Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Cancer Discov. 2012 Sep;2(9):775–777. doi: 10.1158/2159-8290.CD-12-0312

Paracrine signaling between carcinoma cells and mesenchymal stem cells generates cancer stem cell niche via epithelial-mesenchymal transition

Kati Räsänen 1, Meenhard Herlyn 1
PMCID: PMC3646332  NIHMSID: NIHMS467292  PMID: 22969117

Summary

Li and colleagues present data that cancer cell-derived intereleukin-1 induces prostaglandin E2 and cytokine secretion in mesenchymal stem cells (MSC) to activate β-catenin signaling in the cancer cell. This paracrine signaling between carcinoma cells and MSC leads to the creation of a cancer stem cell niche via epithelial-mesenchymal transition.


Tumorigenesis is a multistep process, and carcinomas – cancers of epithelial origin – were long viewed as a clonal disease. However, the emergence of tumor microenvironment's role in cancer progression first proposed ten years ago by Bissell and colleagues (1) brought into light the need to study each component of the stroma. The tumor stroma is the compartment that serves as a tissue framework, consisting the vasculature (endothelial, smooth muscle cells and pericytes), inflammatory and immune cells (macrophages, lymphocytes and dendritic cells), and cancer associated fibroblasts (CAFs). Mesenchymal stem cells (MSCs) are recruited from the bone marrow to the stroma, a process that is mediated mainly by inflammatory factors present in tumor microenvironment (2). Once recruited to the tumor, MSCs can act as precursors for CAFs, which represents a distinct cell type characterized by heterogeneous expression of alpha-smooth muscle actin, fibroblast activation protein and fibroblast specific protein 1 (α-SMA, FAP and FSP1, respectively). CAFs secrete a variety of inflammatory cytokines, growth factors and proteinases, all of which contribute to tumor progression. These factors include interleukins (ILs), chemokine C-X-C motif ligand cytokines (CXCLs), vascular endothelial and hepatocyte growth factors (VEGF and HGF) and matrix metalloproteinases (MMPs) that affect the surrounding components of the tumor microenvironment, including cancer cells and cells of vasculature and immunity. In addition, factors secreted by CAFs modify the extracellular matrix (ECM) releasing collagens (COL), fibronectin (FN) and periostatin (POSTN) that further act on the cancer cells (3).

Although the existence of cancer stem cells (CSCs, or alternatively called tumor initiating cells, TICs) in carcinomas has been commonly accepted, the origin of CSCs remains under debate, and epithelial-mesenchymal transition (EMT) has been proposed as one possible mechanism (4). The hierarchical model of clonal evolution does not take into account the heterogeneity frequently observed in cancers, thus the role of the tumor microenvironment creating dynamic subpopulations presents an alternative model (5). In this issue of Cancer Discovery, Li and colleagues provide evidence that MSCs can directly promote EMT in the carcinoma cells that in turn leads to entrance into a stem-cell like state (6).

Using transwell co-culture assays the authors demonstrated significant increase in cyclo-oxygenase 2 (COX-2) expression that led to enzymatic conversion of arachidonic acid to prostaglandin E2 (PGE2) in MSCs. Concomitantly, they observed secretion of interleukin 1 (IL-1) by a panel of human colorectal and breast cancer cell lines. Similar increases in PGE2 production in MSC cells wer observed when recombinant IL-1 was used. Conversely, IL-1 neutralizing antibody or recombinant antagonist of the IL-1 receptor (IL-1ra) decreased the ability of MSCs to produce PGE2. Notably, the intracellular levels IL-1 and IL-1ra were heterogeneous between the cell lines, and only cells that secreted IL-1 were able to induce PGE2 in MSCs. In concert with the COX-2 and PGE2, levels of Gro-α (aka CXCL1), IL-6 and IL-8 were induced in MSCs by LoVo colorectal carcinoma cell conditioned medium or by adding exogenous IL-1 to the MSC cultures. IL-1 shRNA expressing LoVo cells attenuated the PGE2 production; levels of Gro-α, IL-6 and IL-8 were also decreased in MSCs. Further, COX-1/COX-2 inhibitor indomethacin and PGE2 cell surface receptor antagonists AH6809 and GW627368X (against EP2 and EP4, respectively), reduced the production of the aforementioned cytokines in both LoVo conditioned medium and IL-1 treated MSC cells. These data confirmed the necessary role of cancer cell-derived IL-1 in driving PGE2 synthesis in the MSCs and that the cytokine production in the MSCs was dependent on the autocrine secretion of PGE2. In line with these results, a recent study by Alcolea and colleagues showed that head and neck squamous cell carcinoma-derived IL-1 leads to upregulation of COX-2 and PGE2 biosynthesis in fibroblasts (7).

Previous work by the Weinberg laboratory has proposed a possible presence of mesenchymal-like cells which via EMT have gained stem-cell properties in mammary carcinoma (8). In that study EMT was induced either by transforming growth factor beta (TGF-β), a well-characterized inducer of EMT, or by forced expression of the EMT transcription factors Twist or Slug. This led to a cadherin switch from E- to N-cadherin expression, and de novo synthesis of mesenchymal proteins vimentin and fibronectin. Importantly, the majority of the mesenchymal cells acquired a CD44hi / CD24lo expression pattern that is frequently observed in mammary stem cells. In light of this, Li and colleagues investigated whether MSC co-cultures lead to EMT and a CSC phenotype in the carcinoma cells. FACS analysis of colon carcinoma / MSC co-cultures revealed significant decrease of E-cadherin expression in LoVo cells, with simultaneous upregulation of Snail, vimentin and fibronectin, thus confirming induction of EMT in the cancer cells. Similar results were obtained when LoVo cells were treated with PGE2 together with Gro-α, IL-6 and IL-8. However, PGE2 or the cytokines alone were not able to elicit a complete induction of EMT, thus suggesting the requirement of multiple signals acting at the same time. The mesenchymal phenotype obtained via EMT significantly augmented LoVo cell migration in a transwell assay; effect that was reversed by the selective COX-2 inhibitor NS398, and partially rescued by addition of PGE2.

In vivo experiments conducted by Li and colleagues revealed that co-injecting carcinoma cells with MSCs formed more invasive tumors and led to intravasation of LoVo cells into microvessels. Further, extreme limiting dilution analysis (ELDA) uncovered that the frequency of TICs increased by several magnitudes in xenografts containing both LoVo and MSC cells, corroborating data published by Rudnick and colleagues showing the expansion of mammary CSC population by PGE2 -secreting fibroblasts (9). What remains to be answered by these studies is whether the increased number of CSCs contributes also to metastasis, which is implied by the presence of carcinoma cells in the nearby microvessels.

Aldehyde dehydrogenase 1 (ALDH1) expression is considered to be a marker for identifying cancer stem cells. ALDH1 acts as a detoxifying enzyme in cells, thus contributing to therapeutic resistance (10). Co-culturing colorectal carcinoma cells with MSCs increased ALDH1 expression in the cancer cells and expanded the ALDHhi subpopulation, but only if the cancer cells were in continuous presence of MSC cells. To further characterize this subpopulation, the authors used a second commonly accepted CSC marker, CD133, a transmembrane glycoprotein known to be expressed in hematopoietic stem cells. As with the ALDHhi subset, the number of ALDHhi /CD133+ LoVo cells increased in MSC co-cultures. Further dissecting the role of this stem cell-like population in tumor propagation, ALDHhi cells were injected subcutaneously into nude mice. ELDA calculations revealed increased TIC frequency in ALDHhi LoVo cells, effect that was further augmented by co-injection of MSCs. Tumor initiating capacity of ALDHlo carcinoma cells was also increased by the presence of MSC cells. This effect was mediated by the PGE2 produced in the MSCs, and the EP4 antagonist was able to reduce the levels of ALDH1 in the LoVo cells. In addition, ex vivo treatment of LoVo cells with PGE2 increased the number of ALDHhi cells and TIC frequency in SCID mice. This effect was abolished when PGE2 synthesis was inhibited, either with small molecule inhibitors or with RNAi directed against COX-2. In the xenografts, ALDHhi LoVo cells were surrounded by COX-2- and FSP-expressing MSC cells, confirming the notion that within the tumor microenvironment MSCs act as precursors for CAFs (3). These data suggests a critical role of MSC-derived PGE2 in maintaining the ALDHhi cancer stem cell-like subpopulation in carcinomas. Further, in clinical samples COX-2 expression correlated with IL-1 expression, with higher expression reflecting more aggressive breast and colon carcinoma phenotypes.

To further dissect the mechanism by which PGE2 elicits these functions, Li and colleagues investigated the downstream signaling pathways that are known to maintain the CSC phenotype, among them β-catenin signaling. In the absence of Wnt, glycogen synthase kinase 3 (GSK-3) phosphorylates β-catenin at Ser33/Ser37/Thr41 residues, thus marking it for proteosomal degradation. PGE2 treatment of LoVo cells activated Akt at Thr473, which in turn increased β-catenin phosphorylation at Ser552 residue, thus rendering it inaccessible for GSK-3. In addition, PGE2 increased GSK-3 Ser21 phosphorylation, thus decreasing the GSK-3 activity. The EP4 receptor antagonist GW627368X was able to reverse these effects, leading to degradation of β-catenin. Similar effects were observed when PGE2-induced ALDHhi LoVo cells were treated with the phosphatidylinositol 3-kinase inhibitor LY294002, confirming the role of Akt in mediating β-catenin signaling. Downregulation of E-cadherin, orchestrated by PGE2-induced EMT, released β-catenin bound in adherens junctions and enabled its translocation into the nucleus, thus upregulating β-catenin / TCF target genes, including pluripotency regulators Nanog, Oct4 and Sox2. This data further strengthens the observation that PGE2 plays a role in creating and maintaining a cancer stem cell niche via EMT.

The cancer stem cell model has manifested new implications for cancer therapy. In addition of ALDH, many CSCs have been found to express adenosine triphosphate-binding cassette (ABC) transporters which efficiently pump out various chemotherapeutic drugs, leading to resistance (5,10). In addition, other components of the tumor microenviroment are known to confer drug resistance, such as leaky blood vessels generated by reactivation of angiogenesis in response to targeted therapy. The key findings of Li and colleagues, put in context with other interactions between carcinoma cells and components of the tumor microenvironment discussed in this commentary, are summarized in Figure 1. As it becomes clear from the schematic illustration, a variety of factors contributes to the generation of a cancer stem cell niche, and importantly all of these subpopulations should be targeted in order to achieve effective therapeutic responses. Relevantly, the data presented here by Li and colleagues offers an insight into how targeting one component of the tumor could affect another component, and thus lead to tumor regression. IL-1 inhibitors are used clinically to treat inflammatory conditions, and are currently in early clinical trials for various solid cancers. In addition, COX-2 inhibitors are currently investigated for treatment of carcinomas, although severe side effects have been reported. In the future the goal of personalized therapy should focus in targeting the various cell types present in the tumor microenvironment, thus emphasizing the need of systematic preclinical testing of combination therapies.

Figure 1.

Figure 1

Key findings of Li and colleagues illustrating the interactions of cancer cells with MSCs that creates a cancer stem cell niche via EMT (left side of the dashed line) and how it supports previously published data regarding tumor initiating cells and tumor – stroma interactions discussed in the commentary (right hand side of the dashed line). For abbreviations, see text.

Acknowledgments

Grant support. Work in the authors’ laboratory is supported by grants from the National Cancer Institute (CA025874, CA114046, CA098101 and CA047159).

Footnotes

Disclosure of potential conflicts of interest. No potential conflicts of interest.

REFERENCES

  • 1.Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1:46–54. doi: 10.1038/35094059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hogan NM, Dwyer RM, Joyce MR, Kerin M. Mesenchymal stem cells in the colorectal tumor microenvironment: Recent progress and implications. Int J Cancer. 2012;131:1–7. doi: 10.1002/ijc.27458. [DOI] [PubMed] [Google Scholar]
  • 3.Rasanen K, Vaheri A. Activation of fibroblasts in cancer stroma. Exp Cell Res. 2010;316:2713–22. doi: 10.1016/j.yexcr.2010.04.032. [DOI] [PubMed] [Google Scholar]
  • 4.Ansieau S. EMT in breast cancer stem cell generation. Cancer Letters. 2012 May 22; doi: 10.1016/j.canlet.2012.05.014. [Epub ahead of print] DOI:10.1016/j.canlet.2012.05.014. [DOI] [PubMed] [Google Scholar]
  • 5.Fukunaga-Kalabis M, Roesch A, Herlyn M. From cancer stem cells to tumor maintenance in melanoma. J Invest Dermatol. 2011;131:1600–4. doi: 10.1038/jid.2011.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li H-J, Reinhardt F, Herschman HR, Weinberg RA. Cancer-stimulated mesenchymal stem cells create a carcinoma stem-cell niche via prostaglandinn E2 signaling. Cancer Discovery. 2012 doi: 10.1158/2159-8290.CD-12-0101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Alcolea S, Anton R, Camacho M, Soler M, Alfranca A, Aviles-Jurado F-X, et al. Interaction between head and neck squamous cell carcinoma cells and fibroblasts in the biosynthesis of PGE2. J Lipid Res. 2012;53:630–42. doi: 10.1194/jlr.M019695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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(4):704–15. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rudnick JA, Arendt LM, Klebba I, Hinds JW, Iyer V, Gupta PB, et al. Functional heterogeneity of breast fibroblasts is defined by a prostaglandin secretory phenotype that promotes expansion of cancer-stem like cells. PLoS One. 2011;6:e24605. doi: 10.1371/journal.pone.0024605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Biddle A, Mackenzie IC. Cancer stem cells and EMT in carcinoma. Cancer Metastasis Rev. 2012;31:285–93. doi: 10.1007/s10555-012-9345-0. [DOI] [PubMed] [Google Scholar]

RESOURCES