You are on page 1of 12

Immunity

Review

Alternative Activation of Macrophages:


Mechanism and Functions
Siamon Gordon1,* and Fernando O. Martinez1,*
1Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK

*Correspondence: siamon.gordon@path.ox.ac.uk (S.G.), fernando.martinezestrada@path.ox.ac.uk (F.O.M.)


DOI 10.1016/j.immuni.2010.05.007

The concept of an alternative pathway of macrophage activation has stimulated interest in its definition,
mechanism, and functional significance in homeostasis and disease. We assess recent research in this field,
argue for a restricted definition, and explore pathways by which the T helper 2 (Th2) cell cytokines interleukin-
4 (IL-4) and IL-13 mediate their effects on macrophage cell biology, their biosynthesis, and responses to
a normal and pathological microenvironment. The stage is now set to gain deeper insights into the role of
alternatively activated macrophages in immunobiology.

Introduction tion factor STAT6 in their signaling (Heller et al., 2008). These
The concept of an alternative pathway of macrophage activation and other experiments showed that these Th2 cytokines were
induced by the T helper 2 (Th2) cytokines interleukin-4 (IL-4) and not simply inhibitory, but subtle modulators of macrophage func-
IL-13, distinct from interferon-g (IFN-g)-mediated classical acti- tions. We proposed that IL-4 induced an ‘‘alternative’’ form of
vation, has gained considerable ground over the past decade. activation in macrophages (Stein et al., 1992).
Analysis of marker molecules in the mouse has revealed a stereo- Subsequently, other workers introduced the term M1 and M2
typic signature of expressed genes induced by IL-4 and IL-13, to parallel macrophage activation with T helper cell polarization,
contrasting with characteristic changes induced by IFN-g, which subdivided M2 classes into subgroups, and in some cases,
mediates Th1 cell-type activation of macrophages. Phenotypes termed all nonclassical activation phenotypes as ‘‘alternative’’
associated with alternative activation of macrophages (AAMs) (discussed in Gratchev et al., 2001; Mantovani et al., 2008;
and classical activation of macrophages (CAMs) can be distin- Mosser and Edwards, 2008). We have argued that too loose
guished from the direct effects of microbial stimuli, such as lipo- a definition obscures the study of mechanism and discovery of
polysaccharide, which induce innate macrophage activation, potential therapeutic agents. Even a restricted definition of IL-4
often through Toll-like receptors (TLRs). Innate stimuli are able and IL-13 dependence needs to take into account the complex
to synergize with CAMs, to achieve full expression of macro- effects of cell differentiation, interactions with other receptors,
phage effector pathways. and signaling pathways, and the role of tissue microenvironment
Investigators have implicated AAMs in a range of physiologic in modulating the macrophage phenotype. Monocyte and mac-
and pathological processes, including homeostasis, inflamma- rophage heterogeneity and nomenclature are discussed below
tion, repair, metabolic functions, and malignancy. This is in addi- and elsewhere (Gabrilovich and Nagaraj, 2009; Geissmann
tion to their established role in immunity, hypersensitivity, allergy, et al., 2010).
parasitic infections, and their sequelae, such as fibrosis. Recent
genome-wide analysis, although broadly supporting earlier stud- Analytic Approaches
ies with limited numbers of markers, has underscored the There are two distinct, complementary approaches to the
heterogeneity and plasticity of macrophage gene expression, study of AAMs. The first, ‘‘forward analysis,’’ flows from recep-
adding to the confusing nomenclature of monocyte and tissue tor-dependent recognition, through signal transduction, gene
macrophage subpopulations (Geissmann et al., 2010). In this expression, and protein production. The second, ‘‘reverse anal-
review, we assess current knowledge, point to the need to ysis,’’ deduces from phenotypic markers the mechanism of initi-
restrict the concept of AAM, and consider newer findings ation. Studies with isolated macrophages and in situ analysis
regarding mechanisms and therapeutic potential. in wild-type and genetically modified mice made it possible to
Our own interest in IL-4 modulation of the macrophage pheno- combine these approaches. Forward analysis utilizes cytokine,
type arose in the late 1980s from experiments with human blood receptor, or signaling-deficient animals to define an AAM pheno-
monocyte-derived macrophages and mouse peritoneal macro- type, including Arginase 1 (ARG1), Ym1 and Ym2, Fizz1, MRC1,
phages. At the time, the role of this cytokine was believed to and selected chemokines (Loke et al., 2002). Signature markers,
be mainly anti-inflammatory, resulting from its ability to suppress in combination, have then been used to infer an AAM pathway
TNF and IL-6 production in macrophages. Whereas macrophage and mechanism (reverse analysis).
mannose receptor (MRC1) expression and its endocytic function There are obvious caveats to the use of reverse analysis alone,
were selectively downregulated by IFN-g (Mokoena and Gordon, as required in human studies in situ: substantial species differ-
1985), it was upregulated by IL-4 (Stein et al., 1992) and IL-13 ences exist in available markers, and established markers such
(Doyle et al., 1994) and depended in vitro and in vivo on as Arg1 can be induced by other pathways (El Kasmi et al.,
IL4Ra1, the common IL-4 and IL-13 receptor alpha chain (Line- 2008). Even clusters of multiple signature markers can be inde-
han et al., 2003). Others showed a critical role for the transcrip- pendent of the IL-4 and IL-13 receptor pathway (Daley et al.,

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 593


Immunity

Review

2010; Stout, 2010). We lack markers for human AAM, but matory cytokines, such as IL-10 and TGF-b. Proinflammatory
these should become available with current microarray and cytokines include IL-1b, TNF, and IL-6. Through potent mem-
proteomic studies. Combined forward and reverse analysis brane protein assemblies they generate reactive oxygen, nitro-
in the mouse will validate the link between recognition and gen, and arachidonate metabolites.
response. With humans, our dilemma mimics the problem of The study of chromatin structure and epigenetic control of
diagnosis in clinical medicine, where a cluster of symptoms and gene expression in macrophages is in its infancy. Given the
signs cannot be assigned to a particular cause without sup- range of receptors and potential ligands, it may be invidious to
portive evidence. single out IFN-g, IL-4, and IL-13 receptor pathways as central
regulators of macrophage activation. However, given their often
Toward an Integrated Approach polarized production in the immune response, these contrasting
Before turning to the pathways and functions of AAM, we states of activation provide an important conceptual framework.
summarize general features of monocyte and macrophage Other receptors participate in forming signaling platforms and
heterogeneity (Gordon, 2008). Macrophages arise from hemo- complexes, e.g., G protein-coupled receptors and integrins.
poietic progenitors (Geissmann et al., 2010), which differentiate, Crosstalk at all levels impinges on altered transcription. It is
directly or via circulating monocytes, into subpopulations of therefore possible to conceive an infinite number of potential
tissue macrophages and closely related myeloid dendritic cells phenotypes; however, the conserved, stereotypic macrophage-
(Steinman and Idoyaga, 2010), the latter specialized to present specific signature for IFN-g versus IL-4- and IL-13-dependent
antigens to naive T helper lymphocytes. Osteoclasts arise from activation provides useful insights into physiologic and patho-
monocyte precursors to form multinucleated giant cells, impli- logic mechanisms.
cated in resorption of bone. Monocyte and macrophages share Figure 1 presents a schematic model of macrophage activa-
properties with granulocytes, especially neutrophils, and to tion. The first stage, differentiation, depends on growth factors
lesser extent with B lymphocytes. Monocyte and macrophages such as GM-CSF or M-CSF; the second, priming by IFN-g
migrate through several body compartments, including bone (CAMs) or IL-4 and IL-13 (AAMs); and the third, a localizing stim-
marrow, blood, lymphoid, and all nonhematopoietic tissues. ulus delivered by a TLR or analogous receptor. This additional
Resident macrophages are present in organs constitutively, in stimulus promotes induction of a full complement of classical
the absence of overt inflammation, and perform trophic as or alternative activation functions. In the case of AAMs, this
well as homeostatic roles in the removal of apoptotic cells, may arise in a Th2 cell-biased environment, e.g., chronic para-
serving as sentinels of injury and infection. Tissue macrophages sitic infection, followed by a secondary microbial challenge.
can replicate locally, but are terminally differentiated, turning Counteracting negative controls contribute to a fourth stage of
over at different rates, depending on the stimulus and tissue resolution and repair. Once established, some properties of full
environment. alternative activation can be reversible (plasticity) whereas
Macrophages differ morphologically and phenotypically in others may be irreversible (Stout et al., 2005).
organs such as liver, spleen, lung, gut, and brain, interacting Cytokines in AAM Induction
with matrix and other cell types. They are active in biosynthesis IL-4 and IL-13 are the prototypical direct inducers of AAMs, but
and express a wide range of receptors, recognizing foreign as other cytokines such as IL-33 and IL-25 amplify AAM induction
well as normal and abnormal cells and host-derived products. indirectly, through Th2 cells (for review on CD4+ T cell heteroge-
Through constitutive and induced endocytosis, phagocytosis, neity, see O’Shea and Paul, 2010). IL-4 and IL-13 are produced
and secretion of various products, including cytokines, growth by various innate cells, including non-B non-T cells (Moro et al.,
factors, and metabolites, they perform both trophic and toxic 2009; Neill et al., 2010), mast cells, basophils, eosinophils, NKT
functions, serving as a widely dispersed mononuclear phago- cells, and even macrophages themselves. Apart from activated
cyte system during development and throughout adult life. Th2 cells and B cells, epithelia and tumor cells are potential sour-
They contribute to tissue remodeling, host defense in innate ces in tissues. The availability of reporter mice has contributed to
and adaptive immunity, and many disease processes. the analysis of IL-4 production in vivo, during allergic inflamma-
Monocytes are the main source of newly recruited tissue tion and infection (Mohrs et al., 2005).
macrophages in infection, granulomata, atherosclerosis, Alz- The role of IL-5, another Th2 cell cytokine that binds to distinct
heimer’s disease, and tumors. Differentiation and activation of receptors on macrophages, is unclear. IL-10, produced by CD4+
macrophages depend on specific growth and differentiation Th2 cells, macrophages, and B cells as well as tumor cells, is
factors, their receptors, signaling pathways, and transcription a potent downregulator of macrophage gene expression, modu-
factors. Macrophages migrate to local sites of injury and infec- lating IL-4 and IL-13 and IFN-g actions. IL-21 enhances AAMs by
tion, contributing to acute and chronic inflammation, locally driving IL-4R expression (Pesce et al., 2006). IL-33, a Th2 cell
and systemically. In addition they acquire enhanced cytotoxic, product, binds to ST2L, a subunit of the IL-33 receptor, to
antimicrobial, and inhibitory activities, initiate repair, and resolve amplify Th2 cytokine production and AAM induction (Kurow-
inflammation. ska-Stolarska et al., 2009).
Macrophages express opsonic and a range of nonopsonic IL-25, a member of the IL-17 family also produced by Th2
receptors, TLRs, RIG-I-like and Nod-like sensing receptors, on cells, induces production of multipotent hemopoietic progeni-
their surface and in vacuolar and cytosolic compartments. Other tors in bone marrow and at sites of extramedullary hemopoiesis,
receptors include families of regulatory molecules, lectins, and giving rise to a range of myeloid cells, including monocytes and
scavenger receptors. Their secretory repertoire includes anti- macrophages (Saenz et al., 2010). IL-25 activates antigen-
bacterial and proteolytic enzymes, chemokines, and anti-inflam- specific responses in mucosal tissues, such as the colon. In

594 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.


Immunity

Review

AAMs, depending on specific ligand-receptor interactions, and


their interplay is largely unexplored. The signaling pathways acti-
RESOLUTION vated by the Type I and II receptors involve STAT6, although
other STATs can participate (Bhattacharjee et al., 2006).
IL-10, TGF-β, lipoxins
STAT6 plays a central role in gene regulation by IL-4 and IL-13,
including the differentiation of Th2 cells, production of IgE, che-
ACTIVATION mokines, and mucus at sites of allergic inflammation.
The differential effects of IL-4 and IL-13 on different macro-
TLR stimuli phage populations from mice and humans have been examined
by correlating the magnitude of response with the expression of
each receptor (Junttila et al., 2008). Early, low amounts of IL-4
PRIMING promote a Th2 cell type response; as the stimulus persists,
IFN-γ<-> IL-4 IL-13 becomes sufficient to induce responses in both hemopoi-
etic and nonhemopoietic cells, making it the dominant Th2
effector cell cytokine.
DIFFERENTIATION Type I and Type II receptor complexes have also been
compared in mouse bone marrow-cultured macrophages and
M-CSF <-> GM-CSF human monocytoid cells (Heller et al., 2008). IL-4 action via the
Type I receptor was more potent with subtle differences in
signaling and target gene activation, which bear on the contribu-
tion of each cytokine to pathophysiology, e.g., in the airway.
Type I IL-4R selectively activated IRS-2, which interacts with
the adaptor Grb-2 and with PI-3 kinase, thereby initiating
Figure 1. Paradigm of Macrophage Activation multiple signaling pathways. IL-4 more readily induced a subset
Macrophages undergo activation to fulfill specific functional roles during of commonly used mouse AAM signature genes (Arg1, Fizz1,
inflammation and its resolution. The phenotype is imparted by the complex
mixture of cytokines, metabolites, plasma proteins, and microbial ligands Ym1), whereas Mrc1 mRNA was induced comparably by IL-4
present in the inflammatory milieu. The development into mature and fully acti- and IL-13. The expression of each receptor polypeptide influ-
vated macrophages can be artificially divided into successive stages. In this enced its affinity for each cytokine.
scheme we have placed these stimuli in a hierarchical order.
A first phase is envisaged in which the recruited monocytes mature into
IL-13Ra1-deficient cells were used to define distinct require-
macrophages. In vitro experiments suggest that the balance of MCSF, ments for IL-13 and IL-4 in AAM induction by parasitic infection
GMCSF, retinoic acids, and lipoproteins determines substantial differences (Ramalingam et al., 2008). Whereas IL-4 could stimulate both
in the phenotype of the mature macrophage. During continuous recruitment,
Type I and Type II receptor signaling, the Type II receptor was
monocytes are exposed to varying concentrations of mediators, inducing
a second phase of priming by cytokines. The priming term derives from the essential for protection against schistosomiasis and resistance
fact that these stimuli per se are not very strong, but influence the inflammatory to Nippostrongylus braziliensis and its expulsion from the gut.
potential of macrophages and their response to other stimuli. During the third Allergen-induced airway hyperreactivity, mucus secretion, and
phase of activation, macrophages reach a mature functional phenotype in
response to microbial and opsonic stimuli such as antibody complexes. fibrosis were IL-13Ra1 dependent, whereas this receptor was
If the macrophage survives its inflammatory task, it should undergo the final redundant for AAM and fibroblast activation. Expression of a
phase commonly referred as to deactivation. We have chosen the term reso- subset of genes, however, was reduced in the IL-13Ra1-defi-
lution, because while the macrophage proinflammatory potential is deacti-
vated, it also undergoes functional changes that allow it to clear debris and
cient mice, including IL-13Ra2, Muc5a, Ccl11, and Ccl3. Surpris-
express general repair functions. IL-10, TGF-b, and a multitude of anti-inflam- ingly, the Th2 cell response was exacerbated, possibly because
matory mediators such as nucleotides, lipoxins, and glucocorticoids are of lack of feedback inhibition. These results show that IL-13 is the
included in this category. more pathogenic cytokine, while consistent with the notion that
IL-4 contributes most to expansion of a Th2 cell response. The
principle, all cytokines that regulate Th1 and Th2 cell responses role of epithelial and fibroblast receptors in vivo requires further
can affect AAMs indirectly. study.
The IL-4 and IL-13 Receptors, Signaling, and Function IL-13 induced phosphorylation of IL-13Ra1 and IL-4Ra1, but
Although IL-4 and IL-13 have many overlapping activities on not of the IL-13Ra2 or IL-2Rgc (Bhattacharjee et al., 2006; Roy
macrophages, they have distinct functions. Receptors for IL-4 et al., 2002). Phosphorylated Jak2 associated with IL-4Ra1 and
are mainly expressed on hematopoietic cells, whereas IL-13 phosphorylated Tyk2 with IL-13Ra1. STAT1, STAT3, STAT5A
receptors are also present on nonhematopoietic cells (Figure 2). and B, and STAT6 were all tyrosine phosphorylated and acti-
IL-4 and IL-13 receptors share a common alpha chain, but there vated in response to IL-13. Furthermore, IL-13 induction of the
are specific IL-13Ra1 and IL-13Ra2 subunits (Kawashima et al., gene that encodes 15-LO (15 lipoxygenase) expression required
2006). The IL-4Ra1 can signal through the common gamma p38 MAP kinase-mediated phosphorylation of STAT1 and
chain (Type I IL-4R) or partner IL-13Ra1 (Type II IL-4R). The STAT3. 15-LO was regulated by a signaling complex with PKCd
IL-13Ra2 chain, thought to form a decoy receptor with extraordi- kinase and both tyrosine and serine phosphorylation of STAT3.
nary affinity for IL-13 (Lupardus et al., 2010), but with little PKCd was thus required for IL-13-induced 15-LO expression
signaling capacity, has been implicated in STAT6-independent and subsequent inhibition of the lipid mediator leukotriene B4.
fibrogenesis and tumor-host interactions (Fichtner-Feigl et al., IL-4 has substantial effects on SHIP1 and SHIP2, phospha-
2006, 2008b). There are therefore three distinct subtypes of tases that skew macrophage polarization (Rauh et al., 2005).

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 595


Immunity

Review
Figure 2. IL-4 and IL-13 Signaling
Type I Type II Machinery
IL-4 binds IL-4Ra and then recruits either IL-2Rg
IL-4 IL-13
chain (to form the type I receptor) or the IL-13Ra1
IL-4
chain (to make the type II receptor), depending
IL-13 upon the cell type (Heller et al., 2008). However,
IL-13 can only signal through type II receptor as
IL-4Rα IL-2Rγ IL-4Rα IL-13Rα1 IL-13Rα2 it binds IL-13Ra1 and recruits IL-4Ra (LaPorte
et al., 2008). The common signaling pathways
shared by both the cytokines involve phosphoryla-
tion of STAT6, which leads to dimerization and
translocation of STAT6 to the nucleus where it
Jak1 Jak3 Jak1 Tyk2 ? regulates the transcription of a number of genes.
The type I receptor is predominantly found in cells
STAT6 IRS-2 STAT6
of hemopoietic origin such as lymphocytes and
Grb2 STAT3 (?) granulocytes, resulting from restricted expression
pattern of the IL-2Rg chain. The type II IL-4 recep-
STAT6 STAT6 tor is mainly expressed by cells of nonhemopoietic
PI3K origin such as epithelial cells and fibroblasts.
P P
Some cell types, such as macrophages, express
both receptor types. IL-13 also binds IL-13Ra2,
Hematopoietic cell Nonhematopoietic cell with an extraordinarily high affinity (Lupardus
et al., 2010), which is thought to function as a
STAT6 STAT6 decoy receptor (Chiaramonte et al., 2003), though
PP PP evidence exists for its potential to signal in
selected circumstances (Fichtner-Feigl et al.,
2006).

STAT6
PP

IL-4 triggers proteosomal degradation of both of these phospha- Before global mRNA and protein repertoire analysis, knowl-
tases in bone marrow-cultured mouse macrophages, enhancing edge of AAM markers was restricted to a few molecules.
the PI3K and Akt kinase pathways (Ruschmann et al., 2010). MRC1, MHC class II, FN1, and CCL22 were upregulated in com-
SHIP represses IL-4 production from basophils, thereby indi- mon in both human and mouse macrophages, leading to the
rectly controlling the AAM phenotype (Kuroda et al., 2009). impression that they responded similarly to IL-4 (Gratchev
The mTOR adaptor is an evolutionarily conserved member of et al., 2001; Mantovani et al., 2005; Martinez et al., 2009). Subse-
the PI3K family that forms the core of distinct TORC1 and quent reports showed that important AAM markers in mouse,
TORC2 signaling complexes, broadly responsible for the regula- such as Fizz1, Ym1, and Ym2, lacked homologs in humans,
tion of metabolism, growth, and proliferation (Delgoffe et al., and others such as Arg1 were not upregulated in human macro-
2009). Recent studies have shown that mTOR is essential in phages by IL-4, despite gene homology.
dictating T helper cell differentiation, TORC1 for Th1 and Th17, Microarrays have made it possible to investigate effects of IL-4
and TORC2 for Th2 cell differentiation. It would be interesting in macrophages of both species; however, given the limited
to examine macrophage polarization in mice after genetic abla- number of published studies in human or mouse macrophages,
tion of these distinct pathways. a consensus of markers is hard to establish, particularly for
There are few reports of epigenetic modification or microRNA humans (Syed et al., 2007; Martinez et al., 2006; Scotton et al.,
analysis in AAMs. Signature marker genes of IL-4-treated mouse 2005). Genes that appear to be modulated in common by IL-4
macrophages, Arg1, Ym1, Fizz1, and MRC1, revealed reciprocal in macrophages, monocytes, and perhaps circulating precur-
changes in histone H3K4 and H3K27 methylation; the latter sors include ALOX15, FN1, CD23a, MAO-A, GAS6, FXIIIA1,
methylation markers were removed by the H3K27 demethylase CCL26, CCL22, and IL-17RB. Many of the expected AA markers
Jmjd3 (Ishii et al., 2009). These changes depended on activation could also be regulated by M-CSF, while a proportion of the
of STAT6, which bound to consensus sites at the Jmjd3 pro- genes were uniquely regulated by IL-4.
moter, contributing decreased di- and trimethylation of H3K27, Cell Biology and Membrane Dynamics in AAM
as well as transcriptional activation of AAM marker genes. In Apart from their effects on specific receptors and antigen
vivo studies with Schistosome egg granulomas were consistent markers, IL-4 and IL-13 profoundly alter membrane flow in
with evidence of chromatin remodeling in AAM. macrophages (Kzhyshkowska and Krusell, 2009; Montaner
The kinase Akt regulates LPS-induced microRNA in macro- et al., 1999; Varin and Gordon, 2009). Human blood monocyte-
phages and is implicated in LPS tolerance (Androulidaki et al., derived macrophages displayed enhanced flow from the
2009). The isoforms Akt1 and Akt2 have differential effects on plasma membrane, through early endosomes into an expanded
TLR4 and SOCS1 signaling in macrophages, depending on the vacuolar compartment, accompanied by enhanced fusion with
microRNAs let7e and miR155. The effect of IL-4 and IL-13 on lysosomes (Montaner et al., 1999). These effects are distinct
Akt isoforms has not been reported. Other studies have impli- from those induced by other cytokines, such as IFN-g and
cated microRNAs in regulation of macrophage activation TNF. Apart from receptor-mediated endocytosis, e.g., via man-
(Taganov et al., 2006; Tili et al., 2007). nose, transferrin, and folate receptors (Puig-Kroger et al.,

596 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.


Immunity

Review

2009), fluid phase endocytosis is also enhanced. The effects of Ym1 and Ym2 belong to a family of chitinase-like molecules.
IL-4 and IL-13 on phagocytosis have been variable (Varin et al., Chilectins are strongly induced by Th2 cell stimuli on macro-
2009). phages as well as epithelium. Chilectins have undergone consid-
In our hands, with mouse peritoneal macrophages, these cyto- erable diversification during evolution, in many cases losing
kines selectively downregulate phagocytosis of a range of parti- enzyme activity, as with Ym1 and Ym2. These molecules bind
cles, by approximately 50%, independent of specific receptor chitin in insects, Helminth eggs, and Nematode pharynx. Chitin
expression (Varin et al., 2009). Uptake of Neisseria meningitidis induces Th2 cell responses (Reese et al., 2007; Voehringer
via Class A Scavenger receptors, of zymosan particles via Dec- et al., 2006) and can be degraded by glucanases, and MRC1
tin-1, and of latex particles is similarly decreased. Reduced contributes to chitin binding.
uptake is associated with reduced Akt phosphorylation, phago- Fizz1, also known as Relma or Retnla, a secreted protein
cytic cup closure, and PI3 Kinase activation. The mechanism produced by macrophages, epithelium, and eosinophils, is
and functional significance of these observations require further highly induced by IL-13. Fizz1 found in lung allergy and asthma
study. may inhibit inflammation (Nair et al., 2009) Fizz / AAM display
In contrast with reduction of phagocytosis in AAMs, IL-4 is exaggerated antigen-specific Th2 cell differentiation. Fizz1 pro-
required for the efficient uptake of apoptotic neutrophils by tein binds to macrophages and effector CD4+ Th2 cells, inhibit-
macrophages from murine gp91 phox-deficient mice, a model ing Th2 cytokine production via Bruton’s tyrosine kinase. Fizz1
of chronic granulomatous disease (Fernandez-Boyanapalli plays a role in metabolism, as well as regulating T cell differenti-
et al., 2009). ation and eosinophil migration. Others have reported a proinflam-
Another feature of IL-4 and IL-13 effects on macrophages is matory role for Fizz1 and related resistin-like molecules (Munitz
the induction of homokaryon formation by macrophage fusion et al., 2008).
(Helming and Gordon, 2009; Moreno et al., 2007). Foreign
body giant cell formation depends on IL-4, whereas osteoclast Involvement of AAMs in Selected Pathological
formation, induced by RANKL cytokine, is inhibited by IL-4 Processes
(Moreno et al., 2003). Independent studies have demonstrated Role of AAM in Inflammation and Immunity
murine multinucleated giant cell formation by IL-4 and IL-13 AAMs are influenced by, and act upon, many processes associ-
and studied its mechanism. Fusion depends on STAT6 signaling ated with inflammation, immunity, and hypersensitivity. AAMs
and is homotypic, i.e., both fusing partners need to express counteract proinflammatory and cellular immune effector mech-
STAT6 (Moreno et al., 2007). Several surface molecules have anisms, serving regulatory as well as inhibitory functions, to
been implicated in fusion, including E-Cadherin, DC-STAMP shape the nature of the response. For example, IL-4 induces
(dendritic cell-specific transmembrane protein), and CD36, expression of SOCS1 in human and mouse macrophages via
which is thought to interact with patches of exposed PS on STAT6. SOCS1 feedback inhibits IL-4 signaling to limit expres-
fusion partners (Helming and Gordon, 2009). Other molecules sion of STAT6 responsive genes and the AAM pathway (Dicken-
such as DAP-12 induce a fusogenic phenotype required on sheets et al., 2007).
both fusion partners (Helming et al., 2008). The effects of IL-4 Although intimately associated with IL-10, TGF-b, and other
and IL-13 are not necessarily associated with enhanced expres- agents of more profound deactivation, IL-4 and IL-13 promote
sion of fusogenic surface molecules, but may result from clus- a distinctive inflammation and cellular immune response. For
tering in the plasma membrane or altered signaling. example, NO generated by IFN-g-induced iNOS is shut down
With human monocyte-derived macrophages, IL-4 and IL-13 in macrophages (it is induced in epithelial targets), with a shift
accelerate fusion in vitro, but other culture variables, e.g., to Arg1. Interactions with mast cells, basophils, eosinophils,
substratum, growth factors, and serum, also play a role. The and NKT cells, as well as IgE and selected subclasses of IgG,
significance of giant cell formation in granulomatous inflamma- promote allergy and hypersensitivity. Together they influence
tion is obscure, nor is it known whether IL-4 and IL-13 play vascular permeability, angiogenesis, cell recruitment, smooth
a role in macrophage fusion in nonparasitic infections, such as muscle contraction, goblet cell secretion, mucus production,
the prototypical Langhans giant cells of tuberculosis. and collagen deposition by myofibroblasts (Locksley, 2010).
Function of Signature Markers in AAM Several questions remain understudied. For example, the
In the case of enzyme markers such as Arg1, there has been interactions between AAMs and other cells of the innate and
extensive investigation of its possible role in alternative versus acquired immune system, by contact, and secretion, require
classical activation (De’Broski et al., 2010; El Kasmi et al., attention. Do AAMs influence DCs, which themselves are
2008; Herbert et al., 2004; Pesce et al., 2009a). Although its affected by Th2 cytokines, and can AAMs function as antigen-
upregulation in the mouse can be a useful indicator of IL-4 and presenting cells (Loke et al., 2000; Rodriguez-Sosa et al.,
IL-13 effects on macrophages, it can be upregulated by other 2002)? Is the reported upregulation of MHCII significant? Apart
pathways. Its demonstration in human macrophages has been from modulating inflammation, can AAM kill microbial and host
problematic. Its possible role in fibrosis is described below. targets, particularly parasites? Arg1 has been put forward as
Enhanced expression and endocytosis of MRC1 are conserved a candidate. Do IL-4-induced homokaryons participate in this
features in both mouse and human macrophages; enhanced process? Perhaps killing is indirect through eosinophils or
uptake of pathogens through carbohydrate recognition may neutrophils. In sum, the metabolic and secretory activities of
contribute to infection. However, its immunologic role is AAM favor trophic rather than lytic functions, clearance of
unknown. A possible clue is via a role in Th17 cell differentiation apoptotic corpses rather than induction of necrosis, and induc-
(van de Veerdonk et al., 2009). tion of tolerance rather than autoimmunity.

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 597


Immunity

Review

Inflammasome Activation subverted by pathogen-induced reprogramming of the macro-


Little is known about inflammasome activation and IL-1b release phage to acquire an AAM phenotype. Instead of the host-protec-
by AAMs, compared with CAMs. Decrease of caspase 1 expres- tive CAMs, it induces many characteristics of AAMs, by unknown
sion and pro-IL-1b processing has been described for human sensing receptors within the phagocytic vacuole and, on escape
monocytes stimulated with IL-13 (Scotton et al., 2005). A sim- into the cytoplasm, by additional cytosolic sensors. The macro-
ilar downregulation is observed in some human macrophage phages are induced to produce IL-4 and IL-13, which amplify the
models. An intriguing study by Pelegrin and Surprenant exam- AAM phenotype through autocrine and paracrine mechanisms.
ined the role of extracellular ATP and purinergic receptors in The canonical signature markers are induced, depending on
a spectrum of polarized macrophages. In intermediate, but not the IL-4 and IL-13 common alpha chain receptor and STAT6,
fully AAMs, P2X7R activation uncouples NLRP3 inflammasome incapacitating the cell’s antimicrobial responses, and allowing
maturation (Pelegrin and Surprenant, 2009). ATP acts, instead, FT to replicate freely.
through pyrophosphate to inhibit IL-1b release. Similar though less dramatic exploitation of AAMs may con-
Role of AAMs in Infection, Fibrosis, tribute to persistent mycobacterial infection (Kahnert et al.,
and Immunopathology 2006). Another issue is the secondary infection of Th2 cell-
The generation and role of AAMs has been studied extensively in biased infected hosts with opportunistic or pathogenic bacteria.
parasitic disease models (e.g., Anthony et al., 2006; Fallon et al., Challenge of IL-4 or IL-13 pretreated macrophages with Neisse-
2006; Hesse et al., 2001; Holscher et al., 2006; Reece et al., ria meningitidis can potentiate the release of proinflammatory
2006), providing the best evidence for a regulatory, protective cytokines (Varin et al., 2009); AAMs can thus contribute to septic
role of AAMs in these Th2 cell-dominated infections. Earlier shock.
studies, culminating in macrophage-specific genetic ablation Pathogens such as Aspergillus and Candida utilize TLR-2
experiments, implicated the common IL-4Ra1 chain in Nippos- and TLR-6 and the beta glucan receptor Dectin-1 to infect
trongylus resistance. IL-13 is critical for worm expulsion and macrophages (Brown, 2006). AAMs express enhanced Dec-
also plays an important role in Schistosome egg granuloma tin-1 as well as mannose receptor, which is delivered to the
formation and fibrosis. Murine markers of AAMs have provided nascent phagosome intracellularly (Heinsbroek et al., 2009).
a remarkably conserved signature of gene expression, consid- Dectin-1 contains a hemi-ITAM (immunoreceptor tyrosine-
ering the diversity of organisms studied. Parasite-specific based activation) motif in its cytoplasmic domain, essential
features are associated with particular sites of migration during for signaling, through Syk kinase and CARD9, thus inducing
complex life cycles, e.g., in skin, lung, gut, and liver, illustrating proinflammatory cytokines and activating a respiratory burst.
the importance of tissue microenvironment in tuning the Th2 The receptor contributes to host resistance to Candida and
cell response. AAMs respond to Th2 cytokines and in turn regu- Pneumocystis. However, it is not clear whether AAMs are bene-
late T helper cell functions. Delayed healing or excessive fibrosis ficial or deleterious to the host, because other innate (comple-
occurs in mice with dysfunctional AAMs (Wilson and Wynn, ment, neutrophils) and adaptive mechanisms, including clas-
2009). sical activation by IFN-g and Th17 cell activation, come into
Major questions remain unresolved regarding the role, if any, play.
of AAM in parasite killing and in activation or suppression of Respiratory syncytial virus (RSV), a major cause of morbidity in
different T cell subsets. Can AAM serve as parasite antigen-pre- children, employs a similar mechanism of AAM induction to that
senting cells for T cells? Chemokines produced by AAMs described for Francisella, although in this instance the outcome
promote recruitment and activation of eosinophils, mast cells, is beneficial to host repair and diminishes host pathology
and basophils, themselves sources of Th2 cytokines, as well (Figure 3B; Shirey et al., 2010). Studies with mouse macro-
as neutrophils. phages in vitro and with cotton rats in vivo show that AAMs
Interactions of AAMs with antibody subclasses promoted by play a role in the response to infection. The macrophages are
Th2 cytokines have received less attention, but include upregu- induced to express the marker signature of AAMs, which
lation of FcR epsilon for IgE. The range of organisms studied in depends on the IL-4 and IL-13 receptor common alpha chain
the mouse includes H. polygyrus and flatworms, and well- and STAT6, as well as TLR-4 and IFN-b. Infection in humans
studied unicellular parasites include Leishmania and Trypano- may also involve Th2 cytokines and AAMs. Although markers
soma brucei. In humans, filariasis has attracted investigators, are insufficient to establish their role, other cell types, including
but the lack of reliable AAM markers has hampered progress. innate and adaptive immune and epithelial cells, also modulate
Further details on the diversity of host responses to parasitic the host response.
infection can be found in excellent reviews (Jenkins and Allen, Since the earliest reports on AAMs, it has been assumed that
2010; Wilson and Wynn, 2009). these cells promote repair of host tissues after inflammation.
A striking example of the way in which a bacterial pathogen Induction of Arg1 by IL-4 and IL-13 has been implicated in
can subvert the host response to favor its intracellular survival collagen deposition (Hesse et al., 2001) and degradation
is provided by Francisella tularensis (Ft), the causal agent of tular- (reviewed in Wilson and Wynn, 2009). Imbalance in production
aemia (Shirey et al., 2008), illustrated in Figure 3A. This highly and catabolism, associated with prolonged IL-13 effects on
virulent coccobacillus has been studied in the form of a vaccine macrophages, promotes excessive fibrosis. Wynn and his
LVS strain, attenuated for humans but still pathogenic for mice. colleagues and Murray, who has produced a conditional macro-
It enters macrophages by a TLR-2-dependent mechanism, phage-specific deficiency of Arg1, provide evidence to support
utilizing mannose and scavenger receptors, and initiates a tran- as well as question these hypotheses (El Kasmi et al., 2008;
sient inflammatory response, but the host CAM reaction is then Pesce et al., 2009a, 2009b).

598 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.


Immunity

Review

A
Ft IL-4, IL-13

TLR2 IL-4, IL-13


(requires IL-4Rα and STAT6)
Inhibit Ft
replication TLR2-dependent increase in:
FIZZ1, Ym1, mannose receptor,
arginase 1

Macrophage Alternatively
activated macrophage Promotes Ft replication
Exogenous
and endogenous IFN-β IL-10
Proinflammatory cytokines
Kills host

B RSV IL-4, IL-13


Antiviral
Increase IL-4Rα
IFN-β
IL-4, IL-13 TLR4-dependent increase in:
(requires IL-4Rα and STAT6) FIZZ1, Ym1, mannose receptor,
arginase 1

Lung macrophage Alternatively


Steroids
activated macrophage Repair of tissue damage
COX-2
IL-10
Parecoxib
? Predisposition to
PGs Pathology airway hyperreactivity

Figure 3. Alternative Activation Amplification Loops in Infection


(A) Model for the role of AAMs during Francisella tularensis (Ft) LVS infection. Ft infection of macrophages results in an initial proinflammatory response through
a TLR2-dependent pathway (Cole et al., 2007), as well as an unknown intracellular sensor that leads to IFN-b production (Cole et al., 2008) and activation of
caspase-1 through AIM2 and ASC sensors (Cole et al., 2008; Rathinam et al., 2010). IFN-b acts to inhibit bacterial replication, but production of IL-4 and
IL-13 differentiates the macrophages to an AAM phenotype through an IL-4Ra- and TLR2-dependent mechanism that results in a microenvironment that
promotes Ft replication, which is ultimately detrimental to the host.
(B) Model for the role of AAM during RSV infection. RSV infection of macrophages leads to an early induction of proinflammatory cytokines and COX-2, which has
been shown to mediate the observed lung pathology (Richardson et al., 2005). After this early proinflammatory period, macrophages produce their own IL-4 and
IL-13 and differentiate into AAM through an IL-4Ra-, TLR4-, and IFN-b-dependent mechanism. This is thought to promote repair of damage caused by the earlier
inflammatory process by inducing AAM markers including Arg1, FIZZ1, and Ym1. IL-10 acts to downregulate production of inflammatory cytokines.
Figure adapted from Shirey et al. (2010) with permission.

Although sterile foreign body reactions can depend on IL-4, properties of AAMs. Polyamines contribute to induction of
IL-4Ra1 chain, and STAT6, repair of surgical wounds was inde- Cadherin-1, after IL-4 treatment in vitro or Taenia crassiceps or
pendent of this pathway (Daley et al., 2010). The IL-13-specific allergen-induced activation in vivo (Van den Bossche et al.,
receptor was not investigated although it has been shown to 2009). Cadherin-1 expression at the macrophage surface,
be essential for fibrosis in a parasitic model of S. mansoni gran- together with catenin, mediates homotypic (cell fusion) and
uloma formation (Ramalingam et al., 2008). As noted, Arg1 can heterotypic interactions with CD103+ T cells. Alternative activa-
be induced by a variety of different pathways, and cells other tion of macrophages has also been implicated in fibrosis after
than macrophages also express receptors for IL-13. The reduc- gamma herpes virus infection in mice (Gangadharan et al., 2008).
tion of fibrosis in egg granulomata in macrophage-selective It is not clear to what extent Th2 cytokines, AAMs, or fibro-
ablation of the IL-13Ra1, in which induction of AAM is unaf- blasts contribute to noninfectious causes of fibrosis or whether
fected, confirms the existence of AAM-independent pathways combined targeting of IL-13 and TGF-b might prevent or reverse
of fibrogenesis, and also independent of TGF-b or MMP9, which fibrosis.
are enhanced in this model. A remarkable example of a pathogenic role of IL-13 in chronic
Macrophage-specific ablation of Arg1 enhances S. mansoni obstructive pulmonary disease (COPD) has been reported
egg-induced fibrosis, an unexpected result. In wild-type mice, (Holtzman et al., 2009; Kim et al., 2008). COPD is a major cause
granulomata appear in liver after 7–10 weeks and diminish of morbidity and mortality, in which lung failure is associated with
subsequently by Arg1-dependent downregulation, which is chronic inflammation, mucous cell metaplasia, bronchial hyper-
absent in Arg1-deficient mice. Macrophages express several reactivity, and fibrosis. The condition is often attributed to aller-
transporters such as Cat2 (Thompson et al., 2008) as well as gens and adaptive immune mechanisms, but this study provides
enzymes involved in L-arginine uptake and metabolism, but convincing evidence that an initial transient acute viral infection
not a complete urea cycle. Ornithine decarboxylase, hydroxy- in the lung can be followed after a period by activation of NKT
proline, and polyamines have been implicated in additional cells, production of IL-13 by these cells, and by macrophages

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 599


Immunity

Review

themselves. The macrophages upregulate IL-13Ra1 expression reduced in macrophages from the hearts of IL-13 or doubly defi-
and become alternatively activated by an autocrine or paracrine cient IL-13 and IL-4, but not IL-4-deficient, mice.
mechanism. PPARs and AAMs: Role in Metabolism, Inflammation,
In an experimental mouse model of Sendai virus infection, and Atherosclerosis
bronchiolitis is associated with mucus production and airway Since the early observation by Glass and colleagues that PPAR
hyperreactivity to methacholine. After the virus becomes barely gamma is strongly induced by IL-4, PPARs have been investi-
detectable, NKT cells expressing semi-invariant Valpha14-J- gated in relation to lipid metabolism, macrophage polarization,
alpha 18 TCR produce IL-13 through a CD1d-glycolipid-depen- obesity and insulin resistance, and atherogenesis (reviewed by
dent mechanism, initiating a vicious circle of AAM amplification. Glass [Glass and Saijo, 2010] and Chawla [Chawla, 2010]).
In the mouse, the characteristic signature markers Arg1, Ym1 Macrophages express PPARg and PPARd; macrophage-spe-
and Ym2, Fizz1, Mmp12, and Alox12e are now present. cific deficient mice have been utilized in bone marrow chimera
An analogous mechanism has been proposed for human experiments of experimental atherosclerosis. Although there
COPD, with patient transplant-derived material to demonstrate has been progress in analyzing the control of inflammation, the
the presence of NKT cells and IL-13 message and protein in therapeutic application of PPAR ligand agonists via these recep-
macrophages (Holtzman et al., 2009). The critical role of IL-13 tors remains elusive. Macrophages in white adipose tissue
in the mouse model was demonstrated by macrophage deple- in obese versus lean mice differ in phenotype, with features of
tion studies and blockade with soluble IL-13R2 protein. This CAMs in the former and of AAMs in the latter. By contrast,
provocative study shifts the pathogenic mechanism to the innate lipid-laden foam cells in atherosclerotic plaques have a distinct
immune system. Improved AAM markers are needed for human phenotype, not necessarily corresponding to the binary classifi-
tissue analysis and prospective studies in human subjects. cation of M1- and M2-polarized cells. AAMs could play a role in
Plasma Chitinase-1, a member of the chitinase family, allows smooth muscle cell migration and plaque rupture.
quantitative stratification of COPD patients (Agapov et al., 2009). IL-4 enhances uptake and oxidation of fatty acids and biogen-
The question arises of genetic susceptibility to these sequelae of esis of mitochondria, via STAT6, regulating programs controlled
common virus infections, as does the possibility of therapeutic by PPARg and the coactivator protein PGC-1b. PPARg is
targeting of the IL-13 pathway. dispensable for induction of AAM but required to sustain it. It
LPS tolerance, a refractory state in responses of the host and plays a more complex role in limiting proinflammatory gene
macrophages to repeated challenge with LPS, is known to expression by LPS, preventing turnover of NCoR corepressor
involve NF-kB. Patients with sepsis can display features resem- complexes and keeping such genes in the ‘‘off’’ position in
bling alternative activation of monocytes. Porta and colleagues AAMs, thus controlling the initiation, magnitude, and duration
have shown that tolerance and alternative activation (M2 polari- of inflammation.
zation) are related processes mediated by p50 homodimers, PPARg regulates primarily the metabolic phenotype of AAMs,
which lack a transcription activation domain of members of the whereas PPARd regulates the immune repertoire. PPARd is
NF-kB pathway (Porta et al., 2009). Studies were performed a sensor of triglyceride VLDL and contributes to macrophage
with human monocyte-derived macrophages and with mouse numbers and phenotype in liver as well as adipose tissue.
primary macrophages, in vitro and in vivo. P50 was shown to Mono-unsaturated fatty acids, such as oleic acid, synergize
be a negative regulator of classical, M1 polarization and of with IL-4 to enhance expression of AAM signature genes; PPARd
IFN-b production, but played a positive, nonredundant role in may also play a role in the proliferative and anti-inflammatory
induction of several signature M2-associated genes. Markers effects of IL-4 on macrophages.
included selected chemokines (CCL2, CCL17, and CCL22),
Arg1, Ym1, and Fizz1. There were some discrepancies between Role of AAMs in Tumorigenesis
human and mouse phenotypes, however, and no reported Macrophages are a prominent component of the stroma and
involvement of IL-4 and IL-13 expression or of their receptors. leukocyte infiltrate in human and experimental mouse tumors
P50 / mice lacked the characteristic AAM phenotype associ- (DeNardo et al., 2009; Gocheva et al., 2010; Grivennikov et al.,
ated with Taenia crassiceps infection, consistent with the overall 2010; Hallam et al., 2009; Joyce and Pollard, 2009; Mantovani
hypothesis. et al., 2008; Sinha et al., 2007). They interact with malignant
In another model, the prototypical proinflammatory function of epithelial, blood vessels and lymphatics, lymphocytes, innate
TLR4 can be subverted by house dust mite allergen, which cells, and extracellular matrix. They have been implicated in
mimics MD2, its coreceptor, to induce IL-4 production (Tromp- carcinogenesis, the angiogenic switch, local invasion, and
ette et al., 2009). metastasis. Systemic features of malignancy have also been
Although IL-13 has been associated with disease-promoting ascribed to macrophages, locally and outside the actual tumor
activities, it can also play a protective role by virtue of its ability bed. It is established that they contribute trophic functions to
to regulate macrophages in an autoimmune myocarditis model the emergence of nascent tumor clones, phagocytose apoptotic
of disease (Cihakova et al., 2008). Infection of mice with Cox- tumor cells, influence the tissue response to hypoxia and,
sackie virus B3 induces a self-limiting myocarditis in most mouse together with regulatory T lymphocytes, suppress Th1 cell and
strains, but a few strains develop severe myocardiopathy, with cytotoxic T lymphocyte antitumor responses.
Th1 cell type inflammation, myosin antibodies, and fibrosis, The nature of tumor-associated macrophages (TAMs) has
especially in IL-13 / , but not IL-4 / , BALB/c mice. T cells are become of great interest, though confusing because of pheno-
not known to express IL-13Ra, and the protective role of IL-13 typic heterogeneity. TAMs have many properties of AAMs;
has been attributed to macrophages. MRC1 expression was myeloid-derived suppressor cells (MDSC) are less defined and

600 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.


Immunity

Review

in mouse contain infiltrating GR-1 intermediate CD11b+ mono- ruff et al., 2009). Various strategies have been employed to target
cytes, as well as GR-1 high CD11b+ granulocytic cells (Gabrilo- IL-13 through receptor-specific antibodies, chimeric proteins,
vich and Nagaraj, 2009). Some MDSC express F4/80 in the or soluble receptor (Blease, 2008). Proof-of-concept animal
mouse and IL-4Ra, enabling them to respond to both IL-4 and and early human studies have been undertaken, to ameliorate
IL-13. Innate and adaptive T cells as well as tumor cells can asthma and idiopathic lung fibrosis. Species differences need
produce these cytokines, and some tumors also express the to be borne in mind, because in contrast with the mouse, human
IL-4 and IL-13 alpha receptor (Mandruzzato et al., 2009). decoy IL-13 receptor is not secreted from cells (O’Toole et al.,
Several recent studies with experimental tumors in mice 2008). Novel strategies include functional variants based on
(transplantable as well as oncogene-induced) provide striking analysis of human SNPs that demonstrate altered affinity for
evidence that TAMs have much in common with AAMs. They IL-13Ra2 and cell penetrant proteins or small molecules that
contribute to local invasion through cathepsin B and S, promote target the signaling pathway through STAT 6. Structure-function
angiogenesis and tumor growth through VEGF, and induce studies should aid in the development of IL-13-specific
recruitment of other hemopoietic cells, through chemokines therapeutic agents (LaPorte et al., 2008; Mitchell et al., 2010;
(Gocheva et al., 2010). AAMs as well as tumor cells themselves Lupardus et al., 2010).
orchestrate tumor growth and spread through M-CSF, TNF,
IL-10, TGF-b, and other products. The evidence that alternative Conclusion and Further Questions
activation of macrophages is important in promoting tumorigen- Although there has been striking progress in understanding the
esis includes expression of murine signature markers and mechanisms and functions of AAM, much remains to be discov-
dependence on the IL-4 and IL-13 common receptor alpha ered. We have argued in favor of a restricted definition, shown
chain. Blockade of this receptor or its genetic ablation may the value of in situ analysis in mouse and man, and pointed
therefore shift the host-tumor balance toward tumor rejection to the possibility of selective targeting of pathogenic pathways
by Th1 cell-dependent effector mechanisms, such as IL-12 through IL-13. Major human diseases, including infection,
versus IL-10 production. A recent study (Weiss et al., 2009) inflammation, fibrosis, and malignancy, demonstrate the two-
combined treatment with IL-2 and anti-CD40 to induce a switch edged nature of AAM function at different stages of disease.
from M2 to M1 macrophage functions, depending on CCR2 and The need for validated human markers is apparent. Perhaps
IFN-g. The contribution of TAM to this balance varied in different less obvious is a clearer understanding of what determines the
tumor models and could affect either primary or secondary spectrum of innate, classical, alternative, and other activation
growth. B cell-dependent mechanisms have been demonstrated phenotypes in macrophage populations and individual cells.
in selected tumors (Andreu et al., 2010), with possible interac- The trophic, antigen-presenting, and killing functions of AAMs
tions with AAM Fc receptors. need better characterization. Finally, does alternative activation
IL-13 and its receptors have been implicated in a different of macrophages extend to other innate cells, in particular den-
model of suppression of host immune effector responses to dritic, NKT, and even nonhematopoietic cells? As in other fields
tumors (Fichtner-Feigl et al., 2008a). IL-4 or IL-13, acting through of immunobiology, the alternative may indeed be the antecedent
the IL13Ra1, is able to upregulate expression of the IL-13Ra2 path in evolution.
polypeptide. TNF and NF-kB contribute in parallel to the
STAT6 pathway. IL-13 acts on GR1 intermediate CD11b high ACKNOWLEDGMENTS
monocytic cells, through variant AP1 transcription factor, to
produce TGF-b. This pathway is responsible for suppression of We thank colleagues for sharing unpublished results, Stefanie Vogel and Keri
tumor rejection by Th1 cell-type immune response. Treatment Ann Shirey for Figure 3, and Thiru Ramalingam for Figure 2. This review was
prepared while S.G. was a visiting NCI/NIAID scholar and he thanks Giorgio
with anti-TNF or siRNA to block IL-13Ra2 reduces metastasis Trinchieri and Alan Sher for their hospitality. F.O.M. was supported by the
and promotes survival. British Heart Foundation.
In summary, AAMs are important candidates for therapeutic
modulation of the tumor-host inflammatory interaction. How- REFERENCES
ever, AAM markers are urgently required to establish their contri-
bution in naturally occurring human malignancy. Agapov, E., Battaile, J.T., Tidwell, R., Hachem, R., Patterson, G.A., Pierce, R.A.,
Atkinson, J.J., and Holtzman, M.J. (2009). Macrophage chitinase 1 stratifies
chronic obstructive lung disease. Am. J. Respir. Cell Mol. Biol. 41, 379–384.
Therapeutic Interventions
Andreu, P., Johansson, M., Affara, N.I., Pucci, F., Tan, T., Junankar, S., Korets,
As knowledge of the pathophysiologic pathways of IL-4 and L., Lam, J., Tawfik, D., DeNardo, D.G., et al. (2010). FcRgamma activation
IL-13 in human disease has grown, attempts are being made regulates inflammation-associated squamous carcinogenesis. Cancer Cell
to achieve therapeutic blockade. In a well-controlled phase 2a 17, 121–134.
trial (Wenzel et al., 2007), recombinant human pitrakinra showed Androulidaki, A., Iliopoulos, D., Arranz, A., Doxaki, C., Schworer, S., Zachar-
promising results in ameliorating late-phase asthmatic response ioudaki, V., Margioris, A.N., Tsichlis, P.N., and Tsatsanis, C. (2009). The kinase
Akt1 controls macrophage response to lipopolysaccharide by regulating
to allergen challenge in patients, by aerosol and subcutaneous microRNAs. Immunity 31, 220–231.
routes. This agent, previously tested in cynomolgus monkeys,
is a potent inhibitor of binding of both IL-4 and IL-13 to IL-4R Anthony, R.M., Urban, J.F., Jr., Alem, F., Hamed, H.A., Rozo, C.T., Boucher,
J.L., Van Rooijen, N., and Gause, W.C. (2006). Memory T(H)2 cells induce alter-
receptor complexes, offering an advantage over previous natively activated macrophages to mediate protection against nematode
attempts to inhibit IL-4 alone, IL-5, or IgE. parasites. Nat. Med. 12, 955–960.
Selective blockade of IL-13 signaling could offer further Bhattacharjee, A., Xu, B., Frank, D.A., Feldman, G.M., and Cathcart, M.K.
advantages, e.g., in a subpopulation of asthma patients (Wood- (2006). Monocyte 15-lipoxygenase expression is regulated by a novel

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 601


Immunity

Review
cytosolic signaling complex with protein kinase C delta and tyrosine-phos- Fichtner-Feigl, S., Terabe, M., Kitani, A., Young, C.A., Fuss, I., Geissler, E.K.,
phorylated Stat3. J. Immunol. 177, 3771–3781. Schlitt, H.J., Berzofsky, J.A., and Strober, W. (2008a). Restoration of tumor
immunosurveillance via targeting of interleukin-13 receptor-alpha 2. Cancer
Blease, K. (2008). Therapeutics targeting IL-13 for the treatment of pulmonary Res. 68, 3467–3475.
inflammation and airway remodeling. Curr. Opin. Investig. Drugs 9,
1180–1184. Fichtner-Feigl, S., Young, C.A., Kitani, A., Geissler, E.K., Schlitt, H.J., and
Strober, W. (2008b). IL-13 signaling via IL-13R alpha2 induces major down-
Brown, G.D. (2006). Dectin-1: A signalling non-TLR pattern-recognition stream fibrogenic factors mediating fibrosis in chronic TNBS colitis. Gastroen-
receptor. Nat. Rev. Immunol. 6, 33–43. terology 135, 2003–2013.
Chawla, A. (2010). Control of macrophage activation and function by PPARs.
Gabrilovich, D.I., and Nagaraj, S. (2009). Myeloid-derived suppressor cells as
Circ. Res. 106, 1559–1569.
regulators of the immune system. Nat. Rev. Immunol. 9, 162–174.
Chiaramonte, M.G., Mentink-Kane, M., Jacobson, B.A., Cheever, A.W., Whit-
ters, M.J., Goad, M.E., Wong, A., Collins, M., Donaldson, D.D., Grusby, M.J., Gangadharan, B., Hoeve, M.A., Allen, J.E., Ebrahimi, B., Rhind, S.M., Dutia,
and Wynn, T.A. (2003). Regulation and function of the interleukin 13 receptor B.M., and Nash, A.A. (2008). Murine gammaherpesvirus-induced fibrosis is
alpha 2 during a T helper cell type 2-dominant immune response. J. Exp. associated with the development of alternatively activated macrophages.
Med. 197, 687–701. J. Leukoc. Biol. 84, 50–58.

Cihakova, D., Barin, J.G., Afanasyeva, M., Kimura, M., Fairweather, D., Berg, Geissmann, F., Manz, M.G., Jung, S., Sieweke, M.H., Merad, M., and Ley, K.
M., Talor, M.V., Baldeviano, G.C., Frisancho, S., Gabrielson, K., et al. (2008). (2010). Development of monocytes, macrophages, and dendritic cells.
Interleukin-13 protects against experimental autoimmune myocarditis by Science 327, 656–661.
regulating macrophage differentiation. Am. J. Pathol. 172, 1195–1208.
Glass, C.K., and Saijo, K. (2010). Nuclear receptor transrepression pathways
Cole, L.E., Shirey, K.A., Barry, E., Santiago, A., Rallabhandi, P., Elkins, K.L., that regulate inflammation. Nat. Rev. Immunol. 10, 365–376.
Puche, A.C., Michalek, S.M., and Vogel, S.N. (2007). Toll-like receptor 2-medi-
ated signaling requirements for Francisella tularensis live vaccine strain infec- Gocheva, V., Wang, H.W., Gadea, B.B., Shree, T., Hunter, K.E., Garfall, A.L.,
tion of murine macrophages. Infect. Immun. 75, 4127–4137. Berman, T., and Joyce, J.A. (2010). IL-4 induces cathepsin protease activity
in tumor-associated macrophages to promote cancer growth and invasion.
Cole, L.E., Santiago, A., Barry, E., Kang, T.J., Shirey, K.A., Roberts, Z.J., Genes Dev. 24, 241–255.
Elkins, K.L., Cross, A.S., and Vogel, S.N. (2008). Macrophage proinflammatory
response to Francisella tularensis live vaccine strain requires coordination of Gordon, S. (2008). Macrophages and phagocytosis. In Fundamental Immu-
multiple signaling pathways. J. Immunol. 180, 6885–6891. nology, W.E. Paul, ed. (Philadelphia, PA: Lippincott Raven), p. 547.
Daley, J.M., Brancato, S.K., Thomay, A.A., Reichner, J.S., and Albina, J.E. Gratchev, A., Schledzewski, K., Guillot, P., and Goerdt, S. (2001). Alternatively
(2010). The phenotype of murine wound macrophages. J. Leukoc. Biol. 87, activated antigen-presenting cells: Molecular repertoire, immune regulation,
59–67. and healing. Skin Pharmacol. Appl. Skin Physiol. 14, 272–279.
De’Broski, R.H., Orekev, T., Roloson, A., Ilies, M., Perkins, C., O’Brien, W.,
Cederbaum, S., Christianson, S.W., Zimmermann, N., Rothenberg, M.E., Grivennikov, S.I., Greten, F.R., and Karin, M. (2010). Immunity, inflammation,
and cancer. Cell 140, 883–899.
and Finkelman, F.D. (2010). Arginine I suppreses IL-12/IL-23p40-driven intes-
tinal inflammation during acute schistosomiasis. J. Immunol., in press.
Published online May 5, 2010. 10.4049/jimmunol.0902009. Hallam, S., Escorcio-Correia, M., Soper, R., Schultheiss, A., and Hagemann, T.
(2009). Activated macrophages in the tumour microenvironment-dancing to
Delgoffe, G.M., Kole, T.P., Zheng, Y., Zarek, P.E., Matthews, K.L., Xiao, B., the tune of TLR and NF-kappaB. J. Pathol. 219, 143–152.
Worley, P.F., Kozma, S.C., and Powell, J.D. (2009). The mTOR kinase differen-
tially regulates effector and regulatory T cell lineage commitment. Immunity 30, Heinsbroek, S.E., Kamen, L.A., Taylor, P.R., Brown, G.D., Swanson, J., and
832–844. Gordon, S. (2009). Actin and phosphoinositide recruitment to fully formed
Candida albicans phagosomes in mouse macrophages. J. Innate Immunol.
DeNardo, D.G., Barreto, J.B., Andreu, P., Vasquez, L., Tawfik, D., Kolhatkar, 1, 244–253.
N., and Coussens, L.M. (2009). CD4(+) T cells regulate pulmonary metastasis
of mammary carcinomas by enhancing protumor properties of macrophages. Heller, N.M., Qi, X., Junttila, I.S., Shirey, K.A., Vogel, S.N., Paul, W.E., and Kee-
Cancer Cell 16, 91–102. gan, A.D. (2008). Type I IL-4Rs selectively activate IRS-2 to induce target gene
expression in macrophages. Sci. Signal. 1, ra17.
Dickensheets, H., Vazquez, N., Sheikh, F., Gingras, S., Murray, P.J., Ryan, J.J.,
and Donnelly, R.P. (2007). Suppressor of cytokine signaling-1 is an IL-4-induc- Helming, L., and Gordon, S. (2009). Molecular mediators of macrophage
ible gene in macrophages and feedback inhibits IL-4 signaling. Genes Immun. fusion. Trends Cell Biol. 19, 514–522.
8, 21–27.
Helming, L., Tomasello, E., Kyriakides, T.R., Martinez, F.O., Takai, T., Gordon,
Doyle, A.G., Herbein, G., Montaner, L.J., Minty, A.J., Caput, D., Ferrara, P., and S., and Vivier, E. (2008). Essential role of DAP12 signaling in macrophage
Gordon, S. (1994). Interleukin-13 alters the activation state of murine programming into a fusion-competent state. Sci. Signal. 1, ra11.
macrophages in vitro: Comparison with interleukin-4 and interferon-gamma.
Eur. J. Immunol. 24, 1441–1445. Herbert, D.R., Holscher, C., Mohrs, M., Arendse, B., Schwegmann, A., Rad-
wanska, M., Leeto, M., Kirsch, R., Hall, P., Mossmann, H., et al. (2004). Alter-
El Kasmi, K.C., Qualls, J.E., Pesce, J.T., Smith, A.M., Thompson, R.W., Henao-
native macrophage activation is essential for survival during schistosomiasis
Tamayo, M., Basaraba, R.J., Konig, T., Schleicher, U., Koo, M.S., et al. (2008).
and downmodulates T helper 1 responses and immunopathology. Immunity
Toll-like receptor-induced arginase 1 in macrophages thwarts effective immu-
20, 623–635.
nity against intracellular pathogens. Nat. Immunol. 9, 1399–1406.

Fallon, P.G., Ballantyne, S.J., Mangan, N.E., Barlow, J.L., Dasvarma, A., Hesse, M., Modolell, M., La Flamme, A.C., Schito, M., Fuentes, J.M., Cheever,
Hewett, D.R., McIlgorm, A., Jolin, H.E., and McKenzie, A.N. (2006). Identifica- A.W., Pearce, E.J., and Wynn, T.A. (2001). Differential regulation of nitric oxide
tion of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: Granulomatous
and IL-13 at the onset of helminth expulsion. J. Exp. Med. 203, 1105–1116. pathology is shaped by the pattern of L-arginine metabolism. J. Immunol. 167,
6533–6544.
Fernandez-Boyanapalli, R.F., Frasch, S.C., McPhillips, K., Vandivier, R.W.,
Harry, B.L., Riches, D.W., Henson, P.M., and Bratton, D.L. (2009). Impaired Holscher, C., Arendse, B., Schwegmann, A., Myburgh, E., and Brombacher, F.
apoptotic cell clearance in CGD due to altered macrophage programming is (2006). Impairment of alternative macrophage activation delays cutaneous
reversed by phosphatidylserine-dependent production of IL-4. Blood 113, leishmaniasis in nonhealing BALB/c mice. J. Immunol. 176, 1115–1121.
2047–2055.
Holtzman, M.J., Byers, D.E., Benoit, L.A., Battaile, J.T., You, Y., Agapov, E.,
Fichtner-Feigl, S., Strober, W., Kawakami, K., Puri, R.K., and Kitani, A. (2006). Park, C., Grayson, M.H., Kim, E.Y., and Patel, A.C. (2009). Immune pathways
IL-13 signaling through the IL-13alpha2 receptor is involved in induction of for translating viral infection into chronic airway disease. Adv. Immunol. 102,
TGF-beta1 production and fibrosis. Nat. Med. 12, 99–106. 245–276.

602 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.


Immunity

Review
Ishii, M., Wen, H., Corsa, C.A., Liu, T., Coelho, A.L., Allen, R.M., Carson, W.F., Martinez, F.O., Helming, L., and Gordon, S. (2009). Alternative activation of
IV, Cavassani, K.A., Li, X., Lukacs, N.W., et al. (2009). Epigenetic regulation of macrophages: An immunologic functional perspective. Annu. Rev. Immunol.
the alternatively activated macrophage phenotype. Blood 114, 3244–3254. 27, 451–483.

Jenkins, S.J., and Allen, J.E. (2010). Similarity and diversity in macrophage Mitchell, J., Dimov, V., and Townley, R.G. (2010). IL-13 and the IL-13 receptor
activation by nematodes, trematodes, and cestodes. J. Biomed. Biotechnol. as therapeutic targets for asthma and allergic disease. Curr. Opin. Investig.
2010, 262609. Drugs 11, 527–534.

Joyce, J.A., and Pollard, J.W. (2009). Microenvironmental regulation of metas- Mohrs, K., Wakil, A.E., Killeen, N., Locksley, R.M., and Mohrs, M. (2005).
tasis. Nat. Rev. Cancer 9, 239–252. A two-step process for cytokine production revealed by IL-4 dual-reporter
mice. Immunity 23, 419–429.
Junttila, I.S., Mizukami, K., Dickensheets, H., Meier-Schellersheim, M.,
Yamane, H., Donnelly, R.P., and Paul, W.E. (2008). Tuning sensitivity to IL-4 Mokoena, T., and Gordon, S. (1985). Human macrophage activation. Modula-
and IL-13: Differential expression of IL-4Ralpha, IL-13Ralpha1, and gammac tion of mannosyl, fucosyl receptor activity in vitro by lymphokines, gamma and
regulates relative cytokine sensitivity. J. Exp. Med. 205, 2595–2608. alpha interferons, and dexamethasone. J. Clin. Invest. 75, 624–631.
Kahnert, A., Seiler, P., Stein, M., Bandermann, S., Hahnke, K., Mollenkopf, H., Montaner, L.J., da Silva, R.P., Sun, J., Sutterwala, S., Hollinshead, M., Vaux,
and Kaufmann, S.H. (2006). Alternative activation deprives macrophages of D., and Gordon, S. (1999). Type 1 and type 2 cytokine regulation of macro-
a coordinated defense program to Mycobacterium tuberculosis. Eur. J. Immu- phage endocytosis: differential activation by IL-4/IL-13 as opposed to IFN-
nol. 36, 631–647. gamma or IL-10. J. Immunol. 162, 4606–4613.
Kawashima, R., Kawamura, Y.I., Kato, R., Mizutani, N., Toyama-Sorimachi, N., Moreno, J.L., Kaczmarek, M., Keegan, A.D., and Tondravi, M. (2003). IL-4
and Dohi, T. (2006). IL-13 receptor alpha2 promotes epithelial cell regeneration suppresses osteoclast development and mature osteoclast function by
from radiation-induced small intestinal injury in mice. Gastroenterology 131, a STAT6-dependent mechanism: Irreversible inhibition of the differentiation
130–141. program activated by RANKL. Blood 102, 1078–1086.
Kim, E.Y., Battaile, J.T., Patel, A.C., You, Y., Agapov, E., Grayson, M.H., Moreno, J.L., Mikhailenko, I., Tondravi, M.M., and Keegan, A.D. (2007). IL-4
Benoit, L.A., Byers, D.E., Alevy, Y., Tucker, J., et al. (2008). Persistent activa- promotes the formation of multinucleated giant cells from macrophage precur-
tion of an innate immune response translates respiratory viral infection into sors by a STAT6-dependent, homotypic mechanism: Contribution of E-cad-
chronic lung disease. Nat. Med. 14, 633–640. herin. J. Leukoc. Biol. 82, 1542–1553.
Kuroda, E., Ho, V., Ruschmann, J., Antignano, F., Hamilton, M., Rauh, M.J., Moro, K., Yamada, T., Tanabe, M., Takeuchi, T., Ikawa, T., Kawamoto, H.,
Antov, A., Flavell, R.A., Sly, L.M., and Krystal, G. (2009). SHIP represses the Furusawa, J., Ohtani, M., Fujii, H., and Koyasu, S. (2009). Innate production
generation of IL-3-induced M2 macrophages by inhibiting IL-4 production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid
from basophils. J. Immunol. 183, 3652–3660. cells. Nature 463, 540–544.
Kurowska-Stolarska, M., Stolarski, B., Kewin, P., Murphy, G., Corrigan, C.J., Mosser, D.M., and Edwards, J.P. (2008). Exploring the full spectrum of macro-
Ying, S., Pitman, N., Mirchandani, A., Rana, B., van Rooijen, N., et al. (2009). phage activation. Nat. Rev. Immunol. 8, 958–969.
IL-33 amplifies the polarization of alternatively activated macrophages that
contribute to airway inflammation. J. Immunol. 183, 6469–6477. Munitz, A., Waddell, A., Seidu, L., Cole, E.T., Ahrens, R., Hogan, S.P., and
Rothenberg, M.E. (2008). Resistin-like molecule alpha enhances myeloid cell
Kzhyshkowska, J., and Krusell, L. (2009). Cross-talk between endocytic clear-
activation and promotes colitis. J. Allergy Clin. Immunol. 122, 1200–1207.
ance and secretion in macrophages. Immunobiology 214, 576–593.
Nair, M.G., Du, Y., Perrigoue, J.G., Zaph, C., Taylor, J.J., Goldschmidt, M.,
LaPorte, S.L., Juo, Z.S., Vaclavikova, J., Colf, L.A., Qi, X., Heller, N.M., Kee-
Swain, G.P., Yancopoulos, G.D., Valenzuela, D.M., Murphy, A., et al. (2009).
gan, A.D., and Garcia, K.C. (2008). Molecular and structural basis of cytokine
Alternatively activated macrophage-derived RELM-{alpha} is a negative regu-
receptor pleiotropy in the interleukin-4/13 system. Cell 132, 259–272.
lator of type 2 inflammation in the lung. J. Exp. Med. 206, 937–952.
Linehan, S.A., Coulson, P.S., Wilson, R.A., Mountford, A.P., Brombacher, F.,
Neill, D.R., Wong, S.H., Bellosi, A., Flynn, R.J., Daly, M., Langford, T.K., Bucks,
Martinez-Pomares, L., and Gordon, S. (2003). IL-4 receptor signaling is
C., Kane, C.M., Fallon, P.G., Pannell, R., et al. (2010). Nuocytes represent
required for mannose receptor expression by macrophages recruited to gran-
a new innate effector leukocyte that mediates type-2 immunity. Nature 464,
ulomata but not resident cells in mice infected with Schistosoma mansoni. Lab.
1367–1370.
Invest. 83, 1223–1231.

Locksley, R.M. (2010). Asthma and allergic inflammation. Cell 140, 777–783. O’Shea, J.J., and Paul, W.E. (2010). Mechanisms underlying lineage commit-
ment and plasticity of helper CD4+ T cells. Science 327, 1098–1102.
Loke, P., MacDonald, A.S., and Allen, J.E. (2000). Antigen-presenting cells
recruited by Brugia malayi induce Th2 differentiation of naive CD4(+) T cells. O’Toole, M., Legault, H., Ramsey, R., Wynn, T.A., and Kasaian, M.T. (2008).
Eur. J. Immunol. 30, 1127–1135. A novel and sensitive ELISA reveals that the soluble form of IL-13R-alpha2 is
not expressed in plasma of healthy or asthmatic subjects. Clin. Exp. Allergy
Loke, P., Nair, M.G., Parkinson, J., Guiliano, D., Blaxter, M., and Allen, J.E. 38, 594–601.
(2002). IL-4 dependent alternatively-activated macrophages have a distinctive
in vivo gene expression phenotype. BMC Immunol. 3, 7. Pelegrin, P., and Surprenant, A. (2009). Dynamics of macrophage polarization
reveal new mechanism to inhibit IL-1beta release through pyrophosphates.
Lupardus, P.J., Birnbaum, M.E., and Garcia, K.C. (2010). Molecular basis for EMBO J. 28, 2114–2127.
shared cytokine recognition revealed in the structure of an unusually high
affinity complex between IL-13 and IL-13Ralpha2. Structure 18, 332–342. Pesce, J., Kaviratne, M., Ramalingam, T.R., Thompson, R.W., Urban, J.F., Jr.,
Cheever, A.W., Young, D.A., Collins, M., Grusby, M.J., and Wynn, T.A. (2006).
Mandruzzato, S., Solito, S., Falisi, E., Francescato, S., Chiarion-Sileni, V., The IL-21 receptor augments Th2 effector function and alternative macro-
Mocellin, S., Zanon, A., Rossi, C.R., Nitti, D., Bronte, V., and Zanovello, P. phage activation. J. Clin. Invest. 116, 2044–2055.
(2009). IL4Ralpha+ myeloid-derived suppressor cell expansion in cancer
patients. J. Immunol. 182, 6562–6568. Pesce, J.T., Ramalingam, T.R., Mentink-Kane, M.M., Wilson, M.S., El Kasmi,
K.C., Smith, A.M., Thompson, R.W., Cheever, A.W., Murray, P.J., and Wynn,
Mantovani, A., Sica, A., and Locati, M. (2005). Macrophage polarization comes T.A. (2009a). Arginase-1-expressing macrophages suppress Th2 cytokine-
of age. Immunity 23, 344–346. driven inflammation and fibrosis. PLoS Pathog. 5, e1000371.

Mantovani, A., Allavena, P., Sica, A., and Balkwill, F. (2008). Cancer-related Pesce, J.T., Ramalingam, T.R., Wilson, M.S., Mentink-Kane, M.M., Thompson,
inflammation. Nature 454, 436–444. R.W., Cheever, A.W., Urban, J.F., Jr., and Wynn, T.A. (2009b). Retnla (relmal-
pha/fizz1) suppresses helminth-induced Th2-type immunity. PLoS Pathog. 5,
Martinez, F.O., Gordon, S., Locati, M., and Mantovani, A. (2006). Transcrip- e1000393.
tional profiling of the human monocyte-to-macrophage differentiation and
polarization: New molecules and patterns of gene expression. J. Immunol. Porta, C., Rimoldi, M., Raes, G., Brys, L., Ghezzi, P., Di Liberto, D., Dieli, F.,
177, 7303–7311. Ghisletti, S., Natoli, G., De Baetselier, P., et al. (2009). Tolerance and M2

Immunity 32, May 28, 2010 ª2010 Elsevier Inc. 603


Immunity

Review
(alternative) macrophage polarization are related processes orchestrated by Steinman, R.M., and Idoyaga, J. (2010). Features of the dendritic cell lineage.
p50 nuclear factor kappaB. Proc. Natl. Acad. Sci. USA 106, 14978–14983. Immunol. Rev. 234, 5–17.

Puig-Kroger, A., Sierra-Filardi, E., Dominguez-Soto, A., Samaniego, R., Cor- Stout, R.D. (2010). Editorial: Macrophage functional phenotypes: No alterna-
cuera, M.T., Gomez-Aguado, F., Ratnam, M., Sanchez-Mateos, P., and Corbi, tives in dermal wound healing? J. Leukoc. Biol. 87, 19–21.
A.L. (2009). Folate receptor beta is expressed by tumor-associated macro-
phages and constitutes a marker for M2 anti-inflammatory/regulatory macro- Stout, R.D., Jiang, C., Matta, B., Tietzel, I., Watkins, S.K., and Suttles, J. (2005).
phages. Cancer Res. 69, 9395–9403. Macrophages sequentially change their functional phenotype in response to
changes in microenvironmental influences. J. Immunol. 175, 342–349.
Ramalingam, T.R., Pesce, J.T., Sheikh, F., Cheever, A.W., Mentink-Kane,
M.M., Wilson, M.S., Stevens, S., Valenzuela, D.M., Murphy, A.J., Yancopou- Syed, F., Huang, C.C., Li, K., Liu, V., Shang, T., Amegadzie, B.Y., Griswold,
los, G.D., et al. (2008). Unique functions of the type II interleukin 4 receptor D.E., Song, X.Y., and Li, L. (2007). Identification of interleukin-13 related
identified in mice lacking the interleukin 13 receptor alpha1 chain. Nat. Immu- biomarkers using peripheral blood mononuclear cells. Biomarkers 12,
nol. 9, 25–33. 414–423.

Rathinam, V.A., Jiang, Z., Waggoner, S.N., Sharma, S., Cole, L.E., Waggoner, Taganov, K.D., Boldin, M.P., Chang, K.J., and Baltimore, D. (2006). NF-kap-
L., Vanaja, S.K., Monks, B.G., Ganesan, S., Latz, E., et al. (2010). The AIM2 paB-dependent induction of microRNA miR-146, an inhibitor targeted to
inflammasome is essential for host defense against cytosolic bacteria and signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA
DNA viruses. Nat. Immunol. 11, 395–402. 103, 12481–12486.
Rauh, M.J., Ho, V., Pereira, C., Sham, A., Sly, L.M., Lam, V., Huxham, L., Min- Thompson, R.W., Pesce, J.T., Ramalingam, T., Wilson, M.S., White, S.,
chinton, A.I., Mui, A., and Krystal, G. (2005). SHIP represses the generation of Cheever, A.W., Ricklefs, S.M., Porcella, S.F., Li, L., Ellies, L.G., and Wynn,
alternatively activated macrophages. Immunity 23, 361–374. T.A. (2008). Cationic amino acid transporter-2 regulates immunity by modu-
lating arginase activity. PLoS Pathog. 4, e1000023.
Reece, J.J., Siracusa, M.C., and Scott, A.L. (2006). Innate immune responses
to lung-stage helminth infection induce alternatively activated alveolar macro- Tili, E., Michaille, J.J., Cimino, A., Costinean, S., Dumitru, C.D., Adair, B., Fab-
phages. Infect. Immun. 74, 4970–4981. bri, M., Alder, H., Liu, C.G., Calin, G.A., and Croce, C.M. (2007). Modulation of
miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimu-
Reese, T.A., Liang, H.E., Tager, A.M., Luster, A.D., Van Rooijen, N., Voeh- lation and their possible roles in regulating the response to endotoxin shock.
ringer, D., and Locksley, R.M. (2007). Chitin induces accumulation in tissue J. Immunol. 179, 5082–5089.
of innate immune cells associated with allergy. Nature 447, 92–96.
Trompette, A., Divanovic, S., Visintin, A., Blanchard, C., Hegde, R.S., Madan,
Richardson, J.Y., Ottolini, M.G., Pletneva, L., Boukhvalova, M., Zhang, S., R., Thorne, P.S., Wills-Karp, M., Gioannini, T.L., Weiss, J.P., and Karp, C.L.
Vogel, S.N., Prince, G.A., and Blanco, J.C. (2005). Respiratory syncytial virus
(2009). Allergenicity resulting from functional mimicry of a Toll-like receptor
(RSV) infection induces cyclooxygenase 2: a potential target for RSV therapy. complex protein. Nature 457, 585–588.
J. Immunol. 174, 4356–4364.
van de Veerdonk, F.L., Marijnissen, R.J., Kullberg, B.J., Koenen, H.J., Cheng,
Rodriguez-Sosa, M., Satoskar, A.R., Calderon, R., Gomez-Garcia, L., Saave-
S.C., Joosten, I., van den Berg, W.B., Williams, D.L., van der Meer, J.W., Joos-
dra, R., Bojalil, R., and Terrazas, L.I. (2002). Chronic helminth infection induces
ten, L.A., and Netea, M.G. (2009). The macrophage mannose receptor induces
alternatively activated macrophages expressing high levels of CCR5 with low
IL-17 in response to Candida albicans. Cell Host Microbe 5, 329–340.
interleukin-12 production and Th2-biasing ability. Infect. Immun. 70, 3656–
3664.
Van den Bossche, J., Bogaert, P., van Hengel, J., Guerin, C.J., Berx, G.,
Movahedi, K., Van den Bergh, R., Pereira-Fernandes, A., Geuns, J.M., Pircher,
Roy, B., Bhattacharjee, A., Xu, B., Ford, D., Maizel, A.L., and Cathcart, M.K.
H., et al. (2009). Alternatively activated macrophages engage in homotypic and
(2002). IL-13 signal transduction in human monocytes: Phosphorylation of
receptor components, association with Jaks, and phosphorylation/activation heterotypic interactions through IL-4 and polyamine-induced E-cadherin/
catenin complexes. Blood 114, 4664–4674.
of Stats. J. Leukoc. Biol. 72, 580–589.

Ruschmann, J., Ho, V., Antignano, F., Kuroda, E., Lam, V., Ibaraki, M., Snyder, Varin, A., and Gordon, S. (2009). Alternative activation of macrophages:
K., Kim, C., Flavell, R.A., Kawakami, T., et al. (2010). Tyrosine phosphorylation Immune function and cellular biology. Immunobiology 214, 630–641.
of SHIP promotes its proteosomal degradation. Exp. Hematol. 38, 392–402.
Varin, A., Mukhopadhyay, S., Herbein, G., and Gordon, S. (2009). Alternative
Saenz, S.A., Siracusa, M.C., Perrigoue, J.G., Spencer, S.P., Urban, J.F., Jr., activation of macrophages by IL-4 impairs phagocytosis of pathogens but
Tocker, J.E., Budelsky, A.L., Kleinschek, M.A., Kastelein, R.A., Kambayashi, potentiates microbial-induced signalling and cytokine secretion. Blood 115,
T., et al. (2010). IL25 elicits a multipotent progenitor cell population that 353–362.
promotes T(H)2 cytokine responses. Nature 464, 1362–1366.
Voehringer, D., Reese, T.A., Huang, X., Shinkai, K., and Locksley, R.M. (2006).
Scotton, C.J., Martinez, F.O., Smelt, M.J., Sironi, M., Locati, M., Mantovani, A., Type 2 immunity is controlled by IL-4/IL-13 expression in hematopoietic non-
and Sozzani, S. (2005). Transcriptional profiling reveals complex regulation of eosinophil cells of the innate immune system. J. Exp. Med. 203, 1435–1446.
the monocyte IL-1 beta system by IL-13. J. Immunol. 174, 834–845.
Weiss, J.M., Back, T.C., Scarzello, A.J., Subleski, J.J., Hall, V.L., Stauffer, J.K.,
Shirey, K.A., Cole, L.E., Keegan, A.D., and Vogel, S.N. (2008). Francisella tular- Chen, X., Micic, D., Alderson, K., Murphy, W.J., and Wiltrout, R.H. (2009).
ensis live vaccine strain induces macrophage alternative activation as Successful immunotherapy with IL-2/anti-CD40 induces the chemokine-medi-
a survival mechanism. J. Immunol. 181, 4159–4167. ated mitigation of an immunosuppressive tumor microenvironment. Proc. Natl.
Acad. Sci. USA 106, 19455–19460.
Shirey, K.A., Pletneva, L.M., Puche, A.C., Keegan, A.D., Prince, G.A., Blanco,
J.C., and Vogel, S.N. (2010). Control of RSV-induced lung injury by alterna- Wenzel, S., Wilbraham, D., Fuller, R., Getz, E.B., and Longphre, M. (2007).
tively activated macrophages is IL-4Ra-. TLR4-, and IFN-b-dependent. Effect of an interleukin-4 variant on late phase asthmatic response to allergen
Mucosal Immunol. 3, 291–300. challenge in asthmatic patients: results of two phase 2a studies. Lancet 370,
1422–1431.
Sinha, P., Clements, V.K., Bunt, S.K., Albelda, S.M., and Ostrand-Rosenberg,
S. (2007). Cross-talk between myeloid-derived suppressor cells and macro- Wilson, M.S., and Wynn, T.A. (2009). Pulmonary fibrosis: Pathogenesis,
phages subverts tumor immunity toward a type 2 response. J. Immunol. etiology and regulation. Mucosal Immunol. 2, 103–121.
179, 977–983.
Woodruff, P.G., Modrek, B., Choy, D.F., Jia, G., Abbas, A.R., Ellwanger, A.,
Stein, M., Keshav, S., Harris, N., and Gordon, S. (1992). Interleukin 4 potently Koth, L.L., Arron, J.R., and Fahy, J.V. (2009). T-helper type 2-driven inflamma-
enhances murine macrophage mannose receptor activity: A marker of alterna- tion defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med.
tive immunologic macrophage activation. J. Exp. Med. 176, 287–292. 180, 388–395.

604 Immunity 32, May 28, 2010 ª2010 Elsevier Inc.

You might also like