Basal-like breast cancer (BLBC) is an aggressive subtype of breast cancer which is often enriched with cancer stem cells (CSC), but the underlying molecular basis for this connection remains elusive. We hypothesized that BLBC cells are able to establish a niche permissive to the maintenance of CSCs and found that tumor cell-derived periostin (POSTN), a component of the extracellular matrix, as well as a corresponding cognate receptor, integrin αvβ3, are highly expressed in a subset of BLBC cell lines as well as in CSC-enriched populations. Furthermore, we demonstrated that an intact periostin–integrin β3 signaling axis is required for the maintenance of breast CSCs. POSTN activates the ERK signaling pathway and regulates NF-κB–mediated transcription of key cytokines, namely IL6 and IL8, which in turn control downstream activation of STAT3. In summary, these findings suggest that BLBC cells have an innate ability to establish a microenvironmental niche supportive of CSCs.

Implications: The findings reported here indicate that POSTN produced by CSCs acts to reinforce the stem cell state through the activation of integrin receptors and the production of key cytokines. Mol Cancer Res; 14(1); 103–13. ©2015 AACR.

This article is featured in Highlights of This Issue, p. 1

Cancer stem cells (CSC) represent a highly malignant subpopulation of tumor cells that have been proposed to drive tumorigenesis, metastasis, disease recurrence and resistance to chemotherapy (1, 2). Breast CSCs (CD44high/CD24low/−) were first identified based on their ability to initiate tumor growth and differentiate into non-CSCs when injected at limiting dilution (3). Since then, various other phenotypic characteristics have been associated with breast CSCs including high aldehyde dehydrogenase (ALDH) activity (4), an enhanced capacity to generate mammospheres, a surrogate marker for self-renewal and stem cells (5, 6), and activation of an epithelial–mesenchymal transition (EMT) program (7, 8). The relevance of CSCs to breast cancer is supported by clinical findings that link markers of their presence (i.e., expression of stem cell–associated genes) to a poor disease prognosis (9) and chemotherapeutic resistance (10).

Several signaling pathways have emerged as critical regulators of the CSC state. Many of these signals constitute important developmental programs that, in the case of breast cancer, may act to sustain a population of CSCs (11, 12). Recently, cytokines, namely IL6 and IL8, have been implicated in the maintenance of breast CSCs (13–16). And conditions such as hypoxia have also been shown to promote the appearance of breast CSCs (17). Therefore, in addition to defined genetic mutations, the surrounding tumor microenvironment may play a major role in the promotion of the CSC state.

Basal-like breast cancer (BLBC) is an aggressive molecular subtype of the disease that is enriched for cells with stem-like features such as a CD44high/CD24low surface marker profile (18), activation of a mesenchymal transcriptional program (19) and resistance to therapy (20). The relevance and underlying basis for this enrichment are not well understood but it raises the possibility that BLBC pathogenesis could be linked to the creation of a tumor microenvironment conducive to the formation or maintenance of CSCs.

Here, we examined the hypothesis that BLBC cells secrete periostin (encoded by the POSTN gene, also known as OSF-2), a mesenchymal extracellular matrix (ECM) protein, to support a population of CSCs. These studies highlight a role for tumor cell-derived POSTN, which signals through integrin αvβ3 to regulate the expression of key cytokines and mediates stemness in BLBC cells.

Cell culture

All M cell lines (MI, MII, MIII) were cultured in DMEM F/12 media containing 5% horse serum, 10 μg/mL insulin, 20 ng/mL EGF, 100 ng/mL cholera toxin, and 0.5 μg/mL hydrocortisone, as previously described (21). SUM159 cell lines were cultured in Ham's F/12 supplemented with 5% FBS, 10 μg/mL insulin, and 0.5 μg/mL hydrocortisone while Hs578T cells were maintained in DMEM containing 10% FBS. All other cell lines were cultured as described previously (22). Mammospheres were grown in MammoCult media (Stem Cell Technologies) containing 0.48 μg/mL hydrocortisone and 1% methylcellulose. All cells were grown in the presence of penicillin/streptomycin (1%) and CO2 (5%) at 37°C in a humidified incubator.

Antibodies and reagents

A detailed list of all antibodies and reagents used is provided in the Supplementary Data.

Generation of knockdown lines

Stable knockdown of periostin or integrin β3 was achieved by transduction of lentiviral vectors (pLKO.1) followed by selection with puromycin (2 μg/mL). shRNA sequences can be found in the Supplementary Data.

qRT-PCR

Total RNA was isolated with TRIzol (Life Technologies) and cDNA synthesized using random primers and SuperScript II Reverse Transcriptase (Life Technologies). qRT-PCR was performed with Power SYBR Green PCR Master Mix (Life Technologies) and run on an Applied Biosystems 7900HT Sequence Detection System. Relative RNA expression was calculated using the ΔΔCt method and normalized to β-actin. Primer sequences are listed in the Supplementary Data.

ELISAs

Secreted periostin was measured with an ELISA kit (AdipoGen) according to the manufacturer's protocol. Protein was concentrated using Amicon centrifugation filters (30K membrane, Millipore) before analysis. To measure cytokine production, cells were starved in serum-reduced media (0.1% serum) and conditioned media collected 48 hours later. IL8 protein was quantified using a Quantikine ELISA Kit (R&D Systems), whereas IL-6 quantification was performed with a CyPlex (CyVek, Inc.).

Western blotting

Protein was isolated from cells on ice with RIPA buffer containing protease and phosphatase inhibitor cocktails (Roche). For examination of signaling events, cells were grown in serum-reduced media (0.1% serum) for 24 or 48 hours before protein isolation. Proteins were separated by SDS-PAGE and immobilized on PVDF membranes. Secondary antibodies were conjugated to peroxidase and detected by chemiluminescence with ECL solution (PerkinElmer).

Flow cytometry

For examination of cell surface proteins, cells were trypsinized, washed in FACS buffer (PBS with 0.2% BSA, 0.09% sodium azide and 1 mmol/L EDTA) and stained with antibody for 30 minutes at 4°C. To quantify the percentage of ALDH-positive cells, the ALDEFLUOR assay was used (Stem Cell Technologies). Briefly, cells were trypsinized, suspended in assay buffer, and treated for 30 minutes with the ALDEFLUOR reagent (BODIPY-aminoacetaldehyde, BAAA; 3 μmol/L), in the presence or absence of the ALDH inhibitor diethylaminobenzaldehyde (DEAB; 37.5 μmol/L). Samples were analyzed on FACScan, FACSCalibur, LSR II or Accuri C6 (BD) cytometers. Cells were sorted using a different buffer (PBS, 2% horse serum, 1 mmol/L ETDA) with a MoFlo (Beckman Coulter).

Animal studies

Tumor xenograft experiments were conducted under the approval of the Institutional Animal Care and Use Committee at Boston University School of Medicine (Boston, MA). Bioluminescent cells were generated using a pMSCV-Luc-PGK-hygro plasmid (Addgene-8782). Fifty thousand cells were injected subcutaneously, in the absence of Matrigel (in 0.1 mL of serum-free F/12 media), into 6- to 8 week-old NOD/SCID mice (The Jackson Laboratory). Animals were monitored on a weekly basis using an IVIS Spectrum Imaging System (Caliper) for a period of 6 months, unless tumor growth necessitated euthanasia.

Gene expression microarray and bioinformatic analysis

Total RNA was isolated using TRizol (Life Technologies) and cleaned up with the RNeasy Mini kit (Qiagen). Biotin labeling was performed using the Ambion WT Expression Kit (Life Technologies) and the GeneChip WT Terminal Labeling and Controls Kit (Affymetrix), followed by hybridization to GeneChip Human Gene 2.0 ST arrays (Affymetrix). Raw CEL files were normalized using the Robust Multiarray Average [PubMed ID 14960456]. Differential gene expression between shPN and shGFP was assessed using the moderated t test implemented in the “limma” package. Raw CEL files from GEO Series GSE21653 were normalized in the same manner, and differential expression between molecular subtypes was assessed using the Welch t test. All microarray analyses were performed using the R environment for statistical computing.

Luciferase reporter assay

Cells were cotransfected with the 3× KB-L NF-κB luciferase reporter plasmid (Addgene #26699) and a Renilla-luciferase plasmid using X-tremeGENE HP DNA transfection reagent (Roche). Twenty-four hours after transfection, cells were starved in reduced serum medium (0.1% FBS) for 16 hours. TNFα treatment (20 ng/mL, R&D Systems) served as a positive control. For experiments that involved inhibition of the ERK pathway, inhibitors were added 24 hours after transfection and luminescence was measured 24 hours later. Luminescence was quantified with the Dual-Luciferase Reporter Assay (Promega) using a GloMax microplate reader. Firefly luciferase signal was normalized to the Renilla luciferase signal to determine relative luminescence units (RLU).

Statistical analysis

Statistical analysis was performed using a two-tailed paired Student t test. A P value less than 0.05 was considered statistically significant. Error bars represent ± SEM.

Characterization of cancer stem cell traits in M cells

To identify tumor cell-derived factors that could support the maintenance of breast CSCs, we examined a well-established cell line model of breast cancer progression (23, 24). This model system consists of three cell lines: MCF10A (MI) and two derivatives of this line, MCF10AT1k.cl2 (MII) and MCF10CA1h (MIII). These cell lines exhibit increasing degrees of malignancy (24). Previous characterization by our laboratory has indicated that while MI and MII cells display numerous epithelial features, MIII cells clearly exhibit a phenotype associated with activation of an EMT program (21).

Given the connection between EMT and CSCs (7, 8), we wondered whether MIII cells also exhibit features associated with the stem cell state. In support of this notion, MIII cells predominantly displayed the CD44high/CD24low surface phenotype (Fig. 1A) and preferentially formed mammospheres, doing so with nearly a 9-fold greater efficiency than MII cells (Fig. 1B). Because the MIII cell line displayed various traits reminiscent of breast CSCs, we reasoned that it could serve as a good model to identify regulators of the CSC state.

Figure 1.

Overexpression of POSTN and ITGB3 in BLBC cells suggests a functional role for this signaling axis. A, flow-cytometric analysis of CD44 and CD24 surface expression in MI, MII, and MIII cells. The graph shows the percentage of CD44high/CD24low cells in each line, n = 3. B, mammosphere formation assay with MII and MIII cells, n = 3. C, expression of POSTN in the M cell lines was measured using qRT-PCR. D, Western blot analysis for POSTN in the conditioned media of the indicated cell lines. Ponceau staining was used to show protein loading. E, POSTN mRNA was measured by qRT-PCR in MIII cells before and after growth as mammospheres. F, POSTN mRNA was measured by qRT-PCR in MII cells sorted as either CD44high/CD24low or CD44high/CD24high. G, flow-cytometric analysis of surface levels of integrin αvβ3 in the M cell lines. Gray shading indicates isotype control. Numbers in parentheses indicate the percentage of positive cells in each line. H, expression of POSTN and ITGB3 was assessed by qRT-PCR. Red indicates luminal cell lines while black indicates basal-like cell lines. The circle shows the subset of cells that express high levels of both genes. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Figure 1.

Overexpression of POSTN and ITGB3 in BLBC cells suggests a functional role for this signaling axis. A, flow-cytometric analysis of CD44 and CD24 surface expression in MI, MII, and MIII cells. The graph shows the percentage of CD44high/CD24low cells in each line, n = 3. B, mammosphere formation assay with MII and MIII cells, n = 3. C, expression of POSTN in the M cell lines was measured using qRT-PCR. D, Western blot analysis for POSTN in the conditioned media of the indicated cell lines. Ponceau staining was used to show protein loading. E, POSTN mRNA was measured by qRT-PCR in MIII cells before and after growth as mammospheres. F, POSTN mRNA was measured by qRT-PCR in MII cells sorted as either CD44high/CD24low or CD44high/CD24high. G, flow-cytometric analysis of surface levels of integrin αvβ3 in the M cell lines. Gray shading indicates isotype control. Numbers in parentheses indicate the percentage of positive cells in each line. H, expression of POSTN and ITGB3 was assessed by qRT-PCR. Red indicates luminal cell lines while black indicates basal-like cell lines. The circle shows the subset of cells that express high levels of both genes. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Close modal

Periostin is highly expressed in basal-like breast cancer stem cells

Because all of the M model cell lines share the same genetic lineage, we hypothesized that altered expression of secreted microenvironmental factors may contribute to the CSC-like features of MIII cells. Our previous gene expression profiling studies (21) revealed that periostin (POSTN) was overexpressed in the MIII cell line. This was particularly interesting because POSTN is regulated by Twist (25) and TGFβ (26), two factors that promote passage through an EMT and acquisition of stem cell phenotypes (7). We validated that POSTN is a transcriptional target of the TGFβ pathway in this model of breast cancer, as treatment of MII cells with TGFβ1 resulted in a strong increase in POSTN transcription (Supplementary Fig. S1A). Conversely, active TGFβ signaling was necessary to sustain POSTN expression in MIII cells (Supplementary Fig. S1B).

Using qRT-PCR, we confirmed that the level of POSTN mRNA was significantly increased in MIII cells compared with MI or MII cells (Fig. 1C). As expected, this increase in transcription was correlated with enhanced secretion of POSTN into the surrounding media (Fig. 1D). Moreover, we found that POSTN was highly expressed in populations enriched for CSCs. For instance, MIII cells grown as mammospheres, which are predominately formed by CSCs (6), expressed nearly 10 times more POSTN than their adherent counterparts (Fig. 1E). In a second approach, when MII cells were fractionated based on the surface expression of CD44 and CD24, we found that cells within the CD44high/CD24low (CSC) population expressed significantly more POSTN than cells within the CD44high/CD24high (non-CSC) fraction (Fig. 1F). Overall, in the MCF10A breast cancer model, high POSTN expression directly correlated with multiple phenotypes attributed to CSCs.

Basal-like breast cancer cells express a periostin–integrin β3 signaling axis

In examining gene expression profiles of the M cell lines, we noted that, in addition to POSTN, MIII cells also expressed high levels of integrin β3 (ITGB3), a subunit of the integrin αvβ3 complex that is a known receptor for POSTN (27). Indeed, we found that the surface levels of the functional integrin αvβ3 complex were abundant on MIII cells but nearly absent from MI and MII cells (Fig. 1G). The coordinated expression of the receptor-ligand combination required for the activity of an intact signaling axis suggested that, in MIII cells, POSTN could potentially signal in an autocrine fashion.

Wishing to expand on this observation, we examined whether such an axis was present in other breast cancer cells. To this end, we measured the mRNA levels of both POSTN and ITGB3 in a panel of nine breast cancer cell lines. A high level of expression of both genes was detected in four cell lines: BT549, SUM1315, SUM159, and Hs578T cells (Fig. 1H). Notably, and similar to MIII cells (21), all four of these cell lines fall under the molecular classification of BLBC cells (22). In a subset of these lines, we verified that the increase in POSTN and ITGB3 expression correlated with enhanced POSTN secretion (Supplementary Fig. S2A) and increased surface levels of the integrin αvβ3 receptor complex (Supplementary Fig. S2B). These data indicated that a periostin–integrin β3 signaling axis is preserved, and potentially operative, in a subset of BLBC cells.

Tumor cell–derived POSTN and ITGB3 maintain breast cancer stem cells

To test the relevance of POSTN expression to CSCs, we opted to focus on the SUM159 line because it has a well-defined CSC population (28). We generated SUM159 cells with stable knockdown of POSTN (shPN) or ITGB3 (shBeta3) through expression of corresponding shRNA constructs (Supplementary Fig. S3A and S3B). Knockdown of POSTN or ITGB3 in SUM159 cells did not have an effect on cell proliferation or apoptosis (Supplementary Fig. S3C and S3D) and did not lead to any morphologic changes or reversion of the EMT program. (Supplementary Fig. S3E and S3F). However, POSTN knockdown resulted in a significant reduction in the ability of SUM159 cells to generate mammospheres, with shPN cells forming 72% fewer mammospheres than control (shGFP) cells (Fig. 2A). Similarly, SUM159 shBeta3 cells also exhibited a significant reduction in mammosphere formation potential (Fig. 2A). Notably, the most dramatic effect of POSTN knockdown was observed in the well-established (28), ALDH-positive CSC population of SUM159 cells. Under conditions of reduced serum (0.1% serum), which are often used to analyze the effect of ECM proteins and integrin signaling (29), close to 15% of SUM159 shGFP were ALDH-positive. On the other hand, only 2% of the cells were ALDH positive in SUM159 shPN cells, representing a 7.4-fold reduction in this highly tumorigenic cell population (Fig. 2B). Furthermore, SUM159 shBeta3 cells had a 2.9-fold reduction in the ALDH-positive CSC population (Fig. 2B). Importantly, we verified these findings with an additional shRNA hairpin directed at either POSTN or ITGB3 (Supplementary Fig. S4A–S4C). These results suggest that BLBC cells rely on POSTN and ITGB3 to sustain a population of CSCs.

Figure 2.

POSTN signaling is required to maintain breast CSCs. A, SUM159 control (shGFP) and periostin (shPN) or integrin β3 (shBeta3) knockdown cells were grown as mammospheres and quantified, n = 2. Representative images of mammospheres are shown on the right. B, quantification of the percentage of ALDH-positive cells in SUM159 shGFP, shPN, and shBeta3 cells after 48 hours of growth in serum-reduced (0.1% serum) media, n = 3. Representative ALDH plots are shown on the right. DEAB, an inhibitor of ALDH, was used as a gating control. C, MIII cells were sorted by flow cytometry based on the surface expression of the integrin αvβ3 complex (LM609) before seeding an equal number of cells for mammosphere growth, n = 3. D, SUM159 shGFP and shPN cells were seeded as mammospheres at limiting dilution, n = 3. E, the percentage of animals that developed tumors 6 months after subcutaneous injection of the indicated cell lines (5 × 104 cells), n = 4 animals per group. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Figure 2.

POSTN signaling is required to maintain breast CSCs. A, SUM159 control (shGFP) and periostin (shPN) or integrin β3 (shBeta3) knockdown cells were grown as mammospheres and quantified, n = 2. Representative images of mammospheres are shown on the right. B, quantification of the percentage of ALDH-positive cells in SUM159 shGFP, shPN, and shBeta3 cells after 48 hours of growth in serum-reduced (0.1% serum) media, n = 3. Representative ALDH plots are shown on the right. DEAB, an inhibitor of ALDH, was used as a gating control. C, MIII cells were sorted by flow cytometry based on the surface expression of the integrin αvβ3 complex (LM609) before seeding an equal number of cells for mammosphere growth, n = 3. D, SUM159 shGFP and shPN cells were seeded as mammospheres at limiting dilution, n = 3. E, the percentage of animals that developed tumors 6 months after subcutaneous injection of the indicated cell lines (5 × 104 cells), n = 4 animals per group. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Close modal

Additional evidence for this notion was obtained from experiments with the MIII cell line. Here too, knockdown of POSTN (Supplementary Fig. S5A) led to a significant reduction in mammosphere-forming potential; MIII shPN cells generated 52% fewer mammospheres than control cells (Supplementary Fig. S5B). Notably, when MIII cells were sorted based on the surface level of integrin αvβ3, there was a dramatic difference in the capacity to initiate mammosphere growth, where only MIII cells with high surface levels of αvβ3 could robustly form mammospheres (Fig. 2C). The overlapping phenotypes observed following perturbation of POSTN or ITGB3 imply that these proteins function in a closely related or overlapping pathway to regulate the CSC state in BLBC.

Consistent with the above findings, when shGFP and shPN cells were seeded as mammospheres at limiting dilution we detected a clear reduction in the estimated stem cell frequency, with the sphere formation efficiency of 100 shGFP control cells being roughly equal to that of 1000 shPN cells (Fig. 2D). Importantly, knockdown of POSTN impaired the ability of SUM159 cells to initiate tumors when injected at limiting numbers (5 × 104 cells). Six months after injection, 50% of mice had developed tumors in the control group while no tumors were detected in animals injected with SUM159 shPN cells (Fig. 2E). This result is consistent with our in vitro findings and lend support to the idea that tumor cell-derived periostin is crucial for tumor initiation, the salient characteristic of CSCs.

Periostin regulates cytokine production via ERK signaling and NF-κB

To understand the molecular events downstream of POSTN, we performed genome-wide expression profiling of SUM159 shGFP and shPN cells and used Gene Set Enrichment Analysis (GSEA) to identify the major pathways that were altered upon POSTN knockdown in SUM159 cells. This analysis revealed six gene sets, containing a preponderance of cytokines, that were significantly repressed in SUM159 shPN cells (Supplementary Fig. S6A). Given the connection between cytokines and breast CSCs (13), we wished to investigate this further. Close examination of the twenty genes that comprise the cytokine pathway gene set highlighted five cytokines that were clearly repressed in SUM159 shPN cells compared with shGFP cells: IL1A, IL8, IL6, IL18, and IL16 (Fig. 3A). Of these five cytokines, IL1A, IL8, and IL6 were found at the leading edge of other downregulated gene sets (Supplementary Fig. S6B), implying that they may represent particularly important mediators downstream of POSTN.

Figure 3.

POSTN signaling regulates cytokine production. A, heatmap depicting expression of the 20 genes contained in BioCarta Cytokine Network gene set in shGFP and shPN cells. Gene expression values are zero-centered by row and arranged in order by the moderated t statistic. Colors represent: blue, below mean expression; red, above mean expression. B, expression of IL1A, IL6, and IL8 was measured in the indicated SUM159 cell lines by qRT-PCR. RNA was isolated from cells grown in serum-reduced media for 48 hours. C, IL8 protein was quantified from the conditioned media of the indicated cell lines after 48 hours of growth in serum-reduced (0.1% serum) media. D, IL6 protein was quantified from the conditioned media of the indicated cell lines after 48 hours of growth in serum-reduced (0.1% serum) media. E, expression of POSTN and IL6 was assessed by qRT-PCR in a panel of six basal-like breast cancer cell lines (Fig. 1H). F, Western blot analysis of STAT3 phosphorylation (tyrosine 705) and total STAT3 in the indicated SUM159 cell lines. Protein was isolated from cells grown in serum-reduced media (0.1% serum) for 48 hours. G, rescue of STAT3 signaling with conditioned media. Conditioned media were collected from shGFP and shPN cells and used to treat the indicated SUM159 cell lines following growth in serum-reduced media for 48 hours. After starvation, the cells were transiently exposed (60 minutes) to conditioned media (CM) from either shGFP or shPN cells; serum-reduced media served as a mock control. Western blot analysis shows the levels of phosphorylated STAT3 (tyrosine 705), total STAT3, and β-actin. **, P < 0.01; error bars represent SEM.

Figure 3.

POSTN signaling regulates cytokine production. A, heatmap depicting expression of the 20 genes contained in BioCarta Cytokine Network gene set in shGFP and shPN cells. Gene expression values are zero-centered by row and arranged in order by the moderated t statistic. Colors represent: blue, below mean expression; red, above mean expression. B, expression of IL1A, IL6, and IL8 was measured in the indicated SUM159 cell lines by qRT-PCR. RNA was isolated from cells grown in serum-reduced media for 48 hours. C, IL8 protein was quantified from the conditioned media of the indicated cell lines after 48 hours of growth in serum-reduced (0.1% serum) media. D, IL6 protein was quantified from the conditioned media of the indicated cell lines after 48 hours of growth in serum-reduced (0.1% serum) media. E, expression of POSTN and IL6 was assessed by qRT-PCR in a panel of six basal-like breast cancer cell lines (Fig. 1H). F, Western blot analysis of STAT3 phosphorylation (tyrosine 705) and total STAT3 in the indicated SUM159 cell lines. Protein was isolated from cells grown in serum-reduced media (0.1% serum) for 48 hours. G, rescue of STAT3 signaling with conditioned media. Conditioned media were collected from shGFP and shPN cells and used to treat the indicated SUM159 cell lines following growth in serum-reduced media for 48 hours. After starvation, the cells were transiently exposed (60 minutes) to conditioned media (CM) from either shGFP or shPN cells; serum-reduced media served as a mock control. Western blot analysis shows the levels of phosphorylated STAT3 (tyrosine 705), total STAT3, and β-actin. **, P < 0.01; error bars represent SEM.

Close modal

We used qRT-PCR to confirm the repression of IL1A, IL6, and IL8 in SUM159 shPN cells; importantly, the expression of all three genes was also significantly reduced in SUM159 shBeta3 cells (Fig. 3B). Further, conditioned media from shPN and shBeta3 cells contained significantly less IL8 and IL6 protein than media conditioned from shGFP control cells (Fig. 3C and D). Of these three cytokines, IL6 expression was most closely correlated with the expression of POSTN in a panel of six BLBC cell lines (Fig. 3E), suggesting that the regulation of IL6 expression by POSTN may be common in BLBC cells.

In light of the reduced cytokine levels, and because STAT3 is a transducer of cytokine signaling that has been linked to the maintenance of breast CSCs (15), we were prompted to investigate the STAT3 pathway in cells with knockdown of POSTN or ITGB3. Indeed, although SUM159 shGFP cells exhibited relatively high levels of active, phosphorylated STAT3, both SUM159 shPN and shBeta3 cells had markedly reduced levels of phosphorylated STAT3 (Fig. 3F). The impaired transcription of IL6 and IL8, along with reduced activation of STAT3, was also verified using an additional hairpin directed at POSTN or ITGB3 (Supplementary Fig. S4D and S4E).

To test whether impaired production of secreted factors was responsible for this effect, we collected conditioned media from both SUM159 shGFP and shPN cells, and then treated the three cell lines with this or serum-reduced media (mock) for 60 minutes. The diminished activation of STAT3 in the knockdown cells could be rescued by transient exposure to media conditioned from shGFP cells, but not media conditioned from shPN cells (Fig. 3G). These data indicate that impaired production of secreted factors from SUM159 shPN and shBeta3 cells, ostensibly the cytokines mentioned above, is responsible for the observed reduction in STAT3 signaling, rather than any inherent intracellular defects. In addition, we found that treatment of shPN cells with media conditioned from control shGFP cells led to a 1.5-fold increase in the ALDH-positive subpopulation (Supplementary Fig. S7A). This effect was not observed in shBeta3 cells, suggesting that both POSTN and cytokine signaling were required to maintain this population.

Next, we wished to determine the proximal signals downstream of the POSTN–ITGB3 pathway that were responsible for controlling cytokine production. Our approach to achieve this aim was to examine the expression profiles of SUM159 shGFP and shPN cells using Ingenuity Pathway Analysis (IPA) software to look for upstream regulatory networks that might help to explain the differential gene expression profiles between these two cell lines. Interestingly, we noted that, in comparison with the control shGFP cells, SUM159 shPN cells exhibited significant activation of gene expression profiles characteristic of cells treated with two ERK pathway inhibitors (Supplementary Fig. S7B and S7C). This suggested that cells with disrupted POSTN signaling might show impaired activation of the ERK pathway. Consistent with this, cells with knockdown of POSTN or ITGB3 were found to have dramatically reduced levels of phosphorylated, active ERK1/2 (Fig. 4A). This defect in ERK signaling appears to be upstream of cytokine transcription, as pharmacologic inhibition of the ERK pathway led to significantly reduced expression of IL6, IL8, and IL1A (Fig. 4B and Supplementary Fig. S7D). In line with a recent report that linked POSTN signaling to activation of the NF-κB pathway (30), we found that knockdown of POSTN or ITGB3 impaired the transcriptional activity of NF-κB as assessed by a luciferase reporter assay (Fig. 4C). This result was verified using an independent shRNA hairpin directed at POSTN or ITGB3. (Supplementary Fig. S4F). Furthermore, pharmacologic disruption of ERK signaling also caused a decrease in NF-κB transcriptional activity (Fig. 4D).

Figure 4.

POSTN controls cytokine production through the ERK and NF-κB pathway. A, the indicated SUM159 cells were grown in serum-reduced media (0.1% serum) for 24 hours before protein isolation. Western blot analysis shows the levels of phosphorylated ERK1/2 (threonine 202/tyrosine 204) and total ERK1/2. B, SUM159 shGFP cells were treated with DMSO (mock) or an ERK pathway inhibitor (PD184325) for 24 hours before RNA isolation. IL1A, IL6, and IL8 mRNA levels were measured by qRT-PCR. C, the activity of a transfected NF-κB luciferase reporter was measured in the indicated cell lines after 16 hours of growth in serum-reduced media (0.1% serum). Relative luminescence units (RLU) were calculated based on cotransfection of a Renilla luciferase construct. TNFα treatment served as a positive control, n = 3. D, the activity of a transfected NF-κB luciferase reporter was measured in SUM159 shGFP cells 24 hours after treatment with the indicated inhibitors, n = 3. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Figure 4.

POSTN controls cytokine production through the ERK and NF-κB pathway. A, the indicated SUM159 cells were grown in serum-reduced media (0.1% serum) for 24 hours before protein isolation. Western blot analysis shows the levels of phosphorylated ERK1/2 (threonine 202/tyrosine 204) and total ERK1/2. B, SUM159 shGFP cells were treated with DMSO (mock) or an ERK pathway inhibitor (PD184325) for 24 hours before RNA isolation. IL1A, IL6, and IL8 mRNA levels were measured by qRT-PCR. C, the activity of a transfected NF-κB luciferase reporter was measured in the indicated cell lines after 16 hours of growth in serum-reduced media (0.1% serum). Relative luminescence units (RLU) were calculated based on cotransfection of a Renilla luciferase construct. TNFα treatment served as a positive control, n = 3. D, the activity of a transfected NF-κB luciferase reporter was measured in SUM159 shGFP cells 24 hours after treatment with the indicated inhibitors, n = 3. *, P < 0.05; **, P < 0.01; error bars represent SEM.

Close modal

To test whether the connection between POSTN-controlled cytokine expression and CSCs is applicable outside of the SUM159 line, we also knocked down POSTN in Hs578T cells (Supplementary Fig. S8A), another BLBC cell line (22) that coexpresses high levels of both POSTN and integrin αvβ3 (Fig. 1H and Supplementary Fig. S2A and S2B). Consistent with our previous observations, Hs578T cells with knockdown of POSTN formed 57.9% fewer mammospheres (Supplementary Fig. S8B) and showed a 3-fold reduction in the ALDH-positive subpopulation (Supplementary Fig. S8C). The loss of these CSC characteristics was also associated with reduced IL6 expression (Supplementary Fig. S8D) and impaired activation of the STAT3 pathway (Supplementary Fig. S8E). This suggests that the functional relevance of POSTN-regulated cytokine pathways may be more broadly applicable to other basal-like CSCs.

Periostin is associated with a poor prognosis in basal-like breast cancer

We reasoned that cancers with an operative POSTN-ITGB3 signaling axis might be enriched with CSCs and therefore associated with a worse clinical prognosis. On the basis of the fact that ITGB3 marks a population of luminal progenitor cells, the presumed cell-of-origin for basal-like cancers (31), one would predict that this would be limited to a subset of POSTN-expressing BLBCs, many of which are likely to be positive for ITGB3. In accordance with this notion, in silico Kaplan–Meier analysis of a public breast cancer dataset (32) revealed that high POSTN expression was associated with reduced relapse-free survival in basal-like (HR = 1.42, P = 0.0084), but not luminal A (HR = 1.08, P = 0.4), breast cancers (Fig. 5A). As POSTN has been suggested to promote angiogenesis through activation of integrin αvβ3 on endothelial cells (33), it is possible that high POSTN expression could be related to neoangiogenesis, which is also a prognostic factor in breast cancer (34). However, the effect of angiogenesis would seem to be independent of molecular subtype, suggesting that the association with relapse-free survival could be based on an alternative explanation. Indeed, our findings are in line with a recent study that found a correlation between POSTN expression, breast CSC content (as assessed by CD44high/CD24low cells), and a worse clinical prognosis (35).

Figure 5.

High POSTN expression is associated with a poor prognosis in basal-like breast cancer. A, in silico Kaplan–Meier analysis of relapse-free survival in basal-like (n = 581) and Luminal A (n = 1,678) breast cancers. Patients were divided into two groups based on the median expression level of POSTN: high expression (red) or low expression (black). High POSTN expression was associated with reduced relapse-free survival in basal-like (left panel, HR = 1.42, P = 0.0084), but not with luminal A (right panel, HR = 1.08, P = 0.4), breast cancers. B, IL6 and IL8 expression in a publicly available dataset of primary breast cancer samples (GSE21653). Differential expression between groups was assessed by Welch t test. *, P < 0.05; ***, P < 0.001.

Figure 5.

High POSTN expression is associated with a poor prognosis in basal-like breast cancer. A, in silico Kaplan–Meier analysis of relapse-free survival in basal-like (n = 581) and Luminal A (n = 1,678) breast cancers. Patients were divided into two groups based on the median expression level of POSTN: high expression (red) or low expression (black). High POSTN expression was associated with reduced relapse-free survival in basal-like (left panel, HR = 1.42, P = 0.0084), but not with luminal A (right panel, HR = 1.08, P = 0.4), breast cancers. B, IL6 and IL8 expression in a publicly available dataset of primary breast cancer samples (GSE21653). Differential expression between groups was assessed by Welch t test. *, P < 0.05; ***, P < 0.001.

Close modal

In addition, we examined the expression of POSTN, IL6, and IL8 in another independent microarray dataset of 226 primary breast cancers (GEO Series GSE21653; ref. 36). In this dataset, POSTN was highly expressed (median percentile = 99.6%) across all molecular subtypes (Supplementary Fig. S9), but IL6 and IL8 levels were significantly elevated in BLBC (Fig. 5B), in agreement with previous findings that have implicated these cytokines in the growth and tumorigenicity of BLBC stem cells (15, 16, 37). Overall, these clinical data are consistent with a model wherein POSTN can control the production of cytokines in the context of BLBC.

EMT-induced cancer cell lines have been successfully used to screen for selective inhibitors of CSCs (38) and we reasoned that MIII cells—which clearly underwent EMT and display numerous other CSC traits—could represent a similar cell line model in which to study regulators of the CSC state. We hypothesized that proteins secreted into the local microenvironment would play a particularly important role in the maintenance of breast CSCs. Consistent with this notion, our previous gene expression profiling study (21) found a correlation between TGFβ-mediated EMT and POSTN expression, and here we focused our efforts on the function of this protein in regulation of the CSC state.

As a matricellular protein, POSTN can potentially serve as a crucial mediator between CSCs and their surrounding microenvironment (39). Yet there is limited mechanistic detail as to how POSTN may function in BLBC cells. We found a POSTN–ITGB3 signaling axis is operational in a number of breast cancer cell lines, and in each case, the cells could be classified molecularly under the basal-like subtype. This is notable for a number of reasons. First, numerous lines of evidence support the idea that BLBC arises from transformation of luminal progenitor cells (40–42), which are specifically marked by expression of ITGB3 (43). Second, BLBC cells share numerous phenotypic similarities with CSCs (18–20), which may be especially true of a related disease subtype, claudin-low breast cancers (44, 45). In this context, we suggest that tumor cell-derived POSTN may be particularly important for the maintenance of a population of CSCs in basal-like disease.

In support of this hypothesis, our results from knockdown experiments, carried out in three different BLBC cell lines, showed that POSTN expression was required to maintain CSC phenotypes. These findings complement work from others, which showed that ectopic expression of POSTN promotes stem-like properties in human mammary epithelial cells (46). Our data also support a major role for the integrin αvβ3 receptor in sustaining mammosphere growth and an ALDH-positive subpopulation. Another study has addressed the relationship between integrin β3 and CSCs and reported that, while ITGB3 is highly expressed in breast CSCs, knockdown of this gene resulted in minimal effects on mammosphere growth (47). Interestingly, these experiments were performed in derivatives of SK-BR-3 cells, a luminal cell line and, as argued above, this specific integrin may be more relevant in the context of BLBC.

We also sought to identify the pathways downstream of the POSTN-ITGB3 signaling axis that are relevant in the context of CSC regulation. Here, we focused on a defined cytokine network, consisting of IL6, IL8, and IL1A, which was repressed following POSTN knockdown. Interestingly, other gene sets significantly repressed in shPN cells included the hematopoietic lineage and stem cell gene sets. As POSTN has been found to localize to stem cell niches in the bone marrow (48), this suggests that POSTN-mediated regulation of cytokine production might also have physiologic relevance to hematopoietic stem cells. Furthermore, our observations are also consistent with a report that demonstrated that POSTN was required for the production of multiple proinflammatory cytokines during chronic skin inflammation (30). Thus, in the context of cancer, breast CSCs may activate a similar program downstream of POSTN. We also found that cells with defective POSTN signaling showed reduced activation of ERK signaling and, consequently, impaired cytokine transcription. This result is in accordance with a recent study, which reported that activation of the MAPK pathway due to DUSP4 loss promoted the formation of CSCs and increased the expression of IL6 and IL8, in this case through the transcription factors ETS-1 and c-JUN (49). Our data suggest that, at least downstream of POSTN, the NF-κB pathway is likely to be responsible for driving cytokine expression, similar to what has been observed during skin inflammation (30). Nevertheless, NF-κB transcriptional activity appears to lie downstream of the ERK pathway, raising the possibility that POSTN-mediated regulation of the CSC state may be susceptible to interference by MAPK inhibitors.

Overall, this leads to a model in which the POSTN-ITGB3 signaling axis regulates the production of cytokines, which in turn activate STAT3 signaling. This model is compelling because of the rapidly accumulating evidence that links cytokines, especially IL6 and IL8, to the regulation of breast CSCs (15, 16, 28, 50). These alterations may be further understood when considering recent studies that have reported on the cellular plasticity that exists between non-CSCs and CSCs (14, 51). For instance, it has been shown that the IL6-STAT3 pathway can shift this equilibrium in favor of CSCs (14). Thus, one possibility may be that POSTN, at least in part through control of cytokine production, is able to tilt this equilibrium to promote the formation and/or maintenance of CSCs.

In addition to the experiments presented here, others have recently found that POSTN plays a role in the regulation of breast CSCs during the process of metastatic colonization (48). It has also been shown to act as a factor derived from neovascular tip cells in the perivascular niche, where it can serve to break tumor cell dormancy (52). Our work, while consistent with the central idea that POSTN is required for the maintenance of CSCs at sites of tumor cell dissemination, also differs in important aspects. First, we found that, in a subset of basal-like cancer lines, POSTN can be tumor cell derived, potentially allowing these cells to generate their own functional niche and rendering them more independent from their surrounding microenvironment. Similarly, POSTN has recently been reported to be secreted by glioblastoma stem cells (53). Second, our data highlight a major role for integrin αvβ3 in facilitating POSTN signaling, which represents an important regulatory pathway for the maintenance of breast CSCs. Indeed, a recent study found integrin β3 to be a marker of CSCs and a mediator of resistance to targeted therapy, not only in breast cancer, but also in lung and pancreatic cancers (54). Therefore, it is conceivable that periostin could contribute to CSC maintenance and resistance to therapy in breast as well as other carcinomas. Finally, our study implies that a POSTN signaling network can, in principle, act in the primary tumor as well, helping to maintain a fraction of CSCs at earlier stages of tumor progression (Fig. 6). Therefore, we suggest that a subset of BLBCs can establish their own microenvironmental niche supportive of breast CSCs through the production of tumor cell-derived POSTN, which might be clinically relevant as a biomarker or therapeutic target.

Figure 6.

Working model of POSTN signaling in basal-like breast cancer. Cancer cells which coexpress POSTN (in response to TGFβ) and integrin αvβ3 are able to activate downstream ERK signaling, which regulates the production of cytokines via NF-κB. POSTN may also be able to activate NF-κB directly (30). These soluble signals act to maintain a population of CSCs that may be responsible for the recurrence of cancers associated with a worse clinical prognosis. We suggest that this signaling network is most likely to be operative in a subset of basal-like breast cancers. Note, POSTN has also been shown to interact with Wnt1 and Wnt3A to enhance CSC maintenance (48).

Figure 6.

Working model of POSTN signaling in basal-like breast cancer. Cancer cells which coexpress POSTN (in response to TGFβ) and integrin αvβ3 are able to activate downstream ERK signaling, which regulates the production of cytokines via NF-κB. POSTN may also be able to activate NF-κB directly (30). These soluble signals act to maintain a population of CSCs that may be responsible for the recurrence of cancers associated with a worse clinical prognosis. We suggest that this signaling network is most likely to be operative in a subset of basal-like breast cancers. Note, POSTN has also been shown to interact with Wnt1 and Wnt3A to enhance CSC maintenance (48).

Close modal

No potential conflicts of interest were disclosed.

Conception and design: A.W. Lambert, S. Ozturk, C.K. Wong, P. Papageorgis, S. Thiagalingam

Development of methodology: A.W. Lambert, C.K. Wong, S. Ozturk, R. Raghunathan, B.M. Reinhard, H.M. Abdolmaleky, S. Thiagalingam

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.W. Lambert, C.K. Wong, S. Ozturk, P. Papageorgis, R. Raghunathan, Y. Alekseyev, S. Thiagalingam

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.W. Lambert, C.K. Wong, S. Ozturk, P. Papageorgis, A.C. Gower, B.M. Reinhard, S. Thiagalingam

Writing, review, and/or revision of the manuscript: A.W. Lambert, C.K. Wong, S. Ozturk, P. Papageorgis, B.M. Reinhard, H.M. Abdolmaleky, S. Thiagalingam

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.C. Gower

Study supervision: S. Thiagalingam

The authors thank Drs. David Sherr and Ramon Parsons for generously providing reagents.

This work was supported by grants from Susan G. Komen for the Cure (KG081435) and NIH/NCI (CA165707) (to S. Thiagalingam) and NIH/NCI (CA138509) (to B.M. Reinhard and S. Thiagalingam). A.W. Lambert is a recipient of a predoctoral traineeship award from the Department of Defense, Breast Cancer Research Program (W81XWH-11-1-006). P. Papageorgis was supported with a postdoctoral fellowship from the Research Promotion Foundation, Cyprus (DIDAKTOR/0609/24). This work was also supported by a seed grant from the Boston University Genome Science Institute as well as the Boston University Flow Cytometry Core Facility, and the core facilities of the Boston University Clinical and Translational Science Institute (CTSA award UL1-TR000157).

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.
Shackleton
M
,
Quintana
E
,
Fearon
ER
,
Morrison
SJ
. 
Heterogeneity in cancer: cancer stem cells versus clonal evolution
.
Cell
2009
;
138
:
822
9
.
2.
Visvader
JE
,
Lindeman
GJ
. 
Cancer stem cells in solid tumours: accumulating evidence and unresolved questions
.
Nat Rev Cancer
2008
;
8
:
755
68
.
3.
Al-Hajj
M
,
Wicha
MS
,
Benito-Hernandez
A
,
Morrison
SJ
,
Clarke
MF
. 
Prospective identification of tumorigenic breast cancer cells
.
Proc Natl Acad Sci U S A
2003
;
100
:
3983
8
.
4.
Ginestier
C
,
Hur
MH
,
Charafe-Jauffret
E
,
Monville
F
,
Dutcher
J
,
Brown
M
, et al
ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome
.
Cell Stem Cell
2007
;
1
:
555
67
.
5.
Dontu
G
,
Abdallah
WM
,
Foley
JM
,
Jackson
KW
,
Clarke
MF
,
Kawamura
MJ
, et al
In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells
.
Genes Dev
2003
;
17
:
1253
70
.
6.
Ponti
D
,
Costa
A
,
Zaffaroni
N
,
Pratesi
G
,
Petrangolini
G
,
Coradini
D
, et al
Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties
.
Cancer Res
2005
;
65
:
5506
11
.
7.
Mani
SA
,
Guo
W
,
Liao
MJ
,
Eaton
EN
,
Ayyanan
A
,
Zhou
AY
, et al
The epithelial-mesenchymal transition generates cells with properties of stem cells
.
Cell
2008
;
133
:
704
15
.
8.
Morel
AP
,
Lievre
M
,
Thomas
C
,
Hinkal
G
,
Ansieau
S
,
Puisieux
A
. 
Generation of breast cancer stem cells through epithelial-mesenchymal transition
.
PLoS ONE
2008
;
3
:
e2888
.
9.
Liu
R
,
Wang
X
,
Chen
GY
,
Dalerba
P
,
Gurney
A
,
Hoey
T
, et al
The prognostic role of a gene signature from tumorigenic breast-cancer cells
.
N Engl J Med
2007
;
356
:
217
26
.
10.
Li
X
,
Lewis
MT
,
Huang
J
,
Gutierrez
C
,
Osborne
CK
,
Wu
MF
, et al
Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy
.
J Natl Cancer Inst
2008
;
100
:
672
9
.
11.
Takebe
N
,
Ivy
SP
. 
Controversies in cancer stem cells: targeting embryonic signaling pathways
.
Clin Cancer Res
2010
;
16
:
3106
12
.
12.
Reya
T
,
Morrison
SJ
,
Clarke
MF
,
Weissman
IL
. 
Stem cells, cancer, and cancer stem cells
.
Nature
2001
;
414
:
105
11
.
13.
Korkaya
H
,
Liu
S
,
Wicha
MS
. 
Breast cancer stem cells, cytokine networks, and the tumor microenvironment
.
J Clin Invest
2011
;
121
:
3804
9
.
14.
Iliopoulos
D
,
Hirsch
HA
,
Wang
G
,
Struhl
K
. 
Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion
.
Proc Natl Acad Sci U S A
2011
;
108
:
1397
402
.
15.
Marotta
LL
,
Almendro
V
,
Marusyk
A
,
Shipitsin
M
,
Schemme
J
,
Walker
SR
, et al
The JAK2/STAT3 signaling pathway is required for growth of CD44(+)CD24(−) stem cell-like breast cancer cells in human tumors
.
J Clin Invest
2011
;
121
:
2723
35
.
16.
Ginestier
C
,
Liu
S
,
Diebel
ME
,
Korkaya
H
,
Luo
M
,
Brown
M
, et al
CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts
.
J Clin Invest
2010
;
120
:
485
97
.
17.
Conley
SJ
,
Gheordunescu
E
,
Kakarala
P
,
Newman
B
,
Korkaya
H
,
Heath
AN
, et al
Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia
.
Proc Natl Acad Sci U S A
2012
;
109
:
2784
9
.
18.
Honeth
G
,
Bendahl
PO
,
Ringner
M
,
Saal
LH
,
Gruvberger-Saal
SK
,
Lovgren
K
, et al
The CD44+/CD24− phenotype is enriched in basal-like breast tumors
.
Breast Cancer Res
2008
;
10
:
R53
.
19.
Sarrio
D
,
Rodriguez-Pinilla
SM
,
Hardisson
D
,
Cano
A
,
Moreno-Bueno
G
,
Palacios
J
. 
Epithelial-mesenchymal transition in breast cancer relates to the basal-like phenotype
.
Cancer Res
2008
;
68
:
989
97
.
20.
Creighton
CJ
,
Li
X
,
Landis
M
,
Dixon
JM
,
Neumeister
VM
,
Sjolund
A
, et al
Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features
.
Proc Natl Acad Sci U S A
2009
;
106
:
13820
5
.
21.
Papageorgis
P
,
Lambert
AW
,
Ozturk
S
,
Gao
F
,
Pan
H
,
Manne
U
, et al
Smad signaling is required to maintain epigenetic silencing during breast cancer progression
.
Cancer Res
2010
;
70
:
968
78
.
22.
Neve
RM
,
Chin
K
,
Fridlyand
J
,
Yeh
J
,
Baehner
FL
,
Fevr
T
, et al
A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes
.
Cancer Cell
2006
;
10
:
515
27
.
23.
Santner
SJ
,
Dawson
PJ
,
Tait
L
,
Soule
HD
,
Eliason
J
,
Mohamed
AN
, et al
Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cells
.
Breast Cancer Res Treat
2001
;
65
:
101
10
.
24.
Strickland
LB
,
Dawson
PJ
,
Santner
SJ
,
Miller
FR
. 
Progression of premalignant MCF10AT generates heterogeneous malignant variants with characteristic histologic types and immunohistochemical markers
.
Breast Cancer Res Treat
2000
;
64
:
235
40
.
25.
Oshima
A
,
Tanabe
H
,
Yan
T
,
Lowe
GN
,
Glackin
CA
,
Kudo
A
. 
A novel mechanism for the regulation of osteoblast differentiation: transcription of periostin, a member of the fasciclin I family, is regulated by the bHLH transcription factor, twist
.
J Cell Biochem
2002
;
86
:
792
804
.
26.
Horiuchi
K
,
Amizuka
N
,
Takeshita
S
,
Takamatsu
H
,
Katsuura
M
,
Ozawa
H
, et al
Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor beta
.
J Bone Miner Res
1999
;
14
:
1239
49
.
27.
Ruan
K
,
Bao
S
,
Ouyang
G
. 
The multifaceted role of periostin in tumorigenesis
.
Cell Mol Life Sci
2009
;
66
:
2219
30
.
28.
Charafe-Jauffret
E
,
Ginestier
C
,
Iovino
F
,
Wicinski
J
,
Cervera
N
,
Finetti
P
, et al
Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature
.
Cancer Res
2009
;
69
:
1302
13
.
29.
King
WG
,
Mattaliano
MD
,
Chan
TO
,
Tsichlis
PN
,
Brugge
JS
. 
Phosphatidylinositol 3-kinase is required for integrin-stimulated AKT and Raf-1/mitogen-activated protein kinase pathway activation
.
Mol Cell Biol
1997
;
17
:
4406
18
.
30.
Masuoka
M
,
Shiraishi
H
,
Ohta
S
,
Suzuki
S
,
Arima
K
,
Aoki
S
, et al
Periostin promotes chronic allergic inflammation in response to Th2 cytokines
.
J Clin Invest
2012
;
122
:
2590
600
.
31.
Prat
A
,
Perou
CM
. 
Mammary development meets cancer genomics
.
Nat Med
2009
;
15
:
842
4
.
32.
Gyorffy
B
,
Lanczky
A
,
Eklund
AC
,
Denkert
C
,
Budczies
J
,
Li
Q
, et al
An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients
.
Breast Cancer Res Treat
2010
;
123
:
725
31
.
33.
Shao
R
,
Bao
S
,
Bai
X
,
Blanchette
C
,
Anderson
RM
,
Dang
T
, et al
Acquired expression of periostin by human breast cancers promotes tumor angiogenesis through up-regulation of vascular endothelial growth factor receptor 2 expression
.
Mol Cell Biol
2004
;
24
:
3992
4003
.
34.
Weidner
N
,
Semple
JP
,
Welch
WR
,
Folkman
J
. 
Tumor angiogenesis and metastasis–correlation in invasive breast carcinoma
.
N Engl J Med
1991
;
324
:
1
8
.
35.
Xu
D
,
Xu
H
,
Ren
Y
,
Liu
C
,
Wang
X
,
Zhang
H
, et al
Cancer stem cell-related gene periostin: a novel prognostic marker for breast cancer
.
PLoS ONE
2012
;
7
:
e46670
.
36.
Sabatier
R
,
Finetti
P
,
Cervera
N
,
Lambaudie
E
,
Esterni
B
,
Mamessier
E
, et al
A gene expression signature identifies two prognostic subgroups of basal breast cancer
.
Breast Cancer Res Treat
2011
;
126
:
407
20
.
37.
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
.
38.
Gupta
PB
,
Onder
TT
,
Jiang
G
,
Tao
K
,
Kuperwasser
C
,
Weinberg
RA
, et al
Identification of selective inhibitors of cancer stem cells by high-throughput screening
.
Cell
2009
;
138
:
645
59
.
39.
Bornstein
P
,
Sage
EH
. 
Matricellular proteins: extracellular modulators of cell function
.
Curr Opin Cell Biol
2002
;
14
:
608
16
.
40.
Lim
E
,
Vaillant
F
,
Wu
D
,
Forrest
NC
,
Pal
B
,
Hart
AH
, et al
Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers
.
Nat Med
2009
;
15
:
907
13
.
41.
Molyneux
G
,
Geyer
FC
,
Magnay
FA
,
McCarthy
A
,
Kendrick
H
,
Natrajan
R
, et al
BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells
.
Cell Stem Cell
2010
;
7
:
403
17
.
42.
Proia
TA
,
Keller
PJ
,
Gupta
PB
,
Klebba
I
,
Jones
AD
,
Sedic
M
, et al
Genetic predisposition directs breast cancer phenotype by dictating progenitor cell fate
.
Cell Stem Cell
2011
;
8
:
149
63
.
43.
Asselin-Labat
ML
,
Sutherland
KD
,
Barker
H
,
Thomas
R
,
Shackleton
M
,
Forrest
NC
, et al
Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation
.
Nat Cell Biol
2007
;
9
:
201
9
.
44.
Herschkowitz
JI
,
Zhao
W
,
Zhang
M
,
Usary
J
,
Murrow
G
,
Edwards
D
, et al
Comparative oncogenomics identifies breast tumors enriched in functional tumor-initiating cells
.
Proc Natl Acad Sci U S A
2012
;
109
:
2778
83
.
45.
Prat
A
,
Parker
JS
,
Karginova
O
,
Fan
C
,
Livasy
C
,
Herschkowitz
JI
, et al
Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer
.
Breast Cancer Res
2010
;
12
:
R68
.
46.
Wang
X
,
Liu
J
,
Wang
Z
,
Huang
Y
,
Liu
W
,
Zhu
X
, et al
Periostin contributes to the acquisition of multipotent stem cell-like properties in human mammary epithelial cells and breast cancer cells
.
PLoS ONE
2013
;
8
:
e72962
.
47.
Yu
F
,
Deng
H
,
Yao
H
,
Liu
Q
,
Su
F
,
Song
E
. 
Mir-30 reduction maintains self-renewal and inhibits apoptosis in breast tumor-initiating cells
.
Oncogene
2010
;
29
:
4194
204
.
48.
Malanchi
I
,
Santamaria-Martinez
A
,
Susanto
E
,
Peng
H
,
Lehr
HA
,
Delaloye
JF
, et al
Interactions between cancer stem cells and their niche govern metastatic colonization
.
Nature
2012
;
481
:
85
9
.
49.
Balko
JM
,
Schwarz
LJ
,
Bhola
NE
,
Kurupi
R
,
Owens
P
,
Miller
TW
, et al
Activation of MAPK pathways due to DUSP4 loss promotes cancer stem cell-like phenotypes in basal-like breast cancer
.
Cancer Res
2013
;
73
:
6346
58
.
50.
Iliopoulos
D
,
Hirsch
HA
,
Struhl
K
. 
An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation
.
Cell
2009
;
139
:
693
706
.
51.
Gupta
PB
,
Fillmore
CM
,
Jiang
G
,
Shapira
SD
,
Tao
K
,
Kuperwasser
C
, et al
Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells
.
Cell
2011
;
146
:
633
44
.
52.
Ghajar
CM
,
Peinado
H
,
Mori
H
,
Matei
IR
,
Evason
KJ
,
Brazier
H
, et al
The perivascular niche regulates breast tumour dormancy
.
Nat Cell Biol
2013
;
15
:
807
17
.
53.
Zhou
W
,
Ke
SQ
,
Huang
Z
,
Flavahan
W
,
Fang
X
,
Paul
J
, et al
Periostin secreted by glioblastoma stem cells recruits M2 tumor-associated macrophages and promotes malignant grwoth
.
Nat Cell Biol
2015
;
17
:
170
82
.
54.
Seguin
L
,
Kato
S
,
Franovic
A
,
Camargo
MF
,
Lesperance
J
,
Elliott
KC
, et al
An integrin beta(3)-KRAS-RalB complex drives tumour stemness and resistance to EGFR inhibition
.
Nat Cell Biol
2014
;
16
:
457
68
.