Macrophage CD40 signaling: A pivotal regulator of disease protection and pathogenesis

Macrophage CD40 signaling: A pivotal regulator of disease protection and pathogenesis

Seminars in Immunology 21 (2009) 257–264 Contents lists available at ScienceDirect Seminars in Immunology journal homepage: www.elsevier.com/locate/...

469KB Sizes 0 Downloads 25 Views

Seminars in Immunology 21 (2009) 257–264

Contents lists available at ScienceDirect

Seminars in Immunology journal homepage: www.elsevier.com/locate/ysmim

Review

Macrophage CD40 signaling: A pivotal regulator of disease protection and pathogenesis Jill Suttles ∗ , Robert D. Stout Department of Microbiology and Immunology, University of Louisville School of Medicine, 319 Abraham Flexner Way, Louisville, KY 40292, USA

a r t i c l e Keywords: CD40 CD154 Macrophage Monocyte Inflammation Cell signaling

i n f o

a b s t r a c t Macrophages reside in all tissues as resident populations and as immigrants recruited in response to tissue injury, inflammation or pathogen invasion. Under normal conditions, macrophages contribute to tissue homeostasis and provide innate immune surveillance. Both macrophages and their progenitors, bone marrow-derived monocytes, constitutively express the tumor necrosis factor receptor superfamily member, CD40, and are capable of a robust response to CD40 ligation resulting in the induction or enhancement of expression of genes with a predominantly pro-inflammatory function. CD40 signaling in macrophages in the context of host responses to pathogens plays a crucial role in host defense. However, macrophage responses to CD40 ligation in the context of autoimmune and cardiovascular disease contribute to disease pathogenesis. In this review, we discuss the role of CD40 in both protective and destructive processes, including the signaling pathways engaged and the factors capable of modulating CD40 signal transduction. © 2009 Elsevier Ltd. All rights reserved.

1. Introduction Macrophages reside in all tissues of the body where they provide innate immune surveillance and contribute to tissue homeostasis. Macrophages originate from bone marrow-derived mature monocytes that migrate to tissues in response to chemokine signaling, either for replenishment of the tissue resident population, or in response to tissue insult [1–4]. Upon entry into tissues monocytes/macrophages acquire unique functional profiles in response to the tissue environment. Even within a tissue, macrophages display heterogeneity dependent on the microanatomical location in that tissue [5–7]. This feature of macrophages is representative of their remarkable plasticity and range of activities that can be initiated and maintained by environmental signals [8]. Macrophages are capable of a highly diverse repertoire of functional activities, including phagocytosis, generation of microbicidal oxygen and nitrogen radicals, production of metalloproteinases, and production of both inflammatory and anti-inflammatory cytokines as well as factors promoting tissue growth and angiogenesis [8]. Both monocytes in circulation and macrophages in tissues are exposed to a wide variety of potential stimuli, including cytokines, chemokines, hormones, lipid mediators, and immunoglobulins, all of which have the capacity to modulate macrophage behavior. In addition to these various soluble factors, it was recognized over two decades ago that

∗ Corresponding author. Tel.: +1 502 852 5144; fax: +1 502 852 7531. E-mail address: [email protected] (J. Suttles). 1044-5323/$ – see front matter © 2009 Elsevier Ltd. All rights reserved. doi:10.1016/j.smim.2009.05.011

monocytes and macrophages can be activated solely via cell contact with activated T cells [9,10]. A number of receptor–ligand pairs were reported to contribute to this activity, including membrane TNF␣ [11]. However, it was subsequently found that T cells that do not express membrane TNF␣ are equally effective in contact-dependent stimulation of macrophages [12]. Although previously considered as primarily a B cell surface receptor, CD40 was then viewed as a likely candidate receptor on monocytes/macrophages for the signal received by activated T cells. The expression of CD40 on human monocytes and murine macrophages and its functional ligation via CD154 expressed on activated CD4+ T cells was revealed [13–15], and followed by the conclusive finding that CD4+ T cells isolated from mice deficient for expression of CD154 were greatly impaired in ability to activate macrophage TNF␣ expression and generation of nitric oxide [16]. The discovery of the CD40:CD154 interaction as a mechanism of macrophage activation greatly expanded the view of the contribution of CD40 signaling to infectious and autoimmune inflammatory diseases. The subsequent identification of CD154 on platelets, mast cells and basophils [17,18], and the discovery of a soluble form of CD154 [19], further extend the possible scenarios in which ligation of CD40 expressed by monocytes and macrophages may play a role in disease progression or management. 2. Functional outcomes of macrophage CD40 stimulation The first studies of monocyte and macrophage CD40 responses revealed a primarily pro-inflammatory outcome of CD40 signal transduction. Monocytes and macrophages for the most part

258

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

display similar functional outcomes to CD40 ligation, although the level of responsiveness may differ. Differences in responses based on the tissue source of cells are also apparent and likely reflect the priming effect of the tissue environment. Even within tissues or organs, macrophage responses may vary based on location within that tissue or organ. For example, T cell contact-dependent activation of lung tissue macrophages, but not alveolar macrophages, induces expression of matrix metalloproteinases [20]. Ligation of CD40 on monocytes and macrophages results in the efficient induction of synthesis of proinflammatory cytokines including IL-1␣ and IL-1␤ [14], TNF␣ [16,21], IL-6 [13,21,22], IL-12 [23,24], as well as chemokines, including IL-8 [21], CCL2, CCL3, CCL4 and CCL5 [25]. Ligation of CD40 on monocytes/macrophages also upregulates expression of MHC class II and co-stimulatory molecules CD80 and CD86, as well as CD40, itself [21]. Other effector functions of monocytes/macrophages induced via CD40 stimulation include the expression of matrix metalloproteinases (as mentioned above) [20,26–28] and production of nitric oxide (NO) [15,16]. In addition, CD40 stimulation of monocytes has also been shown to rescue them from apoptosis induced via serum deprivation [29]. Thus, upon exposure to CD154, monocytes and macrophages can be directed towards a functional phenotype that promotes antigen presentation and proinflammatory activity. However, many of the studies examining the functional outcomes of CD40 stimulation of macrophages have been performed in vitro, therefore not accounting for the contribution of the tissue environment. Studies using soluble forms of CD154, T cell membranes or fixed T cells do not take into consideration the cytokine milieu. For example, both Th1 and Th2 clones have been shown to activate macrophages [10,12,30], yet the outcome is quite different due to the presence or absence of IFN␥ or IL-4/IL-10. IFN␥ provides a second stimulus for the production of NO, thus, typically, in its absence CD40 ligation will not result in NO production [10]. The presence of IL-4 and IL-10, however, will downregulate the proinflammatory influence of CD40 ligation [31]. Stimulus of CD40 on macrophages in tissues with a high TGF␤ content will also have unique consequences. For example, many tumor types express CD154 as well as secrete TGF␤. Tumor-associated macrophages (TAMs) are maintained in a tumor-supportive state and there is evidence that CD40 signaling may contribute to this unique state of activation of TAMs (Stout and Suttles, unpublished observations). In contrast, treatment of tumor-bearing mice with an agonistic anti-CD40 mAb activates macrophage cytostatic activity and suppresses tumor cell growth [32,33]. Thus, the nature of the delivery of CD40 stimulus, and the tissue environment, result in variable consequences that can be either protective from disease or contribute to disease progression. As we discuss later, the CD40:CD154 interaction involving T cells is reciprocal and contributes to the functional plasticity of both cell types, thus directing, or re-directing, immune responses. 3. Monocyte/macrophage CD40 signal transduction Despite the heterogeneity of monocytes and macrophages, analysis of expression of CD40 on circulating monocytes (human) or in vitro generated murine macrophages by flow cytometry typically reveals homogeneous expression of CD40 that can be upregulated by cytokine stimulus [13,14]. Despite the fairly low levels of CD40 expression exhibited by resting monocytes/macrophages, these cells are readily activated by CD40 ligation. However, the requirements for activation of monocytes/macrophages through CD40 appear to be more stringent than reported for B cells, in that typically agonistic anti-CD40 IgG antibodies, alone, are not efficient activators of signal transduction in monocytes/macrophages, nor are monomeric forms of soluble CD154. More extensive crosslinking is required, such as can be achieved by use of IgM anti-CD154 antibody, cross-linking of anti-CD40 antibodies with

anti-Ig antibody, use of activated T cell membrane preparations, CD154 transfectants, or multimeric soluble forms of the ligand [14,22,34]. Possibly due to the limited availability of reagents that effectively activate macrophage-expressed CD40, studies of CD40 signaling in monocytes/macrophages are not extensive. Two independent early studies of monocyte/macrophage CD40 signaling revealed a tyrosine kinase-dependent pathway leading to activation of the mitogen-activated protein kinases (MAPKs) ERK1/2 which proved critical to the induction of proinflammatory cytokine and chemokine expression (i.e., IL-1, TNF␣, IL-6, and IL-8) [31,35]. Low-level CD40-mediated activation of the MAPK Jun-N-terminal kinase (JNK) was found in one of these studies [35], whereas both reports found no enhancement of MAPK p38 phosphorylation. Later studies reported p38 activation in monocytes in response to soluble CD154 stimulation and demonstrated a role of p38 in the induction of iNOS, COX-2 and IL-12 expression [36,37]. The discrepancies in the signaling outcomes observed in studies of CD40 ligation of monocytes and macrophages is likely due to the variation in the stimuli used. For example, a report by Mathur et al. [37] suggested that the strength of CD40 signaling dictated signaling outcome, with weak signals inducing ERK1/2 phosphorylation and IL-10 production, with stronger signals resulting in p38 activation and induction of IL-12. The tyrosine kinase activity rapidly induced by CD40 ligation was later attributed to Src kinase, which serves as an upstream initiator of pathways leading to both ERK1/2 and NF-␬B activation [22]. CD40 ligation on macrophages has also been shown to activate a phosphoinositide 3-kinase/Akt pathway that contributes to induction of IL-10 [38], which may occur via similar mechanisms as proposed for signaling via Toll-like receptor 4 (TLR4) [39]. There is also evidence for a role of the serine/threonine kinase Tpl2 (tumor progression locus 2), also known as Cot/MAP3K8, as an upstream initiator of the activation of ERK1/2, but not of JNK, p38 in response to macrophage CD40 ligation [40]. CD40 has no intrinsic catalytic activity and relies on the recruitment of adaptor proteins of the TNFR-associated factor (TRAF) family for signal transduction. Ligation of CD40 causes its oligomerization, which leads to recruitment of specific TRAF proteins, of which six have been identified. TRAF family members share a common stretch of amino acids at the carboxy terminus designated as the TRAF domain, which is further divided into two sub-regions. The carboxy terminus of the TRAF domain, TRAF-C, mediates homoand hetero-dimerization with other TRAF proteins and also to the receptor that recruits them. With the exception of TRAF1, all TRAFs contain a ring finger and 5–6 zinc finger-like motifs N-terminal to the TRAF domain, which presumably play a role in NF-␬B and c-Jun N-terminal kinase (JNK) activation [41]. The cytoplasmic domain of human CD40 has a proximal TRAF6 binding site and a more distal TRAF2/3/5 binding site. Although TRAF:CD40 interactions and the roles of TRAFs in B cell CD40 signaling has been evaluated in substantial detail [42,43], the specific functions of TRAFs in CD40 signal transduction in monocytes/macrophages have not been fully explored. Human monocytes express all TRAF family members constitutively [35]. Studies to date suggest that TRAF6 is likely the major player in mediating a wide range of proinflammatory functions initiated by CD40 ligation on monocytes and macrophages, as well as myeloid DC [22,44]. The role of TRAF family members in mediating CD40 signaling in macrophages was evaluated by transfection of a CD40-deficient macrophage cell line with wild-type human CD40, or with mutant forms of CD40 that contained disrupted TRAF binding sites [22]. It was shown that ligation of either wild-type CD40, or a CD40 mutant unable to bind TRAF2/3/5, resulted in effective stimulation of inflammatory cytokine production by macrophages, whereas ligation of a CD40 mutant lacking a functional TRAF6 binding site did not. An intact TRAF6 bindings site was found to be required for CD40-mediated activation of ERK1/2, as well as

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

I␬B kinase (IKK) and NF-␬B. Introduction of a dominant negative TRAF6 into a wild-type (CD40+ ) macrophage cell line resulted in abrogation of CD40-mediated induction of inflammatory cytokine synthesis. The crucial role of TRAF6 in these pathways was corroborated by the use of a cell permeable peptide corresponding to the TRAF6 binding motif of CD40. This peptide, capable of blocking the TRAF6:CD40 interaction, inhibited CD40 activation of ERK1/2, IKK and inflammatory cytokine production [22]. Blockade of the TRAF6:CD40 interaction did not inhibit the initial phosphorylation of Src kinase induced through CD40, suggesting that the requirement for TRAF6 is downstream of Src in the signaling pathways leading to ERK1/2 and NF-␬B activation. As mentioned above, the serine/threonine kinase Tpl2 has also been implicated in CD40-induction of ERK1/2 phosphorylation in macrophages. Macrophages deficient for Tpl2 expression showed impaired ERK1/2 activation [40]. In this study, recruitment of Tpl2 in a complex with CD40 and TRAF6 was demonstrated in fibroblasts, which may be the case as well in macrophages, although this has not yet been directly shown. TRAF6 has also been shown to be critical to CD40-induced autophagy in a model of Toxoplasma gondii infection of macrophages [45,46]. This is due, in part, to the TRAF6-dependency of TNF␣ production induced in response to CD40 ligation [22]. CD40 and TNF␣ work synergistically to promote vacuole–lysosome fusion through autophagy, resulting killing of this intracellular pathogen [46].

259

Surface plasmon resonance studies revealed that TRAF6 has a low affinity for CD40 and requires extensive multimerization of CD40 to mediate signaling events [47]. Thus, the identification of TRAF6 as an important signal transducer for CD40 in monocytes/macrophages may explain the requirement for extensive cross-linking to initiate CD40 signaling in these cell types. Although TRAF6 clearly plays a key role in CD40 signaling in monocytes and macrophages, the disruption of the TRAF2,3,5 reduces selected signaling events, as well, indicating that TRAF interactions with region contribute to CD40-mediated signaling. For example, the absence of an intact TRAF2,3,5 binding site results in reduced TNF␣ production in response to CD40 ligation, suggesting that TRAFs interacting with this binding region, though not essential for TNF␣ expression, contribute to this outcome [22]. Treatment of macrophages with a cell-permeable TRAF2,3,5 binding peptide also resulted in reduced expression of TNF␣, (J. Suttles, unpublished data). The signaling pathways engaged by CD40 ligation on monocytes/macrophages, and their functional outcomes are shown in Fig. 1. The common use of TRAF6 as adapter for signaling by both CD40 as well as TLR-mediated proinflammatory signaling pathways presents interesting possibilities for signaling cross-talk or cross-inhibition. It was found that the phenomenon of LPS tolerance, whereby macrophages stimulated by LPS are refractory to a secondary stimulus with LPS, also applies to CD40 signaling, as well. As discussed below, this phenomenon could be a major contributor

Fig. 1. Signaling pathways and functional outcomes of CD40 stimulation of monocytes/macrophages. TRAF6 is required for, and TRAF2 contributes to NF-␬B activation. ERK1/2 are activated via a Src, TRAF6-mediated pathway. Both the NF-␬B and ERK1/2 pathways contribute to a large number of primarily pro-inflammatory activities of monocytes and macrophages. Activation of ERK1/2 is also responsible for monocyte survival. Activation of p38 plays a role in activation of iNOS expression and PI3K activity contributes to induction of IL-10 synthesis.

260

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

to the immunosuppression associated with sepsis. Sinistro and colleagues found that stimulation of monocytes with LPS resulted in a reduced response to a subsequent stimulation via CD40 ligation, including reduced IL-12 and TNF␣ production [48]. Importantly, similar results were seen when monocytes from patients undergoing sepsis were evaluated. Monocytes from septic patients showed similar defects in TNF␣, IL-12, and IL-1␤ production, as well reduced expression of CD80 and CD86 [49]. We found that pre-incubation with CD154 results in strong suppression of a secondary LPS stimulus, indicating that the tolerance effect is bidirectional. CD40 simulation resulted in robust induction of SOCS3, which may be responsible for this inhibitory effect (J. Suttles, unpublished data). Not unexpectedly, CD40 stimulation of macrophages also results in a greatly reduced response to a subsequent stimulation of CD40. This could be due to a combination of receptor endocytosis, TRAF degradation, and induction SOCS3 expression. CD40 signal transduction in monocytes/macrophages can be modulated by a variety of cytokines, chemokines and hormones. The anti-inflammatory cytokines IL-4 and IL-10 have been shown to inhibit CD40-mediated activation of ERK1/2 and induction of IL1␤ synthesis, and were synergistic in this regard, suggesting that they may act at different points in the CD40 signaling pathway [31]. This was also evident by the finding that IL-4, but not IL-10 inhibited CD40-mediated rescue of monocytes from apoptosis [50]. Both IL-4 and IL-10 were also found to inhibit CD40-mediated induction of iNOS expression and PGE2 production by monocytes, but, again, this appeared to be due to different mechanisms. IL-10 significantly inhibited CD40-induced activation of the ERK1/2, p38, and NF-␬B pathways, whereas inhibition by IL-4 was limited to the ERK1/2 pathway [36]. Interestingly, although treatment of monocytes with IL-l0 strongly downregulated IL-12 production, treatment with IL-4 or IL-13 was found to enhance expression of IL-12 p40, and production of IL-12 p70 [51]. Thus, these two typically anti-inflammatory cytokines appear to differ in their ability to modulate CD40 signaling in monocytes/macrophages and act via differing mechanisms that have not been fully explored. Mechanisms of action likely include roles of suppressors of cytokine synthesis (SOCS) [52,53], activation of phosphatases [54] and activation of AMP-activated protein kinase, AMPK, recently identified as a potent suppressor of macrophage inflammatory signaling pathways that is activated by IL-4 and IL-10 [55]. Monocyte/macrophage inflammatory function has shown to be inhibited by the nuclear receptor superfamily members, peroxisome-proliferator activated receptors (PPARs), in particular PPAR␥ [56]. Although studies to date have focused on the ability of PPAR␥ to suppress a subset of TLR-regulated genes, it has been shown that macrophages that display elevated PPAR␥ activity, show greatly reduced NF-␬B activity and proinflammatory cytokine production induced by CD40 ligation [57] and PPAR␥ agonists inhibit CD40-induced activation of IL-12 production in DC [58]. Factors that enhance CD40 signaling include CCL2/MCP-1, which was shown to act synergistically with CD40 ligation in the induction of COX2 expression by macrophages [59]. Of course, the regulation of CD40 expression is a critical control point for reducing or enhancing the contribution of this receptor to monocyte/macrophage function. CD40 is an NF-␬B-regulated gene and, not surprisingly, it appears that proinflammatory stimuli upregulate CD40 expression, whereas typically anti-inflammatory stimuli downregulate CD40 expression [60]. CD40 expression on macrophages, as well as on numerous other cell types, is positively influenced by IFN␥. Thus the infiltration of IFN␥-producing cells serves to recruit cells of the tissue environment (e.g., resident tissue macrophages, endothelial cells, vascular smooth muscle cells, and fibroblasts) to become active participants in the inflammatory response [61]. Monocyte/macrophage expression of CD40 was shown to be enhanced by treatment with GM-CSF and IL-3, as well as via stim-

ulation with LPS [13,62–64]. IFN␥ and LPS stimulated expression of CD40 on macrophages and microglial cells can be negatively regulated by TGF␤, IL-4, and IL-10 [65–67]. IFN␥ enhancement of CD40 expression on monocytes/macrophages can also be inhibited by statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) [68–70], which may contribute to their antiinflammatory action [69].

4. Role of macrophage CD40 function in autoimmune inflammatory disease The most well-studied role of CD40 expression on myeloid cells is its function as a critical co-stimulatory molecule in myeloid DC:T cell interactions during the process of antigen presentation. Blockade of the CD40:CD154 interaction has been shown to inhibit onset of inflammatory disease in numerous animal models, including experimental autoimmune encephalomyelitis (EAE), collagen-induced arthritis (CIA), thyroiditis, uveitis, inflammatory bowel disease, and diabetes [71–77]. Although blockade of CD40:CD154 interactions can disrupt DC:T cell interactions and inhibit induction of disease, following activation of autoreactive T cells, T cell interactions with monocytes and macrophages at the sites of inflammation are a substantive contributor to the pathogenesis of autoimmune inflammatory disease. There are a number of examples in which blockade of CD40:CD154 interactions ameliorates ongoing disease. For example, in the relapsing EAE (SJL) model, treatment of mice with anti-CD154 antibody either during peak disease, or during remission was found to inhibit disease progression [78]. Anti-CD154 antibody treatment also inhibited disease in adoptive transfer recipients of encephalitogenic T cells, suggesting that blockade of CD40 activation of microglial cells (the resident macrophages of the brain) and infiltrating peripheral macrophages, contributes to the therapeutic effect of this treatment on ongoing disease [78]. It is cautioned that the abrogation of disease using this strategy in the relapsing EAE model may be transient if there is continued inflammatory stimulus, e.g., via epitope spreading [79]. Studies employing CD40 bone marrow chimeric mice have also revealed the importance of the CD40:CD154 interaction occurring between T cells and macrophages/microglial cells in the CNS. Using the myelin oligodendrocyte glycoprotein (MOG)-induced model of EAE, Becher et al. found that development of CNS inflammation was reduced in CD40+/+ → CD40−/− chimeric mice, even though T cell activation appeared normal [80]. Interestingly, infiltration of activated T cells into CNS was also impaired, suggesting that CD40 expression in CNS is required for production of inflammatory mediators/chemokines necessary for recruitment of T cells. These influences of CD40 expression in CNS are most likely attributable to the microglial compartment. In a similar study conducted by Ponomarev and colleagues, induction of EAE by adoptive transfer of encephalitogenic T cells resulted in reduced diseases in the absence of CD40 expression in CNS, which was accompanied by reduced infiltration of macrophages into CNS. The authors concluded that during the course of disease, microglial cells are induced to express MHC class II, CD40 and CD86 in a CD40-independent manner. A second, CD40-dependent activation step was required for disease progression, including expansion of encephalitogenic T cells and efficient recruitment of macrophages from the periphery [81]. Macrophages are also key contributors to the pathogenesis of rheumatoid arthritis (RA) [82] and models of collagen-induced arthritis also have shown dependency on the CD40:CD154 interaction for disease [72]. A recent report demonstrated that delivery of liposome carrier encapsulated CD40-directed antisense oligonucleotides reduced clinical severity of ongoing disease in murine CIA [83]. The efficacy of this approach, specifically with regard to the treatment of ongoing disease, was attributed to the tropism

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

of the reagent for macrophages and monocytes, in addition to DC. It is important to consider that the T cell macrophage interaction is reciprocal. In autoimmune inflammatory disease such as EAE/multiple sclerosis and CIA/rheumatoid arthritis, as well as inflammatory bowel disease, a role of both Th1 and Th17 Th subsets has been established [84] and, in the case of EAE, T cells expressing both IFN␥ and IL-17 are present during disease [85]. There is new appreciation for the potential plasticity of T cells and evidence suggests that interactions of T cells with monocytes/macrophages at the sites of inflammation can direct, or re-direct T cell responses. In a recent report, it was shown that monocytes derived from inflamed joints of patients with active RA, unlike monocytes from peripheral blood, specifically drove Th17 responses in a cell contact-dependent manner [86]. The cell contact-dependency suggests a possible role of CD40:CD154 interactions. Although not shown in this study, it is possible that the cytokine milieu of inflamed tissue may modify the response of monocytes/macrophages to CD40 ligation resulting in a specialized reciprocal stimulation of the CD154-expressing T cell. Overall, the functional significance of CD40 expression on monocytes and macrophages in the context of inflammatory autoimmune disease is very broad-reaching, including not only the wide range of activities of monocytes/macrophages induced through CD40 ligation, but also the reciprocal effects on T cell function.

5. Contributions of macrophage CD40 signaling to atherosclerosis Atherosclerosis is an inflammatory disease of complex etiology that displays many common features with autoimmune inflammatory diseases, including infiltration of T cells and monocytes/macrophage to the target site, in this case the vessel wall, and the recruitment of non-hematopoietic cells into the inflammatory response. Mouse models of atherosclerosis, including mice deficient for apolipoprotein E (ApoE) critical for cholesterol metabolism, and mice lacking low-density lipoprotein receptor (LDLR) have provided useful tools for the evaluation of the processes underlying atherogenesis. Formation of atherosclerotic plaque is initiated by infiltration of monocytes and T cells, with monocyte differentiation to macrophages occurring in the tissue, accompanied by a Th1 type inflammatory response. The production of inflammatory cytokines such as IFN␥ and TNF␣ induce expression of CD40 on endothelial cells and vascular smooth muscle cells, rendering them targets for CD154-expressing cells [87–89]. Although co-expression of both CD40 and CD154 on macrophages, endothelial cells and vascular smooth muscle cells in atherosclerosis has been reported [90], other laboratories have found CD154 expression to be exclusive to T cells and platelets, and absent on resident cells of the vasculature [88,91]. The importance of CD154 to the pathogenesis of atherosclerosis was shown by several studies employing the mouse models mentioned above and using either an antibody blockade approach to disrupt CD154-induced signaling, or use of CD154 deficient mice backcrossed onto the appropriate background (i.e., ApoE-deficient, LDLR-deficient). For example, Mach et al. [92], found that treatment of LDLR-deficient mice (fed a high cholesterol diet) with antiCD154, reduced the lipid content and size of aortic atherosclerotic lesions, as well as inhibited infiltration of macrophages and T cells. Lutgens et al. used mice deficient for CD154, backcrossed onto an ApoE-deficient background and examined both early and advanced lesions, concluding that initial lesion formation was not impacted, but late stage atherosclerotic changes were affected, with advanced plaques displaying reduced lipid content and a more stable plaque phenotype [93]. The source of CD154 most relevant to atherosclerosis was questioned by reports that the atherosclerotic plaque phenotype was not affected when CD154-deficient bone marrow

261

chimeras were generated on an LDLR-deficient background [94,95], which suggested that non-hematopoietic cells were the origin of CD154. Adding additional complexity are recent findings indicating that CD154 interacts with receptors other than CD40, including the integrins Mac-1 (CD11/CD18) and ␣5␤1 [96,97]. It has been reported that CD40-deficient/LDLR-deficient mice were not protected from development of atherosclerosis as compared to mice deficient for LDLR alone, suggesting to these authors that the contribution of CD154 to the disease is independent of CD40 and reliant on CD154:Mac-1 interactions [96]. However, this interpretation contrasts with numerous studies demonstrating that CD40-deficient mice display macrophage dysfunction, and that cross-linking of anti-CD40 antibodies results in macrophage signal transduction leading to a number of strongly pro-atherogenic outcomes. The elevated expression of CD40 on numerous cell types, including macrophages, in inflamed vascular tissue in atherosclerosis suggests a functional association with disease progression that requires further exploration. 6. Macrophage CD40 signaling in host defense CD40, in its role as a key co-stimulatory molecule in T cell:APC interactions, is required for the induction of effective immune responses against invading pathogens via directing Th1, Th2 and Th17 responses [98–100]. However, CD40 ligation of macrophages also contributes directly towards the induction of anti-microbial activities. As discussed above, CD40 ligation of macrophages effectively induces proinflammatory cytokine production [13,14,61], and CD40 ligation in the presence of IFN␥ induces full effector function of macrophages including production of NO [15,16]. These activities have been shown to contribute to responses against parasitic, fungal and bacterial infection. For example, mice deficient for CD154 infected with parasitic protozoan, Leishmania amazonensis, displayed a higher susceptibility to infection than wild-type mice, which was associated with impairment of both T cell and macrophage functions. In addition to the inability to generate a protective immune response, macrophage production of NO was impaired [101]. It has also been shown that infection by Trypanosoma cruzi is reduced by addition of exogenous CD154 [102], and this protection involved CD154 stimulation of macrophage NO production in addition to the induction of IL-12. In addition, infection of mice with T. cruzi transfected with CD154, resulted in greatly reduced parasitemia and mortality as compared with the control organism [103]. These authors also demonstrated that treatment of mice with small molecules that mimic trimeric CD154 (miniCD40Ls) can also successfully amplify host resistance to T cruzi. Importantly, this response was evident in wild-type, but not CD40-deficient mice, indicating that CD40 was the specific target of these CD154 mimetics [104]. Likewise, CD40:CD154 interactions were shown to contribute to restraint of growth of T. gondii, even in the absence of IFN␥ [105]. In this study, IFN␥-deficient mice infected with T. gondii were treated in vivo with agonistic anti-CD40 which was found to decrease parasite load. An investigation of the mechanism underlying this phenomenon revealed that in vivo ligation of CD40 increased TNF␣ production by T. gondii-infected macrophages, which appeared sufficient for anti-toxoplasmacidal activity [105]. This may be due to the described synergy between CD40 and TNF␣ in inducing macrophage effector function, which can bypass the need for IFN␥ [106]. T. gondii, as does many intracellular pathogens, survives in macrophages by residing in vacuoles and avoiding vacuole–lysosome fusion. Interestingly, an additional mechanism of CD40-mediated anti-microbial activity observed in this model was the ability of CD40 stimulation to induce autophagydependent vacuole–lysosome fusion [45], as mentioned previously. This TRAF6-dependent CD40-mediated mechanism may apply to

262

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

other organisms that use similar evasive strategies for survival in macrophages [46]. Macrophage CD40 signaling contributes similarly to defense against fungal infections. CD154-deficient mice infected via intratracheal inoculation with Cryptococcus neoformans displayed a greatly reduced inflammatory response in the lung, corresponding to an increased fungal burden, as compared to wild-type mice. These observations were accompanied by impaired macrophage NO production and impaired anti-microbial activity, as well as reduced IL-12 production [107]. Similarly, when CD154-deficient mice were subjected to Candida albicans infection, a lower fungal load in kidneys was observed as compared to wild-type mice, which could be mimicked by neutralizing anti-CD154 antibody. The CD154deficient mice showed lower plasma TNF␣ levels and reduced NO production by C. albicans-stimulated peritoneal macrophages was observed, which was discerned as the key mechanism for increased susceptibility to infection [108]. There is evidence that CD40 signaling also plays a role in bacterial sepsis, however, in this case it is exacerbating, rather than protective. Gold and colleagues subjected CD40-deficient mice to the cecal ligation and puncture model of polymicrobial sepsis and found that CD40-deficient mice displayed improved survival and reduced evidence of organ injury [109]. However, further studies by this group found that mice deficient for CD154 did not show this level of protection and evidence was presented indicating that CD40 stimulation in this model may be primarily via heat shock protein 70 (Hsp70) ligation of macrophage CD40 and subsequent induction of IL-12 production [110,111]. An outcome of sepsis is suppression of macrophage function such that responses to secondary stimuli are blunted. Although this may be a mechanism that could protect the host from uncontrolled inflammation and septic shock, it can result in a state of immunosuppression that renders the host unable to control a secondary infection. The cross-tolerance of TLR and CD40 creates a scenario by which attempts of CD154+ T cells to reactivate macrophages to combat infection would be impaired. Thus, this cross-tolerance effect could be viewed and an immunoevasive mechanism employed by bacteria. Interference of macrophage CD40 expression or interference with CD40 signaling machinery would favor pathogen survival. As an example, although CD40 ligation on macrophages typically results in IL-12 production, in Leishmania major infected macrophages, CD40 ligation preferentially induces IL-10. There is evidence that this may be a result of Leishmania’s propensity to deplete membrane cholesterol, which results in disruption of the assembly of IL-12-inducing CD40 signaling components in lipid rafts [112]. The net result is the creation of an environment that favors L. major survival.

7. Conclusions Macrophages display a remarkably diverse array of functions and adapt rapidly to changes in their microenvironment [8]. They are the most versatile of leukocyte populations and are present in nearly all tissues where they help maintain tissue homeostasis and provide defense against invading pathogens. However, their ubiquitous presence and their sensitivity to a broad range of stimuli also targets them as easy recruits into disease pathologies. Although macrophages are primarily considered in the context of innate responses, e.g., as responders to TLR stimulation, the functional expression of CD40 on macrophage confers upon them the ability to play a significant role in the outcome of adaptive immune responses. Antigen-activated CD154+ T cells can drive macrophage effector functions to provide antigen-driven protective effects in host defense. On the other hand, in the case of autoimmune inflammatory disease, autoantigen-activated CD154+ T cells can activate macrophage effector functions resulting in tis-

sue destruction. The ability to manipulate macrophage function, hence redirecting their behavior, has suggested that macrophages are an excellent targets for disease-modifying therapies. For example, macrophage-targeted blockade of CD40 signaling could provide benefit for ongoing autoimmune inflammatory disease. References [1] van Furth R, Cohn ZA. The origin and kinetics of mononuclear phagocytes. J Exp Med 1968;128:415–35. [2] van Furth R, Diesselhoff-den Dulk MC, Mattie H. Quantitative study on the production and kinetics of mononuclear phagocytes during an acute inflammatory reaction. J Exp Med 1973;138:1314–30. [3] Kennedy DW, Abkowitz JL. Mature monocytic cells enter tissues and engraft. Proc Natl Acad Sci USA 1998;95:14944–9. [4] Kennedy DW, Abkowitz JL. Kinetics of central nervous system microglial and macrophage engraftment: analysis using a transgenic bone marrow transplantation model. Blood 1997;90:986–93. [5] Laskin DL, Weinberger B, Laskin JD. Functional heterogeneity in liver and lung macrophages. J Leukoc Biol 2001;70:163–70. [6] Guillemin GJ, Brew BJ. Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. J Leukoc Biol 2004;75:388–97. [7] Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev Immunol 2005;5:606–16. [8] Stout RD, Suttles J. Functional plasticity of macrophages: reversible adaptation to changing microenvironments. J Leukoc Biol 2004;76:509–13. [9] Sypek JP, Panosian CB, Wyler DJ. Cell contact-mediated macrophage activation for antileishmanial defense. II. Identification of effector cell phenotype and genetic restriction. J Immunol 1984;133:3351–7. [10] Stout RD, Bottomly K. Antigen-specific activation of effector macrophages by IFN-gamma producing (TH1) T cell clones. Failure of IL-4-producing (TH2) T cell clones to activate effector function in macrophages. J Immunol 1989;142:760–5. [11] Sypek JP, Wyler DJ. Antileishmanial defense in macrophages triggered by tumor necrosis factor expressed on CD4+ T lymphocyte plasma membrane. J Exp Med 1991;174:755–9. [12] Suttles J, Miller RW, Tao X, Stout RD. T cells which do not express membrane tumor necrosis factor-alpha activate macrophage effector function by cell contact-dependent signaling of macrophage tumor necrosis factor-alpha production. Eur J Immunol 1994;24:1736–42. [13] Alderson MR, Armitage RJ, Tough TW, Strockbine L, Fanslow WC, Spriggs MK. CD40 expression by human monocytes: regulation by cytokines and activation of monocytes by the ligand for CD40. J Exp Med 1993;178: 669–74. [14] Wagner Jr DH, Stout RD, Suttles J. Role of the CD40–CD40 ligand interaction in CD4+ T cell contact-dependent activation of monocyte interleukin-1 synthesis. Eur J Immunol 1994;24:3148–54. [15] Tian L, Noelle RJ, Lawrence DA. Activated T cells enhance nitric oxide production by murine splenic macrophages through gp39 and LFA-1. Eur J Immunol 1995;25:306–9. [16] Stout RD, Suttles J, Xu J, Grewal IS, Flavell RA. Impaired T cellmediated macrophage activation in CD40 ligand-deficient mice. J Immunol 1996;156:8–11. [17] Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G, et al. CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells. Nature 1998;391:591–4. [18] Gauchat JF, Henchoz S, Mazzei G, Aubry JP, Brunner T, Blasey H, et al. Induction of human IgE synthesis in B cells by mast cells and basophils. Nature 1993;365:340–3. [19] Henn V, Steinbach S, Buchner K, Presek P, Kroczek RA. The inflammatory action of CD40 ligand (CD154) expressed on activated human platelets is temporally limited by coexpressed CD40. Blood 2001;98:1047–54. [20] Ferrari-Lacraz S, Nicod LP, Chicheportiche R, Welgus HG, Dayer JM. Human lung tissue macrophages, but not alveolar macrophages, express matrix metalloproteinases after direct contact with activated T lymphocytes. Am J Respir Cell Mol Biol 2001;24:442–51. [21] Kiener PA, Moran-Davis P, Rankin BM, Wahl AF, Aruffo A, Hollenbaugh D. Stimulation of CD40 with purified soluble gp39 induces proinflammatory responses in human monocytes. J Immunol 1995;155:4917–25. [22] Mukundan L, Bishop GA, Head KZ, Zhang L, Wahl LM, Suttles J. TNF receptor-associated factor 6 is an essential mediator of CD40-activated proinflammatory pathways in monocytes and macrophages. J Immunol 2005;174:1081–90. [23] Kennedy MK, Picha KS, Fanslow WC, Grabstein KH, Alderson MR, Clifford KN, et al. CD40/CD40 ligand interactions are required for T cell-dependent production of interleukin-12 by mouse macrophages. Eur J Immunol 1996;26: 370–8. [24] Shu U, Kiniwa M, Wu CY, Maliszewski C, Vezzio N, Hakimi J, et al. Activated T cells induce interleukin-12 production by monocytes via CD40–CD40 ligand interaction. Eur J Immunol 1995;25:1125–8. [25] Kornbluth RS, Kee K, Richman DD. CD40 ligand (CD154) stimulation of macrophages to produce HIV-1-suppressive beta-chemokines. Proc Natl Acad Sci USA 1998;95:5205–10.

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264 [26] Wu L, Fan J, Matsumoto S, Watanabe T. Induction and regulation of matrix metalloproteinase-12 by cytokines and CD40 signaling in monocyte/macrophages. Biochem Biophys Res Commun 2000;269:808–15. [27] Malik N, Greenfield BW, Wahl AF, Kiener PA. Activation of human monocytes through CD40 induces matrix metalloproteinases. J Immunol 1996;156:3952–60. [28] Mach F, Schonbeck U, Bonnefoy JY, Pober JS, Libby P. Activation of monocyte/macrophage functions related to acute atheroma complication by ligation of CD40: induction of collagenase, stromelysin, and tissue factor. Circulation 1997;96:396–9. [29] Suttles J, Evans M, Miller RW, Poe JC, Stout RD, Wahl LM. T cell rescue of monocytes from apoptosis: role of the CD40–CD40L interaction and requirement for CD40-mediated induction of protein tyrosine kinase activity. J Leukoc Biol 1996;60:651–7. [30] Stout RD, Suttles J. T cell–macrophage cognate interaction in the activation of macrophage effector function by Th2 cells. J Immunol 1993;150:5330–7. [31] Suttles J, Milhorn DM, Miller RW, Poe JC, Wahl LM, Stout RD. CD40 signaling of monocyte inflammatory cytokine synthesis through an ERK1/2-dependent pathway. A target of interleukin (il)-4 and il-10 anti-inflammatory action. J Biol Chem 1999;274:5835–42. [32] Buhtoiarov IN, Lum H, Berke G, Paulnock DM, Sondel PM, Rakhmilevich AL. CD40 ligation activates murine macrophages via an IFN-gammadependent mechanism resulting in tumor cell destruction in vitro. J Immunol 2005;174:6013–22. [33] Lum HD, Buhtoiarov IN, Schmidt BE, Berke G, Paulnock DM, Sondel PM, et al. In vivo CD40 ligation can induce T-cell-independent antitumor effects that involve macrophages. J Leukoc Biol 2006;79:1181–92. [34] Kilinc MO, Mukundan L, Yolcu ES, Singh NP, Suttles J, Shirwan H. Generation of a multimeric form of CD40L with potent immunostimulatory activity using streptavidin as a chaperon. Exp Mol Pathol 2006;80:252–61. [35] Pearson LL, Castle BE, Kehry MR. CD40-mediated signaling in monocytic cells: up-regulation of tumor necrosis factor receptor-associated factor mRNAs and activation of mitogen-activated protein kinase signaling pathways. Int Immunol 2001;13:273–83. [36] Inoue Y, Otsuka T, Niiro H, Nagano S, Arinobu Y, Ogami E, et al. Novel regulatory mechanisms of CD40-induced prostanoid synthesis by IL-4 and IL-10 in human monocytes. J Immunol 2004;172:2147–54. [37] Mathur RK, Awasthi A, Wadhone P, Ramanamurthy B, Saha B. Reciprocal CD40 signals through p38MAPK and ERK-1/2 induce counteracting immune responses. Nat Med 2004;10:540–4. [38] Foey AD, Feldmann M, Brennan FM. CD40 ligation induces macrophage IL-10 and TNF-alpha production: differential use of the PI3K and p42/44 MAPKpathways. Cytokine 2001;16:131–42. [39] Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol 2005;6:777–84. [40] Eliopoulos AG, Wang CC, Dumitru CD, Tsichlis PN. Tpl2 transduces CD40 and TNF signals that activate ERK and regulates IgE induction by CD40. EMBO J 2003;22:3855–64. [41] Inoue J, Ishida T, Tsukamoto N, Kobayashi N, Naito A, Azuma S, et al. Tumor necrosis factor receptor-associated factor (TRAF) family: adapter proteins that mediate cytokine signaling. Exp Cell Res 2000;254:14–24. [42] Bishop GA. The multifaceted roles of TRAFs in the regulation of B-cell function. Nat Rev Immunol 2004;4:775–86. [43] Bishop GA, Moore CR, Xie P, Stunz LL, Kraus ZJ. TRAF proteins in CD40 signaling. Adv Exp Med Biol 2007;597:131–51. [44] Mackey MF, Wang Z, Eichelberg K, Germain RN. Distinct contributions of different CD40 TRAF binding sites to CD154-induced dendritic cell maturation and IL-12 secretion. Eur J Immunol 2003;33:779–89. [45] Andrade RM, Wessendarp M, Gubbels MJ, Striepen B, Subauste CS. CD40 induces macrophage anti-Toxoplasma gondii activity by triggering autophagydependent fusion of pathogen-containing vacuoles and lysosomes. J Clin Invest 2006;116:2366–77. [46] Subauste CS, Andrade RM, Wessendarp M. CD40-TRAF6 and autophagydependent anti-microbial activity in macrophages. Autophagy 2007;3: 245–8. [47] Pullen SS, Labadia ME, Ingraham RH, McWhirter SM, Everdeen DS, Alber T, et al. High-affinity interactions of tumor necrosis factor receptor-associated factors (TRAFs) and CD40 require TRAF trimerization and CD40 multimerization. Biochemistry 1999;38:10168–77. [48] Sinistro A, Ciaprini C, Natoli S, Sussarello E, Carducci FC, Almerighi C, et al. Lipopolysaccharide desensitizes monocytes–macrophages to CD40 ligand stimulation. Immunology 2007;122:362–70. [49] Sinistro A, Almerighi C, Ciaprini C, Natoli S, Sussarello E, Di FS, et al. Downregulation of CD40 ligand response in monocytes from sepsis patients. Clin Vaccine Immunol 2008;15:1851–8. [50] Poe JC, Wagner Jr DH, Miller RW, Stout RD, Suttles J. IL-4 and IL-10 modulation of CD40-mediated signaling of monocyte IL-1beta synthesis and rescue from apoptosis. J Immunol 1997;159:846–52. [51] Bullens DM, Kasran A, Thielemans K, Bakkus M, Ceuppens JL. CD40L-induced IL-12 production is further enhanced by the Th2 cytokines IL-4 and IL-13. Scand J Immunol 2001;53:455–63. [52] Berlato C, Cassatella MA, Kinjyo I, Gatto L, Yoshimura A, Bazzoni F. Involvement of suppressor of cytokine signaling-3 as a mediator of the inhibitory effects of IL-10 on lipopolysaccharide-induced macrophage activation. J Immunol 2002;168:6404–11.

263

[53] Dickensheets H, Vazquez N, Sheikh F, Gingras S, Murray PJ, Ryan JJ, et al. Suppressor of cytokine signaling-1 is an IL-4-inducible gene in macrophages and feedback inhibits IL-4 signaling. Genes Immun 2007;8:21–7. [54] Jiang H, Harris MB, Rothman P. IL-4/IL-13 signaling beyond JAK/STAT. J Allergy Clin Immunol 2000;105:1063–70. [55] Sag D, Carling D, Stout RD, Suttles J. Adenosine 5 -monophosphate-activated protein kinase promotes macrophage polarization to an anti-inflammatory functional phenotype. J Immunol 2008;181:8633–41. [56] Straus DS, Glass CK. Anti-inflammatory actions of PPAR ligands: new insights on cellular and molecular mechanisms. Trends Immunol 2007;28:551–8. [57] Makowski L, Brittingham KC, Reynolds JM, Suttles J, Hotamisligil GS. The fatty acid-binding protein, aP2, coordinates macrophage cholesterol trafficking and inflammatory activity. Macrophage expression of aP2 impacts peroxisome proliferator-activated receptor gamma and IkappaB kinase activities. J Biol Chem 2005;280:12888–95. [58] Faveeuw C, Fougeray S, Angeli V, Fontaine J, Chinetti G, Gosset P, et al. Peroxisome proliferator-activated receptor gamma activators inhibit interleukin-12 production in murine dendritic cells. FEBS Lett 2000;486:261–6. [59] Futagami S, Tatsuguchi A, Hiratsuka T, Shindo T, Horie A, Hamamoto T, et al. Monocyte chemoattractant protein 1 and CD40 ligation have a synergistic effect on vascular endothelial growth factor production through cyclooxygenase 2 upregulation in gastric cancer. J Gastroenterol 2008;43:216–24. [60] Benveniste EN, Nguyen VT, Wesemann DR. Molecular regulation of CD40 gene expression in macrophages and microglia. Brain Behav Immun 2004;18:7–12. [61] Stout RD, Suttles J. The many roles of CD40 in cell-mediated inflammatory responses. Immunol Today 1996;17:487–92. [62] Qin H, Wilson CA, Lee SJ, Zhao X, Benveniste EN. LPS induces CD40 gene expression through the activation of NF-kappaB and STAT-1alpha in macrophages and microglia. Blood 2005;106:3114–22. [63] Wu W, Alexis NE, Bromberg PA, Jaspers I, Peden DB. Mechanisms of LPSinduced CD40 expression in human peripheral blood monocytic cells. Biochem Biophys Res Commun 2009;379:573–7. [64] Wu W, Alexis NE, Chen X, Bromberg PA, Peden DB. Involvement of mitogenactivated protein kinases and NFkappaB in LPS-induced CD40 expression on human monocytic cells. Toxicol Appl Pharmacol 2008;228:135–43. [65] Nguyen VT, Walker WS, Benveniste EN. Post-transcriptional inhibition of CD40 gene expression in microglia by transforming growth factor-beta. Eur J Immunol 1998;28:2537–48. [66] Nguyen VT, Benveniste EN. IL-4-activated STAT-6 inhibits IFN-gammainduced CD40 gene expression in macrophages/microglia. J Immunol 2000;165:6235–43. [67] Qin H, Wilson CA, Roberts KL, Baker BJ, Zhao X, Benveniste EN. IL-10 inhibits lipopolysaccharide-induced CD40 gene expression through induction of suppressor of cytokine signaling-3. J Immunol 2006;177:7761–71. [68] Garlichs CD, John S, Schmeisser A, Eskafi S, Stumpf C, Karl M, et al. Upregulation of CD40 and CD40 ligand (CD154) in patients with moderate hypercholesterolemia. Circulation 2001;104:2395–400. [69] Youssef S, Stuve O, Patarroyo JC, Ruiz PJ, Radosevich JL, Hur EM, et al. The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature 2002;420:78–84. [70] Lee SJ, Qin H, Benveniste EN. Simvastatin inhibits IFN-gamma-induced CD40 gene expression by suppressing STAT-1alpha. J Leukoc Biol 2007;82:436–47. [71] Gerritse K, Laman JD, Noelle RJ, Aruffo A, Ledbetter JA, Boersma WJ, et al. CD40–CD40 ligand interactions in experimental allergic encephalomyelitis and multiple sclerosis. Proc Natl Acad Sci USA 1996;93:2499–504. [72] Durie FH, Fava RA, Foy TM, Aruffo A, Ledbetter JA, Noelle RJ. Prevention of collagen-induced arthritis with an antibody to gp39, the ligand for CD40. Science 1993;261:1328–30. [73] Carayanniotis G, Masters SR, Noelle RJ. Suppression of murine thyroiditis via blockade of the CD40–CD40L interaction. Immunology 1997;90:421–6. [74] Bagenstose LM, Agarwal RK, Silver PB, Harlan DM, Hoffmann SC, Kampen RL, et al. Disruption of CD40/CD40–ligand interactions in a retinal autoimmunity model results in protection without tolerance. J Immunol 2005;175:124–30. [75] Namba K, Ogasawara K, Kitaichi N, Morohashi T, Sasamoto Y, Kotake S, et al. Amelioration of experimental autoimmune uveoretinitis by pretreatment with a pathogenic peptide in liposome and anti-CD40 ligand monoclonal antibody. J Immunol 2000;165:2962–9. [76] Danese S, Sans M, Fiocchi C. The CD40/CD40L costimulatory pathway in inflammatory bowel disease. Gut 2004;53:1035–43. [77] Balasa B, Krahl T, Patstone G, Lee J, Tisch R, McDevitt HO, et al. CD40 ligand–CD40 interactions are necessary for the initiation of insulitis and diabetes in nonobese diabetic mice. J Immunol 1997;159:4620–7. [78] Howard LM, Miga AJ, Vanderlugt CL, Dal Canto MC, Laman JD, Noelle RJ, et al. Mechanisms of immunotherapeutic intervention by anti-CD40L (CD154) antibody in an animal model of multiple sclerosis. J Clin Invest 1999;103:281–90. [79] Howard LM, Miller SD. Immunotherapy targeting the CD40/CD154 costimulatory pathway for treatment of autoimmune disease. Autoimmunity 2004;37:411–8. [80] Becher B, Durell BG, Miga AV, Hickey WF, Noelle RJ. The clinical course of experimental autoimmune encephalomyelitis and inflammation is controlled by the expression of CD40 within the central nervous system. J Exp Med 2001;193:967–74. [81] Ponomarev ED, Shriver LP, Dittel BN. CD40 expression by microglial cells is required for their completion of a two-step activation process during central nervous system autoimmune inflammation. J Immunol 2006;176:1402–10.

264

J. Suttles, R.D. Stout / Seminars in Immunology 21 (2009) 257–264

[82] Drexler SK, Kong PL, Wales J, Foxwell BM. Cell signalling in macrophages, the principal innate immune effector cells of rheumatoid arthritis. Arthritis Res Ther 2008;10:216. [83] Andreakos E, Rauchhaus U, Stavropoulos A, Endert G, Wendisch V, Benahmed AS, et al. Amphoteric liposomes enable systemic antigen-presenting celldirected delivery of CD40 antisense and are therapeutically effective in experimental arthritis. Arthritis Rheum 2009;60:994–1005. [84] Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev 2008;223:87–113. [85] Suryani S, Sutton I. An interferon-gamma-producing Th1 subset is the major source of IL-17 in experimental autoimmune encephalitis. J Neuroimmunol 2007;183:96–103. [86] Evans HG, Gullick NJ, Kelly S, Pitzalis C, Lord GM, Kirkham BW, et al. In vivo activated monocytes from the site of inflammation in humans specifically promote Th17 responses. Proc Natl Acad Sci USA 2009. [87] Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 2006;6:508–19. [88] Szabolcs MJ, Cannon PJ, Thienel U, Chen R, Michler RE, Chess L, et al. Analysis of CD154 and CD40 expression in native coronary atherosclerosis and transplant associated coronary artery disease. Virchows Arch 2000;437: 149–59. [89] Mukundan L, Milhorn DM, Matta B, Suttles J. CD40-mediated activation of vascular smooth muscle cell chemokine production through a Src-initiated, MAPK-dependent pathway. Cell Signal 2004;16:375–84. [90] Mach F, Schonbeck U, Sukhova GK, Bourcier T, Bonnefoy JY, Pober JS, et al. Functional CD40 ligand is expressed on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for CD40–CD40 ligand signaling in atherosclerosis. Proc Natl Acad Sci USA 1997;94:1931–6. [91] Buchner K, Henn V, Grafe M, de Boer OJ, Becker AE, Kroczek RA. CD40 ligand is selectively expressed on CD4+ T cells and platelets: implications for CD40CD40L signalling in atherosclerosis. J Pathol 2003;201:288–95. [92] Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature 1998;394:200–3. [93] Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS, Koteliansky VE, et al. Requirement for CD154 in the progression of atherosclerosis. Nat Med 1999;5:1313–6. [94] Bavendiek U, Zirlik A, LaClair S, MacFarlane L, Libby P, Schonbeck U. Atherogenesis in mice does not require CD40 ligand from bone marrow-derived cells. Arterioscler Thromb Vasc Biol 2005;25:1244–9. [95] Smook ML, Heeringa P, Damoiseaux JG, Daemen MJ, de Winther MP, Gijbels MJ, et al. Leukocyte CD40L deficiency affects the CD25(+) CD4 T cell population but does not affect atherosclerosis. Atherosclerosis 2005;183: 275–82. [96] Zirlik A, Maier C, Gerdes N, MacFarlane L, Soosairajah J, Bavendiek U, et al. CD40 ligand mediates inflammation independently of CD40 by interaction with Mac-1. Circulation 2007;115:1571–80. [97] Leveille C, Bouillon M, Guo W, Bolduc J, Sharif-Askari E, El-Fakhry Y, et al. CD40 ligand binds to alpha5beta1 integrin and triggers cell signaling. J Biol Chem 2007;282:5143–51.

[98] Mackey MF, Barth Jr RJ, Noelle RJ. The role of CD40/CD154 interactions in the priming, differentiation, and effector function of helper and cytotoxic T cells. J Leukoc Biol 1998;63:418–28. [99] Iezzi G, Sonderegger I, Ampenberger F, Schmitz N, Marsland BJ, Kopf M. CD40–CD40L cross-talk integrates strong antigenic signals and microbial stimuli to induce development of IL-17-producing CD4+ T cells. Proc Natl Acad Sci USA 2009;106:876–81. [100] Perona-Wright G, Jenkins SJ, O’Connor RA, Zienkiewicz D, McSorley HJ, Maizels RM, et al. A pivotal role for CD40-mediated IL-6 production by dendritic cells during IL-17 induction in vivo. J Immunol 2009;182:2808–15. [101] Soong L, Xu JC, Grewal IS, Kima P, Sun J, Longley BJ, et al. Disruption of CD40CD40 ligand interactions results in an enhanced susceptibility to Leishmania amazonensis infection. Immunity 1996;4:263–73. [102] Chaussabel D, Jacobs F, de JJ, de VM, Carlier Y, Thielemans K, et al. CD40 ligation prevents Trypanosoma cruzi infection through interleukin-12 upregulation. Infect Immun 1999;67:1929–34. [103] Chamekh M, Vercruysse V, Habib M, Lorent M, Goldman M, Allaoui A, et al. Transfection of Trypanosoma cruzi with host CD40 ligand results in improved control of parasite infection. Infect Immun 2005;73:6552–61. [104] Habib M, Rivas MN, Chamekh M, Wieckowski S, Sun W, Bianco A, et al. Cutting edge: small molecule CD40 ligand mimetics promote control of parasitemia and enhance T cells producing IFN-gamma during experimental Trypanosoma cruzi infection. J Immunol 2007;178:6700–4. [105] Subauste CS, Wessendarp M. CD40 restrains in vivo growth of Toxoplasma gondii independently of gamma interferon. Infect Immun 2006;74:1573–9. [106] Andrade RM, Wessendarp M, Portillo JA, Yang JQ, Gomez FJ, Durbin JE, et al. TNF receptor-associated factor 6-dependent CD40 signaling primes macrophages to acquire antimicrobial activity in response to TNF-alpha. J Immunol 2005;175:6014–21. [107] Pietrella D, Lupo P, Perito S, Mosci P, Bistoni F, Vecchiarelli A. Disruption of CD40/CD40L interaction influences the course of Cryptococcus neoformans infection. FEMS Immunol Med Microbiol 2004;40:63–70. [108] Netea MG, Meer JW, Verschueren I, Kullberg BJ. CD40/CD40 ligand interactions in the host defense against disseminated Candida albicans infection: the role of macrophage-derived nitric oxide. Eur J Immunol 2002;32:1455–63. [109] Gold JA, Parsey M, Hoshino Y, Hoshino S, Nolan A, Yee H, et al. CD40 contributes to lethality in acute sepsis: in vivo role for CD40 in innate immunity. Infect Immun 2003;71:3521–8. [110] Nolan A, Weiden MD, Hoshino Y, Gold JA. Cd40 but not CD154 knockout mice have reduced inflammatory response in polymicrobial sepsis: a potential role for Escherichia coli heat shock protein 70 in CD40-mediated inflammation in vivo. Shock 2004;22:538–42. [111] Wang Y, Kelly CG, Karttunen JT, Whittall T, Lehner PJ, Duncan L, et al. CD40 is a cellular receptor mediating mycobacterial heat shock protein 70 stimulation of CC-chemokines. Immunity 2001;15:971–83. [112] Rub A, Dey R, Jadhav M, Kamat R, Chakkaramakkil S, Majumdar S, et al. Cholesterol depletion associated with Leishmania major infection alters macrophage CD40 signalosome composition and effector function. Nat Immunol 2009;10:273–80.