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:

GPR15-mediated T cell recruitment during acute viral myocarditis facilitated virus elimination and improved outcome

Abstract

Viral myocarditis is characterized by infiltration of mononuclear cells essential for virus elimination. GPR15 has been identified as a homing receptor for regulatory T cells in inflammatory intestine diseases, but its role in inflammatory heart diseases is still elusive. Here we show that GPR15 deficiency impairs coxsackievirus B3 elimination, leading to adverse cardiac remodeling and dysfunction. Delayed recruitment of regulatory T cells in GPR15-deficient mice was accompanied by prolonged persistence of cytotoxic and regulatory T cells. In addition, RNA sequencing revealed prolonged inflammatory response and altered chemotaxis in knockout mice. In line, we identified GPR15 and its ligand GPR15L as an important chemokine receptor–ligand pair for the recruitment of regulatory and cytotoxic T cells. In summary, the insufficient virus elimination might be caused by a delayed recruitment of T cells as well as delayed interferon-γ expression, resulting in a prolonged inflammatory response and an adverse outcome in GPR15-deficient mice.

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: Impact of GPR15 deficiency during the subacute phase of CVB3-induced myocarditis 16 days p.i.
Fig. 2: Impact of GPR15 deficiency during acute phase of CVB3-induced myocarditis 5 days, 6 days and 7 days p.i.
Fig. 3: Analysis of immune cell infiltration in LV tissue during the acute phase 5 days, 6 days and 7 days after CVB3 infection.
Fig. 4: Identification of DEGs comparing cardiac tissue samples from Gpr15gfp/gfp and WT mice at 6 days and 7 days after CVB3 infection.
Fig. 5: Upregulated GO terms related to T-cell-mediated immune response and chemotaxis in infected Gpr15gfp/gfp mice on day 7 p.i.
Fig. 6: GPR15-expressing splenocytes and their interactions with the ligand GPR15L.
Fig. 7: IFNγ in different T cell subtypes and its influence on virus load and replication in HL1 cells.

Similar content being viewed by others

Data availability

RNA sequencing data are available under GSE248521. Source data of all main figures are provided.

Code availability

The code used for processing of RNA sequencing data (poly(A) tail clipping) is available at https://github.com/AGLindner/GPR15-RNA-Sequencing.

References

  1. Fung, G., Luo, H., Qiu, Y., Yang, D. & McManus, B. Myocarditis. Circ. Res. 118, 496–514 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Blauwet, L. A. & Cooper, L. T. Myocarditis. Prog. Cardiovasc. Dis. 52, 274–288 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Cooper, L. T. Jr Myocarditis. N. Engl. J. Med. 360, 1526–1538 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Schultz, J. C., Hilliard, A. A., Cooper, L. T. Jr. & Rihal, C. S. Diagnosis and treatment of viral myocarditis. Mayo Clin. Proc. 84, 1001–1009 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Fabre, A. & Sheppard, M. N. Sudden adult death syndrome and other non-ischaemic causes of sudden cardiac death. Heart 92, 316–320 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Kuhl, U. et al. High prevalence of viral genomes and multiple viral infections in the myocardium of adults with ‘idiopathic’ left ventricular dysfunction. Circulation 111, 887–893 (2005).

    Article  PubMed  Google Scholar 

  7. Kandolf, R. et al. Mechanisms and consequences of enterovirus persistence in cardiac myocytes and cells of the immune system. Virus Res. 62, 149–158 (1999).

    Article  CAS  PubMed  Google Scholar 

  8. Bouin, A. et al. Enterovirus persistence in cardiac cells of patients with idiopathic dilated cardiomyopathy is linked to 5′ terminal genomic RNA-deleted viral populations with viral-encoded proteinase activities. Circulation 139, 2326–2338 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lammermann, T. & Kastenmuller, W. Concepts of GPCR-controlled navigation in the immune system. Immunol. Rev. 289, 205–231 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Dubyak, G. R. GPCRs in innate and adaptive immune responses. In GPCRs: Structure, Function, and Drug Discovery (eds Jastrzebska, B. & Park. P.S.-H.) 429–461 (Elsevier, 2020).

  11. Heiber, M. et al. A novel human gene encoding a G-protein-coupled receptor (GPR15) is located on chromosome 3. Genomics 32, 462–465 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Deng, H. K., Unutmaz, D., KewalRamani, V. N. & Littman, D. R. Expression cloning of new receptors used by simian and human immunodeficiency viruses. Nature 388, 296–300 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Nguyen, L. P. et al. Role and species-specific expression of colon T cell homing receptor GPR15 in colitis. Nat. Immunol. 16, 207–213 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Lahl, K., Sweere, J., Pan, J. & Butcher, E. Orphan chemoattractant receptor GPR15 mediates dendritic epidermal T-cell recruitment to the skin. Eur. J. Immunol. 44, 2577–2581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kim, S. V. et al. GPR15-mediated homing controls immune homeostasis in the large intestine mucosa. Science 340, 1456–1459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jegodzinski, L. et al. The G protein-coupled receptor (GPR) 15 counteracts antibody-mediated skin inflammation. Front. Immunol. 11, 1858 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Fischer, A. et al. Differential effects of α4β7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo. Gut 65, 1642–1664 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Ocon, B. et al. A mucosal and cutaneous chemokine ligand for the lymphocyte chemoattractant receptor GPR15. Front. Immunol. 8, 1111 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Pan, B. et al. The fifth epidermal growth factor like region of thrombomodulin alleviates LPS-induced sepsis through interacting with GPR15. Thromb. Haemost. 117, 570–579 (2017).

    Article  PubMed  Google Scholar 

  20. Suply, T. et al. A natural ligand for the orphan receptor GPR15 modulates lymphocyte recruitment to epithelia. Sci. Signal. 10, eaal0180 (2017).

  21. Becher, P. M. et al. Cardiac function remains impaired despite reversible cardiac remodeling after acute experimental viral myocarditis. J. Immunol. Res. 2017, 6590609 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bacmeister, L. et al. Inflammation and fibrosis in murine models of heart failure. Basic Res. Cardiol. 114, 19 (2019).

    Article  PubMed  Google Scholar 

  23. Weinzierl, A. O. et al. Effective chemokine secretion by dendritic cells and expansion of cross-presenting CD4/CD8+ dendritic cells define a protective phenotype in the mouse model of coxsackievirus myocarditis. J. Virol. 82, 8149–8160 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lindner, D. et al. Cardiac fibroblasts aggravate viral myocarditis: cell specific coxsackievirus B3 replication. Mediators Inflamm. 2014, 519528 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Supek, F., Bosnjak, M., Skunca, N. & Smuc, T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS ONE 6, e21800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bauer, M. The role of GPR15 function in blood and vasculature. Int. J. Mol. Sci. 22, 10824 (2021).

  27. Huan, T. et al. A whole-blood transcriptome meta-analysis identifies gene expression signatures of cigarette smoking. Hum. Mol. Genet. 25, 4611–4623 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Haase, T. et al. Novel DNA methylation sites influence GPR15 expression in relation to smoking. Biomolecules 8, 74 (2018).

  29. Haase, T. et al. G protein-coupled receptor 15 expression is associated with myocardial infarction. Int. J. Mol. Sci. 24, 180 (2022).

  30. Fairweather, D., Kaya, Z., Shellam, G. R., Lawson, C. M. & Rose, N. R. From infection to autoimmunity. J. Autoimmun. 16, 175–186 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Machado, F. S. et al. CCR5 plays a critical role in the development of myocarditis and host protection in mice infected with Trypanosoma cruzi. J. Infect. Dis. 191, 627–636 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Muller, I. et al. CX3CR1 knockout aggravates Coxsackievirus B3-induced myocarditis. PLoS ONE 12, e0182643 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Pappritz, K. et al. Immunomodulation by adoptive regulatory T-cell transfer improves Coxsackievirus B3-induced myocarditis. FASEB J. https://doi.org/10.1096/fj.201701408R (2018).

  34. Shi, Y. et al. Regulatory T cells protect mice against coxsackievirus-induced myocarditis through the transforming growth factor β–coxsackie-adenovirus receptor pathway. Circulation 121, 2624–2634 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Henke, A., Huber, S., Stelzner, A. & Whitton, J. L. The role of CD8+ T lymphocytes in coxsackievirus B3-induced myocarditis. J. Virol. 69, 6720–6728 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kang, S., Brown, H. M. & Hwang, S. Direct antiviral mechanisms of interferon-gamma. Immune Netw. 18, e33 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Fairweather, D. et al. Interferon-γ protects against chronic viral myocarditis by reducing mast cell degranulation, fibrosis, and the profibrotic cytokines transforming growth factor-β1, interleukin-1β, and interleukin-4 in the heart. Am. J. Pathol. 165, 1883–1894 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Becher, P. M. et al. Role of Toll-like receptors and interferon regulatory factors in different experimental heart failure models of diverse etiology: IRF7 as novel cardiovascular stress-inducible factor. PLoS ONE 13, e0193844 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hinrichs, S. et al. Precursor proadrenomedullin influences cardiomyocyte survival and local inflammation related to myocardial infarction. Proc. Natl Acad. Sci. USA 115, E8727–E8736 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Westermann, D. et al. Selective PDE5A inhibition with sildenafil rescues left ventricular dysfunction, inflammatory immune response and cardiac remodeling in angiotensin II-induced heart failure in vivo. Basic Res. Cardiol. 107, 308 (2012).

    Article  PubMed  Google Scholar 

  41. National Research Council. Guide for the Care and Use of Laboratory Animals 8th edn (National Academies Press, 2011).

  42. Pacher, P., Nagayama, T., Mukhopadhyay, P., Batkai, S. & Kass, D. A. Measurement of cardiac function using pressure–volume conductance catheter technique in mice and rats. Nat. Protoc. 3, 1422–1434 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bacmeister, L. et al. Assessment of PEEP-ventilation and the time point of parallel-conductance determination for pressure–volume analysis under β-adrenergic stimulation in mice. Front. Cardiovasc. Med. 6, 36 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lindner, D. et al. Protective function of STAT3 in CVB3-induced myocarditis. Cardiol. Res. Pract. 2012, 437623 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the \(2^{-{\Delta}{\Delta}{C}_T}\) method. Methods 25, 402–408 (2001).

  46. Zawada, A. M. et al. Massive analysis of cDNA ends (MACE) and miRNA expression profiling identifies proatherogenic pathways in chronic kidney disease. Epigenetics 9, 161–172 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17 https://doi.org/10.14806/ej.17.1.200 (2011).

  48. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Alexa, A. & Rahnenfuhrer, J. Analysis for Gene Ontology. R package version 2.42.0; https://bioconductor.org/packages/topGO (2020).

  51. Gu, Z. & Hübschmann, D. simplifyEnrichment: A Bioconductor Package for Clustering and Visualizing Functional Enrichment Results. Genomics Proteomics Bioinformatics 21, 190–202 (2023).

    Article  PubMed  Google Scholar 

  52. Lin, D. An information-theoretic definition of similarity. ICML ʼ98: Proc. of the Fifteenth International Conference on Machine Learning 98, 296–304 (1998).

    Google Scholar 

  53. Pedersen T. ggforce: Accelerating ‘ggplot2’. version 3.3.3; https://ggforce.data-imaginist.com, https://github.com/thomasp85/ggforce (2021).

  54. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).

  55. Walter, W., Sanchez-Cabo, F. & Ricote, M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics 31, 2912–2914 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Scherschel, K. et al. Characterization of the HCN interaction partner TRIP8b/PEX5R in the intracardiac nervous system of TRIP8b-deficient and wild-type mice. Int. J. Mol. Sci. 22, 4772 (2021).

  57. Brauninger, H. et al. Cardiac SARS-CoV-2 infection is associated with pro-inflammatory transcriptomic alterations within the heart. Cardiovasc. Res. 118, 542–555 (2022).

    Article  PubMed  Google Scholar 

  58. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Voss, S. et al. Macrophage migration inhibitory factor (MIF) expression increases during myocardial infarction and supports pro-inflammatory signaling in cardiac fibroblasts. Biomolecules 9, 38 (2019).

  60. Lindner, D. et al. Cardiac fibroblasts support cardiac inflammation in heart failure. Basic Res. Cardiol. 109, 428 (2014).

  61. Plendl, J., Sinowatz, F. & Auerbach, R. A transformed murine myocardial vascular endothelial cell clone: characterization of cells in vitro and of tumours derived from clone in situ. Virchows Arch 426, 619–628 (1995).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the core unit for cytometry and cell sorting; the UKE Microscopy Imaging Facility at the University Hospital Centre Hamburg-Eppendorf for providing flow cytometers, microscopes and support; and GenXPro for performing MACE analyses and bioinformatics. Furthermore, the authors thank O. E. Meyfarth and R. Scholz for technical assistance. We greatly appreciate the assistance of K. Hartmann and S. Krasemann (HEXT Mouse Pathology Core Facility, UKE Hamburg) in processing histological samples. We thank the Lighthouse Core Facility (LCF) for support with cell sorting and flow cytometry. LCF is funded, in part, by the Medical Faculty, University of Freiburg (project nos. 2021/A2-Fol and 2021/B3-Fol) and the German Research Foundation (DFG) (project no. 450392965). The transgenic DsRed mice were kindly provided by R. Kesselring (Department of General and Visceral Surgery, Medical Center, University of Freiburg). H.W., T.V., T.M., D. Wolf, I.H., D. Westermann and D.L. are members of SFB1425, funded by the DFG (project no. 422681845). This work was supported by the German Centre for Cardiovascular Research (DZHK) (FKZ 81Z0710108 to D. Westermann and D.L.).

Author information

Authors and Affiliations

Authors

Contributions

Conceived and designed the experiments: B.S., H.W., D. Wolf, I.H., D. Westermann and D.L. Performed the experiments: B.S., H.W., L.B., S.K., T.V., T.M., I.Y., A.A. and D.L. Analyzed the data: B.S., H.W., L.B., T.V., T.M., F.E. and D.L. Contributed materials/analysis tools: B.S., H.W., L.B., T.V., T.M., M.A.B., P.M.B., F.E., S.V.K., K.K., P.K., S.B., T.Z., D. Wolf, I.H., D. Westermann and D.L. Wrote the manuscript: B.S., H.W., L.B., D. Westermann and D.L.

Corresponding author

Correspondence to Diana Lindner.

Ethics declarations

Competing interests

All authors declare no competing financial interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

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 Analysis of immune cells in blood, lymph nodes and LV tissue sections.

(a) Representative staining of CD3 (left panel) or CD8 (right panel) in immune cell infiltrates during the acute phase of myocarditis in heart tissue of infected WT and GPR15deficient mice. Only infected mice were stained, n numbers are shown in Fig. 2a. (b) Gene expression of Gpr15 was determined in blood of WT mice. N numbers stated in Fig. 2a. Gene expression of specific immune cell markers for T cells (Cd3) and T cell sub-populations TH (Cd4), TC (Cd8) and Treg cells (Foxp3) was determined in blood of WT and GPR15-deficient mice. Ct-values were normalised to 18S and the corresponding sham (sh) controls (ΔΔCt). 2¬ ΔΔCt values were plotted (geo-mean ± 95% CI). Significance was tested using an unpaired two-tailed t-test with Bonferroni correction. (c) Gene expression of Gpr15 was determined in lymph nodes of WT mice. N numbers stated in Fig. 2a. Gene expression of specific immune cell markers for T cells (Cd3) and T cell sub-populations TH (Cd4), TC (Cd8) and Treg cells (Foxp3) was determined in lymph nodes of WT and GPR15-deficient mice. Ct-values were normalised to Cdkn1b and the corresponding sham (sh) controls (ΔΔCt). 2¬ ΔΔCt values were plotted (geo-mean ± 95% CI). Unpaired two-tailed t-test with Bonferroni correction. Significant, compared * to sh of the same genotype, # between similarly treated groups of different genotypes. (*, **, ***, ****; p < 0.05, 0.01, 0.001, 0.0001)

Source data

Extended Data Fig. 2 Volcano plots to visualise DEGs assigned to the GO term GO:0006915 “response to virus”.

(a) Myocarditis – 6 days: CVB3-infected WT (left) and Gpr15gfp/gfp (right) mice compared to their sham (sh) control 6 days p.i. (b) Myocarditis – 7 days: CVB3-infected WT (left) and Gpr15gfp/gfp (right) mice compared to their sh control 7 days p.i. (c) Comparison of sh WT and sh Gpr15gfp/gfp mice. Transcriptome analysis was performed by MACE-RNA seq of LV tissue. Fold change (FC) and p-value are displayed in the Volcano plot to visualise significant differences in gene expression. Genes assigned to the GO term GO:0006915 “response to virus” were highlighted as follows: (i) Not significantly regulated (black), (ii) significantly upregulated (FC > 1.5 & p-value < 0.05 (light red), p-value < 0.001 (dark red)) or (iii) significantly downregulated (FC < -1.5 & p-value < 0.05 (light blue), p-value < 0.001 (dark blue)). Genes labelled with their symbol are part of this GO term and were quantified by TaqMan analyses and plotted in Fig. 3. Genes not assigned to the GO term GO:0006915 are displayed in grey. DEGs were calculated with DESeq2 using Wald test.

Source data

Extended Data Fig. 3 69 similar regulated DEGs found by comparing DEGs from day 6 and day 7.

Heat maps of the 69 DEGs (58 up- and 11 downregulated in GPR15-deficient mice, FC + /-1.5 and a p-value < 0.001) that were similar regulated on day 6 (left) and 7 (right) p.i. Gene expression is normalized to the mean of the respective CVB3-infected WT group and plotted as fold change (FC, log2) for each sample separately. DEGs were calculated with DESeq2 using Wald test.

Source data

Extended Data Fig. 4 Similarity matrix of significant GO terms of the domain biological process.

957 GO terms that were significantly different on day 6 and, or on day 7 between both infected genotypes were clustered based on semantic similarity using the R package simplify enrichment. “Lin” method was used to calculate the similarity matrix for these GO terms. 15 semantic similarity clusters were determined (right) and were further summarized into 8 main clusters due to strong similarities (left, C1-C8).

Source data

Extended Data Fig. 5 Adhesion assay and analysis of GPR15-mediated T cell functionality.

(a) Gpr15l gene expression in HL1 cardiomyocytes after TNFα stimulation (10 ng/ml, 6 h, n = 4 biological replicates). Ct-values were normalised to Cdnk1b and the corresponding untreated control (ΔΔCt). Unpaired two-tailed t-test. (b) Quiescent or activated T cells were incubated in the presence or absence of the receptor agonist GPR15L (500 nM). Percentage of positive cells for the intracellular markers (GranzymeB (GrB), IL10, IL17, IFNγ, TNF-α) was quantified for three different T cell populations: CD4+CD25-: T helper cells (TH); CD4+CD25+: regulatory T cells (Treg); CD8+: cytotoxic T cells (TC). (c) Thrombomodulin (Thbd) expression after TNFα stimulation in MHEC‐5 T (25 ng/mL, 5 h, n = 4 biological replicates). Ct‐values were normalised to 18 S and the untreated control (ΔΔCt). Gene expression data were plotted as 2-ΔΔCt (geo-mean ± 95% CI). Unpaired two‐tailed t‐test. (d) MHEC‐5 T monolayer were either cultured with growth medium alone or supplemented with 25 ng/mL TNFα for 24 h. Adhesion of primary splenocytes to MHEC‐5 T monolayer (n = 6-13 biological replicates in 3 independent experiments) was investigated during constant flow for 20 min. The adhesion strength of primary splenocytes was analysed after 5 min of flow pause to allow cells to adhere before flow was turned on to remove nonadherent splenocytes. Adherent splenocytes were counted via FIJI (mean ± 95% CI). Unpaired two‐tailed t‐test. Significant, compared * to control or control of the same genotype (*, **,****; p < 0.05, 0.01, 0.0001).

Source data

Supplementary Information

Supplementary Information

Supplementary Figs. 1–5 and Supplementary Tables 1–10

Reporting Summary

Source data

Source Data Fig. 1–7 and Extended Data Fig. 1–5

Source data and exact P value for comparisons

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

Stoffers, B., Wolf, H., Bacmeister, L. et al. GPR15-mediated T cell recruitment during acute viral myocarditis facilitated virus elimination and improved outcome. Nat Cardiovasc Res 3, 76–93 (2024). https://doi.org/10.1038/s44161-023-00401-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-023-00401-z

This article is cited by

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