Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 13.
Published in final edited form as: Immunobiology. 2011 Jun 30;217(5):468–475. doi: 10.1016/j.imbio.2011.06.009

Macrophage diversity in cardiac inflammation: a review

Jobert G Barin 1,2, Noel R Rose 2,3, Daniela Čiháková 3
PMCID: PMC4292796  NIHMSID: NIHMS316587  PMID: 21820754

Abstract

Cardiac inflammatory disease represents a significant public health burden, and interesting questions of immunopathologic science and clinical inquiry. Novel insights into the diverse programming and functions within the macrophage lineages in recent years have yielded a view of these cells as dynamic effectors and regulators of immunity, host defense, and inflammatory disease. In this review, we examine and discuss recent investigations into the complex participation of mononuclear phagocytic cells in the pathology of animal models of myocarditis.

Keywords: myocarditis, macrophage, monocyte, autoimmunity

Introduction

Mononuclear phagocytic cells were characterized over a century ago in pioneering studies by Metchnikoff – heralding the advent of our understanding of cell-mediated immunity (1). In the century since, investigations have described a diversity of related cells falling under the banner of “macrophage”, with activities specialized and adapted for effector and regulatory functions in host defense and immunopathologic disease (2). The predominant paradigm for macrophage biology in innate immunity for the past half-century has been the mononuclear phagocyte system (MPS) model, established by van Furth and others (3). The model holds that macrophages in peripheral tissues arise from blood borne precursors, monocytes, which in turn arise from bone marrow precursors that are distinct from lymphocyte precursors. While the model has undergone extensive revision, refinement and complication, the essential notion is still valid (4). In addition to phagocytosis and other microbicidal roles, macrophages are known to also participate in endogenous homeostatic functions, such as scavenging (5).

Macrophage involvement in autoimmune disease, particularly inflammatory autoimmunities, has long been suspected. This review summarizes current understanding of the diverse functions and plasticity of macrophages as applied to addressing these longstanding issues. Current research emphasizes an important role for macrophages in the inflammatory component of cardiovascular diseases. Roles for inflammatory macrophage recruitment have been proposed for hypertensive cardiac remodeling (6), and atherosclerosis (7). Similarly, infectious agents associated with cardiac inflammation elicit macrophage and monocyte invasion of the heart, as reported in Lyme carditis (8), simian immunodeficiency virus carditis (9), human immunodeficiency virus carditis (10, 11), Chagas disease (12), and rheumatic carditis (13). An analysis of autopsy samples from sudden death cases revealed that across several infectious agents associated with myocarditis, macrophage and monocyte infiltration was among the most common findings (14).

Macrophage diversity - T cell- and tumor-derived signals

While commonly considered integral to “innate” host defense, macrophages are also responsive to instructive cues from CD4+ T cell-derived signals. Interferon-gamma (IFNγ) was first identified as a potent macrophage-activating factor, inducing peroxide production and microbicidal activity in human peripheral blood monocyte-derived macrophages (15). Macrophages are still regarded as major effectors of Th1-associated host defenses. IFNγ control of macrophage activation is associated with microbicidal and tumoricidal activity (16, 17) through the upregulation of lysozymal proteolytic enzymes and oxygen radicals (18). Macrophages from IFNγ-deficient mice have been shown to be defective in both antigen-presentation and the production of iNOS-dependent nitric oxide (19-21). Martinez and colleagues have categorically proposed a distinction between these “classically-activated” or M1-type macrophages, and those elicited by purely innate recognition of microbial pathogen-associated molecular patterns (22); this “innate activation” phenotype express some inflammatory cytokines, but lack enhanced phagocytotic uptake, and express a different program of scavenging and uptake receptors, such as MARCO (Scara2) (23).

Th2-associated cytokines were at first thought to suppress macrophage function, as IL4 downregulated superoxide production and the production of IL1β (24). The activation of macrophages by IL4 was demonstrated in 1992 by Stein and colleagues, who reported that IL4 induced the expression of macrophage mannose receptor (MϕMR, CD206), upregulation of MHC Class II, proliferation, and syncytiation in macrophages (25). Additional transcriptional targets upregulated by IL4 or IL13 signaling in macrophages have since been identified, including the Type A Scavenger receptor (SR-A, CD204), the chitinase-like lectins Ym1/2, Resistin-like alpha (HIMF, FIZZ1, RELMα), and arginase-1 (26-28). The resulting paradigm categorizes T cell-elicited macrophages into two major categories, IFNγ-driven “classically-activated” macrophages, and IL4/IL13-elicited “alternatively-activated” macrophages, thought to be specialized for wound healing, fibrotic and scavenger programs, particularly evident in Th2 responses to helminthic, protozoan and other parasitic infections (29).

Investigations in tumor models have provided evidence for a potentially related macrophage phenotype – the “M2” macrophage. The evasive, immunosuppressive tumor microenvironment is associated with the production of IL10, TGFβ, glucocorticoids, and vitamin D3, which in turn elicit a poorly activated macrophage phenotype in situ, characterized by expression of Mϕ-MR and diminished acquisition of a dendritic cell-like antigen-presenting phenotype (30, 31). Mantovani and colleagues have further termed these variations of alternative-activation of macrophages “M2” phenotypes, respectively (32). These macrophage phenotypes are associated with immunosuppressive and angiogenic functions, elicited by tumors evading immune surveillance (33). Macrophage recruitment to tumors has been found to correlate with poor prognosis, implicating tumor-associated macrophages (TAMs) in critical mechanisms of tumor evasion (34). Recent gene profiling experiments from a breast tumor model have shown that transcriptional profiles among TAMs are surprisingly similar to fetal macrophages, suggesting that the tumorigenic environment adaptively elicits embryonic developmental functions of macrophages as an escape mechanism (35). The complex interactions of macrophages with the tumor microenvironment remain a highly active area of investigation (36).

The relationship of M2 cells and canonical “alternatively-activated” macrophages remains contentious – some authors will use the terms interchangeably, whereas others resist the integration of these models (Refs). The prevailing consensus at present is that these categorizations describe polar simplifications of what is likely to represent a spectrum of macrophage phenotypes in vivo, with varying degrees of specialization for microbicidal, surveillance, antigen-presenting, fibrotic, healing, and immunoregulatory functions (37, 38). Moreover, macrophages are thought to be highly plastic, readily programmable integrators of a variety of potentially competing pro-inflammatory, anti-inflammatory, regulatory, and modulatory signaling pathways (39).

Macrophage diversity – different precursors for different functions

In 2003, Geissman and Littman described the phenotypic markers defining circulating monocytic precursors that distinguish subsets of monocytes in mice. Monocytes which express Ly6C and CCR2 migrate to sites of inflammation, whereas monocytes which express CX3CR1, the receptor for CX3CL1/fractalkine, are precursors for partially self-renewing tissue-resident macrophages, which traffic through peripheral tissues under non-inflammatory homeostatic conditions (40). Importantly, these murine populations approximately mirror the major monocytic populations described in humans, particularly with regard to chemokine receptor expression and inflammatory potential (41).

The spleen has been identified as a critical reservoir of monocytic precursors in inflammatory conditions; egress of these precursors has been shown to be under the control of angiotensin II signaling (42). In experimental ischemic heart injury, early recruitment of Ly6Chi inflammatory macrophages is associated with injury, preceding the recruitment of Ly6Clo resident macrophages, which appear to be involved in myocardial healing (43). To further complicate matters, “inflammatory” monocyte populations may have immunosuppressive or regulatory functions; TNF, iNOS-producing dendritic cells (TipDCs) and myeloid-derived suppressor cells (MDSCs) may be derived from these precursor populations, suggesting that following recruitment, localized microenvironmental signals further tune phenotype and function among MPS-lineage cells (44, 45).

In contrast, Ly6Clo resident-type monocytes have been proposed to be precursors for alternatively-activated macrophages in tissues (46). Extravasated Ly6Clo monocytes bear a transcriptional profile similar to canonical alternatively-activated macrophages (47).

These findings are consistent with the characterization of the osteopetrotic op/op mouse, which carries a deleterious mutation in M-CSF (48). The strain mutation is named for a characteristic overgrown skeletal malformity, caused by defective osteoclast development (49, 50). However, M-CSF deficiency is not sufficient to completely ablate all macrophages; selected subsets of macrophages differentiate under the control of GM-CSF (51). GM-CSF has been reported to induce a more inflammatory program of transcriptional regulation than M-CSF, including the induction of IL1, IL6, TNFα, IL12 and IL23. These authors described a protective, immunoregulatory phenotype induced in the presence of M-CSF. Notably, these transcriptional programs are plastic, and are readily interconverted by switching CSF signals (52). Similar plasticity has been reported for macrophage differentiation induced by Th1 and Th2 cytokines (53).

From these data, we can synthesize a model wherein constituitive M-CSF expression controls Ly6Clo resident macrophages at steady-state, maintaining largely homeostatic macrophages such as scavenging and passive surveillance. In inflammatory conditions, such as in the case of infection or an autoimmune lesion, Ly6Chi monocytes expand in response to GM-CSF and are recruited to sites of inflammation. These cells are predisposed to inflammatory, antimicrobial, and cytotoxic functions. Resolution of inflammatory responses allows M-CSF levels to return to predominance, allowing Ly6Clo cells to mediate homeostatic repair and healing from inflammatory insult to the tissue.

Due to the developmental relationships between macrophages and dendritic cells, it may also be worthwhile to consider specialization for antigen-presentation and education of adaptive responses as a separate, perhaps orthogonal function of MPS-lineage cells. It has been recently argued that treating macrophages and dendritic cells as functionally independent lineages obscures the shared ontogeny and functional relationships of these cells (54). Indeed, both cell types have been shown to be able to perform functions commonly assumed to be restricted to the other: dendritic cells are capable of phagocytosis and production of oxygen radicals (55-58), while macrophages are capable of cross-presenting intracellular antigens (59-61). In this review, we confine our attention to macrophage populations.

Monocyte and macrophage infiltration in autoimmune disease

Cells of the monocyte and macrophage lineages comprise a substantial fraction, if not the majority, of infiltrating inflammatory cells in EAE (62-64), EAU (65), CIA (66) and experimental myocarditis (67).

Substantial research into macrophage-mediated autoimmune pathology has been pursued in various models of arthritis. Kelchtermans and colleagues observed that neutralization of IL17 during the severe CIA of IFNγR–/– mice diminished the production of GM-CSF in serum, following in vivo challenge with αCD3. These authors found no role for IFNγ in direct control of GM-CSF, arguing that GM-CSF is directly regulated by IL17 (68). In a model of streptococcal cell wall-induced arthritis, Plater-Zyberk and colleagues found that neutralization of GM-CSF by mAb blockade synergized with IL17R deficiency in the control of arthritis, indicating that GM-CSF mediates effector functions independent and non-synonymous with IL17 signaling (69). Campbell and colleagues found that treatment of mice with exogenous M-CSF exacerbated the severity and increased the incidence of CIA. The authors also found that neutralization of M-CSF with mAb, or induction of CIA in M-CSF-deficient op/op mice resulted in less severe disease (70). This group also described a pathogenic role for GM-CSF in CIA, as GM-CSF–/– mice were protected from CIA, characterized by no change in collagen II autoantibodies and diminished delayed-type hypersensitivity responses (66).

Research into the role of CSFs in other autoimmune diseases varies substantially, according to the specific model in question. In experimental lupus nephritis, op/op mice were protected from disease (71). In EAE, GM-CSF–/– mice are protected from disease (72). Enrichment of Ly6Chi inflammatory monocytes by adoptive transfer exacerbated EAE (64). Retroviral transduction of autoreactive T cells with GM-CSF was reported to be sufficient to enhance the severity of EAE (73). A specific role for GM-CSF has been described in the activation of microglia, as a necessary prerequisite for the onset of EAE (74). Reddy and colleagues have described a role for direct GM-CSF signaling in the induction of CD40 and TNFα expression in microglia, the resident macrophage cell type in the CNS (75).

Myocarditis – cellular mediators of autoimmunity, inflammation, and cardiac dysfunction

Cardiac inflammation is among the most common causes of non-congenital, non-ischemic sudden death in otherwise normal, healthy young adults (76, 77). Myocarditis in patients is diagnostically heterogeneous, with limited available treatment options (78, 79). While evidence for a wide variety of etiologies exist for myocarditis, infectious agents – notably various enteroviruses – are among the most common (80). The best studied and characterized model of viral myocarditis is infection with Coxsackievirus B3 (CB3), a common enterovirus (81). Recent evidence suggests persistent expression of viral products subsequent to parvovirus B19 infection may also be a critical determinant of myocarditis (82-84).

The actual mechanistic relationships between viral infection and chronic cardiac inflammatory disease, with particular relevance to progression to heart failure, appears to involve autoimmunity, particularly in the case of CB3 (85-88). In susceptible strains of mice, such as A/J, A.SW, and BALB/c, myocarditis continues well past the clearance of infectious virus due to autoreactive adaptive responses (87, 89, 90). In the same strains of mice, viral infection can be replaced by immunization of animals with cardiac antigen with appropriate adjuvants – this model is termed experimental autoimmune myocarditis (EAM) (91).

Depletion and adoptive transfer studies have unambiguously proven that experimental autoimmune myocarditis in rodents is a CD4+ T cell-dependent disease (92, 93). On one hand, these findings immediately implicate T cell-derived cytokines as critical effectors of autoreactive CD4+ T cells. They also further implicate other hematopoietic-origin cells as signaling targets of these cytokines, as downstream cellular effectors of T cell-driven autoimmune cardiac pathology (94).

Infiltration with total CD45+ leukocytes peaks at day 21 of EAM (Figure 1a). Our laboratory has previously demonstrated that infiltration of the heart by CD4+ T cells negatively correlates with cardiac function, further underscoring the centrality of CD4+ T cells in cardiac autoimmunity (67).

Figure 1.

Figure 1

Monocyte and macrophage infiltration into cardiac tissue in EAM. A)Time course of absolute enumeration of total CD45+ leukocytes. B) Representative gating of monocytes and macrophages from heart-infiltrating CD45+ leukocytes. C) Absolute enumeration of CD11b+F4/80CD45+ monocytes and CD11b+F4/80+CD45+ macrophages. Individual data points represent individual wild type female BALB/c mice immunized on days 0 and 7 with 100 μg/mL MyHCα614-629 in supplemented CFA, as previously described (91). Heart-infiltrating leukocytes were isolated as previously described (67). Bars indicate mean of each group. Asterisks denote significant t-test statistics, compared to day 0 (*, p < 0.05; **, p < 0.005).

Macrophage kinetics in the course of autoimmune heart disease

In autoimmune myocarditis, myeloid-lineage cells have been shown to comprise the bulk of infiltrating leukocytes (94). The majority of these cells express CD11b, indicating their myeloid lineage, but do not all express high levels of Ly6G (one component of the shared epitope Gr1), excluding the possibility that they are neutrophils (67). Similar results were obtained whether BALB/c or A/J mice were studied.

To address the role of macrophages in the natural history of autoimmune myocarditis, we examined the time course of infiltration of various populations of macrophages into the heart during EAM. As a proportion of total leukocytes, macrophages comprise the bulk of cardiac-resident cells in naïve mice (Figure 1b). However, during the course of EAM, influx of other CD45+ hematopoietic cells substantially diminishes the proportion of macrophages in the heart relative to other cell types, although MPS-lineage cells remain among the most numerous. As depicted in Figure 1c, infiltration of CD11b+F4/80 monocytes precedes the infiltration of macrophages into the heart; the peak of monocyte infiltration occurs on day 14 – indicating monocytes are among the first components of the autoaggressive cardiac infiltrate. Macrophage infiltration appears to peak later, on day 21.

Examining the expression of Ly6C on infiltrating macrophages reveals similar kinetic discontinuity. Cardiac infiltration with Ly6Chi inflammatory macrophages peaked on day 14, while the peak of infiltration with Ly6Clo resident macrophages followed on day 21 (Figure 2).

Figure 2.

Figure 2

Cardiac infiltration by macrophage subpopulations in EAM. A) Time course of absolute enumeration of Ly6ChiCD11b+F4/80+CD45+ “inflammatory” macrophages and Ly6CloCD11b+F4/80+CD45+ “resident” macrophages. B) Expression of markers of classical and alternative activation on intracardiac CD11b+F4/80+CD45+-gated macrophages during EAM. Individual data points represent individual wild type female BALB/c mice. Bars indicate mean of each group. Asterisks denote significant t-test statistics, compared to day 0 (*, p < 0.05; **, p < 0.005).

Macrophage balance in the control of inflammatory cardiac disease

In myocarditis, GM-CSF–/– mice were protected from disease, through a specific defect in the ability of GM-CSF–/– dendritic cells to potentiate pathogenic Th17 responses via IL6 signaling (95). Transfer of M-CSF-derived bone marrow-derived macrophages (BMMϕ) during the effector phase of disease was reported to be sufficient for the control of EAM, through a mechanism purported to involve IFNγ-dependent induction of nitric oxide and subsequent apoptosis of autoaggressive T cells (96). This mechanism conflicts with findings that inhibition of iNOS-derived nitric oxide production ameliorates myocarditis in rats (97, 98) and mice (99).

Our laboratory has reported that IL13-deficient mice develop a particularly severe form of myocarditis. In spite of the fact that mouse lymphocytes do not express receptors for IL13, we observed heightened T and B cell responses, suggesting that the protection afforded by IL13 in EAM is mediated indirectly through an innate compartment. Indeed, we observed fewer heart-infiltrating macrophages expressing markers of alternative activation, raising two non-exclusive possibilities: that alternatively-activated macrophages exert protective functions in EAM, or classically-activated macrophages mediate disease pathophysiology (100).

Macrophage infiltration was increased in the enhanced acute viral myocarditis of complement receptor 1/2–deficient mice following infection with CB3 (101). A role for differential activation of macrophages in the inflamed heart has been associated with the sex differences observed in cardiotropic CB3 infection in BALB/c mice – where male mice are more susceptible to increased cardiac inflammation. Li and colleagues reported that intracardiac macrophages from CB3-infected male mice preferentially expressed markers associated with classical or M1 activation, including iNOS, IL12, TNFα, and CD16/32, whereas heart-infiltrating macrophages in females bore a transcriptional profile consistent with alternative or M2 activation, characterized by arginase 1, IL10, and Mϕ-MR (102).

Frisancho-Kiss and colleagues orchiectomized male mice prior to infection with CB3, which diminishes the severity of resulting viral myocarditis with no change in cardiac viral titers – indicating this effect of complement signaling affects cardiac inflammation independently of antiviral immunity. This sex hormone-dependent protection from disease was associated with expansion of an “M2”-like F4/80+Gr1+ macrophage population, which coexpressed increased levels of TIM3, and decreased IL1β and IL10 (103, 104).

Chemokines associated with the recruitment of various MPS-lineage populations have similarly been described as effectors of autoimmune pathophysiology. Through bone marrow chimerization experiments, Mildner and colleagues found that CCR2 expression in the myeloid compartment enables the recruitment of Ly6Chi inflammatory monocytes to the CNS in EAE (105). Blockade of the CCR2 ligands CCL2/MCP1 or CCL3/MIP1α ameliorated autoimmune myocarditis, further underscoring that inflammatory Ly6Chi monocytes serve as precursors for effectors of cardiac inflammation (106).

Interactions of macrophages and other inflammatory cells

While some macrophages are clearly heart-resident (Figure 1), macrophages are also recruited to the inflamed heart in a CD4+ T cell-dependent manner. As shown in our kinetic studies, Ly6Chi “inflammatory” monocytes are among the first immigrants into the inflamed heart in EAM (Figure 2a). Moreover, intracardiac macrophages upregulate ICAM1 concurrent with the downregulation of MϕMR/CD206 (Figure 2b), indicating a switch from a native “alternatively-activated” macrophage environment, to an inflammatory “classically-activated” state over the course of disease. The biphasic recruitment of MPS subpopulations suggests that MPS involvement in EAM pathophysiology is discretely regulated by interactions with other cell types, most notably autoreactive CD4+ T cells. However, data from a variety of disease models suggest that relevant MPS crosstalk is not limited to the T cell compartment.

Treatment of mice infected with CB3 with alpha-galactosylceramide, a potent synthetic agonist of invariant NKT cells, reduced viral replication and cardiac inflammation, associated with expansion of splenic CD11b+F4/80+ cells, pointing to some measure of regulation between NKT cells and the MPS (107). IL6-deficient mice failed to control acute CB3 viremia, leading to increased cardiac infiltration during post-viral chronic myocarditis (108).

There is also evidence that hematopoietic cells, with some measure of “stemness”, are involved in complex regulation of cardiac inflammation. Kania and colleagues have reported a Prominin/CD133+ heart-resident pluripotent hematopoietic cell type with the ability to differentiate into myeloid cell types, including macrophages, or cells that resembled cardiomyocytes both in vitro and in vivo. Intravenous transfer of these cells into mice with EAM limited the development of disease through a mechanism dependent on IFNγ. These authors proposed that microenvironmental signals dictate the lineage commitment of these cells, whether to exert TGFβ-dependent fibrosis through differentiation into a fibroblast-like cell, or protective effects as a nitric oxide-producing macrophage (109, 110).

In contrast, in a mouse model of myocardial infarction, transfer of bone marrow cells led to increased trafficking of macrophages to the heart, associated with increased expression of inflammatory markers and improvement of cardiac functional parameters (111). Together, these data present a novel field of inquiry – the exciting proposition that MPS-lineage cells participate in the control of stem cell renewal and differentiation, and that manipulation of these pathways may present an attractive regenerative therapeutic target for a host of cardiac diseases.

Conclusion

The delicate balance of subpopulations within the macrophage compartment is increasingly appreciated within a diverse range of infectious, tumor, and autoimmune disease models. Pleiotropic functions of macrophages, beyond widely-acknowledged microbicidal and phagocytotic roles, participate in the pathophysiology and regulation of disease, including inflammatory heart disease. As further investigation yields insight into the functional complexity of these cells, it seems increasingly likely that these findings may provide novel tools and targets for modulation of the pathobiology of these diseases.

Table I. Models of macrophage differentiation.

Macrophage population Inducing signals Functions Markers Soluble Products References
Classically-activated (M1) macrophages IFNγ
TLR2
TLR4
TNFα
IL1
IL6
Reactive oxygen production
Phagocytosis
Bactericide
Antigen presentation
Costimulation
Inducible nitric oxide synthase (Nos2, iNOS); MHC Class II; CD80 (B7.1), CD86 (B7.2) CCL2 (MCP1)
CCL3 (MIP1α)
CCL10 (IP10)
IL1β
IL12p70
IL23
TNFα
(37, 112, 113)
Alternatively-activated (M2a) macrophages IL4
IL13
Fibrosis
Wound healing
Scavenging
T cell suppression
Arginase 1 (Arg1); Macrophage mannose receptor (Mrc1, Mϕ-MR, CD206); Macrophage scavenger receptor (Msr1, SR-A, CD204); FcεRII (Fcer2, CD23); YM1 (Chi3l3); YM2 (Chi3l4); RELMα (Retnla, FIZZ1, HIMF); MHC Class II IL1ra
CCL17 (TARC, A17)
CCL18 (AMAC1, MIP4, PARC)
Fibronectin
(29, 37)
Type II, Regulatory (M2b) macrophages IL1/TLR + immune complexes T cell Suppression
Inflammatory resolution
Sphingosine kinase 1 (SPHK1); LIGHT (TNFSF14) CCL1 (I-309)
IL10
(22, 114)
(M2c) macrophages IL10
TGFβ glucocorticoids
T cell Suppression
Angiogenesis
Tumor evasion
Arginase 1 (Arg1); B7-H4 (Vtcn1); Pentraxin 3; Regulator of G-protein signaling-16 (RGS16); Suppressor of cytokine signaling-3 (SOCS3) IL10
VEGF
(115-117)

Continually evolving nomenclature systems complicate the already confusing overlap between putative macrophage subpopulations. This table, while not meant to be comprehensive or, encapsulates the most commonly referenced subsets, with deference towards their original nomenclature. Those proposed by Mantovani, et al. are in parentheses (115).

Acknowledgments

The authors would like to express their gratitude to Monica V. Talor and G. Christian Baldeviano for critical and editorial assistance, and to R. Lee Blosser and Ada Tam for assistance with cytometric analyses.

This work was supported by NIH/NHLBI grants R01 HL70729 and R01 HL67290. JGB is the American Autoimmune-Related Disease Association O'Leary-Wilson Fellow in Autoimmune Disease Research, at the Johns Hopkins Autoimmune Disease Research Center. DC was supported by a research fellowship grant from the Myocarditis Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Metschnikoff E. A disease of Daphnia caused by a yeast. A contribution to the theory of phagocytes as agents for attack on disease-causing organisms. Archiv Pathol Anatomy. 1884;96:177. [Google Scholar]
  • 2.Takahashi K. Development and Differentiation of Macrophages and Related Cells: Historical Review and Current Concepts. Journal of Clinical and Experimental Hematopathology. 2001;41:1. [Google Scholar]
  • 3.van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL. The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ. 1972;46:845. [PMC free article] [PubMed] [Google Scholar]
  • 4.Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science. 327:656. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Pollard JW. Trophic macrophages in development and disease. Nat Rev Immunol. 2009;9:259. doi: 10.1038/nri2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kai H, Kudo H, Takayama N, Yasuoka S, Kajimoto H, Imaizumi T. Large blood pressure variability and hypertensive cardiac remodeling--role of cardiac inflammation. Circ J. 2009;73:2198. doi: 10.1253/circj.cj-09-0741. [DOI] [PubMed] [Google Scholar]
  • 7.Woollard KJ, Geissmann F. Monocytes in atherosclerosis: subsets and functions. Nat Rev Cardiol. 7:77. doi: 10.1038/nrcardio.2009.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Montgomery RR, Booth CJ, Wang X, Blaho VA, Malawista SE, Brown CR. Recruitment of macrophages and polymorphonuclear leukocytes in L:yme carditis. Infect Immun. 2007;75:613. doi: 10.1128/IAI.00685-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yearley JH, Pearson C, Carville A, Shannon RP, Mansfield KG. SIV-associated myocarditis: viral and cellular correlates of inflammation severity. AIDS Res Hum Retroviruses. 2006;22:529. doi: 10.1089/aid.2006.22.529. [DOI] [PubMed] [Google Scholar]
  • 10.Cimarelli A. Journey to the heart of macrophages: the delicate relationship between HIV-1 and a multifaceted cell type. Retrovirology. 7:28. doi: 10.1186/1742-4690-7-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kay DG, Yue P, Hanna Z, Jothy S, Tremblay E, Jolicoeur P. Cardiac disease in transgenic mice expressing human immunodeficiency virus-1 nef in cells of the immune system. Am J Pathol. 2002;161:321. doi: 10.1016/S0002-9440(10)64184-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Melo RC. Acute heart inflammation: ultrastructural and functional aspects of macrophages elicited by Trypanosoma cruzi infection. J Cell Mol Med. 2009;13:279. doi: 10.1111/j.1582-4934.2008.00388.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kemeny E, Grieve T, Marcus R, Sareli P, Zabriskie JB. Identification of mononuclear cells and T cell subsets in rheumatic valvulitis. Clin Immunol Immunopathol. 1989;52:225. doi: 10.1016/0090-1229(89)90174-8. [DOI] [PubMed] [Google Scholar]
  • 14.Cioc AM, Nuovo GJ. Histologic and in situ viral findings in the myocardium in cases of sudden, unexpected death. Mod Pathol. 2002;15:914. doi: 10.1097/01.MP.0000024291.37651.CD. [DOI] [PubMed] [Google Scholar]
  • 15.Nathan CF, Murray HW, Wiebe ME, Rubin BY. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983;158:670. doi: 10.1084/jem.158.3.670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bloom BR, Bennett B. Macrophages and delayed-type hypersensitivity. Semin Hematol. 1970;7:215. [PubMed] [Google Scholar]
  • 17.David JR. Lymphocyte mediators and cellular hypersensitivity. N Engl J Med. 1973;288:143. doi: 10.1056/NEJM197301182880311. [DOI] [PubMed] [Google Scholar]
  • 18.Hu X, Ivashkiv LB. Cross-regulation of signaling pathways by interferon-gamma: implications for immune responses and autoimmune diseases. Immunity. 2009;31:539. doi: 10.1016/j.immuni.2009.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dalton DK, Pitts-Meek S, Keshav S, Figari IS, Bradley A, Stewart TA. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science. 1993;259:1739. doi: 10.1126/science.8456300. [DOI] [PubMed] [Google Scholar]
  • 20.Kleinschmidt WJ, Schultz RM. Similarities of murine gamma interferon and the lymphokine that renders macrophages cytotoxic. J Interferon Res. 1982;2:291. doi: 10.1089/jir.1982.2.291. [DOI] [PubMed] [Google Scholar]
  • 21.Roberts WK, Vasil A. Evidence for the identity of murine gamma interferon and macrophage activating factor. J Interferon Res. 1982;2:519. doi: 10.1089/jir.1982.2.519. [DOI] [PubMed] [Google Scholar]
  • 22.Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453. doi: 10.2741/2692. [DOI] [PubMed] [Google Scholar]
  • 23.Mukhopadhyay S, Chen Y, Sankala M, Peiser L, Pikkarainen T, Kraal G, Tryggvason K, Gordon S. MARCO, an innate activation marker of macrophages, is a class A scavenger receptor for Neisseria meningitidis. Eur J Immunol. 2006;36:940. doi: 10.1002/eji.200535389. [DOI] [PubMed] [Google Scholar]
  • 24.Abramson SL, Gallin JI. IL-4 inhibits superoxide production by human mononuclear phagocytes. J Immunol. 1990;144:625. [PubMed] [Google Scholar]
  • 25.Stein M, Keshav S, Harris N, Gordon S. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med. 1992;176:287. doi: 10.1084/jem.176.1.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Welch JS, Escoubet-Lozach L, Sykes DB, Liddiard K, Greaves DR, Glass CK. TH2 cytokines and allergic challenge induce Ym1 expression in macrophages by a STAT6-dependent mechanism. J Biol Chem. 2002;277:42821. doi: 10.1074/jbc.M205873200. [DOI] [PubMed] [Google Scholar]
  • 27.Loke P, MacDonald AS, Robb A, Maizels RM, Allen JE. Alternatively activated macrophages induced by nematode infection inhibit proliferation via cell-to-cell contact. Eur J Immunol. 2000;30:2669. doi: 10.1002/1521-4141(200009)30:9<2669::AID-IMMU2669>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 28.Munder M, Eichmann K, Moran JM, Centeno F, Soler G, Modolell M. Th1/Th2-regulated expression of arginase isoforms in murine macrophages and dendritic cells. J Immunol. 1999;163:3771. [PubMed] [Google Scholar]
  • 29.Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • 30.Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23:549. doi: 10.1016/s1471-4906(02)02302-5. [DOI] [PubMed] [Google Scholar]
  • 31.Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev. 2006;25:315. doi: 10.1007/s10555-006-9001-7. [DOI] [PubMed] [Google Scholar]
  • 32.Mantovani A, Sozzani S, Locati M, Schioppa T, Saccani A, Allavena P, Sica A. Infiltration of tumours by macrophages and dendritic cells: tumour-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Novartis Found Symp. 2004;256:137. [PubMed] [Google Scholar]
  • 33.Mantovani A, Sica A, Locati M. Macrophage polarization comes of age. Immunity. 2005;23:344. doi: 10.1016/j.immuni.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 34.Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196:254. doi: 10.1002/path.1027. [DOI] [PubMed] [Google Scholar]
  • 35.Ojalvo LS, King W, Cox D, Pollard JW. High-density gene expression analysis of tumor-associated macrophages from mouse mammary tumors. Am J Pathol. 2009;174:1048. doi: 10.2353/ajpath.2009.080676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 141:39. doi: 10.1016/j.cell.2010.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009;27:451. doi: 10.1146/annurev.immunol.021908.132532. [DOI] [PubMed] [Google Scholar]
  • 38.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
  • 40.Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19:71. doi: 10.1016/s1074-7613(03)00174-2. [DOI] [PubMed] [Google Scholar]
  • 41.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 42.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, Kohler RH, Chudnovskiy A, Waterman P, Aikawa E, Mempel TR, Libby P, Weissleder R, Pittet MJ. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA, Pamer EG. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity. 2003;19:59. doi: 10.1016/s1074-7613(03)00171-7. [DOI] [PubMed] [Google Scholar]
  • 45.Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, De Baetselier P, Van Ginderachter JA. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233. doi: 10.1182/blood-2007-07-099226. [DOI] [PubMed] [Google Scholar]
  • 46.Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009;27:669. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
  • 47.Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006;177:7303. doi: 10.4049/jimmunol.177.10.7303. [DOI] [PubMed] [Google Scholar]
  • 48.Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa S, Okamura H, Sudo T, Shultz LD. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature. 1990;345:442. doi: 10.1038/345442a0. [DOI] [PubMed] [Google Scholar]
  • 49.Tanaka S, Takahashi N, Udagawa N, Tamura T, Akatsu T, Stanley ER, Kurokawa T, Suda T. Macrophage colony-stimulating factor is indispensable for both proliferation and differentiation of osteoclast progenitors. J Clin Invest. 1993;91:257. doi: 10.1172/JCI116179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Suda T, Tanaka S, Takahashi N. Macrophage colon-stimulating factor (M-CSF) is essential for differentiation rather than proliferation of osteoclast progenitors. Osteoporos Int. 1993;3 Suppl 1:111. doi: 10.1007/BF01621881. [DOI] [PubMed] [Google Scholar]
  • 51.Naito M, Hayashi S, Yoshida H, Nishikawa S, Shultz LD, Takahashi K. Abnormal differentiation of tissue macrophage populations in ‘osteopetrosis’ (op) mice defective in the production of macrophage colony-stimulating factor. Am J Pathol. 1991;139:657. [PMC free article] [PubMed] [Google Scholar]
  • 52.Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J Immunol. 2007;178:5245. doi: 10.4049/jimmunol.178.8.5245. [DOI] [PubMed] [Google Scholar]
  • 53.Gratchev A, Kzhyshkowska J, Kothe K, Muller-Molinet I, Kannookadan S, Utikal J, Goerdt S. Mphi1 and Mphi2 can be re-polarized by Th2 or Th1 cytokines, respectively, and respond to exogenous danger signals. Immunobiology. 2006;211:473. doi: 10.1016/j.imbio.2006.05.017. [DOI] [PubMed] [Google Scholar]
  • 54.Hume DA. Macrophages as APC and the dendritic cell myth. J Immunol. 2008;181:5829. doi: 10.4049/jimmunol.181.9.5829. [DOI] [PubMed] [Google Scholar]
  • 55.Burgdorf S, Kautz A, Bohnert V, Knolle PA, Kurts C. Distinct pathways of antige uptake and intracellular routing in CD4 and CD8 T cell activation. Science. 2007;316:612. doi: 10.1126/science.1137971. [DOI] [PubMed] [Google Scholar]
  • 56.Qureshi AA, Hosoi J, Xu S, Takashima A, Granstein RD, Lerner EA. Langerhans cells express inducible nitric oxide synthase and produce nitric oxide. J Invest Dermatol. 1996;107:815. doi: 10.1111/1523-1747.ep12330572. [DOI] [PubMed] [Google Scholar]
  • 57.Aiello S, Noris M, Piccinini G, Tomasoni S, Casiraghi F, Bonazzola S, Mister M, Sayegh MH, Remuzzi G. Thymic dendritic cells express inducible nitric oxide synthase and generate nitric oxide in response to self- and alloantigens. J Immunol. 2000;164:4649. doi: 10.4049/jimmunol.164.9.4649. [DOI] [PubMed] [Google Scholar]
  • 58.Powell TJ, Jenkins CD, Hattori R, MacPherson GG. Rat bone marrow-derived dendritic cells, but not ex vivo dendritic cells, secrete nitric oxide and can inhibit T-cell proliferation. Immunology. 2003;109:197. doi: 10.1046/j.1365-2567.2003.01639.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Alatery A, Siddiqui S, Chan M, Kus A, Petrof EO, Basta S. Cross, but not direct, presentation of cell-associated virus antigens by spleen macrophages is influenced by their differentiation state. Immunol Cell Biol. 88:3. doi: 10.1038/icb.2009.90. [DOI] [PubMed] [Google Scholar]
  • 60.Kiesel JR, Buchwald ZS, Aurora R. Cross-presentation by osteoclasts induces FoxP3 in CD8+ T cells. J Immunol. 2009;182:5477. doi: 10.4049/jimmunol.0803897. [DOI] [PubMed] [Google Scholar]
  • 61.Nakayama M, Akiba H, Takeda K, Kojima Y, Hashiguchi M, Azuma M, Yagita H, Okumura K. Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood. 2009;113:3821. doi: 10.1182/blood-2008-10-185884. [DOI] [PubMed] [Google Scholar]
  • 62.Al-Omaishi J, Bashir R, Gendelman HE. The cellular immunology of multiple sclerosis. J Leukoc Biol. 1999;65:444. doi: 10.1002/jlb.65.4.444. [DOI] [PubMed] [Google Scholar]
  • 63.Floris S, Blezer EL, Schreibelt G, Dopp E, van der Pol SM, Schadee-Eestermans IL, Nicolay K, Dijkstra CD, de Vries HE. Blood-brain barrier permeability and monocyte infiltration in experimental allergic encephalomyelitis: a quantitative MRI study. Brain. 2004;127:616. doi: 10.1093/brain/awh068. [DOI] [PubMed] [Google Scholar]
  • 64.King IL, Dickendesher TL, Segal BM. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood. 2009;113:3190. doi: 10.1182/blood-2008-07-168575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Dagkalis A, Wallace C, Xu H, Liebau S, Manivannan A, Stone MA, Mack M, Liversidge J, Crane IJ. Development of experimental autoimmune uveitis: efficient recruitment of monocytes is independent of CCR2. Invest Ophthalmol Vis Sci. 2009;50:4288. doi: 10.1167/iovs.09-3434. [DOI] [PubMed] [Google Scholar]
  • 66.Campbell IK, Rich MJ, Bischof RJ, Dunn AR, Grail D, Hamilton JA. Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice. J Immunol. 1998;161:3639. [PubMed] [Google Scholar]
  • 67.Afanasyeva M, Georgakopoulos D, Belardi DF, Ramsundar AC, Barin JG, Kass DA, Rose NR. Quantitative analysis of myocardial inflammation by flow cytometry in murine autoimmune myocarditis: correlation with cardiac function. Am J Pathol. 2004;164:807. doi: 10.1016/S0002-9440(10)63169-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kelchtermans H, Schurgers E, Geboes L, Mitera T, Van Damme J, Van Snick J, Uyttenhove C, Matthys P. Effector mechanisms of interleukin-17 in collagen-induced arthritis in the absence of interferon-gamma and counteraction by interferon-gamma. Arthritis Res Ther. 2009;11:R122. doi: 10.1186/ar2787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Plater-Zyberk C, Joosten LA, Helsen MM, Koenders MI, Baeuerle PA, van den Berg WB. Combined blockade of granulocyte-macrophage colony stimulating factor and interleukin 17 pathways potently suppresses chronic destructive arthritis in a tumour necrosis factor alpha-independent mouse model. Ann Rheum Dis. 2009;68:721. doi: 10.1136/ard.2007.085431. [DOI] [PubMed] [Google Scholar]
  • 70.Campbell IK, Rich MJ, Bischof RJ, Hamilton JA. The colony-stimulating factors and collagen-induced arthritis: exacerbation of disease by M-CSF and G-CSF and requirement for endogenous M-CSF. J Leukoc Biol. 2000;68:144. [PubMed] [Google Scholar]
  • 71.Menke J, Rabacal WA, Byrne KT, Iwata Y, Schwartz MM, Stanley ER, Schwarting A, Kelley VR. Circulating CSF-1 promotes monocyte and macrophage phenotypes that enhance lupus nephritis. J Am Soc Nephrol. 2009;20:2581. doi: 10.1681/ASN.2009050499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.McQualter JL, Darwiche R, Ewing C, Onuki M, Kay TW, Hamilton JA, Reid HH, Bernard CC. Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis. J Exp Med. 2001;194:873. doi: 10.1084/jem.194.7.873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Marusic S, Miyashiro JS, Douhan J, 3rd, Konz RF, Xuan D, Pelker JW, Ling V, Leonard JP, Jacobs KA. Local delivery of granulocyte macrophage colony-stimulating factor by retrovirally transduced antigen-specific T cells leads to severe, chronic experimental autoimmune encephalomyelitis in mice. Neurosci Lett. 2002;332:185. doi: 10.1016/s0304-3940(02)00947-3. [DOI] [PubMed] [Google Scholar]
  • 74.Ponomarev ED, Shriver LP, Maresz K, Pedras-Vasconcelos J, Verthelyi D, Dittel BN. GM-CSF production by autoreactive T cells is required for the activation of microglial cells and the onset of experimental autoimmune encephalomyelitis. J Immunol. 2007;178:39. doi: 10.4049/jimmunol.178.1.39. [DOI] [PubMed] [Google Scholar]
  • 75.Reddy PH, Manczak M, Zhao W, Nakamura K, Bebbington C, Yarranton G, Mao P. Granulocyte-macrophage colony-stimulating factor antibody suppresses microglial activity: implications for anti-inflammatory effects in Alzheimer's disease and multiple sclerosis. J Neurochem. 2009;111:1514. doi: 10.1111/j.1471-4159.2009.06432.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Villacorta-Lyew R, Laselle B, Mazzoncini JP, Merchant E, Buckley PJ. Cardiac pathological conditions in young soldiers: case series. Mil Med. 2008;173:1122. doi: 10.7205/milmed.173.11.1122. [DOI] [PubMed] [Google Scholar]
  • 77.Eckart RE, Scoville SL, Campbell CL, Shry EA, Stajduhar KC, Potter RN, Pearse LA, Virmani R. Sudden death in young adults: a 25-year review of autopsies in military recruits. Ann Intern Med. 2004;141:829. doi: 10.7326/0003-4819-141-11-200412070-00005. [DOI] [PubMed] [Google Scholar]
  • 78.Cooper LT., Jr Myocarditis. The New England journal of medicine. 2009;360:1526. doi: 10.1056/NEJMra0800028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Magnani JW, Dec GW. Myocarditis: current trends in diagnosis and treatment. Circulation. 2006;113:876. doi: 10.1161/CIRCULATIONAHA.105.584532. [DOI] [PubMed] [Google Scholar]
  • 80.Andreoletti L, Leveque N, Boulagnon C, Brasselet C, Fornes P. Viral causes of human myocarditis. Arch Cardiovasc Dis. 2009;102:559. doi: 10.1016/j.acvd.2009.04.010. [DOI] [PubMed] [Google Scholar]
  • 81.Tam PE. Coxsackievirus myocarditis: interplay between virus and host in the pathogenesis of heart disease. Viral Immunol. 2006;19:133. doi: 10.1089/vim.2006.19.133. [DOI] [PubMed] [Google Scholar]
  • 82.Escher F, Modrow S, Sabi T, Kuhl U, Lassner D, Schultheiss HP, Noutsias M. Parvovirus B19 profiles in patients presenting with acute myocarditis and chronic dilated cardiomyopathy. Med Sci Monit. 2008;14:CR589. [PubMed] [Google Scholar]
  • 83.Kuethe F, Lindner J, Matschke K, Wenzel JJ, Norja P, Ploetze K, Schaal S, Kamvissi V, Bornstein SR, Schwanebeck U, Modrow S. 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;49:1660. doi: 10.1086/648074. [DOI] [PubMed] [Google Scholar]
  • 84.Lindner J, Noutsias M, Lassner D, Wenzel J, Schultheiss HP, Kuehl U, Modrow S. Adaptive immune responses against parvovirus B19 in patients with myocardial disease. J Clin Virol. 2009;44:27. doi: 10.1016/j.jcv.2008.09.007. [DOI] [PubMed] [Google Scholar]
  • 85.Kallwellis-Opara A, Dorner A, Poller WC, Noutsias M, Kuhl U, Schultheiss HP, Pauschinger M. Autoimmunological features in inflammatory cardiomyopathy. Clin Res Cardiol. 2007;96:469. doi: 10.1007/s00392-007-0524-x. [DOI] [PubMed] [Google Scholar]
  • 86.Rose NR. Viral damage or ‘molecular mimicry’-placing the blame in myocarditis. Nat Med. 2000;6:631. doi: 10.1038/76199. [DOI] [PubMed] [Google Scholar]
  • 87.Rose NR. Myocarditis: Infection Versus Autoimmunity. J Clin Immunol. 2009 doi: 10.1007/s10875-009-9339-z. [DOI] [PubMed] [Google Scholar]
  • 88.Rose NR, Herskowitz A, Neumann DA, Neu N. Autoimmune myocarditis: a paradigm of post-infection autoimmune disease. Immunol Today. 1988;9:117. doi: 10.1016/0167-5699(88)91282-0. [DOI] [PubMed] [Google Scholar]
  • 89.Fairweather D, Frisancho-Kiss S, Rose NR. Viruses as adjuvants for autoimmunity: evidence from Coxsackievirus-induced myocarditis. Rev Med Virol. 2005;15:17. doi: 10.1002/rmv.445. [DOI] [PubMed] [Google Scholar]
  • 90.Fairweather D, Kaya Z, Shellam GR, Lawson CM, Rose NR. From infection to autoimmunity. J Autoimmun. 2001;16:175. doi: 10.1006/jaut.2000.0492. [DOI] [PubMed] [Google Scholar]
  • 91.Cihakova D, Sharma RB, Fairweather D, Afanasyeva M, Rose NR. Animal models for autoimmune myocarditis and autoimmune thyroiditis. Methods Mol Med. 2004;102:175. doi: 10.1385/1-59259-805-6:175. [DOI] [PubMed] [Google Scholar]
  • 92.Smith SC, Allen PM. Myosin-induced acute myocarditis is a T cell-mediated disease. J Immunol. 1991;147:2141. [PubMed] [Google Scholar]
  • 93.Smith SC, Allen PM. The role of T cells in myosin-induced autoimmune myocarditis. Clin Immunol Immunopathol. 1993;68:100. doi: 10.1006/clin.1993.1103. [DOI] [PubMed] [Google Scholar]
  • 94.Afanasyeva M, Georgakopoulos D, Rose NR. Autoimmune myocarditis: cellular mediators of cardiac dysfunction. Autoimmun Rev. 2004;3:476. doi: 10.1016/j.autrev.2004.08.009. [DOI] [PubMed] [Google Scholar]
  • 95.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;205:2281. doi: 10.1084/jem.20071119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Valaperti A, Marty RR, Kania G, Germano D, Mauermann N, Dirnhofer S, Leimenstoll B, Blyszczuk P, Dong C, Mueller C, Hunziker L, Eriksson U. CD11b+ monocytes abrogate Th17 CD4+ T cell-mediated experimental autoimmune myocarditis. J Immunol. 2008;180:2686. doi: 10.4049/jimmunol.180.4.2686. [DOI] [PubMed] [Google Scholar]
  • 97.Ishiyama S, Hiroe M, Nishikawa T, Abe S, Shimojo T, Ito H, Ozasa S, Yamakawa K, Matsuzaki M, Mohammed MU, Nakazawa H, Kasajima T, Marumo F. Nitric oxide contributes to the progression of myocardial damage in experimental autoimmune myocarditis in rats. Circulation. 1997;95:489. doi: 10.1161/01.cir.95.2.489. [DOI] [PubMed] [Google Scholar]
  • 98.Shin T, Tanuma N, Kim S, Jin J, Moon C, Kim K, Kohyama K, Matsumoto Y, Hyun B. An inhibitor of inducible nitric oxide synthase ameliorates experimental autoimmune myocarditis in Lewis rats. J Neuroimmunol. 1998;92:133. doi: 10.1016/s0165-5728(98)00194-5. [DOI] [PubMed] [Google Scholar]
  • 99.Barin JG, Talor MV, Baldeviano GC, Kimura M, Rose NR, Cihakova D. Mechanisms of IFNgamma regulation of autoimmune myocarditis. Exp Mol Pathol. doi: 10.1016/j.yexmp.2010.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cihakova D, Barin JG, Afanasyeva M, Kimura M, Fairweather D, Berg M, Talor MV, Baldeviano GC, Frisancho S, Gabrielson K, Bedja D, Rose NR. Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation. Am J Pathol. 2008;172:1195. doi: 10.2353/ajpath.2008.070207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.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;176:3516. doi: 10.4049/jimmunol.176.6.3516. [DOI] [PubMed] [Google Scholar]
  • 102.Li K, Xu W, Guo Q, Jiang Z, Wang P, Yue Y, Xiong S. Differential macrophage polarization in male and female BALB/c mice infected with coxsackievirus B3 defines susceptibility to viral myocarditis. Circ Res. 2009;105:353. doi: 10.1161/CIRCRESAHA.109.195230. [DOI] [PubMed] [Google Scholar]
  • 103.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;23:649. doi: 10.1016/j.bbi.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fairweather D, Cihakova D. Alternatively activated macrophages in infection and autoimmunity. J Autoimmun. 2009 doi: 10.1016/j.jaut.2009.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mildner A, Mack M, Schmidt H, Bruck W, Djukic M, Zabel MD, Hille A, Priller J, Prinz M. CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain. 2009;132:2487. doi: 10.1093/brain/awp144. [DOI] [PubMed] [Google Scholar]
  • 106.Goser S, Ottl R, Brodner A, Dengler TJ, Torzewski J, Egashira K, Rose NR, Katus HA, Kaya Z. Critical role for monocyte chemoattractant protein-1 and macrophage inflammatory protein-1alpha in induction of experimental autoimmune myocarditis and effective anti-monocyte chemoattractant protein-1 gene therapy. Circulation. 2005;112:3400. doi: 10.1161/CIRCULATIONAHA.105.572396. [DOI] [PubMed] [Google Scholar]
  • 107.Wu CY, Feng Y, Qian GC, Wu JH, Luo J, Wang Y, Chen GJ, Guo XK, Wang ZJ. alpha-Galactosylceramide protects mice from lethal Coxsackievirus B3 infection and subsequent myocarditis. Clin Exp Immunol. doi: 10.1111/j.1365-2249.2010.04233.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Poffenberger MC, Straka N, El Warry N, Fang D, Shanina I, Horwitz MS. Lack of IL-6 during coxsackievirus infection heightens the early immune response resulting in increased severity of chronic autoimmune myocarditis. PLoS One. 2009;4:e6207. doi: 10.1371/journal.pone.0006207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kania G, Blyszczuk P, Stein S, Valaperti A, Germano D, Dirnhofer S, Hunziker L, Matter CM, Eriksson U. Heart-infiltrating prominin-1+/CD133+ progenitor cells represent the cellular source of transforming growth factor beta-mediated cardiac fibrosis in experimental autoimmune myocarditis. Circ Res. 2009;105:462. doi: 10.1161/CIRCRESAHA.109.196287. [DOI] [PubMed] [Google Scholar]
  • 110.Kania G, Blyszczuk P, Valaperti A, Dieterle T, Leimenstoll B, Dirnhofer S, Zulewski H, Eriksson U. Prominin-1+/CD133+ bone marrow-derived heart-resident cells suppress experimental autoimmune myocarditis. Cardiovasc Res. 2008;80:236. doi: 10.1093/cvr/cvn190. [DOI] [PubMed] [Google Scholar]
  • 111.Sun J, Li SH, Liu SM, Wu J, Weisel RD, Zhuo YF, Yau TM, Li RK, Fazel SS. Improvement in cardiac function after bone marrow cell thearpy is associated with an increase in myocardial inflammation. Am J Physiol Heart Circ Physiol. 2009;296:H43. doi: 10.1152/ajpheart.00613.2008. [DOI] [PubMed] [Google Scholar]
  • 112.Adams DO, Hamilton TA. The cell biology of macrophage activation. Annu Rev Immunol. 1984;2:283. doi: 10.1146/annurev.iy.02.040184.001435. [DOI] [PubMed] [Google Scholar]
  • 113.Mosser DM. The many faces of macrophage activation. J Leukoc Biol. 2003;73:209. doi: 10.1189/jlb.0602325. [DOI] [PubMed] [Google Scholar]
  • 114.Anderson CF, Mosser DM. Cutting edge: biasing immune responses by directing antigen to macrophage Fc gamma receptors. J Immunol. 2002;168:3697. doi: 10.4049/jimmunol.168.8.3697. [DOI] [PubMed] [Google Scholar]
  • 115.Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677. doi: 10.1016/j.it.2004.09.015. [DOI] [PubMed] [Google Scholar]
  • 116.Perrier P, Martinez FO, Locati M, Bianchi G, Nebuloni M, Vago G, Bazzoni F, Sozzani S, Allavena P, Mantovani A. Distinct transcriptional programs activated by interleukin-10 with or without lipopolysaccharide in dendritic cells: induction of the B cell-activating chemokine, CXC chemokine ligand 13. J Immunol. 2004;172:7031. doi: 10.4049/jimmunol.172.11.7031. [DOI] [PubMed] [Google Scholar]
  • 117.Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P, Brumlik M, Cheng P, Curiel T, Myers L, Lackner A, Alvarez X, Ochoa A, Chen L, Zou W. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med. 2006;203:871. doi: 10.1084/jem.20050930. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES