Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 1.
Published in final edited form as: Semin Cancer Biol. 2012 Mar 6;22(4):298–306. doi: 10.1016/j.semcancer.2012.02.010

Tumor Associated Regulatory Dendritic Cells

Yang Ma 1, Galina V Shurin 1, Dmitriy W Gutkin 2, Michael R Shurin 1,3
PMCID: PMC3373995  NIHMSID: NIHMS363039  PMID: 22414911

Abstract

Immune effector and regulatory cells in the tumor microenvironment are key factors in tumor development and progression as the pathogenesis of cancer vitally depends on the multifaceted interactions between various microenvironmental stimuli provided by tumor-associated immune cells. Immune regulatory cells participate in all stages of cancer development from the induction of genomic instability to the maintenance of intratumoral angiogenesis, proliferation and spreading of malignant cells, and formation of premetastatic niches in distal tissues. Dendritic cells in the tumor microenvironment serve as a double-edged sword and, in addition to initiating potent anti-tumor immune responses, may mediate genomic damage, support neovascularization, block anti-tumor immunity and stimulate cancerous cell growth and spreading. Regulatory dendritic cells in cancer may directly and indirectly maintain antigen-specific and non-specific T cell unresponsiveness by controlling T cell polarization, MDSC and Treg differentiation and activity, and affecting specific microenvironmental conditions in premalignant niches. Understanding the mechanisms involved in regulatory dendritic cell polarization and operation and revealing pharmacological means for harnessing these pathways will provide additional opportunities for modifying the tumor microenvironment and improving the efficacy of different therapeutic approaches to cancer.

Keywords: dendritic cells, cancer, immunosuppression, immune escape, tolerance, immune regulatory cells

Introduction: Dendritic cell dichotomy

Dendritic cells (DCs) are potent antigen presenting cells (APCs) that possess the ability to present antigens to antigen-specific naïve T cells, as well as to maintain both innate and adoptive immune responses. DCs are derived from the bone marrow hematopoietic progenitor cells and circulate in the blood as immature precursors prior to migration into the peripheral tissues. Within the specific tissue microenvironment, DCs differentiate further displaying a potential to pick up and process antigens for their subsequent presentation as peptide-major histocompatibility complex (MHC) on the surface of DCs. Upon appropriate stimulation, DCs undergo further maturation and migrate to secondary lymphoid tissues where they present antigens to T cells inducing their clonal proliferation and the immune response [1]. However, specific microenvironmental signaling might prevent maturation of DCs or polarize their differentiation resulting in formation of DC subsets with tolerogenic and immunosuppressive potential accountable for antigen-specific unresponsiveness in the lymphoid organs and in the periphery [2].

Thus, DCs represent a heterogeneous hematopoietic lineage with cell subsets in different tissues showing differential morphology, phenotype and function, and often sharing the ability to stimulate T cell proliferation or induce immune tolerance between various DC subsets depending on the environmental conditions. Although it has been suggested that the so-called `myeloid' versus `lymphoid' subsets of DCs, or conventional (cDCs) versus plasmacytoid (pDCs) subpopulations perform specific stimulatory or tolerogenic function, respectively, these concepts suffer from numerous exceptions in different experimental systems and clinical situations. Even “classically” inducing immune response by activating CD4+ Th1 and Th2 cells and CD8+ CTLs, cDC, including tissue-resident DCs, migratory DCs and inflammatory DCs might exhibit immunosuppressive properties under certain circumstances or in immature stage [35]. Other DC subsets, such as the pDCs, have also been reported to exhibit potent immunosuppressive and tolerogenic properties by blocking proliferation of naïve and antigen-specific CD4+ and CD8+ T cells, supporting polarization and activation of Treg lymphocytes or efficiently presenting antigens to CTL and inducing efficient immune responses [68].

Another commonly accepted paradigm is that functional properties of DCs are maturation-dependent. However, existing evidence suggests that DCs can exist in a multitude of functional states other than simply immature or mature. Additionally, both phenotypically “immature” and “mature” DCs may be conditioned by the microenvironment to maintain either immune tolerance or immunosuppression [9]. Thus, DCs are a specialized group of antigen-presenting cells with high functional plasticity that express immunostimulating or immunosuppressive potential, or both, depending on the consequence and combination of microenvironmental stimuli affecting DC differentiation, maturation, activation and polarization. A wide spectrum of cells and factors in the tumor microenvironment represent an excellent example of differential stimuli that affect all aspects of DC biology and thus control functionality and longevity of all DC subsets.

Dendritic cells in cancer

DCs has been attracting scientific and clinical attention due to their key role in tumor immunity and a potential for being used as biological adjuvants in tumor vaccinations [10]. However, anti-tumor immune responses are often deficient or suboptimal since tumor cells are able to exploit the functional roles of DCs for tumor growth and progression [11]. Suppression and re-polarization of DC function in cancer patients is thought to contribute to the failure of anti-tumor immune responses and consequent disease progression. Subversion of tumor immunity by manipulating the tumor microenvironment and DC subset distribution and function is mediated by various tumor-derived and stromal factors, many of which remain to be identified. Molecular mechanisms of tumor-mediated dysfunction of conventional DC have partly described, although potential signaling pathways responsible for emerging and function of immunosuppressive or tolerogenic DC subsets remain elusive.

Many mechanisms of conventional DC alterations in the tumor microenvironment have been revealed during the last 20 years, and most of them relate to the inhibition of dendropoiesis (DC production), decelerating DC differentiation, induction of functional deficiency of DCs and acceleration of cell death of semi-mature/mature DCs or DC precursors [1215]. For instance, in 1991–1995 several groups reported that the antigen-presenting capacity of lymph node cells was impaired during tumorigenesis [16], that tumor was able to regulate DC attraction and homing at the tumor site and inhibit function of DCs and thus, induction of anti-tumor immunity [17, 18] and that DCs were functionally abnormal in patients with cancer [19]. In 1996, Gabrilovich et al. reported that DCs isolated from tumor-bearing mice displayed reduced ability to induce syngeneic tumor-specific CTLs and stimulate allogeneic T cells [20] and Chaux et al. revealed that tumor-associated DCs express low levels of co-stimulatory molecules [21]. Following these initial findings, other groups demonstrated lower generation of human CD34-derived and CD14-derived DCs in patients with cancer, as well as murine bone marrow-derived DCs in tumor-bearing mice and described a significant decrease in the number of circulating DCs in the peripheral blood of cancer patients [2228].

Elimination of functional cDCs in cancer may be also associated with apoptosis of DCs or acceleration of their turnover. Induction of apoptosis in DCs by tumor-derived factors was first reported by Esche et al. in 1999 [12] and then confirmed by others [2931]. Tumor-mediated cell death of DC precursors [32] and accelerated early apoptosis of DCs [29, 33] were also described. The presence of a significantly higher proportion of apoptotic blood DCs in patients with early stage breast cancer compared to healthy volunteers may also support this mechanism of cDC elimination in the tumor environment [34].

Functional deficiency of cDCs in the tumor environment is the best documented type of DC abnormalities observed in cells harvested from cancer patients and cells generated in cultures treated with tumor cell line-derived factors or primary tumor cells. Inhibited ability of DCs to stimulate allogeneic and syngeneic T cell proliferation, decreased uptake, processing and presentation of antigens, lowered expression of co-stimulatory signal, inefficient motility and migration towards specific chemokines, suppressed endocytic potential and decreased production of IL-12 were repeatedly described for prostate, breast, renal, liver, lung cancer, head and neck squamous cell carcinoma (HNSCC), melanoma, myeloma, leukemia, glioma, neuroblastoma and other tumor types by our and other teams [32, 33, 3540] and repeatedly reviewed [15, 41, 42]. It is important to mention here that functionally deficient DCs are usually not immunosuppressive: they are unable to induce activation of antigen-specific or allogeneic T cells, but do not actively suppress proliferation of pre-activated T cells and do not induce functional tolerance or Treg cell differentiation. However, in specific tumor microenvironment conditions, the loss of function in DCs may, at least in part, be associated with DC polarization and acquisition of tolerogenic and/or immunosuppressive activities.

Regulatory DC subsets in cancer

Tumor-promoted redirection of dendropoiesis and polarization of DC differentiation is commonly associated with the engagement and accumulation of DC subsets that actively block development of anti-tumor immunity, promote appearance of regulatory T cells and myeloid-derived suppressor cells (MDSCs) and support tumor progression by endorsing intratumoral neoangiogenesis and development of metastases. Probably, Enk et al. were the first who in 1997 showed that melanoma-derived factors converted the antigen-presenting function of DCs to tolerance induction against tumor tissue [43].

Although cultured bone marrow-derived immature DCs do not possess immunosuppressive ability, when appropriately conditioned in the tumor microenvironment, in vivo and in vitro, they may inhibit innate and adaptive immunity by various mechanisms. Immature DCs were found at high levels within tumor-infiltrating leukocytes and increased circulating levels of immature DCs have also been reported in the peripheral blood of patients with lung, breast, head and neck and esophageal cancer [44]. Certain subsets of immature DCs fail to provide an appropriate co-stimulatory and cytokine signals to T cells and might induce tolerance through abortive proliferation or anergy of antigen-specific CD4+ and CD8+ T cells or through the generation of regulatory T cells that prevent immune responses by producing IL-10 and TGF-β [5, 4548]. These mechanisms usually account for the deletion of autoreactive T cells, but in the tumor environment may be directed towards the inhibition of anti-tumor immunity. In fact, Ghiringhelli et al. have demonstrated that during tumor progression, a subset of immature myeloid DCs is recruited to draining lymph nodes and selectively promotes the proliferation of Treg cells in a TGF-β-dependent manner [49]. Tumor cells were necessary and sufficient to convert immature DCs into regulatory DCs that secrete TGF-β and stimulate reg cell proliferation. Thus, although it is widely accepted that the ability of DCs to initiate immune responses or induce tolerance is strictly dependent on their maturation state or subsets, increasing evidence suggests that maturation status of DCs should no longer be considered as a distinguishing feature of stimulatory versus regulatory DC phenotype. Several publications describing that mature DCs induce CD4+ T cell tolerance [5052] challenged the model of tolerogenic immature and immunogenic mature differentiation stages in DCs. Furthermore, it seems that regDCs can exist as immature, semi-mature and fully mature DC subpopulations that use different mechanisms for induction of immune tolerance and immune suppression.

Maturation of DCs includes up-regulation of MHC class II and co-stimulatory molecules as well as secretion of proinflammatory cytokines. Mature immunogenic DCs produce large amounts of IL-12, TNF-α, IL-1 and IL-6 [4, 51]. Recently DCs that express high levels of MHC class II and co-stimulatory molecules but do not secrete IL-1β, IL-6, TNF-α and IL-12 have been shown to display tolerogenic rather than stimulatory properties and therefore been referred as semi-mature tolerogenic DCs [4]. Furthermore, Akbari et al. showed that IL-10 producing DCs with a mature phenotype could initiate CD4+ T cell unresponsiveness after exposure to an antigen and induce Treg cells that also produce high amounts of IL-10 [50]. Thus, the IL-10/IL-12 production profile of DCs along with the low grade phenotypic maturation might distinguish between semi-mature DCs with the regulatory properties and fully mature stimulatory DC subpopulations

The ability of mature DCs to induce T cell tolerance was explained by a hypothesis that a specialized subset of mature DCs might actively divert T cell responses towards tolerance [53]. DCs treated with IFN-γ and displaying a mature cell phenotype (CD80+CD83+CD86highHLA-DRhigh) might exert tolerogenic properties due to an additional expression of indoleamine 2,3-dioxygenase (IDO) [54, 55]. These mature regulatory DCs were effective stimulators of T cell proliferation if IDO was blocked with the specific inhibitor 1-methyl tryptophan (1-MT), suggesting that these cells could also act as competent antigen-presenting cells. These data point towards a functional plasticity of mature DCs, allowing them to adopt either suppressive/tolerogenic or activating/immunogenic phenotypes depending on the signals received [56].

Regulatory properties of plasmacytoid DCs

Plasmacytoid DCs are recognized as the main source of IFN-α after challenge with pathogens [57, 58]. They originate in the bone marrow from DC progenitors common to pDCs and cDCs [59, 60] and can be identified as CD4+CD11clinHLADR+ cells expressing CD123/IL-3Rα chain. At the steady state, pDCs circulate in the blood and may enter the lymph nodes through the high endothelial veinules [61, 62]. While present in tissues at low numbers in the healthy steady state, pDCs accumulate in lymphoid and non-lymphoid tissues under different pathological conditions [63].

Plasmacytoid DCs are considered to be crucial effector cells in innate and adaptive immunity and express distinctive pathogen recognition receptors mainly residing in the endosomes and consisting of TLR-7 and TLR-9. The engagement of TLR-7/9 by viral RNA and DNA leads to powerful type I IFN secretion. In addition to IFN, pDCs produce other cytokines, such as TNF-α, IL-6 and CXCL8 and inflammatory chemokines, such as CXCL9, CXCL10, CCL3, CCL4 and CCL5 [64, 65]. Freshly isolated pDCs can present antigens to T cells to a small extent, but upon activation by viruses, CpG, IL-3 and CD40L, they acquire full DC properties and are capable of presenting antigens to CD4+ T cells and cross-prime CD8+ T cells [6669]. Through the release of IFN-α, pDCs can activate NK cells, promote CTL activity and induce differentiation of B lymphocytes into Ig-secreting plasma cells [70].

While the importance of pDCs in innate and adaptive immune responses against pathogens is well established, their role in anti-tumor immunity is not clear. Due to their ability to secrete high levels of type-I IFN and TNF-α, pDCs would appear to have potential to promote anti-tumor immunity: IFN-α has direct antitumor activities by inhibiting tumor cell proliferation and neoangiogenesis and also by promoting immunosurveillance through the activation of B cells, NK cells and macrophages [71]. Theoretically, pDCs have an ability to orchestrate the local immune response to cancer cells by producing IFN, recruiting other immune cells via amplification of the proinflammatory chemokine network and cross-presenting antigens to CD8+ T cells [65, 67, 69]. They can also induce apoptosis of tumor cell lines either directly by secreting TRAIL or indirectly via the effect of IFN-α on other cytotoxic cells [72]. However, numerous experimental and clinical evidence shows exactly the opposite: pDCs possess immunosuppressive and tolerogenic property and, thus, promote tumor growth and progression. This apparent discrepancy might be explained, at least in part, by the homing of pDCs into the tumor and the properties of tumor associated pDCs (TApDC).

As has been mentioned above, the number of pDCs in normal peripheral tissues is low. However, circulating pDCs express multiple chemotactic receptors, including CXCR4 and ChemR23 (CMKLR1) [7]. Malignant tumors have been shown to express high levels of CXCR4 ligand, stromal-derived factor-1 (CXCL12), which likely represents the main axis for pDC accumulation in tumors [7375]. Interaction of stromal cell-derived factor-1 with CXCR4 on pDCs can up-regulate very late antigen-5 for trans-endothelial migration via vascular cell adhesion molecule-1 and can protect pDCs from IL-10-induced apoptosis [73]. Other possible interaction includes CCL20 binding to CCR6 on pDCs that can recruit them to the tumor site [76].

Accumulation of pDCs in tumors has been directly demonstrated in primary carcinomas of different organs (breast, ovary, head and neck, lung, skin, cervix, prostate and liver), as well as cutaneous melanoma and lymphomas [74, 75, 7781]. They commonly represent a minor fraction (10 –15%) of the infiltrating immune cells [7], but at least in some tumors they were found to be the most abundant DC subset [78]. As the well-established methods of isolation and identification of TApDCs are now available, several studies were focused on a functional characterization of these cells. Practically all of the currently existing results point to the same conclusion: TApDCs are defective in IFN-α production and instead secrete immunosuppressive soluble factors responsible for tumor progression. Immunosuppressive and tolerogenic TApDCs were demonstrated in both murine and human prostate carcinoma [57, 80], HNSCC [77] and ovarian carcinoma [75, 78]. It is important to mention that these findings have strong clinical correlations: the results of several studies indicate that prognosis of different types of tumors is inversely related to the number of tumor-infiltrating pDCs [78, 81].

Although the mechanisms of this phenomenon are not known, several possible scenarios can explain its origin. First, it has been proven that TApDCs are disarmed in their ability to produce the required amount of IFN-α to sustain elimination of cancer cells. This functional inhibition of pDCs likely depends on the abundance of ligands to the inhibitory receptors on cancer cells or on cells of the tumor microenvironment [7]. Human pDCs express several receptors that negatively regulate the amplitude of the IFN-α response. One of them, ILT7, recognizes BST2, the protein that is strongly expressed on tumor cell lines and carcinomas [82, 83]. Next, human and mouse pDCs can drive CD4+ T cells to the generation of CD4+CD25+Foxp3+ Treg cells, which leads to anergy and immune suppression, favoring the immune escape of tumor cells [8486]. Recent findings also demonstrate that pDCs infiltrating prostate carcinoma can be polarized to express IDO and other tolerogenic mediators under the control of forkhead box O3 (FOXO3) [80]. Catabolic activity of IDO and arginase in pDCs eventually leads to the inhibition of T cell activation and immunosuppression [87, 88]. Another possible tolerogenic mechanism of TApDCs is the secretion of Granzyme B. It has been shown that under IL-3 and IL-10 exposure, human pDCs release abundant amounts of Granzyme B, which is capable of blocking T cell proliferation [89].

All of these mechanisms underline the active role of the tumor microenvironment in polarizing tolerogenic pDCs. In addition, recent study of Bjorck et al. identified two distinct subsets of pDCs in mice, one of which (CD9+ immature pDCs) was found mainly in the bone marrow and spleen and was responsible for IFN-α production, while the other (CD9 mature pDCs) was present in the peripheral tissues and was tolerogenic [8]. These results indicate that even without the negative influence of the tumor microenvironment TApDCs would not be actively producing IFN or other inflammatory cytokines, but instead induce the tolerance. Additional analysis of these pDC subsets in the tumor milieu should provide interesting insights into our understanding of how tumor-derived factors affect immunogenic and constitutively tolerogenic pDC subpopulations.

Regulatory dendritic cells: Initiating mechanisms and factors

The ability of DCs to coordinate the immune response is not an intrinsic quality of the cell but is rather the result of specific microenvironmental signals, including the local cytokine network and the milieu of soluble factors from the neighboring cells. For instance, tumor-derived IL-10, TGF-βand PgE2 can render DCs to acquire regulatory instead of stimulatory capacities. IL-10, which is produced by many cells in the tumor milieu, including T cells, B cells, macrophages, mast cells and cancerous cells, can inhibits maturation of antigen-presenting cells, reduce expression of MHC class I and II molecules and co-stimulatory molecules and attenuate production of inflammatory cytokines [9092]. CCR-7 dependent migration of DCs to secondary lymphoid tissue is also impaired by the presence of IL-10 [93]. IL-10-treated DCs can be polarized to induce T cell anergy [94]. In fact, tumor-derived IL-10 has been reported to induce tolerance to tumor tissue by changing the phenotypic and functional properties of DCs in the tumor microenvironment [43]. Comparing different factors known to induce regulatory DCs, Boks et al. reported that IL-10-treated human DCs showed most powerful tolerogenic characteristics [95]. Furthermore, in addition to being involved in regDC-induced polarization of Treg cells, IL-10 may participate in Treg cell-mediated polarization of regDCs. Studying the interaction between Treg cells and antigen-presenting cells (APCs), Kryczek et al. demonstrated that Treg cells, but not conventional T lymphocytes, triggered high levels of IL-10 production by APCs, stimulate APC B7-H4 expression, and render APCs immunosuppressive [96]. Initial blockade of B7-H4 reduced the suppressive activity mediated by Treg cell-conditioned APCs. Further, APC-derived, rather than Treg cell-derived, IL-10 was responsible for APC B7-H4 induction, suggesting that Treg cells convey suppressive activity to APCs by stimulating B7-H4 expression through IL-10 [96].

TGF-βis another soluble factor produced in the tumor microenvironment [97, 98] and associated with the tolerance induction. Similar to IL-10, exposure of DCs to TGF-βinhibits their ability to process antigens, migrate to the draining lymph nodes and stimulate tumor-specific T cells [99]. Murine bone marrow-derived DC that were propagated in IL-10 and TGF-β (so-called “alternatively activated” DCs) expressed low levels of TLR4, MHC class II, CD40, CD80, CD86 and IL-12p70, secreted much higher levels of IL-10 and efficiently expanded functional CD4+CD25+Foxp3+ Treg cells [100]. Regulatory DCs could be also produced from bone marrow precursors in the presence of GM-CSF, IL-10, TGF-β1, LPS or TNF-α and they retained their T cell regulatory property in vitro and in vivo even under inflammatory conditions [101]. Interestingly, CCL18 has been reported to differentiate DCs in tolerogenic cells able to prime regulatory T cells [102]. Another minor subpopulation of regDCs has been recently described in murine spleen. These splenic CD19+ DCs that did not express the plasmacytoid DC marker acquired potent IDO-dependent T cell suppressive functions [103].

Tumor-derived PgE2 can also induce DC-mediated T cell tolerance. Increased levels of PgE2 were reported for many solid and hematological malignancies, especially in tumors associated with chronic inflammatory responses, such as colon cancer [104], breast cancer [105, 106] and lymphoma [107]. Interestingly, in addition to the ability to directly suppress CD4+ T cell proliferation by inhibiting the TCR-associated tyrosine kinase Lck [107] and induce regulatory T cells [108110], PgE2 has been also shown to affect DC activity by blocking IL-12 expression [111] and inducing expression of regulatory molecules [112] that both result in impaired T cell stimulation.

Human leukocyte antigen G (HLA-G) molecules, which are normally expressed in cytotrophoblasts and play a key role in maintaining immune tolerance at the maternal-fetal interface, was also reported to be expressed on malignant cells and can be regulated by hypoxia [113, 114]. As DCs expressed immunoglobulin-like transcript 4 (ILT4), an inhibitory receptor capable of interacting with HLA-G, DCs may be tolerized by HLA-G through inhibitory receptor interactions. Indeed, the HLA-G-ILT4 interaction leads to development of tolerogenic DCs with the induction of anergic and immunosuppressive T cells [115]. DCs express a number of inhibitory receptors, which are characterized by the presence of cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Upon receptor engagement, ITIMs are phosphorylated and subsequently activate phosphatases that down-regulate the process of cell activation. Two inhibitory receptors, ILT3 and ILT4, may be connected with the induction of regulatory DCs. Up-regulation of ILT3 and ILT4 was associated with decreased expression of co-stimulatory molecules and induction of T cell anergy by DCs [116].

Regulatory dendritic cells: Initiating signaling pathways

Although the list of tumor-derived and stromal factors involving in the impaired or repolarized DC functions is getting longer, some of them may utilize similar transcription factors and signaling pathways, in particular STAT3, MAP kinase and small Rho GTPases. For instance, treatment of DCs with melanoma-conditioned medium reduced expression of IL-12, MHC class II, and CD40 due to the increased induction of STAT3 [117]. The immunosuppressive effects of tumor-derived factors on DC differentiation were abrogated in cells from STAT3−/− mice and by blocking downstream STAT activation in DC precursors. STAT3 is also involved in the IL-6-mediated regulation of DC activation and maturation [118], the pathway which play an important role in cell interactions in the tumor microenvironment. In fact, it has been recently reported that phosphorylation of STAT3 in DCs by IL-6-producing tumor cells resulted in acquisition of their tolerogenic phenotype [119]. Interestingly, CD4+CD25+FoxP3+ regulatory T cells from tumor-bearing animals could hamper DC function by activating STAT3 and inducing the Smad signaling pathway [120], which was also associated with down-regulation of activation of the transcription factor NF-κB, required TGF-β and IL-10 and resulted in strong inhibition of expression of the co-stimulatory molecules CD80, CD86 and CD40, the production of TNF-α, IL-12, and CCL5/RANTES by DCs. Our data revealed that tumor-induced polarization of immunostimulatory cDCs into immunosuppressive regDCs in vitro was also accompanied by fast activation of STAT3 in treated cells (Shurin, unpublished data). Interestingly, histone deacetylase inhibition has been also reported to alter DCs to assume a tolerogenic phenotype [121]. Recently, Sun et al. demonstrated that histone deacetylase inhibition acetylates and activates STAT-3, which regulates DCs by promoting the transcription of IDO. These findings demonstrate a novel functional role for posttranslational modification of STAT-3 through acetylation [122].

Other members of the STAT family also play a role in DC differentiation and maturation. For example, the STAT6-mediated signaling is constitutively activated in immature DCs and declines as they differentiate into mature DCs, in contrast to STAT1 signaling, which is most robust in mature DCs and required for the efficient antigen presentation [123]. Down-regulation of STAT6 pathway is accompanied by remarkable induction of suppressors of cytokine signaling 1 (SOCS1), SOCS2 and SOCS3 [124]. Therefore, it is possible that cytokine-induced maturation of DCs is under feedback regulation by SOCS proteins and that the switch from activated STAT6 pathway in immature DCs to predominant utilization of STAT1 signals in mature DCs is mediated in part by STAT1-induced SOCS expression [124]. On the other hand, SOCS1 functions as an antigen-presentation attenuator by controlling the tolerogenic state of DCs and the magnitude of antigen presentation [125]. Since SOCS1 restricts DC ability to break self-tolerance and induce anti-tumor immunity by regulating IL-12 production and signaling, it is likely that tumor-derived and/or stroma-derived factors might induce SOCS1 expression in DCs the tumor milieu. Although not proven experimentally, this pathway may operate in the tumor microenvironment limiting the ability of DC to process and present tumor antigens and secrete IL-12 or attenuate tolerogenic potential of tumor-associated DCs.

Recent data from Wang et al. suggest that tumor-induced p38 mitogen-activated protein kinase (MAPK) and Janus kinase (JNK) activation and extracellular regulated kinase (ERK) inhibition in DCs are involved in disregulated DC functioning in cancer patients [126, 127]. Additional studies confirmed these findings and revealed that selective ERK activation induced mouse and human DCs to secrete bioactive TGF-β required for suppression of T cell responses and differentiation of antigen-specific Treg cells [128]. Zhao et al. reported a critical role of constitutively activated p38 MAPK in the acquirement of tolerogenic pattern by DCs during melanoma progression that contributes to the suppression of anti-tumor T cell immune responses [129].

Furthermore, based on the previous data demonstrating that key functions of cDCs are regulated by the family of small Rho GTPases (Cdc42, RhoA and Rac1/2) [130], Tourkova et al. determined whether small Rho GTPases might be affected by tumor-derived factors. They found that impaired endocytic activity of cDCs co-cultured with tumor cells was associated with decreased levels of active Cdc42 and Rac1 [131]. Transduction of DCs with the dominant negative Cdc42 and Rac1 genes also lead to reduced phagocytosis and receptor-mediated endocytosis, while transduction of DCs with the constitutively active Cdc42 and Rac1 genes restored endocytic activity of DCs that were inhibited by the tumors [131]. Following these results, we have recently revealed that polarization of cDCs into immunosuppressive regDCs in tumor-treated cultures can be prevented by toxin B, an inhibitor of small Rho GTPase activity (Zhong et al., submitted). Altogether, these data demonstrate that specific intracellular signal transduction pathways in DCs are responsible for DC differentiation and polarization in the tumor microenvironment that render tolerogenic and immunosuppressive properties. However, it is still unknown whether the involvement of these pathways is tumor-specific or DC subset-specific. Additional studies are required to evaluate the biological significance of multiple signaling activities in regDCs and determine specificity of their activation or suppression in different types of cancer both in vitro and in vivo.

Regulatory dendritic cells: Inhibitory pathways

Tolerogenic and immunosuppressive properties of regDCs are mediated by either direct effects of regDCs on effector T cells or by the induction or activation of other immune regulatory cells, such as Treg cells and MDSCs. Several soluble regDC-derived factors and membrane-bound or intracellular molecules have been revealed to be responsible for these activities. Production of IL-10 and TGF-βby regDCs has been well established and reviewed [132, 133] and the role of these cytokines in polarization and function of regulatory T cells has been also repeatedly discussed elsewhere. Functionally active expression of COX-2 and iNOS in tumor-associated regDCs has been also documented [134], suggesting additional mechanisms of T cell suppression by regDCs. Furthermore, expression of other enzymes in regDC, including arginase and IDO, is also accounted for their immunosuppressive properties.

The enzyme arginase metabolizes L-arginine to L-ornithine and urea. Besides its fundamental role in the hepatic urea cycle, arginase is also expressed in the immune system of mice and man. Myeloid cell arginase-mediated L-arginine depletion profoundly suppresses T cell immune responses and this has emerged as a fundamental mechanism of inflammation-associated immunosuppression [135]. Influence of L-arginine deficiency on the function of T lymphocytes is mediated by down-regulation of the T cell receptor (TCR) ζ chain, a critical signaling element of the TCR, and an arrest of T cells in the G0–G1 phase of the cell cycle, associated with the absence of up-regulated cyclin D3 and cyclin-dependent kinase 4 (cdk4) [135]. Expression of arginase by MDSC has been proven as a key mechanism of MDSC-induced immunosuppression in cancer [136]. However, evidence of arginase expression in tumor-associated regDCs is limited by a very few papers showing that tumor-infiltrating regDCs can inhibit CD8+ T cell function via L-arginine metabolism [88, 134]. Additional results are required to establish the role of this pathway in regDC-mediated immune unresponsiveness in cancer.

Indoleamine-2,3,dioxygenase (IDO) is the rate-limiting enzyme in the tryptophan catabolism, also known as the kynurenine pathway. It degrades the essential amino acid tryptophan thereby leading to an accumulation of its metabolites, the kynurenine [56]. Tryptophan is required for protein synthesis and for the synthesis of serotonin in the nervous system and the gut, as well as for melatonin synthesis in the pineal gland. Lately, IDO, was first reported to be implicated in the inhibition of viruses and intracellular pathogens, whose survival depends on the host tryptophan [137139]. More recently, IDO has been associated with immune tolerance, as the reduction of tryptophan might prevent T cell proliferation and the elevation of metabolites of the tryptophan catabolism might exert toxic effects on immune cells [140, 141]. Since then, numerous reports revealed the role of IDO activation and subsequent tryptophan depletion in the regulation of immune responses during infections and tumorigenesis [142144]. Expression of IDO in DCs was documented and implicated to DC-induced immunosuppression [145, 146]. These studies suggest that IDO-expressing regDCs found at the tumor site and in tumor-draining lymph nodes might help suppress the initiation of immune responses to tumor-derived antigens and perhaps help create systemic tolerance to these antigens [132]. Interestingly, human chorionic gonadotropin (hCG), which serves as an important tumor marker for trophoblastic disease, has been recently shown to up-regulate expression of IDO in DCs [147].

The ability of tumor-derived factors to bias DC differentiation toward providing inhibitory signals to interacting T cells should also be instrumental in tumor immune evasion. For instance, a dominant pathway of immune suppression in ovarian cancer involves tumor-associated and DC-associated B7-H1. The interaction of B7-H1 with PD-1 on tumor-infiltrating T cells is an acceptable theory of immune suppression involving B7-H1 in ovarian cancer [148]. Recent studies suggest that the B7-H1 ligand, programmed death receptor-1 (PD-1), is also expressed on myeloid cells, complicating interpretations of how B7-H1 regulates DC function in the tumor. Krempski et al. have recently found that ovarian cancer-infiltrating DCs progressively expressed increased levels of PD-1 over time in addition to B7-H1 [149]. These PD-1+ B7-H1+ DCs had a classical DC phenotype, but were immature and immunosuppressive. Accumulation of PD-1+ B7-H1+ regDCs in the tumor was associated with suppression of T cell activity and decreased infiltrating T cells in advancing tumors [149].

Tumor-infiltrating DCs may be polarize to express programmed death ligand 1 (PD-L1), a known inhibitor of T cell proliferation. The immunohistochemical analysis of 109 non-small cell lung cancer (NSCLC) tissues and demonstrated that immature CD1a+ DCs express PDL1 and that PD-L1 might be regarded as a poor prognostic factor [150]. Therefore, different mechanisms of immunosuppression are associated with the functional activity of regulatory DCs in the tumor environment. Understanding of these pathways and revealing of how they can co-interact in regDCs should provide additional insights in developing the effective therapeutic approaches to their blockade.

Conclusions

A growing body of evidence reveals DCs with tolerogenic and immunosuppressive activities within different cell subsets, including immature and mature myeloid cells, conventional DCs and pDCs. This suggests that DCs with “tolerogenic” function may be present in specific environments under specific conditions due to unusual differentiational and functional flexibility of DCs. Tumor-induced polarization and differentiation of cDCs and pDCs into immature or mature DCs with the inhibitory and tolerogenic function strongly impair antigen-specific T cell responses. For patients with cancer, the resulting dysfunction or misbalance of the DC system would result in significant deficiency in the induction of anti-tumor immunity, tumor progression and low response to immunotherapy and, probably, other treatment modalities. Given that resident DCs in patients with cancer might be also important for fulfilling the potential of various cancer vaccines, gained knowledge of immunobiology of regulatory DCs in cancer should help find new drugs and agents to selectively block the immunosuppressive pathways and restore the original immunostimulatory function of DCs in the tumor environment.

The term “regulatory” DCs is commonly used interchangeably with “tolerogenic” DCs, creating massive controversy in understanding the nature of DCs with immunosuppressive function, as has been purposely demonstrated in this review. The multiple discrepancies and controversies in phenotypic and functional characterization of the so-called “regulatory” DCs suggest that the use of this term is excessively abused and non-standardized. In fact, conflicting reports further compound the difficulty in understanding the biology of DC subsets with immunosuppressive and/or tolerogenic properties. As has been recently suggested, “One should only assign the term “tolerogenic” to DCs that have been proven to induce immunological tolerance or, at the very least, encourage the formation of functional Tregs under various experimental and pathophysiological conditions. Similarly, to slow the growing perplexity, the term “regulatory” should be restricted to DC subsets with experimentally established immunosuppressive activity in the tumor microenvironment.” [133].

Footnotes

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

Conflict of Interest statement: The authors declare that there are no conflicts of interest.

REFERENCES

  • [1].Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991;9:271–96. doi: 10.1146/annurev.iy.09.040191.001415. [DOI] [PubMed] [Google Scholar]
  • [2].Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
  • [3].Manicassamy S, Pulendran B. Dendritic cell control of tolerogenic responses. Immunol Rev. 2011;241:206–27. doi: 10.1111/j.1600-065X.2011.01015.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Lutz MB, Schuler G. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity? Trends Immunol. 2002;23:445–9. doi: 10.1016/s1471-4906(02)02281-0. [DOI] [PubMed] [Google Scholar]
  • [5].Mahnke K, Schmitt E, Bonifaz L, Enk AH, Jonuleit H. Immature, but not inactive: the tolerogenic function of immature dendritic cells. Immunol Cell Biol. 2002;80:477–83. doi: 10.1046/j.1440-1711.2002.01115.x. [DOI] [PubMed] [Google Scholar]
  • [6].Reizis B, Colonna M, Trinchieri G, Barrat F, Gilliet M. Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Nat Rev Immunol. 2011;11:558–65. doi: 10.1038/nri3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Vermi W, Soncini M, Melocchi L, Sozzani S, Facchetti F. Plasmacytoid dendritic cells and cancer. J Leukoc Biol. 2011;90:681–90. doi: 10.1189/jlb.0411190. [DOI] [PubMed] [Google Scholar]
  • [8].Bjorck P, Leong HX, Engleman EG. Plasmacytoid dendritic cell dichotomy: identification of IFN-alpha producing cells as a phenotypically and functionally distinct subset. J Immunol. 2011;186:1477–85. doi: 10.4049/jimmunol.1000454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Lin A, Schildknecht A, Nguyen LT, Ohashi PS. Dendritic cells integrate signals from the tumor microenvironment to modulate immunity and tumor growth. Immunol Lett. 2010;127:77–84. doi: 10.1016/j.imlet.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • [10].Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–26. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
  • [11].Palucka K, Ueno H, Fay J, Banchereau J. Dendritic cells and immunity against cancer. J Intern Med. 2011;269:64–73. doi: 10.1111/j.1365-2796.2010.02317.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Esche C, Lokshin A, Shurin GV, Gastman BR, Rabinowich H, Watkins SC, Lotze MT, Shurin MR. Tumor's other immune targets: dendritic cells. J Leukoc Biol. 1999;66:336–44. doi: 10.1002/jlb.66.2.336. [DOI] [PubMed] [Google Scholar]
  • [13].Shurin MR. Regulation of dendropoiesis in cancer. Clin Immunol Newsletter. 1999;19:135–9. [Google Scholar]
  • [14].Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metastasis Rev. 2006;25:333–56. doi: 10.1007/s10555-006-9010-6. [DOI] [PubMed] [Google Scholar]
  • [15].Pinzon-Charry A, Maxwell T, Lopez JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83:451–61. doi: 10.1111/j.1440-1711.2005.01371.x. [DOI] [PubMed] [Google Scholar]
  • [16].Alcalay J, Kripke ML. Antigen-presenting activity of draining lymph node cells from mice painted with a contact allergen during ultraviolet carcinogenesis. J Immunol. 1991;146:1717–21. [PubMed] [Google Scholar]
  • [17].Halliday GM, Reeve VE, Barnetson RS. Langerhans cell migration into ultraviolet light-induced squamous skin tumors is unrelated to anti-tumor immunity. J Invest Dermatol. 1991;97:830–4. doi: 10.1111/1523-1747.ep12491503. [DOI] [PubMed] [Google Scholar]
  • [18].Halliday GM, Lucas AD, Barnetson RS. Control of Langerhans' cell density by a skin tumour-derived cytokine. Immunology. 1992;77:13–8. [PMC free article] [PubMed] [Google Scholar]
  • [19].Tas MP, Simons PJ, Balm FJ, Drexhage HA. Depressed monocyte polarization and clustering of dendritic cells in patients with head and neck cancer: in vitro restoration of this immunosuppression by thymic hormones. Cancer Immunol Immunother. 1993;36:108–14. doi: 10.1007/BF01754410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Gabrilovich DI, Nadaf S, Corak J, Berzofsky JA, Carbone DP. Dendritic cells in antitumor immune responses. II. Dendritic cells grown from bone marrow precursors, but not mature DC from tumor-bearing mice, are effective antigen carriers in the therapy of established tumors. Cell Immunol. 1996;170:111–9. doi: 10.1006/cimm.1996.0140. [DOI] [PubMed] [Google Scholar]
  • [21].Chaux P, Moutet M, Faivre J, Martin F, Martin M. Inflammatory cells infiltrating human colorectal carcinomas express HLA class II but not B7–1 and B7–2 costimulatory molecules of the T-cell activation. Lab Invest. 1996;74:975–83. [PubMed] [Google Scholar]
  • [22].Ninomiya T, Akbar SM, Masumoto T, Horiike N, Onji M. Dendritic cells with immature phenotype and defective function in the peripheral blood from patients with hepatocellular carcinoma. J Hepatol. 1999;31:323–31. doi: 10.1016/s0168-8278(99)80231-1. [DOI] [PubMed] [Google Scholar]
  • [23].Sakakura K, Chikamatsu K, Takahashi K, Whiteside TL, Furuya N. Maturation of circulating dendritic cells and imbalance of T-cell subsets in patients with squamous cell carcinoma of the head and neck. Cancer Immunol Immunother. 2006;55:151–9. doi: 10.1007/s00262-005-0697-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Maecker B, Mougiakakos D, Zimmermann M, Behrens M, Hollander S, Schrauder A, Schrappe M, Welte K, Klein C. Dendritic cell deficiencies in pediatric acute lymphoblastic leukemia patients. Leukemia. 2006;20:645–9. doi: 10.1038/sj.leu.2404146. [DOI] [PubMed] [Google Scholar]
  • [25].Ormandy LA, Farber A, Cantz T, Petrykowska S, Wedemeyer H, Horning M, Lehner F, Manns MP, Korangy F, Greten TF. Direct ex vivo analysis of dendritic cells in patients with hepatocellular carcinoma. World J Gastroenterol. 2006;12:3275–82. doi: 10.3748/wjg.v12.i20.3275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Wojas K, Tabarkiewicz J, Jankiewicz M, Rolinski J. Dendritic cells in peripheral blood of patients with breast and lung cancer--a pilot study. Folia Histochem Cytobiol. 2004;42:45–8. [PubMed] [Google Scholar]
  • [27].Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, Clerici M, Greco M, Villa ML. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003;89:1463–72. doi: 10.1038/sj.bjc.6601243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Bellik L, Gerlini G, Parenti A, Ledda F, Pimpinelli N, Neri B, Pantalone D. Role of conventional treatments on circulating and monocyte-derived dendritic cells in colorectal cancer. Clin Immunol. 2006;121:74–80. doi: 10.1016/j.clim.2006.06.011. [DOI] [PubMed] [Google Scholar]
  • [29].Kiertscher SM, Luo J, Dubinett SM, Roth MD. Tumors promote altered maturation and early apoptosis of monocyte-derived dendritic cells. J Immunol. 2000;164:1269–76. doi: 10.4049/jimmunol.164.3.1269. [DOI] [PubMed] [Google Scholar]
  • [30].Yang T, Witham TF, Villa L, Erff M, Attanucci J, Watkins S, Kondziolka D, Okada H, Pollack IF, Chambers WH. Glioma-associated hyaluronan induces apoptosis in dendritic cells via inducible nitric oxide synthase: implications for the use of dendritic cells for therapy of gliomas. Cancer Res. 2002;62:2583–91. [PubMed] [Google Scholar]
  • [31].Peguet-Navarro J, Sportouch M, Popa I, Berthier O, Schmitt D, Portoukalian J. Gangliosides from human melanoma tumors impair dendritic cell differentiation from monocytes and induce their apoptosis. J Immunol. 2003;170:3488–94. doi: 10.4049/jimmunol.170.7.3488. [DOI] [PubMed] [Google Scholar]
  • [32].Katsenelson NS, Shurin GV, Bykovskaia SN, Shogan J, Shurin MR. Human small cell lung carcinoma and carcinoid tumor regulate dendritic cell maturation and function. Mod Pathol. 2001;14:40–5. doi: 10.1038/modpathol.3880254. [DOI] [PubMed] [Google Scholar]
  • [33].Onishi H, Morisaki T, Baba E, Kuga H, Kuroki H, Matsumoto K, Tanaka M, Katano M. Dysfunctional and short-lived subsets in monocyte-derived dendritic cells from patients with advanced cancer. Clin Immunol. 2002;105:286–95. doi: 10.1006/clim.2002.5293. [DOI] [PubMed] [Google Scholar]
  • [34].Pinzon-Charry A, Maxwell T, McGuckin MA, Schmidt C, Furnival C, Lopez JA. Spontaneous apoptosis of blood dendritic cells in patients with breast cancer. Breast Cancer Res. 2006;8:R5. doi: 10.1186/bcr1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Aalamian M, Pirtskhalaishvili G, Nunez A, Esche C, Shurin GV, Huland E, Huland H, Shurin MR. Human prostate cancer regulates generation and maturation of monocyte-derived dendritic cells. Prostate. 2001;46:68–75. doi: 10.1002/1097-0045(200101)46:1<68::aid-pros1010>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • [36].Brown RD, Pope B, Murray A, Esdale W, Sze DM, Gibson J, Ho PJ, Hart D, Joshua D. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7-1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood. 2001;98:2992–8. doi: 10.1182/blood.v98.10.2992. [DOI] [PubMed] [Google Scholar]
  • [37].Shurin GV, Aalamian M, Pirtskhalaishvili G, Bykovskaia S, Huland E, Huland H, Shurin MR. Human prostate cancer blocks the generation of dendritic cells from CD34+ hematopoietic progenitors. Eur Urol. 2001;39(Suppl 4):37–40. doi: 10.1159/000052584. [DOI] [PubMed] [Google Scholar]
  • [38].Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P, Oliviero B, Fogli M, Ferri E, Della Cuna GR, Tura S, Baccarani M, Lemoli RM. Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. Blood. 2002;100:230–7. doi: 10.1182/blood.v100.1.230. [DOI] [PubMed] [Google Scholar]
  • [39].Satthaporn S, Robins A, Vassanasiri W, El-Sheemy M, Jibril JA, Clark D, Valerio D, Eremin O. Dendritic cells are dysfunctional in patients with operable breast cancer. Cancer Immunol Immunother. 2004;53:510–8. doi: 10.1007/s00262-003-0485-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kichler-Lakomy C, Budinsky AC, Wolfram R, Hellan M, Wiltschke C, Brodowicz T, Viernstein H, Zielinski CC. Deficiences in phenotype expression and function of dentritic cells from patients with early breast cancer. Eur J Med Res. 2006;11:7–12. [PubMed] [Google Scholar]
  • [41].Fricke I, Gabrilovich DI. Dendritic cells and tumor microenvironment: a dangerous liaison. Immunol Invest. 2006;35:459–83. doi: 10.1080/08820130600803429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Yang L, Carbone DP. Tumor-host immune interactions and dendritic cell dysfunction. Adv Cancer Res. 2004;92:13–27. doi: 10.1016/S0065-230X(04)92002-7. [DOI] [PubMed] [Google Scholar]
  • [43].Enk AH, Jonuleit H, Saloga J, Knop J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma. Int J Cancer. 1997;73:309–16. doi: 10.1002/(sici)1097-0215(19971104)73:3<309::aid-ijc1>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • [44].Lizee G, Radvanyi LG, Overwijk WW, Hwu P. Improving antitumor immune responses by circumventing immunoregulatory cells and mechanisms. Clin Cancer Res. 2006;12:4794–803. doi: 10.1158/1078-0432.CCR-06-0944. [DOI] [PubMed] [Google Scholar]
  • [45].Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, Rivera M, Ravetch JV, Steinman RM, Nussenzweig MC. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med. 2001;194:769–79. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Kim R, Emi M, Tanabe K. Functional roles of immature dendritic cells in impaired immunity of solid tumour and their targeted strategies for provoking tumour immunity. Clin Exp Immunol. 2006;146:189–96. doi: 10.1111/j.1365-2249.2006.03215.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].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;192:1213–22. doi: 10.1084/jem.192.9.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, Roncarolo MG. Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+CD4+ Tr cells. Blood. 2005;105:1162–9. doi: 10.1182/blood-2004-03-1211. [DOI] [PubMed] [Google Scholar]
  • [49].Ghiringhelli F, Puig PE, Roux S, Parcellier A, Schmitt E, Solary E, Kroemer G, Martin F, Chauffert B, Zitvogel L. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J Exp Med. 2005;202:919–29. doi: 10.1084/jem.20050463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Akbari O, DeKruyff RH, Umetsu DT. Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol. 2001;2:725–31. doi: 10.1038/90667. [DOI] [PubMed] [Google Scholar]
  • [51].Menges M, Rossner S, Voigtlander C, Schindler H, Kukutsch NA, Bogdan C, Erb K, Schuler G, Lutz MB. Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce antigen-specific protection of mice from autoimmunity. J Exp Med. 2002;195:15–21. doi: 10.1084/jem.20011341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].McGuirk P, McCann C, Mills KH. Pathogen-specific T regulatory 1 cells induced in the respiratory tract by a bacterial molecule that stimulates interleukin 10 production by dendritic cells: a novel strategy for evasion of protective T helper type 1 responses by Bordetella pertussis. J Exp Med. 2002;195:221–31. doi: 10.1084/jem.20011288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Shortman K, Heath WR. Immunity or tolerance? That is the question for dendritic cells. Nat Immunol. 2001;2:988–9. doi: 10.1038/ni1101-988. [DOI] [PubMed] [Google Scholar]
  • [54].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;297:1867–70. doi: 10.1126/science.1073514. [DOI] [PubMed] [Google Scholar]
  • [55].Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004;4:762–74. doi: 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
  • [56].Grohmann U, Fallarino F, Puccetti P. Tolerance, DCs and tryptophan: much ado about IDO. Trends Immunol. 2003;24:242–8. doi: 10.1016/s1471-4906(03)00072-3. [DOI] [PubMed] [Google Scholar]
  • [57].Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol. 2005;23:275–306. doi: 10.1146/annurev.immunol.23.021704.115633. [DOI] [PubMed] [Google Scholar]
  • [58].Asselin-Paturel C, Boonstra A, Dalod M, Durand I, Yessaad N, Dezutter-Dambuyant C, Vicari A, O'Garra A, Biron C, Briere F, Trinchieri G. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat Immunol. 2001;2:1144–50. doi: 10.1038/ni736. [DOI] [PubMed] [Google Scholar]
  • [59].Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, Chu FF, Randolph GJ, Rudensky AY, Nussenzweig M. In vivo analysis of dendritic cell development and homeostasis. Science. 2009;324:392–7. doi: 10.1126/science.1170540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Naik SH, Sathe P, Park HY, Metcalf D, Proietto AI, Dakic A, Carotta S, O'Keeffe M, Bahlo M, Papenfuss A, Kwak JY, Wu L, Shortman K. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol. 2007;8:1217–26. doi: 10.1038/ni1522. [DOI] [PubMed] [Google Scholar]
  • [61].Cella M, Jarrossay D, Facchetti F, Alebardi O, Nakajima H, Lanzavecchia A, Colonna M. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat Med. 1999;5:919–23. doi: 10.1038/11360. [DOI] [PubMed] [Google Scholar]
  • [62].Yoneyama H, Matsuno K, Zhang Y, Nishiwaki T, Kitabatake M, Ueha S, Narumi S, Morikawa S, Ezaki T, Lu B, Gerard C, Ishikawa S, Matsushima K. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int Immunol. 2004;16:915–28. doi: 10.1093/intimm/dxh093. [DOI] [PubMed] [Google Scholar]
  • [63].Matta BM, Castellaneta A, Thomson AW. Tolerogenic plasmacytoid DC. Eur J Immunol. 2010;40:2667–76. doi: 10.1002/eji.201040839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Decalf J, Fernandes S, Longman R, Ahloulay M, Audat F, Lefrerre F, Rice CM, Pol S, Albert ML. Plasmacytoid dendritic cells initiate a complex chemokine and cytokine network and are a viable drug target in chronic HCV patients. J Exp Med. 2007;204:2423–37. doi: 10.1084/jem.20070814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Penna G, Vulcano M, Roncari A, Facchetti F, Sozzani S, Adorini L. Cutting edge: differential chemokine production by myeloid and plasmacytoid dendritic cells. J Immunol. 2002;169:6673–6. doi: 10.4049/jimmunol.169.12.6673. [DOI] [PubMed] [Google Scholar]
  • [66].Schlecht G, Garcia S, Escriou N, Freitas AA, Leclerc C, Dadaglio G. Murine plasmacytoid dendritic cells induce effector/memory CD8+ T-cell responses in vivo after viral stimulation. Blood. 2004;104:1808–15. doi: 10.1182/blood-2004-02-0426. [DOI] [PubMed] [Google Scholar]
  • [67].Mouries J, Moron G, Schlecht G, Escriou N, Dadaglio G, Leclerc C. Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation. Blood. 2008;112:3713–22. doi: 10.1182/blood-2008-03-146290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Villadangos JA, Young L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity. 2008;29:352–61. doi: 10.1016/j.immuni.2008.09.002. [DOI] [PubMed] [Google Scholar]
  • [69].Hoeffel G, Ripoche AC, Matheoud D, Nascimbeni M, Escriou N, Lebon P, Heshmati F, Guillet JG, Gannage M, Caillat-Zucman S, Casartelli N, Schwartz O, De la Salle H, Hanau D, Hosmalin A, Maranon C. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity. 2007;27:481–92. doi: 10.1016/j.immuni.2007.07.021. [DOI] [PubMed] [Google Scholar]
  • [70].Jego G, Pascual V, Palucka AK, Banchereau J. Dendritic cells control B cell growth and differentiation. Curr Dir Autoimmun. 2005;8:124–39. doi: 10.1159/000082101. [DOI] [PubMed] [Google Scholar]
  • [71].Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6:836–48. doi: 10.1038/nri1961. [DOI] [PubMed] [Google Scholar]
  • [72].Chaperot L, Blum A, Manches O, Lui G, Angel J, Molens JP, Plumas J. Virus or TLR agonists induce TRAIL-mediated cytotoxic activity of plasmacytoid dendritic cells. J Immunol. 2006;176:248–55. doi: 10.4049/jimmunol.176.1.248. [DOI] [PubMed] [Google Scholar]
  • [73].Zou W, Machelon V, Coulomb-L'Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, Pustilnik T, Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001;7:1339–46. doi: 10.1038/nm1201-1339. [DOI] [PubMed] [Google Scholar]
  • [74].Vermi W, Bonecchi R, Facchetti F, Bianchi D, Sozzani S, Festa S, Berenzi A, Cella M, Colonna M. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas. J Pathol. 2003;200:255–68. doi: 10.1002/path.1344. [DOI] [PubMed] [Google Scholar]
  • [75].Wilke CM, Kryczek I, Zou W. Antigen-presenting cell (APC) subsets in ovarian cancer. Int Rev Immunol. 2011;30:120–6. doi: 10.3109/08830185.2011.567362. [DOI] [PubMed] [Google Scholar]
  • [76].Charles J, Di Domizio J, Salameire D, Bendriss-Vermare N, Aspord C, Muhammad R, Lefebvre C, Plumas J, Leccia MT, Chaperot L. Characterization of circulating dendritic cells in melanoma: role of CCR6 in plasmacytoid dendritic cell recruitment to the tumor. J Invest Dermatol. 2010;130:1646–56. doi: 10.1038/jid.2010.24. [DOI] [PubMed] [Google Scholar]
  • [77].Hartmann E, Wollenberg B, Rothenfusser S, Wagner M, Wellisch D, Mack B, Giese T, Gires O, Endres S, Hartmann G. Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer. Cancer Res. 2003;63:6478–87. [PubMed] [Google Scholar]
  • [78].Labidi-Galy SI, Sisirak V, Meeus P, Gobert M, Treilleux I, Bajard A, Combes JD, Faget J, Mithieux F, Cassignol A, Tredan O, Durand I, Menetrier-Caux C, Caux C, Blay JY, Ray-Coquard I, Bendriss-Vermare N. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011;71:5423–34. doi: 10.1158/0008-5472.CAN-11-0367. [DOI] [PubMed] [Google Scholar]
  • [79].Thiel A, Pries R, Jeske S, Trenkle T, Wollenberg B. Effect of head and neck cancer supernatant and CpG-oligonucleotides on migration and IFN-alpha production of plasmacytoid dendritic cells. Anticancer Res. 2009;29:3019–25. [PubMed] [Google Scholar]
  • [80].Watkins SK, Zhu Z, Riboldi E, Shafer-Weaver KA, Stagliano KE, Sklavos MM, Ambs S, Yagita H, Hurwitz AA. FOXO3 programs tumor-associated DCs to become tolerogenic in human and murine prostate cancer. J Clin Invest. 2011;121:1361–72. doi: 10.1172/JCI44325. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [81].Treilleux I, Blay JY, Bendriss-Vermare N, Ray-Coquard I, Bachelot T, Guastalla JP, Bremond A, Goddard S, Pin JJ, Barthelemy-Dubois C, Lebecque S. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res. 2004;10:7466–74. doi: 10.1158/1078-0432.CCR-04-0684. [DOI] [PubMed] [Google Scholar]
  • [82].Cao W, Bover L. Signaling and ligand interaction of ILT7: receptor-mediated regulatory mechanisms for plasmacytoid dendritic cells. Immunol Rev. 2010;234:163–76. doi: 10.1111/j.0105-2896.2009.00867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Cai D, Cao J, Li Z, Zheng X, Yao Y, Li W, Yuan Z. Up-regulation of bone marrow stromal protein 2 (BST2) in breast cancer with bone metastasis. BMC Cancer. 2009;9:102. doi: 10.1186/1471-2407-9-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].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;117:2570–82. doi: 10.1172/JCI31911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ito T, Yang M, Wang YH, Lande R, Gregorio J, Perng OA, Qin XF, Liu YJ, Gilliet M. Plasmacytoid dendritic cells prime IL-10-producing T regulatory cells by inducible costimulator ligand. J Exp Med. 2007;204:105–15. doi: 10.1084/jem.20061660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Chen W, Liang X, Peterson AJ, Munn DH, Blazar BR. The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J Immunol. 2008;181:5396–404. doi: 10.4049/jimmunol.181.8.5396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Muller AJ, Prendergast GC. Indoleamine 2,3-dioxygenase in immune suppression and cancer. Curr Cancer Drug Targets. 2007;7:31–40. doi: 10.2174/156800907780006896. [DOI] [PubMed] [Google Scholar]
  • [88].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;69:3086–94. doi: 10.1158/0008-5472.CAN-08-2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Jahrsdorfer B, Vollmer A, Blackwell SE, Maier J, Sontheimer K, Beyer T, Mandel B, Lunov O, Tron K, Nienhaus GU, Simmet T, Debatin KM, Weiner GJ, Fabricius D. Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion. Blood. 2010;115:1156–65. doi: 10.1182/blood-2009-07-235382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].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;174:915–24. doi: 10.1084/jem.174.4.915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O'Garra A. IL-10 inhibits cytokine production by activated macrophages. J Immunol. 1991;147:3815–22. [PubMed] [Google Scholar]
  • [92].Willems F, Marchant A, Delville JP, Gerard C, Delvaux A, Velu T, de Boer M, Goldman M. Interleukin-10 inhibits B7 and intercellular adhesion molecule-1 expression on human monocytes. Eur J Immunol. 1994;24:1007–9. doi: 10.1002/eji.1830240435. [DOI] [PubMed] [Google Scholar]
  • [93].Takayama T, Morelli AE, Onai N, Hirao M, Matsushima K, Tahara H, Thomson AW. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability. J Immunol. 2001;166:7136–43. doi: 10.4049/jimmunol.166.12.7136. [DOI] [PubMed] [Google Scholar]
  • [94].Steinbrink K, Wolfl M, Jonuleit H, Knop J, Enk AH. Induction of tolerance by IL-10-treated dendritic cells. J Immunol. 1997;159:4772–80. [PubMed] [Google Scholar]
  • [95].Boks MA, Kager-Groenland JR, Haasjes MS, Zwaginga JJ, van Ham SM, Ten Brinke A. IL-10-generated tolerogenic dendritic cells are optimal for functional regulatory T cell induction - A comparative study of human clinical-applicable DC. Clin Immunol. 2011 doi: 10.1016/j.clim.2011.11.011. in press. [DOI] [PubMed] [Google Scholar]
  • [96].Kryczek I, Wei S, Zou L, Zhu G, Mottram P, Xu H, Chen L, Zou W. Cutting edge: induction of B7–H4 on APCs through IL-10: novel suppressive mode for regulatory T cells. J Immunol. 2006;177:40–4. doi: 10.4049/jimmunol.177.1.40. [DOI] [PubMed] [Google Scholar]
  • [97].Fischer JR, Darjes H, Lahm H, Schindel M, Drings P, Krammer PH. Constitutive secretion of bioactive transforming growth factor beta 1 by small cell lung cancer cell lines. Eur J Cancer. 1994;30A:2125–9. doi: 10.1016/0959-8049(94)00364-b. [DOI] [PubMed] [Google Scholar]
  • [98].Rodeck U, Bossler A, Graeven U, Fox FE, Nowell PC, Knabbe C, Kari C. Transforming growth factor beta production and responsiveness in normal human melanocytes and melanoma cells. Cancer Res. 1994;54:575–81. [PubMed] [Google Scholar]
  • [99].Kobie JJ, Wu RS, Kurt RA, Lou S, Adelman MK, Whitesell LJ, Ramanathapuram LV, Arteaga CL, Akporiaye ET. Transforming growth factor beta inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines. Cancer Res. 2003;63:1860–4. [PubMed] [Google Scholar]
  • [100].Lan YY, Wang Z, Raimondi G, Wu W, Colvin BL, de Creus A, Thomson AW. “Alternatively activated” dendritic cells preferentially secrete IL-10, expand Foxp3+CD4+ T cells, and induce long-term organ allograft survival in combination with CTLA4-Ig. J Immunol. 2006;177:5868–77. doi: 10.4049/jimmunol.177.9.5868. [DOI] [PubMed] [Google Scholar]
  • [101].Sato K, Yamashita N, Baba M, Matsuyama T. Regulatory dendritic cells protect mice from murine acute graft-versus-host disease and leukemia relapse. Immunity. 2003;18:367–79. doi: 10.1016/s1074-7613(03)00055-4. [DOI] [PubMed] [Google Scholar]
  • [102].Azzaoui I, Yahia SA, Chang Y, Vorng H, Morales O, Fan Y, Delhem N, Ple C, Tonnel AB, Wallaert B, Tsicopoulos A. CCL18 differentiates dendritic cells in tolerogenic cells able to prime regulatory T cells in healthy subjects. Blood. 2011;118:3549–58. doi: 10.1182/blood-2011-02-338780. [DOI] [PubMed] [Google Scholar]
  • [103].Mellor AL, Baban B, Chandler PR, Manlapat A, Kahler DJ, Munn DH. Cutting edge: CpG oligonucleotides induce splenic CD19+ dendritic cells to acquire potent indoleamine 2,3-dioxygenase-dependent T cell regulatory functions via IFN Type 1 signaling. J Immunol. 2005;175:5601–5. doi: 10.4049/jimmunol.175.9.5601. [DOI] [PubMed] [Google Scholar]
  • [104].Rigas B, Goldman IS, Levine L. Altered eicosanoid levels in human colon cancer. J Lab Clin Med. 1993;122:518–23. [PubMed] [Google Scholar]
  • [105].Benoit V, Relic B, Leval Xd X, Chariot A, Merville MP, Bours V. Regulation of HER-2 oncogene expression by cyclooxygenase-2 and prostaglandin E2. Oncogene. 2004;23:1631–5. doi: 10.1038/sj.onc.1207295. [DOI] [PubMed] [Google Scholar]
  • [106].Pockaj BA, Basu GD, Pathangey LB, Gray RJ, Hernandez JL, Gendler SJ, Mukherjee P. Reduced T-cell and dendritic cell function is related to cyclooxygenase-2 overexpression and prostaglandin E2 secretion in patients with breast cancer. Ann Surg Oncol. 2004;11:328–39. doi: 10.1245/aso.2004.05.027. [DOI] [PubMed] [Google Scholar]
  • [107].Chemnitz JM, Driesen J, Classen S, Riley JL, Debey S, Beyer M, Popov A, Zander T, Schultze JL. Prostaglandin E2 impairs CD4+ T cell activation by inhibition of lck: implications in Hodgkin's lymphoma. Cancer Res. 2006;66:1114–22. doi: 10.1158/0008-5472.CAN-05-3252. [DOI] [PubMed] [Google Scholar]
  • [108].Akasaki Y, Liu G, Chung NH, Ehtesham M, Black KL, Yu JS. Induction of a CD4+ T regulatory type 1 response by cyclooxygenase-2-overexpressing glioma. J Immunol. 2004;173:4352–9. doi: 10.4049/jimmunol.173.7.4352. [DOI] [PubMed] [Google Scholar]
  • [109].Baratelli F, Lin Y, Zhu L, Yang SC, Heuze-Vourc'h N, Zeng G, Reckamp K, Dohadwala M, Sharma S, Dubinett SM. Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. J Immunol. 2005;175:1483–90. doi: 10.4049/jimmunol.175.3.1483. [DOI] [PubMed] [Google Scholar]
  • [110].Sharma S, Yang SC, Zhu L, Reckamp K, Gardner B, Baratelli F, Huang M, Batra RK, Dubinett SM. Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+ CD25+ T regulatory cell activities in lung cancer. Cancer Res. 2005;65:5211–20. doi: 10.1158/0008-5472.CAN-05-0141. [DOI] [PubMed] [Google Scholar]
  • [111].Kalinski P, Vieira PL, Schuitemaker JH, de Jong EC, Kapsenberg ML. Prostaglandin E(2) is a selective inducer of interleukin-12 p40 (IL-12p40) production and an inhibitor of bioactive IL-12p70 heterodimer. Blood. 2001;97:3466–9. doi: 10.1182/blood.v97.11.3466. [DOI] [PubMed] [Google Scholar]
  • [112].von Bergwelt-Baildon MS, Popov A, Saric T, Chemnitz J, Classen S, Stoffel MS, Fiore F, Roth U, Beyer M, Debey S, Wickenhauser C, Hanisch FG, Schultze JL. CD25 and indoleamine 2,3-dioxygenase are up-regulated by prostaglandin E2 and expressed by tumor-associated dendritic cells in vivo: additional mechanisms of T-cell inhibition. Blood. 2006;108:228–37. doi: 10.1182/blood-2005-08-3507. [DOI] [PubMed] [Google Scholar]
  • [113].Wilczynski JR. Cancer and pregnancy share similar mechanisms of immunological escape. Chemotherapy. 2006;52:107–10. doi: 10.1159/000092537. [DOI] [PubMed] [Google Scholar]
  • [114].Mouillot G, Marcou C, Zidi I, Guillard C, Sangrouber D, Carosella ED, Moreau P. Hypoxia Modulates HLA-G Gene Expression in Tumor Cells. Hum Immunol. 2007;68:277–85. doi: 10.1016/j.humimm.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • [115].Ristich V, Liang S, Zhang W, Wu J, Horuzsko A. Tolerization of dendritic cells by HLA-G. Eur J Immunol. 2005;35:1133–42. doi: 10.1002/eji.200425741. [DOI] [PubMed] [Google Scholar]
  • [116].Chang CC, Ciubotariu R, Manavalan JS, Yuan J, Colovai AI, Piazza F, Lederman S, Colonna M, Cortesini R, Dalla-Favera R, Suciu-Foca N. Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol. 2002;3:237–43. doi: 10.1038/ni760. [DOI] [PubMed] [Google Scholar]
  • [117].Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, Bhattacharya R, Gabrilovich D, Heller R, Coppola D, Dalton W, Jove R, Pardoll D, Yu H. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004;10:48–54. doi: 10.1038/nm976. [DOI] [PubMed] [Google Scholar]
  • [118].Park SJ, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa S, Kamimura D, Ueda N, Iwakura Y, Ishihara K, Murakami M, Hirano T. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J Immunol. 2004;173:3844–54. doi: 10.4049/jimmunol.173.6.3844. [DOI] [PubMed] [Google Scholar]
  • [119].Alshamsan A. Induction of tolerogenic dendritic cells by IL-6-secreting CT26 colon carcinoma. Immunopharmacol Immunotoxicol. 2012 doi: 10.3109/08923973.2011.625034. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • [120].Larmonier N, Marron M, Zeng Y, Cantrell J, Romanoski A, Sepassi M, Thompson S, Chen X, Andreansky S, Katsanis E. Tumor-derived CD4(+)CD25(+) regulatory T cell suppression of dendritic cell function involves TGF-beta and IL-10. Cancer Immunol Immunother. 2007;56:48–59. doi: 10.1007/s00262-006-0160-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Misaki K, Morinobu A, Saegusa J, Kasagi S, Fujita M, Miyamoto Y, Matsuki F, Kumagai S. Histone deacetylase inhibition alters dendritic cells to assume a tolerogenic phenotype and ameliorates arthritis in SKG mice. Arthritis Res Ther. 2011;13:R77. doi: 10.1186/ar3339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Sun Y, Chin YE, Weisiger E, Malter C, Tawara I, Toubai T, Gatza E, Mascagni P, Dinarello CA, Reddy P. Cutting edge: Negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3. J Immunol. 2009;182:5899–903. doi: 10.4049/jimmunol.0804388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Pilz A, Kratky W, Stockinger S, Simma O, Kalinke U, Lingnau K, von Gabain A, Stoiber D, Sexl V, Kolbe T, Rulicke T, Muller M, Decker T. Dendritic cells require STAT-1 phosphorylated at its transactivating domain for the induction of peptide-specific CTL. J Immunol. 2009;183:2286–93. doi: 10.4049/jimmunol.0901383. [DOI] [PubMed] [Google Scholar]
  • [124].Jackson SH, Yu CR, Mahdi RM, Ebong S, Egwuagu CE. Dendritic cell maturation requires STAT1 and is under feedback regulation by suppressors of cytokine signaling. J Immunol. 2004;172:2307–15. doi: 10.4049/jimmunol.172.4.2307. [DOI] [PubMed] [Google Scholar]
  • [125].Evel-Kabler K, Song XT, Aldrich M, Huang XF, Chen SY. SOCS1 restricts dendritic cells' ability to break self tolerance and induce antitumor immunity by regulating IL-12 production and signaling. J Clin Invest. 2006;116:90–100. doi: 10.1172/JCI26169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Wang S, Yang J, Qian J, Wezeman M, Kwak LW, Yi Q. Tumor evasion of the immune system: inhibiting p38 MAPK signaling restores the function of dendritic cells in multiple myeloma. Blood. 2006;107:2432–9. doi: 10.1182/blood-2005-06-2486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Wang S, Hong S, Yang J, Qian J, Zhang X, Shpall E, Kwak LW, Yi Q. Optimizing immunotherapy in multiple myeloma: Restoring the function of patients' monocyte-derived dendritic cells by inhibiting p38 or activating MEK/ERK MAPK and neutralizing interleukin-6 in progenitor cells. Blood. 2006;108:4071–7. doi: 10.1182/blood-2006-04-016980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Arce F, Breckpot K, Stephenson H, Karwacz K, Ehrenstein MR, Collins M, Escors D. Selective ERK activation differentiates mouse and human tolerogenic dendritic cells, expands antigen-specific regulatory T cells, and suppresses experimental inflammatory arthritis. Arthritis Rheum. 2011;63:84–95. doi: 10.1002/art.30099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Zhao F, Falk C, Osen W, Kato M, Schadendorf D, Umansky V. Activation of p38 mitogen-activated protein kinase drives dendritic cells to become tolerogenic in ret transgenic mice spontaneously developing melanoma. Clin Cancer Res. 2009;15:4382–90. doi: 10.1158/1078-0432.CCR-09-0399. [DOI] [PubMed] [Google Scholar]
  • [130].Shurin GV, Tourkova IL, Chatta GS, Schmidt G, Wei S, Djeu JY, Shurin MR. Small rho GTPases regulate antigen presentation in dendritic cells. J Immunol. 2005;174:3394–400. doi: 10.4049/jimmunol.174.6.3394. [DOI] [PubMed] [Google Scholar]
  • [131].Tourkova IL, Shurin GV, Wei S, Shurin MR. Small Rho GTPases Mediate Tumor-induced Inhibition of endocytic Activity of Dendritic Cells. J Immunol. 2007;178:7787–93. doi: 10.4049/jimmunol.178.12.7787. [DOI] [PubMed] [Google Scholar]
  • [132].Shurin MR, Naiditch H, Zhong H, Shurin GV. Regulatory dendritic cells: new targets for cancer immunotherapy. Cancer Biol Ther. 2011;11:988–92. doi: 10.4161/cbt.11.11.15543. [DOI] [PubMed] [Google Scholar]
  • [133].Shurin GV, Ouellette CE, Shurin MR. Regulatory dendritic cells in the tumor immunoenvironment. Cancer Immunol Immunother. 2012;61:223–30. doi: 10.1007/s00262-011-1138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Liu Q, Zhang C, Sun A, Zheng Y, Wang L, Cao X. Tumor-educated CD11bhighIalow regulatory dendritic cells suppress T cell response through arginase I. J Immunol. 2009;182:6207–16. doi: 10.4049/jimmunol.0803926. [DOI] [PubMed] [Google Scholar]
  • [135].Munder M. Arginase: an emerging key player in the mammalian immune system. Br J Pharmacol. 2009;158:638–51. doi: 10.1111/j.1476-5381.2009.00291.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer J. 2010;16:348–53. doi: 10.1097/PPO.0b013e3181eb3358. [DOI] [PubMed] [Google Scholar]
  • [137].Yoshida R, Urade Y, Tokuda M, Hayaishi O. Induction of indoleamine 2,3-dioxygenase in mouse lung during virus infection. Proc Natl Acad Sci U S A. 1979;76:4084–6. doi: 10.1073/pnas.76.8.4084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Pfefferkorn ER. Interferon gamma blocks the growth of Toxoplasma gondii in human fibroblasts by inducing the host cells to degrade tryptophan. Proc Natl Acad Sci U S A. 1984;81:908–12. doi: 10.1073/pnas.81.3.908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Byrne GI, Lehmann LK, Landry GJ. Induction of tryptophan catabolism is the mechanism for gamma-interferon-mediated inhibition of intracellular Chlamydia psittaci replication in T24 cells. Infect Immun. 1986;53:347–51. doi: 10.1128/iai.53.2.347-351.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Moffett JR, Namboodiri MA. Tryptophan and the immune response. Immunol Cell Biol. 2003;81:247–65. doi: 10.1046/j.1440-1711.2003.t01-1-01177.x. [DOI] [PubMed] [Google Scholar]
  • [141].MacKenzie CR, Heseler K, Muller A, Daubener W. Role of indoleamine 2,3-dioxygenase in antimicrobial defence and immuno-regulation: tryptophan depletion versus production of toxic kynurenines. Curr Drug Metab. 2007;8:237–44. doi: 10.2174/138920007780362518. [DOI] [PubMed] [Google Scholar]
  • [142].Mellor AL, Keskin DB, Johnson T, Chandler P, Munn DH. Cells expressing indoleamine 2,3-dioxygenase inhibit T cell responses. J Immunol. 2002;168:3771–6. doi: 10.4049/jimmunol.168.8.3771. [DOI] [PubMed] [Google Scholar]
  • [143].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;11:312–9. doi: 10.1038/nm1196. [DOI] [PubMed] [Google Scholar]
  • [144].Popov A, Abdullah Z, Wickenhauser C, Saric T, Driesen J, Hanisch FG, Domann E, Raven EL, Dehus O, Hermann C, Eggle D, Debey S, Chakraborty T, Kronke M, Utermohlen O, Schultze JL. Indoleamine 2,3-dioxygenase-expressing dendritic cells form suppurative granulomas following Listeria monocytogenes infection. J Clin Invest. 2006;116:3160–70. doi: 10.1172/JCI28996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114:280–90. doi: 10.1172/JCI21583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Kwon HK, Lee CG, So JS, Chae CS, Hwang JS, Sahoo A, Nam JH, Rhee JH, Hwang KC, Im SH. Generation of regulatory dendritic cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune disorders. Proc Natl Acad Sci U S A. 2010;107:2159–64. doi: 10.1073/pnas.0904055107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Ueno A, Cho S, Cheng L, Wang J, Hou S, Nakano H, Santamaria P, Yang Y. Transient upregulation of IDO in dendritic cells by human chorionic gonadotropin downregulates autoimmune diabetes. Diabetes. 2007;56:1686–93. doi: 10.2337/db06-1727. [DOI] [PubMed] [Google Scholar]
  • [148].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;9:562–7. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • [149].Krempski J, Karyampudi L, Behrens MD, Erskine CL, Hartmann L, Dong H, Goode EL, Kalli KR, Knutson KL. Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer. J Immunol. 2011;186:6905–13. doi: 10.4049/jimmunol.1100274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol. 2011;28:682–8. doi: 10.1007/s12032-010-9515-2. [DOI] [PubMed] [Google Scholar]

RESOURCES