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:

Maintenance of the marginal-zone B cell compartment specifically requires the RNA-binding protein ZFP36L1

Abstract

RNA-binding proteins of the ZFP36 family are best known for inhibiting the expression of cytokines through binding to AU-rich elements in the 3′ untranslated region and promoting mRNA decay. Here we identified an indispensable role for ZFP36L1 as the regulator of a post-transcriptional hub that determined the identity of marginal-zone B cells by promoting their proper localization and survival. ZFP36L1 controlled a gene-expression program related to signaling, cell adhesion and locomotion; it achieved this in part by limiting expression of the transcription factors KLF2 and IRF8, which are known to enforce the follicular B cell phenotype. These mechanisms emphasize the importance of integrating transcriptional and post-transcriptional processes by RNA-binding proteins for maintaining cellular identity among closely related cell types.

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

Figure 1: MZ B cells specifically require ZFP36L1.
Figure 2: B cell–intrinsic requirement for ZFP36L1.
Figure 3: ZFP36L1 is selectively required for the maintenance of MZ and MZP B cells.
Figure 4: Gene-expression analysis following deletion of Zfp36l1.
Figure 5: ZFP36L1 enforces MZ B cell identity.
Figure 6: ZFP36L1 regulates IRF8 expression.
Figure 7: ZFP36L1 regulates KLF2.
Figure 8: ZFP36L1 controls the localization of MZ B cells in the splenic MZ.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. 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. 9, 54–62 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Arnon, T.I., Horton, R.M., Grigorova, I.L. & Cyster, J.G. Visualization of splenic marginal zone B-cell shuttling and follicular B-cell egress. Nature 493, 684–688 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Song, J. et al. Extracellular matrix of secondary lymphoid organs impacts on B-cell fate and survival. Proc. Natl. Acad. Sci. USA 110, E2915–E2924 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Simonetti, G. et al. IRF4 controls the positioning of mature B cells in the lymphoid microenvironments by regulating NOTCH2 expression and activity. J. Exp. Med. 210, 2887–2902 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fasnacht, N. et al. Specific fibroblastic niches in secondary lymphoid organs orchestrate distinct Notch-regulated immune responses. J. Exp. Med. 211, 2265–2279 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Cerutti, A., Cols, M. & Puga, I. Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes. Nat. Rev. Immunol. 13, 118–132 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pillai, S. & Cariappa, A. The follicular versus marginal zone B lymphocyte cell fate decision. Nat. Rev. Immunol. 9, 767–777 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Martin, F. & Kearney, J.F. Marginal-zone B cells. Nat. Rev. Immunol. 2, 323–335 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Srivastava, B., Quinn, W.J. III, Hazard, K., Erikson, J. & Allman, D. Characterization of marginal zone B cell precursors. J. Exp. Med. 202, 1225–1234 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kleiman, E. et al. Distinct transcriptomic features are associated with transitional and mature B-cell populations in the mouse spleen. Front. Immunol. 6, 30 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Tan, J.B. et al. Lunatic and manic fringe cooperatively enhance marginal zone B cell precursor competition for delta-like 1 in splenic endothelial niches. Immunity 30, 254–263 (2009).

    Article  PubMed  CAS  Google Scholar 

  12. Witt, C.M., Won, W.J., Hurez, V. & Klug, C.A. Notch2 haploinsufficiency results in diminished B1 B cells and a severe reduction in marginal zone B cells. J. Immunol. 171, 2783–2788 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Feng, J. et al. IFN regulatory factor 8 restricts the size of the marginal zone and follicular B cell pools. J. Immunol. 186, 1458–1466 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Hart, G.T., Wang, X., Hogquist, K.A. & Jameson, S.C. Krüppel-like factor 2 (KLF2) regulates B-cell reactivity, subset differentiation, and trafficking molecule expression. Proc. Natl. Acad. Sci. USA 2, 1–6 (2010).

    Google Scholar 

  15. Winkelmann, R. et al. B cell homeostasis and plasma cell homing controlled by Krüppel-like factor 2. Proc. Natl. Acad. Sci. USA 108, 710–715 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Vu, T.T. et al. Impaired B cell development in the absence of Krüppel-like factor 3. J. Immunol. 187, 5032–5042 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Clipson, A. et al. KLF2 mutation is the most frequent somatic change in splenic marginal zone lymphoma and identifies a subset with distinct genotype. Leukemia 29, 1177–1185 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Turner, M., Galloway, A. & Vigorito, E. Noncoding RNA and its associated proteins as regulatory elements of the immune system. Nat. Immunol. 15, 484–491 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Belver, L., de Yébenes, V.G. & Ramiro, A.R. MicroRNAs prevent the generation of autoreactive antibodies. Immunity 33, 713–722 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kramer, N.J. et al. Altered lymphopoiesis and immunodeficiency in miR-142 null mice. Blood 125, 3720–3730 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Yuan, J., Nguyen, C.K., Liu, X., Kanellopoulou, C. & Muljo, S.A. Lin28b reprograms adult bone marrow hematopoietic progenitors to mediate fetal-like lymphopoiesis. Science 335, 1195–1200 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhou, Y. et al. Lin28b promotes fetal B lymphopoiesis through the transcription factor Arid3a. J. Exp. Med. 212, 569–580 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brooks, S.A. & Blackshear, P.J. Tristetraprolin (TTP): interactions with mRNA and proteins, and current thoughts on mechanisms of action. Biochim. Biophys. Acta 1829, 666–679 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hodson, D.J. et al. Deletion of the RNA-binding proteins ZFP36L1 and ZFP36L2 leads to perturbed thymic development and T lymphoblastic leukemia. Nat. Immunol. 11, 717–724 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Galloway, A. et al. RNA-binding proteins ZFP36L1 and ZFP36L2 promote cell quiescence. Science 352, 453–459 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Kwon, K. et al. Instructive role of the transcription factor E2A in early B lymphopoiesis and germinal center B cell development. Immunity 28, 751–762 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Bell, S.E. et al. The RNA binding protein Zfp36l1 is required for normal vascularisation and post-transcriptionally regulates VEGF expression. Dev. Dyn. 235, 3144–3155 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Stumpo, D.J. et al. Chorioallantoic fusion defects and embryonic lethality resulting from disruption of Zfp36L1, a gene encoding a CCCH tandem zinc finger protein of the Tristetraprolin family. Mol. Cell. Biol. 24, 6445–6455 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Allman, D. et al. Resolution of three nonproliferative immature splenic B cell subsets reveals multiple selection points during peripheral B cell maturation. J. Immunol. 167, 6834–6840 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Vogel, K.U., Bell, L.S., Galloway, A., Ahlfors, H. & Turner, M. The RNA-binding proteins Zfp36l1 and Zfp36l2 enforce the thymic β-selection checkpoint by limiting DNA damage response signaling and cell cycle progression. J. Immunol. 197, 2673–2685 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Oliver, A.M., Martin, F., Gartland, G.L., Carter, R.H. & Kearney, J.F. Marginal zone B cells exhibit unique activation, proliferative and immunoglobulin secretory responses. Eur. J. Immunol. 27, 2366–2374 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Leadbetter, E.A. et al. NK T cells provide lipid antigen-specific cognate help for B cells. Proc. Natl. Acad. Sci. USA 105, 8339–8344 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Grajales-Reyes, G.E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8α+ conventional DC clonogenic progenitor. Nat. Immunol. 16, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Winkelmann, R., Sandrock, L., Kirberg, J., Jäck, H.M. & Schuh, W. KLF2--a negative regulator of pre-B cell clonal expansion and B cell activation. PLoS One 9, e97953 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Yeo, J.C. et al. Klf2 is an essential factor that sustains ground state pluripotency. Cell Stem Cell 14, 864–872 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Revilla-i-Domingo, R. et al. The B-cell identity factor Pax5 regulates distinct transcriptional programmes in early and late B lymphopoiesis. EMBO J. 31, 3130–3146 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Keene, J.D. RNA regulons: coordination of post-transcriptional events. Nat. Rev. Genet. 8, 533–543 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Kaymak, E. & Ryder, S.P. RNA recognition by the Caenorhabditis elegans oocyte maturation determinant OMA-1. J. Biol. Chem. 288, 30463–30472 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Farley, B.M. & Ryder, S.P. POS-1 and GLD-1 repress glp-1 translation through a conserved binding-site cluster. Mol. Biol. Cell 23, 4473–4483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tamburino, A.M., Ryder, S.P. & Walhout, A.J.M. A compendium of Caenorhabditis elegans RNA binding proteins predicts extensive regulation at multiple levels. G3-Genes Genom. Genet. 3, 297–304 (2013).

    CAS  Google Scholar 

  41. Hoek, K.L. et al. Follicular B cell trafficking within the spleen actively restricts humoral immune responses. Immunity 33, 254–265 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Santio, N.M. et al. Pim kinases promote migration and metastatic growth of prostate cancer xenografts. PLoS One 10, e0130340 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Grundler, R. et al. Dissection of PIM serine/threonine kinases in FLT3-ITD-induced leukemogenesis reveals PIM1 as regulator of CXCL12-CXCR4-mediated homing and migration. J. Exp. Med. 206, 1957–1970 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jain, M., Zhang, L., Patterson, E.E. & Kebebew, E. KIAA0101 is overexpressed, and promotes growth and invasion in adrenal cancer. PLoS One 6, e26866 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Aranda, J.F. et al. MYADM regulates Rac1 targeting to ordered membranes required for cell spreading and migration. Mol. Biol. Cell 22, 1252–1262 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Aranda, J.F. et al. MYADM controls endothelial barrier function through ERM-dependent regulation of ICAM-1 expression. Mol. Biol. Cell 24, 483–494 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Devis, L. et al. Activated leukocyte cell adhesion molecule (ALCAM) is a marker of recurrence and promotes cell migration, invasion and metastasis in early stage endometrioid endometrial cancer. J. Pathol. 241, 475–487 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Nalvarte, I., Damdimopoulos, A.E., Rüegg, J. & Spyrou, G. The expression and activity of thioredoxin reductase 1 splice variants v1 and v2 regulate the expression of genes associated with differentiation and adhesion. Biosci. Rep. 35, e00269 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Hsiao, S.P. & Chen, S.L. Myogenic regulatory factors regulate M-cadherin expression by targeting its proximal promoter elements. Biochem. J. 428, 223–233 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Reginato, M.J. et al. Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat. Cell Biol. 5, 733–740 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Hobeika, E. et al. Testing gene function early in the B cell lineage in mb1-cre mice. Proc. Natl. Acad. Sci. USA 103, 13789–13794 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. de Luca, C. et al. Complete rescue of obesity, diabetes, and infertility in db/db mice by neuron-specific LEPR-B transgenes. J. Clin. Invest. 115, 3484–3493 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bouillet, P. et al. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 286, 1735–1738 (1999).

    CAS  PubMed  Google Scholar 

  55. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Anders, S., Pyl, P.T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  57. 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  CAS  Google Scholar 

  58. Robinson, J.T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Thorvaldsdóttir, H., Robinson, J.T. & Mesirov, J.P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  PubMed  CAS  Google Scholar 

  60. Chen, J., Bardes, E.E., Aronow, B.J. & Jegga, A.G. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 37, W305–W311 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Supek, F., Bošnjak, M., Škunca, N. & Šmuc, T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS One 6, e21800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gautier, L., Cope, L., Bolstad, B.M. & Irizarry, R.A. affy--analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20, 307–315 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Busslinger (Research Institute of Molecular Pathology) for the mice expressing CD23-Cre; M. Reth (Max Planck Institute for Immunobiology and Epigenetics) for the mice expressing Mb1-Cre; T. Ludwig (Columbia University) for the mice expressing R26ERT2−Cre; and D. Kioussis (National Institute for Medical Research) for the mice expressing CD2-iCre; K. Bates, R. Walker, A. Davies and L. Waugh and members of the Biological Services Unit for technical support; D. Bell and P. Tolar for help with immunofluorescence analysis; and members of the Turner laboratory, L. Webb, M. Linterman and T. Arnon for advice. Supported by a GlaxoSmithKline-CASE studentship, the Biotechnology and Biological Sciences Research Council, The Medical Research Council and Bloodwise.

Author information

Authors and Affiliations

Authors

Contributions

R.N. designed and did most experiments; H.A. performed bioinformatics analysis; A.S. performed iCLIP analysis; A.G., D.J.H., R.W. and S.E.B. helped with mouse experiments; G.S.B. provided NP–α-GalCer; C.N.C. and A.F.C. performed some histology; and R.N. and M.T. planned the project and wrote the manuscript.

Corresponding author

Correspondence to Martin Turner.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Targeting strategy for the generation of Zfp36fl/fl mice.

(a) The Zfp36 gene consists of two exons (grey boxes). The targeting vector was designed to introduce loxP sites flanking exon 2, which contains the tandem zinc finger RNA binding domain, and a Neomycin selection cassette (neo) flanked by two FRT sites. The targeting construct was introduced into the Zfp36 locus of BRUCE4 embryonic stem cells (C57BL/6 origin) by homologous recombination. Positive clones were identified via Southern blotting confirming correct targeting at both 5’ and 3’ ends. Subsequent crossing of mice possessing the targeted allele to FlpE mice, removing the neo cassette generated the desired Zfp36fl/fl mice. The same gene targeting strategy was used to produce Zfp36l1fl/fl and Zfp36l2fl/fl mice (b) Introduction of the CD2-iCre transgene into homozygous Zfp36fl/fl mice caused a tissue specific deletion of exon 2, which was confirmed by Southern blotting.

Supplementary Figure 2 Peritoneal cavity B-1 cells require ZFP36L1, and the GFP–ZFP36L1 fusion protein is expressed from the transitional 2 B cell stage in R26GFPZFP36L1Cd23-Cre+ mice.

(a) Flow cytometry analysis of peritoneal cavity B cells from Zfp36l1fl/flMb1Cre/+ and Zfp36l1fl/flMb1+/+ mice. Numbers indicate percentage of cells within gate. Data represent at least 8 mice per genotype. (b) Number of B220loCD19+ B1 B cells and B220+CD19lo B2 B cells, in the peritoneal cavity as assessed by flow cytometry. (c) Flow cytometry analysis of peritoneal cavity B cells from Zfp36l1fl/flMb1Cre/+ and Zfp36l1fl/flMb1+/+ mice. Numbers indicate percentage of cells within gate. Dot plots are pre-gated B220intCD19hi cells. Data represent at least 8 mice per genotype. (d) Number of B220loCD19+CD5+IgM+ B1a B cells and B220loCD19+CD5-IgM+ B1b B cells, in the peritoneal cavity as assessed by flow cytometry. (e) Flow cytometry analysis of GFP-ZFP36L1 fusion protein expression in splenic B cell subsets from R26GFPZFP36L1Cd23-Cre+mice. Sample sizes: b, d (Zfp36l1fl/flMb1+/+ 10, Zfp36l1fl/flMb1Cre/+ 8). *P ≤ 0.05, **P ≤ 0.01 Mann-Whitney. Data pooled from 2 experiments (b, d; mean).

Supplementary Figure 3 MZ B cells require ZFP36L1 to persist over time.

(a) qPCR analysis showing relative Zfp36l1 exon 2 abundance in purified B cells from day 7 post tamoxifen treatment of Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/+ mice. (b) Absolute numbers of MZ B cells Fo B cells assessed by flow cytometry in mice as described in a. (c) qPCR analysis showing relative Zfp36l1 exon 2 abundance in purified B cells from day 10 post tamoxifen treatment of Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/+ mice. (d) Absolute numbers of MZ B cells Fo B cells assessed by flow cytometry in mice as described in c. (e) qPCR analysis showing relative Zfp36l1 exon 2 abundance in purified B cells from day 14 post tamoxifen treatment of Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/+ mice. (f) Absolute numbers of MZ B cells Fo B cells assessed by flow cytometry in mice as described in e. Sample sizes: a-f (4). *P ≤ 0.05, Mann-Whitney. Data from 1 experiment (a, c, e; mean + s.e.m.), data from 1 experiment (b, d, f; mean).

Supplementary Figure 4 ZFP36L1 controls turnover of MZ B cells.

(a) BrdU incorporation into splenic B cell subsets in Zfp36lfl/flMb1Cre/+ and Zfp36l1fl/flMb1+/+ mice fed BrdU in water for 14 days (b) BrdU incorporation into splenic B cell subsets from R26GFPZFP36L1Cd23-Cre+ and R26GFPZFP36L1Cd23-Cre- mice fed BrdU in water for 14 days. Sample sizes: a (Zfp36l1fl/flMb1+/+ 10, Zfp36lfl/flMb1Cre/+ 9), b (R26GFPZFP36L1Cd23-Cre- 4, R26GFPZFP36L1Cd23-Cre+ 6). *P ≤ 0.05, **P ≤ 0.01, Mann-Whitney. Data pooled from 2 independent experiments (a; mean), data from 1 experiment (b; mean).

Supplementary Figure 5 Validation of RNA-seq analysis and top regulated transcripts.

(a) Reads mapping to Zfp36l1. Reads were mapped using TopHat, and are displayed using IGV. Enrichment of uniquely aligning deduplicated reads shown in grey. The top 4 rows show reads in control (Zfp36l1+/+R26ERT2-Cre/ERT2-Cre) replicates, and the bottom 4 rows show reads in KO (Zfp36l1fl/flR26ERT2-Cre/+) replicates. (b) Normalised read counts for Zfp36 and Zfp36l2 in MZ B cells from tamoxifen treated Zfp36l1+/+R26ERT2-Cre/ERT2-Cre and Zfp36l1fl/flR26ERT2-Cre/+ mice. (c) A clustered heatmap showing the log2 transformed RPKM values of 50 most regulated transcripts based on their log2FoldChange in the comparison of Zfp36l1fl/flR26ERT2-Cre/+ MZ B cells to their Zfp36l1+/+R26ERT2-Cre/ERT2-Cre controls. Blue color indicates low, yellow medium and red high expressed genes. Dendrogram shows clustering of the genotypes. Sample sizes: a, b (4). Data from 1 experiment (b; mean + s.e.m.)

Supplementary Figure 6 Normal expression of p27 and Ki67 and DAPI profiles in ZFP36L1-deficient MZ B cells.

(a) Flow cytometry analysis showing p27 staining in MZ B cells from Zfp36l1fl/flMb1Cre/+ mice and Zfp36l1fl/flMb1+/+ controls (left panel) and tamoxifen treated Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/+ control chimeras (right panel). Isotype control sample is shown for both sets of analysis. (b, c) Flow cytometry analysis showing the proportion of p27+ MZ B cells in Zfp36l1fl/flMb1+/+ and Zfp36l1fl/flMb1Cre/+ mice (b), tamoxifen treated Zfp36l1+/+R26ERT2-Cre/+ and Zfp36l1fl/flR26ERT2-Cre/+ chimeric mice (c). (d) Flow cytometry analysis showing Ki67 staining in MZ, Fo and GC B cells from Zfp36l1fl/flMb1Cre/+ mice and Zfp36l1fl/flMb1+/+ littermate controls. FMO control sample is shown for all sets of analysis. (e) Proportion of Ki67+ cells in Zfp36l1fl/flMb1Cre/+ and Zfp36l1fl/flMb1+/+. (g) DAPI staining in MZ and Fo B cells from Zfp36l1fl/flMb1Cre/+ mice and Zfp36l1fl/flMb1+/+ littermate controls. Numbers on the graph indicate the percentage of cells in the gate. (h, i) Proportion of cells in the S/G2-M phase of the cell cycle in MZ (h) and Fo B cells (i). Sample sizes: b (Zfp36l1fl/flMb1+/+ 9, Zfp36l1fl/flMb1cre/+ 7), c (Zfp36l1+/+R26ERT2-Cre/+ 6, Zfp36l1fl/flR26ERT2-Cre/+ 8), e, h, i (5) *P ≤ 0.05, Mann-Whitney. Data pooled from 2 independent experiments (b; mean), data from 1 experiment (c, e, h, i; mean).

Supplementary Figure 7 ZFP36L1 regulates localization, identity and survival.

(a) Immunofluorescence analysis of spleen sections from Zfp36l1fl/flR26ERT2-Cre/+ mice and R26GFPZFP36L1Cd23-Cre+ mice. Sections were stained with anti-CD1d (red) and anti-IgD (green). Images were captured at 20x magnification, scale bars represent 50μm. White arrows on the R26GFPZFP36L1Cd23-Cre+ image indicate colocalization of the IgD and CD1d, white arrows on the Zfp36l1fl/flR26ERT2-Cre/+ image indicate single stained cells only. (b) Day 4 NP-specific IgM endpoint titres from mice immunized with NP-α-GalCer. (c) Schematic model depicting the mechanism by which ZFP36L1 regulates survival, migration and identity of MZ B cells through a network of transcription factors which include KLF2 and IRF8. Sample sizes: b (3), Mann-Whitney. Data pooled from 1 experiment (b; mean).

Supplementary Figure 8 ZFP36L1 controls genes that encode products involved in the trafficking of mature B cells.

(a) Venn diagram indicating the number of overlapping genes, which are increased in Zfp36l1fl/flR26ERT2-Cre/+ (KO) MZ B cells when compared to Zfp36l1+/+R26ERT2-Cre/ERT2-Cre (WT) MZ B cells, and decreased in R26GFPZFP36L1Cd23-Cre+ (GFP) Fo when compared to R26GFPZFP36L1Cd23-Cre- (WT) B cells using a padj<0.05 cutoff. (b) Pathway analysis of genes which are increased in expression in KO MZ B cells and decreased in expression in GFP expressing Fo B cells as defined in a using a padj<0.05 cutoff. (c, d) S1pr1 mRNA expression in Zfp36l1fl/flR26ERT2-Cre/+ MZ B cells and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre control MZ B cells (c), and R26GFPZFP36L1Cd23-Cre+ and R26GFPZFP36L1Cd23-Cre- Fo B cells (d). (e) Follicular area in image analysis was defined using CD169 (MOMA-1) staining (green). Follicular area is shown by yellow line. MFI was then calculated for this area. MZ area was defined as CD1d+ (magenta) area outside MOMA-1 (green) and IgD (not shown) staining. MZ area is shown by white line. Background fluorescence (grey box) was calculated and subtracted from MFI. Sample sizes: c, d (4), Benjamini Hochburg adjusted p values calculated using DESeq2. Data from 1 experiment (c, d; mean + s.e.m.).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 7–9 (PDF 1735 kb)

Supplementary Table 1

Differentially expressed genes in MZ B cells from Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice. Differentially expressed genes (P adj < 0.01) in comparisons of MZ B cells from tamoxifen-treated Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice. A 1.5-fold change cutoff has been applied. (XLSX 62 kb)

Supplementary Table 2

Upregulated genes (P adj < 0.01) for GSEA showing iCLIP hits and AREs. Genes that are increased in expression (P adj < 0.01) in comparisons of MZ B cells from tamoxifen-treated Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice. Hits in iCLIP analysis and ARE found in the 3′ UTR of the gene are indicated. An FDR of 0.05 was used for the iCLIP data. (XLSX 87 kb)

Supplementary Table 3

Downregulated genes (P adj < 0.01) for GSEA showing iCLIP hits and AREs. Genes that are decreased in expression (P adj < 0.01) in comparisons of MZ B cells from tamoxifen-treated Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice. Hits in iCLIP analysis and ARE found in the 3′ UTR of the gene are indicated. An FDR of 0.05 was used for the iCLIP data. (XLSX 75 kb)

Supplementary Table 4

Differentially expressed genes in MZ B cells from Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice compared to wild-type MZ and Fo B cells. Differentially expressed genes (P adj < 0.01, and subject to a 1.5-fold cutoff) in comparisons of Zfp36l1fl/flR26ERT2-Cre/+ (KO) MZ B cells and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre control (WT) MZ B cells to wild-type Fo B cell gene expression. Gene lists have been separated into each quadrant of the correlation dot plot. Zfp36l1 has been removed from this analysis. (XLSX 65 kb)

Supplementary Table 5

Differentially expressed genes in Fo B cells from R26GFPZFP36L1Cd23-Cre+ and R26GFPZFP36L1Cd23-Cre–versus wild-type MZ and Fo B cells. Differentially expressed genes (P adj < 0.01, and subject to a 1.5-fold cutoff) in comparisons of R26GFPZFP36L1Cd23-Cre+ (GFP) Fo B cells and R26GFPZFP36L1Cd23-Cre– control Fo B cells to wild-type MZ B cell gene expression. Gene lists have been separated into each quadrant of the correlation dot plot. Zfp36l1 has been removed from this analysis. (XLSX 57 kb)

Supplementary Table 6

Differentially expressed genes in comparisons of MZ B cells from Zfp36l1fl/flR26ERT2-Cre/+ and Zfp36l1+/+R26ERT2-Cre/ERT2-Cre mice to Fo B cells from R26GFPZFP36L1Cd23-Cre+ and R26GFPZFP36L1Cd23-Cre– mice. Differentially expressed genes (P adj < 0.05) found in RNA-seq analysis comparing MZ B cells from Zfp36l1fl/flR26ERT2-Cre/+ to Zfp36l1+/+R26ERT2-Cre/ERT2-Cre controls and RNA-seq analysis comparing GFP–ZFP36L1-positive Fo B cells to Fo B cells from littermate controls. Gene lists have been separated into each quadrant of the correlation dot plot. Zfp36l1 has been removed from this analysis. (XLSX 77 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Newman, R., Ahlfors, H., Saveliev, A. et al. Maintenance of the marginal-zone B cell compartment specifically requires the RNA-binding protein ZFP36L1. Nat Immunol 18, 683–693 (2017). https://doi.org/10.1038/ni.3724

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3724

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