Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells (original) (raw)

Functional properties of tumor-induced CD11b+ myeloid cells. In our studies we used initially the C26 colon carcinoma transduced to release mouse GM-CSF (C26-GM). The amount of GM-CSF produced by the C26-GM cell line was within the range of production by human and mouse tumor cell lines (22). Once inoculated s.c., C26-GM induced the rapid mobilization of myelomonocytic cells in the blood and spleen, and, 9 days after s.c. implantation, the spleen contained 2 main CD11b+ subsets, one expressing only CD11b and the other positive for both CD11b and Gr-1 antigens (22). Both fractions possessed suppressive activity on antigen-activated CD8+ T lymphocytes, whereas CD11b+ cells from tumor-free mice were not suppressive (see Supplemental Figure 1; supplemental material available online with this article; doi:10.1172/JCI28828DS1).

CD11b+ cells purified from the spleens of tumor-bearing mice were added as third party to CD8+ T lymphocytes (CL4 cells) expressing a transgenic TCR specific for the influenza HA. These lymphocytes had been primed in vivo with a vaccinia virus encoding HA and labeled with CFSE before culture to monitor cell division. The cultures were stimulated for 60 hours with the HA-derived immunodominant peptide, and clonotypic proliferation was evaluated as CFSE dilution by flow cytometry (Figure 1A). Dividing cells were plotted as a fraction of the total clonotypic CL4 cells (Figure 1B). In the presence of cognate peptide, CL4 cells underwent more than 4 divisions, whereas tumor-induced CD11b+ splenocytes reduced antigen-specific T cell division to 1–3 cycles (Figure 1B). Cell death and reduced cell recovery during the culture of CL4 T cells in the presence of tumor-conditioned CD11b+ splenocytes indicate that antigen-activated clonotypic T cells eventually underwent apoptosis (Supplemental Figure 2) as suggested by previous findings in various models where MSCs have been studied (1, 27).

MSCs inhibit antigen-induced T lymphocyte proliferation but not IFN-γ releaFigure 1

MSCs inhibit antigen-induced T lymphocyte proliferation but not IFN-γ release. (A) CFSE-labeled splenocytes derived from mice transferred with HA-specific CD8+ (CL4) T cells and primed with HA-encoding vaccinia virus were stimulated with the HA peptide in the absence (No CD11b) or in the presence of CD11b+ cells magnetically purified from either the spleen or the tumor infiltrate of tumor-bearing mice and admixed at a 1:10 ratio. The cultures were stimulated for 60 hours with the relevant peptide, and the clonotypic proliferation was evaluated as CFSE dilution by flow cytometry. (B) Data are presented as the mean percentage of CL4 cells in each cycle from triplicate wells (± SEM); 1 of 2 representative experiments is reported. (C) MSCs do not suppress IFN-γ production from naive or antigen-experienced (memory) CD8+ T cells. MSCs were isolated from the spleens of normal or tumor-bearing mice, and 6 × 105 cells were admixed with 3 × 106 splenocytes containing naive (2.4 × 105) or in vivo antigen-experienced (2.1 × 105) HA-specific CD8+ T cells stimulated with the relevant peptide in an ELISPOT assay. Data (mean ± SEM) are derived from triplicate wells of 1 representative experiment.

In addition to examining the proliferative activity, we sought to determine the extent to which MSCs could impact early effector functions. Naive or memory CL4 T cells were cocultured with normal or tumor-induced CD11b+ splenocytes and stimulated with the HA-specific peptide. IFN-γ production was determined by ELISPOT. No change could be seen between normal and tumor-derived CD11b+ cells (Figure 1C); this is consistent with the fact that MSCs do not block the early events of T cell activation (28). As we show below, early events of lymphocyte activation likely deliver important signals favoring the maturation of CD11b+ cells into a fully suppressive phenotype.

Gene expression profiling of CD11b+ splenocytes. Use of different cell surface markers has not been sufficient to dissect further the heterogeneity of MSCs and to identify the cells endowed with the immunoregulatory properties. Therefore, we performed a genome-wide profiling approach using Affymetrix GeneChips. The RNA was extracted from CD11b+ cells enriched from pooled spleens of normal mice (about 5–10 spleens), and CD11b+ cells isolated from the spleens of single C26-GM tumor–bearing mice. These were analyzed immediately (time 0) or after a 24-hour incubation in complete medium. In vitro incubation was performed because we and others have shown that culture of MSCs increases their suppressive activity (1, 2, 5, 14), which suggests the existence of an internal differentiation/maturation program. Three biological mRNA replicates were hybridized for each group. The raw data have been deposited, and processed values and complete gene lists are available (Gene Expression Omnibus [GEO] accession no. GSE5455; http://www.ncbi.nlm.nih.gov/geo/).

Microarray analysis confirmed that the populations analyzed clustered in separate groups for a number of upregulated and downregulated genes (Supplemental Results, Supplemental Figures 3–5, and Supplemental Tables 2–8). CD11b+ splenocytes freshly isolated from tumor-bearing mice differed from the normal counterparts in regard to the presence of 2 main signatures. One identifies genes coding either for enzymes characterizing polymorphonuclear cells or for molecules associated with acute inflammatory responses (Table 1). The other signature comprises cytokines, membrane molecules, and markers associated with alternative activation of macrophages. This cluster included chitinase 3–like 3 (also known as Ym-1), complement component 4, IL-6, IL-10, IL-1α, IL-1 receptor II, TGF-β–induced transcript 4, and the C-type mannose receptor (25, 29). The A2b adenosine receptor is not unique for macrophages, since it is expressed in mast cells as well, but it has been associated with cytokine-induced macrophage deactivation (30). Taken together, these microarray data suggest that CD11b+ splenocytes comprise both inflammatory granulocytes and a subset of monocytes/macrophages. Other signatures could not be clearly associated with a peculiar cell type, and no clear marker of T, B, and NK lymphocytes was discernible (Supplemental Results and GEO accession no. GSE5455).

Table 1

Transcripts upregulated in CD11b+ splenocytes sorted from the spleens of tumor-bearing miceA

Comparison of CD11b+ splenocytes at time 0 and after 24 hours confirmed the existence of a cellular program leading to alternative macrophage activation. One of the most upregulated genes after 24 hours of culture, in fact, was Arg1, coding for the enzyme Arg1, which is a well-recognized marker of alternatively activated macrophages and also a critical enzyme for the inhibitory activity exerted by MSCs on activated T lymphocytes (21). Other genes were found to be spontaneously upregulated during the 24-hour culture that belong to the pathway activated by Th2-type cytokines such as IL-4 and IL-13 (Table 1). Accordingly, the mRNAs for most of these genes were upregulated by treatment of a cloned MSC population with IL-4 (Supplemental Table 9).

The program activated in CD11b+ splenocytes from tumor-bearing mice is more complex than a plain alternative activation, since many overexpressed genes are under the control of type I and type II IFNs. In particular, Nos2 was also spontaneously upregulated (Table 1), which confirmed our previous findings of the unique, concomitant expression of both enzymes of l-arginine metabolism in tumor-induced CD11b+ splenocytes (10, 22). Taken together, these data indicate MSCs as a peculiar population that eludes the rigid classification of classically or alternatively activated macrophages and suggest the presence of MSC-released autocrine factors driving in vitro MSC maturation/differentiation.

CD11b+ splenocytes from tumor-bearing mice produce IFN-γ and IL-13 and require these cytokines to activate the suppressive program. We speculated that the gene expression profiles emerging after in vitro culture of tumor-induced CD11b+ splenocytes could result from the autocrine action of cytokines released by the same cells. IL-13 and IFN-γ were detectable in the supernatant of CD11b+ splenocytes isolated from tumor-bearing mice and cultured for 48 hours (160 ± 25 and 22,000 ± 235 pg/ml per 105 cells, respectively), and intracellular staining confirmed that a sizeable fraction of CD11b+ splenocytes from tumor-bearing but not from tumor-free mice produced both IFN-γ and IL-13 (Figure 2). Accordingly, CD11b+ splenocytes isolated from tumor-bearing IFN-γ KO mice produced IL-13, suggesting that expression of the 2 cytokines is uncoupled (Figure 2). IL-4 was not detected by ELISA, ELISPOT, or intracellular staining (data not shown).

CD11b+ splenocytes from tumor-bearing mice produce both IFN-γ and IL-13.Figure 2

CD11b+ splenocytes from tumor-bearing mice produce both IFN-γ and IL-13. CD11b+ cells isolated from the spleens of normal BALB/c and different tumor-bearing mice (WT BALB/c, IFN-γ–/–, CD1–/–, and RAG–/–γc–/– KO mice) were analyzed by flow cytometry for intracellular presence of IL-13 (top panels) and IFN-γ (bottom panels). Open histograms indicate staining with IL-13 or IFN-γ antibody; filled histograms indicate staining with isotype-matched antibody controls. Data are representative of at least 3 determinations in separate experiments. MFI, mean fluorescence intensity (a log scale of 100–104 was used).

To address further the role of Th1 and Th2 cytokines in the regulation of MSC function, we analyzed the in vitro generation of alloreactive CTLs in the presence of CD11b+ cells isolated from the spleens of tumor-bearing BALB/c mice, WT or deficient for either IFN-γ or IL-4. When tumor-induced CD11b+ cells were added to WT BALB/c lymphocytes stimulated with γ-irradiated C57BL/6 allogeneic cells, CTL generation was inhibited by either WT or IL-4 KO CD11b+ cells (Figure 3A). On the other hand, CD11b+ cells from IFN-γ KO mice were not suppressive but rather increased the number of alloantigen-specific CTLs recovered from the cultures. This indicates that the ability of CD11b+ cells to release IFN-γ but not IL-4 is important for the suppressive properties of tumor-induced CD11b+ cells. We then asked whether the release of cytokines from activated T cells could play a role in the suppressive pathways. Responder T cells from either IFN-γ KO (Figure 3B) or IL-4 KO (Figure 3C) mice were mixed with allogeneic targets in the presence of CD11b+ cells from the different strains. Lack of IFN-γ production by alloantigen-stimulated T cells completely abrogated the suppressive activity, regardless of the origin of the tumor-induced CD11b+ cells (Figure 3B). In contrast, the T cells’ inability to produce IL-4 did not alter the suppression by third-party suppressors unless CD11b+ cells from IFN-γ KO mice were used in the assay (Figure 3C). These results confirmed the requirement for IFN-γ production by both activated T lymphocytes and tumor-induced CD11b+ cells and completely ruled out any contribution of IL-4 in CD11b+-mediated suppression.

Evaluation of cytokines and cytokine receptors required by tumor-induced CDFigure 3

Evaluation of cytokines and cytokine receptors required by tumor-induced CD11b+ cells to inhibit generation of alloreactive and tumor-specific CTLs. CD11b+ sorted cells from tumor-bearing WT or different KO mice were added at a final concentration of 3% to a mixed leukocyte culture set up with BALB/c effectors stimulated with an equal number of C57BL/6 splenocytes. After 5 days, cytotoxic activity was tested in a 5-hour 51Cr-release assay against either a syngeneic control target (CT26, H-2d) or an allogeneic target (MBL-2, H-2b). Effector lymphocytes were taken from WT (A), IFN-γ KO (B), or IL-4 KO (C) mice. LU30, defined as the number of lymphocytes necessary to achieve 30% specific lysis of 2 × 103 target cells in a 5-hour assay, was calculated on the basis of the total number of viable cells recovered from the cultures. Data are expressed as the ratio between the LU30 measured in cultures containing the third-party cells and in control cultures set up in the absence of third-party cells. Data are mean ± SEM from 3 experiments.

Having excluded IL-4, we considered the involvement of IL-13, since this cytokine plays a role in the alternative macrophage activation and induces ARG1 (25). We used mice deficient in the IL-4 receptor (IL-4R) α chain (CD124). The IL-4R is a heterodimeric complex composed of different chains. In the type I IL-4R, IL-4Rα and a common γ chain form a complex binding only IL-4, whereas the type 2 IL-4R is composed of IL-4Rα and IL-13R1 chains and binds both IL-4 and IL-13 (31). CD11b+ cells were isolated from IL-4Rα KO tumor-bearing mice and added as a third party to the allo-cultures. The presence of IL-4Rα on CD11b+ cells appeared to be critical for their suppressive activity (Figure 3A). Since IL-4 neither was released nor appeared involved in suppression, it is logical to assume that the receptor was activated by IL-13.

To exclude a possible role of contaminating NK cells as a source of IFN-γ among the sorted CD11b+ splenocytes, we isolated CD11b+ cells from the spleens of Rag2–/–γc–/– KO mice, which lack NK cells in addition to B and T lymphocytes (32). We also used CD1–/– mice to obtain CD11b+ cells devoid of any contaminating NKT cells that might account for the IL-13 production. Berzofsky and colleagues, in fact, have shown that NKT cells releasing IL-13 can activate immunosuppressive CD11b+Gr-1+ cells in some mouse tumor models (9, 33).

Tumor-induced CD11b+ cells from KO spleens had an intact suppressive activity on alloreactive CTLs (Figure 3A) and released IL-13 and IFN-γ as well as the WT counterparts (Figure 2). These data confirm that our results unveil a different pathway of suppression, not dependent on either NK or NKT cells. Moreover, since Rag2–/–γc–/– KO mice are also deficient in T and B lymphocytes and lack the common γ chain that is a component of the type I IL-4R, we can conclude that (a) the immunosuppressive properties of CD11b+ cells are a likely outcome of the direct activity of tumor-released factors on the myeloid precursors without requiring any additional intervention from cells of the innate or adaptive immune system; and (b) type I IL-4R, which binds only IL-4, does not play any role in the myeloid-dependent suppression. We can thus conclude that a successful suppression of CTL activity by tumor-induced CD11b+ cells depends on the integration of signals coming from the IFN-γ, released both by activated T lymphocytes and from the same CD11b+ cells, and IL-4Rα expressed on suppressor cells. These findings are further confirmed by the inability of IFN-γ–/– and IL-4Rα–/– CD11b+ splenocytes of tumor-bearing mice to inhibit the proliferation of purified CD8+ T cells stimulated with anti-CD3 and anti-CD28 mAbs (Supplemental Figure 1B).

The IFN-γ effect seems paradoxical in that CTLs produce IFN-γ, which in turn may act in trans to inhibit CTL function of other CTLs. To address further the role of this cytokine, we added either antibodies blocking IFN-γ or the recombinant cytokines to allo-cultures stimulated in the presence of either WT or IFN-γ KO MSCs (Supplemental Figure 6). These experimental data show that (a) inclusion of antibodies against mouse IFN-γ does not elevate dramatically the lytic response in normal allo-cultures devoid of MSCs, whereas it does restore completely immune responsiveness in cultures containing MSCs; and (b) exogenous IFN-γ can efficiently replace the cytokine produced by either T lymphocytes or MSCs (derived from either the spleen or tumor infiltrate) in sustaining T lymphocyte inhibition. The total amount of cytokine added to the culture to obtain this effect was rather high (50 ng/ml), in keeping with the notion that substantial and prolonged release of IFN-γ is necessary to exert the inhibitory effect on CTL activity.

CD11b+IL-4Rα+ cells constitute a subset of inflammatory monocytes able to produce IL-13 and IFN-γ. Having identified IL-4Rα as a possible MSC marker, we separated tumor-induced CD11b+ splenocytes according to its expression. This procedure distinguished 2 subpopulations of CD11b+ cells, the monocytes (IL-4Rα+) and the polymorphonuclear cells (IL-4Rα–) (Figure 4A). With this approach, we could use a single cell surface marker to differentiate the 2 main signatures unveiled by microchip analysis. The IL-4Rα+ fraction also contained some cells with a more immature morphology consistent with myelomonocytic precursors. The IL-4Rα+ but not the IL-4Rα– sorted cells constitutively released IL-13 and IFN-γ and suppressed the generation of alloreactive CTLs (Figure 4, B and C), indicating that the expression of IL-4Rα can indeed be considered a marker of tumor-induced, circulating MSCs.

Morphological and functional characterization of tumor-induced CD11b+IL-4RαFigure 4

Morphological and functional characterization of tumor-induced CD11b+IL-4Rα+ and CD11b+IL-4Rα– cells. Splenocytes were sorted by microbeads to select CD11b+ cells, and this cellular population was subsequently sorted by flow cytometry to select IL-4Rα+ (left panels) and IL-4Rα– (right panels) cells. (A) The sorted cells were stained with H&E and May-Grünwald-Giemsa stain (M-GG). Pictures were taken under microscopy at ×40 magnification. CD11b+IL-4Rα+ cells showed a monocytic morphology, while CD11b+IL-4Rα– cells comprised granulocytes at various differentiation stages, including immature cells with band morphology. (B) Both populations were analyzed by flow cytometry for their expression of IL-13 (top panels) and IFN-γ (bottom panels). Open histograms indicate staining with IL-13 or IFN-γ antibody; filled histograms indicate isotype-matched antibody control. Data are representative of at least 3 determinations in separate experiments. (C) The 2 populations were added at a final concentration of 3% to alloantigen-stimulated cultures (see above). CD11b+IL-4Rα+ but not CD11b+IL-4Rα– cells suppressed the generation of alloreactive CTLs (P < 0.01 and P = 0.48, respectively). Results are shown as the fraction of LU30 measured in the control allo-cultures lacking third-party CD11b+ cells. Data are mean ± SEM from 3 experiments.

In mice bearing a 9-day-old s.c. C26-GM tumor, CD11b+IL-4Rα+ monocytes are present not only in the spleen but also in the blood, where they can constitute a large fraction of the total mononuclear cells (48.2% ± 6.52% in tumor-bearing versus 1.2% ± 0.68% in tumor-free mice). Circulating mouse monocytes comprise at least 2 distinct subpopulations: CD11b+Gr-1–CCR2–CX3CR1high monocytes are precursors of resident macrophages in normal tissues, whereas CD11b+Gr-1+CCR2+CX3CR1low cells are “inflammatory” monocytes that home to sites of inflammation, where they serve as a reservoir of dendritic cells and activated macrophages (34, 35). Flow cytometry analysis on sorted CD11b+ cells from tumor-bearing hosts indicates that the IL-4Rα+ splenocytes express a low level of CX3CR1 and are positive for CCR2 and CD62L (Figure 5A). Moreover, about half of the CD11b+IL-4Rα+ monocytes are Gr-1+, bona fide inflammatory monocytes (Figure 5A). However, Gr-1+ and Gr-1– fractions might be part of the same continuum, since CD11b+Gr-1+ cells can differentiate in vitro and in vivo into CD11b–single positive cells (1, 2, 5, 14, 35).

Phenotypic characterization of tumor-induced CD11b+IL-4Rα+ cells.Figure 5

Phenotypic characterization of tumor-induced CD11b+IL-4Rα+ cells. (A) CD11b+ cells from spleens of mice inoculated 9 days earlier with C26-GM tumor cell lines were enriched through immunomagnetic microbeads and stained with the indicated antibody. FS, forward scatter; SS, side scatter. (B) Blood cells (top rows) and spleen cells (bottom rows) from mice of various strains inoculated with different tumors were stained for CD11b and IL-4Rα. The following tumor cell lines were used: the BALB/c-derived colon carcinoma CT26 (closely related to the C26 tumor used in our previous experiments) and mammary carcinoma 4T1; and the C57BL/6–derived EL-4 lymphoma and MCA203 fibrosarcoma. Data are from a single experiment representative of 3. (C) CD11b+ cells were sorted from the spleens of tumor-bearing WT or KO BALB/c mice and added at a final concentration of 3% to a mixed leukocyte culture set up as previously described (top panels). CD11b+ cells sorted from the spleens of tumor-bearing C57BL/6 mice were added at a final concentration of 3% to a mixed leukocyte culture set up with C57BL/6 effectors stimulated with an equal number of γ-irradiated BALB/c splenocytes, in the presence or absence of anti–IFN-γ and/or anti–IL-13 mAbs added at the beginning and at the third day of culture (bottom panels).

To evaluate whether CD11b+IL-4Rα+ cells could be induced by unmanipulated tumors, we analyzed the blood and the spleen of mice bearing s.c. tumors known to induce systemic immune dysfunction (2, 5, 10, 14, 17). Tumors were allowed to grow until they reached about 1 cm2, and then pools of blood samples and spleens were stained. In all cases, blood and spleen contained various numbers of CD11b+IL-4Rα+ cells, whereas these cells were very few in the tumor-free BALB/c and C57BL/6 mice (Figure 5B).

Even though CT26, a colon carcinoma closely related to the C26 cell line from which the C26-GM tumor model was originated, did not induce high numbers of CD11b+ splenocytes, they were equally suppressive on a per-cell basis (Figure 5C), indicating that GM-CSF transduction was basically enhancing an innate property of the tumor cells. More importantly, CD11b+ cells in the spleens of CT26 tumor–bearing mice behaved as did the cells described previously, since they required the ability to produce IFN-γ and the expression of IL-4Rα in order to be immunosuppressive (Figure 5C). Similar results were obtained with the CD11b+ cells enriched from spleens of BALB/c mice bearing the mammary carcinoma 4T1 (Figure 5C).

Since some key KO mice were not available in the H-2b background, we used a different approach for MCA203 and EL-4 tumors growing in syngeneic C57BL/6 mice. We added mAbs blocking either IL-13 or IFN-γ antibodies to allo-cultures containing, as a third party, CD11b+ splenocytes from tumor-bearing mice. Whereas alloreactive T lymphocytes were generated in standard allo-cultures from tumor-free mice, there was a complete loss of this alloresponse in cultures receiving CD11b+ splenocytes conditioned by either MCA203 or EL-4 tumors. Active cytotoxic effectors could be recovered by addition of either anti–IFN-γ or anti–IL-13 mAbs but not a control mAb, which suggests that each cytokine had a key function in sustaining the suppressive response; moreover, the combination of blocking mAbs resulted in an additive/synergistic activity, since it allowed to double the number of alloreactive T lymphocytes recovered in the cultures (Figure 5C). Regardless of the tumor type and the mouse genetic background, these data indicate that the interplay between the antithetical cytokines IFN-γ and IL-13 (via IL-4Rα) underlies the immunosuppressive pathway.

IL-4Rα expression is required for the in vivo inhibitory activity of myeloid cells in tumor-bearing hosts. To prove that IL-4Rα is not just a cell marker but is critical for the function of suppressive myeloid cells, we inoculated s.c. C26-GM cells in LysMCreIL-4R–/flox mice that have a targeted knockout of the IL-4Rα in macrophages and neutrophils (36). Tumors grew similarly in IL-4Rα conditional KO mice and control littermates. The transfer of CD8+ T lymphocytes purified from mice immunized with γ-irradiated C26-GM cells marginally affected the tumor development in control mice but completely prevented the tumor growth in LysMCreIL-4R–/flox mice (Figure 6A). Moreover, CD8+ T lymphocytes staining with a tetramer loaded with AH1 peptide (AH1-TET), one of the major C26-GM–associated antigens (10, 37), were increased in the blood of LysMCreIL-4R–flox but not of control mice after adoptive transfer to tumor-bearing hosts (Figure 6B). This difference was even more evident when the total number of AH1-TET+ cells in the blood was considered, because of a reduction in the number of CD3+CD8+ T cells circulating in the blood of control mice (Figure 6C). We estimated the number of circulating tumor-specific CD8+ T cells, taking into account the published figures of circulating white blood cells in BALB/c mice (38). We transferred only 57,500 AH1-specific CD8+ T lymphocytes, and we calculated a total of 144,503 (± 44,579) AH1-TET+CD3+CD8+ T cells circulating in the blood of LysMCreIL-4R–/flox tumor-bearing mice 8 days after adoptive transfer. These mathematical considerations indicate that tumor-specific T cells expanded in LysMCreIL-4R–/flox tumor-bearing mice but not in control littermates (Figure 6C). Thus, tumor-specific CD8+ T lymphocytes are inhibited by C26-GM tumor growth in vivo, and this inhibition critically requires the expression of IL-4Rα in myeloid cells.

Adoptive cell transfer is effective only in mice with a myeloid-restrictedFigure 6

Adoptive cell transfer is effective only in mice with a myeloid-restricted inactivation of IL-4Rα. (A) LysMCreIL-4R–/flox mice and control littermates were challenged s.c. on day 0 with C26-GM cells and were given, 24 hours later, CD8+ T cells purified from the spleens and lymph nodes of mice vaccinated 14 days before with γ-irradiated C26-GM cells. Tumor size is indicated as the product of the 2 main perpendicular diameters measured with a caliper. Mice with tumors had to be euthanized on day 12, since they were showing severe signs of distress. (B) The CD3+CD8+ cells positive for the Ld tetramer loaded with AH1 peptide were determined in the blood of tumor-bearing mice 8 days after tumor inoculation. One dot plot for each group is reported. Numbers indicate the percentages of AH1-TET+ cells among gated CD3+CD8+ T lymphocytes (mean ± SEM, n = 5–6). (C) The absolute numbers of CD3+CD8+AH1-TET+ cells were calculated for the total number of circulating white blood cells (WBCs) and reported as mean ± SEM. The background staining given by control β-gal876–884/Ld TET was subtracted for each determination. (D) IFN-γ–/– and WT BALB/c mice were challenged s.c. on day 0 with C26-GM cells and given, 24 hours later, CD8+ T cells purified from the spleens and lymph nodes of IFN-γ–/– and WT BALB/c mice, vaccinated 14 days before with γ-irradiated C26-GM cells. Results, expressed as survival rates after tumor challenge, are from 1 representative experiment with 6 mice per group.

We also evaluated the importance of IFN-γ by adoptively transferring tumor-specific CD8+ T cells isolated from either WT or IFN-γ–/– mice to WT or IFN-γ–/– tumor-bearing mice (Figure 6D). In this way, we could evaluate the relevance of IFN-γ produced by antitumor T cells or by cells of the tumor-conditioned host. In agreement with the in vitro data, a full antitumor activity of adoptive immunotherapy could be obtained in the complete absence of IFN-γ, since the IFN-γ–/– mice receiving IFN-γ–/– CD8+ T cells were completely cured and survived for the entire observation period (Figure 6D; P = 0.001). Moreover, about 65% of IFN-γ–/– mice bearing C26-GM were cured by transfer of WT CD8+ T cells (P = 0.008), suggesting that the endogenous production of IFN-γ is more relevant that the release of IFN-γ by effector lymphocytes in regulating tumor-induced suppression in vivo (Figure 6D).

IL-13 and IFN-γ collaborate to activate the suppressive metabolic pathways in tumor-induced CD11b+ cells. Findings in KO mice and experiments with blocking mAbs indicate that MSCs can integrate the signals received from IL-13 and IFN-γ to activate their suppressive pathways. We evaluated whether antagonist cytokines such as IL-13 and IFN-γ could have a synergistic or additive effect on MSCs. Tumor-induced CD11b+ splenocytes cultured in vitro progressively lack the expression of IL-4Rα+ as a consequence of the autocrine release and binding of IL-13 to this receptor (Figure 7A). Receptor loss, in fact, was delayed by addition of an IL-13 blocking mAb (Figure 7A). Inhibition of IL-4Rα downregulation was also achieved with the addition of IFN-γ to CD11b+ cell cultures. This effect was likely independent from an IFN-γ–mediated inhibition of IL-13 release, since the combination of IFN-γ and IL-13 blocking mAbs had a synergistic effect on IL-4Rα persistence (Figure 7A). Moreover, IFN-γ did not modify the amount of secreted IL-13 monitored by ELISA in the supernatant of CD11b+ cells during the in vitro culture (data not shown). The IL-4Rα was negligible in CD11b+ cells enriched from the spleen of tumor-free mice, and IFN-γ did not affect its expression levels (Figure 7A).

IFN-γ and IL-13 cooperate in the activation of immunosuppressive pathways.Figure 7

IFN-γ and IL-13 cooperate in the activation of immunosuppressive pathways. (A) Kinetics of IL-4Rα expression at the cell surface of tumor-induced CD11b+ splenocytes. CD11b+ splenocytes from tumor-free (normal) or tumor-bearing mice (tumor) were incubated for 72 hours alone, with IFN-γ, with a neutralizing antibody against mouse IL-13, or with both. Isotype-matched antibodies were included as negative controls. IL-4Rα protein expression on the cell surface was analyzed at various time points by flow cytofluorimetry and reported as the mean percentage of positive cells ± SEM of 3 separate determinations. Differences between diverging kinetics values were statistically significant (P < 0.05). (BD) Effect of CD11b+ cell exposure to IL-13 and IFN-γ. CD11b+ cells were sorted from the spleens of tumor-free and tumor-bearing mice. These last cells were analyzed immediately (Fresh) or cultured for 48 hours in medium alone (No cytokine) or in the presence of IFN-γ, IL-13, or both. Some samples were treated either with IFN-γ for 24 hours followed by addition of IL-13 for a further 24 hours (IFN-γ → IL-13) or with the inverse cytokine combination (IL-13 → IFN-γ). Cells were then analyzed for expression of Arg1 and Nos2 mRNA by real-time PCR (B), for Arg1 and Nos2 protein levels by Western blotting (C), and for relative enzyme activity (D). Log2FC is the log-transformed gene expression value of the target gene, normalized to an endogenous reference and relative to a calibrator sample. ND, not done. Data for real-time-PCR, Western blot, and enzyme activity are from the same experiment representative of 3.

To address further the basis of IL-13 and IFN-γ cooperation in MSC suppression, we cultured tumor-induced CD11b+ splenocytes for 48 hours in complete medium with or without IFN-γ or IL-13. We examined the effects different combinations of IFN-γ and IL-13 would have on the expression of the enzymes Arg1 and Nos2, which are the key molecules responsible for MSC-dependent suppression of T cells (Figure 7, B and C).

Fresh and cultured CD11b+ splenocytes isolated from tumor-bearing mice showed Nos2 mRNA upregulation, while the relative proteins remained below the detection limits of the immunoblot (Figure 7, B and C). Arg1, which is strongly upregulated after 24 hours in cultured CD11b+ cells (Table 1 and unpublished data), returned to basal levels after 48-hour culture (Figure 7B). As expected, when each cytokine was added separately, IFN-γ caused preferential expression of Nos2, whereas IL-13 upregulated Arg1. Importantly, when the cytokines were added simultaneously or in the sequence IL-13 followed by IFN-γ and vice versa, the transcripts, proteins, and functional activities of the 2 enzymes were upregulated rather than inhibited (Figure 7, B and C). The cooperation between IL-13 and IFN-γ, inducing concomitant expression and activation of Arg1 and Nos2, thus identifies a unique property of MSCs. In fact, in inflammatory peritoneal macrophages, IL-13 pretreatment was shown to suppress Nos2 protein (but not mRNA) expression and activity induced by IFN-γ and LPS (39).

Intratumoral CD11b+ cells are potent immune suppressors. It has been suggested that tumor-infiltrating myeloid suppressors are different from circulating precursors from which they derive in various respects: they are promptly suppressive, not requiring in vitro culture, and express enzymatically active Arg1 and Nos2 that are critical for the immunosuppressive pathway since pharmacological interference with both enzymes restores the T lymphocyte response (10, 14). We thus evaluated phenotypic and functional properties of tumor-infiltrating myeloid suppressors. CD11b+ cells isolated from the cellular infiltrate of C26-GM nodules were potent immune suppressors, since they completely prevented cell division of clonotypic CL4 cells (Figure 1B) stimulated by the cognate antigen and induced their extensive cell death by apoptosis (Supplemental Figure 2). As described by others, CD11b+ cells are mostly Gr-1– and express a variable amount of the marker F4/80 (Figure 8A), suggesting a differentiation toward mature macrophages (14, 35, 40). However, a relevant fraction of tumor-associated CD11b+ cells possessed the IL-4Rα (69% ± 1.59%, n = 3). Interestingly, whereas CD62L expression and CX3CR1 expression were similar to those observed in splenic CD11b+ cells, the CCR2 was absent (Figure 8A), a likely consequence of the local production of the chemokine CCL2, detected by RT-PCR in the very same intratumoral CD11b+ cells (data not shown).

Phenotypic and functional characterization of CD11b+ cells infiltrating theFigure 8

Phenotypic and functional characterization of CD11b+ cells infiltrating the tumor. (A) CD11b+ cells were separated from the disaggregated nodules of C26-GM tumor and stained with the indicated mAbs. (B) ARG1 and NOS2 protein expression level in CD11b+ cells isolated from tumors of WT, IL-4Rα–/–, and IFN-γ–/– BALB/c mice, analyzed immediately (Fresh) and after 24 hours of culture in standard medium. (C) Cytotoxicity of WT and IFN-γ–/– BALB/c splenocytes (Responder), stimulated with an equal number of γ-irradiated C57BL/6 splenocytes and cocultured without (None) or with CD11b+ cells sorted from tumors of WT, IL-4Rα–/–, and IFN-γ–/– BALB/c mice. Data are the mean of 2 separate experiments.

Tumor-infiltrating CD11b+ cells constitutively expressed Arg1 and Nos2 protein, and this expression was further enhanced by the in vitro culture in standard medium. As expected, a reduction of Arg1 protein levels was observed in fresh and cultured CD11b+ cells isolated from tumor-bearing IL-4Rα–/– mice. Interestingly, reduction in the level of Nos2 protein was detected in both IFN-γ–/– and IL-4Rα–/– mice (Figure 8B). Despite the slight phenotypic differences described above, intratumoral CD11b+ cells behaved functionally as did the splenic counterpart isolated in tumor-bearing mice: in fact, suppression of alloreactive T cells required IFN-γ release by activated T cells, autocrine production of IFN-γ, and the activation of the IL-13/IL-4Rα signaling pathway in CD11b+ cells (Figure 8C).