huber
SA
,
Kupperman
J
,
Newell
MK
.
Estradiol prevents and testosterone promotes Fas-dependent apoptosis in CD4+ Th2 cells by altering Bcl 2 expression
.
Lupus
.
1999
:
8
(
5
):
384
387
. https://doi.org/10.1177/096120339900800511

79

Huber
SA
.
Coxsackievirus B3-induced myocarditis: infection of females during the estrus phase of the ovarian cycle leads to activation of T regulatory cells
.
Virology
.
2008
:
378
(
2
):
292
298
. https://doi.org/10.1016/j.virol.2008.05.015

80

Lyden
DC
,
Olszewski
J
,
Feran
M
,
Job
LP
,
Huber
SA
.
Coxsackievirus B-3-induced myocarditis. Effect of sex steroids on viremia and infectivity of cardiocytes
.
Am J Pathol
.
1987
:
126
(
3
):
432
438
.

81

Frisancho-Kiss
S
,
Coronado
MJ
,
Frisancho
JA
,
Lau
VM
,
Rose
NR
,
Klein
SL
,
Fairweather
D
.
Gonadectomy of male BALB/c mice increases Tim-3(+) alternatively activated M2 macrophages, Tim-3(+) T cells, Th2 cells and Treg in the heart during acute coxsackievirus-induced myocarditis
.
Brain Behav Immun
.
2009
:
23
(
5
):
649
657
. https://doi.org/10.1016/j.bbi.2008.12.002

82

Su
N
,
Yue
Y
,
Xiong
S
.
Monocytic myeloid-derived suppressor cells from females, but not males, alleviate CVB3-induced myocarditis by increasing regulatory and CD4(+)IL-10(+) T cells
.
Sci Rep
.
2016
:
6
(
1
):
22658
. https://doi.org/10.1038/srep22658

83

Rettew
JA
,
Huet
YM
,
Marriott
I
.
Estrogens augment cell surface TLR4 expression on murine macrophages and regulate sepsis susceptibility in vivo
.
Endocrinology
.
2009
:
150
(
8
):
3877
3884
. https://doi.org/10.1210/en.2009-0098

84

Seillet
C
,
Laffont
S
,
Trémollières
F
,
Rouquié
N
,
Ribot
C
,
Arnal
JF
,
Douin-Echinard
V
,
Gourdy
P
,
Guéry
J-C
.
The TLR-mediated response of plasmacytoid dendritic cells is positively regulated by estradiol in vivo through cell-intrinsic estrogen receptor α signaling
.
Blood
.
2012
:
119
(
2
):
454
464
. https://doi.org/10.1182/blood-2011-08-371831

85

Rettew
JA
,
Huet-Hudson
YM
,
Marriott
I
.
Testosterone reduces macrophage expression in the mouse of toll-like receptor 4, a trigger for inflammation and innate immunity
.
Biol Reprod
.
2008
:
78
(
3
):
432
437
. https://doi.org/10.1095/biolreprod.107.063545

86

Bouman
A
,
Heineman
MJ
,
Faas
MM
.
Sex hormones and the immune response in humans
.
Hum Reprod Update
.
2005
:
11
(
4
):
411
423
. https://doi.org/10.1093/humupd/dmi008

87

Malkin
CJ
,
Pugh
PJ
,
Jones
RD
,
Kapoor
D
,
Channer
KS
,
Jones
TH
.
The effect of testosterone replacement on endogenous inflammatory cytokines and lipid profiles in hypogonadal men
.
J Clin Endocrinol Metab
.
2004
:
89
(
7
):
3313
3318
. https://doi.org/10.1210/jc.2003-031069

88

Bobjer
J
,
Katrinaki
M
,
Tsatsanis
C
,
Lundberg Giwercman
Y
,
Giwercman
A
.
Negative association between testosterone concentration and inflammatory markers in young men: a nested cross-sectional study
.
PLoS One
.
2013
:
8
(
4
):
e61466
. https://doi.org/10.1371/journal.pone.0061466

89

Kalinchenko
SY
,
Tishova
YA
,
Mskhalaya
GJ
,
Gooren
LJ
,
Giltay
EJ
,
Saad
F
.
Effects of testosterone supplementation on markers of the metabolic syndrome and inflammation in hypogonadal men with the metabolic syndrome: the double-blinded placebo-controlled Moscow study
.
Clin Endocrinol (Oxf)
.
2010
:
73
(
5
):
602
612
. https://doi.org/10.1111/j.1365-2265.2010.03845.x

90

Silvestre-Roig
C
,
Fridlender
ZG
,
Glogauer
M
,
Scapini
P
.
Neutrophil diversity in health and disease
.
Trends Immunol
.
2019
:
40
(
7
):
565
583
. https://doi.org/10.1016/j.it.2019.04.012

91

Futosi
K
,
Fodor
S
,
Mócsai
A
.
Neutrophil cell surface receptors and their intracellular signal transduction pathways
.
Int Immunopharmacol
.
2013
:
17
(
3
):
638
650
. https://doi.org/10.1016/j.intimp.2013.06.034

92

Zhang
Q
,
Zhang
H
,
Yan
X
,
Ma
S
,
Yao
X
,
Shi
Y
, et al.
Neutrophil infiltration and myocarditis in patients with severe COVID-19: a post-mortem study
.
Front Cardiovasc Med
.
2022
:
9
:
1026866
. https://doi.org/10.3389/fcvm.2022.1026866

93

Rivadeneyra
L
,
Charó
N
,
Kviatcovsky
D
,
de la Barrera
S
,
Gómez
RM
,
Schattner
M
.
Role of neutrophils in CVB3 infection and viral myocarditis
.
J Mol Cell Cardiol
.
2018
:
125
:
149
161
. https://doi.org/10.1016/j.yjmcc.2018.08.029

94

Hiraiwa
H
,
Morimoto
R
,
Ando
R
,
Ito
R
,
Araki
T
,
Mizutani
T
,
Kazama
S
,
Kimura
Y
,
Oishi
H
,
Kuwayama
T
, et al.
Recurrent fulminant non-rheumatic streptococcal myocarditis proven by endomyocardial biopsy and autopsy
.
J Cardiol Cases
.
2022
:
26
(
1
):
62
65
. https://doi.org/10.1016/j.jccase.2022.02.004

95

Sheshachalam
A
,
Srivastava
N
,
Mitchell
T
,
Lacy
P
,
Eitzen
G
.
Granule protein processing and regulated secretion in neutrophils
.
Front Immunol
.
2014
:
5
:
448
. https://doi.org/10.3389/fimmu.2014.00448

96

Lacy
P
.
Mechanisms of degranulation in neutrophils
.
Allergy Asthma Clin Immunol
.
2006
:
2
(
3
):
98
108
. https://doi.org/10.1186/1710-1492-2-3-98

97

Sengeløv
H
,
Kjeldsen
L
,
Borregaard
N
.
Control of exocytosis in early neutrophil activation
.
J Immunol
.
1993
:
150
(
4
):
1535
1543
. https://doi.org/10.4049/jimmunol.150.4.1535

98

Herrmann
L
,
Schelletter
L
,
Hoffrogge
R
,
Niehaus
K
,
Rudolph
V
,
Farr
M
.
Human coxsackie- and adenovirus receptor is a putative target of neutrophil elastase-mediated shedding
.
Mol Biol Rep
.
2022
:
49
(
4
):
3213
3223
. https://doi.org/10.1007/s11033-022-07153-2

99

Klopf
J
,
Brostjan
C
,
Eilenberg
W
,
Neumayer
C
.
Neutrophil extracellular traps and their implications in cardiovascular and inflammatory disease
.
Int J Mol Sci
.
2021
:
22
(
2
):
559
. https://doi.org/10.3390/ijms22020559

100

Kolaczkowska
E
,
Kubes
P
.
Neutrophil recruitment and function in health and inflammation
.
Nat Rev Immunol
.
2013
:
13
(
3
):
159
175
. https://doi.org/10.1038/nri3399

101

Ling
S
,
Xu
JW
.
NETosis as a pathogenic factor for heart failure
.
Oxid Med Cell Longev
.
2021
:
2021
:
6687096
. https://doi.org/10.1155/2021/6687096

102

Branzk
N
,
Lubojemska
A
,
Hardison
SE
,
Wang
Q
,
Gutierrez
MG
,
Brown
GD
,
Papayannopoulos
V
.
Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens
.
Nat Immunol
.
2014
:
15
(
11
):
1017
1025
. https://doi.org/10.1038/ni.2987

103

Carai
P
,
González
LF
,
Van Bruggen
S
,
Spalart
V
,
De Giorgio
D
,
Geuens
N
,
Martinod
K
,
Jones
EAV
,
Heymans
S
.
Neutrophil inhibition improves acute inflammation in a murine model of viral myocarditis
.
Cardiovasc Res
.
2023
:
118
(
17
):
3331
3345
. https://doi.org/10.1093/cvr/cvac052

104

Weckbach
LT
,
Grabmaier
U
,
Uhl
A
,
Gess
S
,
Boehm
F
,
Zehrer
A
,
Pick
R
,
Salvermoser
M
,
Czermak
T
,
Pircher
J
, et al.
Midkine drives cardiac inflammation by promoting neutrophil trafficking and NETosis in myocarditis
.
J Exp Med
.
2019
:
216
(
2
):
350
368
. https://doi.org/10.1084/jem.20181102

105

Li
B
,
Cao
X
,
Ai
G
,
Liu
Y
,
Lv
C
,
Jin
L
,
Xu
R
,
Zhao
G
,
Yuan
H
.
Interleukin-37 alleviates myocardial injury induced by coxsackievirus B3 via inhibiting neutrophil extracellular traps formation
.
Int Immunopharmacol
.
2022
:
113
(
Pt A
):
109343
. https://doi.org/10.1016/j.intimp.2022.109343

106

Chrysanthopoulou
A
,
Mitroulis
I
,
Apostolidou
E
,
Arelaki
S
,
Mikroulis
D
,
Konstantinidis
T
,
Sivridis
E
,
Koffa
M
,
Giatromanolaki
A
,
Boumpas
DT
, et al.
Neutrophil extracellular traps promote differentiation and function of fibroblasts
.
J Pathol
.
2014
:
233
(
3
):
294
307
. https://doi.org/10.1002/path.4359

107

Tillack
K
,
Breiden
P
,
Martin
R
,
Sospedra
M
.
T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses
.
J Immunol
.
2012
:
188
(
7
):
3150
3159
. https://doi.org/10.4049/jimmunol.1103414

108

Flo
TH
,
Smith
KD
,
Sato
S
,
Rodriguez
DJ
,
Holmes
MA
,
Strong
RK
,
Akira
S
,
Aderem
A
.
Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron
.
Nature
.
2004
:
432
(
7019
):
917
921
. https://doi.org/10.1038/nature03104

109

Devireddy
LR
,
Gazin
C
,
Zhu
X
,
Green
MR
.
A cell-surface receptor for lipocalin 24p3 selectively mediates apoptosis and iron uptake
.
Cell
.
2005
:
123
(
7
):
1293
1305
. https://doi.org/10.1016/j.cell.2005.10.027

110

Chen
W
,
Li
W
,
Zhang
Z
,
Tang
X
,
Wu
S
,
Yao
G
,
Li
K
,
Wang
D
,
Xu
Y
,
Feng
R
, et al.
Lipocalin-2 exacerbates lupus nephritis by promoting th1 cell differentiation
.
J Am Soc Nephrol
.
2020
:
31
(
10
):
2263
2277
. https://doi.org/10.1681/ASN.2019090937

111

Ding
L
,
Hanawa
H
,
Ota
Y
,
Hasegawa
G
,
Hao
K
,
Asami
F
,
Watanabe
R
,
Yoshida
T
,
Toba
K
,
Yoshida
K
, et al.
Lipocalin-2/neutrophil gelatinase-B associated lipocalin is strongly induced in hearts of rats with autoimmune myocarditis and in human myocarditis
.
Circ J
.
2010
:
74
(
3
):
523
530
. https://doi.org/10.1253/circj.CJ-09-0485

112

Grabie
N
,
Hsieh
DT
,
Buono
C
,
Westrich
JR
,
Allen
JA
,
Pang
H
,
Stavrakis
G
,
Lichtman
AH
.
Neutrophils sustain pathogenic CD8+ T cell responses in the heart
.
Am J Pathol
.
2003
:
163
(
6
):
2413
2420
. https://doi.org/10.1016/S0002-9440(10)63596-1

113

Scapini
P
,
Bazzoni
F
,
Cassatella
MA
.
Regulation of B-cell-activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils
.
Immunol Lett
.
2008
:
116
(
1
):
1
6
. https://doi.org/10.1016/j.imlet.2007.11.009

114

Jackson
DJ
,
Akuthota
P
,
Roufosse
F
.
Eosinophils and eosinophilic immune dysfunction in health and disease
.
Eur Respir Rev
.
2022
:
31
(
163
):
210150
. https://doi.org/10.1183/16000617.0150-2021

115

Diny
NL
,
Rose
NR
,
Čiháková
D
.
Eosinophils in autoimmune diseases
.
Front Immunol
.
2017
:
8
:
484
. https://doi.org/10.3389/fimmu.2017.00484

116

Melo
RC
,
Liu
L
,
Xenakis
JJ
,
Spencer
LA
.
Eosinophil-derived cytokines in health and disease: unraveling novel mechanisms of selective secretion
.
Allergy
.
2013
:
68
(
3
):
274
284
. https://doi.org/10.1111/all.12103

117

Simon
HU
,
Yousefi
S
,
Germic
N
,
Arnold
IC
,
Haczku
A
,
Karaulov
AV
,
Simon
D
,
Rosenberg
HF
.
The cellular functions of eosinophils: collegium internationale allergologicum (CIA) update 2020
.
Int Arch Allergy Immunol
.
2020
:
181
(
1
):
11
23
. https://doi.org/10.1159/000504847

118

Oakley
CM
,
Olsen
GJ
.
Eosinophilia and heart disease
.
Br Heart J
.
1977
:
39
(
3
):
233
237
. https://doi.org/10.1136/hrt.39.3.233

119

Almusawi
H
,
daSilva-deAbreu
A
,
Zerpa
T
,
Mandras
SA
,
Eiswirth
C
.
Elusive diagnosis of eosinophilic myocarditis: a case series
.
Curr Probl Cardiol
.
2021
:
46
(
10
):
100849
. https://doi.org/10.1016/j.cpcardiol.2021.100849

120

Kuchynka
P
,
Palecek
T
,
Masek
M
,
Cerny
V
,
Lambert
L
,
Vitkova
I
,
Linhart
A
.
Current diagnostic and therapeutic aspects of eosinophilic myocarditis
.
Biomed Res Int
.
2016
:
2016
:
2829583
. https://doi.org/10.1155/2016/2829583

121

Hoshino
N
,
Yanase
M
,
Ichiyasu
T
,
Kuwahara
K
,
Kawai
H
,
Muramatsu
T
,
Ishii
H
,
Tsukamoto
T
,
Morimoto
S-i
,
Izawa
H
.
An autopsy case report of fulminant myocarditis: following mRNA COVID-19 vaccination
.
J Cardiol Cases
.
2022
:
26
(
6
):
391
394
. https://doi.org/10.1016/j.jccase.2022.06.006

122

Fettrelet
T
,
Gigon
L
,
Karaulov
A
,
Yousefi
S
,
Simon
HU
.
The Enigma of eosinophil degranulation
.
Int J Mol Sci
.
2021
:
22
(
13
):
7091
. https://doi.org/10.3390/ijms22137091

123

Tai
PC
,
Ackerman
SJ
,
Spry
CJ
,
Dunnette
S
,
Olsen
EG
,
Gleich
GJ
.
Deposits of eosinophil granule proteins in cardiac tissues of patients with eosinophilic endomyocardial disease
.
Lancet
.
1987
:
1
(
8534
):
643
647
. https://doi.org/10.1016/S0140-6736(87)90412-0

124

deMello
DE
,
Liapis
H
,
Jureidini
S
,
Nouri
S
,
Kephart
GM
,
Gleich
GJ
.
Cardiac localization of eosinophil-granule major basic protein in acute necrotizing myocarditis
.
N Engl J Med
.
1990
:
323
(
22
):
1542
1545
. https://doi.org/10.1056/NEJM199011293232207

125

Diny
NL
,
Baldeviano
GC
,
Talor
MV
,
Barin
JG
,
Ong
S
,
Bedja
D
,
Hays
AG
,
Gilotra
NA
,
Coppens
I
,
Rose
NR
, et al.
Eosinophil-derived IL-4 drives progression of myocarditis to inflammatory dilated cardiomyopathy
.
J Exp Med
.
2017
:
214
(
4
):
943
957
. https://doi.org/10.1084/jem.20161702

126

Nagase
H
,
Ueki
S
,
Fujieda
S
.
The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis
.
Allergol Int
.
2020
:
69
(
2
):
178
186
. https://doi.org/10.1016/j.alit.2020.02.002

127

Liew
FY
,
Pitman
NI
,
McInnes
IB
.
Disease-associated functions of IL-33: the new kid in the IL-1 family
.
Nat Rev Immunol
.
2010
:
10
(
2
):
103
110
. https://doi.org/10.1038/nri2692

128

Abston
ED
,
Barin
JG
,
Cihakova
D
,
Bucek
A
,
Coronado
MJ
,
Brandt
JE
,
Bedja
D
,
Kim
JB
,
Georgakopoulos
D
,
Gabrielson
KL
, et al.
IL-33 independently induces eosinophilic pericarditis and cardiac dilation: sT2 improves cardiac function
.
Circ Heart Fail
.
2012
:
5
(
3
):
366
375
. https://doi.org/10.1161/CIRCHEARTFAILURE.111.963769

129

Diny
NL
,
Hou
X
,
Barin
JG
,
Chen
G
,
Talor
MV
,
Schaub
J
,
Russell
SD
,
Klingel
K
,
Rose
NR
,
Č
D
.
Macrophages and cardiac fibroblasts are the main producers of eotaxins and regulate eosinophil trafficking to the heart
.
Eur J Immunol
.
2016
:
46
(
12
):
2749
2760
. https://doi.org/10.1002/eji.201646557

130

Ueki
S
,
Konno
Y
,
Takeda
M
,
Moritoki
Y
,
Hirokawa
M
,
Matsuwaki
Y
,
Honda
K
,
Ohta
N
,
Yamamoto
S
,
Takagi
Y
, et al.
Eosinophil extracellular trap cell death-derived DNA traps: their presence in secretions and functional attributes
.
J Allergy Clin Immunol
.
2016
:
137
(
1
):
258
267
. https://doi.org/10.1016/j.jaci.2015.04.041

131

Yousefi
S
,
Gold
JA
,
Andina
N
,
Lee
JJ
,
Kelly
AM
,
Kozlowski
E
,
Schmid
I
,
Straumann
A
,
Reichenbach
J
,
Gleich
GJ
, et al.
Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense
.
Nat Med
.
2008
:
14
(
9
):
949
953
. https://doi.org/10.1038/nm.1855

132

Sim
MS
,
Kim
HJ
,
Bae
I
,
Kim
C
,
Chang
HS
,
Choi
Y
,
Lee
D-H
,
Park
H-S
,
Chung
IY
.
Calcium ionophore-activated platelets induce eosinophil extracellular trap formation
.
Allergol Int
.
2022
:
72
(
3
):
466
476
. https://doi.org/10.1016/j.alit.2022.12.002

133

Patella
V
,
Marinò
I
,
Lampärter
B
,
Arbustini
E
,
Adt
M
,
Marone
G
.
Human heart mast cells. Isolation, purification, ultrastructure, and immunologic characterization
.
J Immunol
.
1995
:
154
(
6
):
2855
2865
. https://doi.org/10.4049/jimmunol.154.6.2855

134

Ye
L
,
Shi
S
,
Chen
W
.
Innate immunity in pancreatic cancer: lineage tracing and function
.
Front Immunol
.
2022
:
13
:
1081919
. https://doi.org/10.3389/fimmu.2022.1081919

135

Galli
SJ
,
Gaudenzio
N
,
Tsai
M
.
Mast cells in inflammation and disease: recent progress and ongoing concerns
.
Annu Rev Immunol
.
2020
:
38
(
1
):
49
77
. https://doi.org/10.1146/annurev-immunol-071719-094903

136

Higuchi
H
,
Hara
M
,
Yamamoto
K
,
Miyamoto
T
,
Kinoshita
M
,
Yamada
T
,
Uchiyama
K
,
Matsumori
A
.
Mast cells play a critical role in the pathogenesis of viral myocarditis
.
Circulation
.
2008
:
118
(
4
):
363
372
. https://doi.org/10.1161/CIRCULATIONAHA.107.741595

137

Luo
Y
,
Zhang
H
,
Yu
J
,
Wei
L
,
Li
M
,
Xu
W
.
Stem cell factor/mast cell/CCL2/monocyte/macrophage axis promotes coxsackievirus B3 myocarditis and cardiac fibrosis by increasing Ly6C(high) monocyte influx and fibrogenic mediators production
.
Immunology
.
2022
:
167
(
4
):
590
605
. https://doi.org/10.1111/imm.13556

138

Raj
S
,
Unsworth
LD
.
Targeting active sites of inflammation using inherent properties of tissue-resident mast cells
.
Acta Biomater
.
2023
:
159
:
21
37
. https://doi.org/10.1016/j.actbio.2023.01.024

139

Mukai
K
,
Tsai
M
,
Saito
H
,
Galli
SJ
.
Mast cells as sources of cytokines, chemokines, and growth factors
.
Immunol Rev
.
2018
:
282
(
1
):
121
150
. https://doi.org/10.1111/imr.12634

140

Varricchi
G
,
Marone
G
,
Kovanen
PT
.
Cardiac mast cells: underappreciated immune cells in cardiovascular homeostasis and disease
.
Trends Immunol
.
2020
:
41
(
8
):
734
746
. https://doi.org/10.1016/j.it.2020.06.006

141

Galli
SJ
,
Grimbaldeston
M
,
Tsai
M
.
Immunomodulatory mast cells: negative, as well as positive, regulators of immunity
.
Nat Rev Immunol
.
2008
:
8
(
6
):
478
486
. https://doi.org/10.1038/nri2327

142

Stasiak
A
,
Gola
J
,
Kraszewska
K
,
Mussur
M
,
Kobos
J
,
Mazurek
U
,
Stark
H
,
Fogel
WA
.
Experimental autoimmune myocarditis in rats and therapeutic histamine H1 - H4 receptor inhibition
.
J Physiol Pharmacol
.
2018
:
69
(
6
):
889
900
. https://doi.org/10.26402/jpp.2018.6.13

143

Möllerherm
H
,
von Köckritz-Blickwede
M
,
Branitzki-Heinemann
K
.
Antimicrobial activity of mast cells: role and relevance of extracellular DNA traps
.
Front Immunol
.
2016
:
7
:
265
. https://doi.org/10.3389/fimmu.2016.00265

144

Bruno
KA
,
Macomb
LP
,
Morales-Lara
AC
,
Mathews
JE
,
Frisancho
JA
,
Yang
AL
,
Di Florio
DN
,
Edenfield
BH
,
Whelan
ER
,
Salomon
GR
, et al.
Sex-Specific effects of plastic caging in murine viral myocarditis
.
Int J Mol Sci
.
2021
:
22
(
16
):
8834
. https://doi.org/10.3390/ijms22168834

145

Geiger
TL
,
Sun
JC
.
Development and maturation of natural killer cells
.
Curr Opin Immunol
.
2016
:
39
:
82
89
. https://doi.org/10.1016/j.coi.2016.01.007

146

Wang
F
,
Cui
Y
,
He
D
,
Gong
L
,
Liang
H
.
Natural killer cells in sepsis: friends or foes?
Front Immunol
.
2023
:
14
:
1101918
. https://doi.org/10.3389/fimmu.2023.1101918

147

Márquez
ME
,
Millet
C
,
Stekman
H
,
Conesa
A
,
Deglesne
PA
,
Toro
F
,
Sanctis
JD
,
Blanca
I
.
CD16 cross-linking induces increased expression of CD56 and production of IL-12 in peripheral NK cells
.
Cell Immunol
.
2010
:
264
(
1
):
86
92
. https://doi.org/10.1016/j.cellimm.2010.05.002

148

Perera Molligoda Arachchige AS
.
Human NK cells: from development to effector functions
.
Innate Immun
.
2021
:
27
(
3
):
212
229
. https://doi.org/10.1177/17534259211001512

149

Lanier
LL
.
Up on the tightrope: natural killer cell activation and inhibition
.
Nat Immunol
.
2008
:
9
(
5
):
495
502
. https://doi.org/10.1038/ni1581

150

Prager
I
,
Watzl
C
.
Mechanisms of natural killer cell-mediated cellular cytotoxicity
.
J Leukoc Biol
.
2019
:
105
(
6
):
1319
1329
. https://doi.org/10.1002/JLB.MR0718-269R

151

Ramírez-Labrada
A
,
Pesini
C
,
Santiago
L
,
Hidalgo
S
,
Calvo-Pérez
A
,
Oñate
C
,
Andrés-Tovar
A
,
Garzón-Tituaña
M
,
Uranga-Murillo
I
,
Arias
MA
, et al.
All about (NK cell-mediated) death in two acts and an unexpected encore: initiation, execution and activation of adaptive immunity
.
Front Immunol
.
2022
:
13
:
896228
. https://doi.org/10.3389/fimmu.2022.896228

152

Feehan
DD
,
Jamil
K
,
Polyak
MJ
,
Ogbomo
H
,
Hasell
M
,
Li
SS
,
Xiang
RF
,
Parkins
M
,
Trapani
JA
,
Harrison
JJ
, et al.
Natural killer cells kill extracellular Pseudomonas aeruginosa using contact-dependent release of granzymes B and H
.
PLoS Pathog
.
2022
:
18
(
2
):
e1010325
. https://doi.org/10.1371/journal.ppat.1010325

153

Sankar
J
,
Arora
S
,
Joshi
G
,
Kumar
R
.
Pore-forming proteins and their role in cancer and inflammation: mechanistic insights and plausible druggable targets
.
Chem Biol Interact
.
2022
:
366
:
110127
. https://doi.org/10.1016/j.cbi.2022.110127

154

Nussbaum
JC
,
Van Dyken
SJ
,
von Moltke
J
,
Cheng
LE
,
Mohapatra
A
,
Molofsky
AB
,
Thornton
EE
,
Krummel
MF
,
Chawla
A
,
Liang
H-E
, et al.
Type 2 innate lymphoid cells control eosinophil homeostasis
.
Nature
.
2013
:
502
(
7470
):
245
248
. https://doi.org/10.1038/nature12526

155

Fauriat
C
,
Long
EO
,
Ljunggren
HG
,
Bryceson
YT
.
Regulation of human NK-cell cytokine and chemokine production by target cell recognition
.
Blood
.
2010
:
115
(
11
):
2167
2176
. https://doi.org/10.1182/blood-2009-08-238469

156

Caligiuri
MA
.
Human natural killer cells
.
Blood
.
2008
:
112
(
3
):
461
469
. https://doi.org/10.1182/blood-2007-09-077438

157

Zhou
N
,
Yue
Y
,
Xiong
S
.
Sex hormone contributes to sexually dimorphic susceptibility in CVB3-induced viral myocarditis via modulating IFN-γ(+) NK cell production
.
Can J Cardiol
.
2018
:
34
(
4
):
492
501
. https://doi.org/10.1016/j.cjca.2018.01.002

158

Loebel
M
,
Holzhauser
L
,
Hartwig
JA
,
Shukla
PC
,
Savvatis
K
,
Jenke
A
,
Gast
M
,
Escher
F
,
Becker
SC
,
Bauer
S
, et al.
The forkhead transcription factor foxo3 negatively regulates natural killer cell function and viral clearance in myocarditis
.
Eur Heart J
.
2018
:
39
(
10
):
876
887
. https://doi.org/10.1093/eurheartj/ehx624

159

Ong
S
,
Ligons
DL
,
Barin
JG
,
Wu
L
,
Talor
MV
,
Diny
N
,
Fontes
JA
,
Gebremariam
E
,
Kass
DA
,
Rose
NR
, et al.
Natural killer cells limit cardiac inflammation and fibrosis by halting eosinophil infiltration
.
Am J Pathol
.
2015
:
185
(
3
):
847
861
. https://doi.org/10.1016/j.ajpath.2014.11.023

160

Barnig
C
,
Cernadas
M
,
Dutile
S
,
Liu
X
,
Perrella
MA
,
Kazani
S
,
Wechsler
ME
,
Israel
E
,
Levy
BD
.
Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma
.
Sci Transl Med
.
2013
:
5
(
174
):
174ra26
. https://doi.org/10.1126/scitranslmed.3004812

161

Awad
A
,
Yassine
H
,
Barrier
M
,
Vorng
H
,
Marquillies
P
,
Tsicopoulos
A
,
Duez
C
.
Natural killer cells induce eosinophil activation and apoptosis
.
PLoS One
.
2014
:
9
(
4
):
e94492
. https://doi.org/10.1371/journal.pone.0094492

162

Molofsky
AB
,
Van Gool
F
,
Liang
HE
,
Van Dyken
SJ
,
Nussbaum
JC
,
Lee
J
,
Bluestone
JA
,
Locksley
RM
.
Interleukin-33 and interferon-γ counter-regulate group 2 innate lymphoid cell activation during immune perturbation
.
Immunity
.
2015
:
43
(
1
):
161
174
. https://doi.org/10.1016/j.immuni.2015.05.019

163

Wei
Y
,
Bingyu
W
,
Lei
Y
,
Xingxing
Y
.
The antifibrotic role of natural killer cells in liver fibrosis
.
Exp Biol Med (Maywood)
.
2022
:
247
(
14
):
1235
1243
. https://doi.org/10.1177/15353702221092672

164

Muhanna
N
,
Abu Tair
L
,
Doron
S
,
Amer
J
,
Azzeh
M
,
Mahamid
M
,
Friedman
S
,
Safadi
R
.
Amelioration of hepatic fibrosis by NK cell activation
.
Gut
.
2011
:
60
(
1
):
90
98
. https://doi.org/10.1136/gut.2010.211136

165

Hintermann
E
,
Bayer
M
,
Pfeilschifter
JM
,
Luster
AD
,
Christen
U
.
CXCL10 Promotes liver fibrosis by prevention of NK cell mediated hepatic stellate cell inactivation
.
J Autoimmun
.
2010
:
35
(
4
):
424
435
. https://doi.org/10.1016/j.jaut.2010.09.003

166

Weizman
OE
,
Adams
NM
,
Schuster
IS
,
Krishna
C
,
Pritykin
Y
,
Lau
C
,
Degli-Esposti
MA
,
Leslie
CS
,
Sun
JC
,
O'Sullivan
TE
.
ILC1 confer early host protection at initial sites of viral infection
.
Cell
.
2017
:
171
(
4
):
795
808.e12
. https://doi.org/10.1016/j.cell.2017.09.052

167

Spits
H
,
Bernink
JH
,
Lanier
L
.
NK Cells and type 1 innate lymphoid cells: partners in host defense
.
Nat Immunol
.
2016
:
17
(
7
):
758
764
. https://doi.org/10.1038/ni.3482

168

Jia
Y
,
Fang
X
,
Zhu
X
,
Bai
C
,
Zhu
L
,
Jin
M
,
Wang
X
,
Hu
M
,
Tang
R
,
Chen
Z
.
IL-13(+) type 2 innate lymphoid cells correlate with asthma control status and treatment response
.
Am J Respir Cell Mol Biol
.
2016
:
55
(
5
):
675
683
. https://doi.org/10.1165/rcmb.2016-0099OC

169

Melo-Gonzalez
F
,
Hepworth
MR
.
Functional and phenotypic heterogeneity of group 3 innate lymphoid cells
.
Immunology
.
2017
:
150
(
3
):
265
275
. https://doi.org/10.1111/imm.12697

170

Bracamonte-Baran
W
,
Chen
G
,
Hou
X
,
Talor
MV
,
Choi
HS
,
Davogustto
G
,
Taegtmeyer
H
,
Sung
J
,
Hackam
DJ
,
Nauen
D
, et al.
Non-cytotoxic cardiac innate lymphoid cells are a resident and quiescent type 2-commited population
.
Front Immunol
.
2019
:
10
:
634
. https://doi.org/10.3389/fimmu.2019.00634

171

Serbina
NV
,
Jia
T
,
Hohl
TM
,
Pamer
EG
.
Monocyte-mediated defense against microbial pathogens
.
Annu Rev Immunol
.
2008
:
26
(
1
):
421
452
. https://doi.org/10.1146/annurev.immunol.26.021607.090326

172

Ożańska
A
,
Szymczak
D
,
Rybka
J
.
Pattern of human monocyte subpopulations in health and disease
.
Scand J Immunol
.
2020
:
92
(
1
):
e12883
. https://doi.org/10.1111/sji.12883

173

Ziegler-Heitbrock
L
.
The CD14+ CD16 + blood monocytes: their role in infection and inflammation
.
J Leukoc Biol
.
2007
:
81
(
3
):
584
592
. https://doi.org/10.1189/jlb.0806510

174

Hou
X
,
Chen
G
,
Bracamonte-Baran
W
,
Choi
HS
,
Diny
NL
,
Sung
J
,
Hughes
D
,
Won
T
,
Wood
MK
,
Talor
MV
, et al.
The cardiac microenvironment instructs divergent monocyte fates and functions in myocarditis
.
Cell Rep
.
2019
:
28
(
1
):
172
89.e7
. https://doi.org/10.1016/j.celrep.2019.06.007

175

Cros
J
,
Cagnard
N
,
Woollard
K
,
Patey
N
,
Zhang
SY
,
Senechal
B
,
Puel
A
,
Biswas
SK
,
Moshous
D
,
Picard
C
, et al.
Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors
.
Immunity
.
2010
:
33
(
3
):
375
386
. https://doi.org/10.1016/j.immuni.2010.08.012

176

Belge
KU
,
Dayyani
F
,
Horelt
A
,
Siedlar
M
,
Frankenberger
M
,
Frankenberger
B
,
Espevik
T
,
Ziegler-Heitbrock
L
.
The proinflammatory CD14 + CD16 + DR++ monocytes are a major source of TNF
.
J Immunol
.
2002
:
168
(
7
):
3536
3542
. https://doi.org/10.4049/jimmunol.168.7.3536

177

Jakubzick
CV
,
Randolph
GJ
,
Henson
PM
.
Monocyte differentiation and antigen-presenting functions
.
Nat Rev Immunol
.
2017
:
17
(
6
):
349
362
. https://doi.org/10.1038/nri.2017.28

178

Palani
S
,
Elima
K
,
Ekholm
E
,
Jalkanen
S
,
Salmi
M
.
Monocyte stabilin-1 suppresses the activation of th1 lymphocytes
.
J Immunol
.
2016
:
196
(
1
):
115
123
. https://doi.org/10.4049/jimmunol.1500257

179

Ko
HJ
,
Brady
JL
,
Ryg-Cornejo
V
,
Hansen
DS
,
Vremec
D
,
Shortman
K
,
Zhan
Y
,
Lew
AM
.
GM-CSF-responsive monocyte-derived dendritic cells are pivotal in Th17 pathogenesis
.
J Immunol
.
2014
:
192
(
5
):
2202
2209
. https://doi.org/10.4049/jimmunol.1302040

180

Leuschner
F
,
Courties
G
,
Dutta
P
,
Mortensen
LJ
,
Gorbatov
R
,
Sena
B
,
Novobrantseva
TI
,
Borodovsky
A
,
Fitzgerald
K
,
Koteliansky
V
, et al.
Silencing of CCR2 in myocarditis
.
Eur Heart J
.
2015
:
36
(
23
):
1478
1488
. https://doi.org/10.1093/eurheartj/ehu225

181

Carai
P
,
Papageorgiou
AP
,
Van Linthout
S
,
Deckx
S
,
Velthuis
S
,
Lutgens
E
,
Wijnands
E
,
Tschöpe
C
,
Schmuttermaier
C
,
Kzhyshkowska
J
, et al.
Stabilin-1 mediates beneficial monocyte recruitment and tolerogenic macrophage programming during CVB3-induced viral myocarditis
.
J Mol Cell Cardiol
.
2022
:
165
:
31
39
. https://doi.org/10.1016/j.yjmcc.2021.12.009

182

Hwang
N
,
Huh
Y
,
Bu
S
,
Seo
KJ
,
Kwon
SH
,
Kim
JW
,
Yoon
BK
,
Ahn
H-S
,
Fang
S
.
Single-cell sequencing of PBMC characterizes the altered transcriptomic landscape of classical monocytes in BNT162b2-induced myocarditis
.
Front Immunol
.
2022
:
13
:
979188
. https://doi.org/10.3389/fimmu.2022.979188

183

Jimenez
J
,
Lavine
KJ
.
The dynamic role of cardiac macrophages in aging and disease
.
Curr Cardiol Rep
.
2022
:
24
(
8
):
925
933
. https://doi.org/10.1007/s11886-022-01714-4

184

Jaquenod De Giusti
C
,
Ure
AE
,
Rivadeneyra
L
,
Schattner
M
,
Gomez
RM
.
Macrophages and galectin 3 play critical roles in CVB3-induced murine acute myocarditis and chronic fibrosis
.
J Mol Cell Cardiol
.
2015
:
85
:
58
70
. https://doi.org/10.1016/j.yjmcc.2015.05.010

185

Shapouri-Moghaddam
A
,
Mohammadian
S
,
Vazini
H
,
Taghadosi
M
,
Esmaeili
SA
,
Mardani
F
,
Seifi
B
,
Mohammadi
A
,
Afshari
JT
,
Sahebkar
A
.
Macrophage plasticity, polarization, and function in health and disease
.
J Cell Physiol
.
2018
:
233
(
9
):
6425
6440
. https://doi.org/10.1002/jcp.26429

186

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
. https://doi.org/10.1016/j.yjmcc.2016.12.010

187

Wang
Y
,
Li
M
,
Chen
J
,
Yu
Y
,
Yu
Y
,
Shi
H
,
Liu
X
,
Chen
Z
,
Chen
R
,
Ge
J
.
Macrophage CAPN4 regulates CVB3-induced cardiac inflammation and injury by promoting NLRP3 inflammasome activation and phenotypic transformation to the inflammatory subtype
.
Free Radic Biol Med
.
2023
:
208
:
430
444
. https://doi.org/10.1016/j.freeradbiomed.2023.08.032

188

Shin
HH
,
Jeon
ES
,
Lim
BK
.
Macrophage-specific coxsackievirus and adenovirus receptor deletion enhances macrophage M1 polarity in CVB3-induced myocarditis
.
Int J Mol Sci
.
2023
:
24
(
6
):
5309
. https://doi.org/10.3390/ijms24065309

189

Cihakova
D
,
Barin
JG
,
Afanasyeva
M
,
Kimura
M
,
Fairweather
D
,
Berg
M
,
Talor
MV
,
Baldeviano
GC
,
Frisancho
S
,
Gabrielson
K
, et al.
Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation
.
Am J Pathol
.
2008
:
172
(
5
):
1195
1208
. https://doi.org/10.2353/ajpath.2008.070207

190

Ponce
NE
,
Sanmarco
LM
,
Eberhardt
N
,
García
MC
,
Rivarola
HW
,
Cano
RC
,
Aoki
MP
.
CD73 Inhibition shifts cardiac macrophage polarization toward a microbicidal phenotype and ameliorates the outcome of experimental chagas cardiomyopathy
.
J Immunol
.
2016
:
197
(
3
):
814
823
. https://doi.org/10.4049/jimmunol.1600371

191

Amoah
BP
,
Yang
H
,
Zhang
P
,
Su
Z
,
Xu
H
.
Immunopathogenesis of myocarditis: the interplay between cardiac fibroblast cells, dendritic cells, macrophages and CD4+ T cells
.
Scand J Immunol
.
2015
:
82
(
1
):
1
9
. https://doi.org/10.1111/sji.12298

192

Bajpai
G
,
Schneider
C
,
Wong
N
,
Bredemeyer
A
,
Hulsmans
M
,
Nahrendorf
M
,
Epelman
S
,
Kreisel
D
,
Liu
Y
,
Itoh
A
, et al.
The human heart contains distinct macrophage subsets with divergent origins and functions
.
Nat Med
.
2018
:
24
(
8
):
1234
1245
. https://doi.org/10.1038/s41591-018-0059-x

193

Nicolás-Ávila
JA
,
Lechuga-Vieco
AV
,
Esteban-Martínez
L
,
Sánchez-Díaz
M
,
Díaz-García
E
,
Santiago
DJ
,
Rubio-Ponce
A
,
Li
JLY
,
Balachander
A
,
Quintana
JA
, et al.
A network of macrophages supports mitochondrial homeostasis in the heart
.
Cell
.
2020
:
183
(
1
):
94
109.e23
. https://doi.org/10.1016/j.cell.2020.08.031

194

Müller
I
,
Pappritz
K
,
Savvatis
K
,
Puhl
K
,
Dong
F
,
El-Shafeey
M
,
Hamdani
N
,
Hamann
I
,
Noutsias
M
,
Infante-Duarte
C
, et al.
CX3CR1 Knockout aggravates coxsackievirus B3-induced myocarditis
.
PLoS One
.
2017
:
12
(
8
):
e0182643
. https://doi.org/10.1371/journal.pone.0182643

195

Mosser
DM
,
Hamidzadeh
K
,
Goncalves
R
.
Macrophages and the maintenance of homeostasis
.
Cell Mol Immunol
.
2021
:
18
(
3
):
579
587
. https://doi.org/10.1038/s41423-020-00541-3

196

Hulsmans
M
,
Clauss
S
,
Xiao
L
,
Aguirre
AD
,
King
KR
,
Hanley
A
,
Hucker
WJ
,
Wülfers
EM
,
Seemann
G
,
Courties
G
, et al.
Macrophages facilitate electrical conduction in the heart
.
Cell
.
2017
:
169
(
3
):
510
22.e20
. https://doi.org/10.1016/j.cell.2017.03.050

197

Matheis
F
,
Muller
PA
,
Graves
CL
,
Gabanyi
I
,
Kerner
ZJ
,
Costa-Borges
D
,
Ahrends
T
,
Rosenstiel
P
,
Mucida
D
.
Adrenergic signaling in muscularis macrophages limits infection-induced neuronal loss
.
Cell
.
2020
:
180
(
1
):
64
78.e16
. https://doi.org/10.1016/j.cell.2019.12.002

198

Hart
DN
,
Fabre
JW
.
Demonstration and characterization of ia-positive dendritic cells in the interstitial connective tissues of rat heart and other tissues, but not brain
.
J Exp Med
.
1981
:
154
(
2
):
347
361
. https://doi.org/10.1084/jem.154.2.347

199

Satpathy
AT
,
Kc
W
,
Albring
JC
,
Edelson
BT
,
Kretzer
NM
,
Bhattacharya
D
,
Murphy
TL
,
Murphy
KM
.
Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages
.
J Exp Med
.
2012
:
209
(
6
):
1135
1152
. https://doi.org/10.1084/jem.20120030

200

Hilligan
KL
,
Ronchese
F
.
Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses
.
Cell Mol Immunol
.
2020
:
17
(
6
):
587
599
. https://doi.org/10.1038/s41423-020-0465-0

201

Steinman
RM
.
Decisions about dendritic cells: past, present, and future
.
Annu Rev Immunol
.
2012
:
30
(
1
):
1
22
. https://doi.org/10.1146/annurev-immunol-100311-102839

202

Ganguly
D
,
Haak
S
,
Sisirak
V
,
Reizis
B
.
The role of dendritic cells in autoimmunity
.
Nat Rev Immunol
.
2013
:
13
(
8
):
566
577
. https://doi.org/10.1038/nri3477

203

Macri
C
,
Pang
ES
,
Patton
T
,
O'Keeffe
M
.
Dendritic cell subsets
.
Semin Cell Dev Biol
.
2018
:
84
:
11
21
. https://doi.org/10.1016/j.semcdb.2017.12.009

204

Van der Borght
K
,
Lambrecht
BN
.
Heart macrophages and dendritic cells in sickness and in health: a tale of a complicated marriage
.
Cell Immunol
.
2018
:
330
:
105
113
. https://doi.org/10.1016/j.cellimm.2018.03.011

205

Pistulli
R
,
Andreas
E
,
König
S
,
Drobnik
S
,
Kretzschmar
D
,
Rohm
I
,
Lichtenauer
M
,
Heidecker
B
,
Franz
M
,
Mall
G
, et al.
Characterization of dendritic cells in human and experimental myocarditis
.
ESC Heart Fail
.
2020
:
7
(
5
):
2305
2317
. https://doi.org/10.1002/ehf2.12767

206

Clemente-Casares
X
,
Hosseinzadeh
S
,
Barbu
I
,
Dick
SA
,
Macklin
JA
,
Wang
Y
,
Momen
A
,
Kantores
C
,
Aronoff
L
,
Farno
M
, et al.
A CD103(+) conventional dendritic cell surveillance system prevents development of overt heart failure during subclinical viral myocarditis
.
Immunity
.
2017
:
47
(
5
):
974
89.e8
. https://doi.org/10.1016/j.immuni.2017.10.011

207

Rahnefeld
A
,
Ebstein
F
,
Albrecht
N
,
Opitz
E
,
Kuckelkorn
U
,
Stangl
K
,
Rehm
A
,
Kloetzel
PM
,
Voigt
A
.
Antigen-presentation capacity of dendritic cells is impaired in ongoing enterovirus myocarditis
.
Eur J Immunol
.
2011
:
41
(
9
):
2774
2781
. https://doi.org/10.1002/eji.201041039

208

Guerra-de-Blas
PDC
,
Cruz-González
D
,
Martínez-Shio
EB
,
González-Amaro
R
,
González-Pacheco
H
,
Layseca-Espinosa
E
,
Escobedo-Uribe
CD
,
Monsiváis-Urenda
AE
.
Altered phenotype of circulating dendritic cells and regulatory T cells from patients with acute myocarditis
.
J Immunol Res
.
2022
:
2022
:
8873146
. https://doi.org/10.1155/2022/8873146

209

Chen
P
,
Chen
R
,
Yang
Y
,
Yu
Y
,
Xie
Y
,
Zou
Y
,
Ge
J
,
Chen
H
.
Coxsackievirus B3 infection promotes generation of myeloid dendritic cells from bone marrow and accumulation in the myocardium
.
Int Immunopharmacol
.
2009
:
9
(
11
):
1304
1312
. https://doi.org/10.1016/j.intimp.2009.07.014

210

Eriksson
U
,
Kurrer
MO
,
Sonderegger
I
,
Iezzi
G
,
Tafuri
A
,
Hunziker
L
,
Suzuki
S
,
Bachmaier
K
,
Bingisser
RM
,
Penninger
JM
, et al.
Activation of dendritic cells through the interleukin 1 receptor 1 is critical for the induction of autoimmune myocarditis
.
J Exp Med
.
2003
:
197
(
3
):
323
331
. https://doi.org/10.1084/jem.20021788

211

Sonderegger
I
,
Iezzi
G
,
Maier
R
,
Schmitz
N
,
Kurrer
M
,
Kopf
M
.
GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival
.
J Exp Med
.
2008
:
205
(
10
):
2281
2294
. https://doi.org/10.1084/jem.20071119

212

Van der Borght
K
,
Scott
CL
,
Martens
L
,
Sichien
D
,
Van Isterdael
G
,
Nindl
V
,
Saeys
Y
,
Boon
L
,
Ludewig
B
,
Gillebert
TC
, et al.
Myocarditis elicits dendritic cell and monocyte infiltration in the heart and self-antigen presentation by conventional type 2 dendritic cells
.
Front Immunol
.
2018
:
9
:
2714
. https://doi.org/10.3389/fimmu.2018.02714

213

Eriksson
U
,
Ricci
R
,
Hunziker
L
,
Kurrer
MO
,
Oudit
GY
,
Watts
TH
,
Sonderegger
I
,
Bachmaier
K
,
Kopf
M
,
Penninger
JM
.
Dendritic cell-induced autoimmune heart failure requires cooperation between adaptive and innate immunity
.
Nat Med
.
2003
:
9
(
12
):
1484
1490
. https://doi.org/10.1038/nm960

214

Tajiri
K
,
Yonebayashi
S
,
Li
S
,
Ieda
M
.
Immunomodulatory role of tenascin-C in myocarditis and inflammatory cardiomyopathy
.
Front Immunol
.
2021
:
12
:
624703
. https://doi.org/10.3389/fimmu.2021.624703

215

Ness
S
,
Lin
S
,
Gordon
JR
.
Regulatory dendritic cells, T cell tolerance, and dendritic cell therapy for immunologic disease
.
Front Immunol
.
2021
:
12
:
633436
. https://doi.org/10.3389/fimmu.2021.633436

216

Li
X
,
Yang
A
,
Huang
H
,
Zhang
X
,
Town
J
,
Davis
B
, et al.
Induction of type 2T helper cell allergen tolerance by IL-10-differentiated regulatory dendritic cells
.
Am J Respir Cell Mol Biol
.
2010
:
42
(
2
):
190
199
. https://doi.org/10.1165/rcmb.2009-0023OC

217

Li
S
,
Tajiri
K
,
Murakoshi
N
,
Xu
D
,
Yonebayashi
S
,
Okabe
Y
,
Yuan
Z
,
Feng
D
,
Inoue
K
,
Aonuma
K
, et al.
Programmed death-ligand 2 deficiency exacerbates experimental autoimmune myocarditis in mice
.
Int J Mol Sci
.
2021
:
22
(
3
):
1426
. https://doi.org/10.3390/ijms22031426

218

Gregori
S
,
Tomasoni
D
,
Pacciani
V
,
Scirpoli
M
,
Battaglia
M
,
Magnani
CF
,
Hauben
E
,
Roncarolo
M-G
.
Differentiation of type 1T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway
.
Blood
.
2010
:
116
(
6
):
935
944
. https://doi.org/10.1182/blood-2009-07-234872

219

Takenaka
MC
,
Quintana
FJ
.
Tolerogenic dendritic cells
.
Semin Immunopathol
.
2017
:
39
(
2
):
113
120
. https://doi.org/10.1007/s00281-016-0587-8

220

Zhang
M
,
Zheng
Y
,
Sun
Y
,
Li
S
,
Chen
L
,
Jin
X
,
Hou
X
,
Liu
X
,
Chen
Q
,
Li
J
, et al.
Knockdown of NEAT1 induces tolerogenic phenotype in dendritic cells by inhibiting activation of NLRP3 inflammasome
.
Theranostics
.
2019
:
9
(
12
):
3425
3442
. https://doi.org/10.7150/thno.33178

221

Yang
S
,
Li
W
,
Liu
W
,
Gao
C
,
Zhou
B
,
Li
S
,
Li
Y
,
Kong
Y
.
IL-10 gene modified dendritic cells induced antigen-specific tolerance in experimental autoimmune myocarditis
.
Clin Immunol
.
2006
:
121
(
1
):
63
73
. https://doi.org/10.1016/j.clim.2006.06.009

222

Li
WM
,
Liu
W
,
Gao
C
,
Zhou
BG
,
Yang
SS
,
Wang
Z
,
Zhang
R-h
,
Gan
R-t
,
Kong
Y-h
,
LI
Y
, et al.
Antigen-specific tolerance induced by IL-10 gene modified immature dendritic cells in experimental autoimmune myocarditis in rats
.
Chin Med J (Engl)
.
2006
:
119
(
19
):
1646
1652
. https://doi.org/10.1097/00029330-200610010-00009

223

Lee
JH
,
Kim
TH
,
Park
HE
,
Lee
EG
,
Jung
NC
,
Song
JY
,
Seo
HG
,
Seung
K-B
,
Chang
K
,
Lim
D-S
.
Myosin-primed tolerogenic dendritic cells ameliorate experimental autoimmune myocarditis
.
Cardiovasc Res
.
2014
:
101
(
2
):
203
210
. https://doi.org/10.1093/cvr/cvt246

224

Wu
J
,
Liu
M
,
Mang
G
,
Yu
S
,
Chen
Q
,
Li
T
,
Wang
Y
,
Meng
Y
,
Tang
X
,
Zheng
Y
, et al.
Protosappanin A protects against experimental autoimmune myocarditis, and induces metabolically reprogrammed tolerogenic DCs
.
Pharmacol Res
.
2019
:
146
:
104269
. https://doi.org/10.1016/j.phrs.2019.104269

225

Villar
J
,
Segura
E
.
Decoding the heterogeneity of human dendritic cell subsets
.
Trends Immunol
.
2020
:
41
(
12
):
1062
1071
. https://doi.org/10.1016/j.it.2020.10.002

226

Zheng
SY
,
Dong
JZ
.
Role of toll-like receptors and th responses in viral myocarditis
.
Front Immunol
.
2022
:
13
:
843891
. https://doi.org/10.3389/fimmu.2022.843891

227

Eriksson
U
,
Kurrer
MO
,
Bingisser
R
,
Eugster
HP
,
Saremaslani
P
,
Follath
F
,
Marsch
S
,
Widmer
U
.
Lethal autoimmune myocarditis in interferon-gamma receptor-deficient mice: enhanced disease severity by impaired inducible nitric oxide synthase induction
.
Circulation
.
2001
:
103
(
1
):
18
21
. https://doi.org/10.1161/01.CIR.103.1.18

228

Nindl
V
,
Maier
R
,
Ratering
D
,
De Giuli
R
,
Züst
R
,
Thiel
V
,
Scandella
E
,
Di Padova
F
,
Kopf
M
,
Rudin
M
, et al.
Cooperation of Th1 and Th17 cells determines transition from autoimmune myocarditis to dilated cardiomyopathy
.
Eur J Immunol
.
2012
:
42
(
9
):
2311
2321
. https://doi.org/10.1002/eji.201142209

229

Shioi
T
,
Matsumori
A
,
Sasayama
S
.
Persistent expression of cytokine in the chronic stage of viral myocarditis in mice
.
Circulation
.
1996
:
94
(
11
):
2930
2937
. https://doi.org/10.1161/01.CIR.94.11.2930

230

da Silva
MV
,
de Almeida
VL
,
de Oliveira
WD
,
Matos Cascudo
NC
,
de Oliveira
PG
,
da Silva
CA
,
da Silva
ACS
,
Dos Reis Monteiro
MLG
,
Correa
RRM
, et al.
Upregulation of cardiac IL-10 and downregulation of IFN-γ in Balb/c IL-4(-/-) in acute chagasic myocarditis due to Colombian strain of trypanosoma cruzi
.
Mediators Inflamm
.
2018
;
2018
:
3421897
. https://doi.org/10.1155/2018/3421897

231

Michailowsky
V
,
Silva
NM
,
Rocha
CD
,
Vieira
LQ
,
Lannes-Vieira
J
,
Gazzinelli
RT
.
Pivotal role of interleukin-12 and interferon-gamma axis in controlling tissue parasitism and inflammation in the heart and central nervous system during trypanosoma cruzi infection
.
Am J Pathol
.
2001
:
159
(
5
):
1723
1733
. https://doi.org/10.1016/S0002-9440(10)63019-2

232

Song
J
,
Chen
X
,
Cheng
L
,
Rao
M
,
Chen
K
,
Zhang
N
,
Meng
J
,
Li
M
,
Liu
Z-Q
,
Yang
P-C
.
Vitamin D receptor restricts T helper 2-biased inflammation in the heart
.
Cardiovasc Res
.
2018
:
114
(
6
):
870
879
. https://doi.org/10.1093/cvr/cvy034

233

Afanasyeva
M
,
Wang
Y
,
Kaya
Z
,
Park
S
,
Zilliox
MJ
,
Schofield
BH
,
Hill
SL
,
Rose
NR
.
Experimental autoimmune myocarditis in A/J mice is an interleukin-4-dependent disease with a Th2 phenotype
.
Am J Pathol
.
2001
:
159
(
1
):
193
203
. https://doi.org/10.1016/S0002-9440(10)61685-9

234

Chen
X
,
Zeng
XH
,
Wang
M
,
Chen
L
,
Zhang
N
,
Rao
M
,
Yang
P-C
,
Song
J
.
Bcl2-like protein 12 is required for the aberrant T helper-2 polarization in the heart by enhancing interleukin-4 expression and compromising apoptotic machinery in CD4+ T cells
.
Circulation
.
2018
:
138
(
22
):
2559
2568
. https://doi.org/10.1161/CIRCULATIONAHA.118.033890

235

Futamatsu
H
,
Suzuki
J
,
Mizuno
S
,
Koga
N
,
Adachi
S
,
Kosuge
H
,
Maejima
Y
,
Hirao
K
,
Nakamura
T
,
Isobe
M
.
Hepatocyte growth factor ameliorates the progression of experimental autoimmune myocarditis: a potential role for induction of T helper 2 cytokines
.
Circ Res
.
2005
:
96
(
8
):
823
830
. https://doi.org/10.1161/01.RES.0000163016.52653.2e

236

Long
Q
,
Liao
YH
,
Xie
Y
,
Liang
W
,
Cheng
X
,
Yuan
J
,
Yu
M
.
Coxsackievirus B3 directly induced Th17 cell differentiation by inhibiting Nup98 expression in patients with acute viral myocarditis
.
Front Cell Infect Microbiol
.
2016
:
6
:
171
. https://doi.org/10.3389/fcimb.2016.00171

237

Chen
J
,
Yang
F
,
Shi
S
,
Liu
X
,
Qin
F
,
Wei
X
,
Huang
Y
,
Liang
W
,
Miao
L
.
The severity of CVB3-induced myocarditis can be improved by blocking the orchestration of NLRP3 and Th17 in Balb/c mice
.
Mediators Inflamm
.
2021
:
2021
:
5551578
. https://doi.org/10.1155/2021/5551578

238

Liang
W
,
Xie
BK
,
Ding
PW
,
Wang
M
,
Yuan
J
,
Cheng
X
,
Liao
Y-H
,
Yu
M
.
Sacubitril/valsartan alleviates experimental autoimmune myocarditis by inhibiting Th17 cell differentiation independently of the NLRP3 inflammasome pathway
.
Front Pharmacol
.
2021
:
12
:
727838
. https://doi.org/10.3389/fphar.2021.727838

239

Yuan
J
,
Yu
M
,
Lin
QW
,
Cao
AL
,
Yu
X
,
Dong
JH
,
Wang
J-P
,
Zhang
J-H
,
Wang
M
,
Guo
H-P
, et al.
Neutralization of IL-17 inhibits the production of anti-ANT autoantibodies in CVB3-induced acute viral myocarditis
.
Int Immunopharmacol
.
2010
:
10
(
3
):
272
276
. https://doi.org/10.1016/j.intimp.2009.11.010

240

Gergely
TG
,
Kucsera
D
,
Tóth
VE
,
Kovács
T
,
Sayour
NV
,
Drobni
ZD
,
Ruppert
M
,
Petrovich
B
,
Ágg
B
,
Onódi
Z
, et al.
Characterization of immune checkpoint inhibitor-induced cardiotoxicity reveals interleukin-17A as a driver of cardiac dysfunction after anti-PD-1 treatment
.
Br J Pharmacol
.
2023
:
180
(
6
):
740
761
. https://doi.org/10.1111/bph.15984

241

Sonderegger
I
,
Röhn
TA
,
Kurrer
MO
,
Iezzi
G
,
Zou
Y
,
Kastelein
RA
,
Bachmann
MF
,
Kopf
M
.
Neutralization of IL-17 by active vaccination inhibits IL-23-dependent autoimmune myocarditis
.
Eur J Immunol
.
2006
:
36
(
11
):
2849
2856
. https://doi.org/10.1002/eji.200636484

242

Cruz-Adalia
A
,
Jiménez-Borreguero
LJ
,
Ramírez-Huesca
M
,
Chico-Calero
I
,
Barreiro
O
,
López-Conesa
E
,
Fresno
M
,
Sánchez-Madrid
F
,
Martín
P
.
CD69 Limits the severity of cardiomyopathy after autoimmune myocarditis
.
Circulation
.
2010
:
122
(
14
):
1396
1404
. https://doi.org/10.1161/CIRCULATIONAHA.110.952820

243

Baldeviano
GC
,
Barin
JG
,
Talor
MV
,
Srinivasan
S
,
Bedja
D
,
Zheng
D
,
Gabrielson
K
,
Iwakura
Y
,
Rose
NR
,
Cihakova
D
.
Interleukin-17A is dispensable for myocarditis but essential for the progression to dilated cardiomyopathy
.
Circ Res
.
2010
:
106
(
10
):
1646
1655
. https://doi.org/10.1161/CIRCRESAHA.109.213157

244

Lu
Y
,
Zhao
N
,
Wu
Y
,
Yang
S
,
Wu
Q
,
Dong
Q
,
Du
Y
.
Inhibition of PGK1 attenuates autoimmune myocarditis by reprogramming CD4+ T cells metabolism
.
Cardiovasc Res
.
2023:
119
(
6
):
1377
1389
. https://doi.org/10.1093/cvr/cvad029

245

Myers
JM
,
Cooper
LT
,
Kem
DC
,
Stavrakis
S
,
Kosanke
SD
,
Shevach
EM
,
Fairweather
D
,
Stoner
JA
,
Cox
CJ
,
Cunningham
MW
.
Cardiac myosin-Th17 responses promote heart failure in human myocarditis
.
JCI Insight
.
2016
:
1
(
9
):
e85851
. https://doi.org/10.1172/jci.insight.85851

246

Ono
M
,
Shimizu
J
,
Miyachi
Y
,
Sakaguchi
S
.
Control of autoimmune myocarditis and multiorgan inflammation by glucocorticoid-induced TNF receptor family-related protein(high), Foxp3-expressing CD25+ and CD25- regulatory T cells
.
J Immunol
.
2006
:
176
(
8
):
4748
4756
. https://doi.org/10.4049/jimmunol.176.8.4748

247

Shi
Y
,
Fukuoka
M
,
Li
G
,
Liu
Y
,
Chen
M
,
Konviser
M
,
Chen
X
,
Opavsky
MA
,
Liu
PP
.
Regulatory T cells protect mice against coxsackievirus-induced myocarditis through the transforming growth factor beta-coxsackie-adenovirus receptor pathway
.
Circulation
.
2010
:
121
(
24
):
2624
2634
. https://doi.org/10.1161/CIRCULATIONAHA.109.893248

248

Cao
Y
,
Xu
W
,
Xiong
S
.
Adoptive transfer of regulatory T cells protects against coxsackievirus B3-induced cardiac fibrosis
.
PLoS One
.
2013
:
8
(
9
):
e74955
. https://doi.org/10.1371/journal.pone.0074955

249

Akhmerov
A
,
Rogers
R
,
de Couto
G
,
Valle
J
,
Li
L
,
Ibrahim
A
,
Sanchez
L
,
Zhang
R
,
Lin
Y-N
,
Liu
W
, et al.
Regulatory T cell activation, proliferation, and reprogramming induced by extracellular vesicles
.
J Heart Lung Transplant
.
2021
:
40
(
11
):
1387
1395
. https://doi.org/10.1016/j.healun.2021.06.005

250

Shao
PP
,
Liu
CJ
,
Xu
Q
,
Zhang
B
,
Li
SH
,
Wu
Y
,
Sun
Z
,
Cheng
L-F
.
Eplerenone reverses cardiac fibrosis via the suppression of tregs by inhibition of Kv1.3 channel
.
Front Physiol
.
2018
:
9
:
899
. https://doi.org/10.3389/fphys.2018.00899

251

Xie
Y
,
Gong
C
,
Bo
L
,
Jiang
S
,
Kan
H
,
Song
W
,
Zhao
J
,
Li
Y
.
Treg responses are associated with PM2.5-induced exacerbation of viral myocarditis
.
Inhal Toxicol
.
2015
:
27
(
6
):
281
286
. https://doi.org/10.3109/08958378.2015.1040139

252

Li
Y-C
,
Gu
XH
,
Ge
L-S
,
Zhou
DP
,
Xing
C
,
Guo
XL
,
Pan
L-L
,
Song
S-Y
,
Yu
L-L
,
Chen
G-Y
, et al.
Vagus nerve plays a pivotal role in CD4+ T cell differentiation during CVB3-induced murine acute myocarditis
.
Virulence
.
2021
:
12
(
1
):
360
376
. https://doi.org/10.1080/21505594.2020.1869384

253

Barbosa
CD
,
Canto
FB
,
Gomes
A
,
Brandao
LM
,
Lima
JR
,
Melo
GA
,
Granato
A
,
Neves
EG
,
Dutra
WO
,
Oliveira
A-C
, et al.
Cytotoxic CD4(+) T cells driven by T-cell intrinsic IL-18R/MyD88 signaling predominantly infiltrate trypanosoma cruzi-infected hearts
.
Elife
.
2022
:
11
:
e74636
. https://doi.org/10.7554/eLife.74636

254

Qing
K
,
Weifeng
W
,
Fan
Y
,
Yuluan
Y
,
Yu
P
,
Yanlan
H
.
Distinct different expression of Th17 and Th9 cells in coxsackie virus B3-induced mice viral myocarditis
.
Virol J
.
2011
:
8
(
1
):
267
. https://doi.org/10.1186/1743-422X-8-267

255

Lowin
B
,
Hahne
M
,
Mattmann
C
,
Tschopp
J
.
Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways
.
Nature
.
1994
:
370
(
6491
):
650
652
. https://doi.org/10.1038/370650a0

256

Ruby
J
,
Ramshaw
I
.
The antiviral activity of immune CD8+ T cells is dependent on interferon-gamma
.
Lymphokine Cytokine Res
.
1991
:
10
(
5
):
353
358
.

257

Ramsay
AJ
,
Ruby
J
,
Ramshaw
IA
.
A case for cytokines as effector molecules in the resolution of virus infection
.
Immunol Today
.
1993
:
14
(
4
):
155
157
. https://doi.org/10.1016/0167-5699(93)90277-R

258

Klingel
K
,
Schnorr
JJ
,
Sauter
M
,
Szalay
G
,
Kandolf
R
.
beta2-microglobulin-associated regulation of interferon-gamma and virus-specific immunoglobulin G confer resistance against the development of chronic coxsackievirus myocarditis
.
Am J Pathol
.
2003
:
162
(
5
):
1709
1720
. https://doi.org/10.1016/S0002-9440(10)64305-2

259

Bartlett
EJ
,
Lenzo
JC
,
Sivamoorthy
S
,
Mansfield
JP
,
Cull
VS
,
James
CM
.
Type I IFN-beta gene therapy suppresses cardiac CD8+ T-cell infiltration during autoimmune myocarditis
.
Immunol Cell Biol
.
2004
:
82
(
2
):
119
126
. https://doi.org/10.1046/j.0818-9641.2004.01234.x

260

Axelrod
ML
,
Meijers
WC
,
Screever
EM
,
Qin
J
,
Carroll
MG
,
Sun
X
,
Tannous
E
,
Zhang
Y
,
Sugiura
A
,
Taylor
BC
, et al.
T cells specific for α-myosin drive immunotherapy-related myocarditis
.
Nature
.
2022
:
611
(
7937
):
818
826
. https://doi.org/10.1038/s41586-022-05432-3

261

Massilamany
C
,
Gangaplara
A
,
Basavalingappa
RH
,
Rajasekaran
RA
,
Khalilzad-Sharghi
V
,
Han
Z
,
Othman
S
,
Steffen
D
,
R
J
.
Localization of CD8 T cell epitope within cardiac myosin heavy chain-α334-352 that induces autoimmune myocarditis in A/J mice
.
Int J Cardiol
.
2016
:
202
:
311
321
. https://doi.org/10.1016/j.ijcard.2015.09.016

262

Bracamonte-Baran
W
,
Gilotra
NA
,
Won
T
,
Rodriguez
KM
,
Talor
MV
,
Oh
BC
,
Griffin
J
,
Wittstein
I
,
Sharma
K
,
Skinner
J
, et al.
Endothelial stromal PD-L1 (programmed death ligand 1) modulates CD8(+) T-cell infiltration after heart transplantation
.
Circ Heart Fail
.
2021
:
14
(
10
):
e007982
. https://doi.org/10.1161/CIRCHEARTFAILURE.120.007982

263

Komarowska
I
,
Coe
D
,
Wang
G
,
Haas
R
,
Mauro
C
,
Kishore
M
,
Cooper
D
,
Nadkarni
S
,
Fu
H
,
Steinbruchel
DA
, et al.
Hepatocyte growth factor receptor c-Met instructs T cell cardiotropism and promotes T cell migration to the heart via autocrine chemokine release
.
Immunity
.
2015
:
42
(
6
):
1087
1099
. https://doi.org/10.1016/j.immuni.2015.05.014

264

Fanti
S
,
Stephenson
E
,
Rocha-Vieira
E
,
Protonotarios
A
,
Kanoni
S
,
Shahaj
E
,
Longhi
MP
,
Vyas
VS
,
Dyer
C
,
Pontarini
E
, et al.
Circulating c-Met-expressing memory T cells define cardiac autoimmunity
.
Circulation
.
2022
:
146
(
25
):
1930
1945
. https://doi.org/10.1161/CIRCULATIONAHA.121.055610

265

Shiromizu
CM
,
Jancic
CC
.
Γδ T lymphocytes: an effector cell in autoimmunity and infection
.
Front Immunol
.
2018
:
9
:
2389
. https://doi.org/10.3389/fimmu.2018.02389

266

Huber
SA
,
Sartini
D
,
Exley
M
.
Vgamma4(+) T cells promote autoimmune CD8(+) cytolytic T-lymphocyte activation in coxsackievirus B3-induced myocarditis in mice: role for CD4(+) Th1 cells
.
J Virol
.
2002
:
76
(
21
):
10785
10790
. https://doi.org/10.1128/JVI.76.21.10785-10790.2002

267

Huber
SA
.
Depletion of gammadelta+ T cells increases CD4+ FoxP3 (T regulatory) cell response in coxsackievirus B3-induced myocarditis
.
Immunology
.
2009
:
127
(
4
):
567
576
. https://doi.org/10.1111/j.1365-2567.2008.03034.x

268

Huber
S
,
Shi
C
,
Budd
RC
.
Gammadelta T cells promote a Th1 response during coxsackievirus B3 infection in vivo: role of Fas and Fas ligand
.
J Virol
.
2002
:
76
(
13
):
6487
6494
. https://doi.org/10.1128/JVI.76.13.6487-6494.2002

269

Pieper
K
,
Grimbacher
B
,
Eibel
H
.
B-cell biology and development
.
J Allergy Clin Immunol
.
2013
:
131
(
4
):
959
971
. https://doi.org/10.1016/j.jaci.2013.01.046

270

Neu
SD
,
Dittel
BN
.
Characterization of definitive regulatory B cell subsets by cell surface phenotype, function and context
.
Front Immunol
.
2021
:
12
:
787464
. https://doi.org/10.3389/fimmu.2021.787464

271

Mena
I
,
Perry
CM
,
Harkins
S
,
Rodriguez
F
,
Gebhard
J
,
Whitton
JL
.
The role of B lymphocytes in coxsackievirus B3 infection
.
Am J Pathol
.
1999
:
155
(
4
):
1205
1215
. https://doi.org/10.1016/S0002-9440(10)65223-6

272

Jarasch-Althof
N
,
Wiesener
N
,
Schmidtke
M
,
Wutzler
P
,
Henke
A
.
Antibody-dependent enhancement of coxsackievirus B3 infection of primary CD19+ B lymphocytes
.
Viral Immunol
.
2010
:
23
(
4
):
369
376
. https://doi.org/10.1089/vim.2010.0018

273

Lee
WS
,
Wheatley
AK
,
Kent
SJ
,
DeKosky
BJ
.
Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies
.
Nat Microbiol
.
2020
:
5
(
10
):
1185
1191
. https://doi.org/10.1038/s41564-020-00789-5

274

Neumann
DA
,
Rose
NR
,
Ansari
AA
,
Herskowitz
A
.
Induction of multiple heart autoantibodies in mice with coxsackievirus B3- and cardiac myosin-induced autoimmune myocarditis
.
J Immunol
.
1994
:
152
(
1
):
343
350
. https://doi.org/10.4049/jimmunol.152.1.343

275

Esfandiarei
M
,
McManus
BM
.
Molecular biology and pathogenesis of viral myocarditis
.
Annu Rev Pathol
.
2008
:
3
(
1
):
127
155
. https://doi.org/10.1146/annurev.pathmechdis.3.121806.151534

276

Malkiel
S
,
Factor
S
,
Diamond
B
.
Autoimmune myocarditis does not require B cells for antigen presentation
.
J Immunol
.
1999
:
163
(
10
):
5265
5268
. https://doi.org/10.4049/jimmunol.163.10.5265

277

Cen
Z
,
Li
Y
,
Wei
B
,
Wu
W
,
Huang
Y
,
Lu
J
.
The role of B cells in regulation of Th cell differentiation in coxsackievirus B3-induced acute myocarditis
.
Inflammation
.
2021
:
44
(
5
):
1949
1960
. https://doi.org/10.1007/s10753-021-01472-5

278

Wei
B
,
Lu
F
,
Kong
Q
,
Huang
Y
,
Huang
K
,
Wu
W
.
Trehalose induces B cell autophagy to alleviate myocardial injury via the AMPK/ULK1 signalling pathway in acute viral myocarditis induced by coxsackie virus B3
.
Int J Biochem Cell Biol
.
2022
:
146
:
106208
. https://doi.org/10.1016/j.biocel.2022.106208

279

Huang
Y
,
Wei
B
,
Gao
X
,
Deng
Y
,
Wu
W
.
Expression of CD80 and CD86 on B cells during coxsackievirus B3-induced acute myocarditis
.
Cent Eur J Immunol
.
2019
:
44
(
4
):
364
369
. https://doi.org/10.5114/ceji.2019.92786

280

Wei
B
,
Deng
Y
,
Huang
Y
,
Gao
X
,
Wu
W
.
IL-10-producing B cells attenuate cardiac inflammation by regulating Th1 and Th17 cells in acute viral myocarditis induced by coxsackie virus B3
.
Life Sci
.
2019
:
235
:
116838
. https://doi.org/10.1016/j.lfs.2019.116838

281

Lu
J
,
Cen
Z
,
Tang
Q
,
Dong
J
,
Qin
L
,
Wu
W
.
The absence of B cells disrupts splenic and myocardial Treg homeostasis in coxsackievirus B3-induced myocarditis
.
Clin Exp Immunol
.
2022
:
208
(
1
):
1
11
. https://doi.org/10.1093/cei/uxac015

282

Li
Y
,
Huang
Y
,
Wu
W
,
Wei
B
,
Qin
L
.
B cells increase myocardial inflammation by suppressing M2 macrophage polarization in coxsackie virus B3-induced acute myocarditis
.
Inflammation
.
2019
:
42
(
3
):
953
960
. https://doi.org/10.1007/s10753-018-0950-0

283

Cooper
LT
Jr.
Sex and autoimmunity in acute myocarditis: time for a refresh
.
Eur J Heart Fail
.
2022
:
24
(
6
):
1045
1046
. https://doi.org/10.1002/ejhf.2529

Author notes

Conflict of interest The authors declare that they have no conflict of interest.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.