Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2011 Sep 15;60(11):1529–1541. doi: 10.1007/s00262-011-1104-5

Dual biological effects of the cytokines interleukin-10 and interferon-γ

Cailin Moira Wilke 1,2, Shuang Wei 1, Lin Wang 1,3, Ilona Kryczek 1, John Kao 4,5, Weiping Zou 1,2,5,6,
PMCID: PMC11029274  PMID: 21918895

Abstract

It is generally thought that each cytokine exerts either immune stimulatory (inflammatory) or immune inhibitory (antiinflammatory or regulatory) biological activities. However, multiple cytokines can enact both inhibitory and stimulatory effects on the immune system. Two of these cytokines are interleukin (IL)-10 and interferon-gamma (IFNγ). IL-10 has demonstrated antitumor immunity even though it has been known for years as an immunoregulatory protein. Generally perceived as an immune stimulatory cytokine, IFNγ can also induce inhibitory molecule expression including B7-H1 (PD-L1), indoleamine 2,3-dioxygenase (IDO), and arginase on multiple cell populations (dendritic cells, tumor cells, and vascular endothelial cells). In this review, we will summarize current knowledge of the dual roles of both of these cytokines and stress the previously underappreciated stimulatory role of IL-10 and inhibitory role of IFNγ in the context of malignancy. Our progressive understanding of the dual effects of these cytokines is important for dissecting cytokine-associated pathology and provides new avenues for developing effective immune therapy against human diseases, including cancer.

Keywords: Regulatory T cell, IL-10, IFNγ, B7-H1, Inflammation, Cancer

IL-10

IL-10 was first identified in 1988 as a product of T helper type 2 (Th2) cells that inhibited cytokine production from T helper type 1 (Th1) cells [1]. In addition to Th2 cells, IL-10 is produced by other cell populations, including macrophages and dendritic cells (DCs), B cells [2], keratinocytes, mast cells [3, 4], and several subsets of T cells, including regulatory T cells (Tregs) [5, 6], and T helper 17 cells (Th17) [7]. Tregs release IL-10 as one of their many immunosuppressive activities [6, 8, 9]. IL-10 targets T cells and antigen-presenting cells (APC) and mediates immune regulation [1012]. Here, we provide a summary of the defined immunoregulatory roles of IL-10 and emphasize the heretofore unappreciated immunostimulatory roles of IL-10 (Table 1).

Table 1.

Interleukin-10

Effect Mechanism Context References
Immune inhibitory effects
Prevents DC trafficking to lymph node Autocrine signaling in DCs Mycobacterium infection [18]
Inhibition of TAA responses CD8+ Treg production of IL-10 Cancer [5]
Increased susceptibility to re-infection, chronic infection, transplanted tumors IL-10 in lungs, systemic IL-10, IL-10 from Tregs, exogenous/ectopically expressed IL-10 Health, infection, cancer [2530]
Suppression of NK and NKT cell activity αβ and γδ T-cell production of IL-10 Cancer [37]
Decreasing CTL, NK killing IL-10 from tumor cells and other populations, downregulation of MHC on tumors, increase in non-classical MHC on tumors Cancer [3841]
Impedes T-cell proliferation Tumor-infiltrating IL-10+ monocytes Cancer [35]
Increases in CTLA-4 expression IL-10 in tumor environment Cancer [42]
Maintenance of FoxP3 expression and Treg suppressive function Myeloid-derived IL-10 Colitis [20]
Maintenance of intestinal homeostasis Treg and mucosal IL-10 Health [6, 2124]
Constrains Th17 populations IL-10 modulation of IL-1 expression in DC Health, colitis Wilke, under review
Decreases TAA-specific killing, but increases activation of iNKT cells IL-10+ neutrophils in tumor-bearing hosts Cancer [36]
Non-immune effects
Protects neurons from toxin-induced damage IL-10 signaling in spinal cord neurons [60]
Inhibition of autophagy in macrophages Exogenous IL-10 treatment [61]
Increases angiogenesis Possibly through regulation of VEGF, NO expression Hypoxia [62]
Immune stimulatory effects
Increases MHC II expression on B cells, B cell survival, proliferation, Ig production Exogenous IL-10 Health [4649]
Supports growth and proliferation of T cells, induces CTL precursors and increases CTL function In combination with IL-2 and IL-4, IL-10 promotes CTL Health [51, 52]
Tumor cells rejected, tumor metastases decreased IL-10-transfected tumor cells; decrease in tumor MHC, increased NK lysis of tumor Cancer [5456]
Increased activated memory T cells, Ag-specific T cell proliferation and IFNγ production IL-10 administered to tumor-bearing mice Cancer [58]
Limits immunosuppressive cell populations in the tumor environment Limits IL-1 expression in MDSC, Treg induction Cancer [59]

IL-10 and immunoregulation

IL-10 enacts most of its immunosuppressive activity indirectly, via effects on APCs. IL-10 downregulates major histocompatibility molecule (MHC) [13] and B7 co-stimulatory molecule expression [14] and promotes inhibitory B7-H1 expression on APCs [15]. It limits proinflammatory cytokine and chemokine expression in APCs but can also directly affect CD4+ T cells by limiting their activation, proliferation, and cytokine production [10, 11, 16, 17]. Interestingly, a study with mycobacterium demonstrated that autocrine IL-10 signaling in DCs can prevent their trafficking to lymph nodes [18]. This impedes the recruitment of naïve T cells to the draining lymph nodes of infected tissue, as well as polarization of these same T cells to a Th1 phenotype.

It has recently been shown that IL-10 is associated with the suppressive effects and development of Tregs. Human tumor-associated CD8+ Tregs produce IL-10 and inhibit tumor-associated antigen (TAA)-specific T-cell responses. We have shown that these CD8+ Tregs may be induced by plasmacytoid DCs [5]. CD4+ Tregs also produce IL-10 and mediate immune suppression through IL-10 [6, 8, 9, 19]. In addition to the importance of IL-10 in Treg-mediated suppression, myeloid-derived IL-10 maintains Forkhead box P3 (FoxP3) expression and suppressive function of Tregs in mice with colitis [20]. It indicates that IL-10 is involved in Treg development in the gut. We have also observed that IL-10 is essential in constraining Th17 cell population development in mice and in patients with Crohn’s disease through its control of IL-1 expression (Wilke et al., unpublished data). Indeed, several studies have revealed the roles of IL-10 in mediating intestinal homeostasis through its immune regulatory function [6, 2124].

Although much of our current knowledge regarding the in vivo immune regulatory functions of IL-10 arises from studies of infectious disease models [2530], the immune suppressive effects of IL-10 have also been documented in tumor-bearing hosts. In several preclinical models, blockade of IL-10 signaling in tumor or T cells has been shown to enhance antitumor immunity [3133]. Tumor-infiltrating myeloid cells are capable of producing large amounts of IL-10 [34, 35]. In addition to myeloid cells [34, 35], IL-10 may be derived from tumor-associated neutrophils. A very recent manuscript demonstrated the ability of the acute phase protein serum amyloid A-1 (SAA-1) to induce IL-10-secreting neutrophils that inhibited antigen-specific CD8+ T-cell expansion in melanoma patients [36]. IL-10 can suppress NK and NKT cell activity and downregulate MHC molecule expression on tumor cells and DCs, thereby hindering CTL-mediated killing [3740].Tumor cell-derived IL-10 has been associated with an increased expression of the non-classical HLA class Ib molecule HLA-G. This also obstructs host CTL cytolytic activity [41]. Furthermore, it has been reported that the suppressive effects of IL-10 may be linked to the action of CTLA-4 [42] and cyclooxygenase (COX)-2 [4345], two other molecules of interest in the study of tumor immunity. Altogether, it is evident that IL-10 can target multiple immune cell subsets and mediate immune suppression through several modes of action.

IL-10 and immunostimulation

The first immune stimulatory function of IL-10 was observed in B cells. Mouse splenic B cells upregulate their expression of MHC II upon treatment with either human or mouse rIL-10 [46]. IL-10 also serves as a survival factor for B cells and increases their antibody production. Early experiments on human B cells demonstrated that IL-10 served as a co-stimulatory factor for B cell proliferation and synergized with IL-4 to expand B cell cultures [47]. Additionally, IL-10 treatment stimulated B cell production of the immunoglobulins (Ig)M, IgG, and IgA, including autoimmune antibodies [48]. A subsequent study showed that IL-10 supported viability of human germinal center B cells and induced synthesis of B-cell lymphoma 2 (Bcl-2) [49], a protein that was already known to play a key role in the rescue of germinal center B cells from apoptosis [50].

IL-10 can also mediate immune stimulatory effects on T cells. MacNeil et al. first showed that IL-10, along with IL-2 and/or IL-4, supported the growth and proliferation of both mature and immature T cells [51]. Shortly thereafter, more specific effects of IL-10 on CD8+ T cells were explored. IL-10 was found to be capable of inducing cytotoxic lymphocyte precursors and augmenting cytotoxic function [52]. IL-10 is therefore a growth and differentiation factor for CD8+ T cells.

A few laboratories have investigated the stimulatory role of IL-10 in the context of tumor immunity [53]. Suzuki et al. showed that tumor cells transfected with murine IL-10 grew more slowly in vivo and were frequently rejected by the host animals [54]. The following year, IL-10 was discovered to inhibit tumor metastasis in both experimental and spontaneous tumor models via effects on natural killer (NK) cells [55]. Although the precise mechanisms involved were not explored in this paper, a subsequent study from the Fulton group demonstrated that IL-10 downregulated MHC I expression on tumor cells and in doing so, supported NK cell-mediated tumor cell lysis [56]; without expression of self MHC on target cells, the inhibitory signal to NK cells is abolished and lysis ensues [57]. A more recent paper found that recombinant human (rh)IL-10 treatment in immunized mice after tumor challenge significantly enhanced antitumor immunity and vaccine efficacy. Three weeks after IL-10 administration, splenic CD8+CD44hiCD122+ (activated memory) T cells had increased and antigen-specific in vitro proliferation was enhanced. Additionally, antigen-specific IFNγ production was increased in animals challenged with tumor and then treated with IL-10 when compared to animals only given tumor challenge. Interestingly, the effect of IL-10 on CTL function could be enhanced by CD4+ T-cell depletion. This suggests that IL-10 may have opposing effects on CD8+ and CD4+ T cells in a tumor model [58] or that the stimulatory effect of IL-10 may be more pronounced in the absence of CD4+ Tregs.

IL-10 may also target myeloid-derived suppressor cells (MDSCs) and support tumor immunity. We have recently observed that chemically induced tumors, transplanted tumor growth, and metastasis are increased in IL-10-deficient mice. Ablation of endogenous IL-10 increased the expression of MHC on APCs and promoted the development of MDSCs and Tregs in tumor-bearing hosts; this may explain the surprising phenotype of IL-10-deficient mice [59]. It is of note, then, to recognize that the functions of IL-10 are complex and context-dependent, and in a given system, we may only see the net outcome of multiple effects.

IL-10 and “non-immune effects”

IL-10 has recently been shown to protect neurons from toxin-induced damage. Activation of the IL-10R in spinal cord neurons allowed Il-10 to signal through JAK/STAT and PI3-K pathways, upregulating the antiapoptotic proteins Bcl-2 and Bcl-xL, blocking cytochrome C release and cleavage of caspases. These effects were observed using the in vitro model of glutamine-induced apoptosis [60]. IL-10 is also involved in the process of autophagy, a mechanism whereby cells maintain homeostasis. Starvation and exposure to certain stimuli can induce autophagy or “self-eating” of excess macromolecules or organelles in the cytosol. Park and colleagues recently investigated the effects of IL-10 treatment on starved murine macrophages and demonstrated that IL-10 and IL-10 receptor signaling through the PI3 K pathway inhibited the in vitro induction of autophagy [61]. Further studies are required to determine whether this is also the case in vivo. Studies in our laboratory have demonstrated that IL-10 can suppress tumor angiogenesis through its control of the pro-angiogenic factor IL-1 [59]. However, there is evidence that IL-10 may be pro-angiogenic [62]. In a model of ischemia-induced pathological angiogenesis in mice, IL-10 deficiency resulted in reduced retinal blood vessel formation. Macrophages from IL-10−/− mice expressed decreased levels of VEGF and NO in comparison with those from wild-type mice [62]. Therefore, IL-10 can directly and indirectly mediate non-immune effects.

IFNγ

IFNγ, originally termed “macrophage activating factor,” was first described (along with IFN-α and IFN-β) as a mediator that interfered with viral replication [63, 64]. IFNγ is produced primarily by NK cells, CD4+ and CD8+ T cells, and NKT cells [65, 66]. In many of these populations, IL-12 and IL-18 can induce or further increase the production of IFNγ [65, 6770]. IFNγ plays key roles in host defense (including antiviral and antibacterial defense) and antitumor immunity. Notably, there are different interpretations of how IFNγ contributes to antitumor effects. While some scientists believe that antigen-specific T cells control tumors via secretion of IFNγ [71], others argue that it is rather an innate immune response involved in the control of carcinogen-induced tumors [72]. In contrast to its immune stimulatory role, IFNγ is also capable of downregulating immune responses; in this way, it can minimize immune-mediated tissue and organ damage in the context of autoimmunity and infectious immunity. However, the immune regulatory roles of IFNγ may result in immune evasion and hamper antitumor immunity in the context of malignancy (Table 2).

Table 2.

Interferon-γ

Effect Mechanism Context References
Immune inhibitory effects
Decreases effector T-cell generation and function Stimulates B7-H1 expression on APC, endothelial and epithelial cells Cancer [15, 86, 121124]
Limits T-cell activation and function Induces non-cognate MHC class I on tumor cells Cancer [150]
Induces T-cell dysfunction Induces IDO expression in APC and other cell types Cancer [125132]
Supports immunosuppressive functions of tumor-associated myeloid cells May induce arginase expression in these populations Cancer [134138, 141145]
Increases tumor evasion of CTL lysis Increases expression of non-classical MHC and decreases antigen expression on tumor cells Cancer [150152]
Prolongs survival and immune evasion of melanoma cells IFNγ+ macrophages invade irradiated skin, genes induced in melanocytes by IFNγ include CTLA-4 and non-classical MHC Cancer [153155]
Undermines secondary antitumor CD8+ T response Mediates contraction of CD4+ T-cell population via apoptosis Cancer [154]
Non-immune effects
Tumor suppressor functions, minimizes subsequent sensitivity to IFNg, increasing otential for metastasis Signaling induces expression of SOCS1 Cancer [147, 148]
Inhibits vascularization IFNγ stimulates IP-10 and MIG, angiogenesis inhibitors Cancer, organ transplantation [165168]
Regulates HSC repopulation IFNγ secretion by immune and other cells Homeostasis, infection [96]
Immune stimulatory effects
Decreases susceptibility to infection Maintenance of TNFα signaling, Ag-specific IgG2a responses, Ag-specific proliferation Bacterial and viral infection [9095]
Improves resistance to chemically induced and spontaneous tumors IFNγ signaling in tumor cells increases immunogenicity Cancer [98101]
Activates antitumor potential of macrophages Stimulates IL-12 secretion and tumor cell killing by macrophage Cancer [104106]
Supports proliferation, activation, lytic capability, and production of effector cytokines in NK and NKT cells IFNγ production by activated NKT cells Cancer [107, 108]
Recruits CTLs to tumor environment Polarizes Th0 to Th1, which recruit CTL to tumor Cancer [113]
Blocks Treg expansion Causes cell cycle arrest in Tregs Cancer [116]
Mediates T cell and NK cell tumor infiltration Stimulates productions of chemokines MIG and IP-10 Cancer [113, 117, 118]
Decreases tumor growth, promotes antitumor immunity Mediates angiostasis, maintains tumor cell immunogenicity (dying tumor cells release TAAs) Cancer [98, 162, 165169]
Promotes Th17 cell expansion and function Induces IL-1 and IL-23 Autoimmune [86]

IFNγ and immunostimulation

It is well known that IFNγ stimulates MHC and associated chaperone molecule expression on target cells [7376]. IFNγ induces the replacement of proteasome subunits by “immunoproteasome” subunits, thereby increasing the quantity and diversity of peptides presented on MHC I complexes to CD8+ T cells [77]. IFNγ activates the lysosome and increases antibacterial activity in macrophages [78]. It serves as a chemoattractant and repellant for lymphocytes [79], controls isotype switching [80] and antibody production in B cells, and directs the growth and differentiation of several cell types [8183]. IFNγ is a well-known product of Th1 cells and plays a key role in skewing of naïve T cells to a Th1 phenotype, both through inhibition of IL-4 production [84] and induction of IL-12 secretion [85]. Interestingly, although IFNγ may suppress Th17 cell development from naïve T cells in mice, IFNγ can strongly stimulate the expression of IL-1 and IL-23 by myeloid APCs and promote memory Th17 cell expansion in humans [86]. This may explain why Th1 and Th17 cells coexist in the microenvironments of chronic inflammation and cancer [87]. This phenomenon is functionally linked to the development of human autoimmune pathologies, including psoriasis and chronic bowel diseases [88, 89].

IFNγ is, perhaps, best known for its antiviral and antibacterial effects in infected hosts. In 1993, it was observed that IFNγ receptor-deficient mice had an increased susceptibility to Listeria monocytogenes and vaccinia virus infection [90]. It is clear that IFNγ plays a vital role in viral control of infected hosts [91] and in the establishment of a long-term antiviral state [92]. Perhaps, not surprisingly, patients with IFNγ signaling deficiency are more susceptible to infection [9395]. This increased sensitivity is associated with disrupted TNFα production, which would normally result from intact IFNγ signaling. Furthermore, IFNγ signaling is required to mobilize hematopoietic stem cells (HSCs) to proliferate and repopulate immune populations in mice with long-term systemic Mycobacterium avium infection. IFNγ-deficient HSCs proliferate more slowly. This indicates that homeostatic levels of IFNγ regulate HSC activity and in turn affect immune cell repopulation and immune responses [96].

There is an increasing body of evidence demonstrating varied roles of IFNγ in antitumor immunity, reviewed in [97] and elsewhere. Early studies showed that progressively growing Meth A fibrosarcoma cells were rejected in lipopolysaccharide-treated mice and that this rejection was abrogated following inhibition of IFNγ signaling [98]. Subsequently, it was found that IFNγ-deficient mice and IFNγ receptor-deficient mice were more susceptible than their wild-type counterparts to methylcholanthrene (MCA)-induced tumor formation [99, 100]. It has been suggested that the cooperation between IFNγ and lymphocytes is crucial for host protection from carcinogen-induced and spontaneous tumorigenesis [101]. However, there is controversy regarding the contribution of lymphocytes and T-cell-derived IFNγ to tumor initiation and immunosurveillance [102, 103]. Nonetheless, IFNγ is a powerful, multi-effector cytokine, derived from innate immune cells and T cells, that plays different roles in the progressive stages of tumor development.

Of course, many contributions of IFNγ to antitumor immunity are due to its direct effects on components of innate and adaptive immunity. As it was originally described, IFNγ has potent macrophage-activating capabilities; it can induce macrophage killing of multiple tumor cell lines [104] and production of IL-12 by macrophages exposed to bacterial components [105, 106]. Interestingly, NK and NKT cells serve as both sources and targets of IFNγ during the antitumor response. Both IFNγ and IL-12 stimulate the proliferation, lytic capability, and production of effector cytokines in NK and NKT cells. IFNγ secreted from activated NKT cells led to NK cell activation in vivo [107, 108]. In further support of this, blockade of IFNγ signaling in RM-1-bearing mice led to enhanced tumor metastasis, while administration of the same antibody to mice deficient in IL-12p40 or NKT cells had no significant effect on the same [100]. Several reports have documented marked effects of IFNγ on components of the adaptive immune system, including CD8+ T cells [101, 109, 110]. IFNγ also has a key role in promoting the Th1 polarization of CD4+ T helper cells [111, 112], which are crucial in recruiting CTLs to the tumor microenvironment [113]. Early experiments had shown that Stat1−/− mice could not (in contrast to wild-type mice) reject the growth of a P815 mastocytoma subclone or control the development of another subclone engineered to express IL-12 [114]. Stat1−/− T cells exposed to IL-12 produced only half as much IFNγ as wild-type T cells and could not lyse tumor cells. Importantly, Stat6−/− mice could spontaneously reject the same poorly immunogenic subclone of P815, while wild-type mice could not. This rejection was associated with an increased T-cell cytotoxicity and IFNγ production. Because Stat6−/− mice preferentially polarize T cells toward a Th1 phenotype, these data strongly suggest that IFNγ signaling is crucial in determining the Th1 versus Th2 helper T-cell balance and subsequently the efficacy of the in vivo antitumor immune response [115]. The Ley laboratory more recently documented another antitumor function for IFNγ. Their 2009 report showed that IL-12 treatment blocked in vitro Treg expansion in an IL-12 receptor-dependent fashion. IL-12 stimulated IFNγ-mediated inhibition of Treg cell proliferation and decreased IL-2 production. In these experiments, IFNγ signaling caused cell cycle arrest in Treg cells. The data suggest that this IFNγ-dependent mechanism could counteract the ability of Tregs to protect tumors in cancer patients [116]. In addition to its direct effects on immune activation, IFNγ stimulates the production of chemokines, including IP-10 and MIG. IP-10 and MIG are powerful T and NK cell chemoattractants [117, 118], mediate T cell and NK cell tumor infiltration, and indirectly enhance antitumor immunity [113].

IFNγ and immunoregulation

At the same time that IFNγ was being touted as a promising antitumor agent, evidence began to accumulate in support of regulatory roles of IFNγ. IFNγ appears capable of driving several cellular and molecular mechanisms that may underlie tumor initiation and immunosuppression (Table 2). As the clearest example of the duality of IFNγ occurs in malignancy, we review the regulatory mechanisms and roles of IFNγ in the context of tumor.

Effects of IFNγ on B7-H1 (PD-L1)

We have studied ovarian cancer and ovarian cancer-associated DCs and found that both of these cell types express the inhibitory molecule B7-H1 [15, 119]. IFNγ was the most important stimulus of B7-H1 expression on human APC, epithelial cells, and vascular endothelial cells [15, 86]. It is generally assumed that IFNγ is predominantly produced by the Th1 cell subset. However, we observed that Th17 cells also express high levels of IFNγ [113]. It is possible that IFNγ derived from Th1 cells, Th17 cells, and other populations stimulate B7-H1 expression in the tumor microenvironment, thereby hampering antitumor immunity. We then examined the hepatocellular carcinoma (HCC) environment to determine whether this could be verified. We found an increased expression of the inhibitory molecule B7-H1 on Kupffer cells (KC) within the tumor in comparison with the surrounding tissue and an increased PD-1 expression on CD8+ T cells within the tumor microenvironment [120]. Interestingly, B7-H1+ KC and CD8+ T cells were co-localized in HCC stroma. PD-1+CD8+ T cells had decreased proliferative ability and effector function when compared with PD-1 T cells, but B7-H1 neutralization recovered effector T-cell function. These data indicate that B7-H1/PD-1 interactions contribute to immune suppression in human HCC, and it is likely that IFNγ signaling contributes to B7-H1 expression in this setting [121, 122]. A 2007 report showed that B7-H1 expression in CD138+ multiple myeloma cells was induced or augmented by IFNγ and that stimulation by IFNγ inhibited the generation of CTLs. Interestingly, this expression could be reduced via interruption of MyD88, TRAF6, or STAT1 signaling [123]. An increased B7-H1 expression has also been found on cells of high-risk patients with myelodysplastic syndromes (MDS), which are hematologic stem cell disorders characterized by cytopenia, excessive hematopoetic cell apoptosis, and a high risk of progression to acute myeloid leukemia. IFNγ synergized with TNFα to induce B7-H1 expression on MDS blasts, and these blasts proliferated better than B7-H1 blasts. It should be noted that T cells from MDS patients also expressed an increased PD-1. This suggests that the B7-H1 inhibitory signal can be accurately propagated in such patients, where the interaction of these two molecules may contribute to disease progression [124].

Effects of IFNγ on indoleamine-2,3-dioxygenase (IDO)

IDO function has been well studied and defined in the context of tumors [125129]. IDO is expressed in several cell types in the tumor microenvironment, where it minimizes effector T-cell access to tryptophan, an essential amino acid. IDO-expressing DCs can induce Treg-mediated expression of B7-H1 on target DCs and effectively downregulate the T-cell arm of the antitumor response [130]. Interestingly, IFNγ strongly induces IDO production, and both DC- and Treg-derived IFNγ can induce IDO [131] and mediate T-cell dysfunction [128, 132].

Effects of IFNγ on arginase

Arginase, an enzyme, metabolizes the amino acid arginine. Environmental arginine plays a central role in the proliferation and expression of the CD3ζ chain in T cells and is crucial in cytokine production [133, 134]. Some studies have revealed a role of IFNγ in the induction of arginase expression [135138], while others have shown that IFNγ inhibits arginase [139, 140]. While arginase induction by IFNγ may, in fact, be context-dependent, it is important to point out that this enzyme is centrally involved in tumor immunology, where it serves to support the suppressive functions of tumor-associated macrophages [134], DCs [141], and MDSCs [142145]. Perhaps, it is not surprising that arginase expression in MDSCs can also play a beneficial role in immune modulation; MDSC are capable of mediating the suppression of graft-versus-host disease [146].

IFNγ and SOCS1

One of the most important regulatory roles of IFNγ is through its relationship with suppressor of cytokine signaling-1 (SOCS1). SOCS1 expression is induced by a number of cytokines, including IFNγ. Exposure to IFNγ strongly upregulates SOCS1, which renders cells refractory to further cytokine signaling through its ability to inhibit all the janus kinases (JAKs). SOCS1 acts as a tumor suppressor via various modes of oncogene inhibition, including mediation of the ubiquitination and subsequent degradation of potential cellular oncogenes like Vav1, a guanine nucleotide exchange factor [147]. More recent work has demonstrated that SOCS1 can bind to tyrosine 441 (Y441) of the IFNγ receptor subunit 1(IFNγR1), where it mediates some of its negative effects on IFNγ signal transduction. Mice with mutated Y441 are rendered hypersensitive to IFNγ signaling, displaying greater resistance to lung metastases and a more significant antitumor effect when treated with alpha-galactosylceramide (αGalCer) [148]. These mice also had increased MIG serum levels, which may also have contributed to their antitumor phenotype. Via specific modulation of IFNγ signaling, this study both demonstrates one of the traditional, immunostimulatory roles of the cytokine in tumor biology and offers new insight into the control of IFNγ signaling by SOCS1.

Effects of IFNγ on non-classical MHC, antigen presentation, and APCs

An early study showed that IFNγ-treated tumor cells are more aggressive in vivo in mouse models [149]. Recent studies have demonstrated that this is due to the loss of tumor antigen expression and the induction of non-classical MHC expression, which enable tumor cells to evade CTL-mediated lysis [150152]. Furthermore, a recent study has shown that UVB irradiation induces an IFNγ signature in murine melanocytes—a signature that includes multiple genes (like non-classical MHC class Ib antigens) known to participate in immune evasion. The source of IFNγ in this study was a population of pro-tumorigenic macrophages infiltrating irradiated skin; IFNγ+ macrophages have also been found in a majority of human melanoma cases [153]. These tumor-associated macrophages and myeloid cells function as APCs, mediating immune suppression and prolonging the survival of melanoma cells.

Effects of IFNγ on T cells

The studies described above demonstrate the immunoregulatory roles of IFNγ on innate immune components including APCs. While IFNγ is broadly known as a cytokine that supports T-cell immunity in the context of malignancy, there is evidence that this cytokine can also undermine the antitumor response by negatively affecting CD4+ T cells. In 2007, the laboratory of William Murphy reported that after various immunotherapies, IFNγ mediated contraction of the CD4+ T-cell population via the induction of apoptosis [154]. Because CD4+ T cells are essential for the most effective secondary antitumor CD8+ T-cell response, this intriguing observation should be considered in the context of both patient therapy and clinical trial design.

These immune regulatory roles are consistent with several studies in patients with cancer. Intratumoral expression of IFNγ was shown to be associated with a more aggressive phenotype in human melanomas [155]. Most clinical trials of IFNγ treatment did not demonstrate clinical efficacy in patients with melanoma [156159]. Furthermore, multiple clinical trials have demonstrated that IFNγ-treated patients fare worse than untreated patients [160, 161]. Therefore, it is reasonable to point out that the “dark side” of IFNγ action is operative in patients with cancer (Table 2).

IFNγ and “non-immune effects”

Some functions of IFNγ occur outside of the immune system—these include the arrest of cell growth, induction of apoptosis, and inhibition of angiogenesis. Many of these functions target tumor cell lines: cells made unresponsive to IFNγ via genetic manipulation grew more rapidly in vitro and in vivo [98, 162]. This was partially due to a reduction in tumor cell immunogenicity, but also to a decrease in IFNγ-induced angiostasis within the tumor microenvironment. IFNγ has been shown to inhibit proliferation of both human fibrosarcomas and mouse fibroblasts [163, 164], but had no effect on human or murine cells deficient in the Stat1 component of IFNγ signaling pathway. IP-10, the first angiostatic chemokine identified as a product of IFNγ signaling, is a powerful T-cell chemoattractant [117, 118] that works in combination with lymphocytes and other IFNγ-induced chemokines to inhibit tumor vasculogenesis and effect transplanted tumor rejection [165168]. It is interesting to postulate that a subset of progressive tumors develops insensitivity to IFNγ: Certain human tumor cell lines are less sensitive or completely non-reactive toward this cytokine [99]. Quite recently, a report by Briesemeister and colleagues established that tumor initiation can be prevented by local production of IFNγ, but that vascularized tumors are harder to reject the longer they are established. This finding was explained by the reduction in endothelial cells mediated by IFNγ in the local environment and the resulting necrosis of the tumor in question [169]. Therefore, IFNγ- and IFNγ-induced type-1 chemokine IP-10 can directly inhibit tumor angiogenesis and result in tumor cell death. This effect would subsequently promote antitumor immunity due to the increased TAA release from dying tumor cells.

Concluding remarks

In summary, it is important to realize the pluripotency of certain cytokines and refrain from categorizing them according to their main or “classical” function. Because prototypical cytokines involved in inflammation may, in other environments, serve to downregulate immune components, and signature immunosuppressive proteins may also activate certain facets of immunity, it is important to consider specific environmental and chronological contexts. For example, IL-10 is predominantly immunosuppressive in many disease models, particularly infectious disease, where APCs serve as its principal targets [170]. Toll-like receptors are activated, and antigen-specific T-cell priming is induced. In a tumor-bearing host, however, when there is no obvious acute phase of immune response (tumor development is chronic) [127], TAA-specific T cells may slowly be primed, activated, and expanded, along with the accumulation of immune suppressive cell populations, and IL-10 may be gradually induced, suppressing inflammatory cytokines and MDSC accumulation. In this context, IL-10 may promote the antitumor immune response. Although IFNγ is a potent immune effector cytokine, it also induces B7-H1, IDO, arginase, and non-classical MHC expression, which may participate in a feedback mechanism to efficiently downregulate the immune response and avoid tissue and organ damage. In this way, it is easier to imagine such contrasting effects of a particular protein from one disease to another and from the initial stages to the endgame of a single malady. An appreciation for the varied capabilities of individual cytokines will no doubt aid in a more holistic understanding of several human illnesses and perhaps contribute to chronological changes in some cytokine-specific clinical therapies.

Acknowledgments

This work was supported in part by research grants from the NIH/NCI R01 grants and the Ovarian Cancer Research Foundation (WZ) and the NIH through the University of Michigan’s Cancer Center Support Grant (P30CA46592).

Conflict of interest

The authors declare that they have no conflict of interest.

References

  • 1.Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med. 1989;170(6):2081–2095. doi: 10.1084/jem.170.6.2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.O’Garra A, Stapleton G, Dhar V, Pearce M, Schumacher J, Rugo H, Barbis D, Stall A, Cupp J, Moore K, et al. Production of cytokines by mouse B cells: B lymphomas and normal B cells produce interleukin 10. Int Immunol. 1990;2(9):821–832. doi: 10.1093/intimm/2.9.821. [DOI] [PubMed] [Google Scholar]
  • 3.Galli SJ, Kalesnikoff J, Grimbaldeston MA, Piliponsky AM, Williams CM, Tsai M. Mast cells as “tunable” effector and immunoregulatory cells: recent advances. Annu Rev Immunol. 2005;23:749–786. doi: 10.1146/annurev.immunol.21.120601.141025. [DOI] [PubMed] [Google Scholar]
  • 4.Grimbaldeston MA, Nakae S, Kalesnikoff J, Tsai M, Galli SJ. Mast cell-derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B. Nat Immunol. 2007;8(10):1095–1104. doi: 10.1038/ni1503. [DOI] [PubMed] [Google Scholar]
  • 5.Wei S, Kryczek I, Zou L, Daniel B, Cheng P, Mottram P, Curiel T, Lange A, Zou W. Plasmacytoid dendritic cells induce CD8 + regulatory T cells in human ovarian carcinoma. Cancer Res. 2005;65(12):5020–5026. doi: 10.1158/0008-5472.CAN-04-4043. [DOI] [PubMed] [Google Scholar]
  • 6.Asseman C, Mauze S, Leach MW, Coffman RL, Powrie F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J Exp Med. 1999;190(7):995–1004. doi: 10.1084/jem.190.7.995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 2007;8(12):1390–1397. doi: 10.1038/ni1539. [DOI] [PubMed] [Google Scholar]
  • 8.Jonuleit H, Schmitt E, Schuler G, Knop J, Enk AH. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med. 2000;192(9):1213–1222. doi: 10.1084/jem.192.9.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Khoo UY, Proctor IE, Macpherson AJ. CD4 + T cell down-regulation in human intestinal mucosa: evidence for intestinal tolerance to luminal bacterial antigens. J Immunol. 1997;158(8):3626–3634. [PubMed] [Google Scholar]
  • 10.Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008;180(9):5771–5777. doi: 10.4049/jimmunol.180.9.5771. [DOI] [PubMed] [Google Scholar]
  • 11.Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683–765. doi: 10.1146/annurev.immunol.19.1.683. [DOI] [PubMed] [Google Scholar]
  • 12.Mosser DM, Zhang X. Interleukin-10: new perspectives on an old cytokine. Immunol Rev. 2008;226:205–218. doi: 10.1111/j.1600-065X.2008.00706.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.de Waal Malefyt R, Haanen J, Spits H, Roncarolo MG, te Velde A, Figdor C, Johnson K, Kastelein R, Yssel H, de Vries JE. Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class II major histocompatibility complex expression. J Exp Med. 1991;174(4):915–924. doi: 10.1084/jem.174.4.915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ding L, Linsley PS, Huang LY, Germain RN, Shevach EM. IL-10 inhibits macrophage costimulatory activity by selectively inhibiting the up-regulation of B7 expression. J Immunol. 1993;151(3):1224–1234. [PubMed] [Google Scholar]
  • 15.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT, Chen L, Zou W. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9(5):562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • 16.Schandene L, Alonso-Vega C, Willems F, Gerard C, Delvaux A, Velu T, Devos R, de Boer M, Goldman M. B7/CD28-dependent IL-5 production by human resting T cells is inhibited by IL-10. J Immunol. 1994;152(9):4368–4374. [PubMed] [Google Scholar]
  • 17.Joss A, Akdis M, Faith A, Blaser K, Akdis CA. IL-10 directly acts on T cells by specifically altering the CD28 co-stimulation pathway. Eur J Immunol. 2000;30(6):1683–1690. doi: 10.1002/1521-4141(200006)30:6<1683::AID-IMMU1683>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  • 18.Demangel C, Bertolino P, Britton WJ. Autocrine IL-10 impairs dendritic cell (DC)-derived immune responses to mycobacterial infection by suppressing DC trafficking to draining lymph nodes and local IL-12 production. Eur J Immunol. 2002;32(4):994–1002. doi: 10.1002/1521-4141(200204)32:4<994::AID-IMMU994>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 19.Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307. doi: 10.1038/nri1806. [DOI] [PubMed] [Google Scholar]
  • 20.Murai M, Turovskaya O, Kim G, Madan R, Karp CL, Cheroutre H, Kronenberg M. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol. 2009;10(11):1178–1184. doi: 10.1038/ni.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75(2):263–274. doi: 10.1016/0092-8674(93)80068-P. [DOI] [PubMed] [Google Scholar]
  • 22.Davidson NJ, Leach MW, Fort MM, Thompson-Snipes L, Kuhn R, Muller W, Berg DJ, Rennick DM. T helper cell 1-type CD4 + T cells, but not B cells, mediate colitis in interleukin 10-deficient mice. J Exp Med. 1996;184(1):241–251. doi: 10.1084/jem.184.1.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhou P, Streutker C, Borojevic R, Wang Y, Croitoru K. IL-10 modulates intestinal damage and epithelial cell apoptosis in T cell-mediated enteropathy. Am J Physiol Gastrointest Liver Physiol. 2004;287(3):G599–G604. doi: 10.1152/ajpgi.00063.2004. [DOI] [PubMed] [Google Scholar]
  • 24.Jarry A, Bossard C, Bou-Hanna C, Masson D, Espaze E, Denis MG, Laboisse CL. Mucosal IL-10 and TGF-beta play crucial roles in preventing LPS-driven, IFN-gamma-mediated epithelial damage in human colon explants. J Clin Invest. 2008;118(3):1132–1142. doi: 10.1172/JCI32140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hagenbaugh A, Sharma S, Dubinett SM, Wei SH, Aranda R, Cheroutre H, Fowell DJ, Binder S, Tsao B, Locksley RM, Moore KW, Kronenberg M. Altered immune responses in interleukin 10 transgenic mice. J Exp Med. 1997;185(12):2101–2110. doi: 10.1084/jem.185.12.2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Peters N, Sacks D. Immune privilege in sites of chronic infection: Leishmania and regulatory T cells. Immunol Rev. 2006;213:159–179. doi: 10.1111/j.1600-065X.2006.00432.x. [DOI] [PubMed] [Google Scholar]
  • 27.van der Sluijs KF, van Elden LJ, Nijhuis M, Schuurman R, Pater JM, Florquin S, Goldman M, Jansen HM, Lutter R, van der Poll T. IL-10 is an important mediator of the enhanced susceptibility to pneumococcal pneumonia after influenza infection. J Immunol. 2004;172(12):7603–7609. doi: 10.4049/jimmunol.172.12.7603. [DOI] [PubMed] [Google Scholar]
  • 28.Jimenez Mdel P, Walls L, Fierer J. High levels of interleukin-10 impair resistance to pulmonary coccidioidomycosis in mice in part through control of nitric oxide synthase 2 expression. Infect Immun. 2006;74(6):3387–3395. doi: 10.1128/IAI.01985-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fierer J, Walls L, Eckmann L, Yamamoto T, Kirkland TN. Importance of interleukin-10 in genetic susceptibility of mice to Coccidioides immitis. Infect Immun. 1998;66(9):4397–4402. doi: 10.1128/iai.66.9.4397-4402.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kelly JP, Bancroft GJ. Administration of interleukin-10 abolishes innate resistance to Listeria monocytogenes. Eur J Immunol. 1996;26(2):356–364. doi: 10.1002/eji.1830260214. [DOI] [PubMed] [Google Scholar]
  • 31.Kim BG, Joo HG, Chung IS, Chung HY, Woo HJ, Yun YS. Inhibition of interleukin-10 (IL-10) production from MOPC 315 tumor cells by IL-10 antisense oligodeoxynucleotides enhances cell-mediated immune responses. Cancer Immunol Immunother. 2000;49(8):433–440. doi: 10.1007/s002620000123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Matar P, Rozados VR, Gervasoni SI, Scharovsky OG. Down regulation of T-cell-derived IL-10 production by low-dose cyclophosphamide treatment in tumor-bearing rats restores in vitro normal lymphoproliferative response. Int Immunopharmacol. 2001;1(2):307–319. doi: 10.1016/S1567-5769(00)00028-X. [DOI] [PubMed] [Google Scholar]
  • 33.Vicari AP, Chiodoni C, Vaure C, Ait-Yahia S, Dercamp C, Matsos F, Reynard O, Taverne C, Merle P, Colombo MP, O’Garra A, Trinchieri G, Caux C. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti-interleukin 10 receptor antibody. J Exp Med. 2002;196(4):541–549. doi: 10.1084/jem.20020732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Enk AH, Jonuleit H, Saloga J, Knop J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma. Int J Cancer. 1997;73(3):309–316. doi: 10.1002/(SICI)1097-0215(19971104)73:3<309::AID-IJC1>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
  • 35.Loercher AE, Nash MA, Kavanagh JJ, Platsoucas CD, Freedman RS. Identification of an IL-10-producing HLA-DR-negative monocyte subset in the malignant ascites of patients with ovarian carcinoma that inhibits cytokine protein expression and proliferation of autologous T cells. J Immunol. 1999;163(11):6251–6260. [PubMed] [Google Scholar]
  • 36.De Santo C, Arscott R, Booth S, Karydis I, Jones M, Asher R, Salio M, Middleton M, Cerundolo V. Invariant NKT cells modulate the suppressive activity of IL-10-secreting neutrophils differentiated with serum amyloid A. Nat Immunol. 2010;11(11):1039–1046. doi: 10.1038/ni.1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Seo N, Hayakawa S, Tokura Y. Mechanisms of immune privilege for tumor cells by regulatory cytokines produced by innate and acquired immune cells. Semin Cancer Biol. 2002;12(4):291–300. doi: 10.1016/S1044-579X(02)00015-9. [DOI] [PubMed] [Google Scholar]
  • 38.Kurte M, Lopez M, Aguirre A, Escobar A, Aguillon JC, Charo J, Larsen CG, Kiessling R, Salazar-Onfray F. A synthetic peptide homologous to functional domain of human IL-10 down-regulates expression of MHC class I and transporter associated with antigen processing 1/2 in human melanoma cells. J Immunol. 2004;173(3):1731–1737. doi: 10.4049/jimmunol.173.3.1731. [DOI] [PubMed] [Google Scholar]
  • 39.Kim J, Modlin RL, Moy RL, Dubinett SM, McHugh T, Nickoloff BJ, Uyemura K. IL-10 production in cutaneous basal and squamous cell carcinomas. A mechanism for evading the local T cell immune response. J Immunol. 1995;155(4):2240–2247. [PubMed] [Google Scholar]
  • 40.Steinbrink K, Jonuleit H, Muller G, Schuler G, Knop J, Enk AH. Interleukin-10-treated human dendritic cells induce a melanoma-antigen-specific anergy in CD8(+) T cells resulting in a failure to lyse tumor cells. Blood. 1999;93(5):1634–1642. [PubMed] [Google Scholar]
  • 41.Urosevic M, Dummer R. HLA-G and IL-10 expression in human cancer–different stories with the same message. Semin Cancer Biol. 2003;13(5):337–342. doi: 10.1016/S1044-579X(03)00024-5. [DOI] [PubMed] [Google Scholar]
  • 42.Jovasevic VM, Gorelik L, Bluestone JA, Mokyr MB. Importance of IL-10 for CTLA-4-mediated inhibition of tumor-eradicating immunity. J Immunol. 2004;172(3):1449–1454. doi: 10.4049/jimmunol.172.3.1449. [DOI] [PubMed] [Google Scholar]
  • 43.Huang M, Stolina M, Sharma S, Mao JT, Zhu L, Miller PW, Wollman J, Herschman H, Dubinett SM. Non-small cell lung cancer cyclooxygenase-2-dependent regulation of cytokine balance in lymphocytes and macrophages: up-regulation of interleukin 10 and down-regulation of interleukin 12 production. Cancer Res. 1998;58(6):1208–1216. [PubMed] [Google Scholar]
  • 44.Stolina M, Sharma S, Lin Y, Dohadwala M, Gardner B, Luo J, Zhu L, Kronenberg M, Miller PW, Portanova J, Lee JC, Dubinett SM. Specific inhibition of cyclooxygenase 2 restores antitumor reactivity by altering the balance of IL-10 and IL-12 synthesis. J Immunol. 2000;164(1):361–370. doi: 10.4049/jimmunol.164.1.361. [DOI] [PubMed] [Google Scholar]
  • 45.Sharma S, Stolina M, Yang SC, Baratelli F, Lin JF, Atianzar K, Luo J, Zhu L, Lin Y, Huang M, Dohadwala M, Batra RK, Dubinett SM. Tumor cyclooxygenase 2-dependent suppression of dendritic cell function. Clin Cancer Res. 2003;9(3):961–968. [PubMed] [Google Scholar]
  • 46.Go NF, Castle BE, Barrett R, Kastelein R, Dang W, Mosmann TR, Moore KW, Howard M. Interleukin 10, a novel B cell stimulatory factor: unresponsiveness of X chromosome-linked immunodeficiency B cells. J Exp Med. 1990;172(6):1625–1631. doi: 10.1084/jem.172.6.1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rousset F, Garcia E, Defrance T, Peronne C, Vezzio N, Hsu DH, Kastelein R, Moore KW, Banchereau J. Interleukin 10 is a potent growth and differentiation factor for activated human B lymphocytes. Proc Natl Acad Sci USA. 1992;89(5):1890–1893. doi: 10.1073/pnas.89.5.1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Llorente L, Zou W, Levy Y, Richaud-Patin Y, Wijdenes J, Alcocer-Varela J, Morel-Fourrier B, Brouet JC, Alarcon-Segovia D, Galanaud P, Emilie D. Role of interleukin 10 in the B lymphocyte hyperactivity and autoantibody production of human systemic lupus erythematosus. J Exp Med. 1995;181(3):839–844. doi: 10.1084/jem.181.3.839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Levy Y, Brouet JC. Interleukin-10 prevents spontaneous death of germinal center B cells by induction of the bcl-2 protein. J Clin Invest. 1994;93(1):424–428. doi: 10.1172/JCI116977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liu YJ, Mason DY, Johnson GD, Abbot S, Gregory CD, Hardie DL, Gordon J, MacLennan IC. Germinal center cells express bcl-2 protein after activation by signals which prevent their entry into apoptosis. Eur J Immunol. 1991;21(8):1905–1910. doi: 10.1002/eji.1830210819. [DOI] [PubMed] [Google Scholar]
  • 51.MacNeil IA, Suda T, Moore KW, Mosmann TR, Zlotnik A. IL-10, a novel growth cofactor for mature and immature T cells. J Immunol. 1990;145(12):4167–4173. [PubMed] [Google Scholar]
  • 52.Chen WF, Zlotnik A. IL-10: a novel cytotoxic T cell differentiation factor. J Immunol. 1991;147(2):528–534. [PubMed] [Google Scholar]
  • 53.Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol. 2005;78(5):1043–1051. doi: 10.1189/jlb.0705358. [DOI] [PubMed] [Google Scholar]
  • 54.Suzuki T, Tahara H, Narula S, Moore KW, Robbins PD, Lotze MT. Viral interleukin 10 (IL-10), the human herpes virus 4 cellular IL-10 homologue, induces local anergy to allogeneic and syngeneic tumors. J Exp Med. 1995;182(2):477–486. doi: 10.1084/jem.182.2.477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zheng LM, Ojcius DM, Garaud F, Roth C, Maxwell E, Li Z, Rong H, Chen J, Wang XY, Catino JJ, King I. Interleukin-10 inhibits tumor metastasis through an NK cell-dependent mechanism. J Exp Med. 1996;184(2):579–584. doi: 10.1084/jem.184.2.579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kundu N, Fulton AM. Interleukin-10 inhibits tumor metastasis, downregulates MHC class I, and enhances NK lysis. Cell Immunol. 1997;180(1):55–61. doi: 10.1006/cimm.1997.1176. [DOI] [PubMed] [Google Scholar]
  • 57.Karre K. Natural killer cells and the MHC class I pathway of peptide presentation. Semin Immunol. 1993;5(2):127–145. doi: 10.1006/smim.1993.1016. [DOI] [PubMed] [Google Scholar]
  • 58.Fujii S, Shimizu K, Shimizu T, Lotze MT. Interleukin-10 promotes the maintenance of antitumor CD8(+) T-cell effector function in situ. Blood. 2001;98(7):2143–2151. doi: 10.1182/blood.V98.7.2143. [DOI] [PubMed] [Google Scholar]
  • 59.Tanikawa T, Wilke CM, Kryczek I, Chen GY, Kao JY, Zou W (2011) Ablation of interleukin (IL)-10 promotes tumor development, growth and metastasis (in press) [DOI] [PMC free article] [PubMed]
  • 60.Zhou Z, Peng X, Insolera R, Fink DJ, Mata M. Interleukin-10 provides direct trophic support to neurons. J Neurochem. 2009;110(5):1617–1627. doi: 10.1111/j.1471-4159.2009.06263.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Park HJ, Lee SJ, Kim SH, Han J, Bae J, Kim SJ, Park CG, Chun T. IL-10 inhibits the starvation induced autophagy in macrophages via class I phosphatidylinositol 3-kinase (PI3 K) pathway. Mol Immunol. 2011;48(4):720–727. doi: 10.1016/j.molimm.2010.10.020. [DOI] [PubMed] [Google Scholar]
  • 62.Dace DS, Khan AA, Kelly J, Apte RS. Interleukin-10 promotes pathological angiogenesis by regulating macrophage response to hypoxia during development. PLoS One. 2008;3(10):e3381. doi: 10.1371/journal.pone.0003381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147(927):258–267. doi: 10.1098/rspb.1957.0048. [DOI] [PubMed] [Google Scholar]
  • 64.Isaacs A, Lindenmann J, Valentine RC. Virus interference. II. Some properties of interferon. Proc R Soc Lond B Biol Sci. 1957;147(927):268–273. doi: 10.1098/rspb.1957.0049. [DOI] [PubMed] [Google Scholar]
  • 65.Yoshimoto T, Takeda K, Tanaka T, Ohkusu K, Kashiwamura S, Okamura H, Akira S, Nakanishi K. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-gamma production. J Immunol. 1998;161(7):3400–3407. [PubMed] [Google Scholar]
  • 66.Munder M, Mallo M, Eichmann K, Modolell M. Murine macrophages secrete interferon gamma upon combined stimulation with interleukin (IL)-12 and IL-18: a novel pathway of autocrine macrophage activation. J Exp Med. 1998;187(12):2103–2108. doi: 10.1084/jem.187.12.2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tominaga K, Yoshimoto T, Torigoe K, Kurimoto M, Matsui K, Hada T, Okamura H, Nakanishi K. IL-12 synergizes with IL-18 or IL-1beta for IFN-gamma production from human T cells. Int Immunol. 2000;12(2):151–160. doi: 10.1093/intimm/12.2.151. [DOI] [PubMed] [Google Scholar]
  • 68.Okamura H, Tsutsi H, Komatsu T, Yutsudo M, Hakura A, Tanimoto T, Torigoe K, Okura T, Nukada Y, Hattori K, et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature. 1995;378(6552):88–91. doi: 10.1038/378088a0. [DOI] [PubMed] [Google Scholar]
  • 69.Robinson D, Shibuya K, Mui A, Zonin F, Murphy E, Sana T, Hartley SB, Menon S, Kastelein R, Bazan F, O’Garra A. IGIF does not drive Th1 development but synergizes with IL-12 for interferon-gamma production and activates IRAK and NFkappaB. Immunity. 1997;7(4):571–581. doi: 10.1016/S1074-7613(00)80378-7. [DOI] [PubMed] [Google Scholar]
  • 70.Yoshimoto T, Okamura H, Tagawa YI, Iwakura Y, Nakanishi K. Interleukin 18 together with interleukin 12 inhibits IgE production by induction of interferon-gamma production from activated B cells. Proc Natl Acad Sci USA. 1997;94(8):3948–3953. doi: 10.1073/pnas.94.8.3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6(11):836–848. doi: 10.1038/nri1961. [DOI] [PubMed] [Google Scholar]
  • 72.Blankenstein T, Qin Z. Chemical carcinogens as foreign bodies and some pitfalls regarding cancer immune surveillance. Adv Cancer Res. 2003;90:179–207. doi: 10.1016/S0065-230X(03)90006-6. [DOI] [PubMed] [Google Scholar]
  • 73.Wallach D, Fellous M, Revel M. Preferential effect of gamma interferon on the synthesis of HLA antigens and their mRNAs in human cells. Nature. 1982;299(5886):833–836. doi: 10.1038/299833a0. [DOI] [PubMed] [Google Scholar]
  • 74.Johnson DR, Pober JS. Tumor necrosis factor and immune interferon synergistically increase transcription of HLA class I heavy- and light-chain genes in vascular endothelium. Proc Natl Acad Sci USA. 1990;87(13):5183–5187. doi: 10.1073/pnas.87.13.5183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Billiau A, Heremans H, Vandekerckhove F, Dijkmans R, Sobis H, Meulepas E, Carton H. Enhancement of experimental allergic encephalomyelitis in mice by antibodies against IFN-gamma. J Immunol. 1988;140(5):1506–1510. [PubMed] [Google Scholar]
  • 76.Mach B, Steimle V, Martinez-Soria E, Reith W. Regulation of MHC class II genes: lessons from a disease. Annu Rev Immunol. 1996;14:301–331. doi: 10.1146/annurev.immunol.14.1.301. [DOI] [PubMed] [Google Scholar]
  • 77.Groettrup M, Khan S, Schwarz K, Schmidtke G. Interferon-gamma inducible exchanges of 20S proteasome active site subunits: why? Biochimie. 2001;83(3–4):367–372. doi: 10.1016/S0300-9084(01)01251-2. [DOI] [PubMed] [Google Scholar]
  • 78.Nathan CF, Murray HW, Wiebe ME, Rubin BY. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983;158(3):670–689. doi: 10.1084/jem.158.3.670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Flaishon L, Hershkoviz R, Lantner F, Lider O, Alon R, Levo Y, Flavell RA, Shachar I. Autocrine secretion of interferon gamma negatively regulates homing of immature B cells. J Exp Med. 2000;192(9):1381–1388. doi: 10.1084/jem.192.9.1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Finkelman FD, Katona IM, Mosmann TR, Coffman RL. IFN-gamma regulates the isotypes of Ig secreted during in vivo humoral immune responses. J Immunol. 1988;140(4):1022–1027. [PubMed] [Google Scholar]
  • 81.Balkwill F, Taylor-Papadimitriou J. Interferon affects both G1 and S + G2 in cells stimulated from quiescence to growth. Nature. 1978;274(5673):798–800. doi: 10.1038/274798a0. [DOI] [PubMed] [Google Scholar]
  • 82.Xaus J, Cardo M, Valledor AF, Soler C, Lloberas J, Celada A. Interferon gamma induces the expression of p21waf-1 and arrests macrophage cell cycle, preventing induction of apoptosis. Immunity. 1999;11(1):103–113. doi: 10.1016/S1074-7613(00)80085-0. [DOI] [PubMed] [Google Scholar]
  • 83.Shiohara M, Koike K, Nakahata T. Synergism of interferon-gamma and stem cell factor on the development of murine hematopoietic progenitors in serum-free culture. Blood. 1993;81(6):1435–1441. [PubMed] [Google Scholar]
  • 84.Gajewski TF, Fitch FW. Anti-proliferative effect of IFN-gamma in immune regulation. I. IFN-gamma inhibits the proliferation of Th2 but not Th1 murine helper T lymphocyte clones. J Immunol. 1988;140(12):4245–4252. [PubMed] [Google Scholar]
  • 85.Yoshida A, Koide Y, Uchijima M, Yoshida TO. IFN-gamma induces IL-12 mRNA expression by a murine macrophage cell line, J774. Biochem Biophys Res Commun. 1994;198(3):857–861. doi: 10.1006/bbrc.1994.1122. [DOI] [PubMed] [Google Scholar]
  • 86.Kryczek I, Wei S, Gong W, Shu X, Szeliga W, Vatan L, Chen L, Wang G, Zou W. Cutting edge: IFN-gamma enables APC to promote memory Th17 and abate Th1 cell development. J Immunol. 2008;181(9):5842–5846. doi: 10.4049/jimmunol.181.9.5842. [DOI] [PubMed] [Google Scholar]
  • 87.Zou W, Restifo NP. T(H)17 cells in tumour immunity and immunotherapy. Nat Rev Immunol. 2010;10(4):248–256. doi: 10.1038/nri2742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kryczek I, Bruce AT, Gudjonsson JE, Johnston A, Aphale A, Vatan L, Szeliga W, Wang Y, Liu Y, Welling TH, Elder JT, Zou W. Induction of IL-17 + T cell trafficking and development by IFN-gamma: mechanism and pathological relevance in psoriasis. J Immunol. 2008;181(7):4733–4741. doi: 10.4049/jimmunol.181.7.4733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Feng T, Qin H, Wang L, Benveniste EN, Elson CO, Cong Y (2011) Th17 cells induce colitis and promote Th1 cell responses through IL-17 induction of innate IL-12 and IL-23 production. J Immunol [DOI] [PMC free article] [PubMed]
  • 90.Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM, Aguet M. Immune response in mice that lack the interferon-gamma receptor. Science. 1993;259(5102):1742–1745. doi: 10.1126/science.8456301. [DOI] [PubMed] [Google Scholar]
  • 91.Cantin E, Tanamachi B, Openshaw H. Role for gamma interferon in control of herpes simplex virus type 1 reactivation. J Virol. 1999;73(4):3418–3423. doi: 10.1128/jvi.73.4.3418-3423.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Muller U, Steinhoff U, Reis LF, Hemmi S, Pavlovic J, Zinkernagel RM, Aguet M. Functional role of type I and type II interferons in antiviral defense. Science. 1994;264(5167):1918–1921. doi: 10.1126/science.8009221. [DOI] [PubMed] [Google Scholar]
  • 93.Levin M, Newport MJ, D’Souza S, Kalabalikis P, Brown IN, Lenicker HM, Agius PV, Davies EG, Thrasher A, Klein N, et al. Familial disseminated atypical mycobacterial infection in childhood: a human mycobacterial susceptibility gene? Lancet. 1995;345(8942):79–83. doi: 10.1016/S0140-6736(95)90059-4. [DOI] [PubMed] [Google Scholar]
  • 94.Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra BA, Williamson R, Levin M. A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection. N Engl J Med. 1996;335(26):1941–1949. doi: 10.1056/NEJM199612263352602. [DOI] [PubMed] [Google Scholar]
  • 95.Jouanguy E, Lamhamedi-Cherradi S, Lammas D, Dorman SE, Fondaneche MC, Dupuis S, Doffinger R, Altare F, Girdlestone J, Emile JF, Ducoulombier H, Edgar D, Clarke J, Oxelius VA, Brai M, Novelli V, Heyne K, Fischer A, Holland SM, Kumararatne DS, Schreiber RD, Casanova JL. A human IFNGR1 small deletion hotspot associated with dominant susceptibility to mycobacterial infection. Nat Genet. 1999;21(4):370–378. doi: 10.1038/7701. [DOI] [PubMed] [Google Scholar]
  • 96.Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature. 2010;465(7299):793–797. doi: 10.1038/nature09135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ikeda H, Old LJ, Schreiber RD. The roles of IFN gamma in protection against tumor development and cancer immunoediting. Cytokine Growth Factor Rev. 2002;13(2):95–109. doi: 10.1016/S1359-6101(01)00038-7. [DOI] [PubMed] [Google Scholar]
  • 98.Dighe AS, Richards E, Old LJ, Schreiber RD. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity. 1994;1(6):447–456. doi: 10.1016/1074-7613(94)90087-6. [DOI] [PubMed] [Google Scholar]
  • 99.Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ, Schreiber RD. Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci USA. 1998;95(13):7556–7561. doi: 10.1073/pnas.95.13.7556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Street SE, Cretney E, Smyth MJ. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood. 2001;97(1):192–197. doi: 10.1182/blood.V97.1.192. [DOI] [PubMed] [Google Scholar]
  • 101.Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, Schreiber RD. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410(6832):1107–1111. doi: 10.1038/35074122. [DOI] [PubMed] [Google Scholar]
  • 102.Qin Z, Blankenstein T (2004) A cancer immunosurveillance controversy. Nat Immunol 5 (1):3-4 (author reply 4–5) [DOI] [PubMed]
  • 103.Schreiber TH, Podack ER. A critical analysis of the tumour immunosurveillance controversy for 3-MCA-induced sarcomas. Br J Cancer. 2009;101(3):381–386. doi: 10.1038/sj.bjc.6605198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Schreiber RD, Celada A, Buchmeier N. The role of interferon-gamma in the induction of activated macrophages. Ann Inst Pasteur Immunol. 1986;137C(2):203–206. doi: 10.1016/S0771-050X(86)80028-6. [DOI] [PubMed] [Google Scholar]
  • 105.Dighe AS, Campbell D, Hsieh CS, Clarke S, Greaves DR, Gordon S, Murphy KM, Schreiber RD. Tissue-specific targeting of cytokine unresponsiveness in transgenic mice. Immunity. 1995;3(5):657–666. doi: 10.1016/1074-7613(95)90136-1. [DOI] [PubMed] [Google Scholar]
  • 106.Murphy TL, Cleveland MG, Kulesza P, Magram J, Murphy KM. Regulation of interleukin 12 p40 expression through an NF-kappa B half-site. Mol Cell Biol. 1995;15(10):5258–5267. doi: 10.1128/mcb.15.10.5258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Carnaud C, Lee D, Donnars O, Park SH, Beavis A, Koezuka Y, Bendelac A. Cutting edge: cross-talk between cells of the innate immune system: NKT cells rapidly activate NK cells. J Immunol. 1999;163(9):4647–4650. [PubMed] [Google Scholar]
  • 108.Eberl G, MacDonald HR. Selective induction of NK cell proliferation and cytotoxicity by activated NKT cells. Eur J Immunol. 2000;30(4):985–992. doi: 10.1002/(SICI)1521-4141(200004)30:4<985::AID-IMMU985>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  • 109.Badovinac VP, Tvinnereim AR, Harty JT. Regulation of antigen-specific CD8 + T cell homeostasis by perforin and interferon-gamma. Science. 2000;290(5495):1354–1358. doi: 10.1126/science.290.5495.1354. [DOI] [PubMed] [Google Scholar]
  • 110.Whitmire JK, Tan JT, Whitton JL. Interferon-gamma acts directly on CD8 + T cells to increase their abundance during virus infection. J Exp Med. 2005;201(7):1053–1059. doi: 10.1084/jem.20041463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Bradley LM, Dalton DK, Croft M. A direct role for IFN-gamma in regulation of Th1 cell development. J Immunol. 1996;157(4):1350–1358. [PubMed] [Google Scholar]
  • 112.Scott P. IFN-gamma modulates the early development of Th1 and Th2 responses in a murine model of cutaneous leishmaniasis. J Immunol. 1991;147(9):3149–3155. [PubMed] [Google Scholar]
  • 113.Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, Huang E, Finlayson E, Simeone D, Welling TH, Chang A, Coukos G, Liu R, Zou W. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 2009;114(6):1141–1149. doi: 10.1182/blood-2009-03-208249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Fallarino F, Gajewski TF. Cutting edge: differentiation of antitumor CTL in vivo requires host expression of Stat1. J Immunol. 1999;163(8):4109–4113. [PubMed] [Google Scholar]
  • 115.Kacha AK, Fallarino F, Markiewicz MA, Gajewski TF. Cutting edge: spontaneous rejection of poorly immunogenic P1. HTR tumors by Stat6-deficient mice. J Immunol. 2000;165(11):6024–6028. doi: 10.4049/jimmunol.165.11.6024. [DOI] [PubMed] [Google Scholar]
  • 116.Cao X, Leonard K, Collins LI, Cai SF, Mayer JC, Payton JE, Walter MJ, Piwnica-Worms D, Schreiber RD, Ley TJ. Interleukin 12 stimulates IFN-gamma-mediated inhibition of tumor-induced regulatory T-cell proliferation and enhances tumor clearance. Cancer Res. 2009;69(22):8700–8709. doi: 10.1158/0008-5472.CAN-09-1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Luster AD, Ravetch JV. Biochemical characterization of a gamma interferon-inducible cytokine (IP-10) J Exp Med. 1987;166(4):1084–1097. doi: 10.1084/jem.166.4.1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Luster AD, Leder P. IP-10, a -C-X-C- chemokine, elicits a potent thymus-dependent antitumor response in vivo. J Exp Med. 1993;178(3):1057–1065. doi: 10.1084/jem.178.3.1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8(6):467–477. doi: 10.1038/nri2326. [DOI] [PubMed] [Google Scholar]
  • 120.Wu K, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppression of CD8 + T cells in human hepatocellular carcinoma is mediated by B7-H1/programmed death-1 interactions. Cancer Res. 2009;69(20):8067–8075. doi: 10.1158/0008-5472.CAN-09-0901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wiendl H, Mitsdoerffer M, Schneider D, Chen L, Lochmuller H, Melms A, Weller M. Human muscle cells express a B7-related molecule, B7-H1, with strong negative immune regulatory potential: a novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB J. 2003;17(13):1892–1894. doi: 10.1096/fj.03-0039fje. [DOI] [PubMed] [Google Scholar]
  • 122.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 123.Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, Saudemont A, Quesnel B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296–304. doi: 10.1182/blood-2006-10-051482. [DOI] [PubMed] [Google Scholar]
  • 124.Kondo A, Yamashita T, Tamura H, Zhao W, Tsuji T, Shimizu M, Shinya E, Takahashi H, Tamada K, Chen L, Dan K, Ogata K. Interferon-gamma and tumor necrosis factor-alpha induce an immunoinhibitory molecule, B7-H1, via nuclear factor-kappaB activation in blasts in myelodysplastic syndromes. Blood. 2010;116(7):1124–1131. doi: 10.1182/blood-2009-12-255125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Munn DH, Sharma MD, Lee JR, Jhaver KG, Johnson TS, Keskin DB, Marshall B, Chandler P, Antonia SJ, Burgess R, Slingluff CL, Jr, Mellor AL. Potential regulatory function of human dendritic cells expressing indoleamine 2, 3-dioxygenase. Science. 2002;297(5588):1867–1870. doi: 10.1126/science.1073514. [DOI] [PubMed] [Google Scholar]
  • 126.Carlin JM, Borden EC, Sondel PM, Byrne GI. Biologic-response-modifier-induced indoleamine 2, 3-dioxygenase activity in human peripheral blood mononuclear cell cultures. J Immunol. 1987;139(7):2414–2418. [PubMed] [Google Scholar]
  • 127.Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5(4):263–274. doi: 10.1038/nrc1586. [DOI] [PubMed] [Google Scholar]
  • 128.Mellor AL, Munn DH. Creating immune privilege: active local suppression that benefits friends, but protects foes. Nat Rev Immunol. 2008;8(1):74–80. doi: 10.1038/nri2233. [DOI] [PubMed] [Google Scholar]
  • 129.Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC. Inhibition of indoleamine 2, 3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med. 2005;11(3):312–319. doi: 10.1038/nm1196. [DOI] [PubMed] [Google Scholar]
  • 130.Sharma MD, Baban B, Chandler P, Hou DY, Singh N, Yagita H, Azuma M, Blazar BR, Mellor AL, Munn DH. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2, 3-dioxygenase. J Clin Invest. 2007;117(9):2570–2582. doi: 10.1172/JCI31911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Alegre ML, Puccetti P. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol. 2003;4(12):1206–1212. doi: 10.1038/ni1003. [DOI] [PubMed] [Google Scholar]
  • 132.Muller AJ, Sharma MD, Chandler PR, Duhadaway JB, Everhart ME, Johnson BA, III, Kahler DJ, Pihkala J, Soler AP, Munn DH, Prendergast GC, Mellor AL. Chronic inflammation that facilitates tumor progression creates local immune suppression by inducing indoleamine 2, 3 dioxygenase. Proc Natl Acad Sci USA. 2008;105(44):17073–17078. doi: 10.1073/pnas.0806173105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, Ochoa JB, Ochoa AC. l-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J Immunol. 2003;171(3):1232–1239. doi: 10.4049/jimmunol.171.3.1232. [DOI] [PubMed] [Google Scholar]
  • 134.Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM, Antonia S, Ochoa JB, Ochoa AC. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64(16):5839–5849. doi: 10.1158/0008-5472.CAN-04-0465. [DOI] [PubMed] [Google Scholar]
  • 135.Saio M, Radoja S, Marino M, Frey AB. Tumor-infiltrating macrophages induce apoptosis in activated CD8(+) T cells by a mechanism requiring cell contact and mediated by both the cell-associated form of TNF and nitric oxide. J Immunol. 2001;167(10):5583–5593. doi: 10.4049/jimmunol.167.10.5583. [DOI] [PubMed] [Google Scholar]
  • 136.Kusmartsev SA, Li Y, Chen SH. Gr-1 + myeloid cells derived from tumor-bearing mice inhibit primary T cell activation induced through CD3/CD28 costimulation. J Immunol. 2000;165(2):779–785. doi: 10.4049/jimmunol.165.2.779. [DOI] [PubMed] [Google Scholar]
  • 137.Liscovsky MV, Ranocchia RP, Gorlino CV, Alignani DO, Moron G, Maletto BA, Pistoresi-Palencia MC. Interferon-gamma priming is involved in the activation of arginase by oligodeoxinucleotides containing CpG motifs in murine macrophages. Immunology. 2009;128(1 Suppl):e159–e169. doi: 10.1111/j.1365-2567.2008.02938.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, Zanovello P, Segal DM. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. 2002;168(2):689–695. doi: 10.4049/jimmunol.168.2.689. [DOI] [PubMed] [Google Scholar]
  • 139.Modolell M, Corraliza IM, Link F, Soler G, Eichmann K. Reciprocal regulation of the nitric oxide synthase/arginase balance in mouse bone marrow-derived macrophages by TH1 and TH2 cytokines. Eur J Immunol. 1995;25(4):1101–1104. doi: 10.1002/eji.1830250436. [DOI] [PubMed] [Google Scholar]
  • 140.Hesse M, Modolell M, La Flamme AC, Schito M, Fuentes JM, Cheever AW, Pearce EJ, Wynn TA. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of l-arginine metabolism. J Immunol. 2001;167(11):6533–6544. doi: 10.4049/jimmunol.167.11.6533. [DOI] [PubMed] [Google Scholar]
  • 141.Norian LA, Rodriguez PC, O’Mara LA, Zabaleta J, Ochoa AC, Cella M, Allen PM. Tumor-infiltrating regulatory dendritic cells inhibit CD8 + T cell function via l-arginine metabolism. Cancer Res. 2009;69(7):3086–3094. doi: 10.1158/0008-5472.CAN-08-2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, McDermott D, Quiceno D, Youmans A, O’Neill A, Mier J, Ochoa AC. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res. 2005;65(8):3044–3048. doi: 10.1158/0008-5472.CAN-04-4505. [DOI] [PubMed] [Google Scholar]
  • 143.Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res. 2007;13(2 Pt 2):721s–726s. doi: 10.1158/1078-0432.CCR-06-2197. [DOI] [PubMed] [Google Scholar]
  • 144.Rodriguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–191. doi: 10.1111/j.1600-065X.2008.00608.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, Ochoa AC. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69(4):1553–1560. doi: 10.1158/0008-5472.CAN-08-1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Highfill SL, Rodriguez PC, Zhou Q, Goetz CA, Koehn BH, Veenstra R, Taylor PA, Panoskaltsis-Mortari A, Serody JS, Munn DH, Tolar J, Ochoa AC, Blazar BR. Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood. 2010;116(25):5738–5747. doi: 10.1182/blood-2010-06-287839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ilangumaran S, Rottapel R. Regulation of cytokine receptor signaling by SOCS1. Immunol Rev. 2003;192:196–211. doi: 10.1034/j.1600-065X.2003.00020.x. [DOI] [PubMed] [Google Scholar]
  • 148.Starr R, Fuchsberger M, Lau LS, Uldrich AP, Goradia A, Willson TA, Verhagen AM, Alexander WS, Smyth MJ. SOCS-1 binding to tyrosine 441 of IFN-gamma receptor subunit 1 contributes to the attenuation of IFN-gamma signaling in vivo. J Immunol. 2009;183(7):4537–4544. doi: 10.4049/jimmunol.0901010. [DOI] [PubMed] [Google Scholar]
  • 149.Taniguchi K, Petersson M, Hoglund P, Kiessling R, Klein G, Karre K. Interferon gamma induces lung colonization by intravenously inoculated B16 melanoma cells in parallel with enhanced expression of class I major histocompatibility complex antigens. Proc Natl Acad Sci USA. 1987;84(10):3405–3409. doi: 10.1073/pnas.84.10.3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Cho HI, Lee YR, Celis E (2010) Interferon gamma limits the effectiveness of melanoma peptide vaccines. Blood [DOI] [PMC free article] [PubMed]
  • 151.Morel S, Levy F, Burlet-Schiltz O, Brasseur F, Probst-Kepper M, Peitrequin AL, Monsarrat B, Van Velthoven R, Cerottini JC, Boon T, Gairin JE, Van den Eynde BJ. Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells. Immunity. 2000;12(1):107–117. doi: 10.1016/S1074-7613(00)80163-6. [DOI] [PubMed] [Google Scholar]
  • 152.Gobin SJ, van den Elsen PJ. Transcriptional regulation of the MHC class Ib genes HLA-E, HLA-F, and HLA-G. Hum Immunol. 2000;61(11):1102–1107. doi: 10.1016/S0198-8859(00)00198-1. [DOI] [PubMed] [Google Scholar]
  • 153.Zaidi MR, Davis S, Noonan FP, Graff-Cherry C, Hawley TS, Walker RL, Feigenbaum L, Fuchs E, Lyakh L, Young HA, Hornyak TJ, Arnheiter H, Trinchieri G, Meltzer PS, De Fabo EC, Merlino G. Interferon-gamma links ultraviolet radiation to melanomagenesis in mice. Nature. 2011;469(7331):548–553. doi: 10.1038/nature09666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Berner V, Liu H, Zhou Q, Alderson KL, Sun K, Weiss JM, Back TC, Longo DL, Blazar BR, Wiltrout RH, Welniak LA, Redelman D, Murphy WJ. IFN-gamma mediates CD4 + T-cell loss and impairs secondary antitumor responses after successful initial immunotherapy. Nat Med. 2007;13(3):354–360. doi: 10.1038/nm1554. [DOI] [PubMed] [Google Scholar]
  • 155.Garbe C, Krasagakis K, Zouboulis CC, Schroder K, Kruger S, Stadler R, Orfanos CE. Antitumor activities of interferon alpha, beta, and gamma and their combinations on human melanoma cells in vitro: changes of proliferation, melanin synthesis, and immunophenotype. J Invest Dermatol. 1990;95(6 Suppl):231S–237S. doi: 10.1111/1523-1747.ep12875837. [DOI] [PubMed] [Google Scholar]
  • 156.Creagan ET, Ahmann DL, Long HJ, Frytak S, Sherwin SA, Chang MN. Phase II study of recombinant interferon-gamma in patients with disseminated malignant melanoma. Cancer Treat Rep. 1987;71(9):843–844. [PubMed] [Google Scholar]
  • 157.Ernstoff MS, Trautman T, Davis CA, Reich SD, Witman P, Balser J, Rudnick S, Kirkwood JM. A randomized phase I/II study of continuous versus intermittent intravenous interferon gamma in patients with metastatic melanoma. J Clin Oncol. 1987;5(11):1804–1810. doi: 10.1200/JCO.1987.5.11.1804. [DOI] [PubMed] [Google Scholar]
  • 158.Kopp WC, Smith JW, II, Ewel CH, Alvord WG, Main C, Guyre PM, Steis RG, Longo DL, Urba WJ. Immunomodulatory effects of interferon-gamma in patients with metastatic malignant melanoma. J Immunother Emphas Tumor Immunol. 1993;13(3):181–190. doi: 10.1097/00002371-199304000-00005. [DOI] [PubMed] [Google Scholar]
  • 159.Kowalzick L, Weyer U, Lange P, Breitbart EW. Systemic therapy of advanced metastatic malignant melanoma with a combination of fibroblast interferon-beta and recombinant interferon-gamma. Dermatologica. 1990;181(4):298–303. doi: 10.1159/000247830. [DOI] [PubMed] [Google Scholar]
  • 160.Meyskens FL, Jr, Kopecky K, Samson M, Hersh E, Macdonald J, Jaffe H, Crowley J, Coltman C. Recombinant human interferon gamma: adverse effects in high-risk stage I and II cutaneous malignant melanoma. J Natl Cancer Inst. 1990;82(12):1071. doi: 10.1093/jnci/82.12.1071-a. [DOI] [PubMed] [Google Scholar]
  • 161.Meyskens FL, Jr, Kopecky KJ, Taylor CW, Noyes RD, Tuthill RJ, Hersh EM, Feun LG, Doroshow JH, Flaherty LE, Sondak VK. Randomized trial of adjuvant human interferon gamma versus observation in high-risk cutaneous melanoma: a southwest oncology group study. J Natl Cancer Inst. 1995;87(22):1710–1713. doi: 10.1093/jnci/87.22.1710. [DOI] [PubMed] [Google Scholar]
  • 162.Coughlin CM, Salhany KE, Gee MS, LaTemple DC, Kotenko S, Ma X, Gri G, Wysocka M, Kim JE, Liu L, Liao F, Farber JM, Pestka S, Trinchieri G, Lee WM. Tumor cell responses to IFNgamma affect tumorigenicity and response to IL-12 therapy and antiangiogenesis. Immunity. 1998;9(1):25–34. doi: 10.1016/S1074-7613(00)80585-3. [DOI] [PubMed] [Google Scholar]
  • 163.Chin YE, Kitagawa M, Su WC, You ZH, Iwamoto Y, Fu XY. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1. Science. 1996;272(5262):719–722. doi: 10.1126/science.272.5262.719. [DOI] [PubMed] [Google Scholar]
  • 164.Bromberg JF, Horvath CM, Wen Z, Schreiber RD, Darnell JE., Jr Transcriptionally active Stat1 is required for the antiproliferative effects of both interferon alpha and interferon gamma. Proc Natl Acad Sci USA. 1996;93(15):7673–7678. doi: 10.1073/pnas.93.15.7673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Sgadari C, Angiolillo AL, Cherney BW, Pike SE, Farber JM, Koniaris LG, Vanguri P, Burd PR, Sheikh N, Gupta G, Teruya-Feldstein J, Tosato G. Interferon-inducible protein-10 identified as a mediator of tumor necrosis in vivo. Proc Natl Acad Sci USA. 1996;93(24):13791–13796. doi: 10.1073/pnas.93.24.13791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Arenberg DA, Kunkel SL, Polverini PJ, Morris SB, Burdick MD, Glass MC, Taub DT, Iannettoni MD, Whyte RI, Strieter RM. Interferon-gamma-inducible protein 10 (IP-10) is an angiostatic factor that inhibits human non-small cell lung cancer (NSCLC) tumorigenesis and spontaneous metastases. J Exp Med. 1996;184(3):981–992. doi: 10.1084/jem.184.3.981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Sgadari C, Farber JM, Angiolillo AL, Liao F, Teruya-Feldstein J, Burd PR, Yao L, Gupta G, Kanegane C, Tosato G. Mig, the monokine induced by interferon-gamma, promotes tumor necrosis in vivo. Blood. 1997;89(8):2635–2643. [PubMed] [Google Scholar]
  • 168.Coughlin CM, Salhany KE, Wysocka M, Aruga E, Kurzawa H, Chang AE, Hunter CA, Fox JC, Trinchieri G, Lee WM. Interleukin-12 and interleukin-18 synergistically induce murine tumor regression which involves inhibition of angiogenesis. J Clin Invest. 1998;101(6):1441–1452. doi: 10.1172/JCI1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Briesemeister D, Sommermeyer D, Loddenkemper C, Loew R, Uckert W, Blankenstein T, Kammertoens T. Tumor rejection by local interferon gamma induction in established tumors is associated with blood vessel destruction and necrosis. Int J Cancer. 2011;128(2):371–378. doi: 10.1002/ijc.25350. [DOI] [PubMed] [Google Scholar]
  • 170.Groux H, Bigler M, de Vries JE, Roncarolo MG. Inhibitory and stimulatory effects of IL-10 on human CD8 + T cells. J Immunol. 1998;160(7):3188–3193. [PubMed] [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES