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:

Evidence of innate lymphoid cell redundancy in humans

A Corrigendum to this article was published on 16 November 2016

This article has been updated

Abstract

Innate lymphoid cells (ILCs) have potent immunological functions in experimental conditions in mice, but their contributions to immunity in natural conditions in humans have remained unclear. We investigated the presence of ILCs in a cohort of patients with severe combined immunodeficiency (SCID). All ILC subsets were absent in patients with SCID who had mutation of the gene encoding the common γ-chain cytokine receptor subunit IL-2Rγ or the gene encoding the tyrosine kinase JAK3. T cell reconstitution was observed in patients with SCID after hematopoietic stem cell transplantation (HSCT), but the patients still had considerably fewer ILCs in the absence of myeloablation than did healthy control subjects, with the exception of rare cases of reconstitution of the ILC1 subset of ILCs. Notably, the ILC deficiencies observed were not associated with any particular susceptibility to disease, with follow-up extending from 7 years to 39 years after HSCT. We thus report here selective ILC deficiency in humans and show that ILCs might be dispensable in natural conditions, if T cells are present and B cell function is preserved.

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: Normal abundance of ILCs in the peripheral blood of healthy pediatric and adult subjects.
Figure 2: Severe ILC lymphopenia in patients with SCID who have mutation of JAK3.
Figure 3: Long-term ILC lymphopenia in HSCT-treated patients with SCID.
Figure 4: Intestinal and skin ILCs in HSCT-treated patients with SCID.
Figure 5: Reconstitution of ILCs after engraftment of adult multipotent progenitor cells into Rag2−/−Il2rg−/− mice.

Similar content being viewed by others

Change history

  • 19 October 2016

    In the version of this article initially published, author Mikael Ebbo was missing from the author list. The correct list is as follows: Frédéric Vély1,2,20, Vincent Barlogis3,20, Blandine Vallentin3,20, Bénédicte Neven4–7,20, Christelle Piperoglou1,2, Mikael Ebbo1,8, Thibaut Perchet9,10, Maxime Petit9,10, Nadia Yessaad11, Fabien Touzot5,12, Julie Bruneau5,13, Nizar Mahlaoui4–7, Nicolas Zucchini14, Catherine Farnarier2, Gérard Michel3, Despina Moshous4–7, Stéphane Blanche4–7, Arnaud Dujardin15, Hergen Spits16, Jörg H W Distler17, Andreas Ramming17, Capucine Picard4–7,18, Rachel Golub9,10, Alain Fischer4–7,19,21 & Eric Vivier1,2,21. The correct affiliation list ends as follows: 8APHM, Hôpital de la Timone, Service de Médecine Interne, Marseille, France. 9Institut Pasteur, Unité de Lymphopoièse, INSERM, Paris, France. 10Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France. 11MI-mAbs consortium, Aix-Marseille University, Marseille, France. 12APHP, Hôpital Necker-Enfants Malades, Biotherapy Unit, Paris, France. 13APHP, Hôpital Necker-Enfants Malades, Service d'anatomopathologie, Paris, France. 14BD Biosciences, Le Pont-de-Claix, France. 15Innate-Pharma, Marseille, France. 16Academic Medical Center at the University of Amsterdam, Arizona Amsterdam, the Netherlands. 17Department of Internal Medicine, Rheumatology & Immunology, University of Erlangen-Nuremberg, Erlangen, Germany. 18APHP, Hôpital Necker-Enfants Malades, Study Center of Immunodeficiencies, Paris, France. 19College de France, Paris, France. 20These authors contributed equally to this work. 21These authors jointly directed this work. The correct Author Contributions section ends as follows: "...and M.E., N.M., N.Z., C.F., G. M., D.M., S.B., A.D., H.S. and C. Pic. provided key expertise, reagents or samples." In addition, the description of patient C9 was incorrect in the text and legend for Figure 4b,d. The correct text (in the final paragraph of the fourth subsection of Results) is as follows: "In addition, NKp46+ ILCs and ILC2s were readily observed in tissues from patients treated with myeloablative HSCT, as illustrated by the analysis of skin biopsies from a patient with SCID who had mutation of RAG2 (C10) and was treated with myeloablative HSCT (Fig. 4b,d).... As a control, tissue-resident gut ILCs were observed in a patient with SCID who had mutation of RAG1 (C9) but was treated with 'pheno-related' HSCT (related donor with more than one compatible HLA haplotype but not genetically identical) under non-myeloablative conditions (Fig. 4b,d). The correct Figure 4b,d legend is as follows: "(b) Microscopy of tissue sections from patients who were treated with HSCT (stained as in a): duodenum (left) or colon (right) from C9 (RAG1 mutation) and skin from C10 (RAG2 mutation) (middle), at 15 months (C9) or 4 months (C10) after HSCT....(d) Microscopy of tissue sections from patients with SCID (colon from P5 and skin from P11 (as in a); colon from C9 and skin from C10 (as in b))...." Also, a label was missing under Figure 1d, far left; that should be labelled as 'LinCD127+'. Finally, the description of lung NK cells in the final paragraph of Results was incorrect; that text should read as follows: "Similarly, lung NK cells (LinNKp46+NK1.1+CD127) underwent reconstitution...." These errors have been corrected in the PDF and HTML versions of this article.

References

  1. Klose, C.S. & Artis, D. Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat. Immunol. 17, 765–774 (2016).

    CAS  PubMed  Google Scholar 

  2. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    CAS  PubMed  Google Scholar 

  3. Eberl, G., Colonna, M., Di Santo, J.P. & McKenzie, A.N. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 348, aaa6566 (2015).

    PubMed  PubMed Central  Google Scholar 

  4. Juelke, K. & Romagnani, C. Differentiation of human innate lymphoid cells (ILCs). Curr. Opin. Immunol. 38, 75–85 (2016).

    CAS  PubMed  Google Scholar 

  5. Renoux, V.M. et al. Identification of a human natural killer cell lineage-restricted progenitor in fetal and adult tissues. Immunity 43, 394–407 (2015).

    CAS  PubMed  Google Scholar 

  6. Montaldo, E., Juelke, K. & Romagnani, C. Group 3 innate lymphoid cells (ILC3s): origin, differentiation, and plasticity in humans and mice. Eur. J. Immunol. 45, 2171–2182 (2015).

    CAS  PubMed  Google Scholar 

  7. Scoville, S.D. et al. A progenitor cell expressing transcription factor RORγt generates all human innate lymphoid cell subsets. Immunity 44, 1140–1150 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Buckley, R.H. Molecular defects in human severe combined immunodeficiency and approaches to immune reconstitution. Annu. Rev. Immunol. 22, 625–655 (2004).

    CAS  PubMed  Google Scholar 

  9. Fischer, A., Hacein-Bey-Abina, S. & Cavazzana-Calvo, M. Gene therapy for primary adaptive immune deficiencies. J. Allergy Clin. Immunol. 127, 1356–1359 (2011).

    CAS  PubMed  Google Scholar 

  10. Gennery, A.R. et al. Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: entering a new century, do we do better? J. Allergy Clin. Immunol. 126, 602–610 (2010).

    PubMed  Google Scholar 

  11. Buckley, R.H. Transplantation of hematopoietic stem cells in human severe combined immunodeficiency: longterm outcomes. Immunol. Res. 49, 25–43 (2011).

    PubMed  PubMed Central  Google Scholar 

  12. Fischer, A. Primary immunodeficiency diseases: an experimental model for molecular medicine. Lancet 357, 1863–1869 (2001).

    CAS  PubMed  Google Scholar 

  13. Rankin, L.C. et al. Complementarity and redundancy of IL-22-producing innate lymphoid cells. Nat. Immunol. 17, 179–186 (2016).

    CAS  PubMed  Google Scholar 

  14. Song, C. et al. Unique and redundant functions of NKp46+ ILC3s in models of intestinal inflammation. J. Exp. Med. 212, 1869–1882 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gasteiger, G., Fan, X., Dikiy, S., Lee, S.Y. & Rudensky, A.Y. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science 350, 981–985 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Munneke, J.M. et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 124, 812–821 (2014).

    CAS  PubMed  Google Scholar 

  17. Hazenberg, M.D. & Spits, H. Human innate lymphoid cells. Blood 124, 700–709 (2014).

    CAS  PubMed  Google Scholar 

  18. Vallentin, B. et al. Innate lymphoid cells in cancer. Cancer Immunol. Res. 3, 1109–1114 (2015).

    CAS  PubMed  Google Scholar 

  19. Kløverpris, H.N. et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity 44, 391–405 (2016).

    PubMed  PubMed Central  Google Scholar 

  20. Huntington, N.D., Carpentier, S., Vivier, E. & Belz, G.T. Innate lymphoid cells: parallel checkpoints and coordinate interactions with T cells. Curr. Opin. Immunol. 38, 86–93 (2016).

    CAS  PubMed  Google Scholar 

  21. Buckley, R.H. et al. Human severe combined immunodeficiency: genetic, phenotypic, and functional diversity in one hundred eight infants. J. Pediatr. 130, 378–387 (1997).

    CAS  PubMed  Google Scholar 

  22. Stephan, J.L. et al. Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients. J. Pediatr. 123, 564–572 (1993).

    CAS  PubMed  Google Scholar 

  23. McKenzie, A.N., Spits, H. & Eberl, G. Innate lymphoid cells in inflammation and immunity. Immunity 41, 366–374 (2014).

    CAS  PubMed  Google Scholar 

  24. Diefenbach, A., Colonna, M. & Koyasu, S. Development, differentiation, and diversity of innate lymphoid cells. Immunity 41, 354–365 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    CAS  PubMed  Google Scholar 

  26. Neven, B. et al. Long-term outcome after hematopoietic stem cell transplantation of a single-center cohort of 90 patients with severe combined immunodeficiency. Blood 113, 4114–4124 (2009).

    CAS  PubMed  Google Scholar 

  27. Fischer, A. et al. Severe combined immunodeficiency. A model disease for molecular immunology and therapy. Immunol. Rev. 203, 98–109 (2005).

    CAS  PubMed  Google Scholar 

  28. Robinette, M.L. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat. Immunol. 16, 306–317 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Björklund, A.K. et al. The heterogeneity of human CD127+ innate lymphoid cells revealed by single-cell RNA sequencing. Nat. Immunol. 17, 451–460 (2016).

    PubMed  Google Scholar 

  30. Roan, F. et al. CD4+ group 1 innate lymphoid cells (ILC) form a functionally distinct ilc subset that is increased in systemic sclerosis. J. Immunol. 196, 2051–2062 (2016).

    CAS  PubMed  Google Scholar 

  31. Fuchs, A. et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity 38, 769–781 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Wohlfahrt, T. et al. Type 2 innate lymphoid cell counts are increased in patients with systemic sclerosis and correlate with the extent of fibrosis. Ann. Rheum. Dis. 75, 623–626 (2016).

    CAS  PubMed  Google Scholar 

  33. Koyasu, S. Inflammatory ILC2 cells: disguising themselves as progenitors? Nat. Immunol. 16, 133–134 (2015).

    CAS  PubMed  Google Scholar 

  34. Czechowicz, A., Kraft, D., Weissman, I.L. & Bhattacharya, D. Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science 318, 1296–1299 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Railey, M.D., Lokhnygina, Y. & Buckley, R.H. Long-term clinical outcome of patients with severe combined immunodeficiency who received related donor bone marrow transplants without pretransplant chemotherapy or post-transplant GVHD prophylaxis. J. Pediatr. 155, 834–840.e1 (2009).

    PubMed  PubMed Central  Google Scholar 

  36. Laffort, C. et al. Severe cutaneous papillomavirus disease after haemopoietic stem-cell transplantation in patients with severe combined immune deficiency caused by common gammac cytokine receptor subunit or JAK-3 deficiency. Lancet 363, 2051–2054 (2004).

    CAS  PubMed  Google Scholar 

  37. Goldschmidt, M.H. et al. Severe papillomavirus infection progressing to metastatic squamous cell carcinoma in bone marrow-transplanted X-linked SCID dogs. J. Virol. 80, 6621–6628 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Nishio, H., Matsui, K., Tsuji, H., Tamura, A. & Suzuki, K. Immunolocalisation of the janus kinases (JAK) signal transducers and activators of transcription (STAT) pathway in human epidermis. J. Anat. 198, 581–589 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Recher, M. et al. IL-21 is the primary common γ chain-binding cytokine required for human B-cell differentiation in vivo. Blood 118, 6824–6835 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Huntington, N.D. et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J. Exp. Med. 206, 25–34 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Satoh-Takayama, N. et al. IL-7 and IL-15 independently program the differentiation of intestinal CD3NKp46+ cell subsets from Id2-dependent precursors. J. Exp. Med. 207, 273–280 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kang, J. & Coles, M. IL-7: the global builder of the innate lymphoid network and beyond, one niche at a time. Semin. Immunol. 24, 190–197 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Cavazzana-Calvo, M. et al. Long-term T-cell reconstitution after hematopoietic stem-cell transplantation in primary T-cell-immunodeficient patients is associated with myeloid chimerism and possibly the primary disease phenotype. Blood 109, 4575–4581 (2007).

    CAS  PubMed  Google Scholar 

  44. Ruggeri, L., Aversa, F., Martelli, M.F. & Velardi, A. Allogeneic hematopoietic transplantation and natural killer cell recognition of missing self. Immunol. Rev. 214, 202–218 (2006).

    CAS  PubMed  Google Scholar 

  45. Fan, X. & Rudensky, A.Y. Hallmarks of Tissue-Resident Lymphocytes. Cell 164, 1198–1211 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Perdiguero, E.G. & Geissmann, F. The development and maintenance of resident macrophages. Nat. Immunol. 17, 2–8 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Bando, J.K. & Colonna, M. Innate lymphoid cell function in the context of adaptive immunity. Nat. Immunol. 17, 783–789 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Parry, D.A. et al. A homozygous STIM1 mutation impairs store-operated calcium entry and natural killer cell effector function without clinical immunodeficiency. J. Allergy Clin. Immunol. 137, 955–957 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Boulenouar, S. et al. The residual innate lymphoid cells in NFIL3-Deficient mice support suboptimal maternal adaptations to pregnancy. Front. Immunol. 7, 43 (2016).

    PubMed  PubMed Central  Google Scholar 

  50. Tomasello, E. et al. Mapping of NKp46+ cells in healthy human lymphoid and non-lymphoid tissues. Front. Immunol. 3, 344 (2012).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all patients and their families for participating in the study; C. Bonnafous and N. Anceriz (Innate-Pharma) for monoclonal antibody to NKp46; F. Suarez (Paris), N. Schleinitz (Marseille) and Y. Bertrand (Lyon) for enrolling patients in the study; and the Laboratory of Hematology (P.E. Morange, Marseille) for sample storage. Supported by the European Research Council (THINK Advanced Grant for the E.V. laboratory), the Ligue Nationale contre le Cancer (Equipe Labellisée; E.V. laboratory), institutional grants from INSERM, CNRS and Aix-Marseille University to CIML (E.V. laboratory), the Institut Universitaire de France (E.V.), the European Research Council (Pidimmune Advanced Grant for the A.F. laboratory), institutional grants from INSERM, Paris Descartes University and Collège de France (A.F. laboratory), Investissements d'Avenir Instituts Hospitaliers Universitaires (A.F. laboratory), institutional grants from Institut Pasteur, INSERM, Université Paris Diderot and the Agence Nationale de la Recherche (“Myeloten”; R.G. group), the Institut National du Cancer (R.G. group) and Université Sorbonne Paris Cité (“Mucocell”; R.G. group).

Author information

Authors and Affiliations

Authors

Contributions

F.V., A.F. and E.V. devised and supervised the study, designed the research, and wrote the manuscript, with the help of the other co-authors; V.B., B.V. and B.N. designed the research, performed experiments and analyzed the data; C. Pip., T.P., M.P., N.Y., F.T., J.B., J.H.W.D., A.R. and R.G. performed the experiments and analyzed the data; and M.E., N.M., N.Z., C.F., G. M., D.M., S.B., A.D., H.S. and C. Pic. provided key expertise, reagents or samples.

Corresponding authors

Correspondence to Alain Fischer or Eric Vivier.

Ethics declarations

Competing interests

E.V. is the cofounder of and a shareholder in Innate Pharma.

Integrated supplementary information

Supplementary Figure 1 Correlation analysis of the number of circulating ILCs in healthy subjects.

A Spearman test was used to analyze correlations between ILC subset counts in both healthy adult (n = 30) and pediatric patients (n = 29). * P < 0.01 and ** P < 0.0001.

Supplementary Figure 2 Circulating ILCs in patients with RAG1 deficiency.

Flow cytometric analysis of ILC subsets in human peripheral blood as shown in Fig. 1. Data are from one experiment per indicated samples.

Supplementary Figure 3 Expression of CD5 on ILC1s.

The cell-surface expression of CD5 was assessed by flow cytometry on ILC1s from the patients indicated. The results are expressed as the percentages of CD5+ ILC1 within total peripheral blood ILC1 defined as in Fig. 1. Individual colors represent individual patients. * P < 0.02. NS, not significant (* P > 0.05).

Supplementary Figure 4 Absence of intestinal NKp46+ ILCs in HSCT-treated patients with SCID.

Staining with anti-NKp46 and anti-CD3 antibodies on a representative duodenum biopsy specimen from a HSCT-treated SCID patient. Frozen sections were stained with polyclonal anti-NKp46 serum (green), and anti-CD3 mAb (red). Nuclei were counterstained with DAPI (gray). The data shown are representative of at least two independent experiments on the same patient sample. Scale bar, 100 μM

Supplementary Figure 5 Absence of tissue-resident ILCs in aplastic patients with SCID.

CD3, NKp46 and CD3, CD11b, ICOS stainings of indicated representative specimen from two aplastic RAG1 SCID patients. Scale bar, 50 μM.

Supplementary Figure 6 Reconstitution of ILCs after engraftment of adult multipotent progenitors into Rag2−/−Il2rg−/− host mice.

Multipotent progenitors were sorted from C57BL/6/J adult bone marrow. Both multipotent progenitors (MPP) and lymphoid primed multipotent progenitors (LMPP) were identified in the LSK fraction of the bone marrow. (a) Control of sorting purity. Flow cytometry analysis of (b) lung and (c) and small intestine lamina propria and (d) liver after the reconstitution of irradiated (left panel) and non-irradiated (right panel) CD45.1+ Rag2−/−Il2rg−/− recipient mice. By using CD45.1 and CD45.2 congenic markers, donor-derived (CD45.2+) hematopoietic populations were separated from their recipient (CD45.1+) counterparts. (b) The expression of CD49a and CD49b was assessed on liver NKp46+NK1.1+ populations, to identify ILC1 and NK cells respectively. (c) ILC2 from the lung were identified as LinGata3+IL7Rα+ cells co-expressing ICOS and ST2. ILC1 and NK cells from the lungs were identified as NKp46+NK1.1+IL7Rα+ and NKp46+NK1.1+IL7Rα- respectively (d) CD3, CD19, Thy1, CD4, NKp46, NK1.1, CD49a, CD49b, KLRG1, RORγt, Gata3 and IL7Rα were assessed in the small intestine (SI) to identify NK, ILC1, ILC2 and ILC3 populations of the lamina propria. The data shown are representative of two experiments with three mice for each condition.

Supplementary Figure 7 Normal values of γδT cells and invariant NKT cells in HSCT-treated patients with SCID.

(a) Absolute numbers of γδT cells are indicated as cell numbers per microliter of peripheral blood in children and adult SCID patients (colored circles) or CDC patients (colored triangles).(b) iNKT cells were defined as Vα24+Vβ11+ T cells within the CD3+ gate. A representative staining corresponding to P9 is shown (left panel). The percentage of iNKT among CD3+ T cells has been determined for 5 patients (right panel). Dashed lines represent normal ranges of peripheral blood iNKT in healthy volunteers as previously described1. Individual colors represent individual patients, NS, not significant (* P > 0.05).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1 and 2 (PDF 1622 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vély, F., Barlogis, V., Vallentin, B. et al. Evidence of innate lymphoid cell redundancy in humans. Nat Immunol 17, 1291–1299 (2016). https://doi.org/10.1038/ni.3553

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3553

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