Skip to main content
Top
Published in: Seminars in Immunopathology 1/2021

Open Access 01-02-2021 | Review

Thymic origins of autoimmunity—lessons from inborn errors of immunity

Authors: Rosa Bacchetta, Kenneth Weinberg

Published in: Seminars in Immunopathology | Issue 1/2021

Login to get access

Abstract

During their intrathymic development, nascent T cells are empowered to protect against pathogens and to be operative for a life-long acceptance of self. While autoreactive effector T (Teff) cell progenitors are eliminated by clonal deletion, the intrathymic mechanisms by which thymic regulatory T cell (tTreg) progenitors maintain specificity for self-antigens but escape deletion to exert their regulatory functions are less well understood. Both tTreg and Teff development and selection result from finely coordinated interactions between their clonotypic T cell receptors (TCR) and peptide/MHC complexes expressed by antigen-presenting cells, such as thymic epithelial cells and thymic dendritic cells. tTreg function is dependent on expression of the FOXP3 transcription factor, and induction of FOXP3 gene expression by tTreg occurs during their thymic development, particularly within the thymic medulla. While initial expression of FOXP3 is downstream of TCR activation, constitutive expression is fixed by interactions with various transcription factors that are regulated by other extracellular signals like TCR and cytokines, leading to epigenetic modification of the FOXP3 gene. Most of the understanding of the molecular events underlying tTreg generation is based on studies of murine models, whereas gaining similar insight in the human system has been very challenging. In this review, we will elucidate how inborn errors of immunity illuminate the critical non-redundant roles of certain molecules during tTreg development, shedding light on how their abnormal development and function cause well-defined diseases that manifest with autoimmunity alone or are associated with states of immune deficiency and autoinflammation.
Literature
2.
go back to reference Owen DL, Sjaastad LE, Farrar MA (2019) Regulatory T cell development in the thymus. J Immunol 203:2031–2041PubMedCrossRef Owen DL, Sjaastad LE, Farrar MA (2019) Regulatory T cell development in the thymus. J Immunol 203:2031–2041PubMedCrossRef
3.
go back to reference Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N (2020) Regulatory T cells and human disease. Annu Rev Immunol 38:541–566PubMedCrossRef Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N (2020) Regulatory T cells and human disease. Annu Rev Immunol 38:541–566PubMedCrossRef
4.
go back to reference Ramsdell F, Rudensky AY (2020) Foxp3: a genetic foundation for regulatory T cell differentiation and function. Nat Immunol 21:708–709PubMedCrossRef Ramsdell F, Rudensky AY (2020) Foxp3: a genetic foundation for regulatory T cell differentiation and function. Nat Immunol 21:708–709PubMedCrossRef
5.
go back to reference Tran DQ, Ramsey H, Shevach EM (2007) Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood 110:2983–2990PubMedPubMedCentralCrossRef Tran DQ, Ramsey H, Shevach EM (2007) Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood 110:2983–2990PubMedPubMedCentralCrossRef
7.
go back to reference Roncarolo MG, Gregori S, Bacchetta R, Battaglia M, Gagliani N (2018) The biology of T regulatory type 1 cells and their therapeutic application in immune-mediated diseases. Immunity 49:1004–1019PubMedCrossRef Roncarolo MG, Gregori S, Bacchetta R, Battaglia M, Gagliani N (2018) The biology of T regulatory type 1 cells and their therapeutic application in immune-mediated diseases. Immunity 49:1004–1019PubMedCrossRef
8.
9.
go back to reference Savage PA, Klawon DEJ, Miller CH (2020) Regulatory T cell development. Annu Rev Immunol 38:421–453PubMedCrossRef Savage PA, Klawon DEJ, Miller CH (2020) Regulatory T cell development. Annu Rev Immunol 38:421–453PubMedCrossRef
10.
go back to reference McDonald-McGinn DM, Sullivan KE, Marino B, Philip N, Swillen A, Vorstman JA, Zackai EH, Emanuel BS, Vermeesch JR, Morrow BE, Scambler PJ, Bassett AS (2015) 22q11.2 deletion syndrome. Nat Rev Dis Primers 1:15071PubMedPubMedCentralCrossRef McDonald-McGinn DM, Sullivan KE, Marino B, Philip N, Swillen A, Vorstman JA, Zackai EH, Emanuel BS, Vermeesch JR, Morrow BE, Scambler PJ, Bassett AS (2015) 22q11.2 deletion syndrome. Nat Rev Dis Primers 1:15071PubMedPubMedCentralCrossRef
11.
go back to reference Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD (2017) Humanized mouse models of clinical disease. Annu Rev Pathol 12:187–215PubMedCrossRef Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD (2017) Humanized mouse models of clinical disease. Annu Rev Pathol 12:187–215PubMedCrossRef
12.
go back to reference Seet CS, He C, Bethune MT, Li S, Chick B, Gschweng EH, Zhu Y, Kim K, Kohn DB, Baltimore D, Crooks GM, Montel-Hagen A (2017) Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids. Nat Methods 14:521–530PubMedPubMedCentralCrossRef Seet CS, He C, Bethune MT, Li S, Chick B, Gschweng EH, Zhu Y, Kim K, Kohn DB, Baltimore D, Crooks GM, Montel-Hagen A (2017) Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids. Nat Methods 14:521–530PubMedPubMedCentralCrossRef
13.
go back to reference Bosticardo M, Pala F, Calzoni E, Delmonte OM, Dobbs K, Gardner CL, Sacchetti N, Kawai T, Garabedian EK, Draper D, Bergerson JRE, DeRavin SS, Freeman AF, Gungor T, Hartog N, Holland SM, Kohn DB, Malech HL, Markert ML, Weinacht KG, Villa A, Seet CS, Montel-Hagen A, Crooks GM, Notarangelo LD (2020) Artificial thymic organoids represent a reliable tool to study T-cell differentiation in patients with severe T-cell lymphopenia. Blood Adv 4:2611–2616PubMedPubMedCentralCrossRef Bosticardo M, Pala F, Calzoni E, Delmonte OM, Dobbs K, Gardner CL, Sacchetti N, Kawai T, Garabedian EK, Draper D, Bergerson JRE, DeRavin SS, Freeman AF, Gungor T, Hartog N, Holland SM, Kohn DB, Malech HL, Markert ML, Weinacht KG, Villa A, Seet CS, Montel-Hagen A, Crooks GM, Notarangelo LD (2020) Artificial thymic organoids represent a reliable tool to study T-cell differentiation in patients with severe T-cell lymphopenia. Blood Adv 4:2611–2616PubMedPubMedCentralCrossRef
15.
go back to reference Raffin C, Vo LT, Bluestone JA (2020) Treg cell-based therapies: challenges and perspectives. Nat Rev Immunol 20:158–172PubMedCrossRef Raffin C, Vo LT, Bluestone JA (2020) Treg cell-based therapies: challenges and perspectives. Nat Rev Immunol 20:158–172PubMedCrossRef
16.
go back to reference Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155:1151–1164PubMedCrossRef Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155:1151–1164PubMedCrossRef
17.
go back to reference Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH (2001) Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med 193:1285–1294PubMedPubMedCentralCrossRef Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH (2001) Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med 193:1285–1294PubMedPubMedCentralCrossRef
18.
go back to reference Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA (2001) CD4+CD25high regulatory cells in human peripheral blood. J Immunol 167:1245–1253PubMedCrossRef Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA (2001) CD4+CD25high regulatory cells in human peripheral blood. J Immunol 167:1245–1253PubMedCrossRef
19.
go back to reference Levings MK, Sangregorio R, Roncarolo MG (2001) Human CD25(+)CD4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function. J Exp Med 193:1295–1302PubMedPubMedCentralCrossRef Levings MK, Sangregorio R, Roncarolo MG (2001) Human CD25(+)CD4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function. J Exp Med 193:1295–1302PubMedPubMedCentralCrossRef
20.
go back to reference Barzaghi F, Passerini L, Gambineri E, Ciullini Mannurita S, Cornu T, Kang ES, Choe YH, Cancrini C, Corrente S, Ciccocioppo R, Cecconi M, Zuin G, Discepolo V, Sartirana C, Schmidtko J, Ikinciogullari A, Ambrosi A, Roncarolo MG, Olek S, Bacchetta R (2012) Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun 38:49–58PubMedCrossRef Barzaghi F, Passerini L, Gambineri E, Ciullini Mannurita S, Cornu T, Kang ES, Choe YH, Cancrini C, Corrente S, Ciccocioppo R, Cecconi M, Zuin G, Discepolo V, Sartirana C, Schmidtko J, Ikinciogullari A, Ambrosi A, Roncarolo MG, Olek S, Bacchetta R (2012) Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun 38:49–58PubMedCrossRef
21.
go back to reference Mold JE, Venkatasubrahmanyam S, Burt TD, Michaelsson J, Rivera JM, Galkina SA, Weinberg K, Stoddart CA, McCune JM (2010) Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science 330:1695–1699PubMedPubMedCentralCrossRef Mold JE, Venkatasubrahmanyam S, Burt TD, Michaelsson J, Rivera JM, Galkina SA, Weinberg K, Stoddart CA, McCune JM (2010) Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science 330:1695–1699PubMedPubMedCentralCrossRef
22.
go back to reference Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203:1701–1711PubMedPubMedCentralCrossRef Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203:1701–1711PubMedPubMedCentralCrossRef
23.
go back to reference Hori S (2014) Lineage stability and phenotypic plasticity of Foxp3(+) regulatory T cells. Immunol Rev 259:159–172PubMedCrossRef Hori S (2014) Lineage stability and phenotypic plasticity of Foxp3(+) regulatory T cells. Immunol Rev 259:159–172PubMedCrossRef
24.
go back to reference Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F (2001) Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27:68–73PubMedCrossRef Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F (2001) Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27:68–73PubMedCrossRef
26.
go back to reference Allan SE, Passerini L, Bacchetta R, Crellin N, Dai M, Orban PC, Ziegler SF, Roncarolo MG, Levings MK (2005) The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest 115:3276–3284PubMedPubMedCentralCrossRef Allan SE, Passerini L, Bacchetta R, Crellin N, Dai M, Orban PC, Ziegler SF, Roncarolo MG, Levings MK (2005) The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest 115:3276–3284PubMedPubMedCentralCrossRef
27.
go back to reference Smith EL, Finney HM, Nesbitt AM, Ramsdell F, Robinson MK (2006) Splice variants of human FOXP3 are functional inhibitors of human CD4+ T-cell activation. Immunology 119:203–211PubMedPubMedCentralCrossRef Smith EL, Finney HM, Nesbitt AM, Ramsdell F, Robinson MK (2006) Splice variants of human FOXP3 are functional inhibitors of human CD4+ T-cell activation. Immunology 119:203–211PubMedPubMedCentralCrossRef
28.
go back to reference Joly AL, Seitz C, Liu S, Kuznetsov NV, Gertow K, Westerberg LS, Paulsson-Berne G, Hansson GK, Andersson J (2018) Alternative splicing of FOXP3 controls regulatory T cell effector functions and is associated with human atherosclerotic plaque stability. Circ Res 122:1385–1394PubMedCrossRef Joly AL, Seitz C, Liu S, Kuznetsov NV, Gertow K, Westerberg LS, Paulsson-Berne G, Hansson GK, Andersson J (2018) Alternative splicing of FOXP3 controls regulatory T cell effector functions and is associated with human atherosclerotic plaque stability. Circ Res 122:1385–1394PubMedCrossRef
29.
go back to reference Frith K, Joly AL, Ma CS, Tangye SG, Lohse Z, Seitz C, Verge CF, Andersson J, Gray P (2019) The FOXP3Δ2 isoform supports Treg cell development and protects against severe IPEX syndrome. J Allergy Clin Immunol 144:317–320.e8PubMedCrossRef Frith K, Joly AL, Ma CS, Tangye SG, Lohse Z, Seitz C, Verge CF, Andersson J, Gray P (2019) The FOXP3Δ2 isoform supports Treg cell development and protects against severe IPEX syndrome. J Allergy Clin Immunol 144:317–320.e8PubMedCrossRef
30.
go back to reference Joly AL, Liu S, Dahlberg CI, Mailer RK, Westerberg LS, Andersson J (2015) Foxp3 lacking exons 2 and 7 is unable to confer suppressive ability to regulatory T cells in vivo. J Autoimmun 63:23–30PubMedCrossRef Joly AL, Liu S, Dahlberg CI, Mailer RK, Westerberg LS, Andersson J (2015) Foxp3 lacking exons 2 and 7 is unable to confer suppressive ability to regulatory T cells in vivo. J Autoimmun 63:23–30PubMedCrossRef
32.
go back to reference Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY (2007) Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature 445:936–940PubMedCrossRef Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY (2007) Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature 445:936–940PubMedCrossRef
33.
go back to reference van der Veeken J, Glasner A, Zhong Y, Hu W, Wang Z-M, Bou-Puerto R, Charbonnier L-M, Chatila TA, Leslie CS, Rudensky AY (2020) The transcription factor Foxp3 shapes regulatory T cell identity by tuning the activity of trans-acting intermediaries. Immunity 53:971–984PubMedPubMedCentralCrossRef van der Veeken J, Glasner A, Zhong Y, Hu W, Wang Z-M, Bou-Puerto R, Charbonnier L-M, Chatila TA, Leslie CS, Rudensky AY (2020) The transcription factor Foxp3 shapes regulatory T cell identity by tuning the activity of trans-acting intermediaries. Immunity 53:971–984PubMedPubMedCentralCrossRef
34.
go back to reference Pan F, Yu H, Dang EV, Barbi J, Pan X, Grosso JF, Jinasena D, Sharma SM, McCadden EM, Getnet D, Drake CG, Liu JO, Ostrowski MC, Pardoll DM (2009) Eos mediates Foxp3-dependent gene silencing in CD4+ regulatory T cells. Science 325:1142–1146PubMedPubMedCentralCrossRef Pan F, Yu H, Dang EV, Barbi J, Pan X, Grosso JF, Jinasena D, Sharma SM, McCadden EM, Getnet D, Drake CG, Liu JO, Ostrowski MC, Pardoll DM (2009) Eos mediates Foxp3-dependent gene silencing in CD4+ regulatory T cells. Science 325:1142–1146PubMedPubMedCentralCrossRef
36.
go back to reference Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA (2004) Role of LAG-3 in regulatory T cells. Immunity 21:503–513PubMedCrossRef Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA (2004) Role of LAG-3 in regulatory T cells. Immunity 21:503–513PubMedCrossRef
37.
go back to reference Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA (2011) Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol Rev 241:180–205PubMedPubMedCentralCrossRef Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA (2011) Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol Rev 241:180–205PubMedPubMedCentralCrossRef
38.
go back to reference Mellor AL, Munn DH (2004) IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 4:762–774PubMedCrossRef Mellor AL, Munn DH (2004) IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 4:762–774PubMedCrossRef
39.
go back to reference Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR, Ley TJ (2007) Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 27:635–646PubMedCrossRef Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR, Ley TJ (2007) Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 27:635–646PubMedCrossRef
40.
go back to reference O’Gorman WE, Dooms H, Thorne SH, Kuswanto WF, Simonds EF, Krutzik PO, Nolan GP, Abbas AK (2009) The initial phase of an immune response functions to activate regulatory T cells. J Immunol 183:332–339PubMedCrossRef O’Gorman WE, Dooms H, Thorne SH, Kuswanto WF, Simonds EF, Krutzik PO, Nolan GP, Abbas AK (2009) The initial phase of an immune response functions to activate regulatory T cells. J Immunol 183:332–339PubMedCrossRef
41.
go back to reference Goudy K, Aydin D, Barzaghi F, Gambineri E, Vignoli M, Ciullini Mannurita S, Doglioni C, Ponzoni M, Cicalese MP, Assanelli A, Tommasini A, Brigida I, Dellepiane RM, Martino S, Olek S, Aiuti A, Ciceri F, Roncarolo MG, Bacchetta R (2013) Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol 146:248–261PubMedPubMedCentralCrossRef Goudy K, Aydin D, Barzaghi F, Gambineri E, Vignoli M, Ciullini Mannurita S, Doglioni C, Ponzoni M, Cicalese MP, Assanelli A, Tommasini A, Brigida I, Dellepiane RM, Martino S, Olek S, Aiuti A, Ciceri F, Roncarolo MG, Bacchetta R (2013) Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol 146:248–261PubMedPubMedCentralCrossRef
42.
go back to reference Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, Gasteiger G, Feng Y, Fontenot JD, Rudensky AY (2016) An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17:1322–1333PubMedPubMedCentralCrossRef Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, Gasteiger G, Feng Y, Fontenot JD, Rudensky AY (2016) An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17:1322–1333PubMedPubMedCentralCrossRef
43.
go back to reference Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC (2007) Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204:1257–1265PubMedPubMedCentralCrossRef Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC (2007) Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204:1257–1265PubMedPubMedCentralCrossRef
45.
go back to reference Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30:899–911PubMedCrossRef Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30:899–911PubMedCrossRef
47.
go back to reference Shevyrev D, Tereshchenko V (2019) Treg heterogeneity, function, and homeostasis. Front Immunol 10:3100PubMedCrossRef Shevyrev D, Tereshchenko V (2019) Treg heterogeneity, function, and homeostasis. Front Immunol 10:3100PubMedCrossRef
48.
go back to reference Zemmour D, Zilionis R, Kiner E, Klein AM, Mathis D, Benoist C (2018) Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat Immunol 19:291–301PubMedPubMedCentralCrossRef Zemmour D, Zilionis R, Kiner E, Klein AM, Mathis D, Benoist C (2018) Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat Immunol 19:291–301PubMedPubMedCentralCrossRef
49.
go back to reference Ouyang W, Beckett O, Ma Q, Paik JH, DePinho RA, Li MO (2010) Foxo proteins cooperatively control the differentiation of Foxp3+ regulatory T cells. Nat Immunol 11:618–627PubMedCrossRef Ouyang W, Beckett O, Ma Q, Paik JH, DePinho RA, Li MO (2010) Foxo proteins cooperatively control the differentiation of Foxp3+ regulatory T cells. Nat Immunol 11:618–627PubMedCrossRef
50.
go back to reference Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA (2000) B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immu-noregulatory T cells that control autoimmune diabetes. Immunity 12:431–440PubMedCrossRef Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA (2000) B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immu-noregulatory T cells that control autoimmune diabetes. Immunity 12:431–440PubMedCrossRef
52.
go back to reference Floess S, Freyer J, Siewert C, Baron U, Olek S, Polansky J, Schlawe K, Chang HD, Bopp T, Schmitt E, Klein-Hessling S, Serfling E, Hamann A, Huehn J (2007) Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol 5:e38PubMedPubMedCentralCrossRef Floess S, Freyer J, Siewert C, Baron U, Olek S, Polansky J, Schlawe K, Chang HD, Bopp T, Schmitt E, Klein-Hessling S, Serfling E, Hamann A, Huehn J (2007) Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol 5:e38PubMedPubMedCentralCrossRef
53.
go back to reference Baron U, Floess S, Wieczorek G, Baumann K, Grutzkau A, Dong J, Thiel A, Boeld TJ, Hoffmann P, Edinger M, Turbachova I, Hamann A, Olek S, Huehn J (2007) DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol 37:2378–2389PubMedCrossRef Baron U, Floess S, Wieczorek G, Baumann K, Grutzkau A, Dong J, Thiel A, Boeld TJ, Hoffmann P, Edinger M, Turbachova I, Hamann A, Olek S, Huehn J (2007) DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol 37:2378–2389PubMedCrossRef
54.
go back to reference Yue X, Trifari S, Äijö T, Tsagaratou A, Pastor WA, Zepeda-Martínez JA, Lio CW, Li X, Huang Y, Vijayanand P, Lähdesmäki H, Rao A (2016) Control of Foxp3 stability through modulation of TET activity. J Exp Med 213:377–397PubMedPubMedCentralCrossRef Yue X, Trifari S, Äijö T, Tsagaratou A, Pastor WA, Zepeda-Martínez JA, Lio CW, Li X, Huang Y, Vijayanand P, Lähdesmäki H, Rao A (2016) Control of Foxp3 stability through modulation of TET activity. J Exp Med 213:377–397PubMedPubMedCentralCrossRef
55.
go back to reference Nair VS, Song MH, Ko M, Oh KI (2016) DNA demethylation of the Foxp3 enhancer is maintained through modulation of ten-eleven-translocation and DNA methyltransferases. Mol Cell 39:888–897CrossRef Nair VS, Song MH, Ko M, Oh KI (2016) DNA demethylation of the Foxp3 enhancer is maintained through modulation of ten-eleven-translocation and DNA methyltransferases. Mol Cell 39:888–897CrossRef
56.
go back to reference Nair VS, Oh KI (2014) Down-regulation of Tet2 prevents TSDR demethylation in IL2 deficient regulatory T cells. Biochem Biophys Res Commun 450:918–924PubMedCrossRef Nair VS, Oh KI (2014) Down-regulation of Tet2 prevents TSDR demethylation in IL2 deficient regulatory T cells. Biochem Biophys Res Commun 450:918–924PubMedCrossRef
57.
go back to reference Baron U, Werner J, Schildknecht K, Schulze JJ, Mulu A, Liebert UG, Sack U, Speckmann C, Gossen M, Wong RJ, Stevenson DK, Babel N, Schurmann D, Baldinger T, Bacchetta R, Grutzkau A, Borte S, Olek S (2018) Epigenetic immune cell counting in human blood samples for immunodiagnostics. Sci Transl Med 10:eaan3508PubMedCrossRef Baron U, Werner J, Schildknecht K, Schulze JJ, Mulu A, Liebert UG, Sack U, Speckmann C, Gossen M, Wong RJ, Stevenson DK, Babel N, Schurmann D, Baldinger T, Bacchetta R, Grutzkau A, Borte S, Olek S (2018) Epigenetic immune cell counting in human blood samples for immunodiagnostics. Sci Transl Med 10:eaan3508PubMedCrossRef
59.
60.
go back to reference Hsieh CS, Lee HM, Lio CW (2012) Selection of regulatory T cells in the thymus. Nat Rev Immunol 12:157–167PubMedCrossRef Hsieh CS, Lee HM, Lio CW (2012) Selection of regulatory T cells in the thymus. Nat Rev Immunol 12:157–167PubMedCrossRef
61.
go back to reference Breed ER, Watanabe M, Hogquist KA (2019) Measuring thymic clonal deletion at the population level. J Immunol 202:3226–3233PubMedCrossRef Breed ER, Watanabe M, Hogquist KA (2019) Measuring thymic clonal deletion at the population level. J Immunol 202:3226–3233PubMedCrossRef
62.
go back to reference Vanhanen R, Leskinen K, Mattila IP, Saavalainen P, Arstila TP (2020) Epigenetic and transcriptional analysis supports human regulatory T cell commitment at the CD4+CD8+ thymocyte stage. Cell Immunol 347:104026PubMedCrossRef Vanhanen R, Leskinen K, Mattila IP, Saavalainen P, Arstila TP (2020) Epigenetic and transcriptional analysis supports human regulatory T cell commitment at the CD4+CD8+ thymocyte stage. Cell Immunol 347:104026PubMedCrossRef
63.
go back to reference Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY (2006) An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nat Immunol 7:401–410PubMedCrossRef Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY (2006) An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nat Immunol 7:401–410PubMedCrossRef
64.
go back to reference Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS (2012) A broad range of self-reactivity drives thymic regulatory T cell selection to limit responses to self. Immunity 37:475–486PubMedPubMedCentralCrossRef Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS (2012) A broad range of self-reactivity drives thymic regulatory T cell selection to limit responses to self. Immunity 37:475–486PubMedPubMedCentralCrossRef
65.
go back to reference Danke NA, Koelle DM, Yee C, Beheray S, Kwok WW (2004) Autoreactive T cells in healthy individuals. J Immunol 172:5967–5972PubMedCrossRef Danke NA, Koelle DM, Yee C, Beheray S, Kwok WW (2004) Autoreactive T cells in healthy individuals. J Immunol 172:5967–5972PubMedCrossRef
66.
go back to reference Su LF, Del Alcazar D, Stelekati E, Wherry EJ, Davis MM (2016) Antigen exposure shapes the ratio between antigen-specific Tregs and conventional T cells in human peripheral blood. Proc Natl Acad Sci U S A 113:E6192–E61E8PubMedPubMedCentralCrossRef Su LF, Del Alcazar D, Stelekati E, Wherry EJ, Davis MM (2016) Antigen exposure shapes the ratio between antigen-specific Tregs and conventional T cells in human peripheral blood. Proc Natl Acad Sci U S A 113:E6192–E61E8PubMedPubMedCentralCrossRef
67.
go back to reference Santoni de Sio FR, Passerini L, Restelli S, Valente MM, Pramov A, Maccari ME, Sanvito F, Roncarolo MG, Porteus M, Bacchetta R (2018) Role of human forkhead box P3 in early thymic maturation and peripheral T-cell homeostasis. J Allergy Clin Immunol 142:1909–21 e9PubMedCrossRef Santoni de Sio FR, Passerini L, Restelli S, Valente MM, Pramov A, Maccari ME, Sanvito F, Roncarolo MG, Porteus M, Bacchetta R (2018) Role of human forkhead box P3 in early thymic maturation and peripheral T-cell homeostasis. J Allergy Clin Immunol 142:1909–21 e9PubMedCrossRef
68.
go back to reference Herppich S, Toker A, Pietzsch B, Kitagawa Y, Ohkura N, Miyao T, Floess S, Hori S, Sakaguchi S, Huehn J (2019) Dynamic Imprinting of the Treg cell-specific epigenetic signature in developing thymic regulatory T cells. Front Immunol 10:2382PubMedPubMedCentralCrossRef Herppich S, Toker A, Pietzsch B, Kitagawa Y, Ohkura N, Miyao T, Floess S, Hori S, Sakaguchi S, Huehn J (2019) Dynamic Imprinting of the Treg cell-specific epigenetic signature in developing thymic regulatory T cells. Front Immunol 10:2382PubMedPubMedCentralCrossRef
69.
go back to reference Owen DL, Mahmud SA, Sjaastad LE, Williams JB, Spanier JA, Simeonov DR, Ruscher R, Huang W, Proekt I, Miller CN, Hekim C, Jeschke JC, Aggarwal P, Broeckel U, LaRue RS, Henzler CM, Alegre ML, Anderson MS, August A, Marson A, Zheng Y, Williams CB, Farrar MA (2019) Thymic regulatory T cells arise via two distinct developmental programs. Nat Immunol 20:195–205PubMedPubMedCentralCrossRef Owen DL, Mahmud SA, Sjaastad LE, Williams JB, Spanier JA, Simeonov DR, Ruscher R, Huang W, Proekt I, Miller CN, Hekim C, Jeschke JC, Aggarwal P, Broeckel U, LaRue RS, Henzler CM, Alegre ML, Anderson MS, August A, Marson A, Zheng Y, Williams CB, Farrar MA (2019) Thymic regulatory T cells arise via two distinct developmental programs. Nat Immunol 20:195–205PubMedPubMedCentralCrossRef
70.
go back to reference Owen DL, Mahmud SA, Vang KB, Kelly RM, Blazar BR, Smith KA, Farrar MA (2018) Identification of cellular sources of IL-2 needed for regulatory T cell development and homeostasis. J Immunol 200:3926–3933PubMedCrossRef Owen DL, Mahmud SA, Vang KB, Kelly RM, Blazar BR, Smith KA, Farrar MA (2018) Identification of cellular sources of IL-2 needed for regulatory T cell development and homeostasis. J Immunol 200:3926–3933PubMedCrossRef
71.
go back to reference Hemmers S, Schizas M, Azizi E, Dikiy S, Zhong Y, Feng Y, Altan-Bonnet G, Rudensky AY (2019) IL-2 production by self-reactive CD4 thymocytes scales regulatory T cell generation in the thymus. J Exp Med 216:2466–2478PubMedPubMedCentralCrossRef Hemmers S, Schizas M, Azizi E, Dikiy S, Zhong Y, Feng Y, Altan-Bonnet G, Rudensky AY (2019) IL-2 production by self-reactive CD4 thymocytes scales regulatory T cell generation in the thymus. J Exp Med 216:2466–2478PubMedPubMedCentralCrossRef
72.
go back to reference Vang KB, Yang J, Mahmud SA, Burchill MA, Vegoe AL, Farrar MA (2008) IL-2, -7, and -15, but not thymic stromal lymphopoeitin, redundantly govern CD4+Foxp3+ regulatory T cell development. J Immunol 181:3285–3290PubMedCrossRef Vang KB, Yang J, Mahmud SA, Burchill MA, Vegoe AL, Farrar MA (2008) IL-2, -7, and -15, but not thymic stromal lymphopoeitin, redundantly govern CD4+Foxp3+ regulatory T cell development. J Immunol 181:3285–3290PubMedCrossRef
73.
go back to reference Burchill MA, Yang J, Vang KB, Moon JJ, Chu HH, Lio CW, Vegoe AL, Hsieh CS, Jenkins MK, Farrar MA (2008) Linked T cell receptor and cytokine signaling govern the development of the regulatory T cell repertoire. Immunity 28:112–121PubMedPubMedCentralCrossRef Burchill MA, Yang J, Vang KB, Moon JJ, Chu HH, Lio CW, Vegoe AL, Hsieh CS, Jenkins MK, Farrar MA (2008) Linked T cell receptor and cytokine signaling govern the development of the regulatory T cell repertoire. Immunity 28:112–121PubMedPubMedCentralCrossRef
74.
go back to reference Derbinski J, Schulte A, Kyewski B, Klein L (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol 2:1032–1039PubMedCrossRef Derbinski J, Schulte A, Kyewski B, Klein L (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol 2:1032–1039PubMedCrossRef
75.
go back to reference Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298:1395–1401PubMedCrossRef Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298:1395–1401PubMedCrossRef
76.
go back to reference Finnish-German AC (1997) An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet 17:399–403CrossRef Finnish-German AC (1997) An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet 17:399–403CrossRef
77.
go back to reference Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJ, Lalioti MD, Mullis PE, Antonarakis SE, Kawasaki K, Asakawa S, Ito F, Shimizu N (1997) Positional cloning of the APECED gene. Nat Genet 17:393–398PubMedCrossRef Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJ, Lalioti MD, Mullis PE, Antonarakis SE, Kawasaki K, Asakawa S, Ito F, Shimizu N (1997) Positional cloning of the APECED gene. Nat Genet 17:393–398PubMedCrossRef
78.
go back to reference Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci U S A 105:657–662PubMedPubMedCentralCrossRef Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci U S A 105:657–662PubMedPubMedCentralCrossRef
79.
go back to reference Sansom SN, Shikama-Dorn N, Zhanybekova S, Nusspaumer G, Macaulay IC, Deadman ME, Heger A, Ponting CP, Holländer GA (2014) Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia. Genome Res 24:1918–1931PubMedPubMedCentralCrossRef Sansom SN, Shikama-Dorn N, Zhanybekova S, Nusspaumer G, Macaulay IC, Deadman ME, Heger A, Ponting CP, Holländer GA (2014) Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia. Genome Res 24:1918–1931PubMedPubMedCentralCrossRef
80.
go back to reference Dhalla F, Baran-Gale J, Maio S, Lia Chappell L, Holländer GA, Ponting CP (2020) Biologically indeterminate yet ordered promiscuous gene expression in single medullary thymic epithelial cells. EMBO J 39:e101828PubMedCrossRef Dhalla F, Baran-Gale J, Maio S, Lia Chappell L, Holländer GA, Ponting CP (2020) Biologically indeterminate yet ordered promiscuous gene expression in single medullary thymic epithelial cells. EMBO J 39:e101828PubMedCrossRef
81.
go back to reference Abramson A, Giraud M, Benoist C, Diane MD (2010) Aire’s partners in the molecular control of immunological tolerance. Cell 140:123–135PubMedCrossRef Abramson A, Giraud M, Benoist C, Diane MD (2010) Aire’s partners in the molecular control of immunological tolerance. Cell 140:123–135PubMedCrossRef
82.
go back to reference Pitkänen J, Vähämurto P, Krohn K, Peterson P (2001) Subcellular localization of the autoimmune regulator protein. characterization of nuclear targeting and transcriptional activation domain. J Biol Chem 276:19597–19602PubMedCrossRef Pitkänen J, Vähämurto P, Krohn K, Peterson P (2001) Subcellular localization of the autoimmune regulator protein. characterization of nuclear targeting and transcriptional activation domain. J Biol Chem 276:19597–19602PubMedCrossRef
83.
go back to reference Kumar PG, Laloraya M, Wang CY, Ruan QG, Davoodi-Semiromi A, Kao KJ, She JX (2001) The autoimmune regulator (AIRE) is a DNA-binding protein. J Biol Chem 276:41357–41364PubMedCrossRef Kumar PG, Laloraya M, Wang CY, Ruan QG, Davoodi-Semiromi A, Kao KJ, She JX (2001) The autoimmune regulator (AIRE) is a DNA-binding protein. J Biol Chem 276:41357–41364PubMedCrossRef
84.
go back to reference Org T, Chignola F, Hetenyi C, Gaetani M, Rebane A, Liiv I, Maran U, Mollica L, Bottomley MJ, Musco G, Peterson P (2008) The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression. EMBO Rep 9:370–376PubMedPubMedCentralCrossRef Org T, Chignola F, Hetenyi C, Gaetani M, Rebane A, Liiv I, Maran U, Mollica L, Bottomley MJ, Musco G, Peterson P (2008) The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression. EMBO Rep 9:370–376PubMedPubMedCentralCrossRef
85.
go back to reference Koh AS, Kingston RE, Benoist C, Mathis D (2010) Global relevance of Aire binding to hypomethylated lysine-4 of histone-3. Proc Natl Acad Sci U S A 107:13016–13021PubMedPubMedCentralCrossRef Koh AS, Kingston RE, Benoist C, Mathis D (2010) Global relevance of Aire binding to hypomethylated lysine-4 of histone-3. Proc Natl Acad Sci U S A 107:13016–13021PubMedPubMedCentralCrossRef
86.
go back to reference Waterfield M, Khan IS, Cortez JT, Fan U, Metzger T, Greer A, Fasano K, Martinez-Llordella M, Pollack JL, Erle DJ, Su M, Anderson MS (2014) The transcriptional regulator Aire coopts the repressive ATF7ip-MBD1 complex for the induction of immunotolerance. Nat Immunol 15:258–265PubMedPubMedCentralCrossRef Waterfield M, Khan IS, Cortez JT, Fan U, Metzger T, Greer A, Fasano K, Martinez-Llordella M, Pollack JL, Erle DJ, Su M, Anderson MS (2014) The transcriptional regulator Aire coopts the repressive ATF7ip-MBD1 complex for the induction of immunotolerance. Nat Immunol 15:258–265PubMedPubMedCentralCrossRef
87.
88.
go back to reference Huoh YS, Wu B, Park S, Yang D, Bansal K, Greenwald E, Wong WP, Mathis D, Hur S (2020) Dual functions of Aire CARD multimerization in the transcriptional regulation of T cell tolerance. Nat Commun 11:1625PubMedPubMedCentralCrossRef Huoh YS, Wu B, Park S, Yang D, Bansal K, Greenwald E, Wong WP, Mathis D, Hur S (2020) Dual functions of Aire CARD multimerization in the transcriptional regulation of T cell tolerance. Nat Commun 11:1625PubMedPubMedCentralCrossRef
89.
go back to reference Kekalainen E, Tuovinen H, Joensuu J, Gylling M, Franssila R, Pontynen N, Talvensaari K, Perheentupa J, Miettinen A, Arstila TP (2007) A defect of regulatory T cells in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Immunol 178:1208–1215PubMedCrossRef Kekalainen E, Tuovinen H, Joensuu J, Gylling M, Franssila R, Pontynen N, Talvensaari K, Perheentupa J, Miettinen A, Arstila TP (2007) A defect of regulatory T cells in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Immunol 178:1208–1215PubMedCrossRef
90.
go back to reference Laakso SM, Laurinolli TT, Rossi LH, Lehtoviita A, Sairanen H, Perheentupa J, Kekalainen E, Arstila TP (2010) Regulatory T cell defect in APECED patients is associated with loss of naive FOXP3(+) precursors and impaired activated population. J Autoimmun 35:351–357PubMedCrossRef Laakso SM, Laurinolli TT, Rossi LH, Lehtoviita A, Sairanen H, Perheentupa J, Kekalainen E, Arstila TP (2010) Regulatory T cell defect in APECED patients is associated with loss of naive FOXP3(+) precursors and impaired activated population. J Autoimmun 35:351–357PubMedCrossRef
91.
go back to reference Malchow S, Leventhal DS, Nishi S, Fischer BI, Shen L, Paner GP, Amit AS, Kang C, Geddes JE, Allison JP, Socci ND, Savage PA (2013) Aire-dependent thymic development of tumor-associated regulatory T cells. Science 339:1219–1224PubMedPubMedCentralCrossRef Malchow S, Leventhal DS, Nishi S, Fischer BI, Shen L, Paner GP, Amit AS, Kang C, Geddes JE, Allison JP, Socci ND, Savage PA (2013) Aire-dependent thymic development of tumor-associated regulatory T cells. Science 339:1219–1224PubMedPubMedCentralCrossRef
92.
go back to reference Yang S, Fujikado N, Kolodin D, Benoist C, Mathis D (2015) Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance. Science 348:589–594PubMedPubMedCentralCrossRef Yang S, Fujikado N, Kolodin D, Benoist C, Mathis D (2015) Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance. Science 348:589–594PubMedPubMedCentralCrossRef
93.
go back to reference Malchow S, Leventhal DS, Lee V, Nishi S, Socci ND, Savage PA (2016) Aire enforces immune tolerance by directing autoreactive T cells into the regulatory T cell lineage. Immunity 44:1102–1113PubMedPubMedCentralCrossRef Malchow S, Leventhal DS, Lee V, Nishi S, Socci ND, Savage PA (2016) Aire enforces immune tolerance by directing autoreactive T cells into the regulatory T cell lineage. Immunity 44:1102–1113PubMedPubMedCentralCrossRef
94.
go back to reference Perry JSA, Lio CJ, Kau AL, Nutsch K, Yang Z, Gordon JI, Murphy KM, Hsieh CS (2014) Distinct contributions of Aire and antigen-presenting-cell subsets to the generation of self-tolerance in the thymus. Immunity 41:414–426PubMedPubMedCentralCrossRef Perry JSA, Lio CJ, Kau AL, Nutsch K, Yang Z, Gordon JI, Murphy KM, Hsieh CS (2014) Distinct contributions of Aire and antigen-presenting-cell subsets to the generation of self-tolerance in the thymus. Immunity 41:414–426PubMedPubMedCentralCrossRef
95.
go back to reference Gallegos AM, Bevan MJ (2004) Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med 200:1039–1049PubMedPubMedCentralCrossRef Gallegos AM, Bevan MJ (2004) Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med 200:1039–1049PubMedPubMedCentralCrossRef
96.
go back to reference Sng J, Ayoglu B, Chen JW, Schickel J-N, Ferre EMN, Glauzy S, Romberg N, Hoenig M, Cunningham-Rundles C, Utz PJ, Lionakis MS, Meffre E (2019) AIRE expression controls the peripheral selection of autoreactive B cells. Sci Immunol 4(34):eaav6778PubMedPubMedCentralCrossRef Sng J, Ayoglu B, Chen JW, Schickel J-N, Ferre EMN, Glauzy S, Romberg N, Hoenig M, Cunningham-Rundles C, Utz PJ, Lionakis MS, Meffre E (2019) AIRE expression controls the peripheral selection of autoreactive B cells. Sci Immunol 4(34):eaav6778PubMedPubMedCentralCrossRef
97.
go back to reference Leventhal DS, Gilmore DC, Berger JM, Nishi S, Lee V, Malchow S, Kline DE, Kline J, Vander Griend DJ, Huang H, Socci ND, Savage PA (2016) Dendritic cells coordinate the development and homeostasis of organ-specific regulatory T cells Immunity 44:847–859PubMedPubMedCentralCrossRef Leventhal DS, Gilmore DC, Berger JM, Nishi S, Lee V, Malchow S, Kline DE, Kline J, Vander Griend DJ, Huang H, Socci ND, Savage PA (2016) Dendritic cells coordinate the development and homeostasis of organ-specific regulatory T cells Immunity 44:847–859PubMedPubMedCentralCrossRef
98.
go back to reference Perry JSA, Russler-Germain EV, Zhou YW, Purtha W, Cooper ML, Choi J, Schroeder MA, Salazar V, Egawa T, Lee BC, Abumrad NA, Kim BS, Anderson MS, DiPersio JF, Hsieh CS (2018) Transfer of cell-surface antigens by scavenger receptor CD36 promotes thymic regulatory T cell receptor repertoire development and allo-tolerance. Immunity 48:923–36 e4PubMedPubMedCentralCrossRef Perry JSA, Russler-Germain EV, Zhou YW, Purtha W, Cooper ML, Choi J, Schroeder MA, Salazar V, Egawa T, Lee BC, Abumrad NA, Kim BS, Anderson MS, DiPersio JF, Hsieh CS (2018) Transfer of cell-surface antigens by scavenger receptor CD36 promotes thymic regulatory T cell receptor repertoire development and allo-tolerance. Immunity 48:923–36 e4PubMedPubMedCentralCrossRef
99.
go back to reference Gardner JM, Devoss JJ, Friedman RS, Wong DJ, Tan YX, Zhou X, Johannes KP, Su MA, Chang HY, Krummel MF, Anderson MS (2008) Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 321:843–847PubMedPubMedCentralCrossRef Gardner JM, Devoss JJ, Friedman RS, Wong DJ, Tan YX, Zhou X, Johannes KP, Su MA, Chang HY, Krummel MF, Anderson MS (2008) Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 321:843–847PubMedPubMedCentralCrossRef
100.
go back to reference Gardner JM, Metzger TC, McMahon EJ, Au-Yeung BB, Krawisz AK, Lu W, Price JD, Johannes KP, Satpathy AT, Murphy KM, Tarbell KV, Weiss A, Anderson MS (2013) Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4(+) T cells Immunity 39:560–572PubMedPubMedCentralCrossRef Gardner JM, Metzger TC, McMahon EJ, Au-Yeung BB, Krawisz AK, Lu W, Price JD, Johannes KP, Satpathy AT, Murphy KM, Tarbell KV, Weiss A, Anderson MS (2013) Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4(+) T cells Immunity 39:560–572PubMedPubMedCentralCrossRef
101.
go back to reference Takaba H, Morishita Y, Tomofuji Y, Danks L, Nitta T, Komatsu N, Kodama T, Takayanagi H (2015) Fezf2 orchestrates a thymic program of self-antigen expression for immune tolerance. Cell 163:975–987PubMedCrossRef Takaba H, Morishita Y, Tomofuji Y, Danks L, Nitta T, Komatsu N, Kodama T, Takayanagi H (2015) Fezf2 orchestrates a thymic program of self-antigen expression for immune tolerance. Cell 163:975–987PubMedCrossRef
102.
go back to reference Tomofuji Y, Takaba H, Suzuki HI, Benlaribi R, Martinez CDP, Abe Y, Morishita Y, Okamura T, Taguchi A, Kodama T, Takayanagi H (2020) Chd4 choreographs self-antigen expression for central immune tolerance. Nat Immunol 21:892–901PubMedCrossRef Tomofuji Y, Takaba H, Suzuki HI, Benlaribi R, Martinez CDP, Abe Y, Morishita Y, Okamura T, Taguchi A, Kodama T, Takayanagi H (2020) Chd4 choreographs self-antigen expression for central immune tolerance. Nat Immunol 21:892–901PubMedCrossRef
103.
go back to reference Good RA (1954) Agammaglobulinemia: a provocative experiment of nature. Bull Univ Minn Hosp 26:1–19 Good RA (1954) Agammaglobulinemia: a provocative experiment of nature. Bull Univ Minn Hosp 26:1–19
104.
go back to reference Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Picard C, Puck J, Torgerson TR, Casanova JL, Sullivan KE (2020) Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 40:24–64PubMedPubMedCentralCrossRef Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Picard C, Puck J, Torgerson TR, Casanova JL, Sullivan KE (2020) Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 40:24–64PubMedPubMedCentralCrossRef
105.
go back to reference Bousfiha A, Jeddane L, Picard C, Al-Herz W, Ailal F, Chatila T, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Puck J, Torgerson TR, Casanova JL, Sullivan KE, Tangye SG (2020) Human inborn errors of immunity: 2019 update of the IUIS Phenotypical Classification. J Clin Immunol 40:66–81PubMedPubMedCentralCrossRef Bousfiha A, Jeddane L, Picard C, Al-Herz W, Ailal F, Chatila T, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Puck J, Torgerson TR, Casanova JL, Sullivan KE, Tangye SG (2020) Human inborn errors of immunity: 2019 update of the IUIS Phenotypical Classification. J Clin Immunol 40:66–81PubMedPubMedCentralCrossRef
106.
go back to reference Cepika AM, Sato Y, Liu JM, Uyeda MJ, Bacchetta R, Roncarolo MG (2018) Tregopathies: Monogenic diseases resulting in regulatory T-cell deficiency. J Allergy Clin Immunol 142:1679–1695PubMedCrossRef Cepika AM, Sato Y, Liu JM, Uyeda MJ, Bacchetta R, Roncarolo MG (2018) Tregopathies: Monogenic diseases resulting in regulatory T-cell deficiency. J Allergy Clin Immunol 142:1679–1695PubMedCrossRef
108.
go back to reference Caudy AA, Reddy ST, Chatila T, Atkinson JP, Verbsky JW (2007) CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J Allergy Clin Immunol 119:482–487PubMedCrossRef Caudy AA, Reddy ST, Chatila T, Atkinson JP, Verbsky JW (2007) CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J Allergy Clin Immunol 119:482–487PubMedCrossRef
109.
go back to reference Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, Schickel JN, Tran DQ, Stoddard J, Zhang Y, Frucht DM, Dumitriu B, Scheinberg P, Folio LR, Frein CA, Price S, Koh C, Heller T, Seroogy CM, Huttenlocher A, Rao VK, Su HC, Kleiner D, Notarangelo LD, Rampertaap Y, Olivier KN, McElwee J, Hughes J, Pittaluga S, Oliveira JB, Meffre E, Fleisher TA, Holland SM, Lenardo MJ, Tangye SG, Uzel G (2014) Immune dysregulation in human subjects with heterozygous germline mu- tations in CTLA4. Science 345:1623–1627 Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, Schickel JN, Tran DQ, Stoddard J, Zhang Y, Frucht DM, Dumitriu B, Scheinberg P, Folio LR, Frein CA, Price S, Koh C, Heller T, Seroogy CM, Huttenlocher A, Rao VK, Su HC, Kleiner D, Notarangelo LD, Rampertaap Y, Olivier KN, McElwee J, Hughes J, Pittaluga S, Oliveira JB, Meffre E, Fleisher TA, Holland SM, Lenardo MJ, Tangye SG, Uzel G (2014) Immune dysregulation in human subjects with heterozygous germline mu- tations in CTLA4. Science 345:1623–1627
110.
go back to reference Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, Husami A, Kissell D, Nortman S, Chaturvedi V, Haines H, Young LR, Mo J, Filipovich AH, Bleesing JJ, Mustillo P, Stephens M, Rueda CM, Chougnet CA, Hoebe K, McElwee J, Hughes JD, Karakoc-Aydiner E, Matthews HF, Price S, Su HC, Rao VK, Lenardo MJ, Jordan MB (2015) Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349:436–440PubMedCrossRef Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, Husami A, Kissell D, Nortman S, Chaturvedi V, Haines H, Young LR, Mo J, Filipovich AH, Bleesing JJ, Mustillo P, Stephens M, Rueda CM, Chougnet CA, Hoebe K, McElwee J, Hughes JD, Karakoc-Aydiner E, Matthews HF, Price S, Su HC, Rao VK, Lenardo MJ, Jordan MB (2015) Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349:436–440PubMedCrossRef
111.
go back to reference Flanagan SE, Haapaniemi E, Russell MA, Caswell R, Allen HL, De Franco E, McDonald TJ, Rajala H, Ramelius A, Barton J, Heiskanen K, Heiskanen-Kosma T, Kajosaari M, Murphy NP, Milenkovic T, Seppänen M, Lernmark Å, Mustjoki S, Otonkoski T, Kere J, Morgan NG, Ellard S, Hattersley AT (2014) Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 46:812–814PubMedPubMedCentralCrossRef Flanagan SE, Haapaniemi E, Russell MA, Caswell R, Allen HL, De Franco E, McDonald TJ, Rajala H, Ramelius A, Barton J, Heiskanen K, Heiskanen-Kosma T, Kajosaari M, Murphy NP, Milenkovic T, Seppänen M, Lernmark Å, Mustjoki S, Otonkoski T, Kere J, Morgan NG, Ellard S, Hattersley AT (2014) Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 46:812–814PubMedPubMedCentralCrossRef
112.
go back to reference Cohen AC, Nadeau KC, Tu W, Hwa V, Dionis K, Bezrodnik L, Teper A, Gaillard M, Heinrich J, Krensky AM, Rosenfeld RG, Lewis DB (2006) Decreased accumulation and regulatory function of CD4+ CD25(high) T cells in human STAT5b deficiency. J Immunol 177:2770–2774PubMedCrossRef Cohen AC, Nadeau KC, Tu W, Hwa V, Dionis K, Bezrodnik L, Teper A, Gaillard M, Heinrich J, Krensky AM, Rosenfeld RG, Lewis DB (2006) Decreased accumulation and regulatory function of CD4+ CD25(high) T cells in human STAT5b deficiency. J Immunol 177:2770–2774PubMedCrossRef
113.
go back to reference Roychoudhuri R, Hirahara K, Mousavi K, Clever D, Klebanoff CA, Bonelli M, Sciumè G, Zare H, Vahedi G, Dema B, Yu Z, Liu H, Takahashi H, Rao M, Muranski P, Crompton JG, Punkosdy G, Bedognetti D, Wang E, Hoffmann V, Rivera J, Marincola FM, Nakamura A, Sartorelli V, Kanno Y, Gattinoni L, Muto A, Igarashi K, O’Shea JJ, Restifo NP (2013) BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature 498:506–510PubMedPubMedCentralCrossRef Roychoudhuri R, Hirahara K, Mousavi K, Clever D, Klebanoff CA, Bonelli M, Sciumè G, Zare H, Vahedi G, Dema B, Yu Z, Liu H, Takahashi H, Rao M, Muranski P, Crompton JG, Punkosdy G, Bedognetti D, Wang E, Hoffmann V, Rivera J, Marincola FM, Nakamura A, Sartorelli V, Kanno Y, Gattinoni L, Muto A, Igarashi K, O’Shea JJ, Restifo NP (2013) BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature 498:506–510PubMedPubMedCentralCrossRef
114.
go back to reference Afzali B, Grönholm J, Vandrovcova J, O’Brien C, Sun HW, Vanderleyden I, Davis FP, Khoder A, Zhang Y, Hegazy AN, Villarino AV, Palmer IW, Kaufman J, Watts NR, Kazemian M, Kamenyeva O, Keith J, Sayed A, Kasperaviciute D, Mueller M, Hughes JD, Fuss IJ, Sadiyah MF, Montgomery-Recht K, McElwee J, Restifo NP, Strober W, Linterman MA, Wingfield PT, Uhlig HH, Roychoudhuri R, Aitman TJ, Kelleher P, Lenardo MJ, O’Shea JJ, Cooper N, Laurence ADJ (2017) BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency. Nat Immunol 18:813–823PubMedPubMedCentralCrossRef Afzali B, Grönholm J, Vandrovcova J, O’Brien C, Sun HW, Vanderleyden I, Davis FP, Khoder A, Zhang Y, Hegazy AN, Villarino AV, Palmer IW, Kaufman J, Watts NR, Kazemian M, Kamenyeva O, Keith J, Sayed A, Kasperaviciute D, Mueller M, Hughes JD, Fuss IJ, Sadiyah MF, Montgomery-Recht K, McElwee J, Restifo NP, Strober W, Linterman MA, Wingfield PT, Uhlig HH, Roychoudhuri R, Aitman TJ, Kelleher P, Lenardo MJ, O’Shea JJ, Cooper N, Laurence ADJ (2017) BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency. Nat Immunol 18:813–823PubMedPubMedCentralCrossRef
115.
go back to reference Grant FM, Yang J, Nasrallah R, Clarke J, Sadiyah F, Whiteside SK, Imianowski CJ, Kuo P, Vardaka P, Todorov T, Zandhuis N, Patrascan I, Tough DF, Kometani K, Eil R, Kurosaki T, Okkenhaug K, Roychoudhuri R (2020) BACH2 drives quiescence and maintenance of resting Treg cells to promote homeostasis and cancer immunosuppression. J Exp Med 217:e20190711PubMedPubMedCentralCrossRef Grant FM, Yang J, Nasrallah R, Clarke J, Sadiyah F, Whiteside SK, Imianowski CJ, Kuo P, Vardaka P, Todorov T, Zandhuis N, Patrascan I, Tough DF, Kometani K, Eil R, Kurosaki T, Okkenhaug K, Roychoudhuri R (2020) BACH2 drives quiescence and maintenance of resting Treg cells to promote homeostasis and cancer immunosuppression. J Exp Med 217:e20190711PubMedPubMedCentralCrossRef
116.
go back to reference Fernandez IZ, Baxter RM, Garcia-Perez JE, Vendrame E, Ranganath T, Kong DS, Lundquist K, Nguyen T, Ogolla S, Black J, Galambos C, Gumbart JC, Dawany N, Kelsen JR, de Zoeten EF, Quinones R, Eissa H, Verneris MR, Sullivan KE, Rochford R, Blish CA, Kedl RM, Dutmer CM, Hsieh EWY (2019) A novel human IL2RB mutation results in T and NK cell-driven immune dysregulation. J Exp Med 216:1255–1267 Correction: a novel human IL2RB mutation results in T and NK cell-driven immune dysregulation. 2019. J Exp Med 216:1465 Fernandez IZ, Baxter RM, Garcia-Perez JE, Vendrame E, Ranganath T, Kong DS, Lundquist K, Nguyen T, Ogolla S, Black J, Galambos C, Gumbart JC, Dawany N, Kelsen JR, de Zoeten EF, Quinones R, Eissa H, Verneris MR, Sullivan KE, Rochford R, Blish CA, Kedl RM, Dutmer CM, Hsieh EWY (2019) A novel human IL2RB mutation results in T and NK cell-driven immune dysregulation. J Exp Med 216:1255–1267 Correction: a novel human IL2RB mutation results in T and NK cell-driven immune dysregulation. 2019. J Exp Med 216:1465
117.
go back to reference Zhang Z, Gothe F, Pennamen P, James JR, McDonald D, Mata CP, Modis Y, Alazami AM, Acres M, Haller W, Bowen C, Döffinger R, Sinclair J, Brothers S, Zhang Y, Matthews HF, Naudion S, Pelluard F, Alajlan H, Yamazaki Y, Notarangelo LD, Thaventhiran JE, Engelhardt KR, Al-Mousa H, Hambleton S, Rooryck C, Smith KGC, Lenardo MJ (2019) Human interleukin-2 receptor beta mutations associated with defects in immunity and peripheral tolerance. J Exp Med 216:1311–1327PubMedPubMedCentralCrossRef Zhang Z, Gothe F, Pennamen P, James JR, McDonald D, Mata CP, Modis Y, Alazami AM, Acres M, Haller W, Bowen C, Döffinger R, Sinclair J, Brothers S, Zhang Y, Matthews HF, Naudion S, Pelluard F, Alajlan H, Yamazaki Y, Notarangelo LD, Thaventhiran JE, Engelhardt KR, Al-Mousa H, Hambleton S, Rooryck C, Smith KGC, Lenardo MJ (2019) Human interleukin-2 receptor beta mutations associated with defects in immunity and peripheral tolerance. J Exp Med 216:1311–1327PubMedPubMedCentralCrossRef
118.
go back to reference Serwas NK, Hoeger B, Ardy RC, Stulz SV, Sui Z, Memaran N, Meeths M, Krolo A, Yüce Petronczki Ö, Pfajfer L, Hou TZ, Halliday N, Santos-Valente E, Kalinichenko A, Kennedy A, Mace EM, Mukherjee M, Tesi B, Schrempf A, Pickl WF, Loizou JI, Kain R, Bidmon-Fliegenschnee B, Schickel JN, Glauzy S, Huemer J, Garncarz W, Salzer E, Pierides I, Bilic I, Thiel J, Priftakis P, Banerjee PP, Förster-Waldl E, Medgyesi D, Huber WD, Orange JS, Meffre E, Sansom DM, Bryceson YT, Altman A, Boztug K (2019) Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nat Commun 10:3106PubMedPubMedCentralCrossRef Serwas NK, Hoeger B, Ardy RC, Stulz SV, Sui Z, Memaran N, Meeths M, Krolo A, Yüce Petronczki Ö, Pfajfer L, Hou TZ, Halliday N, Santos-Valente E, Kalinichenko A, Kennedy A, Mace EM, Mukherjee M, Tesi B, Schrempf A, Pickl WF, Loizou JI, Kain R, Bidmon-Fliegenschnee B, Schickel JN, Glauzy S, Huemer J, Garncarz W, Salzer E, Pierides I, Bilic I, Thiel J, Priftakis P, Banerjee PP, Förster-Waldl E, Medgyesi D, Huber WD, Orange JS, Meffre E, Sansom DM, Bryceson YT, Altman A, Boztug K (2019) Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nat Commun 10:3106PubMedPubMedCentralCrossRef
119.
121.
go back to reference Candotti F (2018) Clinical manifestations and pathophysiological mechanisms of the Wiskott-Aldrich Syndrome. J Clin Immunol 38:13–27PubMedCrossRef Candotti F (2018) Clinical manifestations and pathophysiological mechanisms of the Wiskott-Aldrich Syndrome. J Clin Immunol 38:13–27PubMedCrossRef
122.
go back to reference Rowe JH, Delmonte OM, Keles S, Stadinski BD, Dobbs AK, Henderson LA, Yamazaki Y, Allende LM, Bonilla FA, Gonzalez-Granado LI, Celikbilek Celik S, Guner SN, Kapakli H, Yee C, Pai SY, Huseby ES, Reisli I, Regueiro JR, Notarangelo LD (2018) Patients with CD3G mutations reveal a role for human CD3gamma in Treg diversity and suppressive function. Blood 131:2335–2344PubMedPubMedCentralCrossRef Rowe JH, Delmonte OM, Keles S, Stadinski BD, Dobbs AK, Henderson LA, Yamazaki Y, Allende LM, Bonilla FA, Gonzalez-Granado LI, Celikbilek Celik S, Guner SN, Kapakli H, Yee C, Pai SY, Huseby ES, Reisli I, Regueiro JR, Notarangelo LD (2018) Patients with CD3G mutations reveal a role for human CD3gamma in Treg diversity and suppressive function. Blood 131:2335–2344PubMedPubMedCentralCrossRef
123.
go back to reference Godfrey VL, Wilkinson JE, Russell LB (1991) X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol 138:1379–1387PubMedPubMedCentral Godfrey VL, Wilkinson JE, Russell LB (1991) X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol 138:1379–1387PubMedPubMedCentral
124.
go back to reference Powell BR, Buist NRM, Stenzel P (1982) An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr 100:731–737PubMedCrossRef Powell BR, Buist NRM, Stenzel P (1982) An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr 100:731–737PubMedCrossRef
125.
go back to reference Bacchetta R, Barzaghi F, Roncarolo MG (2018) From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann N Y Acad Sci 1417:5–22PubMedCrossRef Bacchetta R, Barzaghi F, Roncarolo MG (2018) From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann N Y Acad Sci 1417:5–22PubMedCrossRef
126.
go back to reference Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ, Nademi Z, Slatter MA, Ulloa ER, Shcherbina A, Roppelt A, Worth A, Silva J, Aiuti A, Murguia-Favela L, Speckmann C, Carneiro-Sampaio M, Fernandes JF, Baris S, Ozen A, Karakoc-Aydiner E, Kiykim A, Schulz A, Steinmann S, Notarangelo LD, Gambineri E, Lionetti P, Shearer WT, Forbes LR, Martinez C, Moshous D, Blanche S, Fisher A, Ruemmele FM, Tissandier C, Ouachee-Chardin M, Rieux-Laucat F, Cavazzana M, Qasim W, Lucarelli B, Albert MH, Kobayashi I, Alonso L, Diaz De Heredia C, Kanegane H, Lawitschka A, Seo JJ, Gonzalez-Vicent M, Diaz MA, Goyal RK, Sauer MG, Yesilipek A, Kim M, Yilmaz-Demirdag Y, Bhatia M, Khlevner J, Richmond Padilla EJ, Martino S, Montin D, Neth O, Molinos-Quintana A, Valverde-Fernandez J, Broides A, Pinsk V, Ballauf A, Haerynck F, Bordon V, Dhooge C, Garcia-Lloret ML, Bredius RG, Kalwak K, Haddad E, Seidel MG, Duckers G, Pai SY, Dvorak CC, Ehl S, Locatelli F, Goldman F, Gennery AR, Cowan MJ, Roncarolo MG, Bacchetta R, Primary immune deficiency treatment C, the Inborn Errors Working Party of the European Society for B, Marrow T (2018) Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol 141:1036–49 e5PubMedCrossRef Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ, Nademi Z, Slatter MA, Ulloa ER, Shcherbina A, Roppelt A, Worth A, Silva J, Aiuti A, Murguia-Favela L, Speckmann C, Carneiro-Sampaio M, Fernandes JF, Baris S, Ozen A, Karakoc-Aydiner E, Kiykim A, Schulz A, Steinmann S, Notarangelo LD, Gambineri E, Lionetti P, Shearer WT, Forbes LR, Martinez C, Moshous D, Blanche S, Fisher A, Ruemmele FM, Tissandier C, Ouachee-Chardin M, Rieux-Laucat F, Cavazzana M, Qasim W, Lucarelli B, Albert MH, Kobayashi I, Alonso L, Diaz De Heredia C, Kanegane H, Lawitschka A, Seo JJ, Gonzalez-Vicent M, Diaz MA, Goyal RK, Sauer MG, Yesilipek A, Kim M, Yilmaz-Demirdag Y, Bhatia M, Khlevner J, Richmond Padilla EJ, Martino S, Montin D, Neth O, Molinos-Quintana A, Valverde-Fernandez J, Broides A, Pinsk V, Ballauf A, Haerynck F, Bordon V, Dhooge C, Garcia-Lloret ML, Bredius RG, Kalwak K, Haddad E, Seidel MG, Duckers G, Pai SY, Dvorak CC, Ehl S, Locatelli F, Goldman F, Gennery AR, Cowan MJ, Roncarolo MG, Bacchetta R, Primary immune deficiency treatment C, the Inborn Errors Working Party of the European Society for B, Marrow T (2018) Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol 141:1036–49 e5PubMedCrossRef
127.
go back to reference Gambineri E, Ciullini Mannurita S, Hagin D, Vignoli M, Anover-Sombke S, DeBoer S, Segundo GRS, Allenspach EJ, Favre C, Ochs HD, Torgerson TR (2018) Clinical, Immunological, and molecular heterogeneity of 173 patients with the phenotype of immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) Syndrome. Front Immunol 9:2411PubMedPubMedCentralCrossRef Gambineri E, Ciullini Mannurita S, Hagin D, Vignoli M, Anover-Sombke S, DeBoer S, Segundo GRS, Allenspach EJ, Favre C, Ochs HD, Torgerson TR (2018) Clinical, Immunological, and molecular heterogeneity of 173 patients with the phenotype of immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) Syndrome. Front Immunol 9:2411PubMedPubMedCentralCrossRef
128.
go back to reference Louie RJ, Tan QK, Gilner JB, Rogers RC, Younge N, Wechsler SB, McDonald MT, Gordon B, Saski CA, Jones JR, Chapman SJ, Stevenson RE, Sleasman JW, Friez MJ (2017) Novel pathogenic variants in FOXP3 in fetuses with echogenic bowel and skin desquamation identified by ultrasound. Am J Med Genet A 173:1219–1225PubMedPubMedCentralCrossRef Louie RJ, Tan QK, Gilner JB, Rogers RC, Younge N, Wechsler SB, McDonald MT, Gordon B, Saski CA, Jones JR, Chapman SJ, Stevenson RE, Sleasman JW, Friez MJ (2017) Novel pathogenic variants in FOXP3 in fetuses with echogenic bowel and skin desquamation identified by ultrasound. Am J Med Genet A 173:1219–1225PubMedPubMedCentralCrossRef
129.
go back to reference Passerini L, Rossi Mel E, Sartirana C, Fousteri G, Bondanza A, Naldini L, Roncarolo MG, Bacchetta R (2013) CD4(+) T cells from IPEX patients convert into functional and stable regulatory T cells by FOXP3 gene transfer. Sci Transl Med 5:215ra174PubMedCrossRef Passerini L, Rossi Mel E, Sartirana C, Fousteri G, Bondanza A, Naldini L, Roncarolo MG, Bacchetta R (2013) CD4(+) T cells from IPEX patients convert into functional and stable regulatory T cells by FOXP3 gene transfer. Sci Transl Med 5:215ra174PubMedCrossRef
130.
go back to reference Van Gool F, Nguyen MLT, Mumbach MR, Satpathy AT, Rosenthal WL, Giacometti S, Le DT, Liu W, Brusko TM, Anderson MS, Rudensky AY, Marson A, Chang HY, Bluestone JA (2019) A mutation in the transcription factor Foxp3 drives T helper 2 effector function in regulatory T cells. Immunity 50:362–77 e6PubMedPubMedCentralCrossRef Van Gool F, Nguyen MLT, Mumbach MR, Satpathy AT, Rosenthal WL, Giacometti S, Le DT, Liu W, Brusko TM, Anderson MS, Rudensky AY, Marson A, Chang HY, Bluestone JA (2019) A mutation in the transcription factor Foxp3 drives T helper 2 effector function in regulatory T cells. Immunity 50:362–77 e6PubMedPubMedCentralCrossRef
131.
go back to reference Bacchetta R, Passerini L, Gambineri E, Dai M, Allan SE, Perroni L, Dagna-Bricarelli F, Sartirana C, Matthes-Martin S, Lawitschka A, Azzari C, Ziegler SF, Levings MK, Roncarolo MG (2006) Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest 116:1713–1722PubMedPubMedCentralCrossRef Bacchetta R, Passerini L, Gambineri E, Dai M, Allan SE, Perroni L, Dagna-Bricarelli F, Sartirana C, Matthes-Martin S, Lawitschka A, Azzari C, Ziegler SF, Levings MK, Roncarolo MG (2006) Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest 116:1713–1722PubMedPubMedCentralCrossRef
132.
go back to reference Passerini L, Olek S, Di Nunzio S, Barzaghi F, Hambleton S, Abinun M, Tommasini A, Vignola S, Cipolli M, Amendola M, Naldini L, Guidi L, Cecconi M, Roncarolo MG, Bacchetta R (2011) Forkhead box protein 3 (FOXP3) mutations lead to increased TH17 cell numbers and regulatory T-cell instability. J Allergy Clin Immunol 128:1376–9 e1PubMedCrossRef Passerini L, Olek S, Di Nunzio S, Barzaghi F, Hambleton S, Abinun M, Tommasini A, Vignola S, Cipolli M, Amendola M, Naldini L, Guidi L, Cecconi M, Roncarolo MG, Bacchetta R (2011) Forkhead box protein 3 (FOXP3) mutations lead to increased TH17 cell numbers and regulatory T-cell instability. J Allergy Clin Immunol 128:1376–9 e1PubMedCrossRef
133.
go back to reference Lim HW, Hillsamer P, Kim CH (2004) Regulatory T cells can migrate to follicles upon T cell activation and suppress GC-Th cells and GC-Th cell-driven B cell responses. J Clin Invest 114:1640–1649PubMedPubMedCentralCrossRef Lim HW, Hillsamer P, Kim CH (2004) Regulatory T cells can migrate to follicles upon T cell activation and suppress GC-Th cells and GC-Th cell-driven B cell responses. J Clin Invest 114:1640–1649PubMedPubMedCentralCrossRef
134.
go back to reference Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, Wang YH, Lim H, Reynolds JM, Zhou XH, Fan HM, Liu ZM, Neelapu SS, Dong C (2011) Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med 17:983–988PubMedPubMedCentralCrossRef Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, Wang YH, Lim H, Reynolds JM, Zhou XH, Fan HM, Liu ZM, Neelapu SS, Dong C (2011) Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med 17:983–988PubMedPubMedCentralCrossRef
135.
136.
go back to reference Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY (2007) Foxp3-dependent programme of regulatory T-cell differentiation. Nature 445:771–775PubMedCrossRef Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY (2007) Foxp3-dependent programme of regulatory T-cell differentiation. Nature 445:771–775PubMedCrossRef
137.
go back to reference Kuczma M, Podolsky R, Garge N, Daniely D, Pacholczyk R, Ignatowicz L, Kraj P (2009) Foxp3-deficient regulatory T cells do not revert into conventional effector CD4+ T cells but constitute a unique cell subset. J Immunol 183:3731–3741PubMedCrossRef Kuczma M, Podolsky R, Garge N, Daniely D, Pacholczyk R, Ignatowicz L, Kraj P (2009) Foxp3-deficient regulatory T cells do not revert into conventional effector CD4+ T cells but constitute a unique cell subset. J Immunol 183:3731–3741PubMedCrossRef
138.
go back to reference Lin W, Haribhai D, Relland LM, Truong N, Carlson MR, Williams CB, Chatila TA (2007) Regulatory T cell development in the absence of functional Foxp3. Nat Immunol 8:359–368PubMedCrossRef Lin W, Haribhai D, Relland LM, Truong N, Carlson MR, Williams CB, Chatila TA (2007) Regulatory T cell development in the absence of functional Foxp3. Nat Immunol 8:359–368PubMedCrossRef
139.
go back to reference Passerini L, Barzaghi F, Curto R, Sartirana C, Barera G, Tucci F, Albarello L, Mariani A, Testoni PA, Bazzigaluppi E, Bosi E, Lampasona V, Neth O, Zama D, Hoenig M, Schulz A, Seidel MG, Rabbone I, Olek S, Roncarolo MG, Cicalese MP, Aiuti A, Bacchetta R (2020) Treatment with rapamycin can restore regulatory T-cell function in IPEX patients. J Allergy Clin Immunol 145:1262–71 e13PubMedCrossRef Passerini L, Barzaghi F, Curto R, Sartirana C, Barera G, Tucci F, Albarello L, Mariani A, Testoni PA, Bazzigaluppi E, Bosi E, Lampasona V, Neth O, Zama D, Hoenig M, Schulz A, Seidel MG, Rabbone I, Olek S, Roncarolo MG, Cicalese MP, Aiuti A, Bacchetta R (2020) Treatment with rapamycin can restore regulatory T-cell function in IPEX patients. J Allergy Clin Immunol 145:1262–71 e13PubMedCrossRef
140.
go back to reference Charbonnier LM, Cui Y, Stephen-Victor E, Harb H, Lopez D, Bleesing JJ, Garcia-Lloret MI, Chen K, Ozen A, Carmeliet P, Li MO, Pellegrini M, Chatila TA (2019) Functional reprogramming of regulatory T cells in the absence of Foxp3. Nat Immunol 20:1208–1219PubMedPubMedCentralCrossRef Charbonnier LM, Cui Y, Stephen-Victor E, Harb H, Lopez D, Bleesing JJ, Garcia-Lloret MI, Chen K, Ozen A, Carmeliet P, Li MO, Pellegrini M, Chatila TA (2019) Functional reprogramming of regulatory T cells in the absence of Foxp3. Nat Immunol 20:1208–1219PubMedPubMedCentralCrossRef
141.
go back to reference Vella A, Cooper JD, Lowe CE, Walker N, Nutland S, Widmer B, Jones R, Ring SM, McArdle W, Pembrey ME, Strachan DP, Dunger DB, Twells RC, Clayton DG, Todd JA (2005) Localization of a type 1 diabetes locus in the IL2RA/CD25 region by use of tag single-nucleotide polymorphisms. Am J Hum Genet 76:773–779PubMedPubMedCentralCrossRef Vella A, Cooper JD, Lowe CE, Walker N, Nutland S, Widmer B, Jones R, Ring SM, McArdle W, Pembrey ME, Strachan DP, Dunger DB, Twells RC, Clayton DG, Todd JA (2005) Localization of a type 1 diabetes locus in the IL2RA/CD25 region by use of tag single-nucleotide polymorphisms. Am J Hum Genet 76:773–779PubMedPubMedCentralCrossRef
142.
go back to reference Bakhtiar S, Fekadu J, Seidel MG, Gambineri E (2019) Allogeneic hematopoietic stem cell transplantation for congenital immune dysregulatory disorders. Front Pediatr 7:461vCrossRef Bakhtiar S, Fekadu J, Seidel MG, Gambineri E (2019) Allogeneic hematopoietic stem cell transplantation for congenital immune dysregulatory disorders. Front Pediatr 7:461vCrossRef
144.
go back to reference Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, Bulashevska A, Petersen BS, Schaffer AA, Gruning BA, Unger S, Frede N, Baumann U, Witte T, Schmidt RE, Dueckers G, Niehues T, Seneviratne S, Kanariou M, Speckmann C, Ehl S, Rensing-Ehl A, Warnatz K, Rakhmanov M, Thimme R, Hasselblatt P, Emmerich F, Cathomen T, Backofen R, Fisch P, Seidl M, May A, Schmitt-Graeff A, Ikemizu S, Salzer U, Franke A, Sakaguchi S, Walker LSK, Sansom DM, Grimbacher B (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20:1410–1416PubMedPubMedCentralCrossRef Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, Bulashevska A, Petersen BS, Schaffer AA, Gruning BA, Unger S, Frede N, Baumann U, Witte T, Schmidt RE, Dueckers G, Niehues T, Seneviratne S, Kanariou M, Speckmann C, Ehl S, Rensing-Ehl A, Warnatz K, Rakhmanov M, Thimme R, Hasselblatt P, Emmerich F, Cathomen T, Backofen R, Fisch P, Seidl M, May A, Schmitt-Graeff A, Ikemizu S, Salzer U, Franke A, Sakaguchi S, Walker LSK, Sansom DM, Grimbacher B (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20:1410–1416PubMedPubMedCentralCrossRef
145.
go back to reference Charbonnier LM, Janssen E, Chou J, Ohsumi TK, Keles S, Hsu JT, Massaad MJ, Garcia-Lloret M, Hanna-Wakim R, Dbaibo G, Alangari AA, Alsultan A, Al-Zahrani D, Geha RS, Chatila TA (2015) Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA. J Allergy Clin Immunol 135:217–227PubMedCrossRef Charbonnier LM, Janssen E, Chou J, Ohsumi TK, Keles S, Hsu JT, Massaad MJ, Garcia-Lloret M, Hanna-Wakim R, Dbaibo G, Alangari AA, Alsultan A, Al-Zahrani D, Geha RS, Chatila TA (2015) Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA. J Allergy Clin Immunol 135:217–227PubMedCrossRef
146.
go back to reference Schwab C, Gabrysch A, Olbrich P, Patino V, Warnatz K, Wolff D, Hoshino A, Kobayashi M, Imai K, Takagi M, Dybedal I, Haddock JA, Sansom DM, Lucena JM, Seidl M, Schmitt-Graeff A, Reiser V, Emmerich F, Frede N, Bulashevska A, Salzer U, Schubert D, Hayakawa S, Okada S, Kanariou M, Kucuk ZY, Chapdelaine H, Petruzelkova L, Sumnik Z, Sediva A, Slatter M, Arkwright PD, Cant A, Lorenz HM, Giese T, Lougaris V, Plebani A, Price C, Sullivan KE, Moutschen M, Litzman J, Freiberger T, van de Veerdonk FL, Recher M, Albert MH, Hauck F, Seneviratne S, Pachlopnik Schmid J, Kolios A, Unglik G, Klemann C, Speckmann C, Ehl S, Leichtner A, Blumberg R, Franke A, Snapper S, Zeissig S, Cunningham-Rundles C, Giulino-Roth L, Elemento O, Duckers G, Niehues T, Fronkova E, Kanderova V, Platt CD, Chou J, Chatila TA, Geha R, McDermott E, Bunn S, Kurzai M, Schulz A, Alsina L, Casals F, Deya-Martinez A, Hambleton S, Kanegane H, Tasken K, Neth O, Grimbacher B (2018) Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol 142:1932–1946PubMedPubMedCentralCrossRef Schwab C, Gabrysch A, Olbrich P, Patino V, Warnatz K, Wolff D, Hoshino A, Kobayashi M, Imai K, Takagi M, Dybedal I, Haddock JA, Sansom DM, Lucena JM, Seidl M, Schmitt-Graeff A, Reiser V, Emmerich F, Frede N, Bulashevska A, Salzer U, Schubert D, Hayakawa S, Okada S, Kanariou M, Kucuk ZY, Chapdelaine H, Petruzelkova L, Sumnik Z, Sediva A, Slatter M, Arkwright PD, Cant A, Lorenz HM, Giese T, Lougaris V, Plebani A, Price C, Sullivan KE, Moutschen M, Litzman J, Freiberger T, van de Veerdonk FL, Recher M, Albert MH, Hauck F, Seneviratne S, Pachlopnik Schmid J, Kolios A, Unglik G, Klemann C, Speckmann C, Ehl S, Leichtner A, Blumberg R, Franke A, Snapper S, Zeissig S, Cunningham-Rundles C, Giulino-Roth L, Elemento O, Duckers G, Niehues T, Fronkova E, Kanderova V, Platt CD, Chou J, Chatila TA, Geha R, McDermott E, Bunn S, Kurzai M, Schulz A, Alsina L, Casals F, Deya-Martinez A, Hambleton S, Kanegane H, Tasken K, Neth O, Grimbacher B (2018) Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol 142:1932–1946PubMedPubMedCentralCrossRef
147.
go back to reference Slatter MA, Engelhardt KR, Burroughs LM, Arkwright PD, Nademi Z, Skoda-Smith S, Hagin D, Kennedy A, Barge D, Flood T, Abinun M, Wynn RF, Gennery AR, Cant AJ, Sansom D, Hambleton S, Torgerson TR (2016) Hematopoietic stem cell transplantation for CTLA4 deficiency. J Allergy Clin Immunol 138:615–9 e1PubMedCrossRef Slatter MA, Engelhardt KR, Burroughs LM, Arkwright PD, Nademi Z, Skoda-Smith S, Hagin D, Kennedy A, Barge D, Flood T, Abinun M, Wynn RF, Gennery AR, Cant AJ, Sansom D, Hambleton S, Torgerson TR (2016) Hematopoietic stem cell transplantation for CTLA4 deficiency. J Allergy Clin Immunol 138:615–9 e1PubMedCrossRef
148.
go back to reference Tesch VK, Abolhassani H, Shadur B, Zobel J, Mareika Y, Sharapova S, Karakoc-Aydiner E, Riviere JG, Garcia-Prat M, Moes N, Haerynck F, Gonzales-Granado LI, Santos Perez JL, Mukhina A, Shcherbina A, Aghamohammadi A, Hammarstrom L, Dogu F, Haskologlu S, Ikinciogullari AI, Kostel Bal S, Baris S, Kilic SS, Karaca NE, Kutukculer N, Girschick H, Kolios A, Keles S, Uygun V, Stepensky P, Worth A, van Montfrans JM, Peters AMJ, Meyts I, Adeli M, Marzollo A, Padem N, Khojah AM, Chavoshzadeh Z, Avbelj Stefanija M, Bakhtiar S, Florkin B, Meeths M, Gamez L, Grimbacher B, Seppanen MRJ, Lankester A, Gennery AR, Seidel MG, Inborn Errors C, Registry Working Parties of the European Society for B, Marrow T, the European Society for I (2020) Long-term outcome of LRBA deficiency in 76 patients after various treatment modalities as evaluated by the immune deficiency and dysregulation activity (IDDA) score. J Allergy Clin Immunol 145:1452–1463PubMedCrossRef Tesch VK, Abolhassani H, Shadur B, Zobel J, Mareika Y, Sharapova S, Karakoc-Aydiner E, Riviere JG, Garcia-Prat M, Moes N, Haerynck F, Gonzales-Granado LI, Santos Perez JL, Mukhina A, Shcherbina A, Aghamohammadi A, Hammarstrom L, Dogu F, Haskologlu S, Ikinciogullari AI, Kostel Bal S, Baris S, Kilic SS, Karaca NE, Kutukculer N, Girschick H, Kolios A, Keles S, Uygun V, Stepensky P, Worth A, van Montfrans JM, Peters AMJ, Meyts I, Adeli M, Marzollo A, Padem N, Khojah AM, Chavoshzadeh Z, Avbelj Stefanija M, Bakhtiar S, Florkin B, Meeths M, Gamez L, Grimbacher B, Seppanen MRJ, Lankester A, Gennery AR, Seidel MG, Inborn Errors C, Registry Working Parties of the European Society for B, Marrow T, the European Society for I (2020) Long-term outcome of LRBA deficiency in 76 patients after various treatment modalities as evaluated by the immune deficiency and dysregulation activity (IDDA) score. J Allergy Clin Immunol 145:1452–1463PubMedCrossRef
149.
go back to reference Goodwin M, Lee E, Lakshmanan U, Shipp S, Froessl L, Barzaghi F, Passerini L, Narula M, Sheikali A, Lee CM, Bao G, Bauer CS, Miller HK, Garcia-Lloret M, Butte MJ, Bertaina A, Shah A, Pavel-Dinu M, Hendel A, Porteus M, Roncarolo MG, Bacchetta R (2020) CRISPR-based gene editing enables FOXP3 gene repair in IPEX patient cells. Sci Adv 6:eaaz0571PubMedPubMedCentralCrossRef Goodwin M, Lee E, Lakshmanan U, Shipp S, Froessl L, Barzaghi F, Passerini L, Narula M, Sheikali A, Lee CM, Bao G, Bauer CS, Miller HK, Garcia-Lloret M, Butte MJ, Bertaina A, Shah A, Pavel-Dinu M, Hendel A, Porteus M, Roncarolo MG, Bacchetta R (2020) CRISPR-based gene editing enables FOXP3 gene repair in IPEX patient cells. Sci Adv 6:eaaz0571PubMedPubMedCentralCrossRef
150.
go back to reference Masiuk KE, Laborada J, Roncarolo MG, Hollis RP, Kohn DB (2019) Lentiviral gene therapy in HSCs restores lineage-specific Foxp3 expression and suppresses autoimmunity in a mouse model of IPEX syndrome. Cell Stem Cell 24:309–17 e7PubMedCrossRef Masiuk KE, Laborada J, Roncarolo MG, Hollis RP, Kohn DB (2019) Lentiviral gene therapy in HSCs restores lineage-specific Foxp3 expression and suppresses autoimmunity in a mouse model of IPEX syndrome. Cell Stem Cell 24:309–17 e7PubMedCrossRef
151.
go back to reference Roth TL, Puig-Saus C, Yu R, Shifrut E, Carnevale J, Li PJ, Hiatt J, Saco J, Krystofinski P, Li H, Tobin V, Nguyen DN, Lee MR, Putnam AL, Ferris AL, Chen JW, Schickel JN, Pellerin L, Carmody D, Alkorta-Aranburu G, Del Gaudio D, Matsumoto H, Morell M, Mao Y, Cho M, Quadros RM, Gurumurthy CB, Smith B, Haugwitz M, Hughes SH, Weissman JS, Schumann K, Esensten JH, May AP, Ashworth A, Kupfer GM, Greeley SAW, Bacchetta R, Meffre E, Roncarolo MG, Romberg N, Herold KC, Ribas A, Leonetti MD, Marson A (2018) Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559:405–409PubMedPubMedCentralCrossRef Roth TL, Puig-Saus C, Yu R, Shifrut E, Carnevale J, Li PJ, Hiatt J, Saco J, Krystofinski P, Li H, Tobin V, Nguyen DN, Lee MR, Putnam AL, Ferris AL, Chen JW, Schickel JN, Pellerin L, Carmody D, Alkorta-Aranburu G, Del Gaudio D, Matsumoto H, Morell M, Mao Y, Cho M, Quadros RM, Gurumurthy CB, Smith B, Haugwitz M, Hughes SH, Weissman JS, Schumann K, Esensten JH, May AP, Ashworth A, Kupfer GM, Greeley SAW, Bacchetta R, Meffre E, Roncarolo MG, Romberg N, Herold KC, Ribas A, Leonetti MD, Marson A (2018) Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559:405–409PubMedPubMedCentralCrossRef
152.
go back to reference Anderson MS, Venanzi ES, Chen Z, Berzins SP, Benoist C, Mathis D (2005) The cellular mechanism of Aire control of T cell tolerance. Immunity 23:227–239PubMedCrossRef Anderson MS, Venanzi ES, Chen Z, Berzins SP, Benoist C, Mathis D (2005) The cellular mechanism of Aire control of T cell tolerance. Immunity 23:227–239PubMedCrossRef
154.
go back to reference Watkin LB, Jessen B, Wiszniewski W, Vece TJ, Jan M, Sha Y, Thamsen M, Santos-Cortez RL, Lee K, Gambin T, Forbes LR, Law CS, Stray-Pedersen A, Cheng MH, Mace EM, Anderson MS, Liu D, Tang LF, Nicholas SK, Nahmod K, Makedonas G, Canter DL, Kwok PY, Hicks J, Jones KD, Penney S, Jhangiani SN, Rosenblum MD, Dell SD, Waterfield MR, Papa FR, Muzny DM, Zaitlen N, Leal SM, Gonzaga-Jauregui C, Baylor-Hopkins Center for Mendelian G, Boerwinkle E, Eissa NT, Gibbs RA, Lupski JR, Orange JS, Shum AK. (2015) COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet 47:654–660PubMedPubMedCentralCrossRef Watkin LB, Jessen B, Wiszniewski W, Vece TJ, Jan M, Sha Y, Thamsen M, Santos-Cortez RL, Lee K, Gambin T, Forbes LR, Law CS, Stray-Pedersen A, Cheng MH, Mace EM, Anderson MS, Liu D, Tang LF, Nicholas SK, Nahmod K, Makedonas G, Canter DL, Kwok PY, Hicks J, Jones KD, Penney S, Jhangiani SN, Rosenblum MD, Dell SD, Waterfield MR, Papa FR, Muzny DM, Zaitlen N, Leal SM, Gonzaga-Jauregui C, Baylor-Hopkins Center for Mendelian G, Boerwinkle E, Eissa NT, Gibbs RA, Lupski JR, Orange JS, Shum AK. (2015) COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet 47:654–660PubMedPubMedCentralCrossRef
155.
go back to reference Vece TJ, Watkin LB, Nicholas S, Canter D, Braun MC, Guillerman RP, Eldin KW, Bertolet G, McKinley S, de Guzman M, Forbes L, Chinn I, Orange JS (2016) Copa syndrome: a novel autosomal dominant immune dysregulatory disease. J Clin Immunol 36:377–387PubMedPubMedCentralCrossRef Vece TJ, Watkin LB, Nicholas S, Canter D, Braun MC, Guillerman RP, Eldin KW, Bertolet G, McKinley S, de Guzman M, Forbes L, Chinn I, Orange JS (2016) Copa syndrome: a novel autosomal dominant immune dysregulatory disease. J Clin Immunol 36:377–387PubMedPubMedCentralCrossRef
156.
go back to reference Deng Z, Law CS, Ho FO, Wang KM, Jones KD, Shin JS, Shum AK (2020) A defect in thymic tolerance causes T cell-mediated autoimmunity in a murine model of COPA syndrome. J Immunol 204:2360–2373PubMedCrossRef Deng Z, Law CS, Ho FO, Wang KM, Jones KD, Shin JS, Shum AK (2020) A defect in thymic tolerance causes T cell-mediated autoimmunity in a murine model of COPA syndrome. J Immunol 204:2360–2373PubMedCrossRef
157.
go back to reference Lepelley A, Martin-Niclos MJ, Le Bihan M, Marsh JA, Uggenti C, Rice GI, Bondet V, Duffy D, Hertzog J, Rehwinkel J, Amselem S, Boulisfane-El Khalifi S, Brennan M, Carter E, Chatenoud L, Chhun S, Coulomb l’Hermine A, Depp M, Legendre M, Mackenzie KJ, Marey J, McDougall C, McKenzie KJ, Molina TJ, Neven B, Seabra L, Thumerelle C, Wislez M, Nathan N, Manel N, Crow YJ, Fremond ML (2020) Mutations in COPA lead to abnormal trafficking of STING to the Golgi and interferon signaling. J Exp Med 217:e20200600PubMedPubMedCentralCrossRef Lepelley A, Martin-Niclos MJ, Le Bihan M, Marsh JA, Uggenti C, Rice GI, Bondet V, Duffy D, Hertzog J, Rehwinkel J, Amselem S, Boulisfane-El Khalifi S, Brennan M, Carter E, Chatenoud L, Chhun S, Coulomb l’Hermine A, Depp M, Legendre M, Mackenzie KJ, Marey J, McDougall C, McKenzie KJ, Molina TJ, Neven B, Seabra L, Thumerelle C, Wislez M, Nathan N, Manel N, Crow YJ, Fremond ML (2020) Mutations in COPA lead to abnormal trafficking of STING to the Golgi and interferon signaling. J Exp Med 217:e20200600PubMedPubMedCentralCrossRef
158.
go back to reference Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, van der Vliet WA, Huys EH, de Jong PJ, Hamel BC, Schoenmakers EF, Brunner HG, Veltman JA, van Kessel AG (2004) Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet 36:955–957PubMedCrossRef Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, van der Vliet WA, Huys EH, de Jong PJ, Hamel BC, Schoenmakers EF, Brunner HG, Veltman JA, van Kessel AG (2004) Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet 36:955–957PubMedCrossRef
159.
go back to reference Gennery AR, Slatter MA, Rice J, Hoefsloot LH, Barge D, McLean-Tooke A, Montgomery T, Goodship JA, Burt AD, Flood TJ, Abinun M, Cant AJ, Johnson D (2008) Mutations in CHD7 in patients with CHARGE syndrome cause T-B + natural killer cell + severe combined immune deficiency and may cause Omenn-like syndrome. Clin Exp Immunol 153:75–80PubMedPubMedCentralCrossRef Gennery AR, Slatter MA, Rice J, Hoefsloot LH, Barge D, McLean-Tooke A, Montgomery T, Goodship JA, Burt AD, Flood TJ, Abinun M, Cant AJ, Johnson D (2008) Mutations in CHD7 in patients with CHARGE syndrome cause T-B + natural killer cell + severe combined immune deficiency and may cause Omenn-like syndrome. Clin Exp Immunol 153:75–80PubMedPubMedCentralCrossRef
160.
go back to reference Guris DL, Duester G, Papaioannou VE, Imamoto A (2006) Dose-dependent interaction of Tbx1 and Crkl and locally aberrant RA signaling in a model of del22q11 syndrome. Dev Cell 10:81–92PubMedCrossRef Guris DL, Duester G, Papaioannou VE, Imamoto A (2006) Dose-dependent interaction of Tbx1 and Crkl and locally aberrant RA signaling in a model of del22q11 syndrome. Dev Cell 10:81–92PubMedCrossRef
161.
go back to reference Zemble R, Luning Prak E, McDonald K, McDonald-McGinn D, Zackai E, Sullivan K (2010) Secondary immunologic consequences in chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome). Clin Immunol 136:409–418PubMedPubMedCentralCrossRef Zemble R, Luning Prak E, McDonald K, McDonald-McGinn D, Zackai E, Sullivan K (2010) Secondary immunologic consequences in chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome). Clin Immunol 136:409–418PubMedPubMedCentralCrossRef
162.
go back to reference Deshpande DR, Demirdag YY, Marsh RA, Sullivan KE, Orange JS, Consortium U. 2020. Relationship between severity of T cell lymphopenia and immune dysregulation in patients with DiGeorge syndrome (22q11.2 Deletions and/or Related TBX1 Mutations): a USIDNET Study. J Clin Immunol Deshpande DR, Demirdag YY, Marsh RA, Sullivan KE, Orange JS, Consortium U. 2020. Relationship between severity of T cell lymphopenia and immune dysregulation in patients with DiGeorge syndrome (22q11.2 Deletions and/or Related TBX1 Mutations): a USIDNET Study. J Clin Immunol
163.
go back to reference Staple L, Andrews T, McDonald-McGinn D, Zackai E, Sullivan KE (2005) Allergies in patients with chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome) and patients with chronic granulomatous disease. Pediatr Allergy Immunol 16:226–230PubMedCrossRef Staple L, Andrews T, McDonald-McGinn D, Zackai E, Sullivan KE (2005) Allergies in patients with chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome) and patients with chronic granulomatous disease. Pediatr Allergy Immunol 16:226–230PubMedCrossRef
164.
go back to reference Di Cesare S, Puliafito P, Ariganello P, Marcovecchio GE, Mandolesi M, Capolino R, Digilio MC, Aiuti A, Rossi P, Cancrini C (2015) Autoimmunity and regulatory T cells in 22q11.2 deletion syndrome patients. Pediatr Allergy Immunol 26:591–594PubMedCrossRef Di Cesare S, Puliafito P, Ariganello P, Marcovecchio GE, Mandolesi M, Capolino R, Digilio MC, Aiuti A, Rossi P, Cancrini C (2015) Autoimmunity and regulatory T cells in 22q11.2 deletion syndrome patients. Pediatr Allergy Immunol 26:591–594PubMedCrossRef
165.
go back to reference Marcovecchio GE, Bortolomai I, Ferrua F, Fontana E, Imberti L, Conforti E, Amodio D, Bergante S, Macchiarulo G, D’Oria V, Conti F, Di Cesare S, Fousteri G, Carotti A, Giamberti A, Poliani PL, Notarangelo LD, Cancrini C, Villa A, Bosticardo M (2019) Thymic epithelium abnormalities in DiGeorge and Down syndrome patients contribute to dysregulation in T cell development. Front Immunol 10:447PubMedPubMedCentralCrossRef Marcovecchio GE, Bortolomai I, Ferrua F, Fontana E, Imberti L, Conforti E, Amodio D, Bergante S, Macchiarulo G, D’Oria V, Conti F, Di Cesare S, Fousteri G, Carotti A, Giamberti A, Poliani PL, Notarangelo LD, Cancrini C, Villa A, Bosticardo M (2019) Thymic epithelium abnormalities in DiGeorge and Down syndrome patients contribute to dysregulation in T cell development. Front Immunol 10:447PubMedPubMedCentralCrossRef
166.
go back to reference Chinn IK, Milner JD, Scheinberg P, Douek DC, Markert ML (2013) Thymus transplantation restores the repertoires of forkhead box protein 3 (FoxP3)+ and FoxP3- T cells in complete DiGeorge anomaly. Clin Exp Immunol 173:140–149PubMedPubMedCentralCrossRef Chinn IK, Milner JD, Scheinberg P, Douek DC, Markert ML (2013) Thymus transplantation restores the repertoires of forkhead box protein 3 (FoxP3)+ and FoxP3- T cells in complete DiGeorge anomaly. Clin Exp Immunol 173:140–149PubMedPubMedCentralCrossRef
167.
go back to reference Ucar O, Tykocinski LO, Dooley J, Liston A, Kyewski B (2013) An evolutionarily conserved mutual interdependence between Aire and microRNAs in promiscuous gene expression. Eur J Immunol 43:1769–1778PubMedPubMedCentralCrossRef Ucar O, Tykocinski LO, Dooley J, Liston A, Kyewski B (2013) An evolutionarily conserved mutual interdependence between Aire and microRNAs in promiscuous gene expression. Eur J Immunol 43:1769–1778PubMedPubMedCentralCrossRef
168.
go back to reference Khan IS, Taniguchi RT, Fasano KJ, Anderson MS, Jeker LT (2014) Canonical microRNAs in thymic epithelial cells promote central tolerance. Eur J Immunol 44:1313–1319PubMedPubMedCentralCrossRef Khan IS, Taniguchi RT, Fasano KJ, Anderson MS, Jeker LT (2014) Canonical microRNAs in thymic epithelial cells promote central tolerance. Eur J Immunol 44:1313–1319PubMedPubMedCentralCrossRef
169.
go back to reference Bernard C, Frih H, Pasquet F, Kerever S, Jamilloux Y, Tronc F, Guibert B, Isaac S, Devouassoux M, Chalabreysse L, Broussolle C, Petiot P, Girard N, Seve P (2016) Thymoma associated with autoimmune diseases: 85 cases and literature review. Autoimmun Rev 15:82–92PubMedCrossRef Bernard C, Frih H, Pasquet F, Kerever S, Jamilloux Y, Tronc F, Guibert B, Isaac S, Devouassoux M, Chalabreysse L, Broussolle C, Petiot P, Girard N, Seve P (2016) Thymoma associated with autoimmune diseases: 85 cases and literature review. Autoimmun Rev 15:82–92PubMedCrossRef
170.
go back to reference Cheng MH, Fan U, Grewal N, Barnes M, Mehta A, Taylor S, Husebye ES, Murphy EJ, Anderson MS (2010) Acquired autoimmune polyglandular syndrome, thymoma, and an AIRE defect. N Engl J Med 362:764–766PubMedPubMedCentralCrossRef Cheng MH, Fan U, Grewal N, Barnes M, Mehta A, Taylor S, Husebye ES, Murphy EJ, Anderson MS (2010) Acquired autoimmune polyglandular syndrome, thymoma, and an AIRE defect. N Engl J Med 362:764–766PubMedPubMedCentralCrossRef
171.
go back to reference Ferre EMN, Break TJ, Burbelo PD, Allgauer M, Kleiner DE, Jin D, Xu Z, Folio LR, Mollura DJ, Swamydas M, Gu W, Hunsberger S, Lee CR, Bondici A, Hoffman KW, Lim JK, Dobbs K, Niemela JE, Fleisher TA, Hsu AP, Snow LN, Darnell DN, Ojaimi S, Cooper MA, Bozzola M, Kleiner GI, Martinez JC, Deterding RR, Kuhns DB, Heller T, Winer KK, Rajan A, Holland SM, Notarangelo LD, Fennelly KP, Olivier KN, Lionakis MS. 2019. Lymphocyte-driven regional immunopathology in pneumonitis caused by impaired central immune tolerance. Sci Transl Med 11 Ferre EMN, Break TJ, Burbelo PD, Allgauer M, Kleiner DE, Jin D, Xu Z, Folio LR, Mollura DJ, Swamydas M, Gu W, Hunsberger S, Lee CR, Bondici A, Hoffman KW, Lim JK, Dobbs K, Niemela JE, Fleisher TA, Hsu AP, Snow LN, Darnell DN, Ojaimi S, Cooper MA, Bozzola M, Kleiner GI, Martinez JC, Deterding RR, Kuhns DB, Heller T, Winer KK, Rajan A, Holland SM, Notarangelo LD, Fennelly KP, Olivier KN, Lionakis MS. 2019. Lymphocyte-driven regional immunopathology in pneumonitis caused by impaired central immune tolerance. Sci Transl Med 11
172.
go back to reference Wolff AS, Karner J, Owe JF, Oftedal BE, Gilhus NE, Erichsen MM, Kampe O, Meager A, Peterson P, Kisand K, Willcox N, Husebye ES (2014) Clinical and serologic parallels to APS-I in patients with thymomas and autoantigen transcripts in their tumors. J Immunol 193:3880–3890PubMedCrossRef Wolff AS, Karner J, Owe JF, Oftedal BE, Gilhus NE, Erichsen MM, Kampe O, Meager A, Peterson P, Kisand K, Willcox N, Husebye ES (2014) Clinical and serologic parallels to APS-I in patients with thymomas and autoantigen transcripts in their tumors. J Immunol 193:3880–3890PubMedCrossRef
173.
go back to reference Hapnes L, Willcox N, Oftedal BE, Owe JF, Gilhus NE, Meager A, Husebye ES, Wolff AS (2012) Radioligand-binding assay reveals distinct autoantibody preferences for type I interferons in APS I and myasthenia gravis subgroups. J Clin Immunol 32:230–237PubMedCrossRef Hapnes L, Willcox N, Oftedal BE, Owe JF, Gilhus NE, Meager A, Husebye ES, Wolff AS (2012) Radioligand-binding assay reveals distinct autoantibody preferences for type I interferons in APS I and myasthenia gravis subgroups. J Clin Immunol 32:230–237PubMedCrossRef
175.
go back to reference Ukena SN, Grosse J, Mischak-Weissinger E, Buchholz S, Stadler M, Ganser A, Franzke A (2011) Acute but not chronic graft-versus-host disease is associated with a reduction of circulating CD4(+)CD25 (high)CD127 (low/-) regulatory T cells. Ann Hematol 90:213–218PubMedCrossRef Ukena SN, Grosse J, Mischak-Weissinger E, Buchholz S, Stadler M, Ganser A, Franzke A (2011) Acute but not chronic graft-versus-host disease is associated with a reduction of circulating CD4(+)CD25 (high)CD127 (low/-) regulatory T cells. Ann Hematol 90:213–218PubMedCrossRef
176.
go back to reference Seemayer TA, Lapp WS, Bolande RP (1977) Thymic involution in murine graft-versus-host reaction. Epithelial injury mimicking human thymic dysplasia. Am J Pathol 88:119–134PubMedPubMedCentral Seemayer TA, Lapp WS, Bolande RP (1977) Thymic involution in murine graft-versus-host reaction. Epithelial injury mimicking human thymic dysplasia. Am J Pathol 88:119–134PubMedPubMedCentral
177.
go back to reference Seddik M, Seemayer TA, Lapp WS (1980) T cell functional defect associated with thymid epithelial cell injury induced by a graft-versus-host reaction. Transplantation 29:61–66PubMedCrossRef Seddik M, Seemayer TA, Lapp WS (1980) T cell functional defect associated with thymid epithelial cell injury induced by a graft-versus-host reaction. Transplantation 29:61–66PubMedCrossRef
178.
go back to reference Rossi S, Blazar BR, Farrell CL, Danilenko DM, Lacey DL, Weinberg KI, Krenger W, Hollander GA (2002) Keratinocyte growth factor preserves normal thymopoiesis and thymic microenvironment during experimental graft-versus-host disease. Blood 100:682–691PubMedCrossRef Rossi S, Blazar BR, Farrell CL, Danilenko DM, Lacey DL, Weinberg KI, Krenger W, Hollander GA (2002) Keratinocyte growth factor preserves normal thymopoiesis and thymic microenvironment during experimental graft-versus-host disease. Blood 100:682–691PubMedCrossRef
179.
go back to reference Dertschnig S, Hauri-Hohl MM, Vollmer M, Hollander GA, Krenger W (2015) Impaired thymic expression of tissue-restricted antigens licenses the de novo generation of autoreactive CD4+ T cells in acute GVHD. Blood 125:2720–2723PubMedPubMedCentralCrossRef Dertschnig S, Hauri-Hohl MM, Vollmer M, Hollander GA, Krenger W (2015) Impaired thymic expression of tissue-restricted antigens licenses the de novo generation of autoreactive CD4+ T cells in acute GVHD. Blood 125:2720–2723PubMedPubMedCentralCrossRef
Metadata
Title
Thymic origins of autoimmunity—lessons from inborn errors of immunity
Authors
Rosa Bacchetta
Kenneth Weinberg
Publication date
01-02-2021
Publisher
Springer Berlin Heidelberg
Published in
Seminars in Immunopathology / Issue 1/2021
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-020-00835-8

Other articles of this Issue 1/2021

Seminars in Immunopathology 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.