Skip to main content

OPINION article

Front. Oncol., 27 September 2018
Sec. Molecular and Cellular Oncology

Does the Sphingosine 1-Phosphate Receptor-1 Provide a Better or Worse Prognostic Outcome for Breast Cancer Patients?

  • Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom

Introduction

The bioactive lipid, sphingosine 1-phosphate (S1P) is formed by the sphingosine kinase-catalyzed phosphorylation of sphingosine. There are two isoforms of sphingosine kinase termed SphK1 and SphK2 that are encoded by different genes and which exhibit distinct sub-cellular localization and biochemical properties to regulate overlapping and non-overlapping biology (1). S1P is either degraded by S1P lyase to produce (E)-2-hexadecenal and phosphoethanolamine or reversibly dephosphorylated by S1P phosphatase to regenerate sphingosine (1). S1P can be exported from cells [through transporter proteins such as Spns2 (2), MFsd2b (3), and some ABC proteins (4)] to bind to a family of five S1P-specific G protein coupled receptors (S1P1−5) (5). There is substantial evidence to suggest that S1P has a critical role in cancer. For example, the knockout of lymphatic endothelial Spns2 reduces pulmonary metastasis via a mechanism that involves induction of a lymphopenia and an increase in effector T cells and natural killer (NK) cells to enhance tumor cell killing in the lung (6). Other evidence supports a role for S1P in transformation, epithelial mesenchymal transition (EMT), invasiveness, cancer cell survival and replicative immortality, tumor neovascularisation, and the Warburg effect (7).

Role of S1P in Defining Breast Cancer Prognosis

Lei et al. (8) used data mining and the Kaplan-Meier plotter (http://kmplot.com/analysis/index.php?p=service&cancer=breast) to establish relationships between messenger RNA levels (rather than protein) of SphK1, SphK2, sphingosine 1-phosphate receptor 1 (S1P1), or sphingosine 1-phosphate receptor 2 (S1P2) and 10 year relapse free survival of non-classified breast cancer (BCA) patients. High SphK1 mRNA in tumors had no effect on 10 year relapse free disease survival, while high SphK2 mRNA was found to have a positive impact in patients with non-classified, or basal type BCA and those who had received adjuvant therapy. Lei et al. (8) also reported that high S1P1 mRNA in tumors was associated with improved survival in patients with non-classified BCA and in those receiving treatment, but had no effect on patients with basal cell type BCA. In addition, improved survival was associated with high S1P2 mRNA in tumors from patients with non-classified BCA, basal cell type BCA and those patients receiving treatment.

We suggest that the interpretation of these findings should be viewed with considerable caution. In this regard, Kaplan-Meier plot analysis relating gene expression, i.e., mRNA transcript, with prognosis does not consider the possibility that mRNA transcript expression may not reflect specific protein levels. Indeed, it is the protein itself that performs the signaling function, and not the mRNA. In this regard, if the turnover of a particular protein is slow, its mRNA levels might be low. Therefore, a reciprocal relationship could exist between mRNA and protein levels. In this case, interpretation of the results by Lei et al. (8) would be completely reversed. For instance, it could be concluded that high expression of S1P1 protein (low mRNA transcript) in BCA tumors is linked with a poorer prognosis. Indeed, this was precisely the findings of Watson et al. (9) where the estrogen receptor positive (ER+) breast cancer tumors of a population of 304 patients was analyzed. In this respect, high protein expression of S1P1 or SphK1 (detected using immunohistochemical staining with anti-S1P1 antibody or anti-SphK1 antibody) in tumors was associated with earlier disease recurrence and reduced disease specific survival times. The difference for S1P1 is not trivial with mean disease recurrence and disease specific survival times being shortened by between 3 and 8 years, respectively.

In addition, a relationship with poor prognosis was evident for both membrane and cytoplasmic localized S1P1; the latter probably reflects receptors that have been endocytosed in response to S1P, and which are capable of persistent signaling (10). In contrast, combined high nuclear localized S1P2 and c-Src protein expression in tumors is associated with increased disease specific survival times (11). Indeed, a thorough analysis of the impact of S1P receptor sub-types on breast cancer prognosis should also consider sub-cellular localization and this can only be performed using analysis of protein with validated anti-S1P receptor antibodies. There is a very substantial body of evidence that demonstrates that S1P1 promotes tumorigenesis through, for example, enhanced cell survival, migration, and angiogensis (7). The role of S1P2 in cancer is more controversial with evidence that it can either promote or inhibit tumorigenesis (12, 13). To rationalize these findings, we have proposed that the positive and negative function of the S1P2 receptor in cancer might be dependent on its sub-cellular localization (14).

Patient stratification based on ER, progesterone receptor and HER2 status is also important. This is exemplified by the finding that high expression of SphK1 protein in tumors actually increases disease specific survival times in ER+/HER2+ patients. Indeed, high expression of SphK1 induces a desensitization/tolerance to HER2-mediated signaling in ER+/HER2+ breast cancer cells (15).

S1P Receptor-Dependent Mechanisms

To provide a mechanistic explanation for the clinical findings, Lei et al. (8) reported the inhibition of colony formation by MCF-7 cells (ER+) or triple negative MDA-MB-231 cells in response to treatment with S1P (4–10 μM) for 2 weeks. Using shRNA knockdown of either S1P1 or S1P2 in MCF-7 and MDA-MB-231 cells, the authors showed that the inhibitory effect of S1P on colony formation was enhanced in both cell types upon S1P2 knockdown whereas S1P1 knockdown reduced the effect in MDA-MB-231 cells and was without effect in MCF-7 cells. However, there was no evidence presented to indicate that the effect of S1P on colony formation actually reflects an inhibition of growth assessed by, for instance, cell-cycle analysis. This is critically important in order to validate the conclusions, because S1P stimulates migration via S1P3 (15) and S1P1 (16). In contrast, the S1P2 receptor has been linked with inhibition of cell migration (17). Therefore, the findings by Lei et al. (8) regarding the effect of shRNA knockdown of S1P2 are in fact consistent with a relief of the inhibitory constraint of S1P2 on the migration of MCF-7 and MDA-MB-231 cells in response to added S1P. This would reduce colony formation by enhancing dispersal and thereby preventing colony residency. The knockdown of S1P1 in reducing the inhibitory effect of S1P on colony formation of MDA-MB-231 cells is consistent with ablation of the positive effect of S1P1 on migration, which would enhance colony formation/residency. Thus, colony formation (without more extensive cell cycle analysis and assessment of migration) does not provide confirmation that S1P inhibits tumorigenesis in vitro. The effect of treatment with S1P for 2 weeks on S1P receptor expression was also not examined by Lei et al. (8).

The major functional S1P receptor type in MCF-7 cells is, in fact, the S1P3 receptor which induces activation of the ERK-1/2 pathway via transactivation of the EGF receptor to promote migration (18, 19). S1P3 expression/function was not analyzed by Lei et al. (8). Furthermore, the EGF receptor kinase is positively linked with tumorigenesis and this is a rationale for the use of EGF receptor kinase inhibitors for the treatment of cancer. Therefore, in contrast to the authors' conclusions, these prior published findings strongly implicate S1P in promoting breast cancer tumorigenesis via an EGF receptor-dependent mechanism. Finally, the study by Lei et al. (8) takes no account of the other responses that S1P regulates through its receptors and which are of relevance to cancer, such as migration, angiogenesis and chemo-resistance.

Conclusion

We conclude that meaningful analysis of the impact of tumor S1P receptor sub-type expression on the survival of breast cancer patients requires analysis of actual protein levels and not mRNA. There is also a need to stratify patient cohorts to consider for instance, ER and HER2 expression. In addition, the effects of S1P on colony formation of MCF-7 and MDA-MB-231 cells requires analysis of cell cycle progression and migration with respect to the function of different S1P receptor sub-types. Moreover, a comprehensive analysis of the protein expression of all S1P receptors in the breast cancer cells studied and their individual biological functions should be interrogated. This is important as it is unknown whether, for instance, knockdown of S1P1 and/or S1P2 has an effect on the expression of the other S1P receptors and whether there is redundancy and/or divergent biological functions of these receptors. Indeed, this appears to be the case as evidenced by lack of effect of S1P1 knockdown in MCF7 cells, where S1P3 may play a more prominent role. Finally, Lei et al. (8) conclude that breast cancer could be treated with S1P1 specific agonists, but provide no in vivo data to support this. In contrast, we argue that the weight of published evidence clearly suggests that such S1P1 specific agonists have the potential to reduce disease specific survival times for breast cancer patients which is counterproductive in terms of therapy.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Pyne S, Adams DR, Pyne NJ. Sphingosine 1-phosphate and sphingosine kinases in health and disease: recent advances. Prog Lipid Res. (2016) 62:93–106. doi: 10.1016/j.plipres.2016.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Hisano Y, Kobayashi N, Yamaguchi A, Nishi T. Mouse SPNS2 functions as a sphingosine-1-phosphate transporter in vascular endothelial cells. PLoS ONE (2012) 7:e3894. doi: 10.1371/journal.pone.0038941

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Vu TM, Ishizu AN, Foo JC, Toh XR, Zhang F, Whee DM, et al. Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets. Nature (2017) 550:524–8. doi: 10.1038/nature24053

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Kobayashi N, Kobayashi N, Yamaguchi A, Nishi T. Characterization of the ATP dependent sphingosine 1-phosphate transporter in rat erythrocytes. J Biol Chem. (2009) 284:21192–200. doi: 10.1074/jbc.M109.006163

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Blaho VA, Hla T. An update on the biology of sphingosine 1-phosphate receptors. J Lipid Res. (2014) 55:1596–608. doi: 10.1194/jlr.R046300

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Weyden LV, Arends MJ, Campbell AD, Bald T, Wardle-Jones H, Griggs N, et al. Genome-wide in vivo screen identifies novel host regulators of metastatic colonisation. Nature (2017) 541:233–6. doi: 10.1038/nature20792

CrossRef Full Text | Google Scholar

7. Pyne NJ, Pyne S. Sphingosine 1-phosphate and cancer. Nat Rev Cancer (2010) 10:489–503. doi: 10.1038/nrc2875

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Lei FJ, Cheng BH, Liao PY, Wang HC, Chang WC, Lai HC, et al. Survival benefit of sphingosin-1-phosphate and receptors expressions in breast cancer patients. Cancer Med. (2018) 7:3743–54. doi: 10.1002/cam4.1609

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Watson C, Long JS, Orange C, Tannahill CL, Mallon E, McGlynn LM, et al. High expression of sphingosine 1-phosphate receptors, S1P1 and S1P3, sphingosine kinase 1, and extracellular signal-regulated kinase-1/2 is associated with development of tamoxifen resistance in estrogen receptor-positive breast cancer patients. Am J Pathol. (2010) 177:2205–15. doi: 10.2353/ajpath.2010.100220

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Pyne NJ, Pyne S. Sphingosine 1-phosphate receptor 1 signalling in mammalian cells. Molecules (2017) 22:E344. doi: 10.3390/molecules22030344

CrossRef Full Text | Google Scholar

11. Ohotski J, Edwards J, Elsberger B, Watson C, Orange C, Mallon E, et al. Identification of novel functional and spatial associations between sphingosine kinase 1, sphingosine 1-phosphate receptors and other signalling proteins that affect prognostic outcome in estrogen receptor-positive breast cancer. Int J Cancer (2013) 132:605–16. doi: 10.1002/ijc.27692

CrossRef Full Text | Google Scholar

12. Lepley D, Paik JH, Hla T, Ferrer F. The G protein-coupled receptor S1P2 regulates Rho/Rho kinase pathway to inhibit tumor cell migration. Cancer Res. (2005) 65:3788–95. doi: 10.1158/0008-5472.CAN-04-2311

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Ponnusamy S, Selvam SP, Mehrotra S, Kawamori T, Snider AJ, Obeid LM, et al. Communication between host organism and cancer cells is transduced by systemic sphingosine kinase 1/sphingosine 1-phosphate signalling to regulate tumour metastasis. EMBO Mol Med. (2012) 4:761–75. doi: 10.1002/emmm.201200244

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Ohotski J, Rosen H, Bittman R, Pyne S, Pyne NJ. Sphingosine kinase 2 prevents the nuclear translocation of sphingosine 1-phosphate receptor-2 and tyrosine 416 phosphorylated c-Src and increases estrogen receptor negative MDA-MB-231 breast cancer cell growth: the role of sphingosine 1-phosphate receptor-4. Cell Signal. (2014) 26:1040–7. doi: 10.1016/j.cellsig.2014.01.023

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Long JS, Edwards J, Watson C, Tovey S, Mair KM, Schiff R, et al. Sphingosine kinase 1 induces tolerance to human epidermal growth factor receptor 2 and prevents formation of a migratory phenotype in response to sphingosine 1-phosphate in estrogen receptor-positive breast cancer cells. Mol Cell Biol. (2010) 30:3827–41. doi: 10.1128/MCB.01133-09

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Waters CM, Long J, Gorshkova I, Fujiwara Y, Connell M, Belmonte KE, et al. Cell migration activated by platelet-derived growth factor receptor is blocked by an inverse agonist of the sphingosine 1-phosphate receptor-1. FASEB J. (2006) 20:509–11. doi: 10.1096/fj.05-4810fje

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Sugimoto N, Takuwa N, Okamoto H, Sakurada S, Takuwa Y. Inhibitory and stimulatory regulation of Rac and cell motility by the G12/13-Rho and Gi pathways integrated downstream of a single G protein-coupled sphingosine-1-phosphate receptor isoform. Mol Cell Biol. (2003) 23:1534–45. doi: 10.1128/MCB.23.5.1534-1545.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Sukocheva O, Wadham C, Holmes A, Albanese N, Verrier E, Feng F, et al. Estrogen transactivates EGFR via the sphingosine 1-phosphate receptor Edg-3: the role of sphingosine kinase-1. J Cell Biol. (2006) 173:301–10. doi: 10.1083/jcb.200506033

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Long JS, Fujiwara Y, Edwards J, Tannahill CL, Tigyi G, Pyne S, et al. Sphingosine 1-phosphate receptor 4 uses HER2 (ERBB2) to regulate extracellular signal regulated kinase-1/2 in MDA-MB-453 breast cancer cells. J Biol Chem. (2010) 285:35957–66. doi: 10.1074/jbc.M110.117945

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Sphingosine 1-phosphate, breast cancer, Sphingosine 1-phosphate receptor, sphingosine kinase, disease specific survival, cell growth, cell migration

Citation: Pyne NJ and Pyne S (2018) Does the Sphingosine 1-Phosphate Receptor-1 Provide a Better or Worse Prognostic Outcome for Breast Cancer Patients? Front. Oncol. 8:417. doi: 10.3389/fonc.2018.00417

Received: 14 July 2018; Accepted: 10 September 2018;
Published: 27 September 2018.

Edited by:

Luisa Lanfrancone, Istituto Europeo di Oncologia s.r.l., Italy

Reviewed by:

Gabriele Multhoff, Technische Universität München, Germany
Julie Valerie Decock, Qatar Biomedical Research Institute, Qatar

Copyright © 2018 Pyne and Pyne. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Nigel J. Pyne, n.j.pyne@strath.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.