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:

Disruption of an antimycobacterial circuit between dendritic and helper T cells in human SPPL2a deficiency

Abstract

Human inborn errors of IFN-γ immunity underlie mycobacterial diseases. We describe patients with Mycobacterium bovis (BCG) disease who are homozygous for loss-of-function mutations of SPPL2A. This gene encodes a transmembrane protease that degrades the N-terminal fragment (NTF) of CD74 (HLA invariant chain) in antigen-presenting cells. The CD74 NTF therefore accumulates in the HLA class II+ myeloid and lymphoid cells of SPPL2a-deficient patients. This toxic fragment selectively depletes IL-12- and IL-23-producing CD1c+ conventional dendritic cells (cDC2s) and their circulating progenitors. Moreover, SPPL2a-deficient memory TH1* cells selectively fail to produce IFN-γ when stimulated with mycobacterial antigens in vitro. Finally, Sppl2a–/– mice lack cDC2s, have CD4+ T cells that produce small amounts of IFN-γ after BCG infection, and are highly susceptible to infection with BCG or Mycobacterium tuberculosis. These findings suggest that inherited SPPL2a deficiency in humans underlies mycobacterial disease by decreasing the numbers of cDC2s and impairing IFN-γ production by mycobacterium-specific memory TH1* cells.

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: Discovery and in vitro characterization of SPPL2A mutations.
Fig. 2: A lack of SPPL2a function leads to the accumulation of CD74 NTF.
Fig. 3: Mature DC immunophenotyping.
Fig. 4: Progenitor DC immunophenotyping and DC in vitro differentiation.
Fig. 5: SPPL2a, IRF8, and CD74 profiling in leukocytes, and CD4+ T cell studies in patients with AR SPPL2a and AD IRF8 deficiencies.
Fig. 6: BCG and MTB-specific CD4+CCR6+ T cells from SPPL2a-mutant individuals display impaired IFN-γ production.
Fig. 7: Sppl2a deficiency causes DC deficiencies and a predisposition to mycobacterial infection in mice.

Similar content being viewed by others

References

  1. Bustamante, J. et al. Mendelian susceptibility to mycobacterial disease: genetic, immunological, and clinical features of inborn errors of IFN-gamma immunity. Semin. Immunol. 26, 454–470 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Casanova, J. L. & Abel, L. Genetic dissection of immunity to mycobacteria: the human model. Annu. Rev. Immunol. 20, 581–620 (2002).

    Article  PubMed  CAS  Google Scholar 

  3. de Beaucoudrey, L. et al. Revisiting human IL-12Rbeta1 deficiency: a survey of 141 patients from 30 countries. Medicine 89, 381–402 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Dupuis, S. et al. Human interferon-gamma-mediated immunity is a genetically controlled continuous trait that determines the outcome of mycobacterial invasion. Immunol. Rev. 178, 129–137 (2000).

    Article  PubMed  CAS  Google Scholar 

  5. Kreins, A. Y. et al. Human TYK2 deficiency: mycobacterial and viral infections without hyper-IgE syndrome. J. Exp. Med. 212, 1641–1662 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Bogunovic, D. et al. Mycobacterial disease and impaired IFN-gamma immunity in humans with inherited ISG15 deficiency. Science 337, 1684–1688 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Bustamante, J. et al. Germline CYBB mutations that selectively affect macrophages in kindreds with X-linked predisposition to tuberculous mycobacterial disease. Nat. Immunol. 12, 213–221 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Boisson-Dupuis, S. et al. Inherited and acquired immunodeficiencies underlying tuberculosis in childhood. Immunol. Rev. 264, 103–120 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Nathan, C. F. et al. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J. Exp. Med. 158, 670–689 (1983).

    Article  PubMed  CAS  Google Scholar 

  10. Prando, C. et al. Inherited IL-12p40 deficiency: genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine 92, 109–122 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Kong, X. F. et al. A novel homozygous p.R1105X mutation of the AP4E1 gene in twins with hereditary spastic paraplegia and mycobacterial disease. PLoS One 8, e58286 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Itan, Y. et al. The mutation significance cutoff: gene-level thresholds for variant predictions. Nat. Methods 13, 109–110 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Voss, M. et al. Mechanism, specificity, and physiology of signal peptide peptidase (SPP) and SPP-like proteases. Biochim. Biophys. Acta 1828, 2828–2839 (2013).

    Article  PubMed  CAS  Google Scholar 

  14. Brady, O. A. et al. Regulated intramembrane proteolysis of the frontotemporal lobar degeneration risk factor, TMEM106B, by signal peptide peptidase-like 2a (SPPL2a). J. Biol. Chem. 289, 19670–19680 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Fluhrer, R. et al. A gamma-secretase-like intramembrane cleavage of TNFα by the GxGD aspartyl protease SPPL2b. Nat. Cell Biol. 8, 894–896 (2006).

    Article  PubMed  CAS  Google Scholar 

  16. Friedmann, E. et al. SPPL2a and SPPL2b promote intramembrane proteolysis of TNFα in activated dendritic cells to trigger IL-12 production. Nat. Cell Biol. 8, 843–848 (2006).

    Article  PubMed  CAS  Google Scholar 

  17. Kirkin, V. et al. The Fas ligand intracellular domain is released by ADAM10 and SPPL2a cleavage in T-cells. Cell Death Differ. 14, 1678–1687 (2007).

    Article  PubMed  CAS  Google Scholar 

  18. Martin, L. et al. Regulated intramembrane proteolysis of Bri2 (Itm2b) by ADAM10 and SPPL2a/SPPL2b. J. Biol. Chem. 283, 1644–1652 (2008).

    Article  PubMed  CAS  Google Scholar 

  19. Beisner, D. R. et al. The intramembrane protease Sppl2a is required for B cell and DC development and survival via cleavage of the invariant chain. J. Exp. Med. 210, 23–30 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Bergmann, H. et al. B cell survival, surface BCR and BAFFR expression, CD74 metabolism, and CD8- dendritic cells require the intramembrane endopeptidase SPPL2A. J. Exp. Med. 210, 31–40 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Schneppenheim, J. et al. The intramembrane protease SPPL2a promotes B cell development and controls endosomal traffic by cleavage of the invariant chain. J. Exp. Med. 210, 41–58 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Fleck, D. et al. Proteolytic processing of neuregulin 1 type III by three intramembrane-cleaving proteases. J. Biol. Chem. 291, 318–333 (2016).

    Article  PubMed  CAS  Google Scholar 

  23. Itan, Y. et al. The human gene connectome as a map of short cuts for morbid allele discovery. Proc. Natl. Acad. Sci. USA 110, 5558–5563 (2013).

    Article  PubMed  CAS  Google Scholar 

  24. Zhang, Y. et al. Genomics is rapidly advancing precision medicine for immunological disorders. Nat. Immunol. 16, 1001–1004 (2015).

    Article  PubMed  CAS  Google Scholar 

  25. Rieux-Laucat, F. Inherited and acquired death receptor defects in human autoimmune lymphoproliferative syndrome. Curr. Dir. Autoimmun. 9, 18–36 (2006).

    PubMed  CAS  Google Scholar 

  26. Ai, J. W. et al. The risk of tuberculosis in patients with rheumatoid arthritis treated with tumor necrosis factor-alpha antagonist: a metaanalysis of both randomized controlled trials and registry/cohort studies. J. Rheumatol. 42, 2229–2237 (2015).

    Article  PubMed  CAS  Google Scholar 

  27. Moreno-De-Luca, A. et al. Adaptor protein complex-4 (AP-4) deficiency causes a novel autosomal recessive cerebral palsy syndrome with microcephaly and intellectual disability. J. Med. Genet. 48, 141–144 (2011).

    Article  PubMed  CAS  Google Scholar 

  28. Schneppenheim, J. et al. Signal-peptide-peptidase-like 2a is required for CD74 intramembrane proteolysis in human B cells. Biochem. Biophys. Res. Commun. 451, 48–53 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Becker-Herman, S. et al. CD74 is a member of the regulated intramembrane proteolysis-processed protein family. Mol. Biol. Cell 16, 5061–5069 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Hambleton, S. et al. IRF8 mutations and human dendritic-cell immunodeficiency. N. Engl. J .Med. 365, 127–138 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Bridgeford, E. C. et al. Agammaglobulinemia and Staphylococcus aureus botryomycosis in a cohort of related sentinel Swiss Webster mice. J. Clin. Microbiol. 46, 1881–1884 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Conley, M. E. et al. Primary B cell immunodeficiencies: comparisons and contrasts. Annu. Rev. Immunol. 27, 199–227 (2009).

    Article  PubMed  CAS  Google Scholar 

  33. Reynolds, G. & Haniffa, M. Human and mouse mononuclear phagocyte networks: a tale of two species? Front. Immunol. 6, 330 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Merad, M. et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

    Article  PubMed  CAS  Google Scholar 

  35. Iwakoshi, N. N. et al. The transcription factor XBP-1 is essential for the development and survival of dendritic cells. J. Exp. Med. 204, 2267–2275 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Dickinson, R. E. et al. The evolution of cellular deficiency in GATA2 mutation. Blood 123, 863–874 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Bigley, V. et al. Dendritic cell analysis in primary immunodeficiency. Curr. Opin. Allergy. Clin. Immunol. 16, 530–540 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Vignali, D. A. & Kuchroo, V. K. IL-12 family cytokines: immunological playmakers. Nat. Immunol. 13, 722–728 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. See, P. et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science 356, eaag3009 (2017).

  40. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

  41. Breton, G. et al. Circulating precursors of human CD1c+ and CD141+ dendritic cells. J. Exp. Med. 212, 401–413 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Ma, C. S. et al. Unique and shared signaling pathways cooperate to regulate the differentiation of human CD4+ T cells into distinct effector subsets. J. Exp. Med. 213, 1589–1608 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Teng, M. W. et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat. Med. 21, 719–729 (2015).

    Article  PubMed  CAS  Google Scholar 

  44. Acosta-Rodriguez, E. V. et al. Surface phenotype and antigenic specificity of human interleukin 17–producing T helper memory cells. Nat. Immunol. 8, 639–646 (2007).

    Article  PubMed  CAS  Google Scholar 

  45. Geiger, R. et al. Human naive and memory CD4+ T cell repertoires specific for naturally processed antigens analyzed using libraries of amplified T cells. J. Exp. Med. 206, 1525–1534 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Marquis, J. F. et al. Interferon regulatory factor 8 regulates pathways for antigen presentation in myeloid cells and during tuberculosis. PLoS Genet. 7, e1002097 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Berghout, J. et al. Irf8-regulated genomic responses drive pathological inflammation during cerebral malaria. PLoS Pathog. 9, e1003491 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Bigley, V. et al. The human syndrome of dendritic cell, monocyte, B and NK lymphoid deficiency. J. Exp. Med. 208, 227–234 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Kindler, V. et al. The inducing role of tumor necrosis factor in the development of bactericidal granulomas during BCG infection. Cell 56, 731–740 (1989).

    Article  PubMed  CAS  Google Scholar 

  50. Keane, J. et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N. Engl. J. Med. 345, 1098–1104 (2001).

    Article  PubMed  CAS  Google Scholar 

  51. Thiant, S. et al. Imatinib mesylate inhibits STAT5 phosphorylation in response to IL-7 and promotes T cell lymphopenia in chronic myelogenous leukemia patients. Blood Cancer J. 7, e551 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Martínez-Barricarte, R. et al. Transduction of Herpesvirus saimiri-transformed T cells with exogenous genes of interest. Curr. Protoc. Immunol. 115, 7.21C.1–7.21C.12 (2016).

    Article  Google Scholar 

  53. Schneppenheim, J. et al. The intramembrane proteases signal peptide peptidase-like 2a and 2b have distinct functions in vivo. Mol. Cell Biol. 34, 1398–1411 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. 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  PubMed  PubMed Central  CAS  Google Scholar 

  56. Wang, K. et al. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Med. Genet. 81, 559–575 (2007).

    CAS  Google Scholar 

  58. Abecasis, G. R. et al. Merlin: rapid analysis of dense genetic maps using sparse gene flow trees. Nat. Genet. 30, 97–101 (2002).

    Article  PubMed  CAS  Google Scholar 

  59. Breton, G. et al. Defining human dendritic cell progenitors by multiparametric flow cytometry. Nat. Protoc. 10, 1407–1422 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Avery, D. T. et al. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J. Exp. Med. 207, 155–171 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Lindestam Arlehamn, C. S. et al. A quantitative analysis of complexity of human pathogen-specific CD4 T cell responses in healthy M. tuberculosis infected South Africans. PLoS Pathog. 12, e1005760 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Gros, P. et al. Genetic control of natural resistance to Mycobacterium bovis (BCG) in mice. J. Immunol. 127, 2417–2421 (1981).

    PubMed  CAS  Google Scholar 

  63. Langlais, D. et al. The macrophage IRF8/IRF1 regulome is required for protection against infections and is associated with chronic inflammation. J. Exp. Med. 213, 585–603 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Ng, S. L. et al. IκB kinase epsilon (IKK(epsilon)) regulates the balance between type I and type II interferon responses. Proc. Natl. Acad. Sci. USA 108, 21170–21175 (2011).

    Article  PubMed  Google Scholar 

  65. Mancino, A. et al. A dual cis-regulatory code links IRF8 to constitutive and inducible gene expression in macrophages. Genes Dev. 29, 394–408 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Barish, G. D. et al. Bcl-6 and NF-kappaB cistromes mediate opposing regulation of the innate immune response. Genes Dev. 24, 2760–2765 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Saliba, D. G. et al. IRF5:RelA interaction targets inflammatory genes in macrophages. Cell Rep. 8, 1308–1317 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Kaikkonen, M. U. et al. Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription. Mol. Cell 51, 310–325 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Ostuni, R. et al. Latent enhancers activated by stimulation in differentiated cells. Cell 152, 157–171 (2013).

    Article  PubMed  CAS  Google Scholar 

  70. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Thorvaldsdottir, H. et al. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Coller, C. Rice, X. Ma, M. Ciancanelli, and G. Vogt for helpful discussions and critical reading. We thank Y. Nemirovskaya, E. Anderson, T. Kochetkov, M. Romanick, L. Amar, C. Patissier, C. Desvallées, M. Woollett, D. Papandrea, A. Gall, and J. Gonzalez for technical and secretarial assistance, and all members of the Laboratory of Human Genetics of Infectious Diseases for helpful discussions. We thank F. Batteux and M. Bahuad from the Laboratory of Immunology, Cochin Hospital, Paris, France, for serological testing of patients, and F. B. Menozzi and the rest of the Microbiology Institute, EOC, Bellinzona, for providing microbial products. X.-F.K. was supported by the Jerome Lejeune Foundation, the Stony Wold-Herbert Fund, the Choh-Hao Li Memorial Fund Scholar Award, and the Shanghai Educational Development Foundation. R.M.B. was funded by a European Molecular Biology Organization (EMBO) long-term fellowship. J.M. was supported by the Charles H. Revson Senior Fellowship in Biomedical Sciences. B.S. received support from the Deutsche Forschungsgemeinschaft as part of the SFB877 and the Cluster of Excellence ‘Inflammation at Interfaces’, and the SCHR 1284/1-1 grant. The Laboratory of Human Genetics of Infectious Diseases is supported by grants from the St. Giles Foundation (J.-L.C.), The Rockefeller University Center for Clinical and Translational Science (grant UL1TR001866 from the National Center for Research Resources and the National Center for Advancing Sciences (NCATS) to R.M.-B. and X.-F.K.), the National Institutes of Health, the National Institute of Allergy and Infectious Diseases (5R01AI089970-02 and 5R37AI095983 to J.-L.C.), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID, and the French National Research Agency (ANR) under the ‘Investments for the future’ program (ANR-10-IAHU-01 to L.A.), ANR-IFNGPHOX (ANR-13-ISV3-0001-01, to J.B.), and ANR-GENMSMD (ANR-16-CE17-0005-01, to J.B.), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Descartes University, and The Rockefeller University. E.K.D., C.S.M., and S.G.T. are supported by research grants and fellowships from the National Health and Medical Research Council of Australia. D.L. was supported by a fellowship from the Fonds de Recherche du Québec Santé. Work in P.G.’s laboratory was supported by a grant from the National Institute of Allergy and Infectious Diseases (R01AI035237-19). The work at the Institute for Research in Biomedicine was supported by grants from the ERC (323183 PREDICT, to F.S.), the Swiss National Science Foundation (170213, to F.S.), and the Helmut Horten Foundation. This work was supported by award U19AI118626, NIH NIAID (to A.S. and F.S.).

Author information

Authors and Affiliations

Authors

Contributions

X.-F.K., R.M.-B., J.K., F.M., T. Lazarov, E.K.D., D. Langlais, C.S.M., G.B., T. Lasseau, L.A., C.T., S.B.-D., M.C.N., D. Latorre, J.M., B.S., K.L., G.R., S.G.T., F.G., F.S., P.G., J.B., and J.-L.C. designed experiments. X.-F.K., R.M.-B., F.M., J.K., T. Lasseau, E.K.D., D. Langlais, G.R., C.T., C.S.M., J.M., G.B., J.M., C.T., T. Lazarov, C.D., K.B.L., and D. Latorre conducted experiments. F.J.-H., Y.I., and L.A. performed bioinformatics analysis. X.-F.K., R.M.-B., F.M., J.K., T. Lazarov, E.K.D., D. Langlais, C.S.M., G.B., S.H., L.A., S.B.D., M.C.N., B.S., G.R., S.G.T., F.G., F.S., K.L., P.G., J.B., and J.-L.C. analyzed and/or interpreted data. A.B., C.A., C.P., K.A., D.M.-V., F.A., A.I., F.D., and I.B. provided samples and performed clinical diagnoses and follow-up of the patients. C.S.L.A., A.S., and B.S. contributed critical materials. J.B. and J.-L.C. supervised the study. X.-F.K., R.M.-B., and J.-L.C. wrote the manuscript and designed the figures, to which all other authors contributed. All authors edited the manuscript and approved its final version. X.-F.K. and R.M.-B. contributed equally. J.K., F.M., T. Lazarov, E.K.D., and C.S.M. contributed equally. G.B., K.B.L., and D. Langlais contributed equally. A.B. and C.A. contributed equally. B.S., M.C.N., and K.L. contributed equally. F.G., S.G.T., P.G., and F.S. contributed equally. J.B. and J.-L.C. contributed equally.

Corresponding author

Correspondence to Jean-Laurent Casanova.

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 Whole-genome linkage, whole-exome sequencing, population genetics, qPCR and effect of SPPL2a deficiency on TNF processing.

a) Combined whole-genome linkage analysis of kindreds A and B. LOD scores are shown in black and blue for alternating chromosomes, and information content is reported as a red trace. SPPL2A maps to the linked region of chromosome 15 indicated with a red arrow. b) The upper panel shows the IGV (https://www.broadinstitute.org/igv/) viewer presentation of the region of the genome containing the mutation for the index cases of both kindreds, in an antisense direction. Mutations are located in the center and the alternative nucleotide is indicated in red. The lower panels show the Sanger sequencing results for all the patients in each family, heterozygous carriers and one WT individual for each of the variants. The mutation site is marked with a dashed square. The shaded squares on the electrophoretograms represent the exonic areas, the rest corresponding to introns. c) Graph of CADD score on the y axis against minor allele frequency (MAF) on the x-axis, for all the heterozygous SPPL2A variants reported in the gnomAD database (http://gnomad.broadinstitute.org). The horizontal dashed line represents the MSC for SPPL2A. d, e) qPCR on cDNA from: d) EBV-B cells from a healthy control, a heterozygous carrier and P1 e) SV40 fibroblasts from a healthy control, a heterozygous carrier and P2 using a probe spanning exons 2-3 of SPPL2A. Mean and SEM is represented in both graphs. f) Schematic representation of the structure and cleavage sites of TNF. The SPPL2a and SPPL2b cleavage site in the transmembrane domain (indicated in black) is mark as a red dashed line. g) Memory and h) naive CD4+ T cells were isolated and stimulated with T cell activation and expansion (TAE) beads alone or under TH1 (TAE beads+IL-12) polarizing conditions. TNF secretion was assessed after and then the cells were re-stimulated with PMA/ionomycin and the percentage of TNF-positive cells was determined, by intracellular staining and flow cytometry. Mean is shown in both graphs.

Supplementary Figure 2 Intracellular versus surface expression of CD74 in B cells, T cells, and monocytes, and flow cytometry gating strategy.

a) Gating strategy used for (b) and (c). b) Representative histogram plots from Fig. 2f showing intracellular CD74 staining for B cells, monocytes and T cells from a healthy control (black), a heterozygous individual (blue) and a SPPL2a-deficient patient (red). c) Representative histogram plots from Fig. 2g showing surface CD74 staining for B cells, monocytes and T cells from a healthy control (black), a heterozygous individual (blue) and a SPPL2a-deficient patient (red). d) Gating strategy for Fig. 2h and 5a. PBMCs were depleted of the populations indicated in the curved arrows by MACS. These populations were analyzed with the gating strategy shown in the panels. After depletion of the CD14+, CD3+, CD19+ and CD56+ populations, the flow-through was analyzed with the markers shown in the gating strategy for analysis of the different DC populations.

Supplementary Figure 3 PBMCs from four healthy controls (WT/WT), two heterozygous carriers (WT/M) and three patients (M/M) were studied.

a) Gating strategy to identify B cell subsets. b) Representative data from FACS analysis for B cell subsets obtained with CD19, CD20, CD10 and CD27 Abs, in WT and SPPL2a-deficient patients. c) Percentages of memory B cells from healthy controls (WT/WT), a healthy WT/WT family member, heterozygous carriers (WT/M) and patients with AR SPPL2a deficiency (M/M) expressing IgG (left panel) or IgA (right panel). Means are indicated with a black line. d). Whiskers (5-95 percentile) graph of the ex vivo IgM, IgG and IgA production by naive and memory B cells from healthy controls, patients and heterozygous carriers following stimulation with CD40L and IL-21. e) BAFF-R expression (upper panel) on naive and memory B cells and HLA-DR expression on monocytes and B cells (lower panel) were assessed by FACS and are presented relative to the results for healthy controls; there were no significant differences between healthy donors, heterozygous carriers and patients. In both graphs means are indicated with a black line.

Supplementary Figure 4 DC immunophenotyping, HLA-DR expression in monocytes and DCs, Flt3 ligand production, and IL-12p40 secretion by whole blood.

a) Example of the gating strategy for Fig. 3a,d,g. b) Graphic representation of CD141+ cDC1s as the percentage of total PBMCs in healthy controls, healthy family members (heterozygous or homozygous carriers), patients with AR SPPL2a deficiency and patients with AD IRF8 deficiency from Fig. 3a. c) Levels of HLA-DR expression in monocytes, CD1c+ cells, cDC1s and pDCs from healthy controls and SPPL2a-deficient patients. Mean and S.E.M. are indicated. d) Flow cytometry histograms of the samples plotted in (c). Numbers indicate the MFI for HLA-DR PerCP-Cy5.5. e) CD1c+ cDCs (cDC2s) from controls and patients had higher levels of HLA-DR and CD11c expression than CD1c cDCs. We assessed the presence of cDC2s in a CD1c-independent manner, by plotting CD11c vs. HLA-DR in live cells, FSC-A vs. SSC-A, FSC-A vs. FSC-W, HLA-DR+CD16 CD14Lin-CD11c+CD1c+ or CD1c. The CD1c population in patients and controls did not display HLA-DRhigh CD11chigh expression, indicating that the smaller CD1c+ cDC2 population in the patients had not shifted to the CD1c gate due to an absence of CD1c expression. These data provide further evidence of a lower levels of (cDC2s) CD1c+, HLA-DRhigh, and CD11chigh cDCs in the patients with AR SPPL2a deficiency or AD IRF8 deficiency. f) Flt3 ligand levels were determined in the plasma of healthy controls, patients with AR SPPL2a deficiency, patients with AD IRF8 deficiency, two heterozygous SPPL2A carriers (P1’s parents) and seven patients carrying heterozygous germline mutations of GATA2 causing monoMAC syndrome, as positive controls. g) IL-12p40 production by whole blood from healthy controls, WT and heterozygous family members and SPPL2a-deficient patients after stimulation with BCG or BCG+IFN-γ. In f and g mean and S.D. are indicated. The p values observed in this figure correspond to an unpaired two tailed t-test with a 95% confidence interval (* p < 0.05, ** p < 0.01).

Supplementary Figure 5 Study of DC progenitors and in vitro differentiation.

a) Example of the gating strategy for the rest of the figure. b) Example of the gating strategy, starting from live single cells in (a), from a healthy control and a SPPL2a-deficient patient used for Fig. 4a. c) Graphic representation of total CD34+ progenitors in healthy controls and patients, obtained from (b) showing the mean and S.D. d) Gating strategy, starting from live single cells in (a), for a control and a SPPL2a-deficient patient for the graphs shown in Fig. 4b. e) Graphic representation of CD34+HLA-DR DC progenitors obtained from (d) showing the mean and S.D. f) Gating strategy of early pre-DCs in healthy controls and SPPL2a-deficient patients, starting from the last gate in (a), the data for which are represented in Fig. 4c.

Supplementary Figure 6 Response to BCG and MTB in CD4+CCR6 T cells.

a) Representation of the percentage of IFN-γ+ cells in the T cell lines from the CCR6+ memory subsets upon stimulation with PMA+ionomycin. The dashed line separates samples analyzed in different experiments. b, c) IFN-γ secretion by T cell lines from CCR6+ memory subsets upon stimulation with PMA+ionomycin in (b) or activating anti-CD3 anti-CD28 Abs in (c). d) Representation of the percentage of IFN-γ+ cells in the T cell lines from CD4+CCR6 memory subsets upon stimulation with PMA+ionomycin. The dashed line separates samples analyzed in different experiments. e, f) IFN-γ secretion by T cell lines from CD4+CCR6 memory subsets upon stimulation with PMA+ionomycin (e) or anti CD3+anti-CD28 Abs (f). g) Frequency of CD4+ T cells reactive against the indicated antigens in the CCR6 memory subsets (containing Th1 and Th2 cells) from controls, patients with AR SPPL2a deficiency and patients with AD IRF8 deficiency. h) IFN-γ production by BCG- or i) MTB-specific CD4+ T cells from the CCR6 memory subsets. The vertical dashed line separates samples run in different experiments. An unpaired two-tailed t-test with a 95% confidence interval was performed (ns non-significative, **** p < 0.0001). j) Immunophenotyping for CCR4, CXCR3 and CCR6 of MTB- and flu-specific T cell clones from P1 and P2, showing that MTB-specific T cells are Th1* (CCR6+CXCR3+), whereas flu-specific T cells are Th1(CCR6CXCR3+). k) MTB-specific T cell clones or flu-specific T cell clones from P1 and P2 were stimulated with MTB peptide pool and MTB lysate or flu vaccine, respectively. EBV-B cells from P1, non-transduced (NT), transduced with a retrovirus generated with an empty vector (EV) or a WT allele of SPPL2A (Fig. 2c) were used as APCs for these T cell clones. Proliferation of these T cell clones was measured by [3H]-thymidine assays and expressed as Δcpm. All graphs in this figure have the mean represented and from g to j also the S.E.M.

Supplementary Figure 7 Regulation of SPPL2a expression by IRF8 and gating for mouse experiments.

a) ChIP-Seq analysis of mouse bone marrow-derived macrophages and peritoneal macrophages left unstimulated, or stimulated with IFN-γ or TLR4 agonists (LPS or KLA). The Sppl2a promoter contains binding sites for STAT1, IRF1 and IRF8. RNA-seq profiles demonstrate an induction of Sppl2a expression following macrophage activation with IFN-γ. b) SPPL2a expression, as assessed by immunobloting (WB) and FACS in EBV-B cells from controls, one STAT1-deficient cell line, one IRF8-deficient cell line and one SPPL2a-deficient cell line. In the immunoblot, GAPDH was used as a loading control and the expression of IRF8, STAT1 and CD74 was also assessed. IRF8/ denotes a cell line carrying the K108E mutation, which completely abolished the function of the protein but not its production (Hambleton et al. 2011). c) Example of the gating strategy used for (d). d) Graphical representation of SPPL2a expression in total lymphocytes, B cells and T cells in healthy controls and AD IRF8-deficient patients. e) Example of a representative FACS experiment gating used for the immunophenotyping graphs shown in Fig. 7 and in the rest of this figure. Hambleton, S. et al. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med 365, 127-138 (2011).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–4

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kong, XF., Martinez-Barricarte, R., Kennedy, J. et al. Disruption of an antimycobacterial circuit between dendritic and helper T cells in human SPPL2a deficiency. Nat Immunol 19, 973–985 (2018). https://doi.org/10.1038/s41590-018-0178-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0178-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