Human FOXN1-deficiency is associated with αβ double-negative and FoxP3+ T-cell expansions that are distinctly modulated upon thymic transplantation

Adriana S Albuquerque, José G Marques, Susana L Silva, Dario Ligeiro, Blythe H Devlin, Jacques Dutrieux, Rémi Cheynier, Claudio Pignata, Rui M M Victorino, M Louise Markert, Ana E Sousa, Adriana S Albuquerque, José G Marques, Susana L Silva, Dario Ligeiro, Blythe H Devlin, Jacques Dutrieux, Rémi Cheynier, Claudio Pignata, Rui M M Victorino, M Louise Markert, Ana E Sousa

Abstract

Forkhead box N1 (FOXN1) is a transcription factor crucial for thymic epithelium development and prevention of its involution. Investigation of a patient with a rare homozygous FOXN1 mutation (R255X), leading to alopecia universalis and thymus aplasia, unexpectedly revealed non-maternal circulating T-cells, and, strikingly, large numbers of aberrant double-negative αβ T-cells (CD4negCD8neg, DN) and regulatory-like T-cells. These data raise the possibility that a thymic rudiment persisted, allowing T-cell development, albeit with disturbances in positive/negative selection, as suggested by DN and FoxP3+ cell expansions. Although regulatory-like T-cell numbers normalized following HLA-mismatched thymic transplantation, the αβDN subset persisted 5 years post-transplantation. Involution of thymus allograft likely occurred 3 years post-transplantation based on sj/βTREC ratio, which estimates intrathymic precursor T-cell divisions and, consequently, thymic explant output. Nevertheless, functional immune-competence was sustained, providing new insights for the design of immunological reconstitution strategies based on thymic transplantation, with potential applications in other clinical settings.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Peripheral T-cells prior to thymus…
Figure 1. Peripheral T-cells prior to thymus transplantation in a patient with R255X FOXN1 mutation.
At day 166 of life there were 10.4% (660 cells/µl) CD4 T-cells, 9.7% (612 cells/µl) CD8 T-cells; and 10.7% (676 cells/µl) DNαβ T-cells in the peripheral blood. CD4+ (A), CD8+ (B), and DNαβ T-cells (C) exhibited a memory-effector activated phenotype and were able to produce significant amounts of IL-2, IFN-γ and IL-4. (D) Percentages of cycling cells, as assessed by Ki-67 expression, within CD8, CD4, DNαβ and γδ T-cells. (E) Dot-plots showing that: FoxP3 expression is restricted to CD3+ cells; that within total FoxP3+ cells (328 cells/µl) there were 67% CD4+, 17% DNαβ and 12% cells co-expressing CD4 and CD8 (DP, DP represented 3% of total αβ cells and 26% of them were FoxP3+) as illustrated by the expression of CD4 and CD8 within total αβ cells (grey) and within FoxP3+ cells (black); the concomitant expression of FoxP3 with other Treg markers (CTLA-4, CD39), or Ki-67 within total CD4+ T-cells; and the lack of cytokine production by FoxP3+ cells. (F) Vβ distribution within FoxP3+ and FoxP3− CD4 T-cells performed at day 254 of life. Graphs show the proportion of FoxP3+ CD4 T-cells and FoxP3− CD4 T-cells belonging to a given Vβ family as assessed by flow cytometry. Dot-plots show analysis after gating on the respective populations; numbers within each quadrant represent the proportion of cells expressing the respective molecules. Cytokine production was assessed after 4 hours stimulation of PBMC with PMA plus Ionomycin in the presence of Brefeldin A.
Figure 2. Immunological reconstitution and Treg recovery…
Figure 2. Immunological reconstitution and Treg recovery after thymic transplantation in a patient with R255X FOXN1 mutation.
(A) Kinetics of the frequency of naïve cells (CD45RA+CD27+) within CD4 and CD8 T-cell subsets; histogram shows CD31 expression within naïve CD4 T-cells at 58 months post-transplant, a marker associated with recent thymic emigrants. (B) Longitudinal quantification of sjTREC (left) and sj/βTREC ratio (right) quantified in total PBMC. (C) Assessment of TCR repertoire by spectratyping analysis of the CDR3 Vβ regions of purified naïve CD4 T-cells at 59 months post-transplant. (D) Representative dot-plots of the longitudinal analysis of the frequency of cells expressing FoxP3 and/or CD25 within total CD4 T-cells (upper dot-plots), and the phenotype of circulating FoxP3+ cells at 36 months post-transplant after successive gates on CD4+ and FoxP3+ T-cells (lower dot-plots); numbers inside quadrants represent the frequency of cells expressing the indicated molecules. (E) Graph shows the proportion of circulating FoxP3+ and FoxP3− CD4 T-cells belonging to each of the Vβ families at 48 months post-transplantation analysed by flow cytometry.
Figure 3. Reduced but functional CD8+ T-cell…
Figure 3. Reduced but functional CD8+ T-cell compartment after fully-mismatched HLA class I thymus transplantation.
(A) Circulating CD8+ αβ T-cells analysed 58 months post-transplantation (5% of total lymphocytes; 83cells/µl) by flow cytometry in terms of: naïve/memory/effector phenotype assessed by CD45RA and CD27 expression; levels of activation markers (CD38 and HLA-DR); IFN-γ and IL-2 production upon PMA+ionomycin stimulation; ex vivo frequency of cycling cells (Ki-67+); and ex vivo frequency of apoptotic cells assessed by annexin V staining. (B) TCR repertoire diversity evaluated by spectratype of CDR3 Vβ regions of purified CD8+ T-cells at 59 months post-transplant. (C) Graph shows the coordinate increase in the proportion of terminally differentiated effector cells defined as CD45RA+CD27neg cells within total CD8+ T-cells during acute varicella infection and its decrease in parallel with its clinical resolution.
Figure 4. Persistence of DNαβ T-cells in…
Figure 4. Persistence of DNαβ T-cells in a patient with R255X FOXN1 mutation after thymic transplantation.
(A) Absolute counts and proportion of DN, CD8+, CD4+ cells within circulating αβ+ T-cells 59 months post-transplantation. Analysis of DNαβ T-cells revealed: (B) a relatively undifferentiated memory phenotype with increased expression of mucosal homing molecules; (C) a skewed repertoire as assessed by spectratype of CDR3 Vβ regions; (D) reduced ability to produce IFN-γ, IL-4, or IL-17 but high IL-2 production upon PMA+ionomycin stimulation; (E) no terminal-effector differentiation according to CD27, CD57 and perforin expression (histogram compares perforin levels within DNαβ, CD8 and CD4 T-cells); (F) reduced levels of the activation markers CD38 and HLA-DR and absence of Treg-associated markers despite the increased levels CD25 expression, Numbers inside dot-plots represent frequency of cells expressing the indicated molecules acquired with FACSCalibur flow cytometer.
Figure 5. Ability of expanded circulating DNαβ…
Figure 5. Ability of expanded circulating DNαβ T-cells to respond to IL-7 and IL-2.
Representative flow cytometric analysis of freshly isolated PBMC from a patient with R255X FOXN1 mutation 59 months after thymic transplantation illustrating (A) the preserved expression of IL-7Rα and increased CD25 expression within DNαβ T-cells; and (B) the high levels of Bcl-2 expression within DNαβ in comparison with CD8 and CD4 T-cells. (C) Up-regulation of p-STAT5 upon 15 min stimulation of PBMC with IL-7 (50 ng/ml), IL-2 (100 U/mL) or IL-15 (25 ng/ml), bars represent p-STAT5 MFI within gated DNαβ, CD8 and CD4 T-cells. Freshly isolated PBMC were cultured for 5-day in the presence of IL-7 (10 ng/ml), IL-2 (10 U/mL), or IL-15 (12.5 ng/ml) or anti-CD3 plus anti-CD28 stimulation and graphs represent the fold change of CD25MFI with respect to medium (D), and the frequency of Ki-67+ cells (E), within gated DNαβ, CD8 and CD4 T-cells. (F) Representative analysis of freshly isolated PBMC illustrating the low levels cycling cells (Ki-67+) despite the increased CD25 expression within gated DNαβ T-cells. Numbers inside dot-plots represent frequency of cells expressing the indicated molecules acquired with FACSCanto (p-STAT5 and 5-day cultures) or FACSCalibur flow cytometers.

References

    1. Nehls M, Pfeifer D, Schorpp M, Hedrich H, Boehm T. New member of the winged-helix protein family disrupted in mouse and rat nude mutations. Nature. 1994;372:103–107.
    1. Blackburn CC, Augustine CL, Li R, Harvey RP, Malin MA, et al. The nu gene acts cell-autonomously and is required for differentiation of thymic epithelial progenitors. Proc Natl Acad Sci U S A. 1996;93:5742–5746.
    1. Chen L, Xiao S, Manley NR. Foxn1 is required to maintain the postnatal thymic microenvironment in a dosage-sensitive manner. Blood. 2009;113:567–574.
    1. Corbeaux T, Hess I, Swann JB, Kanzler B, Haas-Assenbaum A, et al. Thymopoiesis in mice depends on a Foxn1-positive thymic epithelial cell lineage. Proc Natl Acad Sci U S A. 2010;107:16613–16618.
    1. Zook EC, Krishack PA, Zhang S, Zeleznik-Le NJ, Firulli AB, et al. Overexpression of Foxn1 attenuates age-associated thymic involution and prevents the expansion of peripheral CD4 memory T cells. Blood. 2011;118:5723–5731.
    1. Flanagan SP. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet Res. 1966;8:295–309.
    1. Frank J, Pignata C, Panteleyev AA, Prowse DM, Baden H, et al. Exposing the human nude phenotype. Nature. 1999;398:473–474.
    1. Pignata C, Fiore M, Guzzetta V, Castaldo A, Sebastio G, et al. Congenital Alopecia and nail dystrophy associated with severe functional T-cell immunodeficiency in two sibs. Am J Med Genet. 1996;65:167–170.
    1. Pignata C, Gaetaniello L, Masci AM, Frank J, Christiano A, et al. Human equivalent of the mouse Nude/SCID phenotype: long-term evaluation of immunologic reconstitution after bone marrow transplantation. Blood. 2001;97:880–885.
    1. Adriani M, Martinez-Mir A, Fusco F, Busiello R, Frank J, et al. Ancestral founder mutation of the nude (FOXN1) gene in congenital severe combined immunodeficiency associated with alopecia in southern Italy population. Ann Hum Genet. 2004;68:265–268.
    1. Markert ML, Marques JG, Neven B, Devlin BH, McCarthy EA, et al. First use of thymus transplantation therapy for FOXN1 deficiency (nude/SCID): a report of 2 cases. Blood. 2011;117:688–696.
    1. Markert ML, Devlin BH, Alexieff MJ, Li J, McCarthy EA, et al. Review of 54 patients with complete DiGeorge anomaly enrolled in protocols for thymus transplantation: outcome of 44 consecutive transplants. Blood. 2007;109:4539–4547.
    1. Markert ML, Devlin BH, Chinn IK, McCarthy EA, Li YJ. Factors affecting success of thymus transplantation for complete DiGeorge anomaly. Am J Transplant. 2008;8:1729–1736.
    1. Markert ML, Alexieff MJ, Li J, Sarzotti M, Ozaki DA, et al. Postnatal thymus transplantation with immunosuppression as treatment for DiGeorge syndrome. Blood. 2004;104:2574–2581.
    1. Nunes-Cabaco H, Ribot JC, Caramalho I, Serra-Caetano A, Silva-Santos B, et al. Foxp3 induction in human and murine thymus precedes the CD4(+) CD8(+) stage but requires early T-cell receptor expression. Immunol Cell Biol. 2010;88:523–528.
    1. Foxall RB, Soares RS, Albuquerque AS, Cortesao CS, Victorino RM, et al. Increased frequency of CD25dimCD4+ T-cells in HIV-2 infection, a naturally occurring attenuated form of HIV-1. Clin Immunol. 2008;127:158–167.
    1. Azevedo RI, Soares MV, Barata JT, Tendeiro R, Serra-Caetano A, et al. IL-7 sustains CD31 expression in human naive CD4+ T cells and preferentially expands the CD31+ subset in a PI3K-dependent manner. Blood. 2009;113:2999–3007.
    1. Puisieux I, Even J, Pannetier C, Jotereau F, Favrot M, et al. Oligoclonality of tumor-infiltrating lymphocytes from human melanomas. J Immunol. 1994;153:2807–2818.
    1. Garderet L, Dulphy N, Douay C, Chalumeau N, Schaeffer V, et al. The umbilical cord blood alphabeta T-cell repertoire: characteristics of a polyclonal and naive but completely formed repertoire. Blood. 1998;91:340–346.
    1. Dion ML, Poulin JF, Bordi R, Sylvestre M, Corsini R, et al. HIV infection rapidly induces and maintains a substantial suppression of thymocyte proliferation. Immunity. 2004;21:757–768.
    1. Dion ML, Sekaly RP, Cheynier R. Estimating thymic function through quantification of T-cell receptor excision circles. Methods Mol Biol. 2007;380:197–213.
    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. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–1061.
    1. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30:899–911.
    1. Nunes-Cabaco H, Caramalho I, Sepulveda N, Sousa AE. Differentiation of human thymic regulatory T cells at the double positive stage. Eur J Immunol. 2011;41:3604–3614.
    1. Walker MR, Kasprowicz DJ, Gersuk VH, Benard A, Van Landeghen M, et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25− T cells. J Clin Invest. 2003;112:1437–1443.
    1. Morrhaye G, Kermani H, Legros JJ, Baron F, Beguin Y, et al. Impact of growth hormone (GH) deficiency and GH replacement upon thymus function in adult patients. PLoS One. 2009;4:e5668.
    1. Dulude G, Cheynier R, Gauchat D, Abdallah A, Kettaf N, et al. The magnitude of thymic output is genetically determined through controlled intrathymic precursor T cell proliferation. J Immunol. 2008;181:7818–7824.
    1. Nehls M, Kyewski B, Messerle M, Waldschutz R, Schuddekopf K, et al. Two genetically separable steps in the differentiation of thymic epithelium. Science. 1996;272:886–889.
    1. Ikehara S, Pahwa RN, Fernandes G, Hansen CT, Good RA. Functional T cells in athymic nude mice. Proc Natl Acad Sci U S A. 1984;81:886–888.
    1. Guy-Grand D, Azogui O, Celli S, Darche S, Nussenzweig MC, et al. Extrathymic T cell lymphopoiesis: ontogeny and contribution to gut intraepithelial lymphocytes in athymic and euthymic mice. J Exp Med. 2003;197:333–341.
    1. Kennedy JD, Pierce CW, Lake JP. Extrathymic T cell maturation. Phenotypic analysis of T cell subsets in nude mice as a function of age. J Immunol. 1992;148:1620–1629.
    1. Blais ME, Brochu S, Giroux M, Belanger MP, Dulude G, et al. Why T cells of thymic versus extrathymic origin are functionally different. J Immunol. 2008;180:2299–2312.
    1. Schorpp M, Hofmann M, Dear TN, Boehm T. Characterization of mouse and human nude genes. Immunogenetics. 1997;46:509–515.
    1. Schlake T, Schorpp M, Maul-Pavicic A, Malashenko AM, Boehm T. Forkhead/winged-helix transcription factor Whn regulates hair keratin gene expression: molecular analysis of the nude skin phenotype. Dev Dyn. 2000;217:368–376.
    1. Schlake T. The nude gene and the skin. Exp Dermatol. 2001;10:293–304.
    1. Lambolez F, Arcangeli ML, Joret AM, Pasqualetto V, Cordier C, et al. The thymus exports long-lived fully committed T cell precursors that can colonize primary lymphoid organs. Nat Immunol. 2006;7:76–82.
    1. Somech R, Simon AJ, Lev A, Dalal I, Spirer Z, et al. Reduced central tolerance in Omenn syndrome leads to immature self-reactive oligoclonal T cells. J Allergy Clin Immunol. 2009;124:793–800.
    1. Poliani PL, Facchetti F, Ravanini M, Gennery AR, Villa A, et al. Early defects in human T-cell development severely affect distribution and maturation of thymic stromal cells: possible implications for the pathophysiology of Omenn syndrome. Blood. 2009;114:105–108.
    1. Cassani B, Poliani PL, Moratto D, Sobacchi C, Marrella V, et al. Defect of regulatory T cells in patients with Omenn syndrome. J Allergy Clin Immunol. 2010;125:209–216.
    1. Kanaya Y, Ohga S, Ikeda K, Furuno K, Ohno T, et al. Maturational alterations of peripheral T cell subsets and cytokine gene expression in 22q11.2 deletion syndrome. Clin Exp Immunol. 2006;144:85–93.
    1. McLean-Tooke A, Barge D, Spickett GP, Gennery AR. Immunologic defects in 22q11.2 deletion syndrome. J Allergy Clin Immunol. 2008;122:362–367.
    1. Bautista JL, Lio CW, Lathrop SK, Forbush K, Liang Y, et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat Immunol. 2009;10:610–617.
    1. Leung MW, Shen S, Lafaille JJ. TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes. J Exp Med. 2009;206:2121–2130.
    1. Bristeau-Leprince A, Mateo V, Lim A, Magerus-Chatinet A, Solary E, et al. Human TCR alpha/beta+ CD4−CD8− double-negative T cells in patients with autoimmune lymphoproliferative syndrome express restricted Vbeta TCR diversity and are clonally related to CD8+ T cells. J Immunol. 2008;181:440–448.

Source: PubMed

3
구독하다