Abstract
The fact that the immune response to cancer is compromised has been convincingly demonstrated in murine tumor models as well as in cancer patients. The unresponsiveness of the host immune system is one of the major mechanisms of tumor escape as well as an important factor that limits the success of cancer immunotherapy. Inadequate function of professional antigen presenting cells dendritic cells (DC) in cancer is one of the major elements of compromised anti-tumor immune response. Despite substantial progress in recent years, the mechanism of inadequate DC function in cancer still remains unclear. The tumor microenvironment has emerged as an important component contributing to DC malfunction. In this review we will discuss the potential role of tumor microenvironment in DC dysfunction.
Introduction
Dendritic cells (DC) are considered the most powerful antigen presenting cells (APC) and therefore viewed as critical regulators of adaptive immune responses. DCs are a highly heterogenic population of cells with high plasticity sharing common features like morphology and functional characteristics (Banchereau, et al. 1998, Lanzavecchia, et al. 2001). DCs arise from CD34+ bone-marrow stem cells and are classified in two different developmental stages: immature (iDC) and mature (mDC) cells. iDC are localized primarily in peripheral tissues and perform the specialized functions of antigen uptake and processing. On the other hand, mDCs reside in lymphoid organs, where they interact with antigen-specific T cells and initiate immune responses. The chemokine receptor repertoire of iDCs is distinct from mDCs, and this regulates their migration into tissue sites.
Two main populations of DCs are recognized in mouse and human tissues: myeloid DCs (MDCs) and plasmacytoid DCs (PDCs) (Colonna, et al. 2004, Kaech, et al. 2001, Lanzavecchia, et al. 2001, O’Neill, et al. 2004). Early work in humans identified plasmacytoid monocytes (now called plasmacytoid DCs [PDCs]) as cells present in inflamed lymph nodes surrounding high endothelial venules (Facchetti, et al. 1989). These cells have a characteristic phenotype distinct from that of monocytes and myeloid DCs (CD123+, CD45RA+, CD4+, CD11c−, ILT3+, ILT1−, Lin−) and are present at low levels in peripheral blood (where they express additional markers BDCA-2 and 4) (Dzionek, et al. 2000, Grouard, et al. 1997, Olweus, et al. 1997). Experiments using cultured human stem cells, as well as retroviral transduction with genes that interfere with lymphocyte development, indicate that PDCs may have a lymphoid origin (Blom, et al. 2000, Spits, et al. 2000). Recent studies indicate that freshly isolated PDCs stimulated by viruses produce extremely high levels of type I IFNs (Cella, et al. 1999, Siegal, et al. 1999) and selectively prime T cells to produce IFN-γ and IL-10 (i.e. cells with a TH1-like phenotype) (Cella, et al. 2000, Kadowaki, et al. 2000). In humans, the expression of Toll-like receptors TLR7 and TLR9 are restricted to PDC (Edwards, et al. 2003). The path of PDC development and their molecular regulation are not completely understood. The main cytokine required for the development of PDC from hematopoietic stem cells is Fms-like Tyrosine Kinase 3 Ligand (Flt3L) (Blom, et al. 2000, Chen, et al. 2004, Gilliet, et al. 2002). Administration of Flt3L to human volunteers resulted in an increase of PDCs in peripheral blood (Pulendran, et al. 2000). Additionally, Flt3L-deficient mice displayed a smaller amount of PDC whereas Flt3L-transgenic mice had increased levels (Brawand, et al. 2002). Granulocyte colony stimulating factor is also important in PDC biology because it promotes their mobilization from bone marrow (Arpinati, et al. 2000, Pulendran, et al. 2000).
Myeloid DC are characterized through their expression of CD11c, CD33 and absence of CD45RA, CD123 and lineage marker (Kadowaki, et al. 2001, Sieling, et al. 2002). Furthermore, human MDC express a different TLR-repertoire (TLR1, 2, 3, 4, 5, 6, 8, 10) and do not secrete interferon I after viral challenge. According to phenotypical and functional characteristics MDC can be further divided into subpopulations (Grabbe, et al. 2000). Previously, it was believed that PDC (also called DC2) induce a TH2 response, whereas MDC (also termed DC1) induce a TH1 type response (Rissoan, et al. 1999, Vieira, et al. 2000). Now it is accepted that DC are capable of promoting either TH1-, TH2- or TH0/TR1 responses depending on culture conditions and/or origin of activation signal rather than on the ontogeny of DC (Kalinski, et al. 1999).
In early studies it was believed that DCs were exclusively immunogenic, actively initiating or up-regulating immune responses. More recent reports demonstrated that DCs may cover dual functions, and can also exhibit regulatory (suppressive) activity. DCs are able to actively down-regulate an immune response or to induce immune tolerance by influencing the activity of other cell types. It has been seen particularly within the tumor microenvironment that DCs acquire a regulatory function (Curiel, et al. 2004, Curiel, et al. 2003, Gabrilovich, et al. 1996, Munn, et al. 2004a, Zou, et al. 2001). This regulatory capacity is probably not a part of intrinsic nature of a defined DC subset but rather the result of the influence of tumor microenvironment.
Myeloid DCs share the same progenitor cells as monocytes/macrophages, and express myeloid lineage surface markers (Banchereau, et al. 1998). Mature MDCs are potent inducers of T helper 1 (TH1)-type immune responses and also are considered as powerful initiators of tumor associated antigen (TAA)-specific immunity. However, the appearance of functional competent myeloid mDCs inside the tumor is rare as described for human ovarian (Zou, et al. 2001), breast (Bell, et al. 1999, Iwamoto, et al. 2003), prostate (Troy, et al. 1998a) and renal-cell cancers (Troy, et al. 1998b). Multiple factors may be responsible for this phenomenon, such as defective DC recruitment, differentiation, maturation, and survival. There are strong data demonstrating profoundly suppressed differentiation and maturation of myeloid DCs in the presence of factors present in the tumor microenvironment. Tumor cells are able to produce large amounts of vascular endothelial growth factor (VEGF) among other factors (Carmeliet, et al. 2000, Kryczek, et al. 2005). Lack or low levels of IL-12 and/or IFN-γ create an aberrant cytokine pattern in the tumor environment (Freedman, et al. 2004, Kryczek, et al. 2000). The imbalance of cytokines in the tumor microenvironment impairs DC differentiation and maturation. While in vivo myeloid mDCs can induce strong TAA-directed immunity (Labeur, et al. 1999), immature or partially differentiated MDCs induce either T-cell unresponsiveness (Dhodapkar, et al. 1999, Hawiger, et al. 2001) or suppressive regulatory T cells (Treg) (Dhodapkar, et al. 2001, Jonuleit, et al. 2000). Homing of suppressive Treg cells to draining lymph nodes may induce systemically disabled TAA-specific immunity. Incomplete differentiated or immature myeloid DCs therefore can be regarded as regulatory DCs and may function as an important component of the immunosuppressive networks in the tumor microenvironment.
General characteristics of tumor microenvironment and potential role of dendritic cells
Solid tumor is not solely composed of malignant cells, but also includes several non-malignant cell types including endothelial cells, fibroblasts, adipocytes, lymphocytes, macrophages, granulocytes, and immature myeloid cells. This creates a unique microenvironment, which can modify the neoplastic properties of the tumor cells. Studies have shown that the nature of the surrounding cells can modify the outcome of primary oncogenic events in epithelial cells (Coussens, et al. 2000, Coussens, et al. 2002, Iyengar, et al. 2003, Krtolica, et al. 2001, Lin, et al. 2001). It was noted that within the tumor, primarily iDC not mDC (Troy, et al. 1998a, Troy, et al. 1998b) were present while mDC were detected in the marginal zones (Sandel, et al. 2005). Furthermore, labeling experiments revealed that most of the intratumoral DCs remain inside the tumor instead of migrating out (Feijoo, et al. 2005).
An important factor for the tumor development is angiogenesis, a function widely attributed to factors produced by macrophages (Carmeliet, et al. 2000). This process involves a wide range of soluble mediators that are both stimulatory and inhibitory, including basic fibroblast growth factor (bFGF), VEGF, the angiopoietins (ANG1 and ANG2), IL-1, IL-8, tumor necrosis factor-α (TNF-α), thymidine phosphorylase (TP; also known as vascular-derived endothelial growth factor), the matrix metalloproteinases MMP-9 and MMP-2, and nitric oxide (NO) (Boudreau, et al. 2003, Carmeliet, et al. 2000). These molecules, which are expressed in a coordinated spatial and temporal fashion, result in the proliferation and migration of endothelial cells, matrix remodeling and the eventual formation of stabilized vessels (Hanahan, et al. 1996). Macrophages are perfectly designed to promote these processes, as their precursors can migrate into sites where they differentiate into macrophages. These cells can synthesize the required angiogenic molecules on demand in specific locations (Bando, et al. 2000).
Human myeloid DCs, but not plasmacytoid DCs, are the major producers of IL-12. This fact may contrubute to the suppression of tumor neoangiogenesis by myeloid DCs (Curiel, et al. 2004), which is essential for tumor growth and metastasis (Carmeliet, et al. 2000). However, tumor environment seem to lack angiogenesis-inhibitory myeloid DCs, whereas angiogenesis-stimulatory DCs, such as plasmacytoid DCs are present (Conejo-Garcia, et al. 2004, Curiel, et al. 2004). This could be due to the tumor-derived chemokine CXCL12, which attracts and protects plasmacytoid DCs in the tumor microenvironment. These cells in turn are capable of inducing vascularization by spontaneously producing TNF-α and IL-8 (Curiel, et al. 2004). Coukos et al. demonstrated that vascular DCs are recruited by β-defensin, and induce vasculogenesis under the influence of VEGF in mice. Vascular DCs are also found in ovarian tumors (Conejo-Garcia, et al. 2004). Therefore, DCs are relevant not only in tumor immunopathogenesis, but also in tumor vascularization. Optimal vascularization of tumors might require the simultaneous accumulation of vascular DCs and the absence of anti-angiogenic myeloid DCs, as is observed in ovarian tumours. Furthermore, as recently observed, tumor vascularization and growth can also be mediated by DC precursors. These cells can be attracted to the tumor sited by β-defensins and under the influence of VEGF-A may differentiate into endothelial-like cells expressing both DC and endothelial cell markers (Conejo-Garcia, et al. 2004). Therefore, DCs are relevant not only in tumor immunopathogenesis, but also in tumor vascularization.
Modulating factors released by the tumor microenvironment
Ideally, DCs should be recruited to the tumor site to initiate and drive immune responses that would lead to the rejection of the tumor. However, there is now enough evidence indicating that the tumor microenvironment is immunosuppressive (Elgert, et al. 1998, Ohm, et al. 2001). A number of factors that contribute to this suppression are discussed below.
1. Cytokines/chemokines
GM-CSF (granulocyte macrophage colony stimulating factor)
Naba et al. reported that granulocytes, macrophages, and DCs can develop from a common MHC class-II-negative progenitor in the presence of GM-CSF (Inaba, et al. 1993). The concept that DCs can be derived from myeloid precursors was definitively shown by Sallusto and Lanzavecchia (Sallusto, et al. 1994) in a report indicating that human DCs can be differentiated in vitro from monocytes, and has been strengthened more recently by Randolph et al. using an in vivo approach in mice (Randolph, et al. 1999). GM-CSF is released by a large number of tumor cell lines (Bronte, et al. 1999). In mice, chronic exposure to GM-CSF leads to the generation of a cell population expressing the granulocyte and monocyte markers Gr-1 and CD11b (Bronte, et al. 1999, Young, et al. 1991). Most of these cells are immature myeloid cells able to directly suppress antigen-specific T-cell responses (Kusmartsev, et al. 2005). Interestingly, Gr-1+ CD11b+ cells isolated from tumor bearing mice could be differentiated into fully competent mature DC in the absence of tumor-conditioned medium. Furthermore it is important to note that that GM-CSF had been shown to have therapeutic potential as a component of cancer vaccines. The amount of GM-CSF released seems to be the important factor. Serafini et al. showed a strong accumulation of immunosuppressive immature myeloid cells (ImC) in animals vaccinated with a high GM-CSF producing tumor cell line (Serafini 2004).
M-CSF and IL-6
Macrophage colony-stimulating factor (M-CSF) and IL-6 are produced by a large number of tumors and have also been reported to be involved in the tumor-mediated regulation of DC differentiation (Gabrilovich, et al. 1996, Menetrier-Caux, et al. 1998). Renal-cell carcinoma cell lines were shown to release soluble factors that inhibit the differentiation of CD34+ progenitor cells into DCs and instead trigger their differentiation towards monocytic cells. Both neutralizing IL-6- and M-CSF-specific antibodies abolished the impact of renal-cell carcinoma conditioned medium on DC differentiation (1998, Menetrier-Caux, et al. 2001) and the combination of IL-6 and M-CSF displayed a similar effect on inhibition of DC differentiation (Menetrier-Caux, et al. 2001). IL-4 as well as IL-13 improved differentiation of CD34+ cells into DCs affected by the presence of either renal cell carcinoma conditioned medium, IL-6, or M-CSF. It was found that IL-4 rapidly blocked the expression of M-CSF and the IL-6R-transducing chain (gp130), as well as decreased the secondary production of M-CSF, thereby preventing the loss of GM-CSF receptor α-chain expression, which normally occurs during the differentiation of CD34+ cells (Menetrier-Caux, et al. 2001). IL-6 plays an important role in abnormal DC differentiation in multiple myeloma (Ratta, et al. 2002). Furthermore, sera from patients with multiple myeloma inhibited the generation of DCs, which could be reverted by anti-VEGF and/or anti-IL-6 antibodies (Hayashi, et al. 2003). In another recent study, IL-6 was found to suppress DC maturation in vivo and play a major role in maintaining immature DCs (Park, et al. 2004). The suppressive role of IL-6 could be attributed to activation of the transcription factor STAT3.
VEGF (Vascular endothelial growth factor)
VEGF was the first tumor-derived factor described to inhibit DC differentiation. VEGF plays an important role in neovascularization and hematopoiesis during embryogenesis. VEGF is produced by most tumors and increased plasma levels of VEGF in cancer patients correlate with an unfavorable prognosis (Ellis, et al. 1996, Toi, et al. 1996). Involvement of VEGF in tumor-induced defects in DC differentiation was demonstrated in vitro where abrogation of the negative effect of tumor cell conditioned medium on the differentiation of DCs could be achieved by neutralizing VEGF-specific antibodies (Gabrilovich, et al. 1998). These initial findings were confirmed in vivo by administration of recombinant VEGF to tumor-free mice resulting in inhibited DC development and association with increased production of Gr-1+ ImCs (Gabrilovich, et al. 1998). Furthermore, the stimulatory effect of Flt3L on DC production was also abrogated by VEGF (Ohm, et al. 1999). Consequently, in tumor-bearing mice the application of neutralizing VEGF-antibodies resulted in improved DC differentiation and increased number of mDCs (Gabrilovich, et al. 1999, Ishida, et al. 1998). Assembled clinical data supported the important role of VEGF on the observed DC dysfunction: expression of VEGF inversely correlated with DC numbers in tumor tissue (Saito, et al. 1998) and peripheral blood (Almand, et al. 2000, Lissoni, et al. 2001) of cancer patients. Additionally, for patients with gastric cancer or non-small-cell lung cancer, DC differentiation was demonstrated to be negatively affected by VEGF (Fan, et al. 2003, Takahashi 2004). A recent report attributes the inhibitory effect of VEGF on DC to Flt-1 signaling. In the presence of VEGF during culture, DC from wild-type mice displayed an inhibition of antigen presentation capacity, which was not observed for DC from Id1−/− mice under the same experimental settings. Furthermore, addition of neutralizing anti-Flt-1 antibody abrogated the inhibitory effect of VEGF on wild-type DCs (Laxmanan, et al. 2005).
IL-8 (CXCL8)
IL-8 acts as a direct autocrine growth factor for malignant melanoma (Schadendorf, et al. 1993), liver and pancreatic tumors (Miyamoto, et al. 1998), and for colon carcinoma cells (Brew, et al. 2000). Interestingly, in ovarian cancer, the receptors for CXCL8, namely CXCR1/2, have been shown to cross-talk with the epidermal growth factor receptor (EGFR) (Venkatakrishnan, et al. 2000), which links this growth factor pathway with chemokines. Besides its function as a growth factor, IL-8 also exerts chemotactic activity on polymorphonuclear neutrophils (PMNs) (Baggiolini 1998), monocytes (Bonecchi, et al. 2000) and endothelial cells (Schraufstatter, et al. 2003). Both receptors for the CXC chemokine IL-8, CXCR1 and CXCR2, are expressed on monocyte derived DCs. IL-8 was recently reported to be produced by a variety of tumors (hepatocellular carcinoma, colorectal, and pancreatic cancer), which attracts monocyte derived DCs. This effect could be blocked by neutralizing antibodies and the authors suggested involvement of IL-8 in the retention of DCs inside malignant lesions and impairment of DC migration toward lymphoid tissue (Feijoo, et al. 2005).
IL-10
IL-10 plays important role in DC defects in cancer. DCs derived from transgenic mice with IL-10 overexpression have suppressed ability to stimulate allogeneic T-cell and CTL responses as well as IL-12 production (Sharma, et al. 1999). IL-10 might contribute to the conversion of iDC into tolerogenic APCs by decreasing the expression of co-stimulatory molecules (Steinbrink, et al. 1997). Treatment of human DC with IL-10 was found to induce suppression of antigen-specific proliferation of CD4+ and CD8+ T cells via cell-cell contact (Steinbrink, et al. 2002). Furthermore, the blockade of differentiation of monocytes to DCs could be attributed to IL-10, which drives the differentiation process towards a macrophage cell type rather than DC (Allavena, et al. 1998, Buelens, et al. 1997). IL-10 also inhibits the function of Langerhans cells (Beissert, et al. 1995, Enk, et al. 1993, Peguet-Navarro, et al. 1994), monocyte derived DCs, or CD34+ progenitors (Caux, et al. 1994, Steinbrink, et al. 1997). A mouse tumor model revealed that tumor derived IL-10 was responsible for DC dysfunction in vivo. DC function was not affected in IL-10 deficient tumor bearing mice (Yang, et al. 2003). Even though different tumor cells might produce and release IL-10, the majority of IL-10 is probably produced by tumor-associated macrophages (TAM) with some contribution from tumor-infiltrating lymphocytes (Seo, et al. 2001, Sica, et al. 2000).
Gangliosides
Gangliosides are sialic-acid-containing glycosphingolipids that are implicated in the regulation of cellular proliferation and differentiation (Hakomori 2003). A broad range of different gangliosides have been detected in different tissues; including GD2, GD3, and GM3, which seem to be involved in tumor progression. In comparison to the corresponding normal tissues, an abnormal expression pattern of gangliosides was described for neuroblastoma, melanoma, leukemia, lymphoma, and breast tumors (Birkle, et al. 2003). Besides the surface expression on tumor cells gangliosides are also secreted and likely to circulate in the peripheral blood. Tumor-derived gangliosides can inhibit the generation of DCs from mouse bone-marrow progenitors or from human CD34+ precursors (Peguet-Navarro, et al. 2003, Shurin, et al. 2001).
TGF-β (transforming growth factor-β)
Cytokines of the TGF-β family are essential factors in embryonic development and tissue repair. This family includes three types of TGF-β (β1, β2 and β3), inhibins and activins, as well as various bone morphogenetic proteins (BMPs) and mullerian inhibiting substance. Activin βA and TGF-β1 share functions in inflammatory reactions including tissue repair and suppression (Munz, et al. 1999, Rosendahl, et al. 2001). Both cytokines share SMAD2/3 and SMAD4 as intracellular signalling targets of their receptors (Itoh, et al. 2000). In an adoptive transfer model TGF-β revealed its capability of inducing Treg by its ability to generate DCs that promote tolerance in a manner dependent on MHC class II molecules (Alard, et al. 2004). Specifically, generation of Treg cells was attributed to immature DCs, and TGF-β prevents the maturation of DCs by maintaining a low expression of co-stimulatory molecules (Geissmann, et al. 1999, Roncarolo, et al. 2001).
2. Altered glycosylation pattern of tumor associated antigens
MUC1 was shown to transform monocyte derived DC into IL-10high IL-12low producing cells with limited capacity to induce TH1 responses in T-cells. Furthermore, the DC lost the ability to fully mature and rendered T cells anergic or to become suppressor/regulatory cells (Monti, et al. 2004). Previously it was reported that O-glycosylation of MUC1 is responsible for preventing its proteolysis (Hanisch, et al. 2003) and escape of this molecule from processing and presentation (Hiltbold, et al. 2000). Recently, similar results were obtained with a recombinant produced MUC1, which was as strongly sialylated as that expressed by epithelial tumors. This recombinant MUC1 also induced high levels of IL-10 and low levels of IL-12 in monocyte derived DC as well as impaired their development. Furthermore, these DCs had a decreased ability to stimulate allogenic and autologous T cells (Rughetti, et al. 2005).
3. Reactive oxygen species
Reactive oxygen species (ROS) include superoxide, hydrogen peroxide (H2O2), hydroxyl radicals and a variety of their reaction products. Activation of NADPH oxidase, a multicomponent enzyme system which catalyses the NADPH-dependent reduction of oxygen to the superoxide anion (O2−), is the precursor of the other ROS. In resting cells, this multicomponent enzyme system is inactive, and its components are dispersed between the cytosol and the membranes. The flavocytochrome b558 component, which is composed of two subunits, gp91phox and p22phox, is located in the plasma membrane and in specific granules. The other components of the NADPH complex (p47phox, p67phox, p40phox and small G protein Rac2) are cytosol proteins. The activation triggers the phosphorylation of the p47phox, p67phox and p40phox cytosolic components and their translocation to the plasma membrane, where they interact with flavocytochrome b558 (Ago, et al. 1999, Babior 1999, Chanock, et al. 1994, Park, et al. 1992). Concomitantly, Rac2 is dissociated from its inhibitor, RhoGDI, and then interacts with flavocytochrome b558 to form a binding partner for p67phox (Bokoch, et al. 2002). The complete assembly of NADPH oxidase components is crucial for O2− production (Babior 2004).
While low levels of ROS are attributed to be important in proliferation of normal cells and regulation of cellular signaling (Burdon 1995), increased ROS were observed in cancer cells. The biological effects of ROS vary widely in different cells and include modulation of signaling pathways by directly altering the activity of protein kinases and protein phosphatases (PTPases) (Denu, et al. 1998, Monteiro, et al. 1996). Elevated ROS levels during tumor progression leads to constant activation of transcription factors (NFκB and AP-1). Furthermore, the accumulation of ROS in the tumor environment seems to have a strong impact on the development of immune cells infiltrating the tumor tissue by creating a milieu of constant oxidative stress. It is known that oxidative stress triggers expression of Heme oxygenase-1 (HO-1). During the maturation of DC in vitro, expression of HO-1 drastically decreases. Whereas in human tissue the expression is only found in iDC but not in mDC (Chauveau, et al. 2005). Furthermore, the authors could show that chemical-induced oxidative stress inhibited LPS-induced phenotypic maturation and secretion of proinflammatory cytokines, resulting in the inhibition of alloreactive T-cell proliferation, although retaining the ability of these DC to produce IL-10 (Chauveau, et al. 2005). As recently reported, only balanced NF-κB and JNK/AP-1 activity is associated with prolonged DC survival. Low NF-κB and high JNK/AP-1 activity signals cell death in DCs (Kriehuber, et al. 2005). Moreover, this study showed that limiting ROS loads is a major NF-κB function that results in the control of JNK activity in DCs. A current study showed that hydrogen peroxide, at low concentrations, activated p38 but did not alter DC phenotype (Handley, et al. 2005). However, increased concentrations of hydrogen peroxide activated both p38 and JNK, leading to inhibition of tyrosine phosphatases by JNK in DC and induction of apoptosis. JNK inhibitors partially protected the DC from the proapoptotic effects, whereas hydrogen peroxide and LPS synergized in inducing JNK activation and DC apoptosis (Handley, et al. 2005).
Peroxynitrites (ONOO−) are another category of ROS formed as a by-product of the reaction between O2− and NO. They represent an highly potent oxidizing agent that damages biological targets (Radi 2004, Schopfer, et al. 2003) and are able to cross membranes within or between cells faster than they decay (Denicola, et al. 1998). Peroxynitrites can therefore function as unusual intra- and intercellular messengers, because they can induce post-translational protein modifications including the nitration of tyrosine residues in proteins like transcription factors.
4. Indoleamine-2,3-deoxygenase
The expression of IDO (indoleamine-2,3-deoxygenase) in myeloid DC has been described for human as well as murine DC (Munn, et al. 2002). Appearance of IDO+ DC could be demonstrated in vivo in breast tumor tissue as well as draining lymph nodes in patients with melanoma, breast, colon, lung and pancreas cancers (Mellor, et al. 2004b). Catabolism of tryptophan is catalyzed by IDO via oxidation. Tryptophan is essential for proliferation and differentiation of T cells and IDO+ DC contribute to reduction of free environmental tryptophan. This may prevent the clonal expansion of T cells and promote T cell death by apoptosis, anergy, or immune deviation. In studies using immunohistochemistry to detect IDO protein expression in vivo, treatment with CTLA4–immunoglobulin fusion protein was found to upregulate the levels of immunoreactive IDO only by certain subsets of mouse APC in the spleen. This response was restricted mainly to cells in the B220+ (plasmacytoid) and CD8α+ populations of splenic DCs (Mellor, et al. 2003). Studies with APC fractions that were isolated from mice exposed to CTLA4–immunoglobulin confirmed that IDO-dependent T cell suppression was confined to specific DC subsets that express these markers (Mellor, et al. 2004a). Further evaluation will be needed to clarify whether these results describe a single ‘IDO-competent’ population of cells that expresses both markers (that is, B220+ CD8α+ DCs (O’Keeffe, et al. 2002)) or several different populations within the complex mixture of DC subtypes in the spleen. However, an important point is that many more cells express the target ligands for the CTLA4–immunoglobulin fusion protein than actually showed up-regulation of IDO expression. A detailed analysis of IDO-mediated T cell suppression in tumor-draining lymph nodes (Munn, et al. 2004b) showed that although a significant fraction of DCs expressed detectable levels of IDO protein by immunohistochemistry, the functional IDO-mediated suppression was mediated almost entirely by a small, well-defined CD19+ subset among the B220+ plasmacytoid DCs. Furthermore, in humans an IDO+ DC subset could be detected, which also expressed CD123 and CCR6 (Munn, et al. 2002). Whether this particular DC phenotype is a specialized feature of tumor-draining lymph nodes or is a more general phenomenon remains to be elucidated, but these data emphasize that the biologically relevant population of IDO-expressing DCs might be a minor subset. Even within the population of IDO-competent DCs there can be a considerable degree of functional plasticity. Certain pro-inflammatory signals might down-regulate the expression of IDO by cells that would normally express it (Grohmann, et al. 2003, Grohmann, et al. 2001). In contrast, different tolerogenic stimuli might induce IDO expression by different DC populations (Fallarino, et al. 2003, Grohmann, et al. 2003).
5. Stroma-mediated effects on DCs
Recently, it became obvious that the analysis of the tumor stroma is important to better understand tumor biology. At a certain stage of tumor progression, cancer cells start altering residing tissue cells and induce an inflammatory response in order to recruit stromal cells, which are required for tumor progression (Bissell, et al. 2001, Elenbaas, et al. 2001, Tlsty, et al. 2001, Wiseman, et al. 2002). The tumor stroma is a composition of those cell types necessary to built and sustain a tissue. It provides growth factors, blood supply, extracellular matrix, and removes waste and dead cells. The stroma can keep pre-malignant cells in check, and the phenotypically abnormal stroma can support tumor development. The latter point is illustrated by the recent demonstration that the loss of tumor growth factor-β responsiveness in fibroblasts resulted in epithelial neoplasia (Bhowmick, et al. 2004). The released factors also activate surrounding stromal cell types, such as fibroblasts, smooth-muscle cells (De Wever, et al. 2003), and adipocytes (Manabe, et al. 2003), leading to the secretion of additional growth factors and proteases. Concomitant with the altered growth-factor expression, and often induced by their autocrine effect on the tumor cells, cancer cells also start producing proteolytic enzymes (Mueller, et al. 2003, Stetler-Stevenson, et al. 2001), which remodel the extracellular matrix (ECM) and the basement membrane creating a pro-migratory and pro-invasive environment. In addition, degradation of ECM molecules exposes obscure protein domains and generates specific new molecule fragments that can have pro-migratory as well as pro- and anti-angiogenic functions (Kalluri 2003). The remodelled ECM contains matrix metalloproteinases (MMPs) (Brinckerhoff, et al. 2002), which activate cell-surface and ECM-bound growth factors (Egeblad, et al. 2002, McCawley, et al. 2001). These contribute to the extensive crosstalk between the microenvironment and the cancer cells. However, in benign and malignant tumor, no difference in the levels of protease expression was detected. Only when cultured in the presence of stromal fibroblasts did these features correlate with increased expression of MMP1 and MMP9. Interestingly, when co-cultured with stromal cells, only malignant, but not benign, tumor cells exhibited MMP1 and MMP9 production (Borchers, et al. 1997). Furthermore, transplantation of benign tumor cells to mice abolished MMP1 expression, whereas transplantation of tumor cells increased MMP1 expression. Tumor cells also induced production of interstitial collagenase by the host stroma, whereas benign cells did not (Airola, et al. 2001).
Because of these tremendous changes in the tumor stroma, it is conceivable that the stroma also exerts a negative effect on DC development. Recently, it was shown that spleen-derived stromal cells promote the selective development of lin− c-kit+ progenitor cells into CD11clowCD45RB+ regulatory DC, which primarily produce IL-10. These DC had the ability to suppress T cell responses and induce IL-10-producing regulatory T cells in vitro and antigen-specific tolerance in vivo (Svensson, et al. 2004). Furthermore, it could be demonstrated that contact with stromal cells promoted mature DCs to proliferate in a fibronectin-dependent way. Both stromal cell contact and stromal cell-derived transforming growth factor-β induced their differentiation into a new regulatory DC subset, whose in vivo counterpart could be identified in the spleen with similar phenotype and functions. These differentiated DCs secreted nitric oxide, which mediated the suppression of T cell proliferation in response to antigen presentation by mature DCs (Zhang, et al. 2004).
Molecular mechanism involved in DC dysfunction in cancer
Despite the wealth of information regarding various tumor-derived factors affecting DC differentiation, little is known about the molecular mechanisms responsible for DC dysfunction in cancer. One important family of proteins involved in the transition of the immature to mature stage of DC is the STAT family. A fundamental component of several signal-transduction pathways is the Janus activated kinase (JAK) family of tyrosine kinases and signal transducer and activator of transcription (STAT) proteins. These molecules are actively involved in cellular survival, proliferation, differentiation, and apoptosis. In mammals, four members of the JAK family are known (JAK1, JAK2, JAK3 and TYK2) (Rane, et al. 2000). Signal transduction by most of the factors implicated in defective DC function in cancer is mediated by JAKs. Receptor oligomerization induced by cytokine binding to its receptor triggers JAK activation by either auto- or transphosphorylation. The same set of JAKs can be activated by different cytokines though JAKs are not supreme determinants of the specificity of cytokine-mediated signalling (reviewed in Imada, et al. 2000). Subsequent to ligand binding, activated JAKs phosphorylate receptors on target tyrosine residues, generating docking sites for STATs through the STAT Src homology 2 (SH2) domain. Activated JAKs recruite and phosphorylate STATs, which leads to their dimerization and nuclear translocation, where they modulate the expression of target genes. The STAT transcription factor family is comprised of seven members (STAT1, -2, -3, -4, -5A, -5B and -6).
A number of cytokines involved in DC maturation transduce their extracellular signals to the nucleus through activated STAT proteins (Bromberg, et al. 2000, Darnell 1998) and the duration or intensity of the cytokine induced signal is under feedback regulation by a newly described eight-member family of intracellular proteins called suppressors of cytokine signaling (SOCS) (Naka, et al. 1999). SOCS proteins are characterized by the presence of an Src homology 2 domain and a C-terminal conserved domain called the SOCS box, and their inhibitory effects derives from direct interaction with JAKs, thereby preventing recruitment of STATs to the signaling complex (Naka, et al. 1999). SOCS proteins have recently been shown to modulate macrophage effector functions and negatively regulate LPS-induced macrophage activation (Baetz, et al. 2004, Berlato, et al. 2002, Kinjyo, et al. 2002, Nakagawa, et al. 2002), suggesting that they may also be involved in the regulation of DC differentiation and/or maturation.
In mouse DCs, STAT6 plays an important role in regulation of transition from the immature to mature stage. Activation of STAT6 signaling was not detected in freshly isolated precursor DCs (pDCs), while it was found to be constitutively expressed in iDC and its expression disappears in mDC accompanied by increased expression of SOCS1, SOCS2, SOCS3, and Src homology 2-containing protein (Jackson, et al. 2004). STAT6 expression as an early event of activation could also be demonstrated by immunohistochemistry in a subpopulation of human CD1a+ DC in rheumatoid arthritis (Walker, et al. 2005). STAT5 was shown to be constitutively activated at all stages of DC maturation, whereas STAT1 and STAT6 pathways are preferentially used at different stages of DC differentiation and maturation (Jackson, et al. 2004). However, STAT1 activation was detected at all stages of DC development. The tyrosine phosphorylated STAT1 protein was detected mainly in mDCs, implicating the requirement of a more robust STAT1 signaling pathway for functionally mature DCs. The complex function of STAT1 in DC development was explored by analyzing hematopoietic precursor cell (HPC) from STAT1 null mice differentiated into DC. Although STAT1 inhibits CD86 expression in cells at the pDC stage, its inhibitory effects were overridden in mDCs, presumably by maturation signals, such as LPS. In contrast, the STAT1 signaling pathway appears to have no effect on CD40 expression at the pDC stage, but is required for optimal expression of CD40 in mDCs. Contrary to the regulatory effects on CD86 and CD40 expression, STAT1 has no effect on MHC class II expression (Jackson, et al. 2004). Furthermore, STAT1 involvement could be demonstrated in TLR mediated maturation of DC (Hoshino, et al. 2002). Activation of human PDC by CpG was impaired by p38 mitogen-activated protein kinase (MAPK) inhibitor. In the presence of p38 inhibitor, the STAT1 phosphorylation of Y701 as well as S727 in activated PDC disappeared (Takauji, et al. 2002). However, up regulation of SOCS did not affect signaling of STAT1 suggesting that STAT6 and STAT1 are individually regulated during DC maturation (Jackson, et al. 2004).
In vivo, another critical factor for the regulation of DC maturation seems to be IL-6, because IL-6 knockout (KO) mice displayed increased amounts of mDC. Furthermore, IL-6 treatment of DC was shown to abrogate the LPS maturation of DC, which was mediated by its STAT3 activation. Additionally, STAT3 was required for the IL-6-mediated suppression of bone marrow-derived DC activation and/or maturation Thus the IL-6-gp130-STAT3 axis may represent an important regulatory component in DC activation/maturation mechanism (Park, et al. 2004).
Further evidence for the importance of the STAT3 to STAT1 switch on DC maturation was shown by in vitro experiments where overexpression of STAT3 through viral transfection of DC resulted in less IL-2 production by T cells/DC co-cultures (Cheng, et al. 2003). Interestingly, even the development of immature myeloid cells (ImC) into DC seems to be coupled to the transition of STAT3 levels to a more prominent STAT1 milieu. As it was shown in vitro, factors released by tumor cells strongly induce JAK2/STAT3 activation in ImC, which prevented their transition into DC. This blocking effect of the tumor supernatant could be reverted by adding antibodies against tumor-derived factors (Nefedova, et al. 2004).
Summary
Unfortunately, tumors are very effective in evading immune responses. One of the major mechanisms limiting immune recognition is suppression of immune responses. Further insights into the role of dendritic cells during the effector phase of the immune response and the complex interplay of stromal, immune, and tumor cells in the tumor microenvironment represent the future challenges to be conquered in tumor immunology. Gained knowledge in these areas may help to find new drugs to selectively block suppressive pathways and restore the original function of DC. Although the DC system is highly heterogeneous, the differentiation and function of DC populations is largely regulated by exogenous factors. A possible approch to overcome the tumor induced malfunction of DC may be to “override“ the tumor induced signals by a more prominant danger signal such as pathogen derived factors or peptides including CpG, bacterial cell wall component, or viral compounds to actively strenghten the immune response, an area for future research.
References
- Ago T, Nunoi H, Ito T, Sumimoto H. Mechanism for phosphorylation-induced activation of the phagocyte NADPH oxidase protein p47(phox). Triple replacement of serines 303, 304, and 328 with aspartates disrupts the SH3 domain-mediated intramolecular interaction in p47(phox), thereby activating the oxidase. J Biol Chem. 1999;274:33644–33653. doi: 10.1074/jbc.274.47.33644. [DOI] [PubMed] [Google Scholar]
- Airola K, Fusenig NE. Differential stromal regulation of MMP-1 expression in benign and malignant keratinocytes. J Invest Dermatol. 2001;116:85–92. doi: 10.1046/j.1523-1747.2001.00223.x. [DOI] [PubMed] [Google Scholar]
- Alard P, Clark SL, Kosiewicz MM. Mechanisms of tolerance induced by TGF beta-treated APC: CD4 regulatory T cells prevent the induction of the immune response possibly through a mechanism involving TGF beta. Eur J Immunol. 2004;34:1021–1030. doi: 10.1002/eji.200324547. [DOI] [PubMed] [Google Scholar]
- Allavena P, Piemonti L, Longoni D, Bernasconi S, Stoppacciaro A, Ruco L, Mantovani A. IL-10 prevents the differentiation of monocytes to dendritic cells but promotes their maturation to macrophages. Eur J Immunol. 1998;28:359–369. doi: 10.1002/(SICI)1521-4141(199801)28:01<359::AID-IMMU359>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
- Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Carbone DP, Gabrilovich DI. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res. 2000;6:1755–1766. [PubMed] [Google Scholar]
- Arpinati M, Green CL, Heimfeld S, Heuser JE, Anasetti C. Granulocyte-colony stimulating factor mobilizes T helper 2-inducing dendritic cells. Blood. 2000;95:2484–2490. [PubMed] [Google Scholar]
- Babior BM. NADPH oxidase. Curr Opin Immunol. 2004;16:42–47. doi: 10.1016/j.coi.2003.12.001. [DOI] [PubMed] [Google Scholar]
- Babior BM. NADPH oxidase: an update. Blood. 1999;93:1464–1476. [PubMed] [Google Scholar]
- Baetz A, Frey M, Heeg K, Dalpke AH. Suppressor of cytokine signaling (SOCS) proteins indirectly regulate toll-like receptor signaling in innate immune cells. J Biol Chem. 2004;279:54708–54715. doi: 10.1074/jbc.M410992200. [DOI] [PubMed] [Google Scholar]
- Baggiolini M. Chemokines and leukocyte traffic. Nature. 1998;392:565–568. doi: 10.1038/33340. [DOI] [PubMed] [Google Scholar]
- Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
- Bando H, Toi M. Tumor angiogenesis, macrophages, and cytokines. Adv Exp Med Biol. 2000;476:267–284. doi: 10.1007/978-1-4615-4221-6_21. [DOI] [PubMed] [Google Scholar]
- Beissert S, Hosoi J, Grabbe S, Asahina A, Granstein RD. IL-10 inhibits tumor antigen presentation by epidermal antigen-presenting cells. J Immunol. 1995;154:1280–1286. [PubMed] [Google Scholar]
- Bell D, Chomarat P, Broyles D, Netto G, Harb GM, Lebecque S, Valladeau J, Davoust J, Palucka KA, Banchereau J. In Breast Carcinoma Tissue, Immature Dendritic Cells Reside within the Tumor, whereas Mature Dendritic Cells Are Located in Peritumoral Areas. J Exp Med. 1999;190:1417–1426. doi: 10.1084/jem.190.10.1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berlato C, Cassatella MA, Kinjyo I, Gatto L, Yoshimura A, Bazzoni F. Involvement of suppressor of cytokine signaling-3 as a mediator of the inhibitory effects of IL-10 on lipopolysaccharide-induced macrophage activation. J Immunol. 2002;168:6404–6411. doi: 10.4049/jimmunol.168.12.6404. [DOI] [PubMed] [Google Scholar]
- Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, Washington MK, Neilson EG, Moses HL. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303:848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
- Birkle S, Zeng G, Gao L, Yu RK, Aubry J. Role of tumor-associated gangliosides in cancer progression. Biochimie. 2003;85:455–463. doi: 10.1016/s0300-9084(03)00006-3. [DOI] [PubMed] [Google Scholar]
- Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1:46–54. doi: 10.1038/35094059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blom B, Ho S, Antonenko S, Liu YJ. Generation of interferon alpha-producing predendritic cell (Pre-DC)2 from human CD34(+) hematopoietic stem cells. J Exp Med. 2000;192:1785–1796. doi: 10.1084/jem.192.12.1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bokoch GM, Diebold BA. Current molecular models for NADPH oxidase regulation by Rac GTPase. Blood. 2002;100:2692–2696. doi: 10.1182/blood-2002-04-1149. [DOI] [PubMed] [Google Scholar]
- Bonecchi R, Facchetti F, Dusi S, Luini W, Lissandrini D, Simmelink M, Locati M, Bernasconi S, Allavena P, Brandt E, Rossi F, Mantovani A, Sozzani S. Induction of functional IL-8 receptors by IL-4 and IL-13 in human monocytes. J Immunol. 2000;164:3862–3869. doi: 10.4049/jimmunol.164.7.3862. [DOI] [PubMed] [Google Scholar]
- Borchers AH, Steinbauer H, Schafer BS, Kramer M, Bowden GT, Fusenig NE. Fibroblast-directed expression and localization of 92-kDa type IV collagenase along the tumor-stroma interface in an in vitro three-dimensional model of human squamous cell carcinoma. Mol Carcinog. 1997;19:258–266. doi: 10.1002/(sici)1098-2744(199708)19:4<258::aid-mc7>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- Boudreau N, Myers C. Breast cancer-induced angiogenesis: multiple mechanisms and the role of the microenvironment. Breast Cancer Res. 2003;5:140–146. doi: 10.1186/bcr589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brawand P, Fitzpatrick DR, Greenfield BW, Brasel K, Maliszewski CR, De Smedt T. Murine plasmacytoid pre-dendritic cells generated from Flt3 ligand-supplemented bone marrow cultures are immature APCs. J Immunol. 2002;169:6711–6719. doi: 10.4049/jimmunol.169.12.6711. [DOI] [PubMed] [Google Scholar]
- Brew R, Erikson JS, West DC, Kinsella AR, Slavin J, Christmas SE. Interleukin-8 as an autocrine growth factor for human colon carcinoma cells in vitro. Cytokine. 2000;12:78–85. doi: 10.1006/cyto.1999.0518. [DOI] [PubMed] [Google Scholar]
- Brinckerhoff CE, Matrisian LM. Matrix metalloproteinases: a tail of a frog that became a prince. Nature Rev Mol Cell Biol. 2002;3:207–214. doi: 10.1038/nrm763. [DOI] [PubMed] [Google Scholar]
- Bromberg J, Darnell JE., Jr The role of STATs in transcriptional control and their impact on cellular function. Oncogene. 2000;19:2468–2473. doi: 10.1038/sj.onc.1203476. [DOI] [PubMed] [Google Scholar]
- Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu P, Restifo NP. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J Immunol. 1999;162:5728–5737. [PMC free article] [PubMed] [Google Scholar]
- Buelens C, Verhasselt V, De Groote D, Thielemans K, Goldman M, Willems F. Human dendritic cell responses to lipopolysaccharide and CD40 ligation are differentially regulated by interleukin-10. Eur J Immunol. 1997;27:1848–1852. doi: 10.1002/eji.1830270805. [DOI] [PubMed] [Google Scholar]
- Burdon RH. Superoxide and hydrogen peroxide in relation to mammalian cell proliferation. Free Radic Biol Med. 1995;18:775–794. doi: 10.1016/0891-5849(94)00198-s. [DOI] [PubMed] [Google Scholar]
- Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257. doi: 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
- Caux C, Massacrier C, Vanbervliet B, Barthelemy C, Liu YJ, Banchereau J. Interleukin 10 inhibits T cell alloreaction induced by human dendritic cells. Int Immunol. 1994;6:1177–1185. doi: 10.1093/intimm/6.8.1177. [DOI] [PubMed] [Google Scholar]
- Cella M, Facchetti F, Lanzavecchia A, Colonna M. Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization. Nat Immunol. 2000;1:305–310. doi: 10.1038/79747. [DOI] [PubMed] [Google Scholar]
- 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;5:919–923. doi: 10.1038/11360. [DOI] [PubMed] [Google Scholar]
- Chanock SJ, el Benna J, Smith RM, Babior BM. The respiratory burst oxidase. J Biol Chem. 1994;269:24519–24522. [PubMed] [Google Scholar]
- Chauveau C, Remy S, Royer PJ, Hill M, Tanguy-Royer S, Hubert FX, Tesson L, Brion R, Beriou G, Gregoire M, Josien R, Cuturi MC, Anegon I. Heme oxygenase-1 expression inhibits dendritic cell maturation and proinflammatory function but conserves IL-10 expression. Blood. 2005;106:1694–1702. doi: 10.1182/blood-2005-02-0494. [DOI] [PubMed] [Google Scholar]
- Chen W, Antonenko S, Sederstrom JM, Liang X, Chan AS, Kanzler H, Blom B, Blazar BR, Liu YJ. Thrombopoietin cooperates with FLT3-ligand in the generation of plasmacytoid dendritic cell precursors from human hematopoietic progenitors. Blood. 2004;103:2547–2553. doi: 10.1182/blood-2003-09-3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng F, Wang HW, Cuenca A, Huang M, Ghansah T, Brayer J, Kerr WG, Takeda K, Akira S, Schoenberger SP, Yu H, Jove R, Sotomayor EM. A critical role for Stat3 signaling in immune tolerance. Immunity. 2003;19:425–436. doi: 10.1016/s1074-7613(03)00232-2. [DOI] [PubMed] [Google Scholar]
- Colonna M, Trinchieri G, Liu YJ. Plasmacytoid dendritic cells in immunity. Nat Immunol. 2004;5:1219–1226. doi: 10.1038/ni1141. [DOI] [PubMed] [Google Scholar]
- Conejo-Garcia JR, Benencia F, Courreges MC, Kang E, Mohamed-Hadley A, Buckanovich RJ, Holtz DO, Jenkins A, Na H, Zhang L, Wagner DS, Katsaros D, Caroll R, Coukos G. Tumor-infiltrating dendritic cell precursors recruited by a beta-defensin contribute to vasculogenesis under the influence of Vegf-A. Nat Med. 2004;10:950–958. doi: 10.1038/nm1097. [DOI] [PubMed] [Google Scholar]
- Coussens LM, Tinkle CL, Hanahan D, Werb Z. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell. 2000;103:481–490. doi: 10.1016/s0092-8674(00)00139-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou W. Dendritic Cell Subsets Differentially Regulate Angiogenesis in Human Ovarian Cancer. Cancer Res. 2004;64:5535–5538. doi: 10.1158/0008-5472.CAN-04-1272. [DOI] [PubMed] [Google Scholar]
- 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;9:562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
- Darnell JE., Jr Studies of IFN-induced transcriptional activation uncover the Jak-Stat pathway. J Interferon Cytokine Res. 1998;18:549–554. doi: 10.1089/jir.1998.18.549. [DOI] [PubMed] [Google Scholar]
- De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J Pathol. 2003;200:429–447. doi: 10.1002/path.1398. [DOI] [PubMed] [Google Scholar]
- Denicola A, Souza JM, Radi R. Diffusion of peroxynitrite across erythrocyte membranes. Proc Natl Acad Sci U S A. 1998;95:3566–3571. doi: 10.1073/pnas.95.7.3566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denu JM, Tanner KG. Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry. 1998;37:5633–5642. doi: 10.1021/bi973035t. [DOI] [PubMed] [Google Scholar]
- Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001;193:233–238. doi: 10.1084/jem.193.2.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dhodapkar MV, Steinman RM, Sapp M, Desai H, Fossella C, Krasovsky J, Donahoe SM, Dunbar PR, Cerundolo V, Nixon DF, Bhardwaj N. Rapid generation of broad T-cell immunity in humans after a single injection of mature dendritic cells. J Clin Invest. 1999;104:173–180. doi: 10.1172/JCI6909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000;165:6037–6046. doi: 10.4049/jimmunol.165.11.6037. [DOI] [PubMed] [Google Scholar]
- Edwards AD, Diebold SS, Slack EM, Tomizawa H, Hemmi H, Kaisho T, Akira S, Reis e Sousa C. Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8 alpha+ DC correlates with unresponsiveness to imidazoquinolines. Eur J Immunol. 2003;33:827–833. doi: 10.1002/eji.200323797. [DOI] [PubMed] [Google Scholar]
- Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nature Rev Cancer. 2002;2:161–174. doi: 10.1038/nrc745. [DOI] [PubMed] [Google Scholar]
- Elenbaas B, Weinberg RA. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp Cell Res. 2001;264:169–184. doi: 10.1006/excr.2000.5133. [DOI] [PubMed] [Google Scholar]
- Elgert KD, Alleva DG, Mullins DW. Tumor-induced immune dysfunction: the macrophage connection. J Leukoc Biol. 1998;64:275–290. doi: 10.1002/jlb.64.3.275. [DOI] [PubMed] [Google Scholar]
- Ellis LM, Fidler IJ. Angiogenesis and metastasis. Eur J Cancer. 1996;32A:2451–2460. doi: 10.1016/s0959-8049(96)00389-9. [DOI] [PubMed] [Google Scholar]
- Enk AH, Angeloni VL, Udey MC, Katz SI. Inhibition of Langerhans cell antigen-presenting function by IL-10. A role for IL-10 in induction of tolerance. J Immunol. 1993;151:2390–2398. [PubMed] [Google Scholar]
- Facchetti F, de Wolf-Peeters C, van den Oord JJ, de Vos R, Desmet VJ. Plasmacytoid monocytes (so-called plasmacytoid T-cells) in Kikuchi’s lymphadenitis. An immunohistologic study. Am J Clin Pathol. 1989;92:42–50. doi: 10.1093/ajcp/92.1.42. [DOI] [PubMed] [Google Scholar]
- 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;4:1206–1212. doi: 10.1038/ni1003. [DOI] [PubMed] [Google Scholar]
- Fan XH, Han BH, Dong QG, Sha HF, Bao GL, Liao ML. [Vascular endothelial growth factor inhibits dendritic cells from patients with non-small cell lung carcinoma] Zhonghua Jie He He Hu Xi Za Zhi. 2003;26:539–543. [PubMed] [Google Scholar]
- Feijoo E, Alfaro C, Mazzolini G, Serra P, Penuelas I, Arina A, Huarte E, Tirapu I, Palencia B, Murillo O, Ruiz J, Sangro B, Richter JA, Prieto J, Melero I. Dendritic cells delivered inside human carcinomas are sequestered by interleukin-8. Int J Cancer. 2005;116:275–81. doi: 10.1002/ijc.21046. [DOI] [PubMed] [Google Scholar]
- Freedman RS, Deavers M, Liu J, Wang E. Peritoneal inflammation [mdash] a microenvironment for Epithelial Ovarian Cancer (EOC) J Transl Med. 2004;2:23. doi: 10.1186/1479-5876-2-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, Carbone DP. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92:4150–4166. [PubMed] [Google Scholar]
- Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2:1096–1103. doi: 10.1038/nm1096-1096. [DOI] [PubMed] [Google Scholar]
- Gabrilovich DI, Ishida T, Nadaf S, Ohm J, Carbone DP. Antibodies to vascular endothelial growth factor enhance the efficacy of cancer immunotherapy by improving endogenous dendritic cell function. Clin Cancer Res. 1999;5:2963–2970. [PubMed] [Google Scholar]
- Geissmann F, Revy P, Regnault A, Lepelletier Y, Dy M, Brousse N, Amigorena S, Hermine O, Durandy A. TGF-beta 1 prevents the noncognate maturation of human dendritic Langerhans cells. J Immunol. 1999;162:4567–4575. [PubMed] [Google Scholar]
- Gilliet M, Boonstra A, Paturel C, Antonenko S, Xu XL, Trinchieri G, O’Garra A, Liu YJ. The development of murine plasmacytoid dendritic cell precursors is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor. J Exp Med. 2002;195:953–958. doi: 10.1084/jem.20020045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grabbe S, Kampgen E, Schuler G. Dendritic cells: multi-lineal and multi-functional. Immunol Today. 2000;21:431–433. doi: 10.1016/s0167-5699(00)01694-7. [DOI] [PubMed] [Google Scholar]
- Grohmann U, Bianchi R, Orabona C, Fallarino F, Vacca C, Micheletti A, Fioretti MC, Puccetti P. Functional plasticity of dendritic cell subsets as mediated by CD40 versus B7 activation. J Immunol. 2003;171:2581–2587. doi: 10.4049/jimmunol.171.5.2581. [DOI] [PubMed] [Google Scholar]
- Grohmann U, Fallarino F, Bianchi R, Belladonna ML, Vacca C, Orabona C, Uyttenhove C, Fioretti MC, Puccetti P. IL-6 inhibits the tolerogenic function of CD8 alpha+ dendritic cells expressing indoleamine 2,3-dioxygenase. J Immunol. 2001;167:708–714. doi: 10.4049/jimmunol.167.2.708. [DOI] [PubMed] [Google Scholar]
- Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med. 1997;185:1101–1111. doi: 10.1084/jem.185.6.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hakomori S. Structure, organization, and function of glycosphingolipids in membrane. Curr Opin Hematol. 2003;10:16–24. doi: 10.1097/00062752-200301000-00004. [DOI] [PubMed] [Google Scholar]
- Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353–364. doi: 10.1016/s0092-8674(00)80108-7. [DOI] [PubMed] [Google Scholar]
- Handley ME, Thakker M, Pollara G, Chain BM, Katz DR. JNK activation limits dendritic cell maturation in response to reactive oxygen species by the induction of apoptosis. Free Radic Biol Med. 2005;38:1637–1652. doi: 10.1016/j.freeradbiomed.2005.02.022. [DOI] [PubMed] [Google Scholar]
- Hanisch FG, Schwientek T, Von Bergwelt-Baildon MS, Schultze JL, Finn O. O-Linked glycans control glycoprotein processing by antigen-presenting cells: a biochemical approach to the molecular aspects of MUC1 processing by dendritic cells. Eur J Immunol. 2003;33:3242–3254. doi: 10.1002/eji.200324189. [DOI] [PubMed] [Google Scholar]
- 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;194:769–779. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayashi T, Hideshima T, Akiyama M, Raje N, Richardson P, Chauhan D, Anderson KC. Ex vivo induction of multiple myeloma-specific cytotoxic T lymphocytes. Blood. 2003;102:1435–1442. doi: 10.1182/blood-2002-09-2828. [DOI] [PubMed] [Google Scholar]
- Hiltbold EM, Vlad AM, Ciborowski P, Watkins SC, Finn OJ. The Mechanism of Unresponsiveness to Circulating Tumor Antigen MUC1 Is a Block in Intracellular Sorting and Processing by Dendritic Cells. J Immunol. 2000;165:3730–3741. doi: 10.4049/jimmunol.165.7.3730. [DOI] [PubMed] [Google Scholar]
- Hoshino K, Kaisho T, Iwabe T, Takeuchi O, Akira S. Differential involvement of IFN-beta in Toll-like receptor-stimulated dendritic cell activation. Int Immunol. 2002;14:1225–1231. doi: 10.1093/intimm/dxf089. [DOI] [PubMed] [Google Scholar]
- Imada K, Leonard WJ. The JAK-STAT pathway. Mol Immunol. 2000;37:1–11. doi: 10.1016/s0161-5890(00)00018-3. [DOI] [PubMed] [Google Scholar]
- Inaba K, Inaba M, Deguchi M, Hagi K, Yasumizu R, Ikehara S, Muramatsu S, Steinman RM. Granulocytes, macrophages, and dendritic cells arise from a common major histocompatibility complex class II-negative progenitor in mouse bone marrow. Proc Natl Acad Sci U S A. 1993;90:3038–3042. doi: 10.1073/pnas.90.7.3038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishida T, Oyama T, Carbone D, Gabrilovich DI. Defective function of Langerhans cells in tumor-bearing animals is the result of defective maturation from hematopoietic progenitors. J Immunol. 1998;161:4842–4851. [PubMed] [Google Scholar]
- Itoh S, Itoh F, Goumans MJ, Ten Dijke P. Signaling of transforming growth factor-beta family members through Smad proteins. Eur J Biochem. 2000;267:6954–6967. doi: 10.1046/j.1432-1327.2000.01828.x. [DOI] [PubMed] [Google Scholar]
- Iwamoto M, Shinohara H, Miyamoto A, Okuzawa M, Mabuchi H, Nohara T, Gon G, Toyoda M, Tanigawa N. Prognostic value of tumor-infiltrating dendritic cells expressing CD83 in human breast carcinomas. Int J Cancer. 2003;104:92–97. doi: 10.1002/ijc.10915. [DOI] [PubMed] [Google Scholar]
- Iyengar P, Combs TP, Shah SJ, Gouon-Evans V, Pollard JW, Albanese C, Flanagan L, Tenniswood MP, Guha C, Lisanti MP, Pestell RG, Scherer PE. Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene. 2003;22:6408–6423. doi: 10.1038/sj.onc.1206737. [DOI] [PubMed] [Google Scholar]
- Jackson SH, Yu C-R, Mahdi RM, Ebong S, Egwuagu CE. Dendritic Cell Maturation Requires STAT1 and Is under Feedback Regulation by Suppressors of Cytokine Signaling. J Immunol. 2004;172:2307–2315. doi: 10.4049/jimmunol.172.4.2307. [DOI] [PubMed] [Google Scholar]
- 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;192:1213–1222. doi: 10.1084/jem.192.9.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadowaki N, Antonenko S, Lau JY, Liu YJ. Natural interferon alpha/beta-producing cells link innate and adaptive immunity. J Exp Med. 2000;192:219–226. doi: 10.1084/jem.192.2.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadowaki N, Ho S, Antonenko S, Malefyt RW, Kastelein RA, Bazan F, Liu YJ. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869. doi: 10.1084/jem.194.6.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaech SM, Ahmed R. Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naive cells. Nat Immunol. 2001;2:415–422. doi: 10.1038/87720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalinski P, Hilkens CM, Wierenga EA, Kapsenberg ML. T-cell priming by type-1 and type-2 polarized dendritic cells: the concept of a third signal. Immunol Today. 1999;20:561–567. doi: 10.1016/s0167-5699(99)01547-9. [DOI] [PubMed] [Google Scholar]
- Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nature Rev Cancer. 2003;3:422–433. doi: 10.1038/nrc1094. [DOI] [PubMed] [Google Scholar]
- Kinjyo I, Hanada T, Inagaki-Ohara K, Mori H, Aki D, Ohishi M, Yoshida H, Kubo M, Yoshimura A. SOCS1/JAB is a negative regulator of LPS-induced macrophage activation. Immunity. 2002;17:583–591. doi: 10.1016/s1074-7613(02)00446-6. [DOI] [PubMed] [Google Scholar]
- Kriehuber E, Bauer W, Charbonnier AS, Winter D, Amatschek S, Tamandl D, Schweifer N, Stingl G, Maurer D. Balance between NF-kappaB and JNK/AP-1 activity controls dendritic cell life and death. Blood. 2005;106:175–183. doi: 10.1182/blood-2004-08-3072. [DOI] [PubMed] [Google Scholar]
- Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci U S A. 2001;98:12072–12077. doi: 10.1073/pnas.211053698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kryczek I, Grybos M, Karabon L, Klimczak A, Lange A. IL-6 production in ovarian carcinoma is associated with histiotype and biological characteristics of the tumour and influences local immunity. Br J Cancer. 2000;82:621–628. doi: 10.1054/bjoc.1999.0973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kryczek I, Lange A, Mottram P, Alvarez X, Cheng P, Hogan M, Moons L, Wei S, Zou L, Machelon V, Emilie D, Terrassa M, Lackner A, Curiel TJ, Carmeliet P, Zou W. CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res. 2005;65:465–472. [PubMed] [Google Scholar]
- Kusmartsev S, Nagaraj S, Gabrilovich DI. Tumor-Associated CD8+ T Cell Tolerance Induced by Bone Marrow-Derived Immature Myeloid Cells. J Immunol. 2005;175:4583–4592. doi: 10.4049/jimmunol.175.7.4583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Labeur MS, Roters B, Pers B, Mehling A, Luger TA, Schwarz T, Grabbe S. Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage. J Immunol. 1999;162:168–175. [PubMed] [Google Scholar]
- Lanzavecchia A, Sallusto F. The instructive role of dendritic cells on T cell responses: lineages, plasticity and kinetics. Curr Opin Immunol. 2001;13:291–298. doi: 10.1016/s0952-7915(00)00218-1. [DOI] [PubMed] [Google Scholar]
- Laxmanan S, Robertson SW, Wang E, Lau JS, Briscoe DM, Mukhopadhyay D. Vascular endothelial growth factor impairs the functional ability of dendritic cells through Id pathways. Biochem Biophys Res Commun. 2005;334:193–198. doi: 10.1016/j.bbrc.2005.06.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med. 2001;193:727–740. doi: 10.1084/jem.193.6.727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lissoni P, Malugani F, Bonfanti A, Bucovec R, Secondino S, Brivio F, Ferrari-Bravo A, Ferrante R, Vigore L, Rovelli F, Mandala M, Viviani S, Fumagalli L, Gardani GS. Abnormally enhanced blood concentrations of vascular endothelial growth factor (VEGF) in metastatic cancer patients and their relation to circulating dendritic cells, IL-12 and endothelin-1. J Biol Regul Homeost Agents. 2001;15:140–144. [PubMed] [Google Scholar]
- Manabe Y, Toda S, Miyazaki K, Sugihara H. Mature adipocytes, but not preadipocytes, promote the growth of breast carcinoma cells in collagen gel matrix culture through cancer-stromal cell interactions. J Pathol. 2003;201:221–228. doi: 10.1002/path.1430. [DOI] [PubMed] [Google Scholar]
- McCawley LJ, Matrisian LM. Matrix metalloproteinases: they’re not just for matrix anymore! . Curr Opin Cell Biol. 2001;13:534–540. doi: 10.1016/s0955-0674(00)00248-9. [DOI] [PubMed] [Google Scholar]
- Mellor AL, Baban B, Chandler P, Marshall B, Jhaver K, Hansen A, Koni PA, Iwashima M, Munn DH. Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. J Immunol. 2003;171:1652–1655. doi: 10.4049/jimmunol.171.4.1652. [DOI] [PubMed] [Google Scholar]
- Mellor AL, Chandler P, Baban B, Hansen AM, Marshall B, Pihkala J, Waldmann H, Cobbold S, Adams E, Munn DH. Specific subsets of murine dendritic cells acquire potent T cell regulatory functions following CTLA4-mediated induction of indoleamine 2,3 dioxygenase. Int Immunol. 2004a;16:1391–1401. doi: 10.1093/intimm/dxh140. [DOI] [PubMed] [Google Scholar]
- Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004b;4:762–774. doi: 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
- Menetrier-Caux C, Montmain G, Dieu MC, Bain C, Favrot MC, Caux C, Blay JY. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor. Blood. 1998;92:4778–4791. [PubMed] [Google Scholar]
- Menetrier-Caux C, Thomachot MC, Alberti L, Montmain G, Blay JY. IL-4 prevents the blockade of dendritic cell differentiation induced by tumor cells. Cancer Res. 2001;61:3096–3104. [PubMed] [Google Scholar]
- Miyamoto M, Shimizu Y, Okada K, Kashii Y, Higuchi K, Watanabe A. Effect of interleukin-8 on production of tumor-associated substances and autocrine growth of human liver and pancreatic cancer cells. Cancer Immunol Immunother. 1998;47:47–57. doi: 10.1007/s002620050503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monteiro HP, Stern A. Redox modulation of tyrosine phosphorylation-dependent signal transduction pathways. Free Radic Biol Med. 1996;21:323–333. doi: 10.1016/0891-5849(96)00051-2. [DOI] [PubMed] [Google Scholar]
- Monti P, Leone BE, Zerbi A, Balzano G, Cainarca S, Sordi V, Pontillo M, Mercalli A, Di Carlo V, Allavena P, Piemonti L. Tumor-derived MUC1 mucins interact with differentiating monocytes and induce IL-10highIL-12low regulatory dendritic cell. J Immunol. 2004;172:7341–7349. doi: 10.4049/jimmunol.172.12.7341. [DOI] [PubMed] [Google Scholar]
- Mueller MM, Werbowetski T, Del Maestro RF. Soluble factors involved in glioma invasion. Acta Neurochir (Wien) 2003;145:999–1008. doi: 10.1007/s00701-003-0132-0. [DOI] [PubMed] [Google Scholar]
- Munn DH, Mellor AL. IDO and tolerance to tumors. Trends Mol Med. 2004a;10:15–18. doi: 10.1016/j.molmed.2003.11.003. [DOI] [PubMed] [Google Scholar]
- 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. 2004b;114:280–290. doi: 10.1172/JCI21583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;297:1867–1870. doi: 10.1126/science.1073514. [DOI] [PubMed] [Google Scholar]
- Munz B, Hubner G, Tretter Y, Alzheimer C, Werner S. A novel role of activin in inflammation and repair. J Endocrinol. 1999;161:187–193. doi: 10.1677/joe.0.1610187. [DOI] [PubMed] [Google Scholar]
- Naka T, Fujimoto M, Kishimoto T. Negative regulation of cytokine signaling: STAT-induced STAT inhibitor. Trends Biochem Sci. 1999;24:394–398. doi: 10.1016/s0968-0004(99)01454-1. [DOI] [PubMed] [Google Scholar]
- Nakagawa R, Naka T, Tsutsui H, Fujimoto M, Kimura A, Abe T, Seki E, Sato S, Takeuchi O, Takeda K, Akira S, Yamanishi K, Kawase I, Nakanishi K, Kishimoto T. SOCS-1 participates in negative regulation of LPS responses. Immunity. 2002;17:677–687. doi: 10.1016/s1074-7613(02)00449-1. [DOI] [PubMed] [Google Scholar]
- Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, Jove R, Gabrilovich D. Hyperactivation of STAT3 Is Involved in Abnormal Differentiation of Dendritic Cells in Cancer. J Immunol. 2004;172:464–474. doi: 10.4049/jimmunol.172.1.464. [DOI] [PubMed] [Google Scholar]
- Ohm JE, Carbone DP. VEGF as a mediator of tumor-associated immunodeficiency. Immunol Res. 2001;23:263–272. doi: 10.1385/IR:23:2-3:263. [DOI] [PubMed] [Google Scholar]
- Ohm JE, Shurin MR, Esche C, Lotze MT, Carbone DP, Gabrilovich DI. Effect of vascular endothelial growth factor and FLT3 ligand on dendritic cell generation in vivo. J Immunol. 1999;163:3260–3268. [PubMed] [Google Scholar]
- O’Keeffe M, Hochrein H, Vremec D, Caminschi I, Miller JL, Anders EM, Wu L, Lahoud MH, Henri S, Scott B, Hertzog P, Tatarczuch L, Shortman K. Mouse Plasmacytoid Cells: Long-lived Cells, Heterogeneous in Surface Phenotype and Function, that Differentiate Into CD8+ Dendritic Cells Only after Microbial Stimulus. J Exp Med. 2002;196:1307–1319. doi: 10.1084/jem.20021031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olweus J, BitMansour A, Warnke R, Thompson PA, Carballido J, Picker LJ, Lund-Johansen F. Dendritic cell ontogeny: a human dendritic cell lineage of myeloid origin. Proc Natl Acad Sci U S A. 1997;94:12551–12556. doi: 10.1073/pnas.94.23.12551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Neill DW, Adams S, Bhardwaj N. Manipulating dendritic cell biology for the active immunotherapy of cancer. Blood. 2004;104:2235–2246. doi: 10.1182/blood-2003-12-4392. [DOI] [PubMed] [Google Scholar]
- Park JW, Babior BM. The translocation of respiratory burst oxidase components from cytosol to plasma membrane is regulated by guanine nucleotides and diacylglycerol. J Biol Chem. 1992;267:19901–19906. [PubMed] [Google Scholar]
- 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;173:3844–3854. doi: 10.4049/jimmunol.173.6.3844. [DOI] [PubMed] [Google Scholar]
- Peguet-Navarro J, Moulon C, Caux C, Dalbiez-Gauthier C, Banchereau J, Schmitt D. Interleukin-10 inhibits the primary allogeneic T cell response to human epidermal Langerhans cells. Eur J Immunol. 1994;24:884–891. doi: 10.1002/eji.1830240416. [DOI] [PubMed] [Google Scholar]
- 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;170:3488–3494. doi: 10.4049/jimmunol.170.7.3488. [DOI] [PubMed] [Google Scholar]
- Pulendran B, Banchereau J, Burkeholder S, Kraus E, Guinet E, Chalouni C, Caron D, Maliszewski C, Davoust J, Fay J, Palucka K. Flt3-ligand and granulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo. J Immunol. 2000;165:566–572. doi: 10.4049/jimmunol.165.1.566. [DOI] [PubMed] [Google Scholar]
- Radi R. Nitric oxide, oxidants, and protein tyrosine nitration. Proc Natl Acad Sci U S A. 2004;101:4003–4008. doi: 10.1073/pnas.0307446101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Randolph GJ, Inaba K, Robbiani DF, Steinman RM, Muller WA. Differentiation of phagocytic monocytes into lymph node dendritic cells in vivo. Immunity. 1999;11:753–761. doi: 10.1016/s1074-7613(00)80149-1. [DOI] [PubMed] [Google Scholar]
- Rane SG, Reddy ES. Janus kinases: components of multiple signaling pathways. Oncogene. 2000;19:5662–5679. doi: 10.1038/sj.onc.1203925. [DOI] [PubMed] [Google Scholar]
- 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;100:230–237. doi: 10.1182/blood.v100.1.230. [DOI] [PubMed] [Google Scholar]
- Rissoan MC, Soumelis V, Kadowaki N, Grouard G, Briere F, de Waal Malefyt R, Liu YJ. Reciprocal control of T helper cell and dendritic cell differentiation. Science. 1999;283:1183–1186. doi: 10.1126/science.283.5405.1183. [DOI] [PubMed] [Google Scholar]
- Roncarolo MG, Levings MK, Traversari C. Differentiation of T regulatory cells by immature dendritic cells. J Exp Med. 2001;193:F5–9. doi: 10.1084/jem.193.2.f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosendahl A, Checchin D, Fehniger TE, ten Dijke P, Heldin CH, Sideras P. Activation of the TGF-beta/activin-Smad2 pathway during allergic airway inflammation. Am J Respir Cell Mol Biol. 2001;25:60–68. doi: 10.1165/ajrcmb.25.1.4396. [DOI] [PubMed] [Google Scholar]
- Rughetti A, Pellicciotta I, Biffoni M, Backstrom M, Link T, Bennet EP, Clausen H, Noll T, Hansson GC, Burchell JM, Frati L, Taylor-Papadimitriou J, Nuti M. Recombinant Tumor-Associated MUC1 Glycoprotein Impairs the Differentiation and Function of Dendritic Cells. J Immunol. 2005;174:7764–7772. doi: 10.4049/jimmunol.174.12.7764. [DOI] [PubMed] [Google Scholar]
- Saito H, Tsujitani S, Ikeguchi M, Maeta M, Kaibara N. Relationship between the expression of vascular endothelial growth factor and the density of dendritic cells in gastric adenocarcinoma tissue. Br J Cancer. 1998;78:1573–1577. doi: 10.1038/bjc.1998.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor [alpha] J Exp Med. 1994;179:1109–1118. doi: 10.1084/jem.179.4.1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sandel MH, Dadabayev AR, Menon AG, Morreau H, Melief CJM, Offringa R, van der Burg SH, Janssen-van Rhijn CM, Ensink NG, Tollenaar RAEM, van de Velde CJH, Kuppen PJK. Prognostic Value of Tumor-Infiltrating Dendritic Cells in Colorectal Cancer: Role of Maturation Status and Intratumoral Localization. Clin Cancer Res. 2005;11:2576–2582. doi: 10.1158/1078-0432.CCR-04-1448. [DOI] [PubMed] [Google Scholar]
- Schadendorf D, Moller A, Algermissen B, Worm M, Sticherling M, Czarnetzki BM. IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. J Immunol. 1993;151:2667–2675. [PubMed] [Google Scholar]
- Schopfer FJ, Baker PR, Freeman BA. NO-dependent protein nitration: a cell signaling event or an oxidative inflammatory response? . Trends Biochem Sci. 2003;28:646–654. doi: 10.1016/j.tibs.2003.10.006. [DOI] [PubMed] [Google Scholar]
- Schraufstatter IU, Trieu K, Zhao M, Rose DM, Terkeltaub RA, Burger M. IL-8-mediated cell migration in endothelial cells depends on cathepsin B activity and transactivation of the epidermal growth factor receptor. J Immunol. 2003;171:6714–6722. doi: 10.4049/jimmunol.171.12.6714. [DOI] [PubMed] [Google Scholar]
- Seo N, Hayakawa S, Takigawa M, Tokura Y. Interleukin-10 expressed at early tumour sites induces subsequent generation of CD4+ T-regulatory cells and systemic collapse of antitumour immunity. Immunology. 2001;103:449–457. doi: 10.1046/j.1365-2567.2001.01279.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serafini P. High-dose GM-CSF-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells. Cancer Res. 2004;64:6337–6343. doi: 10.1158/0008-5472.CAN-04-0757. [DOI] [PubMed] [Google Scholar]
- Sharma S, Stolina M, Lin Y, Gardner B, Miller PW, Kronenberg M, Dubinett SM. T cell-derived IL-10 promotes lung cancer growth by suppressing both T cell and APC function. J Immunol. 1999;163:5020–5028. [PubMed] [Google Scholar]
- Shurin GV, Shurin MR, Bykovskaia S, Shogan J, Lotze MT, Barksdale EM., Jr Neuroblastoma-derived gangliosides inhibit dendritic cell generation and function. Cancer Res. 2001;61:363–369. [PubMed] [Google Scholar]
- Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, Damme JV, Mantovani A. Autocrine Production of IL-10 Mediates Defective IL-12 Production and NF-{kappa}B Activation in Tumor-Associated Macrophages. J Immunol. 2000;164:762–767. doi: 10.4049/jimmunol.164.2.762. [DOI] [PubMed] [Google Scholar]
- Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, Antonenko S, Liu YJ. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999;284:1835–1837. doi: 10.1126/science.284.5421.1835. [DOI] [PubMed] [Google Scholar]
- Sieling PA, Modlin RL. Toll-like receptors: mammalian "taste receptors" for a smorgasbord of microbial invaders. Curr Opin Microbiol. 2002;5:70–75. doi: 10.1016/s1369-5274(02)00288-6. [DOI] [PubMed] [Google Scholar]
- Spits H, Couwenberg F, Bakker AQ, Weijer K, Uittenbogaart CH. Id2 and Id3 inhibit development of CD34(+) stem cells into predendritic cell (pre-DC)2 but not into pre-DC1. Evidence for a lymphoid origin of pre-DC2. J Exp Med. 2000;192:1775–1784. doi: 10.1084/jem.192.12.1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinbrink K, Graulich E, Kubsch S, Knop J, Enk A. CD4+ and CD8+ anergic T cells induced by interleukin-10-treated human dendritic cells display antigen specific suppressor activity. Blood. 2002;99:2468–2476. doi: 10.1182/blood.v99.7.2468. [DOI] [PubMed] [Google Scholar]
- Steinbrink K, Wolfl M, Jonuleit H, Knop J, Enk AH. Induction of tolerance by IL-10-treated dendritic cells. J Immunol. 1997;159:4772–4780. [PubMed] [Google Scholar]
- Stetler-Stevenson WG, Yu AE. Proteases in invasion: matrix metalloproteinases. Semin Cancer Biol. 2001;11:143–152. doi: 10.1006/scbi.2000.0365. [DOI] [PubMed] [Google Scholar]
- Svensson M, Maroof A, Ato M, Kaye PM. Stromal cells direct local differentiation of regulatory dendritic cells. Immunity. 2004;21:805–816. doi: 10.1016/j.immuni.2004.10.012. [DOI] [PubMed] [Google Scholar]
- Takahashi A. Vascular endothelial growth factor inhibits maturation of dendritic cells induced by lipopolysaccharide, but not by proinflammatory cytokines. Cancer Immunol Immunother. 2004;53:543–550. doi: 10.1007/s00262-003-0466-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takauji R, Iho S, Takatsuka H, Yamamoto S, Takahashi T, Kitagawa H, Iwasaki H, Iida R, Yokochi T, Matsuki T. CpG-DNA-induced IFN-alpha production involves p38 MAPK-dependent STAT1 phosphorylation in human plasmacytoid dendritic cell precursors. J Leukoc Biol. 2002;72:1011–1019. [PubMed] [Google Scholar]
- Tlsty TD, Hein PW. Know thy neighbor: stromal cells can contribute oncogenic signals. Curr Opin Genet Dev. 2001;11:54–59. doi: 10.1016/s0959-437x(00)00156-8. [DOI] [PubMed] [Google Scholar]
- Toi M, Kondo S, Suzuki H, Yamamoto Y, Inada K, Imazawa T, Taniguchi T, Tominaga T. Quantitative analysis of vascular endothelial growth factor in primary breast cancer. Cancer. 1996;77:1101–1106. doi: 10.1002/(sici)1097-0142(19960315)77:6<1101::aid-cncr15>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
- Troy A, Davidson P, Atkinson C, Hart D. Phenotypic characterisation of the dendritic cell infiltrate in prostate cancer. J Urol. 1998a;160:214–219. [PubMed] [Google Scholar]
- Troy A, Summers K, Davidson P, Atkinson C, Hart D. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin Cancer Res. 1998b;4:585–593. [PubMed] [Google Scholar]
- Venkatakrishnan G, Salgia R, Groopman JE. Chemokine receptors CXCR-1/2 activate mitogen-activated protein kinase via the epidermal growth factor receptor in ovarian cancer cells. J Biol Chem. 2000;275:6868–6875. doi: 10.1074/jbc.275.10.6868. [DOI] [PubMed] [Google Scholar]
- Vieira PL, de Jong EC, Wierenga EA, Kapsenberg ML, Kalinski P. Development of Th1-inducing capacity in myeloid dendritic cells requires environmental instruction. J Immunol. 2000;164:4507–4512. doi: 10.4049/jimmunol.164.9.4507. [DOI] [PubMed] [Google Scholar]
- Walker JG, Ahern MJ, Coleman M, Weedon H, Papangelis V, Beroukas D, Roberts-Thomson PJ, Smith MD. Expression of Jak3, STAT1, STAT4 and STAT6 in inflammatory arthritis: Unique Jak3 and STAT4 expression in dendritic cells in seropositive rheumatoid arthritis. Ann Rheum Dis. 2006;65:149–56. doi: 10.1136/ard.2005.037929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiseman BS, Werb Z. Stromal effects on mammary gland development and breast cancer. Science. 2002;296:1046–1049. doi: 10.1126/science.1067431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang AS, Lattime EC. Tumor-induced interleukin 10 suppresses the ability of splenic dendritic cells to stimulate CD4 and CD8 T-cell responses. Cancer Res. 2003;63:2150–2157. [PubMed] [Google Scholar]
- Young M, Wright M, Young M. Antibodies to colony-stimulating factors block Lewis lung carcinoma cell stimulation of immune-suppressive bone marrow cells. Cancer Immunol Immunother. 1991;33:146–152. doi: 10.1007/BF01756134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M, Tang H, Guo Z, An H, Zhu X, Song W, Guo J, Huang X, Chen T, Wang J, Cao X. Splenic stroma drives mature dendritic cells to differentiate into regulatory dendritic cells. Nat Immunol. 2004;5:1124–1133. doi: 10.1038/ni1130. [DOI] [PubMed] [Google Scholar]
- 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;7:1339–1346. doi: 10.1038/nm1201-1339. [DOI] [PubMed] [Google Scholar]