Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA - PubMed (original) (raw)

Case Reports

. 2015 Jan;135(1):217-27.

doi: 10.1016/j.jaci.2014.10.019. Epub 2014 Nov 17.

Erin Janssen 1, Janet Chou 1, Toshiro K Ohsumi 2, Sevgi Keles 1, Joyce T Hsu 1, Michel J Massaad 1, Maria Garcia-Lloret 3, Rima Hanna-Wakim 4, Ghassan Dbaibo 4, Abdullah A Alangari 5, Abdulrahman Alsultan 5, Daifulah Al-Zahrani 6, Raif S Geha 1, Talal A Chatila 7

Affiliations

Case Reports

Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA

Louis-Marie Charbonnier et al. J Allergy Clin Immunol. 2015 Jan.

Abstract

Background: A number of heritable immune dysregulatory diseases result from defects affecting regulatory T (Treg) cell development, function, or both. They include immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, which is caused by mutations in forkhead box P3 (FOXP3), and IPEX-like disorders caused by mutations in IL-2 receptor α (IL2RA), signal transducer and activator of transcription 5b (STAT5b), and signal transducer and activator of transcription 1 (STAT1). However, the genetic defects underlying many cases of IPEX-like disorders remain unknown.

Objective: We sought to identify the genetic abnormalities in patients with idiopathic IPEX-like disorders.

Methods: We performed whole-exome and targeted gene sequencing and phenotypic and functional analyses of Treg cells.

Results: A child who presented with an IPEX-like syndrome and severe Treg cell deficiency was found to harbor a nonsense mutation in the gene encoding LPS-responsive beige-like anchor (LRBA), which was previously implicated as a cause of common variable immunodeficiency with autoimmunity. Analysis of subjects with LRBA deficiency revealed marked Treg cell depletion; profoundly decreased expression of canonical Treg cell markers, including FOXP3, CD25, Helios, and cytotoxic T lymphocyte-associated antigen 4; and impaired Treg cell-mediated suppression. There was skewing in favor of memory T cells and intense autoantibody production, with marked expansion of T follicular helper and contraction of T follicular regulatory cells. Whereas the frequency of recent thymic emigrants and the differentiation of induced Treg cells were normal, LRBA-deficient T cells exhibited increased apoptosis and reduced activities of the metabolic sensors mammalian target of rapamycin complexes 1 and 2.

Conclusion: LRBA deficiency is a novel cause of IPEX-like syndrome and Treg cell deficiency associated with metabolic dysfunction and increased apoptosis of Treg cells.

Keywords: Autoantibodies; LPS-responsive beige-like anchor; T follicular helper cells; T follicular regulatory cells; X-linked syndrome; autoimmunity; enteropathy; forkhead box P3; immune dysregulation; mammalian target of rapamycin complex; polyendocrinopathy; regulatory T cells.

Copyright © 2014 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.

PubMed Disclaimer

Figures

Figure 1

Figure 1

A. Pedigrees of the LRBA-deficient families. Generations are designated by Roman numerals (I–II) and individuals by Arabic numerals. Double lines connecting parents indicate consanguinity. Probands in the respective families are indicated by P1-P6 labeling. Squares, males; circles, females; filled symbols, patients.B. Schematic representation of LRBA protein showing the respective domains and the location of patient mutations. C. Sanger sequencing flurograms showing genomic DNA mutations of patient P1 and P2 as compared to the equivalent DNA sequences of healthy controls (Ctrl). D. Western blot analysis of LRBA protein expression in fibroblasts of patient P1 and lymphocytes of patients P2, P3, P5, and P6 as compared to those of controls. The blots were reprobed for β-actin as a positive control.

Figure 2

Figure 2. LRBA deficiency leads to defect in TR cell frequency and phenotype

A. Representative dot plot analysis of FOXP3 expression in patient P1 vs. a control subject. B. Percent CD4+FOXP3+ T cells in the peripheral blood of healthy controls (n=8; open circles) and LRBA deficient patients (n=6; closed circles). C. Representative flow cytometric histogram plots of FOXP3, CD25, CTLA4 and Helios expression in FOXP3+CD4+ T cells from controls (dotted line) and LRBA deficient patients (solid line). Gray area represents control isotype staining. D. Mean fluorescence intensity of the respective TR cell marker in patient and control subjects. ** p<0.01, *** p<0.001 by unpaired two-tailed Student's _t_-test.

Figure 3

Figure 3. LRBA-deficient TR cells have decreased in vitro suppressive ability

A. Flow cytometric analysis of CellTrace Violet proliferation dye dilution in Teff (CD4+CD25−) cells stimulated with anti-CD2/CD3/CD28 mAb-coated bead in the absence or presence of TR cells (CD4+CD25+CD127low) from healthy controls or LRBA deficient patients. B. Cumulative Results of _in vitro_suppression assays in patient and control subjects. N=6 individuals per group, ****p<0.0001 by unpaired two-tailed Student's _t_-test.C. IL-10 production in patient and control TR cells at baseline and following stimulation with the aCD2/CD3/CD28 or PMA + Ionomycin (Iono) for 4 days. N.D.: Not detected. *P<0.05 by 2-way ANOVA with post-test analysis.

Figure 4

Figure 4. Increased frequency of memory T cells in LRBA-deficient subjects

A. Flow cytometric analysis of circulating CD45RO−CD45RA+ and CD45RO+CD45RA−CD4+ T cells in an LRBA-deficient and a control subject. B. Frequency of activated CD45RO+CD45RA− within the peripheral blood CD4+ T cell pool of LRBA-deficient and control subjects. C. Flow cytometric analysis of circulating CD45RO−CD45RA+ and CD45RO+CD45RA− CD8 T cells in an LRBA-deficient and a control subject. D. Frequency of activated CD45RO+CD45RA− within the peripheral blood CD8+ T cell pool of LRBA-deficient and control subjects.E. Flow cytometric analysis of circulating TEMRA CD45RA+CCR7− and naïve CD45RA+CCR7+ CD8 T cells in an LRBA-deficient and a control subject. F. Frequency of circulating TEMRA CD45RA+CCR7− and naïve CD45RA+CCR7+ within the peripheral blood CD8+ T cell pool of LRBA-deficient and control subjects. *p<0.05, ***p<0.001 by Student’s unpaired two tailed t test.

Figure 5

Figure 5. Increased autoantibodies and dysregulated TFH cell response in LRBA-deficient patients

A. Heat map display of autoantibody reactivity against self-antigens in sera of LRBA-deficient and control subjects, a patient with IPEX and a patient with SLE. A value of 1 (black) is equal to the control average + 1 standard deviation (SD). B. Flow cytometric analysis of circulating TFH(CD4+CXCR5+PD-1hi) cells in patient and control subjects. C. Frequency of TFH cells within the peripheral blood CD4+ T cells of LRBA-deficient and control subjects. D. Flow cytometric overlays of the TFH markers BCL6, ICOS and CD40L within the peripheral blood CD4+CXCR5+PD-1+ cell pool of LRBA-deficient and control subjects. E. Flow cytometric analysis of circulating TFR cells. T cells within the CD4+CXCR5+PD-1hi gate were analyzed for Foxp3 expression. F. Frequencies of TFR cells within the peripheral blood CD4+CXCR5+PD-1+ cell pool. *p<0.05, ***p<0.001 by Student’s unpaired two tailed t test.

Figure 6

Figure 6. LRBA deficiency does not affect the in vitro generation of FOXP3+ iTR cells

A. Representative flow cytometric analyses of CD45RA/RO markers in purified CD4+ T cells (upper panel) and in cell sorted CD3+CD4+CD25−CD45RO−CD45RA+T cells (lower panels) of a control and an LRBA-deficient subject. B. Representative flow cytometric analyses of Foxp3 expression in T cells sorted as in panel A and stimulated in culture with anti-CD2/3/28 mAbs in the absence or presence of TGF-β1. C. Collated frequencies of FOXP3+ iTRcells in control and LRBA-deficient subjects (N=4–5/group). Results are representative of 2 independent experiments.

Figure 7

Figure 7. Increased turnover of LRBA-deficient T cells

A, C E. Flow cytometric analysis of AnnexinV (A), Ki67 (B) and CD31 (C) staining in ex-vivo CD4+Tconv and TR cells from LRBA-deficient and control subjects.B, D, F. Frequencies of AnnexinV+ (B), Ki67+ (D) and CD31+ (F) cells within Tconv and TR cell populations of LRBA-deficient and control subjects. N=4–5 patients and 5–8 controls/group . *p<0.05, **p<0.01 by 2-way ANOVA with post-test analysis.

Figure 8

Figure 8. Impaired mTORC1 and mTORC2 activities in LRBA-deficient Tconv and TR cells

A. Flow cytometric analysis of pS6 of conventional (Tconv) and regulatory (TR) T cells from LRBA-deficient and control subjects before after anti-CD2/3/28 stimulation. B. Mean Fluorescence Intensity (MFI) of pS6 within Tconv and TR cells of LRBA-deficient and control subjects before and after anti-CD2/3/28 stimulation. C. Flow cytometric analysis of p4E-BP1 of Tconv and TR cells from LRBA-deficient and control subjects after anti-CD2/3/28 stimulation. D. MFI of p4E-BP1 within Tconv and TR cells of LRBA-deficient and control subjects before and after anti-CD2/3/28 stimulation. E. Flow cytometric analysis of p-AKT (S473) of Tconv and TR cells from LRBA-deficient and control subjects after anti-CD2/3/28 stimulation. F. MFI of p-AKT (S473) within Tconv and TR cells of LRBA-deficient and control subjects before and after anti-CD2/3/28 stimulation. *p<0.05, **p<0.01 by 2-way ANOVA with post-test analysis.

References

    1. Verbsky JW, Chatila TA. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) and IPEX-related disorders: an evolving web of heritable autoimmune diseases. Curr Opin Pediatr. 2013;25:708–714. - PMC - PubMed
    1. Chatila TA, Blaeser F, Ho N, Lederman HM, Voulgaropoulos C, Helms C, et al. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest. 2000;106:R75–R81. - PMC - PubMed
    1. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–21. - PubMed
    1. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. - PubMed
    1. Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked: forkhead box protein 3 mutations and lack of regulatory T cells. J Allergy Clin Immunol. 2007;120:744–750. quiz 51-2. - PubMed

Publication types

MeSH terms

Substances

Supplementary concepts

Grants and funding

LinkOut - more resources