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:

An essential role for the Zn2+ transporter ZIP7 in B cell development

Abstract

Despite the known importance of zinc for human immunity, molecular insights into its roles have remained limited. Here we report a novel autosomal recessive disease characterized by absent B cells, agammaglobulinemia and early onset infections in five unrelated families. The immunodeficiency results from hypomorphic mutations of SLC39A7, which encodes the endoplasmic reticulum-to-cytoplasm zinc transporter ZIP7. Using CRISPR-Cas9 mutagenesis we have precisely modeled ZIP7 deficiency in mice. Homozygosity for a null allele caused embryonic death, but hypomorphic alleles reproduced the block in B cell development seen in patients. B cells from mutant mice exhibited a diminished concentration of cytoplasmic free zinc, increased phosphatase activity and decreased phosphorylation of signaling molecules downstream of the pre-B cell and B cell receptors. Our findings highlight a specific role for cytosolic Zn2+ in modulating B cell receptor signal strength and positive selection.

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: A novel autosomal recessive agammaglobulinemia caused by mutations in ZIP7.
Fig. 2: Multiple loss-of-function mutations in ZIP7.
Fig. 3: Generation of an allelic series of ZIP7 mutant mice.
Fig. 4: ZIP7 deficiency leads to a B cell-intrinsic failure in development.
Fig. 5: ZIP7 deficiency leads to developmental arrest at the late pre-B to immature B cell transition.
Fig. 6: Reduced cytoplasmic zinc in the presence of ZIP7 mutation.
Fig. 7: Impaired ZIP7 function results in reduced BCR signaling.
Fig. 8: ZIP7-dependent inhibition of B cell phosphatase activity

Similar content being viewed by others

Data availability

RNA-sequencing data generated for this study (Fig. 5 and Supplementary Figs. 4 and 5) have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE108178. Other data that support the findings of this study (including raw data supporting Fig. 1 and Supplementary Fig. 1) are available from the corresponding authors upon reasonable request.

References

  1. Vetrie, D. et al. The gene involved in X-linked agammaglobulinaemia is a member of the SRC family of protein-tyrosine kinases. Nature 361, 226–233 (1993).

    Article  CAS  Google Scholar 

  2. Tsukada, S. et al. Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia. Cell 72, 279–290 (1993).

    Article  CAS  Google Scholar 

  3. Aw, A. & Brown, J. R. Current status of Bruton’s tyrosine kinase inhibitor development and use in B-cell malignancies. Drugs Aging 34, 509–527 (2017).

    Article  CAS  Google Scholar 

  4. Conley, M. E. Genetics of hypogammaglobulinemia: what do we really know? Curr. Opin. Immunol. 21, 466–471 (2009).

    Article  CAS  Google Scholar 

  5. Durandy, A., Kracker, S. & Fischer, A. Primary antibody deficiencies. Nat. Rev. Immunol. 13, 519–533 (2013).

    Article  CAS  Google Scholar 

  6. Conley, M. E. et al. Agammaglobulinemia and absent B lineage cells in a patient lacking the p85alpha subunit of PI3K. J. Exp. Med. 209, 463–470 (2012).

    Article  CAS  Google Scholar 

  7. Feske, S. et al. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 441, 179–185 (2006).

    Article  CAS  Google Scholar 

  8. Li, F. Y. et al. Second messenger role for Mg2 + revealed by human T-cell immunodeficiency. Nature 475, 471–476 (2011).

    Article  CAS  Google Scholar 

  9. Kambe, T., Hashimoto, A. & Fujimoto, S. Current understanding of ZIP and ZnT zinc transporters in human health and diseases. Cell. Mol. Life Sci. 71, 3281–3295 (2014).

    Article  CAS  Google Scholar 

  10. Lichten, L. A. & Cousins, R. J. Mammalian zinc transporters: nutritional and physiologic regulation. Annu. Rev. Nutr. 29, 153–176 (2009).

    Article  Google Scholar 

  11. Hojyo, S. et al. Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength. Proc. Natl Acad. Sci. USA 111, 11786–11791 (2014).

    Article  CAS  Google Scholar 

  12. Taylor, K. M. et al. Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters. Biochem. J. 377(Pt 1), 131–139 (2004).

    Article  CAS  Google Scholar 

  13. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  CAS  Google Scholar 

  14. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014).

    Article  CAS  Google Scholar 

  15. Zhang, T. et al. Crystal structures of a ZIP zinc transporter reveal a binuclear metal center in the transport pathway. Sci. Adv. 3, e1700344 (2017).

    Article  Google Scholar 

  16. Vinkenborg, J. L. et al. Genetically encoded FRET sensors to monitor intracellular Zn2+ homeostasis. Nat. Methods 6, 737–740 (2009).

    Article  CAS  Google Scholar 

  17. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

  18. Hardy, R. R. et al. Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J. Exp. Med. 173, 1213–1225 (1991).

    Article  CAS  Google Scholar 

  19. Dengler, H. S. et al. Distinct functions for the transcription factor Foxo1 at various stages of B cell differentiation. Nat. Immunol. 9, 1388–1398 (2008).

    Article  CAS  Google Scholar 

  20. Tussiwand, R. et al. BAFF-R expression correlates with positive selection of immature B cells. Eur. J. Immunol. 42, 206–216 (2012).

    Article  CAS  Google Scholar 

  21. Holl, T. M., Haynes, B. F. & Kelsoe, G. Stromal cell independent B cell development in vitro: generation and recovery of autoreactive clones. J. Immunol. Methods 354, 53–67 (2010).

    Article  CAS  Google Scholar 

  22. Corfe, S. A., Gray, A. P. & Paige, C. J. Generation and characterization of stromal cell independent IL-7 dependent B cell lines. J. Immunol. Methods 325, 9–19 (2007).

    Article  CAS  Google Scholar 

  23. Chabosseau, P. et al. Mitochondrial and ER-targeted eCALWY probes reveal high levels of free Zn2+. ACS. Chem. Biol. 9, 2111–2120 (2014).

    Article  CAS  Google Scholar 

  24. Maares, M. et al. Characterization of Caco-2 cells stably expressing the protein-based zinc probe eCalwy-5 as a model system for investigating intestinal zinc transport. J. Trace Elem. Med. Biol. 49, 296–304 (2018).

    Article  CAS  Google Scholar 

  25. Ohashi, W. et al. Zinc transporter SLC39A7/ZIP7 promotes intestinal epithelial self-renewal by resolving ER stress. PLoS Genet. 12, e1006349 (2016).

    Article  Google Scholar 

  26. Bin, B. H. et al. Requirement of zinc transporter SLC39A7/ZIP7 for dermal development to fine-tune endoplasmic reticulum function by regulating protein disulfide isomerase. J. Invest. Dermatol. 137, 1682–1691 (2017).

    Article  CAS  Google Scholar 

  27. Strasser, A. et al. Bcl-2 expression promotes B- but not T-lymphoid development in SCID mice. Nature 368, 457 (1994).

    Article  CAS  Google Scholar 

  28. Reth, M. & Nielsen, P. Signaling circuits in early B-cell development. Adv. Immunol. 122, 129–175 (2014).

    Article  CAS  Google Scholar 

  29. Levine, M. H. et al. A B-cell receptor-specific selection step governs immature to mature B cell differentiation. Proc. Natl Acad. Sci. USA 97, 2743–2748 (2000).

    Article  CAS  Google Scholar 

  30. Phan, T. G. et al. B cell receptor-independent stimuli trigger immunoglobulin (Ig) class switch recombination and production of IgG autoantibodies by anergic self-reactive B cells. J. Exp. Med. 197, 845–860 (2003).

    Article  CAS  Google Scholar 

  31. Brautigan, D. L., Bornstein, P. & Gallis, B. Phosphotyrosyl-protein phosphatase. Specific inhibition by Zn. J. Biol. Chem. 256, 6519–6522 (1981).

    CAS  PubMed  Google Scholar 

  32. Haase, H. & Maret, W. Intracellular zinc fluctuations modulate protein tyrosine phosphatase activity in insulin/insulin-like growth factor-1 signaling. Exp. Cell Res. 291, 289–298 (2003).

    Article  CAS  Google Scholar 

  33. Plum, L. M. et al. PTEN-inhibition by zinc ions augments interleukin-2-mediated Akt phosphorylation. Metallomics 6, 1277–1287 (2014).

    Article  CAS  Google Scholar 

  34. Kury, S. et al. Identification of SLC39A4, a gene involved in acrodermatitis enteropathica. Nat. Genet. 31, 239–240 (2002).

    Article  Google Scholar 

  35. Fraker, P. J. & King, L. E. Reprogramming of the immune system during zinc deficiency. Annu. Rev. Nutr. 24, 277–298 (2004).

    Article  CAS  Google Scholar 

  36. Woodruff, G. et al. The zinc iransporter SLC39A7 (ZIP7) is ssential for regulation of cytosolic zinc levels. Mol. Pharmacol. 94, 1092–1100 (2018).

    Article  CAS  Google Scholar 

  37. Groth, C. et al. Protein trafficking abnormalities in Drosophila tissues with impaired activity of the ZIP7 zinc transporter Catsup. Development 140, 3018–3027 (2013).

    Article  CAS  Google Scholar 

  38. Yan, G. et al. Slc39a7/zip7 plays a critical role in development and zinc homeostasis in zebrafish. PLoS ONE 7, e42939 (2012).

    Article  CAS  Google Scholar 

  39. Taniguchi, M. et al. Essential role of the zinc transporter ZIP9/SLC39A9 in regulating the activations of Akt and Erk in B-cell receptor signaling pathway in DT40 cells. PLoS ONE 8, e58022 (2013).

    Article  CAS  Google Scholar 

  40. Ubieta, K. et al. Fra-2 regulates B cell development by enhancing IRF4 and Foxo1 transcription. J. Exp. Med. 214, 2059–2071 (2017).

    Article  CAS  Google Scholar 

  41. Kerdiles, Y. M. et al. Foxo1 links homing and survival of naive T cells by regulating L-selectin, CCR7 and interleukin 7 receptor. Nat. Immunol. 10, 176–184 (2009).

    Article  CAS  Google Scholar 

  42. Geiser, J. et al. Clioquinol synergistically augments rescue by zinc supplementation in a mouse model of acrodermatitis enteropathica. PLoS ONE 8, e72543 (2013).

    Article  CAS  Google Scholar 

  43. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  Google Scholar 

  44. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  45. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

  46. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  Google Scholar 

  47. Sims, D. et al. CGAT: computational genomics analysis toolkit. Bioinformatics 30, 1290–1291 (2014).

    Article  CAS  Google Scholar 

  48. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  Google Scholar 

  49. Liao, Y., Smyth, G. K. & Shi, W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  Google Scholar 

  50. Holl, T. M., Haynes, B. F. & Kelsoe, G. Stromal cell-independent B-cell development in vitro: generation and recovery of autoreactive clones. J Immunol Methods. 354, 53–67 (2010).

    Article  Google Scholar 

  51. Hessels, A. M., Taylor, K. M. & Merkx, M. Monitoring cytosolic and ER Zn(2+) in stimulated breast cancer cells using genetically encoded FRET sensors. Metallomics 8, 211–217 (2016).

    Article  CAS  Google Scholar 

  52. Padilla-Parra, S. et al. Quantitative FRET analysis by fast acquisition time domain FLIM at high spatial resolution in living cells. Biophys. J. 95, 2976–2988 (2008).

    Article  CAS  Google Scholar 

  53. Merola, F. et al. Newly engineered cyan fluorescent proteins with enhanced performances for live cell FRET imaging. Biotechnol. J. 9, 180–191 (2014).

    Article  CAS  Google Scholar 

  54. Markovich, D. Expression cloning and radiotracer uptakes in Xenopus laevis oocytes. Nat. Protoc. 3, 1975–1980 (2008).

    Article  CAS  Google Scholar 

  55. Turk, E. et al. Membrane topology of the human Na+ /glucose cotransporter SGLT1. J. Biol. Chem. 271, 1925–1934 (1996).

    Article  CAS  Google Scholar 

  56. Yoshida, H. et al. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881–891 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank colleagues in the Newcastle University Flow Cytometry and Bioimaging Facilities for assistance. We acknowledge N. Ashley, A. Mead, P. Sopp and C. Waugh for assistance with single cell experiments and flow cytometry, D. Biggs and C. Preece for generation of the mouse models and staff at the Oxford Functional Genomics Facility for animal care. We also thank the National Diagnostic Epidermolysis Bullosa Laboratory (St Thomas’ Hospital, London) and the NIHR Newcastle Biomedical Research Centre. We thank K. Taylor for helpful discussions. This work was supported by the Medical Research Council (MR/J0003042/1, MR/N00275X/1 and MR/L020149/1: DIVA) (C.A., R.J.C., E.F., J.C.C. and G.A.R.); the Sir Jules Thorn Trust (12/JTA) (S.H., D.S., T.S.D. and K.E.); the St Giles Foundation, the Rockefeller University, INSERM, Paris Descartes University, Howard Hughes Medical Institute, National Institutes of Health (5P01AI061093 and 5R01AI104857) and the French National Research Agency (ANR 14-CE15-0009-01) (B.B., S.J.d.J., J.-L.C. and M.E.C.); the Wellcome Trust (WT098424AIA; 090532/Z/09/Z and 207556/Z/17/Z) (P.C., G.A.R., J.R.C., S.P.-P., B.D. and S.H.); Cancer Research UK (C52690/A19270) (C.O. and J.R.C.); Diabetes UK (BDA11/0004210 and BDA/15/0005275) (P.C., G.A.R.); the Northern Counties Kidney Research Fund (14.06) (A.F. and A.W.); the National Health and Medical Research Council of Australia (C.S.M., S.G.T.) and the Ludwig Institute for Cancer Research (E.J.F. and J.C.C.). S.H. is a Wellcome Investigator and R.J.C. is a Principal Investigator of the MRC Human Immunology Unit.

Author information

Authors and Affiliations

Authors

Contributions

C.A. designed and performed experiments, analyzed data and wrote the paper. R.J.C., M.E.C. and S.H. designed experiments, analyzed data and wrote the paper. D.J.S., B.B., T.S.D., M.D.-L., C.O., B.C.L., L.A., R.C., R.B.-P., P.C., M.v.d.B., B.D., A.W. and S.P.-P. designed and performed experiments and analyzed data. K.R.E., J.R.C., J.C.C., G.A.R., S.G.T., J.A.M., S.P. and J.-L.C. designed experiments and analyzed data. B.B., B.v.d.B., K.R.B., A.P.C., D.S., X.X., Y.X. and M.S.K. performed bioinformatic analysis. E.C., T.L.C., A.F., E.J.F., S.J.d.J. and C.S.M. performed experiments. A.J.C., G.K., T.R.L., M.T.d.l.M., J.M.P., R.S.S., M.E.C. and S.H. cared for patients and provided clinical data. All authors reviewed the manuscript

Corresponding authors

Correspondence to Richard J. Cornall, Mary Ellen Conley or Sophie Hambleton.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Allele frequency and predicted severity and evolutionary conservation of mutated residues in SLC39A7.

(a) Population frequency of putative pathogenic alleles of SLC39A7 (ZIP7) from the GNOMAD database13. (b) Combined Annotation Dependent Depletion (CADD)14 scores vs maximum mean allele frequencies of the SLC39A7 variants recorded in this study (red circles) and/or GNOMAD Database (blue circles for predicted null mutations (frameshift/stop/essential splicing), green circle (in-frame insertions and deletions) or gray circle (missense mutations)). None of the predicted null mutations or Inframe INDELS were found in the homozygous state. (c) Alignments of human ZIP7, mouse ZIP7 and Bordetella pertussis ZIP, generated as two pairwise alignments with human ZIP7 in BLAST-P, highlighting residues mutated in patients with agammaglobulinemia (missense, yellow; nonsense, pink) and predicted transmembrane segments (gray)15.

Supplementary Figure 2 Mutant forms of ZIP7 are expressed but display reduced Zn transporter activity.

(a) Fluorescence micrographs revealing expression of endogenous ZIP7 in primary dermal fibroblasts from healthy control or P1, stained with antibody against ZIP7 (red) and DAPI (blue). Scale bar, 20µm. (b) Impaired Zn2+ conductance of mutant forms of ZIP7 expressed in Xenopus oocytes, visualized by zinquin fluorescence. Left, fluorescence micrographs showing Zn2+-related zinquin signal and right, pairwise image analysis in ImageJ, as described in Methods. Images are representative of 4 independent experiments as exemplified in Fig 2h. (c) Western blot of detergent extracts of Xenopus oocytes injected in parallel with those visualized in (b), revealing expression of the recombinant ZIP7 proteins. Images are representative of 3 independent experiments. (d-e) Cytoplasmic Zn2+ concentration in HEK-293T cells stably expressing the genetically encoded cytoplasmic Zn2+ sensor eCALWY-4 and transfected with empty vector (EV), or vectors encoding WT, E363K (d) or P190A (e) ZIP7 proteins. Cytoplasmic Zn2+ concentration was calculated by comparing the steady state live cell fluorescence intensity with maximum and minimum signals obtained in the presence of TPEN and zinc pyrithione respectively, as described in Methods. Total number of cells analyzed was 80 (EV), 55 (WT), 26 (E363K) and 42 (P190A) across 2–4 experiments. Columns show means and bars the standard error. Comparisons were by one-way ANOVA with Bonferroni’s correction; * indicates p=0.0258 and ** p=0.0051.

Supplementary Figure 3 Normal T cell development in ZIP7 P198A/P198A mice.

(a-b) Representative flow cytometry of T cell subsets in the thymus (a) and spleen (b) of WT and ZIP7P198A/P198A (P198A-Hom) mice. Representative of 3 independent experiments (c-d) The percentage of B220+ B cells, naïve (CD62L+CD44-), activated (CD62L-CD44+) and memory (CD62L+CD44+) CD4+ and CD8+ T cells, NK cells (NK1.1+CD3-), neutrophils (B220-CD3-Ly6g+CD11b+Ly6c+) and monocytes (B220-CD3-Ly6c++F4/80-) within the leukocyte populations in wild-type (closed circles) and P198A-Hom (open circles) blood (c) and spleen (d). Representative of three separate experiments; circles represent individual mice, 5 WT and 5 P198A-Hom; bars means and 95% CI, and comparisons by two-way ANOVA with Bonferroni’s correction for multiple comparison; *p<0.0001. (e) The relative proportion of thymic or splenic CD4+ and CD8+ cells, in lethally irradiated mice reconstituted for 8 weeks with 70:30 mixtures of WT or P198A-Hom CD45.2+ and WT CD45.1+ BM. Filled columns show mean percentage CD45.2+ cells and data are representative of 3 experiments.

Supplementary Figure 4 Transcriptomic analysis of WT and ZIP7-deficient B cells from Hardy fraction B.

B220+CD43+CD24+BP1- pro-B cells were sorted flow-cytometrically and whole transcriptome analysis performed by RNAseq on 100 cell samples from WT and P198A-Hom mice. We identified genes whose transcription varied by at least two-fold between WT and P198A-Hom mice with Padj < 0.05 (see Methods). (a-c) Heatmaps show the relative expression level of the subset of these genes that are also differentially expressed during normal B cell development: (a) during the transition from Fractions A to B (reduced, purple and raised, blue), (b) during the transition from Fractions B to C (reduced, red and raised, green) and (c) between Fractions B and D (reduced, yellow and raised, orange), based on the ImmGen database (www.immGen.org). In each heatmap, the far left column indicates the direction of change expected from ImmGen; each of the remaining columns represents one mouse and rows individual transcripts.

Supplementary Figure 5 Transcriptomic analysis of WT and P198A-Hom B cells from Hardy fractions D and E.

B220+CD43-IgM-IgD- late pre-B (FrD) and B220+CD43-IgM+IgD- immature B cells (FrE) were flow-sorted and whole transcriptome analysis performed by RNA-seq on 100 cell samples from WT and P198A-Hom mice. We identified genes whose transcription varied by at least two-fold between WT and P198A-Hom mice with Padj < 0.05 (see Methods) and filtered these against sets of genes that are also differentially expressed at the indicated stages of normal B cell development (www.immGen.org). (a-b) Heatmaps showing the relative expression of the subset of genes that show differential expression between wild-type and P198A-Hom B cells in Hardy Fraction D and also (a) between Fractions C and D (reduced, yellow and raised, red) and (b) between Fractions D and E (reduced, green and raised, blue) of normal mice. (c) Heatmaps showing the relative expression level of the subset of genes that show differential expression between wild-type and P198A-Hom B cells in Hardy Fraction E and also between Fractions D and E (reduced, green and raised, blue) of normal mice. In each heatmap, the far left column indicates the direction of change expected from ImmGen; each of the remaining columns represents one mouse and rows individual transcripts.

Supplementary Figure 6 The B cell developmental arrest caused by ZIP7 deficiency is not associated with endoplasmic reticulum (ER) stress and is not corrected by expression of the pro-survival factor BCL2.

(a) RT-PCR assay for the ER-stress-associated shortened Xbp1-splice variant (Xbp1s). Results show RT-PCR of cDNA from WT (lanes 1–3) or P198A-Hom (lanes 4–6) flow-sorted B cells from FrD and FrE. The controls were tunicamycin treated (Tm) or untreated (Un) mouse embryonic fibroblasts (MEFs). Three mice were studied per genotype in the single experiment shown. (b-c) Representative flow cytometry analysis of B cell development and maturation in BM (b) and spleen (c) of WT and P198A-Hom mice, with and without co-expression of a Bcl2 transgene (upper and lower panels). Gates corresponding to Fr A-F in the BM and total (B220+CD19+), follicular (Fo, CD23+CD21+) and Marginal Zone (MZ, CD23-CD21hi) B cells in the spleen are highlighted. (d-e) Absolute numbers of B cells, gated as in (b-c), in Hardy Fractions in the BM (d) and in splenic subsets (e) from WT, P198A-Hom, WT BCL2 transgenic (BCL2) and P198A-Hom BCL2 transgenic (P198A-Hom BCL2) mice, here n=3 per group and representative of 4 independent experiments. Bars show means and 95% CI; statistical comparison was by two-way ANOVA with Bonferroni correction for multiple comparison, * =p<0.0001.

Supplementary Figure 7 Normal TCR signaling and threshold for activation ZIP7-deficient thymocytes.

(a-c) Mean phospho-specific antibody binding to indicated intracellular signaling molecules downstream of the TCR, 5 min after stimulation of WT (closed circles) and P198A-Hom (open circles) thymocytes with anti-CD3, gated on CD4+CD8+ (a), CD4+ (b) and CD8+ cells (c). (d-f) Mean phospho-specific antibody binding to indicated intracellular signaling molecules 30 min after treatment of WT (closed circles) and P198A-Hom (open circles) thymocytes with the PTEN specific inhibitor BpV(phen), in the absence of TCR stimulation, gated CD4+CD8+ (d), CD4+ (e) and CD8+ cells (f). In this figure, circles represent values from individual mice (n=3 mice per genotype), bars are means of groups and 95% CI. Data are representative of 3 independent experiments.

Supplementary Figure 8 PTEN contributes to phosphatase activity, but PTEN haploinsufficiency does not rescue B cell development in ZIP7 deficiency; normal PLCγ2 kinase expression in P198A-Hom cells.

(a) Reduced phosphatase activity in B cells from PTEN-deficient PTENffMb1Cre (n=2) mice compared with wild-type Mb1Cre controls (n=4). Plots show gating on the pre-B (Fraction D) population in these animals (left panel) and reduced phosphatase activity thereof in the absence of PTEN (middle panel, red fill: PTENffMb1Cre; blue fill: PTENwtMb1Cre); quantification in right panel. Bars show means and 95% CI; n=4 PTENwtMb1Cre and 2 PTENf/fMb1Cre mice; experiment done once. (b) Mean anti-PLCγ2 antibody staining in wild-type/P198A-heterozygote (closed circles) and P198A-Hom (open circles) SWHEL immature (B220+CD43-HEL+IgM+IgD-) B cells and B220-CD43- controls; bars show means and 95% CI (n=3 mice per group, experiment done once). (c-d) B cell numbers in irradiated CD45.1+ B6 mice reconstituted with WT (open triangles), PTENf/+Mb1Cre heterozygote (open circles), P198A-Hom (filled triangles) or P198A-Hom PTENf/+Mb1Cre compound mutant BM (filled circles), gated on Hardy Fractions A-F in the BM (c) and total B220+CD19+, follicular B220+CD19+CD23+CD21+ and marginal B220+CD19+CD23-CD21++ B cells in the spleen (d). Each symbol represents an individual mouse (n= 5 chimeric mice generated per genotype). Bars show means and 95% CI.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Anzilotti, C., Swan, D.J., Boisson, B. et al. An essential role for the Zn2+ transporter ZIP7 in B cell development. Nat Immunol 20, 350–361 (2019). https://doi.org/10.1038/s41590-018-0295-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0295-8

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