Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The sphingosine-1-phosphate receptor 1 mediates the atheroprotective effect of eicosapentaenoic acid

Abstract

Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) have been associated with potential cardiovascular benefits, partly attributed to their bioactive metabolites. However, the underlying mechanisms responsible for these advantages are not fully understood. We previously reported that metabolites of the cytochrome P450 pathway derived from eicosapentaenoic acid (EPA) mediated the atheroprotective effect of ω-3 PUFAs. Here, we show that 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and its receptor, sphingosine-1-phosphate receptor 1 (S1PR1), in endothelial cells (ECs) can inhibit oscillatory shear stress- or tumor necrosis factor-α-induced endothelial activation in cultured human ECs. Notably, the atheroprotective effect of 17,18-EEQ and purified EPA is circumvented in male mice with endothelial S1PR1 deficiency. Mechanistically, the anti-inflammatory effect of 17,18-EEQ relies on calcium release-mediated endothelial nitric oxide synthase (eNOS) activation, which is abolished upon inhibition of S1PR1 or Gq signaling. Furthermore, 17,18-EEQ allosterically regulates the conformation of S1PR1 through a polar interaction with Lys34Nter. Finally, we show that Vascepa, a prescription drug containing highly purified and stable EPA ethyl ester, exerts its cardiovascular protective effect through the 17,18-EEQ–S1PR1 pathway in male and female mice. Collectively, our findings indicate that the anti-inflammatory effect of 17,18-EEQ involves the activation of the S1PR1–Gq–Ca2+–eNOS axis in ECs, offering a potential therapeutic target against atherosclerosis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: 17,18-EEQ inhibits endothelial activation through S1PR1.
Fig. 2: 17,18-EEQ is a ligand of S1PR1.
Fig. 3: 17,18-EEQ inhibits endothelial activation through S1PR1-Gq-Ca2+ signaling.
Fig. 4: 17,18-EEQ inhibits endothelial activation through S1PR1–Gq–Ca2+–eNOS signaling in vitro and in vivo.
Fig. 5: Lys34Nter is involved in 17,18-EEQ-induced S1PR1 activation.
Fig. 6: 17,18-EEQ protects against endothelial activation in vivo.
Fig. 7: S1PR1 is involved in Vascepa-mediated restrainment of endothelial activation in vivo.
Fig. 8: The ameliorative impact of Vascepa on the reduction of aortic atherosclerosis is mediated by endothelial S1PR1.

Similar content being viewed by others

Data availability

Raw data and source data in figures, extended data and tables are provided in the source data file. RNA sequencing data has been deposited in the Gene Expression Omnibus under accession number GSE259394. The details of commercial reagents, antibodies and primer sequences are provided with the supplementary data. Source data are provided with this paper.

References

  1. Budoff, M. J. et al. Effect of icosapent ethyl on progression of coronary atherosclerosis in patients with elevated triglycerides on statin therapy: final results of the EVAPORATE trial. Eur. Heart J. 41, 3925–3932 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Pischon, T. et al. Habitual dietary intake of n-3 and n-6 fatty acids in relation to inflammatory markers among US men and women. Circulation 108, 155–160 (2003).

    Article  PubMed  CAS  Google Scholar 

  3. Erlandson, S. C., McMahon, C. & Kruse, A. C. Structural basis for G protein-coupled receptor signaling. Annu. Rev. Biophys. 47, 1–18 (2018).

    Article  PubMed  CAS  Google Scholar 

  4. Smith, J. S., Lefkowitz, R. J. & Rajagopal, S. Biased signalling: from simple switches to allosteric microprocessors. Nat. Rev. Drug Discov. 17, 243–260 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Kimura, I., Ichimura, A., Ohue-Kitano, R. & Igarashi, M. Free fatty acid receptors in health and disease. Physiol. Rev. 100, 171–210 (2020).

    Article  PubMed  CAS  Google Scholar 

  6. Wang, B. et al. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct. Target Ther. 6, 94 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Liu, Y. et al. Metabolic profiling of murine plasma reveals eicosapentaenoic acid metabolites protecting against endothelial activation and atherosclerosis. Br. J. Pharmacol. 175, 1190–1204 (2018).

    Article  PubMed  CAS  Google Scholar 

  8. Oh, D. Y. et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142, 687–698 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Du, Y. Q. et al. Endogenous Lipid-GPR120 signaling modulates pancreatic islet homeostasis to different extents. Diabetes 71, 1454–1471 (2022).

    Article  PubMed  CAS  Google Scholar 

  10. Mao, C. et al. Unsaturated bond recognition leads to biased signal in a fatty acid receptor. Science 380, eadd6220 (2023).

    Article  PubMed  CAS  Google Scholar 

  11. Lahvic, J. L. et al. Specific oxylipins enhance vertebrate hematopoiesis via the receptor GPR132. Proc. Natl Acad. Sci. USA 115, 9252–9257 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Bokoch, M. P. et al. Ligand-specific regulation of the extracellular surface of a G-protein-coupled receptor. Nature 463, 108–112 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Lin, H. et al. Structures of the ADGRG2–Gs complex in apo and ligand-bound forms. Nat. Chem. Biol. 18, 1196–1203 (2022).

    Article  PubMed  CAS  Google Scholar 

  14. Xiao, P. et al. Tethered peptide activation mechanism of the adhesion GPCRs ADGRG2 and ADGRG4. Nature 604, 771–778 (2022).

    Article  PubMed  CAS  Google Scholar 

  15. Yang, F. et al. Structure, function and pharmacology of human itch receptor complexes. Nature 600, 164–169 (2021).

    Article  PubMed  CAS  Google Scholar 

  16. Ping, Y. Q. et al. Structures of the glucocorticoid-bound adhesion receptor GPR97–Go complex. Nature 589, 620–626 (2021).

    Article  PubMed  CAS  Google Scholar 

  17. O’Sullivan, C. & Dev, K. K. The structure and function of the S1P1 receptor. Trends Pharmacol. Sci. 34, 401–412 (2013).

    Article  PubMed  Google Scholar 

  18. Gupte, T. M., Malik, R. U., Sommese, R. F., Ritt, M. & Sivaramakrishnan, S. Priming GPCR signaling through the synergistic effect of two G proteins. Proc. Natl Acad. Sci. USA 114, 3756–3761 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Li, E. et al. OLFR734 mediates glucose metabolism as a receptor of asprosin. Cell Metab. 30, 319–328.e18 (2019).

    Article  PubMed  CAS  Google Scholar 

  20. Zhao, C. et al. Structural insights into sphingosine-1-phosphate recognition and ligand selectivity of S1PR3–Gi signaling complexes. Cell Res. 32, 218–221 (2022).

    Article  PubMed  CAS  Google Scholar 

  21. Dimmeler, S. et al. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 399, 601–605 (1999).

    Article  PubMed  CAS  Google Scholar 

  22. Wang, S. et al. P2Y2 and Gq/G11 control blood pressure by mediating endothelial mechanotransduction. J. Clin. Invest. 125, 3077–3086 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Balligand, J. L., Feron, O. & Dessy, C. eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol. Rev. 89, 481–534 (2009).

    Article  PubMed  CAS  Google Scholar 

  24. Yuan, Y. et al. Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition. Cell Res. 31, 1263–1274 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hanson, M. A. et al. Crystal structure of a lipid G protein-coupled receptor. Science 335, 851–855 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Xu, Z. et al. Structural basis of sphingosine-1-phosphate receptor 1 activation and biased agonism. Nat. Chem. Biol. 18, 281–288 (2022).

    Article  PubMed  CAS  Google Scholar 

  27. Nagatake, T. et al. The 17,18-epoxyeicosatetraenoic acid–G protein-coupled receptor 40 axis ameliorates contact hypersensitivity by inhibiting neutrophil mobility in mice and cynomolgus macaques. J. Allergy Clin. Immunol. 142, 470–484.e12 (2018).

    Article  PubMed  CAS  Google Scholar 

  28. Kumar, S., Kang, D. W., Rezvan, A. & Jo, H. Accelerated atherosclerosis development in C57Bl6 mice by overexpressing AAV-mediated PCSK9 and partial carotid ligation. Lab. Invest. 97, 935–945 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Bjorklund, M. M. et al. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ. Res. 114, 1684–1689 (2014).

    Article  PubMed  CAS  Google Scholar 

  30. Oni-Orisan, A. et al. Cytochrome P450-derived epoxyeicosatrienoic acids and coronary artery disease in humans: a targeted metabolomics study. J. Lipid Res. 57, 109–119 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Bhatt, D. L. et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N. Engl. J. Med. 380, 11–22 (2019).

    Article  PubMed  CAS  Google Scholar 

  32. Lavie, C. J., Milani, R. V., Mehra, M. R. & Ventura, H. O. Omega-3 polyunsaturated fatty acids and cardiovascular diseases. J. Am. Coll. Cardiol. 54, 585–594 (2009).

    Article  PubMed  CAS  Google Scholar 

  33. Doran, A. C. Inflammation resolution: implications for atherosclerosis. Circ. Res. 130, 130–148 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Hara, S., Tojima, I., Shimizu, S., Kouzaki, H. & Shimizu, T. 17,18-Epoxyeicosatetraenoic acid inhibits TNF-α-induced inflammation in cultured human airway epithelium and LPS-induced murine airway inflammation. Am. J. Rhinol. Allergy 36, 106–114 (2022).

    Article  PubMed  Google Scholar 

  35. Hou, J. et al. Targeted delivery of nitric oxide via a ‘bump-and-hole’-based enzyme-prodrug pair. Nat. Chem. Biol. 15, 151–160 (2019).

    Article  PubMed  CAS  Google Scholar 

  36. Lu, Z. et al. Mitochondrial reactive oxygen species and nitric oxide-mediated cancer cell apoptosis in 2-butylamino-2-demethoxyhypocrellin B photodynamic treatment. Free Radic. Biol. Med. 41, 1590–1605 (2006).

    Article  PubMed  CAS  Google Scholar 

  37. Zhu, Y. et al. LDL induces transcription factor activator protein-1 in human endothelial cells. Arterioscler. Thromb. Vasc. Biol. 18, 473–480 (1998).

    Article  PubMed  CAS  Google Scholar 

  38. Zhang, X., Yang, N., Ai, D. & Zhu, Y. Systematic metabolomic analysis of eicosanoids after omega-3 polyunsaturated fatty acid supplementation by a highly specific liquid chromatography–tandem mass spectrometry-based method. J. Proteome Res. 14, 1843–1853 (2015).

    Article  PubMed  CAS  Google Scholar 

  39. Cheng, J. et al. Autonomous sensing of the insulin peptide by an olfactory G protein-coupled receptor modulates glucose metabolism. Cell Metab. 34, 240–255.e10 (2022).

    Article  PubMed  CAS  Google Scholar 

  40. Olsen, R. H. J. et al. TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat. Chem. Biol. 16, 841–849 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Yang, F. et al. Structural basis of GPBAR activation and bile acid recognition. Nature 587, 499–504 (2020).

    Article  PubMed  CAS  Google Scholar 

  42. Xiao, P. et al. Ligand recognition and allosteric regulation of DRD1–Gs signaling complexes. Cell 184, 943–956.e18 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Qu, C. et al. Ligand recognition, unconventional activation, and G protein coupling of the prostaglandin E2 receptor EP2 subtype. Sci. Adv. 7, eabf1268 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Morris, G. M. et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J. Comput. Chem. 30, 2785–2791 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Huang, J. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2017).

    Article  PubMed  CAS  Google Scholar 

  46. Vanommeslaeghe, K. et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J. Comput. Chem. 31, 671–690 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Yang, Z. et al. Structure of GPR101–Gs enables identification of ligands with rejuvenating potential. Nat. Chem. Biol. 20, 484–492 (2024).

    Article  PubMed  CAS  Google Scholar 

  48. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    Article  PubMed  CAS  Google Scholar 

  49. Van Der Spoel, D. et al. GROMACS: fast, flexible, and free. J. Comput. Chem. 26, 1701–1718 (2005).

    Article  PubMed  Google Scholar 

  50. Kumari, R., Kumar, R., Open Source Drug Discovery Consortium, & Lynn, A. g_mmpbsa—a GROMACS tool for high-throughput MM-PBSA calculations. J. Chem. Inf. Model. 54, 1951–1962 (2014).

    Article  PubMed  CAS  Google Scholar 

  51. Zhang, C. et al. Coupling of Integrin α5 to Annexin A2 by Flow Drives Endothelial Activation. Circ. Res. 127, 1074–1090 (2020).

    Article  PubMed  CAS  Google Scholar 

  52. Li, B. et al. RNA N6-methyladenosine modulates endothelial atherogenic responses to disturbed flow in mice. eLife 11, e69906 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Key R&D Program of China (2019YFA0904200 to J.-P.S. and 2019YFA0802003 to Y.Z.); the National Science Fund for Distinguished Young Scholars Grant (81925003 to D.A.); the National Natural Science Foundation of China (T2321004 to J.-P.S., 82330118 to J.-P.S., 81773704 to J.-P.S., 82300890 to J.C., 82330012 to Y.Z., 82130014 to D.A.); and the Key Research Project of the Beijing Natural Science Foundation, China (Z200019 to J.-P.S.); China National Postdoctoral Program for Innovative Talents (BX20230201 to J.C.); China Postdoctoral Science Foundation (2023M742066 to J.C.). J.-P.S. is also supported by the Tencent New Cornerstone Investigator Program.

Author information

Authors and Affiliations

Authors

Contributions

T.Z., J.C., S.H., C.Z., M.-X.G. and L.-J.Z. performed the experiments and data analysis. D.A., Y.Z. and J.-P.S. designed and supervised the study; D.A., Y.Z. and J.-P.S. wrote the manuscript. All the authors reviewed and approved the final version of the manuscript.

Corresponding authors

Correspondence to Jin-Peng Sun, Yi Zhu or Ding Ai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Aldons Lusis and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Identification of the receptor of 17,18-EEQ and validation of siRNA knockdown efficiency.

a,b, HUVECs pretreated with 1 mM 17,18-EEQ for the indicated time point (6, 12, 24 h) were exposed to OSS (a) for 12 h or TNF-α(b) for 6 h. Representative Western blots and quantitative analysis of vascular cell adhesion molecule 1(VCAM-1) in whole cell lysates (n = 6). c, HUVECs transfected with scramble siRNA and GPR120 siRNA for 48 h were pretreated with 17,18-EEQ (1 mM) for 24 h, then with TNF-α (0.1 ng/ml) for 6 h. Western blot analysis of VCAM-1 and GPR120. Quantification of protein level of VCAM-1 (n = 3). d, HUVECs transfected with scramble siRNA and GPR132 siRNA for 48 h were pretreated with 17,18-EEQ (1 mM) for 24 h, then with TNF-α (0.1 ng/ml) for 6 h. Western blot analysis of VCAM-1 and GPR132. Quantification of protein level of VCAM-1 (n = 3). e, Quantitative PCR analysis was performed to detect 10 GPCRs mRNA levels in HUVECs transfected with scramble siRNA or specific siRNAs for 48 h (n = 3). f, Western blot analysis in candidate GPCRs (ADORA2A, CALCRL, GPR3, ADGRE5, ADRB2, GPR180, GPRC5A, GPR89A, P2RY1, S1PR1)-knockdown or -overexpressing HUVECs to validate antibodies for these 10 GPCRs. g, Whole-cell ELISA measuring the cell surface expression of VCAM-1 in HUVECs transfected with scramble siRNA or 10 GPCR-specific siRNAs for 48 h, then were pretreated with 17,18-EEQ (1 mM) for 24 h and TNF-α (0.1 ng/ml) for 6 h (n = 3). All data are mean±s.e.m. of biologically independent smaples, a-d, one-way ANOVA with Bonferroni’s multiple comparisons post hoc test. e, g, two-tailed Student’s t-tests.

Source data

Extended Data Fig. 2 Saturation binding assay of [3H]-S1P to interact with S1PR1 and incorporation of BRET sensors had no effect on S1PR1 expression.

a, Saturation binding of [3H]-S1P to the membrane fraction from S1PR1 overexpressing-HEK293 cell. The nonspecific binding in the presence of 1 μM unlabeled S1P was subtracted from the total binding to yield the specific binding curve. Representative curves from three biologically independent experiments were shown (n = 3). b, Saturation binding of [3H]-S1P to the membrane fraction from S1PR1 overexpressing-HEK293 cell. The nonspecific binding in the presence of 1 μM unlabeled 17,18-EEQ was subtracted from the total binding to yield the specific binding curve. Representative curves from three independent experiments were shown (n = 3). c, Elisa experiments to determine the expression levels of the wild-type S1PR1 and five FlAsH motif incorporated FLAsH-BRET sensors. Data from three biologically independent experiment (n = 3). All data are mean±s.e.m. of biologically independent smaples, c, one-way ANOVA with Bonferroni’s multiple comparisons post hoc test.

Source data

Extended Data Fig. 3 17,18-EEQ biasedly activated S1PR1-Gq-Ca2+ signaling.

a, HUVECs were treated with 1 μM 17,18-EEQ after S1PR1 knockdown by siNRA. ELISA were performed to measure cAMP levels (n = 3). b, Saturation binding of BODIPY-FL-GTPγS to the membrane fraction from HEK293 cell overexpressing S1PR1 combined with Gq, Gs, Gi, G12, G13 or control vehicle stimulated by 17,18-EEQ (n=3). c-h, Dose response curves of Gα (including Gs, Gi, Gq, G11, G12 and G13)-Gβγ dissociation in S1PR1-overexpressing HEK293 cells in response to stimulation with different agonists of S1PR1 (S1P,17,18-EEQ, AUY954 and SEW2871) (n = 3). i-m, Competitive binding curves of S1P or 17,18-EEQ to S1PR1-5 (n = 3). n,o, The potency of Gα (including Gs, Gi, Gq, G11 and G13)-Gβγ dissociation in response to stimulation with S1P (n) or 17,18-EEQ (o) in S1PR1 overexpressing HEK293 cells. The heatmap is colored according to the value of pEC50 (n = 3). p, The relative fpkm values of Gq and G11 in untreated HUVECs from RNA-seq (n = 3). q,r, HUVECs transfected with scramble siRNA and G11 siRNA were pretreated with 17,18-EEQ, then exposed to OSS (q) or TNF-α(r). The relative mRNA levels of VCAM-1 (vascular cell adhesion molecule 1) (n = 3). s,t, After G11 knockdown, HUVECs treated with 17,18-EEQ were stimulated by OSS (s) or TNF-α (t). Representative western blot and quantification of VCAM-1, G11 and GAPDH (n = 3). u, Effects of G11 siRNA on the 17,18-EEQ-induced IP3 production in HUVECs (n = 3). v, The changes of fluorescence intensity represents Ca2+ concentration in HUVECs treated with 1 μM 17,18-EEQ after S1PR1 silencing (left) (n = 24; 3 independent experiments). Quantification (right) of Ca2+ transient was represented by the relative AUC (n = 3). All data are mean±s.e.m. of biologically independent smaples, ns, no significant difference; ND, signal not detectable. a,q-v two-way ANOVA with Bonferrni’s post hoc test.

Source data

Extended Data Fig. 4 17,18-EEQ biasedly activated S1PR1-Gq-Ca2+ signaling to inhibit endothelial activation.

a, HUVECs transfected with scramble siRNA and S1PR1 siRNA for 48 h, were pretreated with 17,18-EEQ (1 mM) for 24 h, then with TNF-α (0.1 ng/ml) for another 6 h. Representative western blots and quantification of the protein levels of VCAM-1 (n = 6). b,c, Representative blots (b) and quantification (c) of the phosphorylation and protein levels of p-IKKα, p-p65, IκBα in HUVECs transfected by scramble siRNA or S1PR1 siRNA, and then treated with 1 mM 17,18-EEQ, then with 0.1 ng/ml TNF-α (n=6). d,e, Representative images of monocyte-endothelial adhesion (d). HUVECs transfected with scramble siRNA and S1PR1 siRNA for 48 h, were treated with 1 mM 17,18-EEQ for 24 h, then exposed to 0.1 ng/ml TNF-α for 6 h. The number of adhesive cells were quantified from 5 random fields with 10× objective in each experiment and was normalized to static HUVECs (e) (n = 3). f, Effects of YM254890 Gq inhibitor and Ionomycin calcium ionophore on TNF-α-induced VCAM-1 expression in HUVECs treated with 1 μM 17,18-EEQ or PBS. Representative blots of the protein levels of VCAM-1 (n = 6). g,h, After pretreatment with YM254890, Ionomycin, or their combination for 1 h, HUVECs were subjected to 1 μM 17,18-EEQ for 4 h, then exposed to TNF-α for 30 min. Representative blots (g) and quantification (h) of protein levels of p-IKKα, p-p65, IκBα (n = 6). All data are mean±s.e.m. of biologically independent smaples, ns, no significant difference. a,c,e, two-way ANOVA with Bonferroni’s multiple comparisons post hoc test. f,h, one-way ANOVA with Bonferroni’s multiple comparisons post hoc test.

Source data

Extended Data Fig. 5 17,18-EEQ dose and time dependently induced eNOS ser1177 phosphorylation, which was mediated by calcium release.

a,b, HUVECs were seeded on plates pretreated with various concentrations (a) of 17,18-EEQ (1, 10, 100, 1000 nM) for the indicated time points (b) (15, 30, 45, 60 min). Representative blots and quantification of the protein levels of p-eNOS (n = 3). c, HUVECs pretreated with EGTA or DMSO for 30 min were subjected to 1 μM 17,18-EEQ for another 45 min. Representative blot (left panel) of p-eNOS (Ser1177) and total eNOS. Quantification of protein levels of p-eNOS in the right panel (n = 6). d, Strategy for the generation of S1pr1fl/fl mice. e, S1pr1fl/fl mice were injected intraperitoneally with or without 75 mg/kg tamoxifen for five days. Representative immunofluorescence staining images of S1PR1, VE-cadherin and DAPI in mouse carotid arteries, with quantitative analysis in the right panel (n = 3). Scale bar: 20 μm. f, Plasma levels of 17,18-EEQ in S1pr1fl/fl mice followed by 17,18-EEQ (5 μg/kg) intraperitoneal injection (n = 6). All data are mean±s.e.m. of biologically independent smaples, a,b, Kruskal-Wallis test. c, two-way ANOVA with Bonferroni’s multiple comparisons post hoc test. e,f, two-tailed Student’s t-tests.

Source data

Extended Data Fig. 6 The potential binding sites at S1PR1 for 17,18-EEQ by docking.

a, The average Root-mean-square deviation (RMSD) value of 17, 18-EEQ during triplicate 500 ns of MD simulation. b, Dose response curves of key residue mutations for the toggle switch W2696.48 and E1413.49-R1423.50-Y1433.51 of S1PR1 in response to stimulation with 17,18-EEQ (n = 3). c, Effects of key residue mutations of binding pockets of S1PR1 on radioligand binding induced by S1P or 17,18-EEQ. Bars represent differences in calculated potency for each mutant compared with the wild type (n = 3). d, Competitive binding curves of S1P (left) or 17,18-EEQ (right) for the mutants in the binding pockets of S1PR1 (n = 3). e, Barcode representation of stronger interaction patterns in the ligand pocket of S1P-S1PR1 and 17,18-EEQ-S1PR1. Residues of S1PR1 formed both interactions with ligands were indicated in orange. Residues that only show interactions with S1P or 17,18-EEQ were indicated in light blue or light green, respectively. f, Effects of key residue mutations (V194A, L195A, F210A, W269A, L272A, L276A and L297A) of binding pockets of S1PR1 on FlAsH-BRET ratio changes induced by S1P or 17,18-EEQ. Bars represent differences in calculated potency for each mutant compared with the wild type (n = 3). g,h, Representative dose-response curves of the S1P or 17,18-EEQ-induced FlAsH-BRET ratio for the mutants (V194A, L195A, F210A, W269A, L272A, L276A and L297A) in the binding pockets of S1P-S1PR1 (g) or 17,18-EEQ-S1PR1 (h) (n = 3). All data are mean±s.e.m. of biologically independent smaples. c,f, two-sided one-way ANOVA with Tukey’s test (compared with wild type). All data are mean±s.e.m. of biologically independent smaples. c,f, two-sided one-way ANOVA with Tukey’s test (compared with wild type).

Source data

Extended Data Fig. 7 17,18-EEQ has no effect on serum lipid and other regioisomeric EEQs plasma levels in vivo.

a, Hepatic expression of low-density lipoprotein receptor from mice administrated by AAV-ctrl or AAV-PCSK9DY for 28 days (n = 3). b,c, Serum levels of triglyceride (b), total cholesterol (c) in S1pr1fl/fl and S1pr1ECKO mice followed by AAV-PCSK9DY (1 x 1011 VG) administration and western diet (WD). These mice were implanted with an osmotic minipump infusing with 17,18-EEQ (5 μg/kg) for 7 days and subjected to PCAL surgery for another 21 days (n = 8). (d-g) Eight-week-old S1pr1fl/fl and S1pr1ECKO mice administrated by AAV-PCSK9DY (1 x 1011 VG) and fed with WD, were implanted with an osmotic minipump infusing 17,18-EEQ (5 μg/kg) for 7 days and subjected to PCAL surgery for another 21 days. Plasma levels of CYP products 5,6-EEQ (d), 8,9-EEQ (e), 11,12-EEQ (f), 14,15-EEQ (g) were determined by LC-MS/MS (n = 8). h,i, Eight-week-old S1pr1fl/fl and S1pr1ECKO mice administrated by AAV-PCSK9DY (1 x 1011 VG) and fed with WD, were implanted with an osmotic minipump infusing 17,18-EEQ (5 μg/kg) for 7 days and subjected to PCAL surgery for another 7 days. Representative immunofluorescence staining images (h) of t-eNOS, VE-cadherin and DAPI in male mouse carotids. Quantification data (i) of t-eNOS expression (n = 6). Scale bar: 20 μm. The statistical analysis was performed on LCA groups. All data are mean±s.e.m. of biologically independent smaples, ns, no significant difference. a, two-tailed Student’s t-tests. b-g,i, two-way ANOVA with Bonferroni’s multiple comparisons post hoc test.

Source data

Extended Data Fig. 8 Vascepa has no effect on other regioisomeric EEQs plasma levels and t-eNOS expression in vivo.

a-f, Eight-week-old S1PR1fl/fl and S1PR1ECKO male mice followed by AAV-PCSK9DY (1 x 1011 VG) administration and western diet (WD). These mice were orally gavaged with Vascepa (666.7 mg/kg) or olive oil for 7 days and subjected to PCAL surgery for another 21 days. The levels of triglyceride (a), total cholesterol (b), 5,6-EEQ (c), 8,9-EEQ (d), 11,12-EEQ (e), 14,15-EEQ (f) in these mice (n = 8). g, Eight-week-old S1pr1fl/fl and S1pr1ECKO male mice administrated by AAV-PCSK9DY (1 x 1011 VG) and fed with WD, were treated with Vascepa (666.7 mg/kg) or olive oil for 7 days and subjected to PCAL surgery for another 7 days. Representative images (g) and of quantification of t-eNOS (h), VE-cadherin and DAPI in male mouse carotids. (n = 6). Scale bar: 20 μm. The statistical analysis was performed on LCA groups. h, Schematic diagram of the PCAL-induced atherosclerosis model. i-q, The levels of triglycerides (i), total cholesterol (j), EPA (k), 17,18-EEQ (l), 17,18-DHETE (m), 5,6-EEQ (n), 8,9-EEQ (o), 11,12-EEQ (p), 14,15-EEQ (q) in PCSK9DY injected-S1pr1fl/fl and -S1pr1ECKO mice performed with PCAL, were treated with Vascepa (666.7 kg/mg) or Olive oil, and fed with WD for 4 weeks (n = 5). r, Isolated carotid arteries in PCSK9DY injected-S1pr1fl/fl and -S1pr1ECKO mice treated with Vascepa (666.7 kg/mg) or Olive oil (n = 5). Scale bars: 1.5 mm. s-u, Representative images (s) and quantification (t-u) of plaques in ligated carotid arteries of mice were analyzed with H&E staining. Scale bars: 100 μm (n = 5). All data are mean±s.e.m. of biologically independent smaples, ns, no significant difference. a-g,i-q,t,u, two-way ANOVA with Bonferroni’s multiple comparisons post hoc test.

Source data

Extended Data Fig. 9 The beneficial effect of Vascepa on reducing aortic atherosclerosis is mediated by endothelial S1PR1.

a, Schematic diagram of the experimental strategy for the adeno-associated virus (AAV) administration, oral gavage, western diet (WD) feeding, and subsequent analyses for female mice. b-h, Plasma levels of EPA (b), 17,18-EEQ (c), 17,18-DHETE (d), 5,6-EEQ (e), 8,9-EEQ (f), 11,12-EEQ (g), 14,15-EEQ (h) from S1pr1fl/fl and S1pr1ECKO mice followed by PCSK9DY injection with WD feeding for 12 weeks. Vascepa (666.7 kg/mg) or olive oil were orally administered once a day (n = 5). i, Oil Red staining of aortas in PCSK9DY injected-S1pr1fl/fl and -S1pr1ECKO mice treated with Vascepa (666.7 kg/mg) or Olive oil (n = 5). Scale bars: 5 mm. j, Quantification of Oil Red staining of whole aortas, aortic arch, thoracic aorta from PCSK9DY injected-S1pr1fl/fl and -S1pr1ECKO mice (n = 5). k, Oil Red (upper panel) and H&E (lower panel) staining of atherosclerotic lesion areas in cross-sections at the aortic root (n = 5). Scale bars: 200 μm. l, Quantification of aortic root atherosclerotic plaques (n = 5). m-n, The levels of triglycerides (m) and total cholesterol (n) in serum collected from PCSK9DY injected-S1pr1fl/fl and -S1pr1ECKO mice treated with Vascepa (666.7 kg/mg) or Olive oil, and fed with WD for 12 weeks (n = 5). All data are mean±s.e.m. of biologically independent smaples, ns, no significant difference. b-h,j,l-n, two-way ANOVA with Bonferroni’s multiple comparisons post hoc test.

Source data

Extended Data Fig. 10 The mechanism of the anti-inflammatory effect mediated by 17,18-EEQ.

S1PR1 functions as a receptor for 17,18-EEQ, selectively activating Gq-Ca2+-eNOS-NO signaling to inhibit vascular endothelial inflammation and hinder the progression of atherosclerosis.

Supplementary information

Reporting Summary

Supplementary Tables 1 and 2

1. The expression profile of GPCRs in HUVECs from RNA-seq (left) and public dataset (right). 2. Statistical details in all figure panels.

Supplementary Data 1–4

1. Antibodies used for Western blots and Immunofluorescence staining in this study. 2. Chemical reagents used for this study. 3. Primer sequence. 4. siRNA sequences.

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Statistical Source Data.

Uncropped western blots

Uncropped western blots for both main figures and extended data figures.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, T., Cheng, J., He, S. et al. The sphingosine-1-phosphate receptor 1 mediates the atheroprotective effect of eicosapentaenoic acid. Nat Metab 6, 1566–1583 (2024). https://doi.org/10.1038/s42255-024-01070-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-024-01070-3

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing