Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Feb 1.
Published in final edited form as: Semin Liver Dis. 2010 Jul 21;30(3):245–257. doi: 10.1055/s-0030-1255354

Macrophages: Master Regulators of Inflammation and Fibrosis

Thomas A Wynn 1, Luke Barron 1
PMCID: PMC2924662  NIHMSID: NIHMS225126  PMID: 20665377

Abstract

Macrophages are found in close proximity with collagen-producing myofibroblasts and indisputably play a key role in fibrosis. They produce profibrotic mediators that directly activate fibroblasts, including transforming growth factor-β1 and platelet-derived growth factor, and control extracellular matrix turnover by regulating the balance of various matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases. Macrophages also regulate fibrogenesis by secreting chemokines that recruit fibroblasts and other inflammatory cells. With their potential to act in both a pro- and antifibrotic capacity, as well as their ability to regulate the activation of resident and recruited myofibroblasts, macrophages and the factors they express are integrated into all stages of the fibrotic process. These various, and sometimes opposing, functions may be performed by distinct macrophage subpopulations, the identification of which is a growing focus of fibrosis research. Although collagen-secreting myofibroblasts once were thought of as the master “producers” of fibrosis, this review will illustrate how macrophages function as the master “regulators” of fibrosis.

Keywords: Fibrosis, inflammation, collagen, wound healing, stellate cell, myofibroblasts, interleukin-13, transforming growth factor beta, tumor necrosis factor, interleukin-1, interleukin-17, arginase, Relm-alpha, chitinase


Fibrosis results when normal wound-healing responses persist or are not regulated properly, usually in response to some type of repeated injury. For example, the primary causes of liver fibrosis include persistent hepatitis C virus infection, chronic infections with the helminth parasites Schistosoma mansoni and Schistosoma japonicum, alcohol abuse, and nonalcoholic steatohepatitis. Following acute liver injury, a beneficial wound-healing mechanism regenerates damaged parenchymal cells, including hepatocytes, to replace necrotic tissue and apoptotic cells. But, when the cause of injury persists, extracellular matrix (ECM) components like fibrillar collagens accumulate to high levels, ultimately leading to advanced fibrosis or cirrhosis and hepatocellular dysfunction, as well as hepatic insufficiency and portal hypertension caused by increased intrahepatic resistance to blood flow. In diseased liver, most ECM components are produced by hepatic stellate cells (HSCs), also known as lipocytes, Ito cells, or perisinusoidal cells. HSCs respond to injury by differentiating into myofibroblast-like cells with contractile, proinflammatory, and potent fibrogenic activities. In addition, several groups have identified important roles for bone marrow–derived stem cells, circulating fibrocytes, and myofibroblasts derived from epithelial/endothelial mesenchymal transition.14 Because the various types of myofibroblasts are the primary collagen-producing cells in the liver, not surprisingly, they have been the object of decades of research aimed at characterizing the origins and functions of these cells in the fibrotic process.

Recent studies have also identified macrophages as critical regulators of fibrosis. Like myofibroblasts,15 these cells are derived from either resident tissue populations, like Kupffer cells, or from bone marrow immigrants.6,7 Studies now suggest the pathogenesis of fibrosis is tightly regulated by distinct macrophage populations that exert unique functional activities throughout the initiation, maintenance, and resolution phases of fibrosis.

EVIDENCE MACROPHAGES CONTROL FIBROSIS PROGRESSION AND RESOLUTION

Macrophages are almost always found in close proximity with collagen-producing myofibroblasts,810 and there is strong evidence that this interaction is reciprocal.4 Activated HSCs attract and stimulate macrophages with multiple chemokines and macrophage colony stimulating factor (M-CSF), and macrophages produce profibrotic mediators that directly activate fibroblasts, including transforming growth factor (TGF)-β1 and platelet-derived growth factor (PDGF). TGF-β1 is well known as an important inducer of fibrosis in many tissues and organs. In the liver, TGF-β1 promotes fibrosis by prompting HSC differentiation into myofibroblasts, enhancing expression of tissue inhibitors of matrix metalloproteases (TIMPs) that block ECM degradation, and by directly promoting synthesis of interstitial fibrillar collagens.1 PDGF also acts as a potent profibrotic signal by stimulating the proliferation of activated, collagen-producing HSCs.11 Interestingly, although a variety of cell types produce both mediators, several studies have identified macrophages as a critical source of TGF-β1 and PDGF in fibrosis.12,13 Macrophages also regulate fibrosis independently of direct interactions with myofibroblasts. Macrophages present antigens to propagate antigen-specific T-cell responses; produce their own matrix metalloproteases (MMPs) and TIMPs that control ECM turnover; secrete chemokines that recruit fibroblasts and other inflammatory cells; remove dead cells and debris by phagocytosis, which could otherwise trigger proinflammatory and profibrotic signals; and express immunoregulatory mediators like Relm-α/Fizz1/Retnla, chitinase-like proteins, and nitric oxide synthase (NOS)-2 or arginase-1 (Arg-1) that alter the magnitude and duration of the immune response.14 In addition, a growing body of evidence suggests that macrophages and other mediators regulate matrix deposition independently of TGF-β1.1518 Finally, macrophages also play a distinct role in the resolution of fibrosis.19

MACROPHAGES REGULATE FIBROSIS BY FUNCTIONING AS PHAGOCYTIC CELLS

Macrophages have been described classically as cells that phagocytose (engulf and then digest) cellular debris and pathogens. This key activity of macrophages is critical to fibrogenesis; however, whether phagocytosis promotes or slows the progression of fibrosis depends on the type of dead cells that are being engulfed and removed. Tissue injury causes damaged parenchymal cells, like hepatocytes, to undergo either apoptotic or necrotic cell death. Macrophages clear the dead cells in both cases; however, they typically repress inflammation when phagocytosing apoptotic cells and promote inflammation during necrotic cell removal.20,21 Because ingesting dying hepatocytes, and dead cells in general, increases TGF-β1 secretion,2023 in this case the phagocytic activity of macrophages is profibrotic. However, phagocytosis by macrophages can also aid in the regression or resolution of fibrosis because clearing apoptotic myofibroblasts, hepatocytes, and cellular debris often eliminates the stimuli inducing TGF-β1 and other profibrotic factors.2,24 The macrophage-dependent proinflammatory response to necrotic cells similarly holds both pro- and antifibrotic potential: first by recruiting inflammatory cells, which stimulate fibroblast activity and ECM deposition, and then by terminating inflammation by cleaning up tissue debris, including the dying inflammatory cells.25 For example, the phagocytosis of erythrocytes by macrophages and Kupffer cells promotes oxidative stress, inflammation, and fibrosis by depositing iron derived from hemoglobin in the liver.26 Conversely, the anti-fibrotic role of phagocytosis was illustrated nicely with the CCl4-induced liver injury model in which macrophages and Kupffer cells were shown to phagocytose apoptotic hepatocytes and reduce the inflammatory response, thereby shortening the course of injury and preventing the development of fibrosis.27 Likewise, macrophage-mediated phagocytosis of apoptotic cholangiocytes has been shown to promote the reversal of biliary fibrosis.28 Excess matrix components also accumulate from a combination of increased synthesis and secretion of matrix proteins by fibroblasts and from the increased proliferation and accumulation of HSCs at sites of tissue injury. Consequently, the deposition of ECM components is also controlled by macrophages and Kupffer cells removing apoptotic myofibroblasts.29 In addition to clearing apoptotic cells, macrophages can slow the progression of fibrosis by directly eliminating excess collagen from damaged tissues. A recent study suggested this mechanism is facilitated by milk fat globule epidermal growth factor 8 (Mfge8), which binds to and targets collagen for cellular uptake through Mfge8’s discoidin domains.30 Thus, macrophages’ phagocytic activity impacts both the induction and resolution of fibrosis through multiple mechanisms.

ACTIVATED MACROPHAGES PRODUCE AND ACTIVATE TGF-β1

The TGF-β cytokines (TGF-β1, -β2, and -β3) are induced and activated in a variety of fibrotic diseases, with the TGF-β1 isoform suspected of playing a key role in liver fibrogenesis.31 Early studies of liver fibrosis indicated that damaged hepatocytes and activated HSCs were likely the dominant sources of TGF-β1.32,33 These findings suggested an autocrine effect of TGF-β in liver fibrogenesis—that is, the cells that are the primary target of the cytokine produce TGF-β selectively. Subsequent studies, however, revealed that a variety of cell types produce TGF-β1 following liver injury, including hepatocytes, sinusoidal endothelial cells, HSCs, Kupffer cells, CD4+ T cells, bone marrow–derived monocytes, and monocyte-derived macrophages, with the type of cellular injury likely influencing the dominant source of TGF-β and its specific mechanism of action.1,34,35 In addition to inducing fibrosis, TGF-β may also inhibit inflammation and induce apoptosis.36 Thus, TGF-β both positively and negatively regulates fibrosis via pathways that are likely governed by the cells producing the cytokine. For example, TGF-β1-producing regulatory T cells were shown to ameliorate TGF-β1-mediated fibrosis through an interleukin (IL)-10-dependent mechanism.37 In contrast, macrophage-derived TGF-β1 is typically profibrotic. Indeed, numerous studies have identified various macrophage subsets as key producers of TGF-β1,34,35,3840 with the profibrotic cytokine IL-13 playing a key role by both inducing and activating latent TGF-β1 through an MMP9-dependent mechanism.41 Together, these data suggest TGF-β1 activity suppresses inflammation if produced by regulatory T cells but promotes fibrosis when derived from macrophages. The fact that TGF-β1 can simultaneously suppress inflammation but promote collagen synthesis in myofibroblasts likely explains the many contradictory findings reported for this cytokine. These observations present challenges for therapies geared toward TGF-β1 and its signaling pathway and may explain the lack of significant clinical progress with TGF-β-signaling inhibitors in the treatment of fibrotic diseases despite more than 20 years of research on the TGF-β pathway.42,43 Suppressing the profibrotic macrophage, rather than globally attenuating TGF-β1, might provide a more rational approach to ameliorate fibrosis. Indeed, studies have already shown that decreasing the number of TGF-β1-producing macrophages significantly slows the progression of hepatic fibrosis.44

SEVERAL SECRETED FACTORS FROM MACROPHAGES CONTROL FIBROGENESIS

In addition to producing TGF-β1, activated macrophages and Kupffer cells regulate fibrosis by secreting a variety of chemokines, cytokines, and growth factors. Macrophages, like other innate immune cell types, are activated by pathogen-associated molecular patterns that engage an array of pattern-recognition receptors.45 Once stimulated, macrophages secrete factors that recruit myofibroblasts and additional inflammatory cells to sites of tissue injury, and these recruited cells can also secrete a variety of chemokines, cytokines, and growth factors.14,25,4648 Thus, besides TGF-β1, activated macrophages amplify the fibrotic response indirectly by secreting numerous mediators that initiate and maintain the inflammatory cascade. Early studies showed the importance of monocyte chemotactic protein (MCP)-1 in recruiting CC chemokine receptor (CCR)2-expressing monocytes.49 Activated monocytes in turn produce MCP-2 and -3, which have been shown to participate in the development of primary biliary cirrhosis and liver fibrosis by recruiting and activating myofibroblasts and by regulating the production of macrophage-derived MMPs, including MMP2 and MMP9.34,5052 The CC chemokines macrophage inflammatory protein (MIP)-1α, MIP-1β, and regulated upon activation, normal t-cell expressed and secreted (RANTES), together with their corresponding receptors CCR1 and CCR5, are also routinely up-regulated in models of liver fibrogenesis.25 Furthermore, neutralization studies identified important roles for both CCR1- and CCR5-expressing Kupffer cells in the development of liver fibrosis.53 MIP-2 also participates in fibrogenesis by regulating angiogenesis.54 Another notable chemoattractant is osteopontin (OPN).55,56 Studies have shown that OPN recruits macrophages to injured tissues,57 and experiments with OPN−/− mice confirmed an important role for OPN in the development of kidney and liver fibrosis.58,59 However, OPN can also protect mice from CCl4-induced liver fibrosis.60 Regardless of the exact mechanism of action of OPN, these studies nicely illustrate that macrophages regulate fibrosis via mechanisms that are distinct from TGF-β1.61 Indeed, a complex interplay of chemokines and chemokine receptors, produced and expressed by macrophages (Kupffer cells), regulates fibrosis in part by promoting inflammation, angiogenesis, and the recruitment of macrophages and fibroblasts to sites of tissue injury.

Besides secreting TGF-β1 and chemoattractants, macrophages are an important source of yet other factors that regulate fibrosis. One is PDGF, which stimulates the proliferation, survival, and migration of myofibroblasts.62,63 Imatinib mesylate (STI-571, Gleevec), a PDGF receptor tyrosine kinase inhibitor, is accordingly in preclinical development as a possible therapy for chronic hepatic fibrosis.64 Macrophages produce significant amounts of IL-β1 in response to proinflammatory insults, and recent studies suggest that IL-1 myeloid differentiation primary response gene (88)(MyD88) signaling and the inflammasome are essential for fibrogenesis in the lung15,6567 and possibly, the liver.68,69 Another factor is urokinase plasminogen activator, which generates plasmin to directly degrade ECM and assists in the activating MMPs.70,71 Still other studies have shown that galectin-3, a β-galactoside-binding lectin produced by macrophages, is critically involved in the activation of renal fibroblasts. Depletion of galectin-3-positive macrophages reduced fibrosis severity after unilateral ureteric obstruction.72 Macrophages also produce insulin-like growth factor-I, which stimulates the proliferation and survival of myofibroblasts and promotes collagen synthesis by these cells.73 Finally, in some circumstances, macrophages and Kupffer cells secrete IL-4 and IL-13,74,75 which (as discussed in the next section) are believed to function as potent profibrotic cytokines.76

A ROLE FOR TH2 CYTOKINES AND ALTERNATIVELY ACTIVATED MACROPHAGES

The Th2 cytokines IL-4 and IL-13, like TGF-β1, directly stimulate collagen synthesis in mouse and human fibroblasts.7779 They also promote the development of the classic myofibroblast phenotype in human lung fibroblasts.80 In support of these observations, early immune deviation studies during Schistosoma infection, using IL-12 to switch from a Th2 to a Th1 pattern of cytokines, demonstrated that a Th2 response was associated with significant liver fibrosis, and a Th1 response provided protection from fibrosis.81 Similar findings were also observed with the CCl4-induced liver fibrosis model.82 Subsequent studies dissected the individual roles of Th2 cytokines and identified IL-13 as the dominant inducer of hepatic fibrosis in murine schistosomiasis,77,83 although IL-4, IL-5, IL-10, and IL-21 all participate via distinct mechanisms.77,8486 IL-13 has also been linked with the development of liver fibrosis in chronic hepatitis C virus infection.87 Although IL-13 can directly stimulate collagen synthesis in fibroblasts, at least one study suggested that IL-13 promotes fibrosis indirectly, by inducing and activating latent TGF-β1.41 However, results from the schistosomiasis model indicated that IL-13 induces liver fibrosis independently of TGF-β1 signaling,16 which is consistent with findings in humans developing liver fibrosis.87 IL-13 and TGF-β1 have also been shown to cooperate in the activation of myofibroblasts.79

Like myofibroblasts, macrophages respond to Th2 cytokines, and numerous studies have implicated IL-4/IL-13-stimulated macrophages in the pathogenesis of fibrosis,88,89 although their exact contribution remains controversial.14,9092 Gordon and colleagues first noted that macrophages treated with IL-4 developed an alternative activation state that is distinct from “classically activated macrophages” (CAMϕ) exposed to interferon-γ.9395 IL-13 was later shown to have equivalent effects as IL-4.94 In vitro, this phenotype is characterized by elevated major histocompatibility complex class II, mannose receptor (CD206), Ym1, Fizz1/Relm-α, and arginase activity.89,9398 These cells are now commonly termed “alternatively activated macrophages” (AAMϕ) or M2 macrophages.90,95 Studies of schistosomiasis-induced liver fibrosis found that AAMϕ compose 20 to 30% of egg-induced liver granulomas, suggesting that these cells are positioned to play an important regulatory role in hepatic fibrosis.89 Indeed, infected mice treated with Th1-inducing IL-12 had many more nitric oxide (NO)-expressing CAMϕ and much less liver fibrosis than did control mice. Interestingly, however, fibrosis was restored when NO production was blocked, suggesting pro- and antifibrotic roles for AAMϕ and CAMϕ, respectively.88,89 This model was also supported by findings with mice deficient in cationic amino transporter-2 (CAT-2), which imports L-arginine for use in NO production in macrophages.99 CAT-2−/− mice developed exacerbated liver fibrosis following Schistosoma infection, associated with decreased NO production and increased Arg1 activity in AAMϕ and fibroblasts.10 Together, these studies support the hypothesis that the phenotype of the macrophage (classically versus alternatively activated) plays an equal, if not more important, role in the pathogenesis of liver fibrogenesis than does the type of CD4+ T-helper cell response (Th1 versus Th2).76

Several other fibrosis models have also implicated AAMϕ in the pathogenesis of fibrosis. As observed in schistosomiasis, latent herpes virus infection-induced fibrosis is associated with a large population of AAMϕ.100 A silica-induced model of lung fibrosis also found that the IL-4Rα-dependent differentiation of AAMϕ is critical to the induction and maintenance of the CD4 + Th2 response that is required to trigger fibrosis.101 Still others have proposed that AAMϕ are needed for the activation of collagen-producing myofibroblasts, suggesting a positive feedback loop exists between these two cell types.76,89,102 Given the presumably important contribution of AAMϕ to the development of fibrosis, a great deal of recent research has sought to identify the signals that regulate their activation and recruitment to sites of inflammation, with IL-4, IL-13, IL-10, granulocyte-macrophage colony-stimulating factor, IL-21, prostaglandin E2 (PGE2), and Toll-like receptor signaling all playing important roles.85,89,97,103,104 There is also growing interest in understanding the functions of the unique set of genes that characterize alternative activation, including arginase-1 (Arg1), mannose receptor (Mrc1), FIZZ1 (Relm-α/hypoxia-induced mitogenic factor/Retnla), and the large chitinase family of genes (Ym1 [Chi3l3], Ym2 [Chi3l4], acidic mammalian chitinase [Chia], and YKL-40/BRP-3).105107

REGULATION OF FIBROSIS BY MACROPHAGE-ASSOCIATED RELM-α/FIZZ1

The functions of Relm-α in fibrosis are a topic of ongoing research and some controversy. Relm-α (Retnla) is a member of a family of cysteine-rich secreted proteins referred to as resistin-like molecules or found in inflammatory zone, originally identified in the lung.108 The resistin-like family consists of four members: Retnla/Relm-α/FIZZ1, Relm-β/FIZZ2, Resistin/FIZZ3, and Relm-γ/FIZZ4.109 Relm-α is expressed by bronchial epithelial cells and in the walls of the large and small bowel, and was originally hypothesized to regulate obesity and type-2 diabetes.110 Relm-α increases during allergic responses in the lung, as well as in the lung, liver, and/or gut during most helminth infections, largely due to IL-4, IL-13, and Stat6-dependent signaling.107,111,112 Relm-α is also induced in AAMϕ, serving as a useful biomarker of alternative activation.113115 Studies with the bleomycin model of fibrosis revealed that Relm-α is highly induced in the lung during fibrogenesis.116,117 Relm-α-expressing cells activate α-smooth muscle actin and type I collagen expression in fibroblasts via a notch1-dependent but TGF-β1-independent mechanism in vitro.116,117 Relm-α also exerts an antiapoptotic effect on mouse lung fibroblasts. Together, these findings provided evidence that macrophage-derived Relm-α might be involved in fibrogenesis by promoting the differentiation and survival of myofibroblasts.118 A similar hypothesis was also proposed for Relm-β (Retnlb).119 Thus, Relm-α and -βwere both predicted to be important mediators of wound repair and fibrosis at sites of Th2-mediated inflammation.

In addition to the injured lung, Relm-α is also found in abundance following infection with a variety of metazoan parasites.85,107,120123 In schistosomiasis, liver fibrosis and portal hypertension are the primary causes of chronic morbidity and mortality, and Th2 cytokines are essential to the development of fibrosis.83,124 Relm-α is markedly induced in the granulomatous gut, liver, and lungs of S. mansoni-infected and egg-challenged mice85,107 with expression depending on the Th2 immune response.107 Surprisingly, although studies with other helminth parasites linked Relm-α expression with AAMϕ,85,121,125 eosinophils appeared to be the predominant producers of Relm-α during schistosome infection.126 Experiments with Relm-α knockout/reporter mice revealed that Relm-α is not required for the development of helminth-induced CD4+ Th2 responses in the lung, liver, or gut.126 On the contrary, ablating Relm-α facilitated the development of a much stronger Th2 response to S. mansoni eggs, leading to exacerbated liver fibrosis mediated by IL-13. Relm-α similarly reduced inflammation in the lungs of egg-challenged mice.126,127 Therefore, instead of inducing fibrosis, as might have been predicted by prior in vitro studies with recombinant Relm-α/Fizz1 protein,111,116 in vivo Relm-α exhibits protective activity in schistosomiasis-induced fibrosis by functioning as a negative regulator of the Th2 response.126,127 The specific contribution of macrophage-derived Relm-α, however, remains unclear. Conditional deletion of Relm-α in macrophages could help clarify its role in fibrosis. It will also be important to investigate the role of Relm-α in other fibrotic diseases.111

CHITINASES AND CHITOLECTINS REGULATE FIBROSIS

Like Relm-α, chitin-binding proteins are induced in fibrotic lesions, are associated with AAMϕ, and remain an active topic of investigation, yet are inconclusively understood. Active chitinases hydrolyze chitin, an oligosaccharide polymer found in the exoskeletons of insects, crustaceans, fungal cell walls, and in some parasitic helminths, but not vertebrates. Eight human chitinases have been identified, but only two have preserved their chitinase activity, chitotriosidase (CHIT1) and acidic mammalian chitinase (CHIA). Due to mutations in the catalytic domain, the other chitinases have lost their chitinolytic activity and are termed chitolectins.128 Interestingly, expression of the chitinases and chitolectins increases during infectious and inflammatory responses and exhibits important regulatory activity in Th2-type immunity,107,129 although the exact function of the active chitinases remains debated.130,131 Acidic mammalian chitinase (AMCase) is induced in epithelial cells and macrophages by IL-13. Neutralization of AMCase activity has been shown to ameliorate Th2-dependent inflammation, AAMϕ development, airway remodeling, and airway hyperresponsiveness in an aeroallergen asthma model, in part by inhibiting activation of the IL-13 pathway.131 A similar effect was also recently ascribed to the mouse chitolectin breast regression protein-39 (BRP-39; Chi3l1) and its human homologue YKL-40.132 Although AMCase and BRP-39 were both identified as critical mediators of IL-13-induced responses, other studies have shown that IL-4/13-induced AMCase activity inhibits chitin from promoting Th2, AAMϕ, and allergic responses.130 Further study is clearly warranted to better understand the role of AM-Case and the entire chitinase-like family in the development of Th2-driven pathologies.

In addition to allergic-type inflammation, chitinases and chitolectins have been linked with the development of fibrosis in several organs including the liver, likely because of their association with Th2-type immune responses. For example, human chitotriosidase, produced mainly by activated macrophages and Kupffer cells,133 is elevated in patients with nonalcoholic steato-hepatitis.134136 Macrophage-derived chitotriosidase is also overexpressed in lung lavage samples taken from patients with idiopathic pulmonary fibrosis or sarcoidosis, especially from those with progressing disease and significant lung involvement, suggesting that this enzyme plays a role in the pathogenesis of diffuse lung disease-associated fibrosis.137,138 The plasma levels of YKL-40 increase in parallel with the degree of liver fibrosis and so might be useful as a serum biomarker of disease severity.139 However, YKL-40 is also increased in the serum of patients with sarcoidosis, endotoxemia, and some types of cancer,140142 suggesting it may better mark tissue damage, active inflammation, and fibrosis in general rather than distinguishing liver fibrosis, a pattern consistent with YKL-40’s presumed role as a growth factor for fibroblasts and vascular endothelial cells.142 AMCase, chitotriosidase, and BRP-39 are also increased in the lungs and livers of mice embedded with S. mansoni eggs, suggesting that the chitinase family may play an important role in the development IL-13-dependent fibrosis mediated by infections.107,143 Detailed mechanistic studies are needed to elucidate the unique contributions of the chitinases and chitolectins in the pathogenesis of fibrosis.

ARGINASE-1-EXPRESSING MACROPHAGES NEGATIVELY REGULATE FIBROSIS

Although a variety of proteins are differentially expressed between AAMϕ and CAMϕ,105,122,144 the enzymes Arg-1 and NOS-2, respectively, are the key inducible genes commonly used to differentiate these two populations.97,145,146 IL-13-activated macrophages produce the amino acid proline via a mechanism that is highly regulated by Arg-1.89 Because collagen synthesis requires proline,147 this pathway might explain how AAMϕ account for critical steps in wound healing and fibrosis.76,100,107,148150 Indeed, it has been suggested that the profibrotic activity of IL-13 depends on the activation of Arg-1 in macrophages89 However, this hypothesis was recently challenged by a key study conducted with LysMCre IL-4Rα −/flox mice, in which macrophages cannot recognize IL-4 or IL-13 and so cannot become alternatively activated.151 If deprived of AAMϕ, schistosome-infected mice responded by increasing Th1 cytokine production and NOS-2 activity, which exaggerated hepatic and intestinal pathology, impaired egg expulsion, and led to sepsis at the acute stage of infection. However, egg-induced granulomas and liver fibrosis developed normally in the absence of AAMϕ. Therefore, at least in the context of schistosomiasis, AAMϕ are not strictly required for the development of hepatic fibrosis or granuloma formation, but instead avert acute morbidity and mortality by down-regulating proinflammatory cytokine expression in the intestine.151 The specific contribution of macrophage-associated Arg-1, however, could not be discerned in these studies.

Recent studies have tested the function of arginine metabolism in various Th2 disease models using RNA interference, macrophage depletion, and the arginase inhibitors,120,152,153 yet relatively few investigations have examined the role of macrophage-associated Arg-1 in the regulation of fibrosis. Because Arg-1 is predominantly expressed by macrophages85,89 the newly created LysMCre Arg-1−/flox mouse provided an ideal tool to define the role of arginase in the schistosomiasis model of liver fibrosis by selectively deleting ARG1 in macrophages.103 In contrast to the findings with LysMCre IL-4Rα −/flox mice,151 depleting arginase-1 activity specifically in AAMϕ exacerbated the development of liver fibrosis and increased the Th2 immune response without altering the development of AAMϕ.91 The macrophage-specific Arg1 knockout mice also failed to develop endotoxemia or hepatotoxicity and survived acute infection, indicating that Arg-1 expression by AAMϕ is not required to suppress Th1- and NOS-2-mediated morbidity and mortality. Instead, LysMCre Arg-1−/flox mice died at an accelerated rate during the chronic stage of infection and displayed markedly increased granuloma size (i.e., inflammation), liver fibrosis, and portal hypertension. Blood was also frequently found in the intestine, suggesting that bleeding from collateral vessels was contributing to their morbidity and mortality. Thus, although Arg-1 was originally hypothesized to promote fibrosis and portal hypertension, studies with LysMCre Arg-1−/flox mice suggested that the primary role of Arg1-expressing AAMϕ is to slow the progression of schistosomiasis-induced liver disease.91

Th2 cytokine-activated macrophages have often been described as “suppressor” cells.154 Nevertheless, most studies have focused on “myeloid-derived suppressor cells” that are associated with cancers and that cross-regulate type-1 immunity and inhibit the function of classically activated macrophages.155157 Results from the schistosomiasis model suggested that AAMϕ slow the progression of IL-13-dependent liver fibrogenesis by specifically reducing the proliferation and/or activation of CD4+ Th2 cells,91 although an inhibitory effect on myofibroblasts could not be ruled out. Wild-type macrophages activated with IL-13 promoted minimal antigen-specific T-cell proliferation, and macrophages obtained from Arg-1 knockout mice promoted robust T-cell proliferation both before and after stimulation with IL-13.91 T-cell proliferation could also be restored if wild-type AAMϕ were supplemented with exogenous L-arginine, suggesting the suppressive activity of Arg1-expressing AAMϕ was mediated by L-arginine depletion.91,155 The inhibitory cytokines IL-10 and TGF-β1 were not involved, further confirming that L-arginine depletion by AAMϕ was serving as the primary suppressive mechanism. Thus, Arg1-expressing AAMϕ slowed the development of liver fibrosis, at least in part, by suppressing the antigen-specific CD4+ Th2-cell response. This conclusion contrasts with previous reports suggesting AAMϕ are required to induce Th2-cell responses.75,93,158 Given these new findings, it will be important to investigate whether Arg1-expressing macrophages impede ECM deposition in other fibrotic diseases.159,160

A ROLE FOR MACROPHAGES IN THE RESOLUTION AND REVERSAL OF FIBROSIS

Although for many years fibrosis was thought to be a progressive and irreversible process, recent studies have challenged this theory. Perhaps counterintuitively, ongoing inflammation may be the key to reversing fibrosis because inflammatory cells, in particular activated macrophages, are the key sources of MMPs (collagenases) that facilitate ECM degradation. Recruited macrophages and resident Kupffer cells also phagocytose apoptotic myofibroblasts and clean up cellular debris that otherwise perpetuates the fibrotic process. Although the identity and source of the key collagenases that resolve fibrosis remain unclear, accumulating evidence strongly implicates macrophages and the interstitial collagenases MMP1, MMP2, MMP8, MMP9, and MMP13 in the reversal of fibrosis. Experimental depletion of macrophages at the onset of fibrosis resolution (recovery phase) has been shown to retard ECM degradation and the loss of activated HSCs.19 Conversely, transferring macrophages into mice undergoing unilateral ureteral obstruction attenuates the development of renal fibrosis.161 Hepatic macrophages may also play an indirect role in the resolution of fibrosis by recruiting neutrophils, whose collagenases effectively digest ECM components.162 Together, these observations argue that macrophages regulate the resolution and reversal fibrosis through multiple mechanisms, producing MMPs that degrade the ECM, recruiting and/or activating additional collagenase-producing cells and phagocytosing cellular debris. Given that macrophages exert both pro- and antifibrotic activity,19,163 identifying the phenotype, subpopulation, and/or soluble mediator(s) that preferentially activate antifibrotic macrophages will be an important advance in the goal of developing the “holy grail” therapy that can reverse established and progressive fibrosis. The heterogeneity and plasticity of macrophages poses a formidable obstacle to this goal and likely explains why depleting macrophages globally has often yielded conflicting results.164,165 Manipulating distinct macrophage subpopulations may prove critical to understanding the contributions of macrophages to the initiation, maintenance, and resolution phases of fibrosis.19,163

A logical starting point for reversing fibrosis is to identify MMPs and other enzymes able to break down the ECM components that make up fibrotic lesions. Scar-associated macrophages are a major source of MMP13, and studies have shown that the resolution of CCl4-induced hepatic fibrosis is substantially retarded in MMP13-deficient mice.166 Results from the schistosomiasis model also revealed an important antifibrotic role for MMP13 in the development of infection-induced liver fibrosis. In this study, IL-13-mediated fibrosis was dependent on MMP12 (macrophage metalloelastase) and associated with markedly increased expression of collagenases MMP2, MMP9, and MMP13, suggesting that MMP12 was promoting fibrosis by limiting the expression of the ECM-degrading MMPs.167 These findings were consistent with recent studies showing overexpressing MMP9 in macrophages can attenuate pulmonary fibrosis induced by bleomycin,168 and MMP13 produced by Kupffer cells inhibits pig serum-induced liver fibrosis.169 Although numerous investigations have suggested antifibrotic roles for the collagenases, a few have provided conflicting results. Indeed, one study showed that cholestasis-induced liver fibrosis, caused by bile duct ligation, is attenuated in the absence of MMP13. These authors suggested that macrophage-derived MMP13 regulates HSC proliferation and activation, thus identifying a critical profibrotic role for MMP13.170 A similar protective effect was observed when macrophage infiltration was blocked during the induction of liver fibrogenesis in rats.171 These findings reinforce the key point that macrophages and Kupffer cells exhibit both pro- and antifibrotic activity, with the specific experimental model, type of MMP, and phase of the response likely dictating the overall contribution of macrophages to the progression, resolution, and reversal of fibrosis. Together, these observations suggest that instead of shutting down inflammation entirely, the currently favored approach,172174 manipulating the macrophage population or the character of the inflammatory response,2,25,91 might offer a more cogent strategy to treat progressive and advanced fibrotic disease.

SUMMARY AND FUTURE GOALS

It is clear that macrophages are critically involved in both the induction and resolution of fibrosis. Seemingly slight modifications in the pattern of MMP expression can dramatically affect outcomes, with macrophage-derived MMP12 enhancing fibrosis whereas MMP1 and MMP13 display potent antifibrotic activity.166,167 To promote fibrosis, macrophages produce specific MMPs, like MMP9, that degrade the basement membrane and allow inflammatory cells and recruited fibroblasts to enter sites of injury. They also secrete a variety of profibrotic mediators including TGF-β1, PDGF, and many chemokines that recruit and activate inflammatory cells. To negatively regulate fibrosis, macrophages secrete factors that induce myofibroblast apoptosis, remove cellular debris that otherwise perpetuates inflammation, engulf and digest ECM components, and stimulate the production of collagen-degrading MMPs in other cell types including stellate cells, myofibroblasts, and neutrophils. Macrophage-mediated changes in the ECM can also affect the survival of myofibroblasts and so facilitate the termination of progressive fibrosis.175 By expressing Arg-1, macrophages also deplete an essential amino acid that CD4+ T cells and myofibroblasts require to proliferate, thereby facilitating the down-regulation of profibrotic immune responses.91 These observations are consistent with many recent studies that have suggested macrophages are essential to the resolution of fibrosis.

Therefore, although myofibroblasts are typically thought of as the “master mediators” of fibrosis because they synthesize collagen and other ECM components, macrophages play an equally important role by serving as the “master regulators” of myofibroblast function and ECM degradation. A key goal of future research will be to determine when and how distinct subpopulations of macrophages control these disparate functions or whether the same macrophage population can adjust its phenotype over time in coordination with new stimuli found in the local milieu. It will be especially important to test whether recovering from fibrosis requires recruiting a new class of macrophage from the bone marrow.51 In addition to specifying which macrophage subpopulations promote, inhibit, or reverse fibrosis, future research should also elucidate the signals that regulate macrophage phenotypic conversion, differentiation, and recruitment. With our growing appreciation of the complex role of macrophages in fibrogenesis, the global depletion of macrophages no longer seems a viable option to treat highly progressive and established disease.19 Instead, future efforts should focus on identifying the specific macrophage subpopulations that facilitate the resolution and reversal fibrosis as well as their specific mechanism of action.

ABBREVIATIONS

AAMϕ

alternatively activated macrophages

AMCase

acidic mammalian chitinase

Arg-1

arginase-1

BRP-39

breast regression protein-39

CAMϕ

classically activated macrophages

CCR

CC chemokine receptor

CAT-2

cationic amino transporter-2

ECM

extracellular matrix

HSC

hepatic stellate cell

IL

interleukin

MCP

monocyte chemotactic protein

M-CSF

macrophage colony stimulating factor

Mfge8

milk fat globule epidermal growth factor 8

MIP

macrophage inflammatory protein

MMP

matrix metalloprotease

NO

nitric oxide

NOS

nitric oxide synthase

OPN

chemoattractant is osteopontin

PDGF

platelet-derived growth factor

TGF

transforming growth factor

TIMP

tissue inhibitors of matrix metalloprotease

References

  • 1.Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115:209–218. doi: 10.1172/JCI24282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Iredale JP. Models of liver fibrosis: exploring the dynamic nature of inflammation and repair in a solid organ. J Clin Invest. 2007;117:539–548. doi: 10.1172/JCI30542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology. 2008;134:1655–1669. doi: 10.1053/j.gastro.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. 2008;88:125–172. doi: 10.1152/physrev.00013.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wallace K, Burt AD, Wright MC. Liver fibrosis. Biochem J. 2008;411:1–18. doi: 10.1042/BJ20071570. [DOI] [PubMed] [Google Scholar]
  • 6.Luckey SW, Petersen DR. Activation of Kupffer cells during the course of carbon tetrachloride-induced liver injury and fibrosis in rats. Exp Mol Pathol. 2001;71:226–240. doi: 10.1006/exmp.2001.2399. [DOI] [PubMed] [Google Scholar]
  • 7.Ramadori G, Armbrust T. Cytokines in the liver. Eur J Gastroenterol Hepatol. 2001;13:777–784. doi: 10.1097/00042737-200107000-00004. [DOI] [PubMed] [Google Scholar]
  • 8.Ramm GA, Nair VG, Bridle KR, Shepherd RW, Crawford DH. Contribution of hepatic parenchymal and nonparenchymal cells to hepatic fibrogenesis in biliary atresia. Am J Pathol. 1998;153:527–535. doi: 10.1016/S0002-9440(10)65595-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Leicester KL, Olynyk JK, Brunt EM, Britton RS, Bacon BR. CD14-positive hepatic monocytes/macrophages increase in hereditary hemochromatosis. Liver Int. 2004;24:446–451. doi: 10.1111/j.1478-3231.2004.0943.x. [DOI] [PubMed] [Google Scholar]
  • 10.Thompson RW, Pesce JT, Ramalingam T, et al. Cationic amino acid transporter-2 regulates immunity by modulating arginase activity. PLoS Pathog. 2008;4:e1000023. doi: 10.1371/journal.ppat.1000023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Friedman SL, Arthur MJ. Activation of cultured rat hepatic lipocytes by Kupffer cell conditioned medium. Direct enhancement of matrix synthesis and stimulation of cell proliferation via induction of platelet-derived growth factor receptors. J Clin Invest. 1989;84:1780–1785. doi: 10.1172/JCI114362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wahl SM, McCartney-Francis N, Allen JB, Dougherty EB, Dougherty SF. Macrophage production of TGF-beta and regulation by TGF-beta. Ann N Y Acad Sci. 1990;593:188–196. doi: 10.1111/j.1749-6632.1990.tb16111.x. [DOI] [PubMed] [Google Scholar]
  • 13.Bonner JC, Osornio-Vargas AR, Badgett A, Brody AR. Differential proliferation of rat lung fibroblasts induced by the platelet-derived growth factor-AA, -AB, and -BB isoforms secreted by rat alveolar macrophages. Am J Respir Cell Mol Biol. 1991;5:539–547. doi: 10.1165/ajrcmb/5.6.539. [DOI] [PubMed] [Google Scholar]
  • 14.Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214:199–210. doi: 10.1002/path.2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gasse P, Mary C, Guenon I, et al. IL-1R1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J Clin Invest. 2007;117:3786–3799. doi: 10.1172/JCI32285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kaviratne M, Hesse M, Leusink M, et al. IL-13 activates a mechanism of tissue fibrosis that is completely TGF-beta independent. J Immunol. 2004;173:4020–4029. doi: 10.4049/jimmunol.173.6.4020. [DOI] [PubMed] [Google Scholar]
  • 17.Olman MA. Beyond TGF-beta: a prostaglandin promotes fibrosis. Nat Med. 2009;15:1360–1361. doi: 10.1038/nm1209-1360. [DOI] [PubMed] [Google Scholar]
  • 18.Oga T, Matsuoka T, Yao C, et al. Prostaglandin F(2alpha) receptor signaling facilitates bleomycin-induced pulmonary fibrosis independently of transforming growth factor-beta. Nat Med. 2009;15:1426–1430. doi: 10.1038/nm.2066. [DOI] [PubMed] [Google Scholar]
  • 19.Duffield JS, Forbes SJ, Constandinou CM, et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 2005;115:56–65. doi: 10.1172/JCI22675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med. 2009;361:1570–1583. doi: 10.1056/NEJMra0901217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rock KL, Kono H. The inflammatory response to cell death. Annu Rev Pathol. 2008;3:99–126. doi: 10.1146/annurev.pathmechdis.3.121806.151456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101:890–898. doi: 10.1172/JCI1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Takehara T, Tatsumi T, Suzuki T, et al. Hepatocyte-specific disruption of Bcl-xL leads to continuous hepatocyte apoptosis and liver fibrotic responses. Gastroenterology. 2004;127:1189–1197. doi: 10.1053/j.gastro.2004.07.019. [DOI] [PubMed] [Google Scholar]
  • 24.Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102:538–549. doi: 10.1172/JCI1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wynn TA. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Invest. 2007;117:524–529. doi: 10.1172/JCI31487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Otogawa K, Kinoshita K, Fujii H, et al. Erythrophagocytosis by liver macrophages (Kupffer cells) promotes oxidative stress, inflammation, and fibrosis in a rabbit model of steatohepatitis: implications for the pathogenesis of human nonalcoholic steatohepatitis. Am J Pathol. 2007;170:967–980. doi: 10.2353/ajpath.2007.060441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shi J, Aisaki K, Ikawa Y, Wake K. Evidence of hepatocyte apoptosis in rat liver after the administration of carbon tetrachloride. Am J Pathol. 1998;153:515–525. doi: 10.1016/S0002-9440(10)65594-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Patsenker E, Popov Y, Stickel F, Jonczyk A, Goodman SL, Schuppan D. Inhibition of integrin alphavbeta6 on cholangiocytes blocks transforming growth factor-beta activation and retards biliary fibrosis progression. Gastroenterology. 2008;135:660–670. doi: 10.1053/j.gastro.2008.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Douglass A, Wallace K, Parr R, et al. Antibody-targeted myofibroblast apoptosis reduces fibrosis during sustained liver injury. J Hepatol. 2008;49:88–98. doi: 10.1016/j.jhep.2008.01.032. [DOI] [PubMed] [Google Scholar]
  • 30.Atabai K, Jame S, Azhar N, et al. Mfge8 diminishes the severity of tissue fibrosis in mice by binding and targeting collagen for uptake by macrophages. J Clin Invest. 2009;119:3713–3722. doi: 10.1172/JCI40053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sanderson N, Factor V, Nagy P, et al. Hepatic expression of mature transforming growth factor beta 1 in transgenic mice results in multiple tissue lesions. Proc Natl Acad Sci U S A. 1995;92:2572–2576. doi: 10.1073/pnas.92.7.2572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bissell DM, Wang SS, Jarnagin WR, Roll FJ. Cell-specific expression of transforming growth factor-beta in rat liver. Evidence for autocrine regulation of hepatocyte proliferation. J Clin Invest. 1995;96:447–455. doi: 10.1172/JCI118055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol. 2003;200:500–503. doi: 10.1002/path.1427. [DOI] [PubMed] [Google Scholar]
  • 34.Karlmark KR, Weiskirchen R, Zimmermann HW, et al. Hepatic recruitment of the inflammatory Gr1 + monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology. 2009;50:261–274. doi: 10.1002/hep.22950. [DOI] [PubMed] [Google Scholar]
  • 35.Lin SL, Castaño AP, Nowlin BT, Lupher ML, Jr, Duffield JS. Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations. J Immunol. 2009;183:6733–6743. doi: 10.4049/jimmunol.0901473. [DOI] [PubMed] [Google Scholar]
  • 36.Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGF-beta in hepatic fibrosis. Front Biosci. 2002;7:d793–d807. doi: 10.2741/A812. [DOI] [PubMed] [Google Scholar]
  • 37.Kitani A, Fuss I, Nakamura K, Kumaki F, Usui T, Strober W. Transforming growth factor (TGF)-beta1-producing regulatory T cells induce Smad-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGF-beta1-mediated fibrosis. J Exp Med. 2003;198:1179–1188. doi: 10.1084/jem.20030917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schmid-Kotsas A, Gross HJ, Menke A, et al. Lipopolysaccharide-activated macrophages stimulate the synthesis of collagen type I and C-fibronectin in cultured pancreatic stellate cells. Am J Pathol. 1999;155:1749–1758. doi: 10.1016/S0002-9440(10)65490-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cameron RG, Blendis LM, Neuman MG. Accumulation of macrophages in primary sclerosing cholangitis. Clin Biochem. 2001;34:195–201. doi: 10.1016/s0009-9120(01)00215-6. [DOI] [PubMed] [Google Scholar]
  • 40.Xing Z, Tremblay GM, Sime PJ, Gauldie J. Overexpression of granulocyte-macrophage colony-stimulating factor induces pulmonary granulation tissue formation and fibrosis by induction of transforming growth factor-beta 1 and myofibroblast accumulation. Am J Pathol. 1997;150:59–66. [PMC free article] [PubMed] [Google Scholar]
  • 41.Lee CG, Homer RJ, Zhu Z, et al. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor beta(1) J Exp Med. 2001;194:809–821. doi: 10.1084/jem.194.6.809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fattouh R, Jordana M. TGF-beta, eosinophils and IL-13 in allergic airway remodeling: a critical appraisal with therapeutic considerations. Inflamm Allergy Drug Targets. 2008;7:224–236. doi: 10.2174/187152808786848388. [DOI] [PubMed] [Google Scholar]
  • 43.Fattouh R, Midence NG, Arias K, et al. Transforming growth factor-beta regulates house dust mite-induced allergic airway inflammation but not airway remodeling. Am J Respir Crit Care Med. 2008;177:593–603. doi: 10.1164/rccm.200706-958OC. [DOI] [PubMed] [Google Scholar]
  • 44.Ide M, Kuwamura M, Kotani T, Sawamoto O, Yamate J. Effects of gadolinium chloride (GdCl(3)) on the appearance of macrophage populations and fibrogenesis in thioacetamide-induced rat hepatic lesions. J Comp Pathol. 2005;133:92–102. doi: 10.1016/j.jcpa.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 45.Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors and immune recognition. Annu Rev Immunol. 2005;23:901–944. doi: 10.1146/annurev.immunol.23.021704.115816. [DOI] [PubMed] [Google Scholar]
  • 46.Szabo G, Mandrekar P, Dolganiuc A. Innate immune response and hepatic inflammation. Semin Liver Dis. 2007;27:339–350. doi: 10.1055/s-2007-991511. [DOI] [PubMed] [Google Scholar]
  • 47.Meneghin A, Hogaboam CM. Infectious disease, the innate immune response, and fibrosis. J Clin Invest. 2007;117:530–538. doi: 10.1172/JCI30595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Seki E, De Minicis S, Osterreicher CH, et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 2007;13:1324–1332. doi: 10.1038/nm1663. [DOI] [PubMed] [Google Scholar]
  • 49.Marra F, DeFranco R, Grappone C, et al. Expression of monocyte chemotactic protein-1 precedes monocyte recruitment in a rat model of acute liver injury, and is modulated by vitamin E. J Investig Med. 1999;47:66–75. [PubMed] [Google Scholar]
  • 50.Tsuneyama K, Harada K, Yasoshima M, et al. Monocyte chemotactic protein-1, -2, and -3 are distinctively expressed in portal tracts and granulomata in primary biliary cirrhosis: implications for pathogenesis. J Pathol. 2001;193:102–109. doi: 10.1002/1096-9896(2000)9999:9999<::AID-PATH725>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 51.Seki E, de Minicis S, Inokuchi S, et al. CCR2 promotes hepatic fibrosis in mice. Hepatology. 2009;50:185–197. doi: 10.1002/hep.22952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Okuma T, Terasaki Y, Kaikita K, et al. C-C chemokine receptor 2 (CCR2) deficiency improves bleomycin-induced pulmonary fibrosis by attenuation of both macrophage infiltration and production of macrophage-derived matrix metalloproteinases. J Pathol. 2004;204:594–604. doi: 10.1002/path.1667. [DOI] [PubMed] [Google Scholar]
  • 53.Seki E, De Minicis S, Gwak GY, et al. CCR1 and CCR5 promote hepatic fibrosis in mice. J Clin Invest. 2009;119:1858–1870. doi: 10.1172/JCI37444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Keane MP, Belperio JA, Moore TA, et al. Neutralization of the CXC chemokine, macrophage inflammatory protein-2, attenuates bleomycin-induced pulmonary fibrosis. J Immunol. 1999;162:5511–5518. [PubMed] [Google Scholar]
  • 55.Kohan M, Bader R, Puxeddu I, Levi-Schaffer F, Breuer R, Berkman N. Enhanced osteopontin expression in a murine model of allergen-induced airway remodelling. Clin Exp Allergy. 2007;37:1444–1454. doi: 10.1111/j.1365-2222.2007.02801.x. [DOI] [PubMed] [Google Scholar]
  • 56.Berman JS, Serlin D, Li X, et al. Altered bleomycin-induced lung fibrosis in osteopontin-deficient mice. Am J Physiol Lung Cell Mol Physiol. 2004;286:L1311–L1318. doi: 10.1152/ajplung.00394.2003. [DOI] [PubMed] [Google Scholar]
  • 57.Kawashima R, Mochida S, Matsui A, et al. Expression of osteopontin in Kupffer cells and hepatic macrophages and stellate cells in rat liver after carbon tetrachloride intoxication: a possible factor for macrophage migration into hepatic necrotic areas. Biochem Biophys Res Commun. 1999;256:527–531. doi: 10.1006/bbrc.1999.0372. [DOI] [PubMed] [Google Scholar]
  • 58.Persy VP, Verhulst A, Ysebaert DK, De Greef KE, De Broe ME. Reduced postischemic macrophage infiltration and interstitial fibrosis in osteopontin knockout mice. Kidney Int. 2003;63:543–553. doi: 10.1046/j.1523-1755.2003.00767.x. [DOI] [PubMed] [Google Scholar]
  • 59.Sahai A, Malladi P, Melin-Aldana H, Green RM, Whitington PF. Upregulation of osteopontin expression is involved in the development of nonalcoholic steatohepatitis in a dietary murine model. Am J Physiol Gastrointest Liver Physiol. 2004;287:G264–G273. doi: 10.1152/ajpgi.00002.2004. [DOI] [PubMed] [Google Scholar]
  • 60.Lorena D, Darby IA, Gadeau AP, et al. Osteopontin expression in normal and fibrotic liver. altered liver healing in osteopontin-deficient mice. J Hepatol. 2006;44:383–390. doi: 10.1016/j.jhep.2005.07.024. [DOI] [PubMed] [Google Scholar]
  • 61.Wang Y, Mochida S, Kawashima R, et al. Increased expression of osteopontin in activated Kupffer cells and hepatic macrophages during macrophage migration in Propionibacterium acnes-treated rat liver. J Gastroenterol. 2000;35:696–701. doi: 10.1007/s005350070049. [DOI] [PubMed] [Google Scholar]
  • 62.Martinet Y, Rom WN, Grotendorst GR, Martin GR, Crystal RG. Exaggerated spontaneous release of platelet-derived growth factor by alveolar macrophages from patients with idiopathic pulmonary fibrosis. N Engl J Med. 1987;317:202–209. doi: 10.1056/NEJM198707233170404. [DOI] [PubMed] [Google Scholar]
  • 63.Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev. 2004;15:255–273. doi: 10.1016/j.cytogfr.2004.03.006. [DOI] [PubMed] [Google Scholar]
  • 64.Yoshiji H, Noguchi R, Kuriyama S, et al. Imatinib mesylate (STI-571) attenuates liver fibrosis development in rats. Am J Physiol Gastrointest Liver Physiol. 2005;288:G907–G913. doi: 10.1152/ajpgi.00420.2004. [DOI] [PubMed] [Google Scholar]
  • 65.Zhang Y, Lee TC, Guillemin B, Yu MC, Rom WN. Enhanced IL-1 beta and tumor necrosis factor-alpha release and messenger RNA expression in macrophages from idiopathic pulmonary fibrosis or after asbestos exposure. J Immunol. 1993;150:4188–4196. [PubMed] [Google Scholar]
  • 66.Kolb M, Margetts PJ, Anthony DC, Pitossi F, Gauldie J. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J Clin Invest. 2001;107:1529–1536. doi: 10.1172/JCI12568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wilson MS, Madala SK, Ramalingam TR, et al. Bleomycin and IL-1β-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010;207:535–552. doi: 10.1084/jem.20092121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tiggelman AM, Boers W, Linthorst C, Sala M, Chamuleau RA. Collagen synthesis by human liver (myo)fibroblasts in culture: evidence for a regulatory role of IL-1 beta, IL-4, TGF beta and IFN gamma. J Hepatol. 1995;23:307–317. [PubMed] [Google Scholar]
  • 69.Mancini R, Benedetti A, Jezequel AM. An interleukin-1 receptor antagonist decreases fibrosis induced by dimethyl-nitrosamine in rat liver. Virchows Arch. 1994;424:25–31. doi: 10.1007/BF00197389. [DOI] [PubMed] [Google Scholar]
  • 70.Leyland H, Gentry J, Arthur MJ, Benyon RC. The plasminogen-activating system in hepatic stellate cells. Hepatology. 1996;24:1172–1178. doi: 10.1002/hep.510240532. [DOI] [PubMed] [Google Scholar]
  • 71.Salgado S, Garcia J, Vera J, et al. Liver cirrhosis is reverted by urokinase-type plasminogen activator gene therapy. Mol Ther. 2000;2:545–551. doi: 10.1006/mthe.2000.0210. [DOI] [PubMed] [Google Scholar]
  • 72.Henderson NC, Mackinnon AC, Farnworth SL, et al. Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis. Am J Pathol. 2008;172:288–298. doi: 10.2353/ajpath.2008.070726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wynes MW, Riches DW. Induction of macrophage insulin-like growth factor-I expression by the Th2 cytokines IL-4 and IL-13. J Immunol. 2003;171:3550–3559. doi: 10.4049/jimmunol.171.7.3550. [DOI] [PubMed] [Google Scholar]
  • 74.Büttner C, Skupin A, Reimann T, et al. Local production of interleukin-4 during radiation-induced pneumonitis and pulmonary fibrosis in rats: macrophages as a prominent source of interleukin-4. Am J Respir Cell Mol Biol. 1997;17:315–325. doi: 10.1165/ajrcmb.17.3.2279. [DOI] [PubMed] [Google Scholar]
  • 75.Hayashi N, Matsui K, Tsutsui H, et al. Kupffer cells from Schistosoma mansoni-infected mice participate in the prompt type 2 differentiation of hepatic T cells in response to worm antigens. J Immunol. 1999;163:6702–6711. [PubMed] [Google Scholar]
  • 76.Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004;4:583–594. doi: 10.1038/nri1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chiaramonte MG, Donaldson DD, Cheever AW, Wynn TA. An IL-13 inhibitor blocks the development of hepatic fibrosis during a T-helper type 2-dominated inflammatory response. J Clin Invest. 1999;104:777–785. doi: 10.1172/JCI7325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Oriente A, Fedarko NS, Pacocha SE, Huang SK, Lichtenstein LM, Essayan DM. Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther. 2000;292:988–994. [PubMed] [Google Scholar]
  • 79.Murray LA, Argentieri RL, Farrell FX, et al. Hyper-responsiveness of IPF/UIP fibroblasts: interplay between TGFbeta1, IL-13 and CCL2. Int J Biochem Cell Biol. 2008;40:2174–2182. doi: 10.1016/j.biocel.2008.02.016. [DOI] [PubMed] [Google Scholar]
  • 80.Hashimoto S, Gon Y, Takeshita I, Maruoka S, Horie T. IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase-dependent pathway. J Allergy Clin Immunol. 2001;107:1001–1008. doi: 10.1067/mai.2001.114702. [DOI] [PubMed] [Google Scholar]
  • 81.Wynn TA, Cheever AW, Jankovic D, et al. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature. 1995;376:594–596. doi: 10.1038/376594a0. [DOI] [PubMed] [Google Scholar]
  • 82.Shi Z, Wakil AE, Rockey DC. Strain-specific differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc Natl Acad Sci U S A. 1997;94:10663–10668. doi: 10.1073/pnas.94.20.10663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Chiaramonte MG, Cheever AW, Malley JD, Donaldson DD, Wynn TA. Studies of murine schistosomiasis reveal interleukin-13 blockade as a treatment for established and progressive liver fibrosis. Hepatology. 2001;34:273–282. doi: 10.1053/jhep.2001.26376. [DOI] [PubMed] [Google Scholar]
  • 84.Hoffmann KF, Cheever AW, Wynn TA. IL-10 and the dangers of immune polarization: excessive type 1 and type 2 cytokine responses induce distinct forms of lethal immunopathology in murine schistosomiasis. J Immunol. 2000;164:6406–6416. doi: 10.4049/jimmunol.164.12.6406. [DOI] [PubMed] [Google Scholar]
  • 85.Pesce J, Kaviratne M, Ramalingam TR, et al. The IL-21 receptor augments Th2 effector function and alternative macrophage activation. J Clin Invest. 2006;116:2044–2055. doi: 10.1172/JCI27727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Reiman RM, Thompson RW, Feng CG, et al. Interleukin-5 (IL-5) augments the progression of liver fibrosis by regulating IL-13 activity. Infect Immun. 2006;74:1471–1479. doi: 10.1128/IAI.74.3.1471-1479.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Weng HL, Liu Y, Chen JL, et al. The etiology of liver damage imparts cytokines transforming growth factor beta1 or interleukin-13 as driving forces in fibrogenesis. Hepatology. 2009;50:230–243. doi: 10.1002/hep.22934. [DOI] [PubMed] [Google Scholar]
  • 88.Hesse M, Cheever AW, Jankovic D, Wynn TA. NOS-2 mediates the protective anti-inflammatory and antifibrotic effects of the Th1-inducing adjuvant, IL-12, in a Th2 model of granulomatous disease. Am J Pathol. 2000;157:945–955. doi: 10.1016/S0002-9440(10)64607-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hesse M, Modolell M, La Flamme AC, et al. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167:6533–6544. doi: 10.4049/jimmunol.167.11.6533. [DOI] [PubMed] [Google Scholar]
  • 90.Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • 91.Pesce JT, Ramalingam TR, Mentink-Kane MM, et al. Arginase-1-expressing macrophages suppress Th2 cytokine-driven inflammation and fibrosis. PLoS Pathog. 2009;5:e1000371. doi: 10.1371/journal.ppat.1000371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Song E, Ouyang N, Hörbelt M, Antus B, Wang M, Exton MS. Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cell Immunol. 2000;204:19–28. doi: 10.1006/cimm.2000.1687. [DOI] [PubMed] [Google Scholar]
  • 93.Goerdt S, Orfanos CE. Other functions, other genes: alternative activation of antigen-presenting cells. Immunity. 1999;10:137–142. doi: 10.1016/s1074-7613(00)80014-x. [DOI] [PubMed] [Google Scholar]
  • 94.Doyle AG, Herbein G, Montaner LJ, et al. Interleukin-13 alters the activation state of murine macrophages in vitro: comparison with interleukin-4 and interferon-gamma. Eur J Immunol. 1994;24:1441–1445. doi: 10.1002/eji.1830240630. [DOI] [PubMed] [Google Scholar]
  • 95.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;176:287–292. doi: 10.1084/jem.176.1.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gordon S. Macrophages and the immune response. In: Paul WE, editor. Fundamental Immunology. Philadelphia: Lippincott-Raven Publishers; 1999. pp. 533–544. [Google Scholar]
  • 97.Modolell M, Corraliza IM, Link F, Soler G, Eichmann K. Reciprocal regulation of the nitric oxide synthase/arginase balance in mouse bone marrow-derived macrophages by TH1 and TH2 cytokines. Eur J Immunol. 1995;25:1101–1104. doi: 10.1002/eji.1830250436. [DOI] [PubMed] [Google Scholar]
  • 98.Schebesch C, Kodelja V, Müller C, et al. Alternatively activated macrophages actively inhibit proliferation of peripheral blood lymphocytes and CD4 + T cells in vitro. Immunology. 1997;92:478–486. doi: 10.1046/j.1365-2567.1997.00371.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.MacLeod CL. Regulation of cationic amino acid transporter (CAT) gene expression. Biochem Soc Trans. 1996;24:846–852. doi: 10.1042/bst0240846. [DOI] [PubMed] [Google Scholar]
  • 100.Gangadharan B, Hoeve MA, Allen JE, et al. Murine gammaherpesvirus-induced fibrosis is associated with the development of alternatively activated macrophages. J Leukoc Biol. 2008;84:50–58. doi: 10.1189/jlb.0507270. [DOI] [PubMed] [Google Scholar]
  • 101.Migliaccio CT, Buford MC, Jessop F, Holian A. The IL-4Ralpha pathway in macrophages and its potential role in silica-induced pulmonary fibrosis. J Leukoc Biol. 2008;83:630–639. doi: 10.1189/jlb.0807533. [DOI] [PubMed] [Google Scholar]
  • 102.Prasse A, Pechkovsky DV, Toews GB, et al. A vicious circle of alveolar macrophages and fibroblasts perpetuates pulmonary fibrosis via CCL18. Am J Respir Crit Care Med. 2006;173:781–792. doi: 10.1164/rccm.200509-1518OC. [DOI] [PubMed] [Google Scholar]
  • 103.El Kasmi KC, Qualls JE, Pesce JT, et al. Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat Immunol. 2008;9:1399–1406. doi: 10.1038/ni.1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Frey AB. Myeloid suppressor cells regulate the adaptive immune response to cancer. J Clin Invest. 2006;116:2587–2590. doi: 10.1172/JCI29906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Loke P, Nair MG, Guiliano D, et al. IL-4 dependent alternatively-activated macrophages have a distinctive in vivo gene expression phenotype. Biomed Central. 2002;3:7. doi: 10.1186/1471-2172-3-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Raes G, Baetselier PD, Noel W, et al. Differential expression of FIZZ1 and Ym1 in alternatively versus classically activated macrophages. J Leukoc Biol. 2002;71:597–602. [PubMed] [Google Scholar]
  • 107.Sandler NG, Mentink-Kane MM, Cheever AW, Wynn TA. Global gene expression profiles during acute pathogen-induced pulmonary inflammation reveal divergent roles for Th1 and Th2 responses in tissue repair. J Immunol. 2003;171:3655–3667. doi: 10.4049/jimmunol.171.7.3655. [DOI] [PubMed] [Google Scholar]
  • 108.Holcomb IN, Kabakoff RC, Chan B, et al. FIZZ1, a novel cysteine-rich secreted protein associated with pulmonary inflammation, defines a new gene family. EMBO J. 2000;19:4046–4055. doi: 10.1093/emboj/19.15.4046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gerstmayer B, Küsters D, Gebel S, et al. Identification of RELMgamma, a novel resistin-like molecule with a distinct expression pattern. Genomics. 2003;81:588–595. doi: 10.1016/s0888-7543(03)00070-3. [DOI] [PubMed] [Google Scholar]
  • 110.Beltowski J. Adiponectin and resistin—new hormones of white adipose tissue. Med Sci Monit. 2003;9:RA55–RA61. [PubMed] [Google Scholar]
  • 111.Liu T, Jin H, Ullenbruch M, et al. Regulation of found in inflammatory zone 1 expression in bleomycin-induced lung fibrosis: role of IL-4/IL-13 and mediation via STAT-6. J Immunol. 2004;173:3425–3431. doi: 10.4049/jimmunol.173.5.3425. [DOI] [PubMed] [Google Scholar]
  • 112.Stütz AM, Pickart LA, Trifilieff A, Baumruker T, Prieschl-Strassmayr E, Woisetschläger M. The Th2 cell cytokines IL-4 and IL-13 regulate found in inflammatory zone 1/resistin-like molecule alpha gene expression by a STAT6 and CCAAT/enhancer-binding protein-dependent mechanism. J Immunol. 2003;170:1789–1796. doi: 10.4049/jimmunol.170.4.1789. [DOI] [PubMed] [Google Scholar]
  • 113.Nair MG, Cochrane DW, Allen JE. Macrophages in chronic type 2 inflammation have a novel phenotype characterized by the abundant expression of Ym1 and Fizz1 that can be partly replicated in vitro. Immunol Lett. 2003;85:173–180. doi: 10.1016/s0165-2478(02)00225-0. [DOI] [PubMed] [Google Scholar]
  • 114.Edwards JP, Zhang X, Frauwirth KA, Mosser DM. Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol. 2006;80:1298–1307. doi: 10.1189/jlb.0406249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Raes G, Noël W, Beschin A, Brys L, de Baetselier P, Hassanzadeh GH. FIZZ1 and Ym as tools to discriminate between differentially activated macrophages. Dev Immunol. 2002;9:151–159. doi: 10.1080/1044667031000137629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Liu T, Dhanasekaran SM, Jin H, et al. FIZZ1 stimulation of myofibroblast differentiation. Am J Pathol. 2004;164:1315–1326. doi: 10.1016/S0002-9440(10)63218-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Liu T, Hu B, Choi YY, et al. Notch1 signaling in FIZZ1 induction of myofibroblast differentiation. Am J Pathol. 2009;174:1745–1755. doi: 10.2353/ajpath.2009.080618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Chung MJ, Liu T, Ullenbruch M, Phan SH. Antiapoptotic effect of found in inflammatory zone (FIZZ)1 on mouse lung fibroblasts. J Pathol. 2007;212:180–187. doi: 10.1002/path.2161. [DOI] [PubMed] [Google Scholar]
  • 119.Mishra A, Wang M, Schlotman J, et al. Resistin-like molecule-beta is an allergen-induced cytokine with inflammatory and remodeling activity in the murine lung. Am J Physiol Lung Cell Mol Physiol. 2007;293:L305–L313. doi: 10.1152/ajplung.00147.2007. [DOI] [PubMed] [Google Scholar]
  • 120.Anthony RM, Urban JF, Jr, Alem F, et al. Memory T(H)2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med. 2006;12:955–960. doi: 10.1038/nm1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Loke P, Gallagher I, Nair MG, et al. Alternative activation is an innate response to injury that requires CD4 + T cells to be sustained during chronic infection. J Immunol. 2007;179:3926–3936. doi: 10.4049/jimmunol.179.6.3926. [DOI] [PubMed] [Google Scholar]
  • 122.Reece JJ, Siracusa MC, Scott AL. Innate immune responses to lung-stage helminth infection induce alternatively activated alveolar macrophages. Infect Immun. 2006;74:4970–4981. doi: 10.1128/IAI.00687-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Anthony RM, Rutitzky LI, Urban JF, Jr, Stadecker MJ, Gause WC. Protective immune mechanisms in helminth infection. Nat Rev Immunol. 2007;7:975–987. doi: 10.1038/nri2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wilson MS, Mentink-Kane MM, Pesce JT, Ramalingam TR, Thompson R, Wynn TA. Immunopathology of schistosomiasis. Immunol Cell Biol. 2007;85:148–154. doi: 10.1038/sj.icb.7100014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Nair MG, Gallagher IJ, Taylor MD, et al. Chitinase and Fizz family members are a generalized feature of nematode infection with selective upregulation of Ym1 and Fizz1 by antigen-presenting cells. Infect Immun. 2005;73:385–394. doi: 10.1128/IAI.73.1.385-394.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Pesce JT, Ramalingam TR, Wilson MS, et al. Retnla (relmalpha/fizz1) suppresses helminth-induced Th2-type immunity. PLoS Pathog. 2009;5:e1000393. doi: 10.1371/journal.ppat.1000393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Nair MG, Du Y, Perrigoue JG, et al. Alternatively activated macrophage-derived RELM-alpha is a negative regulator of type 2 inflammation in the lung. J Exp Med. 2009;206:937–952. doi: 10.1084/jem.20082048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Guan SP, Mok YK, Koo KN, Chu KL, Wong WS. Chitinases: biomarkers for human diseases. Protein Pept Lett. 2009;16:490–498. doi: 10.2174/092986609788167842. [DOI] [PubMed] [Google Scholar]
  • 129.Sutherland TE, Maizels RM, Allen JE. Chitinases and chitinase-like proteins: potential therapeutic targets for the treatment of T-helper type 2 allergies. Clin Exp Allergy. 2009;39:943–955. doi: 10.1111/j.1365-2222.2009.03243.x. [DOI] [PubMed] [Google Scholar]
  • 130.Reese TA, Liang HE, Tager AM, et al. Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature. 2007;447:92–96. doi: 10.1038/nature05746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Zhu Z, Zheng T, Homer RJ, et al. Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation. Science. 2004;304:1678–1682. doi: 10.1126/science.1095336. [DOI] [PubMed] [Google Scholar]
  • 132.Lee CG, Hartl D, Lee GR, et al. Role of breast regression protein 39 (BRP-39)/chitinase 3-like-1 in Th2 and IL-13-induced tissue responses and apoptosis. J Exp Med. 2009;206:1149–1166. doi: 10.1084/jem.20081271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Boot RG, Renkema GH, Strijland A, van Zonneveld AJ, Aerts JM. Cloning of a cDNA encoding chitotriosidase, a human chitinase produced by macrophages. J Biol Chem. 1995;270:26252–26256. doi: 10.1074/jbc.270.44.26252. [DOI] [PubMed] [Google Scholar]
  • 134.Malaguarnera L. Chitotriosidase: the yin and yang. Cell Mol Life Sci. 2006;63:3018–3029. doi: 10.1007/s00018-006-6269-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Malaguarnera L, Di Rosa M, Zambito AM, dell’Ombra N, Nicoletti F, Malaguarnera M. Chitotriosidase gene expression in Kupffer cells from patients with non-alcoholic fatty liver disease. Gut. 2006;55:1313–1320. doi: 10.1136/gut.2005.075697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Malaguarnera L, Rosa MD, Zambito AM, dell’Ombra N, Marco RD, Malaguarnera M. Potential role of chitotriosidase gene in nonalcoholic fatty liver disease evolution. Am J Gastroenterol. 2006;101:2060–2069. doi: 10.1111/j.1572-0241.2006.00680.x. [DOI] [PubMed] [Google Scholar]
  • 137.Bargagli E, Margollicci M, Luddi A, et al. Chitotriosidase activity in patients with interstitial lung diseases. Respir Med. 2007;101:2176–2181. doi: 10.1016/j.rmed.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 138.Tercelj M, Salobir B, Simcic S, Wraber B, Zupancic M, Rylander R. Chitotriosidase activity in sarcoidosis and some other pulmonary diseases. Scand J Clin Lab Invest. 2009;69:575–578. doi: 10.1080/00365510902829362. [DOI] [PubMed] [Google Scholar]
  • 139.Tran A, Benzaken S, Saint-Paul MC, et al. Chondrex (YKL-40), a potential new serum fibrosis marker in patients with alcoholic liver disease. Eur J Gastroenterol Hepatol. 2000;12:989–993. doi: 10.1097/00042737-200012090-00004. [DOI] [PubMed] [Google Scholar]
  • 140.Johansen JS, Krabbe KS, Møller K, Pedersen BK. Circulating YKL-40 levels during human endotoxaemia. Clin Exp Immunol. 2005;140:343–348. doi: 10.1111/j.1365-2249.2005.02763.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Junker N, Johansen JS, Andersen CB, Kristjansen PE. Expression of YKL-40 by peritumoral macrophages in human small cell lung cancer. Lung Cancer. 2005;48:223–231. doi: 10.1016/j.lungcan.2004.11.011. [DOI] [PubMed] [Google Scholar]
  • 142.Johansen JS, Milman N, Hansen M, Garbarsch C, Price PA, Graudal N. Increased serum YKL-40 in patients with pulmonary sarcoidosis—a potential marker of disease activity? Respir Med. 2005;99:396–402. doi: 10.1016/j.rmed.2004.09.016. [DOI] [PubMed] [Google Scholar]
  • 143.Hoffmann KF, McCarty TC, Segal DH, et al. Disease fingerprinting with cDNA microarrays reveals distinct gene expression profiles in lethal type 1 and type 2 cytokine-mediated inflammatory reactions. FASEB J. 2001;15:2545–2547. doi: 10.1096/fj.01-0306fje. [DOI] [PubMed] [Google Scholar]
  • 144.Troidl C, Möllmann H, Nef H, et al. Classically and alternatively activated macrophages contribute to tissue remodelling after myocardial infarction. J Cell Mol Med. 2009;13:3485–3496. doi: 10.1111/j.1582-4934.2009.00707.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Munder M, Eichmann K, Modolell M. Alternative metabolic states in murine macrophages reflected by the nitric oxide synthase/arginase balance: competitive regulation by CD4 + T cells correlates with Th1/Th2 phenotype. J Immunol. 1998;160:5347–5354. [PubMed] [Google Scholar]
  • 146.Morris SM, Jr, Kepka-Lenhart D, Chen LC. Differential regulation of arginases and inducible nitric oxide synthase in murine macrophage cells. Am J Physiol. 1998;275(5 Pt 1):E740–E747. doi: 10.1152/ajpendo.1998.275.5.E740. [DOI] [PubMed] [Google Scholar]
  • 147.Dunn MA, Rojkind M, Warren KS, Hait PK, Rifas L, Seifter S. Liver collagen synthesis in murine schistosomiasis. J Clin Invest. 1977;59:666–674. doi: 10.1172/JCI108685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Shearer JD, Richards JR, Mills CD, Caldwell MD. Differential regulation of macrophage arginine metabolism: a proposed role in wound healing. Am J Physiol. 1997;272(2 Pt 1):E181–E190. doi: 10.1152/ajpendo.1997.272.2.E181. [DOI] [PubMed] [Google Scholar]
  • 149.Endo M, Oyadomari S, Terasaki Y, et al. Induction of arginase I and II in bleomycin-induced fibrosis of mouse lung. Am J Physiol Lung Cell Mol Physiol. 2003;285:L313–L321. doi: 10.1152/ajplung.00434.2002. [DOI] [PubMed] [Google Scholar]
  • 150.Desguerre I, Mayer M, Leturcq F, Barbet JP, Gherardi RK, Christov C. Endomysial fibrosis in Duchenne muscular dystrophy: a marker of poor outcome associated with macrophage alternative activation. J Neuropathol Exp Neurol. 2009;68:762–773. doi: 10.1097/NEN.0b013e3181aa31c2. [DOI] [PubMed] [Google Scholar]
  • 151.Herbert DR, Hölscher C, Mohrs M, et al. Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. Immunity. 2004;20:623–635. doi: 10.1016/s1074-7613(04)00107-4. [DOI] [PubMed] [Google Scholar]
  • 152.Yang M, Rangasamy D, Matthaei KI, et al. Inhibition of arginase I activity by RNA interference attenuates IL-13-induced airways hyperresponsiveness. J Immunol. 2006;177:5595–5603. doi: 10.4049/jimmunol.177.8.5595. [DOI] [PubMed] [Google Scholar]
  • 153.Zhao A, Urban JF, Jr, Anthony RM, et al. Th2 cytokine-induced alterations in intestinal smooth muscle function depend on alternatively activated macrophages. Gastroenterology. 2008;135:217–225. doi: 10.1053/j.gastro.2008.03.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature. 1996;383:787–793. doi: 10.1038/383787a0. [DOI] [PubMed] [Google Scholar]
  • 155.Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5:641–654. doi: 10.1038/nri1668. [DOI] [PubMed] [Google Scholar]
  • 156.Gallina G, Dolcetti L, Serafini P, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8 + T cells. J Clin Invest. 2006;116:2777–2790. doi: 10.1172/JCI28828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Rodriguez PC, Quiceno DG, Zabaleta J, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64:5839–5849. doi: 10.1158/0008-5472.CAN-04-0465. [DOI] [PubMed] [Google Scholar]
  • 158.Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164:6166–6173. doi: 10.4049/jimmunol.1701141. [DOI] [PubMed] [Google Scholar]
  • 159.Segawa M, Fukada S, Yamamoto Y, et al. Suppression of macrophage functions impairs skeletal muscle regeneration with severe fibrosis. Exp Cell Res. 2008;314:3232–3244. doi: 10.1016/j.yexcr.2008.08.008. [DOI] [PubMed] [Google Scholar]
  • 160.Zimmermann N, King NE, Laporte J, et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest. 2003;111:1863–1874. doi: 10.1172/JCI17912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Nishida M, Okumura Y, Fujimoto S, Shiraishi I, Itoi T, Hamaoka K. Adoptive transfer of macrophages ameliorates renal fibrosis in mice. Biochem Biophys Res Commun. 2005;332:11–16. doi: 10.1016/j.bbrc.2005.04.083. [DOI] [PubMed] [Google Scholar]
  • 162.Harty MW, Papa EF, Huddleston HM, et al. Hepatic macrophages promote the neutrophil-dependent resolution of fibrosis in repairing cholestatic rat livers. Surgery. 2008;143:667–678. doi: 10.1016/j.surg.2008.01.008. [DOI] [PubMed] [Google Scholar]
  • 163.Mitchell C, Couton D, Couty JP, et al. Dual role of CCR2 in the constitution and the resolution of liver fibrosis in mice. Am J Pathol. 2009;174:1766–1775. doi: 10.2353/ajpath.2009.080632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Muriel P, Escobar Y. Kupffer cells are responsible for liver cirrhosis induced by carbon tetrachloride. J Appl Toxicol. 2003;23:103–108. doi: 10.1002/jat.892. [DOI] [PubMed] [Google Scholar]
  • 165.Roggin KK, Papa EF, Kurkchubasche AG, Tracy TF., Jr Kupffer cell inactivation delays repair in a rat model of reversible biliary obstruction. J Surg Res. 2000;90:166–173. doi: 10.1006/jsre.2000.5879. [DOI] [PubMed] [Google Scholar]
  • 166.Fallowfield JA, Mizuno M, Kendall TJ, et al. Scar-associated macrophages are a major source of hepatic matrix metalloproteinase-13 and facilitate the resolution of murine hepatic fibrosis. J Immunol. 2007;178:5288–5295. doi: 10.4049/jimmunol.178.8.5288. [DOI] [PubMed] [Google Scholar]
  • 167.Madala SK, Pesce JT, Ramalingam TR, et al. Matrix metalloproteinase 12-deficiency augments extracellular matrix degrading metalloproteinases and attenuates IL-13-dependent fibrosis. J Immunol. 2010;184:3955–3963. doi: 10.4049/jimmunol.0903008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Cabrera S, Gaxiola M, Arreola JL, et al. Overexpression of MMP9 in macrophages attenuates pulmonary fibrosis induced by bleomycin. Int J Biochem Cell Biol. 2007;39:2324–2338. doi: 10.1016/j.biocel.2007.06.022. [DOI] [PubMed] [Google Scholar]
  • 169.Hironaka K, Sakaida I, Matsumura Y, Kaino S, Miyamoto K, Okita K. Enhanced interstitial collagenase (matrix metalloproteinase-13) production of Kupffer cell by gadolinium chloride prevents pig serum-induced rat liver fibrosis. Biochem Biophys Res Commun. 2000;267:290–295. doi: 10.1006/bbrc.1999.1910. [DOI] [PubMed] [Google Scholar]
  • 170.Uchinami H, Seki E, Brenner DA, D’Armiento J. Loss of MMP 13 attenuates murine hepatic injury and fibrosis during cholestasis. Hepatology. 2006;44:420–429. doi: 10.1002/hep.21268. [DOI] [PubMed] [Google Scholar]
  • 171.Imamura M, Ogawa T, Sasaguri Y, Chayama K, Ueno H. Suppression of macrophage infiltration inhibits activation of hepatic stellate cells and liver fibrogenesis in rats. Gastroenterology. 2005;128:138–146. doi: 10.1053/j.gastro.2004.10.005. [DOI] [PubMed] [Google Scholar]
  • 172.Gross TJ, Hunninghake GW. Idiopathic pulmonary fibrosis. N Engl J Med. 2001;345:517–525. doi: 10.1056/NEJMra003200. [DOI] [PubMed] [Google Scholar]
  • 173.Vaglio A, Salvarani C, Buzio C. Retroperitoneal fibrosis. Lancet. 2006;367:241–251. doi: 10.1016/S0140-6736(06)68035-5. [DOI] [PubMed] [Google Scholar]
  • 174.Berenguer M, Aguilera V, Prieto M, et al. Significant improvement in the outcome of HCV-infected transplant recipients by avoiding rapid steroid tapering and potent induction immunosuppression. J Hepatol. 2006;44:717–722. doi: 10.1016/j.jhep.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 175.Issa R, Zhou X, Trim N, et al. Mutation in collagen-1 that confers resistance to the action of collagenase results in failure of recovery from CCl4-induced liver fibrosis, persistence of activated hepatic stellate cells, and diminished hepatocyte regeneration. FASEB J. 2003;17:47–49. doi: 10.1096/fj.02-0494fje. [DOI] [PubMed] [Google Scholar]

RESOURCES