Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations

Desirée Schubert, Claudia Bode, Rupert Kenefeck, Tie Zheng Hou, James B Wing, Alan Kennedy, Alla Bulashevska, Britt-Sabina Petersen, Alejandro A Schäffer, Björn A Grüning, Susanne Unger, Natalie Frede, Ulrich Baumann, Torsten Witte, Reinhold E Schmidt, Gregor Dueckers, Tim Niehues, Suranjith Seneviratne, Maria Kanariou, Carsten Speckmann, Stephan Ehl, Anne Rensing-Ehl, Klaus Warnatz, Mirzokhid Rakhmanov, Robert Thimme, Peter Hasselblatt, Florian Emmerich, Toni Cathomen, Rolf Backofen, Paul Fisch, Maximilian Seidl, Annette May, Annette Schmitt-Graeff, Shinji Ikemizu, Ulrich Salzer, Andre Franke, Shimon Sakaguchi, Lucy S K Walker, David M Sansom, Bodo Grimbacher, Desirée Schubert, Claudia Bode, Rupert Kenefeck, Tie Zheng Hou, James B Wing, Alan Kennedy, Alla Bulashevska, Britt-Sabina Petersen, Alejandro A Schäffer, Björn A Grüning, Susanne Unger, Natalie Frede, Ulrich Baumann, Torsten Witte, Reinhold E Schmidt, Gregor Dueckers, Tim Niehues, Suranjith Seneviratne, Maria Kanariou, Carsten Speckmann, Stephan Ehl, Anne Rensing-Ehl, Klaus Warnatz, Mirzokhid Rakhmanov, Robert Thimme, Peter Hasselblatt, Florian Emmerich, Toni Cathomen, Rolf Backofen, Paul Fisch, Maximilian Seidl, Annette May, Annette Schmitt-Graeff, Shinji Ikemizu, Ulrich Salzer, Andre Franke, Shimon Sakaguchi, Lucy S K Walker, David M Sansom, Bodo Grimbacher

Abstract

The protein cytotoxic T lymphocyte antigen-4 (CTLA-4) is an essential negative regulator of immune responses, and its loss causes fatal autoimmunity in mice. We studied a large family in which five individuals presented with a complex, autosomal dominant immune dysregulation syndrome characterized by hypogammaglobulinemia, recurrent infections and multiple autoimmune clinical features. We identified a heterozygous nonsense mutation in exon 1 of CTLA4. Screening of 71 unrelated patients with comparable clinical phenotypes identified five additional families (nine individuals) with previously undescribed splice site and missense mutations in CTLA4. Clinical penetrance was incomplete (eight adults of a total of 19 genetically proven CTLA4 mutation carriers were considered unaffected). However, CTLA-4 protein expression was decreased in regulatory T cells (Treg cells) in both patients and carriers with CTLA4 mutations. Whereas Treg cells were generally present at elevated numbers in these individuals, their suppressive function, CTLA-4 ligand binding and transendocytosis of CD80 were impaired. Mutations in CTLA4 were also associated with decreased circulating B cell numbers. Taken together, mutations in CTLA4 resulting in CTLA-4 haploinsufficiency or impaired ligand binding result in disrupted T and B cell homeostasis and a complex immune dysregulation syndrome.

Figures

Figure 1. Genetics and pedigrees of families…
Figure 1. Genetics and pedigrees of families with CTLA4 mutations
(a) Pedigrees of families with CTLA4 mutations. Squares: male subjects; circles: female subjects; black filled symbols: patients with mutation; gray filled symbols: mutation carriers; crossed-out symbols: deceased subjects. CTLA4 was sequenced in all individuals with available gDNA (asterisk). Whole exome sequencing was carried out on subjects with a pink asterisk. (b) Confirmation of the mutations by Sanger sequencing showing cDNA (c) changes and their resulting amino acid (p) changes.
Figure 2. Tissue infiltration and lymphadenopathy in…
Figure 2. Tissue infiltration and lymphadenopathy in patients with CTLA4 mutations
Duodenal biopsies stained for CD4 (patient B.II.4 (a) and A.III.3 (b)). (c) High resolution chest CT scan of the lungs (from patient E.II.3). (d) Pulmonary lymphoid fibrotic lesions stained for CD4 in pulmonary biopsies (from patient E.II.3). (e) Magnetic resonance imaging (MRI) of the pelvic area with two enlarged lymph nodes (arrows) measuring up to 5 cm (from patient A.III.3). (f) Bone marrow biopsy stained for CD4 (from patient B.II.4). (g) MRI of Gadolinium-enhanced lesions (arrows) in the cerebellum (from patient A.III.1) (h) Resected cerebellar lesion stained for CD3 (from patient A.III.1). Scale bars, 50 μm (a,b,d,f,h).
Figure 3. Impact of CTLA4 heterozygosity on…
Figure 3. Impact of CTLA4 heterozygosity on T and B cells
(a) Percentage of naïve CD4+CD45RA+ T cells, CD19+ B cells and CD19+IgM−CD27+ switched memory B cells in the peripheral blood of CTLA4+/− carriers and patients. Gray background indicates normal range. ● Family A, ▴ Family B, ▾ Family C, ∎ Family D. (b) Proportion of IFN-γ+, IL-4+ and and IL-17+ expressing CD3+CD4+CD45RO+ T cells after stimulation of PBMCs with PMA and Ionomycin in healthy CTLA4+/+, CTLA4+/− carriers and CTLA4+/− patients. (c) Percentage of FOXP3+ Treg cells amongst CD4+ T cells in the peripheral blood under resting (ex vivo) conditions or following activation (with beads containing CD3- and CD28-specific antibodies. (d) Representative flow cytometry plots (top) and quantification (bottom) of CTLA-4 expression in CD4+FOXP3+ cells under resting and activated conditions. (Resting: P=0.0130; Activated: P=0.0065). Numbers in quadrants show percentage of CTLA-4 high (top), intermediate (middle) and low (bottom) expressing cells within the FOXP3+ population. Plots in b, c and d show the mean +/− SD; each dot represents one individual. P values were determined by Student’s t test. *P≤0.05; **P≤0.01, ***P≤0.001, ****P≤0.0001.
Figure 4. Impaired transendocytosis, ligand binding and…
Figure 4. Impaired transendocytosis, ligand binding and Treg suppressive activity in CTLA4 heterozygotes
(a) Transendocytosis of CD80-GFP by stimulated primary CD4+FOXP3+ Treg cells in the presence or absence of CTLA-4 blockade. Flow cytometry plots depict CD80-GFP uptake by Treg cells in the absence (upper panels) and presence (lower panels) of CTLA-4 blockade. Dot plot shows the relative CD80-GFP uptake in homozygous versus heterozygous individuals P = 0.0091. (n=9 CTLA4+/+, n=3 CTLA4+/−). (b) (Main panel) Uptake of CD80-Ig (yellow) by CHO cells expressing wild-type and mutant CTLA4. (Right, inset) Flow cytometric analysis of CD80-Ig staining in CHO cells (x axis, CD80-Ig staining; y axis, relative cell number). (Left, inset) CTLA-4 expression (green) in CHO cells, as assessed by staining with an antibody to the C-terminus of CTLA-4. Images are representative of 4 independent experiments. (c) Proliferation of cell trace-labeled CD4+ responder T cells upon co-culture with monocyte-derived dendritic cells and CD3-specific antibodies with or without CD4+CD25+ Treg cells, CTLA-4 Ig or CTLA-4-specific blocking antibodies. Quantification of total proliferating T cell numbers (top) and flow cytometry histograms depicting cell division of responder T cells in suppression assays. P values were determined by Student’s t test. ** P<0.01, *** P<0.001.

References

    1. Al-Herz W, et al. Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency. Front Immunol. 2014;5:162.
    1. Resnick ES, Cunningham-Rundles C. The many faces of the clinical picture of common variable immune deficiency. Curr Opin Allergy Clin Immunol. 2012;12:595–601.
    1. Yong PF, Thaventhiran JE, Grimbacher B. “A rose is a rose is a rose,” but CVID is Not CVID common variable immune deficiency (CVID), what do we know in 2011? Adv Immunol. 2011;111:47–107.
    1. Gathmann B, et al. Clinical picture and treatment of 2212 patients with common variable immunodeficiency. J Allergy Clin Immunol. 2014;134:116–126.
    1. Chen K, et al. Germline mutations in NFKB2 implicate the noncanonical NF-kappaB pathway in the pathogenesis of common variable immunodeficiency. Am J Hum Genet. 2013;93:812–824.
    1. Jou ST, et al. Identification of variations in the human phosphoinositide 3-kinase p110delta gene in children with primary B-cell immunodeficiency of unknown aetiology. Int J Immunogenet. 2006;33:361–369.
    1. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–1061.
    1. Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8:191–197.
    1. Ochs HD, Ziegler SF, Torgerson TR. FOXP3 acts as a rheostat of the immune response. Immunol Rev. 2005;203:156–164.
    1. Tivol EA, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3:541–547.
    1. Waterhouse P, et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270:985–988.
    1. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–336.
    1. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–342.
    1. Friedline RH, et al. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J Exp Med. 2009;206:421–434.
    1. Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302.
    1. Takahashi T, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–310.
    1. Walker LS. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun. 2013;45:49–57.
    1. Wing K, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322:271–275.
    1. Rudd CE. The reverse stop-signal model for CTLA4 function. Nat Rev Immunol. 2008;8:153–160.
    1. Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11:852–863.
    1. Wing K, Yamaguchi T, Sakaguchi S. Cell-autonomous and -non-autonomous roles of CTLA-4 in immune regulation. Trends Immunol. 2011;32:428–433.
    1. Bachmann MF, Kohler G, Ecabert B, Mak TW, Kopf M. Cutting edge: lymphoproliferative disease in the absence of CTLA-4 is not T cell autonomous. J Immunol. 1999;163:1128–1131.
    1. Homann D, et al. Lack of intrinsic CTLA-4 expression has minimal effect on regulation of antiviral T-cell immunity. J Virol. 2006;80:270–280.
    1. Qureshi OS, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332:600–603.
    1. Collins AV, et al. The interaction properties of costimulatory molecules revisited. Immunity. 2002;17:201–210.
    1. Bour-Jordan H, Bluestone JA. Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev. 2009;229:41–66.
    1. Keir ME, Sharpe AH. The B7/CD28 costimulatory family in autoimmunity. Immunol Rev. 2005;204:128–143.
    1. Chambers CA, et al. The role of CTLA-4 in the regulation and initiation of T-cell responses. Immunol Rev. 1996;153:27–46.
    1. Schmidt EM, et al. Ctla-4 controls regulatory T cell peripheral homeostasis and is required for suppression of pancreatic islet autoimmunity. J Immunol. 2009;182:274–282.
    1. Schwartz JC, Zhang X, Fedorov AA, Nathenson SG, Almo SC. Structural basis for co-stimulation by the human CTLA-4/B7-2 complex. Nature. 2001;410:604–608.
    1. Sansom DM, Walker LS. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev. 2006;212:131–148.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264.
    1. Tabares P, et al. Human regulatory T cells are selectively activated by low-dose application of the CD28 superagonist TGN1412/TAB08. Eur J Immunol. 2014;44:1225–1236.
    1. Vincenti F, et al. Costimulation blockade with belatacept in renal transplantation. N Engl J Med. 2005;353:770–781.
    1. Parkes M, Cortes A, van Heel DA, Brown MA. Genetic insights into common pathways and complex relationships among immune-mediated diseases. Nat Rev Genet. 2013;14:661–673.
    1. Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol. 2012;3:211.
    1. Riewaldt J, et al. Severe Developmental B Lymphopoietic Defects in Foxp3-Deficient Mice are Refractory to Adoptive Regulatory T Cell Therapy. Front Immunol. 2012;3:141.
    1. Walker LS, et al. Compromised OX40 function in CD28-deficient mice is linked with failure to develop CXC chemokine receptor 5-positive CD4 cells and germinal centers. J Exp Med. 1999;190:1115–1122.
    1. Angulo I, et al. Phosphoinositide 3-kinase delta gene mutation predisposes to respiratory infection and airway damage. Science. 2013;342:866–871.
    1. Lucas CL, et al. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110delta result in T cell senescence and human immunodeficiency. Nat Immunol. 2014;15:88–97.
    1. Boomer JS, Green JM. An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect Biol. 2010;2:a002436.
    1. Kuehn HS, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345:1623–1627.
    1. Atzeni F, et al. Long-term safety of abatacept in patients with rheumatoid arthritis. Autoimmun Rev. 2013;12:1115–1117.
    1. Wofsy D, Hillson JL, Diamond B. Abatacept for lupus nephritis: alternative definitions of complete response support conflicting conclusions. Arthritis Rheum. 2012;64:3660–3665.
    1. Braig DU, et al. Linkage of autosomal dominant common variable immunodeficiency to chromosome 5p and evidence for locus heterogeneity. Hum Genet. 2003;112:369–378.
    1. Cottingham RW, Jr., Idury RM, Schaffer AA. Faster sequential genetic linkage computations. Am J Hum Genet. 1993;53:252–263.
    1. Lathrop GM, Lalouel JM. Easy calculations of lod scores and genetic risks on small computers. Am J Hum Genet. 1984;36:460–465.
    1. Schaffer AA, Gupta SK, Shriram K, Cottingham RW., Jr. Avoiding recomputation in linkage analysis. Hum Hered. 1994;44:225–237.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079.
    1. McKenna A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303.
    1. Cingolani P, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 2012;6:80–92.
    1. Danecek P, et al. The variant call format and VCFtools. Bioinformatics. 2011;27:2156–2158.
    1. Abbas AR, et al. Immune response in silico (IRIS): immune-specific genes identified from a compendium of microarray expression data. Genes Immun. 2005;6:319–331.
    1. Ashburner M, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–29.
    1. Ehl S, et al. A variant of SCID with specific immune responses and predominance of gamma delta T cells. J Clin Invest. 2005;115:3140–3148.
    1. van Dongen JJ, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia. 2003;17:2257–2317.

Source: PubMed

3
購読する