Keep a Little Fire Burning—The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity
Abstract
:1. Introduction
1.1. Cancer
1.2. The Tumor Microenvironment
2. The Immune System in Cancer
2.1. Tumor-Promoting Inflammation
2.2. Anti-Tumor Immunity
2.3. The Tumor-Supporting Microenvironment
3. S1P Signaling and Immune Cell Dynamics
3.1. S1P Metabolism and Signaling
3.2. S1P and Immune Cell Trafficking
4. S1P Signaling and Tumor Immunity
4.1. S1P Promotes Cancer Development
4.2. Sphingosine Kinases and Tumor Immunity
4.3. S1P Degrading Enzymes and Tumor Immunity
4.4. S1P Export and Tumor Immunity
4.5. S1P Receptors in Tumor Immunity
4.5.1. S1PR1
4.5.2. S1PR2
4.5.3. S1PR3
4.5.4. S1PR4
4.5.5. S1PR5
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
- Anand, P.; Kunnumakkara, A.B.; Sundaram, C.; Harikumar, K.B.; Tharakan, S.T.; Lai, O.S.; Sung, B.; Aggarwal, B.B. Cancer is a preventable disease that requires major lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. [Google Scholar] [CrossRef] [PubMed]
- Trinchieri, G. Cancer and inflammation: An old intuition with rapidly evolving new concepts. Annu. Rev. Immunol. 2012, 30, 677–706. [Google Scholar] [CrossRef] [PubMed]
- GBD 2019 Risk Factors Collaborators. Global burden of 87 risk factors in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease study 2019. Lancet 2020, 396, 1223–1249. [Google Scholar] [CrossRef]
- Hui, L.; Chen, Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett. 2015, 368, 7–13. [Google Scholar] [CrossRef]
- Zitvogel, L.; Kepp, O.; Kroemer, G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat. Rev. Clin. Oncol. 2011, 8, 151–160. [Google Scholar] [CrossRef]
- Zemek, R.M.; Chin, W.L.; Nowak, A.K.; Millward, M.J.; Lake, R.A.; Lesterhuis, W.J. Sensitizing the tumor microenvironment to immune checkpoint therapy. Front. Immunol. 2020, 11, 223. [Google Scholar] [CrossRef] [Green Version]
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [Green Version]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greten, F.R.; Grivennikov, S.I. Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef] [PubMed]
- Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Del Poggetto, E.; Ho, I.L.; Balestrieri, C.; Yen, E.Y.; Zhang, S.; Citron, F.; Shah, R.; Corti, D.; Diaferia, G.R.; Li, C.Y.; et al. Epithelial memory of inflammation limits tissue damage while promoting pancreatic tumorigenesis. Science 2021, 373, eabj0486. [Google Scholar] [CrossRef] [PubMed]
- Nathan, C.; Cunningham-Bussel, A. Beyond oxidative stress: An immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 2013, 13, 349–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Canli, O.; Nicolas, A.M.; Gupta, J.; Finkelmeier, F.; Goncharova, O.; Pesic, M.; Neumann, T.; Horst, D.; Lower, M.; Sahin, U.; et al. Myeloid Cell-derived reactive oxygen species induce Epithelial Mutagenesis. Cancer Cell 2017, 32, 869–883. [Google Scholar] [CrossRef] [Green Version]
- Weigert, A.; Brune, B. Nitric oxide, apoptosis and macrophage polarization during tumor progression. Nitric. Oxide 2008, 19, 95–102. [Google Scholar] [CrossRef]
- Hou, J.; Karin, M.; Sun, B. Targeting cancer-promoting inflammation—Have anti-inflammatory therapies come of age? Nat. Rev. Clin. Oncol. 2021, 18, 261–279. [Google Scholar] [CrossRef]
- Boon, T.; Cerottini, J.C.; Van den Eynde, B.; van der Bruggen, P.; Van Pel, A. Tumor antigens recognized by T lymphocytes. Annu. Rev. Immunol. 1994, 12, 337–365. [Google Scholar] [CrossRef]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
- Matsushita, H.; Vesely, M.D.; Koboldt, D.C.; Rickert, C.G.; Uppaluri, R.; Magrini, V.J.; Arthur, C.D.; White, J.M.; Chen, Y.S.; Shea, L.K.; et al. Cancer exome analysis reveals a T-Cell-dependent mechanism of cancer immunoediting. Nature 2012, 482, 400–404. [Google Scholar] [CrossRef] [PubMed]
- Joseph, C.G.; Darrah, E.; Shah, A.A.; Skora, A.D.; Casciola-Rosen, L.A.; Wigley, F.M.; Boin, F.; Fava, A.; Thoburn, C.; Kinde, I.; et al. Association of the autoimmune disease scleroderma with an immunologic response to cancer. Science 2014, 343, 152–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galon, J.; Bruni, D. Tumor immunology and tumor evolution: Intertwined histories. Immunity 2020, 52, 55–81. [Google Scholar] [CrossRef]
- Martin, T.D.; Patel, R.S.; Cook, D.R.; Choi, M.Y.; Patil, A.; Liang, A.C.; Li, M.Z.; Haigis, K.M.; Elledge, S.J. The adaptive immune system is a major driver of selection for tumor suppressor gene inactivation. Science 2021, 373, 1327–1335. [Google Scholar] [CrossRef] [PubMed]
- Rosenthal, R.; Cadieux, E.L.; Salgado, R.; Bakir, M.A.; Moore, D.A.; Hiley, C.T.; Lund, T.; Tanic, M.; Reading, J.L.; Joshi, K.; et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 2019, 567, 479–485. [Google Scholar] [CrossRef]
- Bruni, D.; Angell, H.K.; Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef] [PubMed]
- Mlecnik, B.; Bindea, G.; Van den Eynde, M.; Galon, J. Metastasis immune-based scores predict patient survival. Oncoimmunology 2020, 9, 1806000. [Google Scholar] [CrossRef] [PubMed]
- Mascaux, C.; Angelova, M.; Vasaturo, A.; Beane, J.; Hijazi, K.; Anthoine, G.; Buttard, B.; Rothe, F.; Willard-Gallo, K.; Haller, A.; et al. Immune evasion before tumour invasion in early lung squamous carcinogenesis. Nature 2019, 571, 570–575. [Google Scholar] [CrossRef] [PubMed]
- Imai, K.; Matsuyama, S.; Miyake, S.; Suga, K.; Nakachi, K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: An 11-year follow-up study of a general population. Lancet 2000, 356, 1795–1799. [Google Scholar] [CrossRef]
- Roithmaier, S.; Haydon, A.M.; Loi, S.; Esmore, D.; Griffiths, A.; Bergin, P.; Williams, T.J.; Schwarz, M.A. Incidence of malignancies in heart and/or lung transplant recipients: A single-institution experience. J. Heart Lung Transplant. 2007, 26, 845–849. [Google Scholar] [CrossRef]
- MacKie, R.M.; Reid, R.; Junor, B. Fatal melanoma transferred in a donated kidney 16 years after melanoma surgery. N. Engl. J. Med. 2003, 348, 567–568. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spencer, C.N.; McQuade, J.L.; Gopalakrishnan, V.; McCulloch, J.A.; Vetizou, M.; Cogdill, A.P.; Khan, M.A.W.; Zhang, X.; White, M.G.; Peterson, C.B.; et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science 2021, 374, 1632–1640. [Google Scholar] [CrossRef] [PubMed]
- Lauby-Secretan, B.; Scoccianti, C.; Loomis, D.; Grosse, Y.; Bianchini, F.; Straif, K.; International Agency for Research on Cancer Handbook Working Group. Body fatness and cancer—Viewpoint of the IARC working group. N. Engl. J. Med. 2016, 375, 794–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sung, H.; Siegel, R.L.; Torre, L.A.; Pearson-Stuttard, J.; Islami, F.; Fedewa, S.A.; Goding Sauer, A.; Shuval, K.; Gapstur, S.M.; Jacobs, E.J.; et al. Global patterns in excess body weight and the associated cancer burden. CA Cancer J. Clin. 2019, 69, 88–112. [Google Scholar] [CrossRef]
- Ringel, A.E.; Drijvers, J.M.; Baker, G.J.; Catozzi, A.; Garcia-Canaveras, J.C.; Gassaway, B.M.; Miller, B.C.; Juneja, V.R.; Nguyen, T.H.; Joshi, S.; et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell 2020, 183, 1848–1866.e1826. [Google Scholar] [CrossRef]
- Kaymak, I.; Williams, K.S.; Cantor, J.R.; Jones, R.G. Immunometabolic interplay in the tumor microenvironment. Cancer Cell 2021, 39, 28–37. [Google Scholar] [CrossRef]
- Weigert, A.; Mora, J.; Sekar, D.; Syed, S.; Brune, B. Killing is not enough: How apoptosis hijacks tumor-associated macrophages to promote cancer progression. Adv. Exp. Med. Biol. 2016, 930, 205–239. [Google Scholar] [CrossRef]
- Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune Cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef]
- Fridman, W.H.; Zitvogel, L.; Sautes-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
- Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.; Turkowski, K.; Mora, J.; Brune, B.; Seeger, W.; Weigert, A.; Savai, R. Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2017, 8, 48436–48452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silvestre-Roig, C.; Hidalgo, A.; Soehnlein, O. Neutrophil heterogeneity: Implications for homeostasis and pathogenesis. Blood 2016, 127, 2173–2181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tazzyman, S.; Lewis, C.E.; Murdoch, C. Neutrophils: Key mediators of tumour angiogenesis. Int. J. Exp. Pathol. 2009, 90, 222–231. [Google Scholar] [CrossRef]
- Noonan, D.M.; De Lerma Barbaro, A.; Vannini, N.; Mortara, L.; Albini, A. Inflammation, inflammatory cells and angiogenesis: Decisions and indecisions. Cancer Metastasis Rev. 2008, 27, 31–40. [Google Scholar] [CrossRef]
- Menetrier-Caux, C.; Ray-Coquard, I.; Blay, J.Y.; Caux, C. Lymphopenia in cancer patients and its effects on response to immunotherapy: An opportunity for combination with Cytokines? J. Immunother. Cancer 2019, 7, 85. [Google Scholar] [CrossRef] [Green Version]
- Hiam-Galvez, K.J.; Allen, B.M.; Spitzer, M.H. Systemic immunity in cancer. Nat. Rev. Cancer 2021, 21, 345–359. [Google Scholar] [CrossRef] [PubMed]
- Spitzer, M.H.; Carmi, Y.; Reticker-Flynn, N.E.; Kwek, S.S.; Madhireddy, D.; Martins, M.M.; Gherardini, P.F.; Prestwood, T.R.; Chabon, J.; Bendall, S.C.; et al. Systemic immunity is required for effective cancer immunotherapy. Cell 2017, 168, 487–502.e415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strub, G.M.; Maceyka, M.; Hait, N.C.; Milstien, S.; Spiegel, S. Extracellular and intracellular actions of sphingosine-1-phosphate. Adv. Exp. Med. Biol. 2010, 688, 141–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendelson, K.; Evans, T.; Hla, T. Sphingosine 1-phosphate signalling. Development 2014, 141, 5–9. [Google Scholar] [CrossRef] [Green Version]
- Pyne, S.; Adams, D.R.; Pyne, N.J. Sphingosine 1-phosphate and sphingosine kinases in health and disease: Recent advances. Prog. Lipid Res. 2016, 62, 93–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pitman, M.R.; Costabile, M.; Pitson, S.M. Recent advances in the development of sphingosine kinase inhibitors. Cell. Signal. 2016, 28, 1349–1363. [Google Scholar] [CrossRef]
- Maceyka, M.; Harikumar, K.B.; Milstien, S.; Spiegel, S. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol. 2012, 22, 50–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weigert, A.; Olesch, C.; Brune, B. Sphingosine-1-Phosphate and macrophage biology-how the sphinx tames the big eater. Front. Immunol. 2019, 10, 1706. [Google Scholar] [CrossRef] [PubMed]
- Hannun, Y.A.; Obeid, L.M. Principles of bioactive lipid signalling: Lessons from sphingolipids. Nat. Rev. Mol. Cell Biol. 2008, 9, 139–150. [Google Scholar] [CrossRef]
- Merrill, A.H., Jr.; Stokes, T.H.; Momin, A.; Park, H.; Portz, B.J.; Kelly, S.; Wang, E.; Sullards, M.C.; Wang, M.D. Sphingolipidomics: A valuable tool for understanding the roles of sphingolipids in biology and disease. J. Lipid Res. 2009, 50, S97–S102. [Google Scholar] [CrossRef] [Green Version]
- Adams, D.R.; Pyne, S.; Pyne, N.J. Sphingosine kinases: Emerging structure-function insights. Trends Biochem. Sci. 2016, 41, 395–409. [Google Scholar] [CrossRef] [Green Version]
- Neubauer, H.A.; Pitson, S.M. Roles, regulation and inhibitors of sphingosine kinase 2. FEBS J. 2013, 280, 5317–5336. [Google Scholar] [CrossRef]
- Le Stunff, H.; Milstien, S.; Spiegel, S. Generation and metabolism of bioactive sphingosine-1-phosphate. J. Cell Biochem. 2004, 92, 882–899. [Google Scholar] [CrossRef]
- Spiegel, S.; Milstien, S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat. Rev. Immunol. 2011, 11, 403–415. [Google Scholar] [CrossRef]
- Alvarez, S.E.; Harikumar, K.B.; Hait, N.C.; Allegood, J.; Strub, G.M.; Kim, E.Y.; Maceyka, M.; Jiang, H.; Luo, C.; Kordula, T.; et al. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 2010, 465, 1084–1088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harikumar, K.B.; Yester, J.W.; Surace, M.J.; Oyeniran, C.; Price, M.M.; Huang, W.C.; Hait, N.C.; Allegood, J.C.; Yamada, A.; Kong, X.; et al. K63-linked polyubiquitination of transcription factor IRF1 is essential for IL-1-induced production of chemokines CXCL10 and CCL5. Nat. Immunol. 2014, 15, 231–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laviad, E.L.; Albee, L.; Pankova-Kholmyansky, I.; Epstein, S.; Park, H.; Merrill, A.H., Jr.; Futerman, A.H. Characterization of ceramide synthase 2: Tissue distribution, substrate specificity, and inhibition by sphingosine 1-phosphate. J. Biol. Chem. 2008, 283, 5677–5684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oertel, S.; Scholich, K.; Weigert, A.; Thomas, D.; Schmetzer, J.; Trautmann, S.; Wegner, M.S.; Radeke, H.H.; Filmann, N.; Brune, B.; et al. Ceramide synthase 2 deficiency aggravates AOM-DSS-induced colitis in mice: Role of colon barrier integrity. Cell Mol. Life Sci. 2017, 74, 3039–3055. [Google Scholar] [CrossRef]
- Barthelmes, J.; de Bazo, A.M.; Pewzner-Jung, Y.; Schmitz, K.; Mayer, C.A.; Foerch, C.; Eberle, M.; Tafferner, N.; Ferreiros, N.; Henke, M.; et al. Lack of ceramide synthase 2 suppresses the development of experimental autoimmune encephalomyelitis by impairing the migratory capacity of neutrophils. Brain Behav. Immun. 2015, 46, 280–292. [Google Scholar] [CrossRef]
- Hait, N.C.; Allegood, J.; Maceyka, M.; Strub, G.M.; Harikumar, K.B.; Singh, S.K.; Luo, C.; Marmorstein, R.; Kordula, T.; Milstien, S.; et al. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science 2009, 325, 1254–1257. [Google Scholar] [CrossRef] [Green Version]
- Hait, N.C.; Maiti, A.; Xu, P.; Qi, Q.; Kawaguchi, T.; Okano, M.; Takabe, K.; Yan, L.; Luo, C. Regulation of hypoxia-inducible factor functions in the nucleus by sphingosine-1-phosphate. FASEB J. 2020, 34, 4293–4310. [Google Scholar] [CrossRef]
- Panneer Selvam, S.; De Palma, R.M.; Oaks, J.J.; Oleinik, N.; Peterson, Y.K.; Stahelin, R.V.; Skordalakes, E.; Ponnusamy, S.; Garrett-Mayer, E.; Smith, C.D.; et al. Binding of the sphingolipid S1P to hTERT stabilizes telomerase at the nuclear periphery by allosterically mimicking protein phosphorylation. Sci. Signal. 2015, 8, ra58. [Google Scholar] [CrossRef] [Green Version]
- Kajimoto, T.; Caliman, A.D.; Tobias, I.S.; Okada, T.; Pilo, C.A.; Van, A.N.; Andrew McCammon, J.; Nakamura, S.I.; Newton, A.C. Activation of atypical protein kinase C by sphingosine 1-phosphate revealed by an aPKC-specific activity reporter. Sci. Signal. 2019, 12, eaat6662. [Google Scholar] [CrossRef] [Green Version]
- Rivera, J.; Proia, R.L.; Olivera, A. The alliance of sphingosine-1-phosphate and its receptors in immunity. Nat. Rev. Immunol. 2008, 8, 753–763. [Google Scholar] [CrossRef] [Green Version]
- Takabe, K.; Kim, R.H.; Allegood, J.C.; Mitra, P.; Ramachandran, S.; Nagahashi, M.; Harikumar, K.B.; Hait, N.C.; Milstien, S.; Spiegel, S. Estradiol induces export of sphingosine 1-phosphate from breast cancer cells via ABCC1 and ABCG2. J. Biol. Chem. 2010, 285, 10477–10486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawahara, A.; Nishi, T.; Hisano, Y.; Fukui, H.; Yamaguchi, A.; Mochizuki, N. The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science 2009, 323, 524–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nieuwenhuis, B.; Luth, A.; Chun, J.; Huwiler, A.; Pfeilschifter, J.; Schafer-Korting, M.; Kleuser, B. Involvement of the ABC-transporter ABCC1 and the sphingosine 1-phosphate receptor subtype S1P3 in the cytoprotection of human fibroblasts by the glucocorticoid dexamethasone. J. Mol. Med. 2009, 87, 645–657. [Google Scholar] [CrossRef] [PubMed]
- Mitra, P.; Oskeritzian, C.A.; Payne, S.G.; Beaven, M.A.; Milstien, S.; Spiegel, S. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc. Natl. Acad. Sci. USA 2006, 103, 16394–16399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vu, T.M.; Ishizu, A.N.; Foo, J.C.; Toh, X.R.; Zhang, F.; Whee, D.M.; Torta, F.; Cazenave-Gassiot, A.; Matsumura, T.; Kim, S.; et al. Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets. Nature 2017, 550, 524–528. [Google Scholar] [CrossRef]
- Blaho, V.A.; Galvani, S.; Engelbrecht, E.; Liu, C.; Swendeman, S.L.; Kono, M.; Proia, R.L.; Steinman, L.; Han, M.H.; Hla, T. HDL-bound sphingosine-1-phosphate restrains lymphopoiesis and neuroinflammation. Nature 2015, 523, 342–346. [Google Scholar] [CrossRef] [Green Version]
- Hla, T.; Venkataraman, K.; Michaud, J. The vascular S1P gradient-cellular sources and biological significance. Biochim. Biophys. Acta 2008, 1781, 477–482. [Google Scholar] [CrossRef] [Green Version]
- Blaho, V.A.; Hla, T. An update on the biology of sphingosine 1-phosphate receptors. J. Lipid Res. 2014, 55, 1596–1608. [Google Scholar] [CrossRef] [Green Version]
- Cartier, A.; Hla, T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019, 366, 6463. [Google Scholar] [CrossRef]
- Graler, M.H.; Bernhardt, G.; Lipp, M. EDG6, a novel G-protein-coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue. Genomics 1998, 53, 164–169. [Google Scholar] [CrossRef]
- Hansen, L.; Lohfink, N.; Vutukuri, R.; Kestner, R.I.; Trautmann, S.; Hecht, M.; Wagner, P.V.; Spitzer, D.; Khel, M.I.; Macas, J.; et al. Endothelial Sphingosine-1-Phosphate Receptor 4 Regulates Blood-Brain Barrier Permeability and Promotes a Homeostatic Endothelial Phenotype. J. Neurosci. 2021. Available online: https://www.jneurosci.org/content/early/2021/12/09/JNEUROSCI.0188-21.2021.abstract?casa_token=FGStSETX4xwAAAAA:LxUmWuElaRYiLREOkZ3agxJD-cEc0q3Lyzk8PDvBTRA11NKNLGX0XHlC-vFRbWAdr1zYjqyQqZBX1Y8 (accessed on 29 December 2021). [CrossRef] [PubMed]
- Walzer, T.; Chiossone, L.; Chaix, J.; Calver, A.; Carozzo, C.; Garrigue-Antar, L.; Jacques, Y.; Baratin, M.; Tomasello, E.; Vivier, E. Natural killer cell trafficking in vivo requires a dedicated sphingosine 1-phosphate receptor. Nat. Immunol. 2007, 8, 1337–1344. [Google Scholar] [CrossRef] [PubMed]
- Idzko, M.; Hammad, H.; van Nimwegen, M.; Kool, M.; Muller, T.; Soullie, T.; Willart, M.A.; Hijdra, D.; Hoogsteden, H.C.; Lambrecht, B.N. Local application of FTY720 to the lung abrogates experimental asthma by altering dendritic cell function. J. Clin. Investig. 2006, 116, 2935–2944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Terai, K.; Soga, T.; Takahashi, M.; Kamohara, M.; Ohno, K.; Yatsugi, S.; Okada, M.; Yamaguchi, T. Edg-8 receptors are preferentially expressed in oligodendrocyte lineage cells of the rat CNS. Neuroscience 2003, 116, 1053–1062. [Google Scholar] [CrossRef]
- van Doorn, R.; Lopes Pinheiro, M.A.; Kooij, G.; Lakeman, K.; van het Hof, B.; van der Pol, S.M.; Geerts, D.; van Horssen, J.; van der Valk, P.; van der Kam, E.; et al. Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier. J. Neuroinflamm. 2012, 9, 133. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.L.; Chang, C.L.; Tang, C.H.; Lin, Y.C.; Ju, T.K.; Huang, W.P.; Lee, H. Extrinsic sphingosine 1-phosphate activates S1P5 and induces autophagy through generating endoplasmic reticulum stress in human prostate cancer PC-3 cells. Cell. Signal. 2014, 26, 611–618. [Google Scholar] [CrossRef]
- Hu, W.M.; Li, L.; Jing, B.Q.; Zhao, Y.S.; Wang, C.L.; Feng, L.; Xie, Y.E. Effect of S1P5 on proliferation and migration of human esophageal cancer cells. World J. Gastroenterol. 2010, 16, 1859–1866. [Google Scholar] [CrossRef]
- Aoki, M.; Aoki, H.; Ramanathan, R.; Hait, N.C.; Takabe, K. Sphingosine-1-phosphate signaling in immune cells and inflammation: Roles and therapeutic potential. Mediat. Inflamm. 2016, 2016, 8606878. [Google Scholar] [CrossRef]
- Don-Doncow, N.; Zhang, Y.; Matuskova, H.; Meissner, A. The emerging alliance of sphingosine-1-phosphate signalling and immune cells: From basic mechanisms to implications in hypertension. Br. J. Pharmacol. 2019, 176, 1989–2001. [Google Scholar] [CrossRef]
- Kono, M.; Mi, Y.; Liu, Y.; Sasaki, T.; Allende, M.L.; Wu, Y.P.; Yamashita, T.; Proia, R.L. The sphingosine-1-phosphate receptors S1P1, S1P2, and S1P3 function coordinately during embryonic angiogenesis. J. Biol. Chem. 2004, 279, 29367–29373. [Google Scholar] [CrossRef] [Green Version]
- Mendelson, K.; Zygmunt, T.; Torres-Vazquez, J.; Evans, T.; Hla, T. Sphingosine 1-phosphate receptor signaling regulates proper embryonic vascular patterning. J. Biol. Chem. 2013, 288, 2143–2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mizugishi, K.; Yamashita, T.; Olivera, A.; Miller, G.F.; Spiegel, S.; Proia, R.L. Essential role for sphingosine kinases in neural and vascular development. Mol. Cell Biol. 2005, 25, 11113–11121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baeyens, A.A.L.; Schwab, S.R. Finding a way out: S1P signaling and immune cell migration. Annu. Rev. Immunol. 2020, 38, 759–784. [Google Scholar] [CrossRef] [PubMed]
- Pinschewer, D.D.; Ochsenbein, A.F.; Odermatt, B.; Brinkmann, V.; Hengartner, H.; Zinkernagel, R.M. FTY720 immunosuppression impairs effector T Cell peripheral homing without affecting induction, expansion, and memory. J. Immunol. 2000, 164, 5761–5770. [Google Scholar] [CrossRef] [Green Version]
- Mandala, S.; Hajdu, R.; Bergstrom, J.; Quackenbush, E.; Xie, J.; Milligan, J.; Thornton, R.; Shei, G.J.; Card, D.; Keohane, C.; et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 2002, 296, 346–349. [Google Scholar] [CrossRef]
- Matloubian, M.; Lo, C.G.; Cinamon, G.; Lesneski, M.J.; Xu, Y.; Brinkmann, V.; Allende, M.L.; Proia, R.L.; Cyster, J.G. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 2004, 427, 355–360. [Google Scholar] [CrossRef]
- Donoviel, M.S.; Hait, N.C.; Ramachandran, S.; Maceyka, M.; Takabe, K.; Milstien, S.; Oravecz, T.; Spiegel, S. Spinster 2, a sphingosine-1-phosphate transporter, plays a critical role in inflammatory and autoimmune diseases. FASEB J. 2015, 29, 5018–5028. [Google Scholar] [CrossRef] [Green Version]
- Skon, C.N.; Lee, J.Y.; Anderson, K.G.; Masopust, D.; Hogquist, K.A.; Jameson, S.C. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 2013, 14, 1285–1293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shiow, L.R.; Rosen, D.B.; Brdickova, N.; Xu, Y.; An, J.; Lanier, L.L.; Cyster, J.G.; Matloubian, M. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 2006, 440, 540–544. [Google Scholar] [CrossRef]
- Mackay, L.K.; Braun, A.; Macleod, B.L.; Collins, N.; Tebartz, C.; Bedoui, S.; Carbone, F.R.; Gebhardt, T. Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J. Immunol. 2015, 194, 2059–2063. [Google Scholar] [CrossRef] [Green Version]
- Keul, P.; Lucke, S.; von Wnuck Lipinski, K.; Bode, C.; Graler, M.; Heusch, G.; Levkau, B. Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circ. Res. 2011, 108, 314–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Debien, E.; Mayol, K.; Biajoux, V.; Daussy, C.; De Aguero, M.G.; Taillardet, M.; Dagany, N.; Brinza, L.; Henry, T.; Dubois, B.; et al. S1PR5 is pivotal for the homeostasis of patrolling monocytes. Eur. J. Immunol. 2013, 43, 1667–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cinamon, G.; Zachariah, M.A.; Lam, O.M.; Foss, F.W., Jr.; Cyster, J.G. Follicular shuttling of marginal zone B cells facilitates antigen transport. Nat. Immunol. 2008, 9, 54–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olesch, C.; Ringel, C.; Brune, B.; Weigert, A. Beyond immune cell migration: The emerging role of the sphingosine-1-phosphate receptor S1PR4 as a modulator of innate immune cell activation. Mediat. Inflamm. 2017, 2017, 6059203. [Google Scholar] [CrossRef] [PubMed]
- Czeloth, N.; Bernhardt, G.; Hofmann, F.; Genth, H.; Forster, R. Sphingosine-1-phosphate mediates migration of mature dendritic cells. J. Immunol. 2005, 175, 2960–2967. [Google Scholar] [CrossRef] [Green Version]
- Weichand, B.; Weis, N.; Weigert, A.; Grossmann, N.; Levkau, B.; Brune, B. Apoptotic cells enhance sphingosine-1-phosphate receptor 1 dependent macrophage migration. Eur. J. Immunol. 2013, 43, 3306–3313. [Google Scholar] [CrossRef]
- Weigert, A.; Weichand, B.; Brune, B. S1P regulation of macrophage functions in the context of cancer. Anticancer Agents Med. Chem. 2011, 11, 818–829. [Google Scholar] [CrossRef]
- Pyne, N.J.; Pyne, S. Sphingosine 1-phosphate and cancer. Nat. Rev. Cancer 2010, 10, 489–503. [Google Scholar] [CrossRef] [Green Version]
- Kunkel, G.T.; Maceyka, M.; Milstien, S.; Spiegel, S. Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond. Nat. Rev. Drug Discov. 2013, 12, 688–702. [Google Scholar] [CrossRef] [Green Version]
- Weigert, A.; Cremer, S.; Schmidt, M.V.; von Knethen, A.; Angioni, C.; Geisslinger, G.; Brune, B. Cleavage of sphingosine kinase 2 by caspase-1 provokes its release from apoptotic cells. Blood 2010, 115, 3531–3540. [Google Scholar] [CrossRef] [Green Version]
- Cuvillier, O.; Ader, I.; Bouquerel, P.; Brizuela, L.; Gstalder, C.; Malavaud, B. Hypoxia, therapeutic resistance, and sphingosine 1-phosphate. Adv. Cancer Res. 2013, 117, 117–141. [Google Scholar] [CrossRef] [PubMed]
- Schneider, G. S1P Signaling in the tumor microenvironment. Adv. Exp. Med. Biol. 2020, 1223, 129–153. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef] [PubMed]
- Dickson, M.A.; Carvajal, R.D.; Merrill, A.H., Jr.; Gonen, M.; Cane, L.M.; Schwartz, G.K. A phase I clinical trial of safingol in combination with cisplatin in advanced solid tumors. Clin. Cancer Res. 2011, 17, 2484–2492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Britten, C.D.; Garrett-Mayer, E.; Chin, S.H.; Shirai, K.; Ogretmen, B.; Bentz, T.A.; Brisendine, A.; Anderton, K.; Cusack, S.L.; Maines, L.W.; et al. A phase I study of ABC294640, a first-in-class sphingosine kinase-2 Inhibitor, in patients with advanced solid tumors. Clin. Cancer Res. 2017, 23, 4642–4650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pal, S.K.; Drabkin, H.A.; Reeves, J.A.; Hainsworth, J.D.; Hazel, S.E.; Paggiarino, D.A.; Wojciak, J.; Woodnutt, G.; Bhatt, R.S. A phase 2 study of the sphingosine-1-phosphate antibody sonepcizumab in patients with metastatic renal cell carcinoma. Cancer 2017, 123, 576–582. [Google Scholar] [CrossRef] [PubMed]
- Visentin, B.; Vekich, J.A.; Sibbald, B.J.; Cavalli, A.L.; Moreno, K.M.; Matteo, R.G.; Garland, W.A.; Lu, Y.; Yu, S.; Hall, H.S.; et al. Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell 2006, 9, 225–238. [Google Scholar] [CrossRef] [Green Version]
- Cartier, A.; Leigh, T.; Liu, C.H.; Hla, T. Endothelial sphingosine 1-phosphate receptors promote vascular normalization and antitumor therapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3157–3166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murdoch, C.; Muthana, M.; Coffelt, S.B.; Lewis, C.E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 2008, 8, 618–631. [Google Scholar] [CrossRef]
- Gazit, S.L.; Mariko, B.; Therond, P.; Decouture, B.; Xiong, Y.; Couty, L.; Bonnin, P.; Baudrie, V.; Le Gall, S.M.; Dizier, B.; et al. Platelet and erythrocyte sources of s1p are redundant for vascular development and homeostasis, but both rendered essential after plasma S1P depletion in anaphylactic shock. Circ. Res. 2016, 119, e110–e126. [Google Scholar] [CrossRef]
- Kawamori, T.; Kaneshiro, T.; Okumura, M.; Maalouf, S.; Uflacker, A.; Bielawski, J.; Hannun, Y.A.; Obeid, L.M. Role for sphingosine kinase 1 in colon carcinogenesis. FASEB J. 2009, 23, 405–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, J.; Nagahashi, M.; Kim, E.Y.; Harikumar, K.B.; Yamada, A.; Huang, W.C.; Hait, N.C.; Allegood, J.C.; Price, M.M.; Avni, D.; et al. Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer. Cancer Cell 2013, 23, 107–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuza, K.; Nagahashi, M.; Shimada, Y.; Nakano, M.; Tajima, Y.; Kameyama, H.; Nakajima, M.; Takabe, K.; Wakai, T. Upregulation of phosphorylated sphingosine kinase 1 expression in colitis-associated cancer. J. Surg. Res. 2018, 231, 323–330. [Google Scholar] [CrossRef] [PubMed]
- Chumanevich, A.A.; Poudyal, D.; Cui, X.; Davis, T.; Wood, P.A.; Smith, C.D.; Hofseth, L.J. Suppression of colitis-driven colon cancer in mice by a novel small molecule inhibitor of sphingosine kinase. Carcinogenesis 2010, 31, 1787–1793. [Google Scholar] [CrossRef] [Green Version]
- De Henau, O.; Rausch, M.; Winkler, D.; Campesato, L.F.; Liu, C.; Cymerman, D.H.; Budhu, S.; Ghosh, A.; Pink, M.; Tchaicha, J.; et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kgamma in myeloid Cells. Nature 2016, 539, 443–447. [Google Scholar] [CrossRef] [Green Version]
- Weigert, A.; Schiffmann, S.; Sekar, D.; Ley, S.; Menrad, H.; Werno, C.; Grosch, S.; Geisslinger, G.; Brune, B. Sphingosine kinase 2 deficient tumor xenografts show impaired growth and fail to polarize macrophages towards an anti-inflammatory phenotype. Int. J. Cancer 2009, 125, 2114–2121. [Google Scholar] [CrossRef]
- Tsuchida, J.; Nagahashi, M.; Nakajima, M.; Katsuta, E.; Rashid, O.M.; Qi, Q.; Yan, L.; Okuda, S.; Takabe, K.; Wakai, T. Sphingosine kinase 1 is associated with immune cell-related gene expressions in human breast cancer. J. Surg. Res. 2020, 256, 645–656. [Google Scholar] [CrossRef]
- Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T cells exhibit distinct features in human breast cancer. Immunity 2016, 45, 1122–1134. [Google Scholar] [CrossRef] [Green Version]
- Strack, E.; Rolfe, P.A.; Fink, A.F.; Bankov, K.; Schmid, T.; Solbach, C.; Savai, R.; Sha, W.; Pradel, L.; Hartmann, S.; et al. Identification of tumor-associated macrophage subsets that are associated with breast cancer prognosis. Clin. Transl. Med. 2020, 10, e239. [Google Scholar] [CrossRef]
- Imbert, C.; Montfort, A.; Fraisse, M.; Marcheteau, E.; Gilhodes, J.; Martin, E.; Bertrand, F.; Marcellin, M.; Burlet-Schiltz, O.; Peredo, A.G.; et al. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nat. Commun. 2020, 11, 437. [Google Scholar] [CrossRef]
- Mrad, M.; Imbert, C.; Garcia, V.; Rambow, F.; Therville, N.; Carpentier, S.; Segui, B.; Levade, T.; Azar, R.; Marine, J.C.; et al. Downregulation of sphingosine kinase-1 induces protective tumor immunity by promoting M1 macrophage response in melanoma. Oncotarget 2016, 7, 71873–71886. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.S.; Sun, W.; Webb, T.J. Sphingosine kinase blockade leads to increased natural killer T cell responses to mantle cell Lymphoma. Cells 2020, 9, 1030. [Google Scholar] [CrossRef] [Green Version]
- Chakraborty, P.; Vaena, S.G.; Thyagarajan, K.; Chatterjee, S.; Al-Khami, A.; Selvam, S.P.; Nguyen, H.; Kang, I.; Wyatt, M.W.; Baliga, U.; et al. Pro-Survival lipid sphingosine-1-phosphate metabolically programs t cells to limit anti-tumor activity. Cell. Rep. 2019, 28, 1879–1893.e1877. [Google Scholar] [CrossRef] [Green Version]
- Degagne, E.; Pandurangan, A.; Bandhuvula, P.; Kumar, A.; Eltanawy, A.; Zhang, M.; Yoshinaga, Y.; Nefedov, M.; de Jong, P.J.; Fong, L.G.; et al. Sphingosine-1-phosphate lyase downregulation promotes colon carcinogenesis through STAT3-activated microRNAs. J. Clin. Investig. 2014, 124, 5368–5384. [Google Scholar] [CrossRef] [Green Version]
- Schwiebs, A.; Herrero, S.J.M.; Schmidt, K.G.; Wiercinska, E.; Anlauf, M.; Ottenlinger, F.; Thomas, D.; Elwakeel, E.; Weigert, A.; Farin, H.F.; et al. Cancer-induced inflammation and inflammation-induced cancer in colon: A role for S1P lyase. Oncogene 2019, 38, 4788–4803. [Google Scholar] [CrossRef]
- Nema, R.; Kumar, A. Sphingosine-1-phosphate catabolizing enzymes predict better prognosis in triple-negative breast cancer patients and correlates with tumor-infiltrating immune cells. Front. Mol. Biosci. 2021, 8, 697922. [Google Scholar] [CrossRef]
- Nema, R.; Shrivastava, A.; Kumar, A. Prognostic role of lipid phosphate phosphatases in non-smoker, lung adenocarcinoma patients. Comput. Biol. Med. 2021, 129, 104141. [Google Scholar] [CrossRef]
- Satyananda, V.; Oshi, M.; Tokumaru, Y.; Maiti, A.; Hait, N.; Matsuyama, R.; Endo, I.; Takabe, K. Sphingosine 1-phosphate (S1P) produced by sphingosine kinase 1 (SphK1) and exported via ABCC1 is related to hepatocellular carcinoma (HCC) progression. Am. J. Cancer Res. 2021, 11, 4394–4407. [Google Scholar]
- Nijnik, A.; Clare, S.; Hale, C.; Chen, J.; Raisen, C.; Mottram, L.; Lucas, M.; Estabel, J.; Ryder, E.; Adissu, H.; et al. The role of sphingosine-1-phosphate transporter Spns2 in immune system function. J. Immunol. 2012, 189, 102–111. [Google Scholar] [CrossRef] [Green Version]
- van der Weyden, L.; Arends, M.J.; Campbell, A.D.; Bald, T.; Wardle-Jones, H.; Griggs, N.; Velasco-Herrera, M.D.; Tuting, T.; Sansom, O.J.; Karp, N.A.; et al. Genome-wide in vivo screen identifies novel host regulators of metastatic colonization. Nature 2017, 541, 233–236. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Tang, Y.; Wang, D.; Zhai, X.; Shen, H.; Zhong, C.; Yao, M.; Jin, A.; Zhou, Z.; Zhou, S.; et al. Sphingosine-1-phosphate transporter spinster homolog 2 is essential for iron-regulated metastasis of hepato cellular carcinoma. Mol. Ther. 2021. [Google Scholar] [CrossRef]
- Lv, L.; Yi, Q.; Yan, Y.; Chao, F.; Li, M. SPNS2 downregulation induces emt and promotes colorectal cancer metastasis via activating akt signaling pathway. Front. Oncol. 2021, 11, 682773. [Google Scholar] [CrossRef]
- Dhodapkar, K.M. Role of tissue-resident memory in intra-tumor heterogeneity and response to immune checkpoint blockade. Front. Immun. 2018, 9, 1655. [Google Scholar] [CrossRef] [Green Version]
- Wei, J.; Ishizuka, J.J. Going viral: HBV-specific CD8(+) tissue-resident memory T cells propagate anti-tumor immunity. Immunity 2021, 54, 1630–1632. [Google Scholar] [CrossRef]
- Priceman, S.J.; Shen, S.; Wang, L.; Deng, J.; Yue, C.; Kujawski, M.; Yu, H. S1PR1 is crucial for accumulation of regulatory T cells in tumors via STAT3. Cell Rep. 2014, 6, 992–999. [Google Scholar] [CrossRef] [Green Version]
- Rathinasamy, A.; Domschke, C.; Ge, Y.; Böhm, H.H.; Dettling, S.; Jansen, D.; Lasitschka, F.; Umansky, L.; Gräler, M.H.; Hartmann, J.; et al. Tumor specific regulatory T cells in the bone marrow of breast cancer patients selectively upregulate the emigration receptor S1P1. Cancer Immunol. Immunother. 2017, 66, 593–603. [Google Scholar] [CrossRef] [Green Version]
- Chongsathidkiet, P.; Jackson, C.; Koyama, S.; Loebel, F.; Cui, X.; Farber, S.H.; Woroniecka, K.; Elsamadicy, A.A.; Dechant, C.A.; Kemeny, H.R.; et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat. Med. 2018, 24, 1459–1468. [Google Scholar] [CrossRef]
- Cheng, F.; Wang, H.W.; Cuenca, A.; Huang, M.; Ghansah, T.; Brayer, J.; Kerr, W.G.; Takeda, K.; Akira, S.; Schoenberger, S.P.; et al. A critical role for Stat3 signaling in immune tolerance. Immunity 2003, 19, 425–436. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Deng, J.; Kujawski, M.; Yang, C.; Liu, Y.; Herrmann, A.; Kortylewski, M.; Horne, D.; Somlo, G.; Forman, S.; et al. STAT3-induced S1PR1 expression is crucial for persistent STAT3 activation in tumors. Nat. Med. 2010, 16, 1421–1428. [Google Scholar] [CrossRef] [Green Version]
- Deng, J.; Liu, Y.; Lee, H.; Herrmann, A.; Zhang, W.; Zhang, C.; Shen, S.; Priceman, S.J.; Kujawski, M.; Pal, S.K.; et al. S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites. Cancer Cell 2012, 21, 642–654. [Google Scholar] [CrossRef] [Green Version]
- Nagahashi, M.; Yamada, A.; Katsuta, E.; Aoyagi, T.; Huang, W.C.; Terracina, K.P.; Hait, N.C.; Allegood, J.C.; Tsuchida, J.; Yuza, K.; et al. Targeting the SphK1/S1P/S1PR1 axis that links obesity, chronic inflammation, and breast cancer metastasis. Cancer Res. 2018, 78, 1713–1725. [Google Scholar] [CrossRef] [Green Version]
- Weichand, B.; Popp, R.; Dziumbla, S.; Mora, J.; Strack, E.; Elwakeel, E.; Frank, A.C.; Scholich, K.; Pierre, S.; Syed, S.N.; et al. S1PR1 on tumor-associated macrophages promotes lymphangiogenesis and metastasis via NLRP3/IL-1β. J. Exp. Med. 2017, 214, 2695–2713. [Google Scholar] [CrossRef]
- Hughes, J.E.; Srinivasan, S.; Lynch, K.R.; Proia, R.L.; Ferdek, P.; Hedrick, C.C. Sphingosine-1-phosphate induces an antiinflammatory phenotype in macrophages. Circ. Res. 2008, 102, 950–958. [Google Scholar] [CrossRef]
- Rodriguez, Y.I.; Campos, L.E.; Castro, M.G.; Aladhami, A.; Oskeritzian, C.A.; Alvarez, S.E. Sphingosine-1 phosphate: A new modulator of immune plasticity in the tumor microenvironment. Front. Oncol. 2016, 6, 218. [Google Scholar] [CrossRef] [Green Version]
- Obinata, H.; Hla, T. Sphingosine 1-phosphate and inflammation. Int. Immunol. 2019, 31, 617–625. [Google Scholar] [CrossRef]
- Human Protein Atlas. 2021. Available online: https://www.proteinatlas.org/ (accessed on 29 December 2021).
- Adada, M.M.; Canals, D.; Jeong, N.; Kelkar, A.D.; Hernandez-Corbacho, M.; Pulkoski-Gross, M.J.; Donaldson, J.C.; Hannun, Y.A.; Obeid, L.M. Intracellular sphingosine kinase 2-derived sphingosine-1-phosphate mediates epidermal growth factor-induced ezrin-radixin-moesin phosphorylation and cancer cell invasion. FASEB J. 2015, 29, 4654–4669. [Google Scholar] [CrossRef] [Green Version]
- Petti, L.; Piontini, A.; Arena, V.; Danese, S.; Vetrano, S. Sphingosine-1-phosphate receptor 2 is a negative regulator of epithelial cell proliferation and intestinal tumorigenesis. FASEB J. 2017, 31, 1046.2. [Google Scholar] [CrossRef]
- Du, W.; Takuwa, N.; Yoshioka, K.; Okamoto, Y.; Gonda, K.; Sugihara, K.; Fukamizu, A.; Asano, M.; Takuwa, Y. S1P2, the G protein-coupled receptor for sphingosine-1-phosphate, negatively regulates tumor angiogenesis and tumor growth in vivo in mice. Cancer Res. 2010, 70, 772–781. [Google Scholar] [CrossRef] [Green Version]
- Cattoretti, G.; Mandelbaum, J.; Lee, N.; Chaves, A.H.; Mahler, A.M.; Chadburn, A.; Dalla-Favera, R.; Pasqualucci, L.; MacLennan, A.J. Targeted disruption of the S1P2 sphingosine 1-phosphate receptor gene leads to diffuse large B-cell lymphoma formation. Cancer Res. 2009, 69, 8686–8692. [Google Scholar] [CrossRef] [Green Version]
- Terlizzi, M.; Colarusso, C.; Ferraro, G.; Monti, M.C.; Rosa, I.D.; Somma, P.; Salvi, R.; Pinto, A.; Sorrentino, R. Intracellular Sphingosine-1-phosphate receptor 3 contributes to lung tumor cell proliferation. Cell Physiol. Biochem. 2021, 55, 539–552. [Google Scholar] [CrossRef]
- Wang, H.; Cai, K.-Y.; Li, W.; Huang, H. Sphingosine-1-phosphate induces the migration and angiogenesis of EPCs through the Akt signaling pathway via sphingosine-1-phosphate receptor 3/platelet-derived growth factor receptor-β. Cell. Mol. Biol. Lett. 2015, 20, 597–611. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Liang, Y.; Chang, W.; Hu, B.; Zhang, Y. Triple Negative Breast Cancer Depends on Sphingosine Kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/sphingosine 1-phosphate receptor 3 (S1PR3)/notch signaling for metastasis. Med. Sci. Monit. J. Exp. Clin. Cancer Res. 2018, 24, 1912–1923. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.; Liu, J.; Lee, J.F.; Zhang, W.; Kandouz, M.; VanHecke, G.C.; Chen, S.; Ahn, Y.H.; Lonardo, F.; Lee, M.J. TGF-β/SMAD3 pathway stimulates sphingosine-1 phosphate receptor 3 expression: Implication of sphingosinE-1 phosphate receptor 3 in lung adenocarcinoma progression. J. Biol. Chem. 2016, 291, 27343–27353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bajwa, A.; Huang, L.; Ye, H.; Dondeti, K.; Song, S.; Rosin, D.L.; Lynch, K.R.; Lobo, P.I.; Li, L.; Okusa, M.D. Dendritic cell Sphingosine 1-phosphate receptor-3 regulates Th1–Th2 polarity in kidney ischemia–reperfusion injury. J. Immunol. 2012, 189, 2584–2596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niessen, F.; Schaffner, F.; Furlan-Freguia, C.; Pawlinski, R.; Bhattacharjee, G.; Chun, J.; Derian, C.K.; Andrade-Gordon, P.; Rosen, H.; Ruf, W. Dendritic cell PAR1-S1P3 signalling couples coagulation and inflammation. Nature 2008, 452, 654–658. [Google Scholar] [CrossRef]
- Xie, S.Z.; Kaufmann, K.B.; Wang, W.; Chan-Seng-Yue, M.; Gan, O.I.; Laurenti, E.; Garcia-Prat, L.; Takayanagi, S.I.; Ng, S.W.K.; Xu, C.; et al. Sphingosine-1-phosphate receptor 3 potentiates inflammatory programs in normal and leukemia stem cells to promote differentiation. Blood Cancer Discov. 2021, 2, 32–53. [Google Scholar] [CrossRef]
- Bien-Möller, S.; Lange, S.; Holm, T.; Böhm, A.; Paland, H.; Küpper, J.; Herzog, S.; Weitmann, K.; Havemann, C.; Vogelgesang, S.; et al. Expression of S1P metabolizing enzymes and receptors correlate with survival time and regulate cell migration in glioblastoma multiforme. Oncotarget 2016, 7, 13031–13046. [Google Scholar] [CrossRef] [Green Version]
- Meshcheryakova, A.; Svoboda, M.; Jaritz, M.; Mungenast, F.; Salzmann, M.; Pils, D.; Cacsire, C.-T.D.; Hager, G.; Wolf, A.; Braicu, E.I.; et al. Interrelations of sphingolipid and lysophosphatidate signaling with immune system in ovarian cancer. Comput. Struct. Biotechnol. J. 2019, 17, 537–560. [Google Scholar] [CrossRef]
- Xiong, Y.; Piao, W.; Brinkman, C.C.; Li, L.; Kulinski, J.M.; Olivera, A.; Cartier, A.; Hla, T.; Hippen, K.L.; Blazar, B.R.; et al. CD4 T cell sphingosine 1-phosphate receptor (S1PR)1 and S1PR4 and endothelial S1PR2 regulate afferent lymphatic migration. Sci. Immunol. 2019, 4, MAR 2019. [Google Scholar] [CrossRef]
- Wang, W.; Graeler, M.H.; Goetzl, E.J. Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration. Faseb. J. 2005, 19, 1731–1733. [Google Scholar] [CrossRef]
- Sekar, D.; Hahn, C.; Brüne, B.; Roberts, E.; Weigert, A. Apoptotic tumor cells induce IL-27 release from human DCs to activate Treg cells that express CD69 and attenuate cytotoxicity. Eur. J. Immunol. 2012, 42, 1585–1598. [Google Scholar] [CrossRef] [Green Version]
- Dillmann, C.; Ringel, C.; Ringleb, J.; Mora, J.; Olesch, C.; Fink, A.F.; Roberts, E.; Brüne, B.; Weigert, A. S1PR4 signaling attenuates ILT 7 internalization to limit IFN-α production by human plasmacytoid dendritic cells. J. Immunol. 2016, 196, 1579–1590. [Google Scholar] [CrossRef] [Green Version]
- Ley, S.; Weigert, A.; Weichand, B.; Henke, N.; Mille-Baker, B.; Janssen, R.A.; Brüne, B. The role of TRKA signaling in IL-10 production by apoptotic tumor cell-activated macrophages. Oncogene 2013, 32, 631–640. [Google Scholar] [CrossRef] [Green Version]
- Olesch, C.; Sirait-Fischer, E.; Berkefeld, M.; Fink, A.F.; Susen, R.M.; Ritter, B.; Michels, B.E.; Steinhilber, D.; Greten, F.R.; Savai, R.; et al. S1PR4 ablation reduces tumor growth and improves chemotherapy via CD8+ T cell expansion. J. Clin. Investig. 2020, 130, 5461–5476. [Google Scholar] [CrossRef]
- Burkard, T.; Dreis, C.; Herrero San Juan, M.; Huhn, M.; Weigert, A.; Pfeilschifter, J.M.; Radeke, H.H. Enhanced CXCR4 expression of human CD8Low T lymphocytes is driven by S1P4. Front. Immunol. 2021, 12, 668884. [Google Scholar] [CrossRef]
- D’Alterio, C.; Buoncervello, M.; Ieranò, C.; Napolitano, M.; Portella, L.; Rea, G.; Barbieri, A.; Luciano, A.; Scognamiglio, G.; Tatangelo, F.; et al. Targeting CXCR4 potentiates anti-PD-1 efficacy modifying the tumor microenvironment and inhibiting neoplastic PD-1. J. Exp. Clin. Cancer Res. 2019, 38, 432. [Google Scholar] [CrossRef] [Green Version]
- Jaafar, F.; Righi, E.; Lindstrom, V.; Linton, C.; Nohadani, M.; Van Noorden, S.; Lloyd, T.; Poznansky, J.; Stamp, G.; Dina, R.; et al. Correlation of CXCL12 expression and FoxP3+ cell infiltration with human papillomavirus infection and clinicopathological progression of cervical cancer. Am. J. Pathol. 2009, 175, 1525–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Simone, V.; Pallone, F.; Monteleone, G.; Stolfi, C. Role of T(H)17 cytokines in the control of colorectal cancer. Oncoimmunology 2013, 2, e26617. [Google Scholar] [CrossRef] [Green Version]
- Schulze, T.; Golfier, S.; Tabeling, C.; Räbel, K.; Gräler, M.H.; Witzenrath, M.; Lipp, M. Sphingosine-1-phospate receptor 4 (S1P4) deficiency profoundly affects dendritic cell function and TH17-cell differentiation in a murine model. FASEB J. 2011, 25, 4024–4036. [Google Scholar] [CrossRef]
- Jaillard, C.; Harrison, S.; Stankoff, B.; Aigrot, M.S.; Calver, A.R.; Duddy, G.; Walsh, F.S.; Pangalos, M.N.; Arimura, N.; Kaibuchi, K.; et al. Edg8/S1P5: An oligodendroglial receptor with dual function on process retraction and cell survival. J. Neurosci. 2005, 25, 1459–1469. [Google Scholar] [CrossRef]
- Mayol, K.; Biajoux, V.; Marvel, J.; Balabanian, K.; Walzer, T. Sequential desensitization of CXCR4 and S1P5 controls natural killer cell trafficking. Blood 2011, 118, 4863–4871. [Google Scholar] [CrossRef] [PubMed]
- Ren, X.; Zhang, Z. Understanding tumor-infiltrating lymphocytes by single cell RNA sequencing. Adv. Immunol. 2019, 144, 217–245. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Li, Q.; Lin, L.; Shou, P.; Keli, L.; Xue, Y.; Hu, M.; Ling, W.; Huang, Y.; Du, L.; et al. MHC Class Ia empowers MHC class Ib-restricted CD8+ T cells with strong tumoricidal capacity. 2021. Available online: https://assets.researchsquare.com/files/rs-1036514/v1/8bc679c1-f19b-4274-b36d-dbda76d11d58.pdf?c=1638813979 (accessed on 29 December 2021). [CrossRef]
- Cheng, Y.; Gunasegaran, B.; Singh, H.D.; Dutertre, C.A.; Loh, C.Y.; Lim, J.Q.; Crawford, J.C.; Lee, H.K.; Zhang, X.; Lee, B.; et al. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity 2021, 54, 1825–1840.e1827. [Google Scholar] [CrossRef] [PubMed]
- Quint, K.; Stiel, N.; Neureiter, D.; Schlicker, H.U.; Nimsky, C.; Ocker, M.; Strik, H.; Kolodziej, M.A. The role of sphingosine kinase isoforms and receptors S1P1, S1P2, S1P3, and S1P5 in primary, secondary, and recurrent glioblastomas. Tumor Biol. 2014, 35, 8979–8989. [Google Scholar] [CrossRef] [PubMed]
- Vasquez, J.C.; Huttner, A.; Zhang, L.; Marks, A.; Chan, A.; Baehring, J.M.; Kahle, K.T.; Dhodapkar, K.M. SOX2 immunity and tissue resident memory in children and young adults with glioma. J. Neuro-Oncol. 2017, 134, 41–53. [Google Scholar] [CrossRef] [PubMed]
- Evrard, M.; Wynne-Jones, E.; Peng, C.; Kato, Y.; Christo, S.N.; Fonseca, R.; Park, S.L.; Burn, T.N.; Osman, M.; Devi, S.; et al. Sphingosine 1-phosphate receptor 5 (S1PR5) regulates the peripheral retention of tissue-resident lymphocytes. J. Exp. Med. 2022, 219, 219. [Google Scholar] [CrossRef]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Willems, J.J.; Arnold, B.P.; Gregory, C.D. Sinister self-sacrifice: The contribution of apoptosis to malignancy. Front. Immunol. 2014, 5, 299. [Google Scholar] [CrossRef] [Green Version]
- Luo, B.; Gan, W.; Liu, Z.; Shen, Z.; Wang, J.; Shi, R.; Liu, Y.; Liu, Y.; Jiang, M.; Zhang, Z.; et al. Erythropoeitin signaling in macrophages promotes dying cell clearance and immune tolerance. Immunity 2016, 44, 287–302. [Google Scholar] [CrossRef] [Green Version]
- Lauber, K.; Ernst, A.; Orth, M.; Herrmann, M.; Belka, C. Dying cell clearance and its impact on the outcome of tumor radiotherapy. Front. Oncol. 2012, 2, 116. [Google Scholar] [CrossRef] [Green Version]
- Kuraishy, A.; Karin, M.; Grivennikov, S.I. Tumor promotion via injury- and death-induced inflammation. Immunity 2011, 35, 467–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baeyens, A.; Fang, V.; Chen, C.; Schwab, S.R. Exit strategies: S1P signaling and T cell migration. Trends Immunol. 2015, 36, 778–787. [Google Scholar] [CrossRef] [Green Version]
- Bryan, A.M.; Del Poeta, M. Sphingosine-1-phosphate receptors and innate immunity. Cell Microbiol. 2018, 20, e12836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ray-Coquard, I.; Cropet, C.; Van Glabbeke, M.; Sebban, C.; Le Cesne, A.; Judson, I.; Tredan, O.; Verweij, J.; Biron, P.; Labidi, I.; et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009, 69, 5383–5391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, S.L.; Buzzai, A.; Rautela, J.; Hor, J.L.; Hochheiser, K.; Effern, M.; McBain, N.; Wagner, T.; Edwards, J.; McConville, R.; et al. Tissue-resident memory CD8(+) T cells promote melanoma-immune equilibrium in skin. Nature 2019, 565, 366–371. [Google Scholar] [CrossRef]
- Baeyens, A.; Bracero, S.; Chaluvadi, V.S.; Khodadadi-Jamayran, A.; Cammer, M.; Schwab, S.R. Monocyte-derived S1P in the lymph node regulates immune responses. Nature 2021, 592, 290–295. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Olesch, C.; Brüne, B.; Weigert, A. Keep a Little Fire Burning—The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity. Int. J. Mol. Sci. 2022, 23, 1289. https://doi.org/10.3390/ijms23031289
Olesch C, Brüne B, Weigert A. Keep a Little Fire Burning—The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity. International Journal of Molecular Sciences. 2022; 23(3):1289. https://doi.org/10.3390/ijms23031289
Chicago/Turabian StyleOlesch, Catherine, Bernhard Brüne, and Andreas Weigert. 2022. "Keep a Little Fire Burning—The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity" International Journal of Molecular Sciences 23, no. 3: 1289. https://doi.org/10.3390/ijms23031289
APA StyleOlesch, C., Brüne, B., & Weigert, A. (2022). Keep a Little Fire Burning—The Delicate Balance of Targeting Sphingosine-1-Phosphate in Cancer Immunity. International Journal of Molecular Sciences, 23(3), 1289. https://doi.org/10.3390/ijms23031289