RAGE is a multifunctional receptor implicated in diverse processes including inflammation and cancer. In this study, we report that RAGE expression is upregulated widely in aggressive triple-negative breast cancer (TNBC) cells, both in primary tumors and in lymph node metastases. In evaluating the functional contributions of RAGE in breast cancer, we found that RAGE-deficient mice displayed a reduced propensity for breast tumor growth. In an established model of lung metastasis, systemic blockade by injection of a RAGE neutralizing antibody inhibited metastasis development. Mechanistic investigations revealed that RAGE bound to the proinflammatory ligand S100A7 and mediated its ability to activate ERK, NF-κB, and cell migration. In an S100A7 transgenic mouse model of breast cancer (mS100a7a15 mice), administration of either RAGE neutralizing antibody or soluble RAGE was sufficient to inhibit tumor progression and metastasis. In this model, we found that RAGE/S100A7 conditioned the tumor microenvironment by driving the recruitment of MMP9-positive tumor-associated macrophages. Overall, our results highlight RAGE as a candidate biomarker for TNBCs, and they reveal a functional role for RAGE/S100A7 signaling in linking inflammation to aggressive breast cancer development. Cancer Res; 75(6); 974–85. ©2015 AACR.

Receptor for advanced glycation endproducts (RAGE) is the signal transduction receptor that senses a variety of signaling molecules (1). The variety of ligands allows RAGE to be implicated in a wide spectrum of pathologic conditions such as inflammation and cancer (1, 2). Epidemiologic and molecular studies, including mouse models, have shown that if inflammation is prolonged, it promotes cancer development (3–6). It is now believed that most solid tumors, including those in the breast, have an inflammatory microenvironment (4, 5). RAGE expression and activation have been shown to be associated with chronic inflammation, which in turn enhances the malignant transformation of various cancers (1, 2, 7–11). However, its role in breast cancer, especially in the modulation of breast cancer microenvironment, is unknown.

RAGE was first described as a receptor for advanced glycation end products (AGE), but it has since been shown to be the receptor for several other molecules involved in innate immunity, including high mobility group box 1 peptide (HMGB-1), amyloid-β peptide, and the S100 family of proteins (1, 2). Phorbol ester 12-O-tetradecanoylphorbol-13-acetate-induced proinflammatory mediators were shown to be decreased in RAGE-deficient mice (7), which suggests that RAGE expression is involved in sustaining inflammation and cancer (1, 2, 7, 9). It has also been well-documented that RAGE ligands bind to RAGE and activate its downstream signaling mechanisms that sustain chronic inflammatory conditions, leading to neoplastic stage (1, 12, 13). It is interesting to note that there is very low or no RAGE expression in normal tissues but enhanced expression in chronic inflammation and cancer (2, 10). Although, these features of RAGE make it an ideal candidate for therapeutic strategies against chronic inflammation, not much is known about its role in breast cancer.

RAGE has been shown to bind to its ligand S100A7 in keratinocytes and leukocytes (14, 15). S100A7 has been shown to be highly expressed in estrogen receptor (ER)α breast cancer (16, 17). It is believed that S100A7 mediates breast cancer growth and metastasis by recruiting proinflammatory cell infiltrates (18, 19). Also, proinflammatory cytokines enhance triple-negative breast cancer (TNBC) growth and metastasis (20). However, very little is known about mechanisms through which RAGE/S100A7 axis modulates tumor microenvironment and enhances breast cancer growth and metastasis.

Macrophages can be divided into subtypes M1 and M2, where M1 macrophages are associated with an anti-cancer phenotype and M2 macrophages express a pro-cancer phenotype (21, 22). Cytokines/chemokines and growth factors modulate the tumor microenvironment, which could directly/indirectly polarize macrophages toward the M2 tumor-associated macrophages (M2-TAM) phenotype (22, 23).

In this investigation, we for the first time show that RAGE is expressed in a panel of aggressive breast cancer cell lines, TNBC, and metastatic lymph node deposits. We also demonstrate that blocking of RAGE reduces tumor metastasis and that RAGE ablation inhibits breast cancer growth. In addition, we show that RAGE mediates its tumor-promoting effects in breast cancer through binding to S100A7. Our findings also uncovered that the RAGE/S100A7 pathway enhanced breast cancer growth and metastasis. These studies further demonstrate that RAGE neutralizing antibodies/soluble RAGE could be used to inhibit breast cancer growth and metastasis, especially in S100A7-expressing invasive cancers. Furthermore, these studies suggest that RAGE could be used as novel biomarker and therapeutic strategy against TNBCs.

Patients

Institutional Review Board of the Ohio State University (OSU; Columbus, OH) has approved protocol for the constructed TNBC tissue microarrays (TMA; n = 173). The clinical and pathologic characteristics of TNBC TMAs have been recently described (24). TMA for lymph node metastasis (BR1008) was obtained from US Biomax, Inc.

Immunohistochemistry, immunofluorescence, and ELISA

Samples from mammary glands and tumors were formalin-fixed and paraffin-embedded (18). Standard IHC techniques were used according to the manufacturer's recommendations (Vector Laboratories) using antibodies against RAGE (Abcam, 1:400) Ki67 (Neomarkers, 1:100), CD31 (Santa Cruz 1:100), F4/80 (AbD Serotec, 1:50), arginase 1 (Santa Cruz, 1:200), and iNOS (Abcam, 1:200) for 60 minutes at room temperature. Vectastain Elite ABC reagents (Vector Laboratories), using avidin DH:biotinylated horseradish peroxidase H complex with 3,3′-diaminobenzidine (Polysciences) and Mayer's hematoxylin (Fisher Scientific), were used for detection of bound antibodies. Staining of TMAs was graded as previously described (25). Immunofluorescence was performed on paraffin-embedded tissues. Briefly, sections were stained with F4/80 (1:75), and MMP9 (R&D Systems, 1:150). Alexa Fluor–conjugated secondary antibodies (Life Technologies) were used to detect primary antibody. Sections were mounted by VECTASHIELD mounting media containing DAPI (Vector Laboratories, Inc.). Images were analyzed by confocal microscopy. Binding of RAGE with human recombinant S100A7 was performed as described (26).

Cancer patient data analysis

High RAGE and S100A7 expressions were defined as overexpression of ager mRNA being greater than 0.5-fold and overexpression of s100a7 being greater than 1.0-fold of SD above the mean, respectively. Association of gene expression alterations was performed on the basis of The Cancer Genome Atlas (TCGA) database by Fisher exact test. Analysis of RAGE expression between basal and non-basal breast cancer samples was based on a subtype-specific breast cancer study (GEO accession GDS2250; ref. 27). For Kaplan–Meier survival analysis, patient samples with RAGE expression values greater than its median were grouped as high RAGE and the other half as low RAGE.

Cell culture

Murine macrophage cell line RAW264.7 and human breast carcinoma cell lines MDA-MB-231, MDA-MB-453, MCF7, T47D, BT-474 were obtained from ATCC. SCP2 cells were kindly provided by Dr. Massague (28). MVT-1 cells (derived from MMTV-c-Myc; MMTV-VEGF bitransgenic mice) were obtained from Dr. Johnson and PyMT cells derived from MMTV-PyMT C57BL/6 mice were obtained from Dr. Hai (OSU; ref. 29). MVT-1 highly metastatic clone, PyMT, Met1, and 4T1 cells were cultured as described (18, 29).

Chemotaxis

Chemotactic assays were performed using Transwell chambers (Costar; 8-μm pore size) as described (18, 30).

Mice

Nude mice were obtained from Charles River. C57B/6 background RAGE−/− mice were kindly provided by Dr. Schmidt (New York University, New York, NY), and TetO-mS100a7a15 mice were kindly provided by Dr. Yuspa (NIH; Bethesda, MD). TetO-mS100a7a15 mice (15) were cross-bred with MMTV-rtTA mice to generate bitransgenic MMTV-mS100a7a15 mice. Knockout and transgenic littermates were genotyped by PCR. Female MMTV-mS100a7a15 mice were fed with doxycycline-chow 1 g/kg (Bio-Serv), and mice with normal diet served as controls. All mice were kept in The OSU's animal facility in compliance with the guidelines and protocols approved by the OSU Institutional Animal Care and Use Committee.

Orthotopic injection assay

MVT-1 or PyMT cells were injected into the mammary glands of transgenic or knockout mice. Transgenic mice injected with MVT-1 cells were either fed with doxycycline-chow 1 g/kg or normal diet (control). Tumors were measured weekly with external calipers and volume was calculated according to the formula V = 0.52 × a2 × b, where a is the smallest superficial diameter and b is the largest superficial diameter. Orthotopically injected animals were sacrificed and tumors were excised (18). RAGE neutralizing antibody (human or murine) and soluble RAGE (human or murine) were purchased from R&D Systems.

FACS analysis

Freshly prepared single-cell suspensions of tumor-infiltrating cells were incubated with anti-F4/80 PE or anti-CD11b APC (18). RAGE expression was analyzed by staining with RAGE antibody (Abcam) followed by Alexa Fluor 488 antibody. After staining, cells were analyzed by FACS Caliber using CellQuest software (BD Biosciences).

Western blot and coimmunoprecipitation

Western blot analysis of cell or tumor lysates was done as described (30). Coimmunoprecipitation was carried out using protein G plus A-agarose beads as described (31), with S100A7 rabbit (Novus Biologicals) and RAGE mouse (Santa Cruz Biotechnology) antibodies.

Luciferase reporter assay

NF-κB activity was determined using NF-κB luciferase reporter assay (Promega) per manufacturer's protocol.

Statistical analysis

To test the association between two categorical variables, χ2 tests or Fisher exact tests were used. For continuous variables, 2-sample t tests were used if two groups were compared, and ANOVAs were used if more than two groups were compared. * or # indicates P < 0.05; ** or ## indicates P < 0.01.

RAGE is expressed in highly metastatic breast cancer cells and its expression correlates with worse clinical prognosis

We analyzed RAGE expression in a panel of breast cancer cell lines. RAGE expression was higher in metastatic TNBC cell lines, whereas low or no RAGE expression was observed in ERα+ breast cancer cell lines (MCF7, T47D, and BT474; Fig. 1A and B), which are weakly metastatic (32–34). These data suggest that RAGE is predominantly expressed in ERα and highly metastatic breast cancer cell lines. To test the correlation of RAGE with ERα status, we analyzed open-access Gene Expression Omnibus (GEO) datasets for the expression of RAGE. In a subtype-specific breast cancer study (GEO accession GDS2250), RAGE expression is significantly enhanced in basal type (majorly TNBC) invasive breast cancer patient tumor samples compared with non-basal type tumors (majorly ERα+ cancer; Fig. 1C). Next, we analyzed open-access dataset for RAGE expression. We found that high RAGE expression was observed in invasive breast cancer (IBC) compared with normal control (Fig. 1D). Furthermore, we analyzed the expression of RAGE in breast cancer TMAs with accompanying outcome data and other clinical information by IHC. We found that 92% of the samples showed high RAGE expression in TNBC tissues (Fig. 1E and F). However, RAGE was expressed only in 29% HER2-positive TMAs (Fig. 1E and F). Using another TMA that contained metastatic and malignant patient samples, we found that metastatic tissue tends to have higher RAGE expression than malignant tissue (P < 0.0001; Fig. 1G). Next, we analyzed open-access dataset for clinical outcome of RAGE expression. We found that high RAGE expression was associated with poor prognosis (Fig. 1H). Taken together, these results show that RAGE expression is associated with highly aggressive and metastatic breast cancers, including TNBC.

Figure 1.

RAGE expression in breast cancer cell lines and patient samples. A, FACS analysis of RAGE in human TNBC, ERα+, and highly metastatic murine mammary tumor cell lines. B, quantification of RAGE expression obtained by FACS. C, RAGE expression in basal and non-basal type breast cancer. RAGE (ager) expression values are normalized to β-actin (actb). D, RAGE expression in normal and invasive breast cancer. E, representative photographs of RAGE expression in TNBC TMA. F, bar graph showing RAGE expression in TNBC (n = 80) and HER2 (n = 33) TMA samples. G, bar graph showing RAGE expression in metastatic breast cancer. We used TMA (n = 100) that contained n = 40 lymph node metastasis, n = 50 malignant, and n = 10 normal tissues. H, expression level of RAGE predicts survival differences by Kaplan–Meier analysis using R2 microarray dataset. Scale bar, 100 μm. #, P < 0.05; ##, P < 0.01.

Figure 1.

RAGE expression in breast cancer cell lines and patient samples. A, FACS analysis of RAGE in human TNBC, ERα+, and highly metastatic murine mammary tumor cell lines. B, quantification of RAGE expression obtained by FACS. C, RAGE expression in basal and non-basal type breast cancer. RAGE (ager) expression values are normalized to β-actin (actb). D, RAGE expression in normal and invasive breast cancer. E, representative photographs of RAGE expression in TNBC TMA. F, bar graph showing RAGE expression in TNBC (n = 80) and HER2 (n = 33) TMA samples. G, bar graph showing RAGE expression in metastatic breast cancer. We used TMA (n = 100) that contained n = 40 lymph node metastasis, n = 50 malignant, and n = 10 normal tissues. H, expression level of RAGE predicts survival differences by Kaplan–Meier analysis using R2 microarray dataset. Scale bar, 100 μm. #, P < 0.05; ##, P < 0.01.

Close modal

Blockade of RAGE inhibits tumor growth and metastasis in vivo

To investigate the role of RAGE in metastasis, we used the IVIS imaging system to analyze the metastatic potential of RAGE-expressing SCP2 cells. We injected highly metastatic single-cell progeny clone 2 (SCP2) of MDA-MB-231 cell lines intracardially into nude mice and blocked RAGE activity with RAGE neutralizing antibody (naRAGE). As shown in Fig. 2A and B, naRAGE treatment significantly reduced the metastatic potential of SCP2 cells as compared with IgG control–treated mice. Next, we elucidated the role of host RAGE on mammary cancer progression and development using the RAGE−/− model. First, we analyzed surface expression of RAGE on PyMT cells by FACS analysis. As shown in Fig. 2C, PyMT cells expressed high RAGE expression. Next, mice were injected with PyMT cells and observed for tumor growth for 35 days (Fig. 2D). Interestingly, PyMT-derived tumor growth was significantly inhibited in RAGE−/− mice as compared with RAGE+/+ mice (P = 0.0125; Fig. 2E and F). To identify the signaling molecules that are associated with breast cancer tumor growth, we examined the expression of ERK, cyclin D1, and MMP2 in PyMT-derived tumor tissues obtained from RAGE−/− mice. We observed reduced phospho-ERK, MMP2, cyclin D1, CD31, and Ki67 expression in RAGE−/− mice compared with RAGE+/+ mice (Fig. 2G and H). Cyclin D1 and Ki67 are markers for active proliferation (35, 36). These data demonstrate that RAGE blockade significantly inhibits mammary tumor development and progression.

Figure 2.

Inhibition of RAGE reduces breast cancer growth and distant metastasis. A, luciferase-positive SCP2 cells (1 × 105/100 μL) were injected intracardially into nude mice (n = 6) pretreated with mouse RAGE neutralizing (naRAGE) or IgG antibody(20 μg/mouse). Representative bioluminescent images (BLI) show comparative metastases of naRAGE- or IgG-treated mice. B, normalized photon flux of mice treated with naRAGE or IgG. C, RAGE expression was analyzed in PyMT cells by FACS. Red, IgG; black, RAGE. D, PyMT (1 × 106) cells were injected into the mammary gland of RAGE+/+ and RAGE−/− mice (n = 5) and tumor volume was measured every week. E, after 5 weeks, tumors were excised from mice and weighed. F, representative photograph of mice showing tumors dissected from different experimental groups. G, PyMT cell line–derived tumors from RAGE+/+ and RAGE−/− mice were subjected to IHC staining for CD31 and Ki67 expression. H, tumor lysates (50 μg) from RAGE+/+ and RAGE−/− mice were subjected to Western blotting using phospho-ERK (P-ERK), total ERK (T-ERK), cyclin D1, and MMP-2 antibodies. GAPDH served as loading control. Data represent mean ± SD of three independent experiments. Scale bar, 100 μm. *, P < 0.05.

Figure 2.

Inhibition of RAGE reduces breast cancer growth and distant metastasis. A, luciferase-positive SCP2 cells (1 × 105/100 μL) were injected intracardially into nude mice (n = 6) pretreated with mouse RAGE neutralizing (naRAGE) or IgG antibody(20 μg/mouse). Representative bioluminescent images (BLI) show comparative metastases of naRAGE- or IgG-treated mice. B, normalized photon flux of mice treated with naRAGE or IgG. C, RAGE expression was analyzed in PyMT cells by FACS. Red, IgG; black, RAGE. D, PyMT (1 × 106) cells were injected into the mammary gland of RAGE+/+ and RAGE−/− mice (n = 5) and tumor volume was measured every week. E, after 5 weeks, tumors were excised from mice and weighed. F, representative photograph of mice showing tumors dissected from different experimental groups. G, PyMT cell line–derived tumors from RAGE+/+ and RAGE−/− mice were subjected to IHC staining for CD31 and Ki67 expression. H, tumor lysates (50 μg) from RAGE+/+ and RAGE−/− mice were subjected to Western blotting using phospho-ERK (P-ERK), total ERK (T-ERK), cyclin D1, and MMP-2 antibodies. GAPDH served as loading control. Data represent mean ± SD of three independent experiments. Scale bar, 100 μm. *, P < 0.05.

Close modal

RAGE mediates its effect in breast cancer cells through S100A7

RAGE has been shown to bind to S100A7 in various immune cells (14). When analyzing the RAGE and S100A7 expression in breast cancer (TCGA), we discovered that RAGE and S100A7 are often simultaneously upregulated in breast tumors among patients of the IBC cohort (P = 0.0055; Fisher exact test), whereas an irrelevant gene ERK (mapk1) showed no correlation with RAGE or S100A7 (Fig. 3A). This co-currency of gene upregulation implies a functional link between RAGE and S100A7 in breast cancer. Furthermore, we observed enhanced expression of RAGE in S100A7-overexpressing MDA-MB-231 cells by Western blot, FACS, and immunofluorescent analyses (Fig. 3B–D). This suggests that RAGE could be a receptor for S100A7 as ligand in breast cancer cells. We further verified the direct interaction between human RAGE and human S100A7 protein using an ELISA-based binding assay, in which EGFR, an irrelevant receptor, was used as a negative control. As shown in Fig. 3E, the total binding was dependent on RAGE-Fc concentration, whereas no binding was observed between EGFR-Fc and S100A7 protein. To further confirm the association of RAGE with S100A7, we performed a coimmunoprecipitation assay. S100A7 coimmunoprecipitated with RAGE in S100A7-overexpressing MDA-MB-231 cells (Fig. 1F). Next, we analyzed the effect of human S100A7 on MDA-MB-231 and SCP2 cell migration and that of its murine paralog mS100A7a15 on MVT-1 cell migration. S100A7 enhanced the migration of MDA-MB-231 and SCP2 cells, respectively, and these effects were significantly abrogated by blocking RAGE using neutralizing antibodies (Fig. 3G and H). We also showed that its murine ortholog mS100a7a15 enhanced migration of MVT-1 cells (Supplementary Fig. S1A). Next, we analyzed the S100A7-induced wound-healing capacity of MDA-MB-231 and MDA-MB-453 cell lines and observed that RAGE neutralizing antibody significantly abrogated this effect (Supplementary Fig. S1B and S1C). In addition, we observed that S100A7 or mS100a7a15 significantly enhanced the invasion of SCP2 and MVT-1 cell lines (Supplementary Fig. S1D and S1E). To examine whether activation of RAGE/S100A7 enhanced downstream signaling in breast cancer cells, we analyzed ERK activation. We showed that S100A7-induced ERK activation was inhibited by blocking S100A7 with soluble RAGE (sRAGE) in MDA-MB-231 and SCP2 cells (Fig. 3I and J). To further confirm these effects, we used RAGE neutralizing antibody to block S100A7-induced ERK activation in MDA-MB-231 cells (Supplementary Fig. S1F). In addition, we observed that sRAGE inhibits S100A7-induced MMP9 activation in SCP2 cells (Supplementary Fig. S1F). NF-κB has also been shown to be the downstream target of RAGE (37). Using NF-κB reporter assay, we showed that S100A7 significantly enhanced NF-κB activity and this effect was inhibited by naRAGE (Fig. 3K). Taken together, these results imply that the RAGE/S100A7/mS100a7a15 signaling axis is necessary for breast cancer cell migration and invasion.

Figure 3.

RAGE receptor binds to S100A7 and enhances the migration of breast cancer cell lines. A, co-currency of ager (rage) and s100a7 gene upregulation (>1.0 SD) was analyzed as described (TCGA, Nature, 2012; ref. 50). An irrelevant gene mapk1 (ERK gene) was used as control, which showed no association with ager and s100a7. B, RAGE expression was analyzed in S100A7-overexpressing MDA-MB-231 (SA7) cells compared with vec control (Vec) as determined by Western blotting. C, flow cytometric analysis. D, immunofluorescence. E, S100A7 binding to RAGE as determined by ELISA using RAGE-Fc or EGFR-Fc proteins. Graph shows mean ± SEM of three independent experiments. F, one milligram of cell lysates from S100A7-overexpressing MDA-MB-231 cells were subjected to immunoprecipitation with IgG or S100A7 and probed with anti-RAGE antibody. G and H, MDA-MB-231 cells (G) and SCP2 cells (H) were pretreated with RAGE neutralizing antibody (naRAGE) or control IgG (10 μg/mL) for 30 minutes before being subjected to recombinant S100A7 (SA7; 50 ng/mL)-induced migration. I and J, MDA-MB-231 (I) and SCP2 (J) cells were pretreated with soluble RAGE (sRAGE, 50 ng/mL) for 30 minutes before stimulation with S100A7 at the indicated times. Cell lysates (50 μg) were subjected to Western blotting using phospho-ERK (P-ERK) and total ERK (T-ERK). K, MDA-MB-231 cells were transfected with either wild-type or NF-κB plasmid for 24 hours, stimulated with S100A7 (SA7, 100 ng/mL) or IgG (10 μg) or naRAGE (10 μg) for additional 24 hours, lysed, and analyzed for luciferase activity. Renilla luciferase vector served as internal control. Graphs represent mean ± SD for each experiment repeated three times with similar results. *, P < 0.05 and **, P < 0.01 versus con; #, P < 0.05 and ##, P < 0.01 versus SA7 IgG.

Figure 3.

RAGE receptor binds to S100A7 and enhances the migration of breast cancer cell lines. A, co-currency of ager (rage) and s100a7 gene upregulation (>1.0 SD) was analyzed as described (TCGA, Nature, 2012; ref. 50). An irrelevant gene mapk1 (ERK gene) was used as control, which showed no association with ager and s100a7. B, RAGE expression was analyzed in S100A7-overexpressing MDA-MB-231 (SA7) cells compared with vec control (Vec) as determined by Western blotting. C, flow cytometric analysis. D, immunofluorescence. E, S100A7 binding to RAGE as determined by ELISA using RAGE-Fc or EGFR-Fc proteins. Graph shows mean ± SEM of three independent experiments. F, one milligram of cell lysates from S100A7-overexpressing MDA-MB-231 cells were subjected to immunoprecipitation with IgG or S100A7 and probed with anti-RAGE antibody. G and H, MDA-MB-231 cells (G) and SCP2 cells (H) were pretreated with RAGE neutralizing antibody (naRAGE) or control IgG (10 μg/mL) for 30 minutes before being subjected to recombinant S100A7 (SA7; 50 ng/mL)-induced migration. I and J, MDA-MB-231 (I) and SCP2 (J) cells were pretreated with soluble RAGE (sRAGE, 50 ng/mL) for 30 minutes before stimulation with S100A7 at the indicated times. Cell lysates (50 μg) were subjected to Western blotting using phospho-ERK (P-ERK) and total ERK (T-ERK). K, MDA-MB-231 cells were transfected with either wild-type or NF-κB plasmid for 24 hours, stimulated with S100A7 (SA7, 100 ng/mL) or IgG (10 μg) or naRAGE (10 μg) for additional 24 hours, lysed, and analyzed for luciferase activity. Renilla luciferase vector served as internal control. Graphs represent mean ± SD for each experiment repeated three times with similar results. *, P < 0.05 and **, P < 0.01 versus con; #, P < 0.05 and ##, P < 0.01 versus SA7 IgG.

Close modal

Blockade of RAGE inhibits mammary tumor growth and metastasis in inducible MMTV-mS100a7a15 mice

To analyze the relevance of RAGE/mS100a7a15 in a mammary tumor growth and metastasis model, we used an MVT-1 syngeneic orthotopic model that recapitulates the stages of human primary tumors. We injected MVT-1 cells into inducible MMTV-mS100a7a15 transgenic mice and blocked RAGE with neutralizing RAGE antibody. Mice were treated with doxycycline-chow (1 g/kg) 1 week before MVT-1 injection to switch on the expression of mS100a7a15. When tumors grew as large as 100 mm3, the doxycycline-treated group was given naRAGE (20 μg/mouse) or IgG (20 μg/mouse) intraperitoneally 3 times a week for 20 days. MMTV-mS100a7a15 mice fed with normal chow were used as a negative control. Inducible mice treated with naRAGE showed reduced tumor progression compared with the IgG-treated group (Fig. 4A–C). To determine whether a blocking ligand of RAGE inhibits mammary tumor progression, we used soluble RAGE (sRAGE). Mice were fed with doxycycline-chow 1 week before the injection of MVT-1 cells into the #4 mammary gland. After day 1 of doxycycline induction, mice were injected intraperitoneally with murine sRAGE (2 μg/mouse) or PBS. MMTV-mS100a7a15 mice who received normal chow served as a negative control. As shown in Fig. 4D–F, sRAGE treatment significantly reduced tumor progression in the doxycycline-induced group as compared with the PBS doxycycline-induced group. Furthermore, we observed that blocking RAGE or mS100a7a15 substantially decreased proliferation and angiogenesis in MVT-1–derived tumors obtained from inducible MMTV-mS100a7a15 mice (Fig. 4G and H). Next, we investigated whether RAGE inhibition reduces surface lung metastases in inducible MMTV-mS100a7a15 mice. We observed a significant decrease in surface lung metastases in the mice treated with naRAGE (Fig. 4I and J) or sRAGE (Fig. 4K and L) in MVT-1–derived tumors obtained from inducible MMTV-mS100a7a15 mice as compared with control groups. Taken together, these results suggest that RAGE plays an important role in mS100a7a15-induced breast cancer progression and metastasis.

Figure 4.

Blockade of RAGE inhibits mammary tumor growth and metastasis in MMTV-mS100a7a15–inducible mice. A, MVT-1 (1 × 105) cells were injected into the mammary gland of doxycycline-treated (+Dox) or untreated (−Dox) MMTV-mS100a7a15 mice (n = 5). Dox-treated mice (n = 10) were injected intraperitoneally with RAGE neutralizing antibody (naRAGE) or control IgG (20 μg/mouse) for every alternate day and tumor volume was measured every week. B, after 28 days, the tumors were excised. C, representative photograph of tumors dissected from different experimental groups. D, MVT-1 (1 × 105) cells were injected into the mammary gland of +Dox or −Dox MMTV-mS100a7a15 mice (n = 5). Dox-treated MMTV-mS100a7a15 mice (n = 10) were either treated with sRAGE (SR, 2 μg/mouse) or PBS for every alternate day and tumor volume was measured every week. E, after 28 days, the tumors were excised. F, representative photograph of tumors dissected from different experimental groups. IHCs were performed for CD31 and Ki67 in tumors from Dox-induced MMTV-mS100a7a15 mice that were either treated with naRAGE or control IgG (G) or SR (H). Data represent mean ± SD of three independent experiments. Scale bar, 100 μm. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS. I, left, representative photograph of lungs dissected from naRAGE-treated groups. Right, hematoxylin and eosin (H&E) staining of metastatic deposits. J, bar graph showing the number of metastatic nodules on the lungs (18). K, left, representative photograph of lungs dissected from SR-treated groups. Right, hematoxylin and eosin (H&E) staining of metastatic deposits. L, bar graph showing the number of metastatic nodules on the lungs. Data represent mean ± SD per experimental group. Scale bar, 20 μm. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS.

Figure 4.

Blockade of RAGE inhibits mammary tumor growth and metastasis in MMTV-mS100a7a15–inducible mice. A, MVT-1 (1 × 105) cells were injected into the mammary gland of doxycycline-treated (+Dox) or untreated (−Dox) MMTV-mS100a7a15 mice (n = 5). Dox-treated mice (n = 10) were injected intraperitoneally with RAGE neutralizing antibody (naRAGE) or control IgG (20 μg/mouse) for every alternate day and tumor volume was measured every week. B, after 28 days, the tumors were excised. C, representative photograph of tumors dissected from different experimental groups. D, MVT-1 (1 × 105) cells were injected into the mammary gland of +Dox or −Dox MMTV-mS100a7a15 mice (n = 5). Dox-treated MMTV-mS100a7a15 mice (n = 10) were either treated with sRAGE (SR, 2 μg/mouse) or PBS for every alternate day and tumor volume was measured every week. E, after 28 days, the tumors were excised. F, representative photograph of tumors dissected from different experimental groups. IHCs were performed for CD31 and Ki67 in tumors from Dox-induced MMTV-mS100a7a15 mice that were either treated with naRAGE or control IgG (G) or SR (H). Data represent mean ± SD of three independent experiments. Scale bar, 100 μm. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS. I, left, representative photograph of lungs dissected from naRAGE-treated groups. Right, hematoxylin and eosin (H&E) staining of metastatic deposits. J, bar graph showing the number of metastatic nodules on the lungs (18). K, left, representative photograph of lungs dissected from SR-treated groups. Right, hematoxylin and eosin (H&E) staining of metastatic deposits. L, bar graph showing the number of metastatic nodules on the lungs. Data represent mean ± SD per experimental group. Scale bar, 20 μm. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS.

Close modal

RAGE/S100A7 axis modulates the tumor microenvironment

Macrophages, especially M2-TAMs, have been shown to enhance tumor growth and metastasis (21, 22, 38, 39). To identify the molecular mechanism of RAGE-mediated breast tumor growth and metastasis, we analyzed macrophage recruitment in the PyMT-derived tumors of RAGE+/+ and RAGE−/− mice. As shown in Supplementary Fig. S2, F4/80/Arg1-positive macrophages were substantially reduced in RAGE−/− PyMT–derived tumors as compared with RAGE+/+ tumors. Next, we observed that mS100a7a15-overexpressing mice significantly increased the recruitment of F4/80+/Arg1 macrophages and RAGE blockage by naRAGE treatment significantly reduced the recruitment of F4/80+/Arg1 macrophages compared with IgG control, as analyzed by IHC (Fig. 5A). In addition, we observed substantial decrease in the recruitment of M2 macrophages in MVT-1–derived tumors pretreated with sRAGE in inducible mice compared with PBS-treated mice (Fig. 5B). We also observed reduced CD11b/F4/80+ TAMs by FACS (Fig. 5C). We further showed decreased expression of iNOS (M1 marker) in primary tumors compared with IgG control (Fig. 5A and B). We also analyzed the infiltrations of macrophages in the lung tissues and observed reduced expression of F4/80+ macrophages and Arg1 expression in naRAGE- or sRAGE-treated MMTV-mS100a7a15–inducible mice when compared with control mice (Supplementary Fig. S3). These studies suggest that blockade of RAGE in mS100a7a15 transgenic mice inhibits tumor growth and metastasis through inhibition of M2 macrophage recruitment.

Figure 5.

RAGE/mS100a7a15 regulates TAM recruitment. A, representative IHC staining of macrophage markers (F4/80+, Arg1, and iNOS) in MVT-1 tumors derived from naRAGE or IgG-treated Dox-induced (+Dox) and uninduced (−Dox) MMTV-mS100a7a15 mice. B, intratumoral macrophages were highlighted with IHC for macrophage markers in MVT-1 tumors derived from sRAGE (SR) or PBS-treated Dox-induced and uninduced MMTV-mS100a7a15 mice. Scale bar, 100 μm. C and D, quantitative analyses of F4/80+ macrophages by FACS in MVT-1 tumors derived from naRAGE or IgG-treated +Dox or −Dox MMTV-mS100a7a15 mice. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS.

Figure 5.

RAGE/mS100a7a15 regulates TAM recruitment. A, representative IHC staining of macrophage markers (F4/80+, Arg1, and iNOS) in MVT-1 tumors derived from naRAGE or IgG-treated Dox-induced (+Dox) and uninduced (−Dox) MMTV-mS100a7a15 mice. B, intratumoral macrophages were highlighted with IHC for macrophage markers in MVT-1 tumors derived from sRAGE (SR) or PBS-treated Dox-induced and uninduced MMTV-mS100a7a15 mice. Scale bar, 100 μm. C and D, quantitative analyses of F4/80+ macrophages by FACS in MVT-1 tumors derived from naRAGE or IgG-treated +Dox or −Dox MMTV-mS100a7a15 mice. *, P < 0.05 and **, P < 0.01 versus Dox (−); #, P < 0.05 and ##, P < 0.01 versus Dox (+) IgG or PBS.

Close modal

Because macrophage recruitment to primary tumors plays an important role in promoting S100A7/mS100a7a15-induced metastasis (18), we wanted to know whether recombinant mS100a7a15 might affect macrophage activity in a RAGE-dependent manner. We analyzed the migration of RAW264.7 (RAW), a macrophage cell line, in the presence of mS100a7a15 recombinant protein. We showed that mS100a7a15 significantly enhanced the migration of RAW and that pretreatment with naRAGE significantly abrogated mS100a7a15-induced migration compared with IgG control (Fig. 6A and B). Although S100A7 has been shown to regulate MMPs in cancer cells (17), its role in the macrophage is not known. We analyzed MMP9 secretion in the presence of mS100a7a15 with or without sRAGE treatment. mS100a7a15-induced MMP9 secretion was enhanced in RAW cells and this effect was diminished in the presence of sRAGE (Fig. 6C). Using double immunofluorescence, we also observed enhanced recruitment of MMP9-expressing F4/80+ macrophages in the MVT-1 tumors of MMTV-mS100a7a15–inducible mice compared with noninducible mice. The recruitment of MMP9+/F4/80+ macrophages was substantially decreased in the naRAGE-treated group compared with IgG-treated MMTV-mS100a7a15–inducible mice (Fig. 6D). We further analyzed the molecular mechanism of the mS100a7a15-induced migration of RAW cells. As shown in Fig. 6E and F, mS100a7a15-induced ERK activation was significantly reduced in the presence of sRAGE treatment. These studies suggest that RAGE receptor regulates mS100a7a15-induced ERK activation in macrophages.

Figure 6.

RAGE/mS100a7a15 regulates MMP9+ macrophages. A, RAGE expression was analyzed on RAW cells by FACS. B, RAW cells were subjected to mS100a7a15 (100 ng/mL)-induced migration in presence of murine RAGE neutralizing (naRAGE) or control IgG antibodies. C, RAW cells were stimulated with mS100a7a15 (100 ng/mL) in the presence or absence of sRAGE (SR, 50 ng/mL) for 24 hours. Conditioned media were analyzed for MMP activity. C, bottom, quantification. D, tumors excised from (−Dox) MMTV-mS100a7a15 mice were subjected to double immunofluorescence for MMP9 (green), F4/80 (red), or DAPI (blue). E, RAW cells were pretreated with naRAGE or IgG antibodies for 1 hour, stimulated with mS100a7a15 (SA15, 100 ng/mL), and subjected to Western blotting for P-ERK. Scale bar, 63 um. F, quantification of Western blot analyses. Data represent mean ± SD per experimental group. * or #, P < 0.05 * versus con; # versus SA15 IgG.

Figure 6.

RAGE/mS100a7a15 regulates MMP9+ macrophages. A, RAGE expression was analyzed on RAW cells by FACS. B, RAW cells were subjected to mS100a7a15 (100 ng/mL)-induced migration in presence of murine RAGE neutralizing (naRAGE) or control IgG antibodies. C, RAW cells were stimulated with mS100a7a15 (100 ng/mL) in the presence or absence of sRAGE (SR, 50 ng/mL) for 24 hours. Conditioned media were analyzed for MMP activity. C, bottom, quantification. D, tumors excised from (−Dox) MMTV-mS100a7a15 mice were subjected to double immunofluorescence for MMP9 (green), F4/80 (red), or DAPI (blue). E, RAW cells were pretreated with naRAGE or IgG antibodies for 1 hour, stimulated with mS100a7a15 (SA15, 100 ng/mL), and subjected to Western blotting for P-ERK. Scale bar, 63 um. F, quantification of Western blot analyses. Data represent mean ± SD per experimental group. * or #, P < 0.05 * versus con; # versus SA15 IgG.

Close modal
Figure 7.

Schematic representation of RAGE-mediated S100A7-induced signaling that regulates breast cancer growth and metastasis. Epithelial cells release S100A7/mS100a7a15, which binds to RAGE and activates signaling cascades that recruit TAMs to the tumor stroma. TAMs in turn enhance growth and metastasis by secreting growth factor, chemokines/cytokines, and MMPs. Blocking of RAGE/S100A7 axis by sRAGE or naRAGE may reduce breast tumor growth and metastasis, especially to lungs.

Figure 7.

Schematic representation of RAGE-mediated S100A7-induced signaling that regulates breast cancer growth and metastasis. Epithelial cells release S100A7/mS100a7a15, which binds to RAGE and activates signaling cascades that recruit TAMs to the tumor stroma. TAMs in turn enhance growth and metastasis by secreting growth factor, chemokines/cytokines, and MMPs. Blocking of RAGE/S100A7 axis by sRAGE or naRAGE may reduce breast tumor growth and metastasis, especially to lungs.

Close modal

Emerging data have implicated importance of RAGE in the pathogenesis of various human disorders including cancers (2, 10). The interactions between RAGE and its ligands trigger the activation of MAPK, JAK/STAT, and NF-κB in various cancers (1, 2). In this work, we identified that RAGE plays a critical role in promoting breast cancer growth and metastasis. We documented that RAGE is highly expressed in human TNBC and murine breast cancer cell lines and weakly expressed in low metastatic ER+ cells. However, there are some conflicting reports regarding the expression of RAGE in the MCF7 cell line (40, 41). It could be due to the difference in the techniques that were used. We detected RAGE expression by FACS analysis. However, other studies detected RAGE expression by Western blot analysis. There is possibility that RAGE detected by Western blotting could be a truncated or soluble form of RAGE. In addition, we showed that RAGE is preferentially expressed in invasive and metastatic tumor deposits. This is consistent with a recent report that demonstrated that high RAGE expression was observed in lymph node and distant metastases patients samples (42). Moreover, we observed that RAGE expression was associated with poor prognosis in breast cancer. We also characterized the role of RAGE in breast cancer progression and metastasis. Using a genetic approach, we showed that RAGE ablation significantly reduced PyMT cell–derived tumor growth while blocking RAGE with neutralizing antibodies inhibits breast cancer visceral metastasis in preclinical mouse models. It has been shown recently that RAGE knockdown by siRNA significantly inhibited tumorigenic potential of MDA-MB-231 cell line (40). RAGE ablation in a triple transgenic model of spontaneous pancreatic cancer has also been shown to delay pancreatic cancer development (9, 43).

Moreover, we demonstrated that RAGE binding to S100A7 enhanced RAGE expression in breast cancer cells. In addition, we provide evidence that S100A7 coimmunoprecipitated with RAGE in S100A7-overexpressing MDA-MB-231 cells. Our bioinformatics data also showed that RAGE is co-overexpressed with S100A7 in human breast cancer tissue. Proinflammatory RAGE ligands, such as S100B, S100A4, and S100A8/A9, have been shown to enhance RAGE expression, and continuous activation of RAGE maintains an inflammatory milieu at the tumor microenvironment (2, 44). Our data suggest that S100A7 enhances activation of NF-κB through RAGE activation. We also showed that blocking RAGE or inhibiting S100A7/mS100a7a15 binding to RAGE by soluble RAGE (sRAGE) inhibits breast cancer cell migration and ERK activation. The sRAGE acts as a decoy that prevents ligands from interacting with the cell surface receptor. The application of sRAGE in vitro and in vivo resulted in an effective blockade of RAGE, in accordance with this decoy mechanism, in a range of animal models (11). It is well documented that S100A8/A9-RAGE axis plays a significant role in breast and colon cancer growth and metastasis by modulating its downstream targets such as ERK1/2 (P44/p42), MAPK, and NF-κB signaling pathways (7, 42, 45).

In the current study, we have shown that RAGE deficiency in the host reduced breast cancer growth by decreasing recruitment of TAMs and tumor angiogenesis. We have shown previously that mS100a7a15 overexpression in mammary glands enhanced mammary tumor growth metastasis through macrophage recruitment (18). To further analyze molecular mechanism of these effects, we blocked RAGE activation in MMTV-mS100a7a15–inducible model by naRAGE or sRAGE. Blocking RAGE reduced macrophage recruitment into the MVT-1–derived tumors in the MMTV-mS100a7a15–inducible model. Our study further revealed that blocking the RAGE/mS100a7a15 axis inhibits M2 marker arginase expression. M2-polarized TAMs are known to drive tumor progression by stimulating angiogenesis and metastasis (21, 38, 46). We did not observe a significant change in CD4/CD3/CD8-positive T cells and other immune cells such as natural killer cells, as detected by FACS in MVT-1–derived tumors obtained from MMTV-mS100a7a15–inducible mice treated with RAGE neutralizing antibody or sRAGE.

We showed that blocking of RAGE inhibits mS100a7a15-induced recruitment and MMP9 activation in macrophages. MMP9 has been shown to degrade the extracellular matrix and release growth factors to enhance angiogenesis (47, 48). Furthermore, it has been shown that MMP9 induction by primary tumors in macrophages and the lung endothelium promotes metastasis, especially to lung (48, 49).

In summary, our study shows that RAGE is highly expressed in basal-type breast cancer, especially TNBC, and is preferentially expressed in invasive and lymph node metastasis tissues. As depicted by our model (Fig. 7), elucidation of the molecular mechanism behind enhanced breast cancer growth and metastasis shows that this is likely due to binding of RAGE to S100A7. In turn, the RAGE/S100A7 axis is responsible for enhanced recruitment of MMP9-positive TAMs. We have also shown that RAGE neutralizing antibody and soluble RAGE significantly decrease tumor growth and metastasis in an inducible mS100a7a15 transgenic mouse model. These data imply that the RAGE/S100A7 signaling axis could be used to inhibit TNBC growth and metastasis. Furthermore, these studies demonstrate that RAGE could be used as a novel biomarker and that neutralizing antibodies/soluble RAGE could be used to develop novel therapeutic strategies against TNBC.

No potential conflicts of interest were disclosed.

Conception and design: M.W. Nasser, N.A. Wani, W.E. Carson III, R.K. Ganju

Development of methodology: M.W. Nasser, N.A. Wani, H. Zhao, L. Padilla, R.K. Ganju

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.W. Nasser, N.A. Wani, D.K. Ahirwar, C.A. Powell, J. Ravi, M. Elbaz, K. Shilo, C. Shapiro, R.K. Ganju

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M.W. Nasser, N.A. Wani, D.K. Ahirwar, C.A. Powell, M. Elbaz, H. Zhao, L. Padilla, X. Zhang, R.K. Ganju

Writing, review, and/or revision of the manuscript: M.W. Nasser, D.K. Ahirwar, C.A. Powell, J. Ravi, L. Padilla, X. Zhang, K. Shilo, M. Ostrowski, C. Shapiro, W.E. Carson III, R.K. Ganju

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.W. Nasser, J. Ravi, M. Elbaz, K. Shilo, M. Ostrowski, R.K. Ganju

Study supervision: R.K. Ganju

The authors thank Kristin Kovach for assistance with IHC. They also thank Drs. Z. Qamri, A. Sneh, D. Chakroborty, and G. Amponsah for technical assistance and S. Adamovich for critical reading of the article.

This work was supported by grants from NIH (CA109527 and CA153490) and Department of Defense to R.K. Ganju. N.A. Wani, D.K. Ahirwar, and H. Zhao were supported by Pelotonia Fellowship from the Comprehensive Cancer Center, OSU. M. Elbaz is supported by Fellowship from Government of Egypt.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sparvero
LJ
,
Asafu-Adjei
D
,
Kang
R
,
Tang
D
,
Amin
N
,
Im
J
, et al
RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation
.
J Transl Med
2009
;
7
:
17
.
2.
Rojas
A
,
Figueroa
H
,
Morales
E
. 
Fueling inflammation at tumor microenvironment: the role of multiligand/RAGE axis
.
Carcinogenesis
2010
;
31
:
334
41
.
3.
de Martel
C
,
Ferlay
J
,
Franceschi
S
,
Vignat
J
,
Bray
F
,
Forman
D
, et al
Global burden of cancers attributable to infections in 2008: a review and synthetic analysis
.
Lancet Oncol
2012
;
13
:
607
15
.
4.
Quail
DF
,
Joyce
JA
. 
Microenvironmental regulation of tumor progression and metastasis
.
Nat Med
2013
;
19
:
1423
37
.
5.
Grivennikov
SI
,
Greten
FR
,
Karin
M
. 
Immunity, inflammation, and cancer
.
Cell
2010
;
140
:
883
99
.
6.
Mantovani
A
,
Allavena
P
,
Sica
A
,
Balkwill
F
. 
Cancer-related inflammation
.
Nature
2008
;
454
:
436
44
.
7.
Gebhardt
C
,
Riehl
A
,
Durchdewald
M
,
Nemeth
J
,
Furstenberger
G
,
Muller-Decker
K
, et al
RAGE signaling sustains inflammation and promotes tumor development
.
J Exp Med
2008
;
205
:
275
85
.
8.
Heijmans
J
,
Buller
NV
,
Hoff
E
,
Dihal
AA
,
van der Poll
T
,
van Zoelen
MA
, et al
Rage signalling promotes intestinal tumourigenesis
.
Oncogene
2013
;
32
:
1202
6
.
9.
Kang
R
,
Loux
T
,
Tang
D
,
Schapiro
NE
,
Vernon
P
,
Livesey
KM
, et al
The expression of the receptor for advanced glycation endproducts (RAGE) is permissive for early pancreatic neoplasia
.
Proc Natl Acad Sci U S A
2012
;
109
:
7031
6
.
10.
Riehl
A
,
Nemeth
J
,
Angel
P
,
Hess
J
. 
The receptor RAGE: Bridging inflammation and cancer
.
Cell Commun Signal
2009
;
7
:
12
.
11.
Taguchi
A
,
Blood
DC
,
del Toro
G
,
Canet
A
,
Lee
DC
,
Qu
W
, et al
Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases
.
Nature
2000
;
405
:
354
60
.
12.
Turovskaya
O
,
Foell
D
,
Sinha
P
,
Vogl
T
,
Newlin
R
,
Nayak
J
, et al
RAGE, carboxylated glycans and S100A8/A9 play essential roles in colitis-associated carcinogenesis
.
Carcinogenesis
2008
;
29
:
2035
43
.
13.
Wang
H
,
Zhang
L
,
Zhang
IY
,
Chen
X
,
Da Fonseca
A
,
Wu
S
, et al
S100B promotes glioma growth through chemoattraction of myeloid-derived macrophages
.
Clin Cancer Res
2013
;
19
:
3764
75
.
14.
Wolf
R
,
Howard
OM
,
Dong
HF
,
Voscopoulos
C
,
Boeshans
K
,
Winston
J
, et al
Chemotactic activity of S100A7 (Psoriasin) is mediated by the receptor for advanced glycation end products and potentiates inflammation with highly homologous but functionally distinct S100A15
.
J Immunol
2008
;
181
:
1499
506
.
15.
Wolf
R
,
Mascia
F
,
Dharamsi
A
,
Howard
OM
,
Cataisson
C
,
Bliskovski
V
, et al
Gene from a psoriasis susceptibility locus primes the skin for inflammation
.
Sci Transl Med
2010
;
2
:
61ra90
.
16.
Al-Haddad
S
,
Zhang
Z
,
Leygue
E
,
Snell
L
,
Huang
A
,
Niu
Y
, et al
Psoriasin (S100A7) expression and invasive breast cancer
.
Am J Pathol
1999
;
155
:
2057
66
.
17.
Sneh
A
,
Deol
YS
,
Ganju
A
,
Shilo
K
,
Rosol
TJ
,
Nasser
MW
, et al
Differential role of psoriasin (S100A7) in estrogen receptor alpha positive and negative breast cancer cells occur through actin remodeling
.
Breast Cancer Res Treat
2013
;
138
:
727
39
.
18.
Nasser
MW
,
Qamri
Z
,
Deol
YS
,
Ravi
J
,
Powell
CA
,
Trikha
P
, et al
S100A7 enhances mammary tumorigenesis through upregulation of inflammatory pathways
.
Cancer Res
2012
;
72
:
604
15
.
19.
West
NR
,
Watson
PH
. 
S100A7 (psoriasin) is induced by the proinflammatory cytokines oncostatin-M and interleukin-6 in human breast cancer
.
Oncogene
2010
;
29
:
2083
92
.
20.
Hartman
ZC
,
Poage
GM
,
den Hollander
P
,
Tsimelzon
A
,
Hill
J
,
Panupinthu
N
, et al
Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8
.
Cancer Res
2013
;
73
:
3470
80
.
21.
Pollard
JW
. 
Tumour-educated macrophages promote tumour progression and metastasis
.
Nat Rev Cancer
2004
;
4
:
71
8
.
22.
Sica
A
,
Mantovani
A
. 
Macrophage plasticity and polarization: in vivo veritas
.
J Clin Invest
2012
;
122
:
787
95
.
23.
Biswas
SK
,
Lewis
CE
. 
NF-kappaB as a central regulator of macrophage function in tumors
.
J Leukoc Biol
2010
;
88
:
877
84
.
24.
Gasparini
P
,
Cascione
L
,
Fassan
M
,
Lovat
F
,
Guler
G
,
Balci
S
, et al
microRNA expression profiling identifies a four microRNA signature as a novel diagnostic and prognostic biomarker in triple negative breast cancers
.
Oncotarget
2014
;
5
:
1174
84
.
25.
Shilo
K
,
Dracheva
T
,
Mani
H
,
Fukuoka
J
,
Sesterhenn
IA
,
Chu
WS
, et al
Alpha-methylacyl CoA racemase in pulmonary adenocarcinoma, squamous cell carcinoma, and neuroendocrine tumors: expression and survival analysis
.
Arch Pathol Lab Med
2007
;
131
:
1555
60
.
26.
Hernandez
JL
,
Padilla
L
,
Dakhel
S
,
Coll
T
,
Hervas
R
,
Adan
J
, et al
Therapeutic targeting of tumor growth and angiogenesis with a novel anti-S100A4 monoclonal antibody
.
PLoS One
2013
;
8
:
e72480
.
27.
Richardson
AL
,
Wang
ZC
,
De Nicolo
A
,
Lu
X
,
Brown
M
,
Miron
A
, et al
X chromosomal abnormalities in basal-like human breast cancer
.
Cancer Cell
2006
;
9
:
121
32
.
28.
Minn
AJ
,
Kang
Y
,
Serganova
I
,
Gupta
GP
,
Giri
DD
,
Doubrovin
M
, et al
Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors
.
J Clin Invest
2005
;
115
:
44
55
.
29.
Wolford
CC
,
McConoughey
SJ
,
Jalgaonkar
SP
,
Leon
M
,
Merchant
AS
,
Dominick
JL
, et al
Transcription factor ATF3 links host adaptive response to breast cancer metastasis
.
J Clin Invest
2013
;
123
:
2893
906
.
30.
Qamri
Z
,
Preet
A
,
Nasser
MW
,
Bass
CE
,
Leone
G
,
Barsky
SH
, et al
Synthetic cannabinoid receptor agonists inhibit tumor growth and metastasis of breast cancer
.
Mol Cancer Ther
2009
;
8
:
3117
29
.
31.
Prasad
A
,
Paruchuri
V
,
Preet
A
,
Latif
F
,
Ganju
RK
. 
Slit-2 induces a tumor-suppressive effect by regulating beta-catenin in breast cancer cells
.
J Biol Chem
2008
;
283
:
26624
33
.
32.
Iorns
E
,
Drews-Elger
K
,
Ward
TM
,
Dean
S
,
Clarke
J
,
Berry
D
, et al
A new mouse model for the study of human breast cancer metastasis
.
PLoS One
2012
;
7
:
e47995
.
33.
Lasfargues
EY
,
Coutinho
WG
,
Redfield
ES
. 
Isolation of two human tumor epithelial cell lines from solid breast carcinomas
.
J Natl Cancer Inst
1978
;
61
:
967
78
.
34.
Lee
TH
,
Seng
S
,
Sekine
M
,
Hinton
C
,
Fu
Y
,
Avraham
HK
, et al
Vascular endothelial growth factor mediates intracrine survival in human breast carcinoma cells through internally expressed VEGFR1/FLT1
.
PLoS Med
2007
;
4
:
e186
.
35.
Wang
TC
,
Cardiff
RD
,
Zukerberg
L
,
Lees
E
,
Arnold
A
,
Schmidt
EV
. 
Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice
.
Nature
1994
;
369
:
669
71
.
36.
Weinstat-Saslow
D
,
Merino
MJ
,
Manrow
RE
,
Lawrence
JA
,
Bluth
RF
,
Wittenbel
KD
, et al
Overexpression of cyclin D mRNA distinguishes invasive and in situ breast carcinomas from non-malignant lesions
.
Nat Med
1995
;
1
:
1257
60
.
37.
Bierhaus
A
,
Schiekofer
S
,
Schwaninger
M
,
Andrassy
M
,
Humpert
PM
,
Chen
J
, et al
Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB
.
Diabetes
2001
;
50
:
2792
808
.
38.
Lin
EY
,
Pollard
JW
. 
Tumor-associated macrophages press the angiogenic switch in breast cancer
.
Cancer Res
2007
;
67
:
5064
6
.
39.
Mantovani
A
,
Sica
A
. 
Macrophages, innate immunity and cancer: balance, tolerance, and diversity
.
Curr Opin Immunol
2010
;
22
:
231
7
.
40.
Radia
AM
,
Yaser
AM
,
Ma
X
,
Zhang
J
,
Yang
C
,
Dong
Q
, et al
Specific siRNA targeting receptor for advanced glycation end products (RAGE) decreases proliferation in human breast cancer cell lines
.
Int J Mol Sci
2013
;
14
:
7959
78
.
41.
Lata
K
,
Mukherjee
TK
. 
Knockdown of receptor for advanced glycation end products attenuate 17alpha-ethinyl-estradiol dependent proliferation and survival of MCF-7 breast cancer cells
.
Biochim Biophys Acta
2014
;
1840
:
1083
91
.
42.
Yin
C
,
Li
H
,
Zhang
B
,
Liu
Y
,
Lu
G
,
Lu
S
, et al
RAGE-binding S100A8/A9 promotes the migration and invasion of human breast cancer cells through actin polymerization and epithelial-mesenchymal transition
.
Breast Cancer Res Treat
2013
;
142
:
297
309
.
43.
DiNorcia
J
,
Lee
MK
,
Moroziewicz
DN
,
Winner
M
,
Suman
P
,
Bao
F
, et al
RAGE gene deletion inhibits the development and progression of ductal neoplasia and prolongs survival in a murine model of pancreatic cancer
.
J Gastrointest Surg
2012
;
16
:
104
12
;
discussion 112
.
44.
Bierhaus
A
,
Humpert
PM
,
Morcos
M
,
Wendt
T
,
Chavakis
T
,
Arnold
B
, et al
Understanding RAGE, the receptor for advanced glycation end products
.
J Mol Med (Berl)
2005
;
83
:
876
86
.
45.
Ichikawa
M
,
Williams
R
,
Wang
L
,
Vogl
T
,
Srikrishna
G
. 
S100A8/A9 activate key genes and pathways in colon tumor progression
.
Mol Cancer Res
2011
;
9
:
133
48
.
46.
Sica
A
,
Allavena
P
,
Mantovani
A
. 
Cancer related inflammation: the macrophage connection
.
Cancer Lett
2008
;
267
:
204
15
.
47.
Kessenbrock
K
,
Plaks
V
,
Werb
Z
. 
Matrix metalloproteinases: regulators of the tumor microenvironment
.
Cell
2010
;
141
:
52
67
.
48.
van Kempen
LC
,
Coussens
LM
. 
MMP9 potentiates pulmonary metastasis formation
.
Cancer Cell
2002
;
2
:
251
2
.
49.
Deng
J
,
Liu
Y
,
Lee
H
,
Herrmann
A
,
Zhang
W
,
Zhang
C
, et al
S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites
.
Cancer Cell
2012
;
21
:
642
54
.
50.
Network TCGA
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.