Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 6.
Published in final edited form as: Curr Opin Pediatr. 2013 Dec;25(6):708–714. doi: 10.1097/MOP.0000000000000029

Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-linked (IPEX) and IPEX-Related Disorders: an Evolving Web of Heritable Autoimmune Diseases

James W Verbsky 1, Talal A Chatila 2
PMCID: PMC4047515  NIHMSID: NIHMS585742  PMID: 24240290

Abstract

Purpose of review

To summarize recent progress in our understanding of Immune Dysregulation, Polyendocrinopathy, enteropathy, X-linked (IPEX) and IPEX-related disorders.

Recent findings

A number of Mendelian disorders of immune dysregulation and autoimmunity have been noted to result from defects in T regulatory (TR) cell development and function. The best characterized of these is Immune Dysregulation, Polyendocrinopathy, enteropathy, X-linked (IPEX), resulting from mutations affecting FOXP3. A number of other gene defects that affect TR cell function also give rise to IPEX-related phenotypes, including loss of function mutations in CD25 and STAT5b and ITCH. Recent progress includes the identification of gain of function mutations in STAT1 as a cause of an IPEX-like disease, emerging FOXP3 genotype/phenotype relationships in IPEX, and the elucidation of a role for the microbiota in the immune dysregulation associated with regulatory T cell deficiency.

Summary

An expanding spectrum of genetic defects that compromise TR cell function underlies human disorders of immune dysregulation and autoimmunity. Collectively, these disorders offer novel insights into pathways of peripheral tolerance and their disruption in autoimmunity.

Keywords: FOXP3, CD25, STAT5, STAT1, ITCH, T regulatory cells, autoimmunity, Tolerance

Introduction

A number of Mendelian disorders of immune dysregulation and autoimmunity have been noted to result from defects in T regulatory cells development and function. The best-characterized of these is Immune Dysregulation, Polyendocrinopathy, enteropathy, X-linked (IPEX), resulting from mutations affecting FOXP3. A number of other gene defects that affect TR cell function also give rise to an IPEX-like phenotype, including loss of function mutations in CD25, STAT5b and ITCH and gain of function mutations in STAT1 (Table 1). This review highlights recent progress in the study of these disorders and the identification of underlying genetic and functional causative abnormalities.

Table 1.

Clinical and laboratory features of IPEX and IPEX-like disorders.

IPEX CD25 Stat5b STAT-1 ITCH

Autoimmunity
Ezcema +++ +++ ++ ++ ++
Enteropathy +++ +++ ++ ++ ++
Endocrinopathy +++ ++ + ++ ++
Allergic Disease +++ + + ++ ++
Cytopenias ++ ++ ++

Lung Disease + ++ +++ + +++

Infections
Yeast ++ +++
Herpes virus +++(EBV/CMV) ++(VZV) ++
Bacterial +/− ++ ++ ++ +

Associated features None None Growth failure Vascular anomalies Dysmorphic Growth failure

Serum Immunoglobulins Elevated Elevated or normal Elevated or normal Low, normal or high Elevated

Serum IgE Elevated Normal or elevated Normal or elevated Normal or mildly elevated Elevated

CD25 expression Normal Absent Normal or low Normal Not tested

CD4+CD45RO Elevated Elevated Elevated Normal or high Not tested

Foxp3 expression Absent or normal Normal or low Normal or low Normal Not tested

IGF-1, IGFBP-3 Normal Normal Low Normal Not tested

Prolactin Normal Normal Elevated Normal Not tested

EBV, Epstein Barr Virus; CMV, cytomegalovirus; VZV, varicella zoster virus

IPEX

IPEX is a heritable autoimmune lymphoproliferative disease caused by loss of function mutation in FOXP3 [13] [reviewed in [46]]. In most patients, IPEX presents early in life with a triad of autoimmune enteropathy, autoimmune endocrinopathy, and eczematous dermatitis [15]. Other autoimmune phenomena include autoimmune cytopenias, liver, and kidney disease. Allergic dysregulation with eczema and food allergy is common, with extremely elevated IgE levels accompanied by intense peripheral eosinophilia and evidence of overt Th2 skewing [1,7].

The immunopathogenesis of IPEX relates to the loss of functional CD4+CD25+ T regulatory (TR) cells, a subset that is critical to the prevention of autoimmunity [8,9]. The majority of TR cells are generated in the thymus, express the transcription factor Foxp3, are selected on high affinity TCR interactions with self antigens, and are referred to as natural TR (nTR) cells[10] [11,12]. Some Foxp3+ TR cells can be also be induced de novo in vivo from peripheral Foxp3 CD4+ cells resulting in a population of cells (iTR cells) that have regulatory properties but with a TCR repertoire distinct from nTR cells [13,14]. This process can also be recapitulated in vitro by TCR activation of naïve CD4+ T cells in the presence of TGF-β and IL-2. iTR cells are particularly enriched at the mucosal surfaces, especially in the gastrointestinal tract, where they are endowed with a TCR repertoire specific for bacterial antigens [15,16]. nTR and iTR cells act in synergy to induce peripheral tolerance [13]. Some iTR cell populations can lose expression of Foxp3 and become effector cells, including Th1 and Th17 cells [17].

Several suppressive mechanisms for TR cells have been demonstrated. These include CTLA4 engagement of B7 molecules on target cells [18,19], expression of immunosuppressive cytokines such as IL-10, TGF-β, and IL-35 [2022], cytotoxicity of target cells through the perforin/granzyme pathway [23], induction of indoleamine 2,3-dioxygenase (IDO) and the catabolism of tryptophan in target cells, as well as consumption of adenosine by expression of CD73, and competition with effector T cells for IL-2 since TR cells constitutively express the high affinity IL-2 receptor CD25 [2427]. Several of these pathways are targeted by mutations in human subjects, including the IL-2 receptor alpha chain (CD25; discussed below), IL-10/IL-10 receptor, and the perforin/granzyme pathway [2830]. Abnormal TR cell function is a key feature of these diseases [31].

Foxp3 is a transcription factor with a winged helix (Forkhead) DNA binding domain, a proline rich N-terminal domain, a C2H2 zinc finger motif, and a leucine zipper domain. The leucine zipper domain has been implicated in the formation homo and heterodimers and higher order assembly of Foxp3 complexes [1,32]. A number of proteins interact with Foxp3 at its N-terminus, including the transcription factors Hif1a, IRF4, and Eos, the histone acetyltransferases Tip60 and p300, and the histone deacetylase HDAC7, which help regulate the levels of Foxp3 protein [32,33]. A linker region between the leucine zipper and c-terminal Forkhead domain interacts with the transcription factor AML1/RUNX1, which cooperates with Foxp3 in the suppression of IL-2 expression [34]. The Forkhead domain itself interacts with the transcription factor NFAT, relevant to the expression in components of the TR cell transcriptome, including IL-2 receptor alpha chain (CD25), GITR and CTLA-4 [35]. FOXP3 may act to induce (e.g. CD25) or suppress (e.g. IL2) gene expression depending on the nature of the interactions with different transcriptional partners at the respective promoter region [8].

Regarding FOXP3 genotype/phenotype relationships in IPEX some conclusions can be drawn from the clinical phenotypes reported for different human FOXP3 mutations [6,36]. Missense mutations, small in-frame deletions, and deletions and mutations in the promoter and 5′ untranslated regions of Foxp3 can be associated with near-normal expression of Foxp3 protein, normal TR cell numbers but with compromised regulatory function, and a milder clinical phenotype. For example, a 1388-bp deletion (g.del–6247–4859) affecting the first untranslated exon and the adjacent intron of FOXP3, a region relevant to the formation of induced TR cells, gave rise to enteropathy, eczema and food allergies and elevated IgE but no endocrinopathy or cytopenias [37]. Similarly, some N-terminal Foxp3 mutations (e.g. Q70H and T108M) gave rise to a milder phenotype without endocrinopathy [38,39]. Leucine zipper domain and Forkhead domain mutations are usually associated with more severe phenotypes [6,36]. It is also likely that the genetic background may affect organ involvement as demonstrated in Foxp3-deficient mice where organ involvement and disease manifestations vary depending on the strain background [40].

The immunopathology of Foxp3 deficiency results from unchecked T cell activation secondary to loss of TR cells that act in a dominant manner to suppress T cell activation [41,42]. This is evidenced by the capacity of perinatal adoptive transfer of CD4+CD25+ TR cells to cure Foxp3-deficient mice from disease [13]. Left untreated, disease in Foxp3 deficient mice evolves aggressively and death ensues within a few weeks after birth [4346]. Disease progression is associated with lymphoid and myeloid hyperplasia and a concomitant intense mixed inflammatory infiltrates involving several organs including the liver, lung pancreas, stomach, skin and the gut [4346]. The overwhelming majority of CD4+ and CD8+ T cells are activated, and Th1, Th2, and Th17 cytokines are abundantly expressed [40]. Inflammation at the mucosal surfaces, including the gut, skin and lungs, may be driven by unrestrained T cell reactivity to antigens of the commensal flora [47,48]. It is attenuated in germ-free Foxp3 deficient mice and by deficiency of the toll-like receptor adaptor MyD88 [48]. In contrast the systemic autoimmunity of Foxp3 deficiency is unaffected by these interventions.

CD25 Deficiency

CD25 deficiency in humans manifests similarities to IPEX but is distinguished by a profound susceptibility to infections. The first description of CD25 deficiency was in a male subject of consanguineous parents who presented with CMV pneumonitis, oral and esophageal candidiasis, adenoviral gastroenteritis, diarrhea and failure to thrive [49,50]. A second case of CD25 deficiency was described in a boy born to unrelated parents who developed severe diarrhea, insulin-dependent diabetes mellitus, and eventual respiratory failure within the first months of life [30]. CMV pneumonitis, enteritis, and EBV lymphoproliferative disease developed. More recently, a third case of CD25 deficiency was described in a female child of consanguineous parents who developed early onset eczema and autoimmune enteropathy that was complicated by CMV, bullous pemphigoid, autoimmune thyroiditis, and alopecia universalis.[51].

The presentation of CD25 deficiency has many features of IPEX, but exhibits unique features including chronic viral, fungal, and bacterial infections [30,49,51]. The autoimmune findings are consistent with the pivotal role of CD25 in TR cell biology [52,53]. IL-2 appears critical for the growth of TR cells, and it has been proposed that one of the key features of IL-2 is to expand TR cells [54,55]. However, two of the reported patients with CD25 deficiency were found to have relatively normal percentages of Foxp3+ cells in the peripheral blood, suggesting that CD25 signaling is not required for the development of TR cells, which is supported in mouse models [55]. Although CD25 is dispensable for the generation of TR cells, deficiency of CD25 leads to widespread immune dysregulation, arguing that functional responses of TR cells critically depend on IL-2. In support of this, in vivo and in vitro treatment of T regulatory cells with IL-2 appears to enhance their suppressive abilities [5557]. In addition, the constitutive expression of CD25 on TR cells allows them to effectively compete for IL-2 and act as a cytokine sink that promotes apoptosis in a Bim-dependent manner in effector cells[58,59]. Finally, IL-2 is essential to the generation of induced TR cell populations [60,61].

The infectious complications in CD25 deficiency also confirm a role for IL-2 in generating effective immunity of T cells [reviewed in [62]]. IL-2 promotes the proliferation and development of TH1 and TH2 cells [6365] [66]. In contrast, IL-2 appears to inhibit Th17 development, but once generated Th17 cells may utilize IL-2 to expand [67,68]. IL-2 also inhibits T follicular helper T cells development [69,70]. IL-2 is important in generating cytotoxic CD8 effector cells as well as memory CD8 cells through the induction of IFN-γ, perforin and granzyme expression[71]. [72]. IL-2 can activate NK cells to become lymphokine-activated killer cells, and IL-2 has been reported to promote B cell response [73,74]. The pleiotropic effects of IL-2 on the effector function of the adaptive immune response helps to explain why individuals lacking CD25 exhibit increased susceptibility to infections, including fungus, bacteria, and viruses.

STAT5b Deficiency

STAT5 consists of two close related proteins, STAT5a and STAT5b, that are the product of two distinct genes and are 90% homologous [75,76]. Complete deficiency in STAT5 proteins in mice results in perinatal lethality since these proteins are involved in the signal transduction of a variety of growth factors, including growth hormone, prolactin, erythropoietin, IL-3, IL-5, and GM-CSF [77,78]. Defects in Stat5a have not been described. Defects in Stat5b have been described to result from deletion, missense and splice junction mutations, and are inherited in an autosomal recessive manner.

The original description of STAT5b deficiency was of an Argentinean girl with growth failure, delayed puberty, prominent forehead, saddle nose, and high pitched voice, all features of growth hormone deficiency [79]. Since then, several other patients have been described [8085]. Common clinical presentations include failure to thrive and growth failure, recurrent infectious pneumonias due to bacterial and viral infections, lymphoid interstitial pneumonitis, severe varicella and recurrent bouts of herpes zoster, and chronic diarrhea and eczema. Laboratory studies usually demonstrate hypergammaglobulinemia, with normal T, B, NK cells number, although the T cells may show severe memory skewing. Foxp3+ TR cells may be normal or slightly decreased. Mitogenic responses may be low, Low IGF-1, low IGFBP-3, and high prolactin are usually present, reflecting defective growth hormone receptor signaling.

Many of the features of STAT5b deficiency are similar to findings in CD25 deficiency, including eczema, chronic diarrhea, thyroiditis, and increased susceptibility to infections, likely due to the fact that IL-2 receptor signaling requires STAT5. Pulmonary disease appears more prominent in STAT5b deficiency, although it is unclear whether this is driven by infectious processes, or is the result of defective immune regulation. Similar to CD25 deficiency, signs of immune activation may be apparent, including hypergammaglobulinemia and increased percentages of CD45RO positive T cells. The severity of disease in STAT5b deficiency appears more variable and less severe compared to CD25 deficiency, likely due to the fact that STAT5a may substitute for some of the functions of Stat5b deficiency. The increased susceptibility to infections and autoimmune manifestations of STAT5b deficiency is likely due to defects in responsiveness to IL-2 as outlined in CD25 deficiency above.

IPEX-like disease due STAT1 Mutations

Autosomal dominant heterozygous gain of function mutations in STAT1 have been found in subjects with mucocutaneous candidiasis. Some of the affected subjects suffered from autoimmunity, mainly autoimmune thyroiditis, but otherwise lacked other characteristics of the AEPECD phenotype, including characteristic end-organ targets of autoimmunity and ectodermal dysplasia [8688]. By screening patients with IPEX like phenotype with or without mucocutaneous candidiasis, Uzel et al identified 5 patients heterozygous gain of function STAT1 mutations [89]. The mutations involved the coiled-coil (R210I, V266I) and DNA binding domains (L358W, T385M (two patients) of STAT1. These mutations resulted in increased STAT1 phosphorylation at the regulatory tyrosine 701, consistent with the gain of function phenotype of the mutations.

Gain of function STAT1 mutations inhibit Th17 cell differentiation, a phenotype associated with heightened susceptibility to mucocutaneous candidiasis. However, the mechanism of IPEX-like disease in some of the patients remains unclear. The number of TR cells and their in vitro suppressor function appeared normal [89]. It is possible that the gain of function STAT1 mutant destabilizes the TR cells by reprograming them into Th1-like cells, but that remains to be determined. IL-10 production by peripheral blood lymphocytes was profoundly decreased, suggesting a role for functional IL-10 deficiency in certain aspects of disease manifestation (such as enteritis).

All five patients presented with eczema and enteropathy, [89]. Three of the patients developed type 1 diabetes, three developed hypothyroidism and or thyroid autoantibodies, and one patient developed growth hormone insufficiency. All patients suffered recurrent infections, with various combinations of sinopulmonary infections and pneumonias (with or without bronchiectasis), herpes virus infections, and blood-borne infections. The patients also suffered form a variety of cardiovascular problems including hypertension, vascular aneurysms and calcifications. Four of the five patients exhibited mucocutaneous candidiasis, suggesting that the disease may occasionally present in the absence of (or cryptic presence) of candidiasis [89]. Disseminated aspergillosis and candidemia have been noted in two patients under conditions of immunosuppression and catheter related, respectively.

ITCH

Human ITCH deficiency was discovered in an extended Amish family with findings of multisystem autoimmunity, dysmorphic features, and developmental abnormalities due to a single adenine nucleotide insertion in codon 132 of ITCH was found, resulting in a frame-shift and premature stop [90]. Ten affected individuals were found to carry this mutation. Autosomal recessive inheritance was apparent in this extended family, and prominent consanguinity was noted. A naturally occurring mutation in Itch, the mouse homologue of human ITCH, recapitulates many features of the human disease, including a severe autoimmune disorder characterized by dermatitis, chronic pulmonary inflammation, alveolar proteinosis, and lymphoid hyperplasia [91]. Studies with Itch-deficient mice have elucidated some of the potential mechanisms of immune dysregulation in these mice. ITCH is a ubiquitin ligase with diverse function in T cells [91]. T cells from Itch−/− mice show an activated phenotype with enhanced proliferation and expression of the TH2 cell cytokines interleukin 4 and IL-5 [92]. IgG1 and IgE levels were elevated, consistent with a prominent TH2 response in these mice. These studies identified JunB, a transcription factor that is involved in TH2 differentiation, as a target of ITCH [93,94]. Other studies have shown that ITCH is upregulated in anergic T cells, and ITCH associates with PLC-γ1 and PKC-θ, two key signaling molecules induced by Ca2+/ calcineurin signaling [95]. Following ITCH-mediated ubiquitination, PLC-γ1 and PKC-θ are targeted to the lysosome for degradation, leading to reduced levels of PLC-γ1 and PKC-θ and defective T cells activation.

ITCH has also been implicated in TR cell function and generation, mainly through its affects on TGF-β signaling and Foxp3 expression in CD4 T cells [96]. The loss of ITCH compromises TGF-β–induced Foxp3 expression and TGF-β–mediated inhibition of T-cell proliferation through the ubiquitination of the transcription factor TIEG1. Upon ubiquitination by ITCH, TIEG1 translocates to the nucleus, and binds GC rich sequences in the Foxp3 promoter [96].

Summary and Future Directions

Despite the progress in identifying several IPEX like diseases and establishing their genetic causations, the underlying defects in a large proportion of subjects who present with IPEX like disorders remain obscure. The advent of global sequencing approaches such as whole genome and exome sequencing promises to identify novel gene defects and exciting new pathways operative in tolerance and autoimmunity.

Key Points.

  • A number of heritable Mendelian disorders of autoimmunity and immune dysregulation involve genes that control various T regulatory (TR) cell functions, including FOXP3, IL2RA, STAT5B, and ITCH.

  • A life-long risk of autoimmune complications attends mutations affecting FOXP3 and related pathways.

  • Gain of function mutations in STAT1, normally associated with mucocutaneous candidiasis, may manifest as an IPEX-like phenotype.

Acknowledgments

The authors acknowledge grant support by the National Institutes of Health to T.A.C. (5R01AI085090) and J.W.V. (1K08AI072023).

Footnotes

The authors declare no conflict of interest.

References

  • 1.Chatila TA, Blaeser F, Ho N, Lederman HM, Voulgaropoulos C, Helms C, Bowcock AM. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest. 2000;106:R75–81. doi: 10.1172/JCI11679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, Levy-Lahad E, Mazzella M, Goulet O, Perroni L, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 3.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–21. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 4.Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, X linked (IPEX) syndrome. J Med Genet. 2002;39:537–545. doi: 10.1136/jmg.39.8.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked: forkhead box protein 3 mutations and lack of regulatory T cells. J Allergy Clin Immunol. 2007;120:744–750. doi: 10.1016/j.jaci.2007.08.044. quiz 751–742. [DOI] [PubMed] [Google Scholar]
  • •6.Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol. 2012;3:211. doi: 10.3389/fimmu.2012.00211. This report presents a comprehensive review of IPEX, the underlying FOXP3 geneotypes and clinicopathological presentations. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nieves DS, Phipps RP, Pollock SJ, Ochs HD, Zhu Q, Scott GA, Ryan CK, Kobayashi I, Rossi TM, Goldsmith LA. Dermatologic and immunologic findings in the immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Arch Dermatol. 2004;140:466–472. doi: 10.1001/archderm.140.4.466. [DOI] [PubMed] [Google Scholar]
  • •8.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–564. doi: 10.1146/annurev.immunol.25.022106.141623. This report, and the one by Benoist et al below, presents a comprehensive review of regulatory T cells and the role of Foxp3 and other factors in their differentiated functions. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •9.Benoist C, Mathis D. Treg cells, life history, and diversity. Cold Spring Harb Perspect Biol. 2012;4:a007021. doi: 10.1101/cshperspect.a007021. This report, and the one by Rudensky et al above, presents a comprehensive review of regulatory T cells and the role of Foxp3 and other factors in their differentiated functions. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lio CW, Hsieh CS. Becoming self-aware: the thymic education of regulatory T cells. Curr Opin Immunol. 2011;23:213–219. doi: 10.1016/j.coi.2010.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hsieh CS, Liang Y, Tyznik AJ, Self SG, Liggitt D, Rudensky AY. Recognition of the peripheral self by naturally arising CD25+ CD4+ T cell receptors. Immunity. 2004;21:267–277. doi: 10.1016/j.immuni.2004.07.009. [DOI] [PubMed] [Google Scholar]
  • 12.Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY. An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nat Immunol. 2006;7:401–410. doi: 10.1038/ni1318. [DOI] [PubMed] [Google Scholar]
  • 13.Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B, Ziegelbauer J, Yassai M, Li SH, Relland LM, et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity. 2011;35:109–122. doi: 10.1016/j.immuni.2011.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •14.Bilate AM, Lafaille JJ. Induced CD4+Foxp3+ regulatory T cells in immune tolerance. Annu Rev Immunol. 2012;30:733–758. doi: 10.1146/annurev-immunol-020711-075043. This report provides a comprehensive review on induced regulatory T cells and their role in peripheral tolerance. [DOI] [PubMed] [Google Scholar]
  • 15.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, Peterson DA, Stappenbeck TS, Hsieh CS. Peripheral education of the immune system by colonic commensal microbiota. Nature. 2011 doi: 10.1038/nature10434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martinez-Llordella M, Ashby M, Nakayama M, Rosenthal W, Bluestone JA. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–1007. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Paust S, Lu L, McCarty N, Cantor H. Engagement of B7 on effector T cells by regulatory T cells prevents autoimmune disease. Proc Natl Acad Sci USA. 2004;101:10398–10403. doi: 10.1073/pnas.0403342101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Asseman C, Mauze S, Leach MW, Coffman RL, Powrie F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. Journal of Experimental Medicine. 1999;190:995–1004. doi: 10.1084/jem.190.7.995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. The Journal of Experimental Medicine. 2001;194:629–644. doi: 10.1084/jem.194.5.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Green EA, Gorelik L, McGregor CM, Tran EH, Flavell RA. CD4+CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes. Proc Natl Acad Sci U S A. 2003;100:10878–10883. doi: 10.1073/pnas.1834400100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 2004;21:589–601. doi: 10.1016/j.immuni.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 24.Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636–645. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
  • 25.Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8:523–532. doi: 10.1038/nri2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–1265. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mellor AL, Chandler P, Baban B, Hansen AM, Marshall B, Pihkala J, Waldmann H, Cobbold S, Adams E, Munn DH. Specific subsets of murine dendritic cells acquire potent T cell regulatory functions following CTLA4-mediated induction of indoleamine 2,3 dioxygenase. Int Immunol. 2004;16:1391–1401. doi: 10.1093/intimm/dxh140. [DOI] [PubMed] [Google Scholar]
  • 28.Glocker EO, Kotlarz D, Klein C, Shah N, Grimbacher B. IL-10 and IL-10 receptor defects in humans. Ann N Y Acad Sci. 2011;1246:102–107. doi: 10.1111/j.1749-6632.2011.06339.x. [DOI] [PubMed] [Google Scholar]
  • 29.Verbsky JW, Grossman WJ. Hemophagocytic lymphohistiocytosis: diagnosis, pathophysiology, treatment, and future perspectives. Ann Med. 2006;38:20–31. doi: 10.1080/07853890500465189. [DOI] [PubMed] [Google Scholar]
  • 30.Caudy AA, Reddy ST, Chatila T, Atkinson JP, Verbsky JW. CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J Allergy Clin Immunol. 2007;119:482–487. doi: 10.1016/j.jaci.2006.10.007. [DOI] [PubMed] [Google Scholar]
  • 31.Verbsky JW, Chatila TA. T-regulatory cells in primary immune deficiencies. Curr Opin Allergy Clin Immunol. 2011;11:539–544. doi: 10.1097/ACI.0b013e32834cb8fa. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li B, Samanta A, Song X, Iacono KT, Brennan P, Chatila TA, Roncador G, Banham AH, Riley JL, Wang Q, et al. FOXP3 is a homo-oligomer and a component of a supramolecular regulatory complex disabled in the human XLAAD/IPEX autoimmune disease. Int Immunol. 2007;19:825–835. doi: 10.1093/intimm/dxm043. [DOI] [PubMed] [Google Scholar]
  • •33.Chatila TA, Williams CB. Foxp3: shades of tolerance. Immunity. 2012;36:693–694. doi: 10.1016/j.immuni.2012.05.011. This report reviews the recent literature on capacity of subtle genotypic changes in Foxp3 to manifest as different autoimmune phenotypes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ono M, Yaguchi H, Ohkura N, Kitabayashi I, Nagamura Y, Nomura T, Miyachi Y, Tsukada T, Sakaguchi S. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature. 2007;446:685–689. doi: 10.1038/nature05673. [DOI] [PubMed] [Google Scholar]
  • 35.Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, Bates DL, Guo L, Han A, Ziegler SF, et al. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell. 2006;126:375–387. doi: 10.1016/j.cell.2006.05.042. [DOI] [PubMed] [Google Scholar]
  • 36.d’Hennezel E, Bin Dhuban K, Torgerson T, Piccirillo CA. The immunogenetics of immune dysregulation, polyendocrinopathy, enteropathy, X linked (IPEX) syndrome. J Med Genet. 2012;49:291–302. doi: 10.1136/jmedgenet-2012-100759. [DOI] [PubMed] [Google Scholar]
  • 37.Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, Hermine O, Vijay S, Gambineri E, Cerf-Bensussan N, et al. Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology. 2007;132:1705–1717. doi: 10.1053/j.gastro.2007.02.044. [DOI] [PubMed] [Google Scholar]
  • 38.Yong PL, Russo P, Sullivan KE. Use of sirolimus in IPEX and IPEX-like children. J Clin Immunol. 2008;28:581–587. doi: 10.1007/s10875-008-9196-1. [DOI] [PubMed] [Google Scholar]
  • 39.De Benedetti F, Insalaco A, Diamanti A, Cortis E, Muratori F, Lamioni A, Carsetti R, Cusano R, De Vito R, Perroni L, et al. Mechanistic associations of a mild phenotype of immunodysregulation, polyendocrinopathy, enteropathy, x-linked syndrome. Clin Gastroenterol Hepatol. 2006;4:653–659. doi: 10.1016/j.cgh.2005.12.014. [DOI] [PubMed] [Google Scholar]
  • 40.Lin W, Truong N, Grossman WJ, Haribhai D, Williams CB, Wang J, Martin MG, Chatila TA. Allergic dysregulation and hyperimmunoglobulinemia E in Foxp3 mutant mice. J Allergy Clin Immunol. 2005;116:1106–1115. doi: 10.1016/j.jaci.2005.08.046. [DOI] [PubMed] [Google Scholar]
  • 41.Clark LB, Appleby MW, Brunkow ME, Wilkinson JE, Ziegler SF, Ramsdell F. Cellular and molecular characterization of the scurfy mouse mutant. J Immunol. 1999;162:2546–2554. [PubMed] [Google Scholar]
  • 42.Blair PJ, Bultman SJ, Haas JC, Rouse BT, Wilkinson JE, Godfrey VL. CD4+CD8- T cells are the effector cells in disease pathogenesis in the scurfy (sf) mouse. J Immunol. 1994;153:3764–3774. [PubMed] [Google Scholar]
  • 43.Lyon MF, Peters J, Glenister PH, Ball S, Wright E. The scurfy mouse mutant has previously unrecognized hematological abnormalities and resembles Wiskott-Aldrich syndrome. Proc Natl Acad Sci U S A. 1990;87:2433–2437. doi: 10.1073/pnas.87.7.2433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol. 1991;138:1379–1387. [PMC free article] [PubMed] [Google Scholar]
  • 45.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 46.Lin W, Truong N, Grossman WJ, Haribhai D, Williams CB, Wang J, MartÍn MG, Chatila TA. Allergic Dysregulation and Hyper Immunoglobulinemia E in Foxp3 Mutant Mice. J Allergy Clin Immunol. 2005 doi: 10.1016/j.jaci.2005.08.046. (in press) [DOI] [PubMed] [Google Scholar]
  • 47.Chinen T, Volchkov PY, Chervonsky AV, Rudensky AY. A critical role for regulatory T cell-mediated control of inflammation in the absence of commensal microbiota. J Exp Med. 2010;207:2323–2330. doi: 10.1084/jem.20101235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••48.Rivas MN, Koh YT, Chen A, Nguyen A, Lee YH, Lawson G, Chatila TA. MyD88 is critically involved in immune tolerance breakdown at environmental interfaces of Foxp3-deficient mice. J Clin Invest. 2012;122:1933–1947. doi: 10.1172/JCI40591. This report demonstrates that the immune dysregulation and inflammation in Foxp3 deficiency can be dissociated into a mucosal inflammatory component, driven by MyD88-dependent mechanisms, and a systemic autoimmune lymphoproliferative component that is MyD88-indpendent. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sharfe N, Dadi HK, Shahar M, Roifman CM. Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor. Proc Natl Acad Sci U S A. 1997;94:3168–3171. doi: 10.1073/pnas.94.7.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Roifman CM. Human IL-2 receptor alpha chain deficiency. Pediatr Res. 2000;48:6–11. doi: 10.1203/00006450-200007000-00004. [DOI] [PubMed] [Google Scholar]
  • 51.Goudy K, Aydin D, Barzaghi F, Gambineri E, Vignoli M, Ciullini MS, Doglioni C, Ponzoni M, Cicalese MP, Assanelli A, et al. Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol. 2013;146:248–261. doi: 10.1016/j.clim.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med. 1996;184:387–396. doi: 10.1084/jem.184.2.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. 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. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 54.Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005;201:723–735. doi: 10.1084/jem.20041982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol. 2005;6:1142–1151. doi: 10.1038/ni1263. [DOI] [PubMed] [Google Scholar]
  • 56.de la Rosa M, Rutz S, Dorninger H, Scheffold A. Interleukin-2 is essential for CD4+CD25+ regulatory T cell function. Eur J Immunol. 2004;34:2480–2488. doi: 10.1002/eji.200425274. [DOI] [PubMed] [Google Scholar]
  • 57.Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ. Interleukin 2 signaling is required for CD4(+) regulatory T cell function. The Journal of Experimental Medicine. 2002;196:851–857. doi: 10.1084/jem.20020190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Barron L, Dooms H, Hoyer KK, Kuswanto W, Hofmann J, O’Gorman WE, Abbas AK. Cutting edge: mechanisms of IL-2-dependent maintenance of functional regulatory T cells. J Immunol. 2010;185:6426–6430. doi: 10.4049/jimmunol.0903940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pandiyan P, Zheng L, Ishihara S, Reed J, Lenardo MJ. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nat Immunol. 2007;8:1353–1362. doi: 10.1038/ni1536. [DOI] [PubMed] [Google Scholar]
  • 60.Davidson TS, DiPaolo RJ, Andersson J, Shevach EM. Cutting Edge: IL-2 is essential for TGF-beta-mediated induction of Foxp3+ T regulatory cells. J Immunol. 2007;178:4022–4026. doi: 10.4049/jimmunol.178.7.4022. [DOI] [PubMed] [Google Scholar]
  • 61.Zheng SG, Wang J, Horwitz DA. Cutting edge: Foxp3+CD4+CD25+ regulatory T cells induced by IL-2 and TGF-beta are resistant to Th17 conversion by IL-6. J Immunol. 2008;180:7112–7116. doi: 10.4049/jimmunol.180.11.7112. [DOI] [PubMed] [Google Scholar]
  • 62.Liao W, Lin JX, Leonard WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity. 2013;38:13–25. doi: 10.1016/j.immuni.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–2088. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 64.Liao W, Lin JX, Wang L, Li P, Leonard WJ. Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat Immunol. 2011;12:551–559. doi: 10.1038/ni.2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Shi M, Lin TH, Appell KC, Berg LJ. Janus-kinase-3-dependent signals induce chromatin remodeling at the Ifng locus during T helper 1 cell differentiation. Immunity. 2008;28:763–773. doi: 10.1016/j.immuni.2008.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liao W, Schones DE, Oh J, Cui Y, Cui K, Roh TY, Zhao K, Leonard WJ. Priming for T helper type 2 differentiation by interleukin 2-mediated induction of interleukin 4 receptor alpha-chain expression. Nat Immunol. 2008;9:1288–1296. doi: 10.1038/ni.1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Laurence A, O’Shea JJ. T(H)-17 differentiation: of mice and men. Nat Immunol. 2007;8:903–905. doi: 10.1038/ni0907-903. [DOI] [PubMed] [Google Scholar]
  • 68.Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, Nussenblatt RB, Gery I, Lee YS, Egwuagu CE. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med. 2007;13:711–718. doi: 10.1038/nm1585. [DOI] [PubMed] [Google Scholar]
  • 69.Ballesteros-Tato A, Leon B, Graf BA, Moquin A, Adams PS, Lund FE, Randall TD. Interleukin-2 inhibits germinal center formation by limiting T follicular helper cell differentiation. Immunity. 2012;36:847–856. doi: 10.1016/j.immuni.2012.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Johnston RJ, Choi YS, Diamond JA, Yang JA, Crotty S. STAT5 is a potent negative regulator of TFH cell differentiation. The Journal of Experimental Medicine. 2012;209:243–250. doi: 10.1084/jem.20111174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ, Rao A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity. 2010;32:79–90. doi: 10.1016/j.immuni.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Williams MA, Tyznik AJ, Bevan MJ. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature. 2006;441:890–893. doi: 10.1038/nature04790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Siegel JP, Sharon M, Smith PL, Leonard WJ. The IL-2 receptor beta chain (p70): role in mediating signals for LAK, NK, and proliferative activities. Science. 1987;238:75–78. doi: 10.1126/science.3116668. [DOI] [PubMed] [Google Scholar]
  • 74.Mingari MC, Gerosa F, Carra G, Accolla RS, Moretta A, Zubler RH, Waldmann TA, Moretta L. Human interleukin-2 promotes proliferation of activated B cells via surface receptors similar to those of activated T cells. Nature. 1984;312:641–643. doi: 10.1038/312641a0. [DOI] [PubMed] [Google Scholar]
  • 75.Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. 293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
  • 76.Ihle JN. The Stat family in cytokine signaling. Curr Opin Cell Biol. 2001;13:211–217. doi: 10.1016/s0955-0674(00)00199-x. [DOI] [PubMed] [Google Scholar]
  • 77.Socolovsky M, Fallon AE, Wang S, Brugnara C, Lodish HF. Fetal anemia and apoptosis of red cell progenitors in Stat5a−/−5b−/− mice: a direct role for Stat5 in Bcl-X(L) induction. Cell. 1999;98:181–191. doi: 10.1016/s0092-8674(00)81013-2. [DOI] [PubMed] [Google Scholar]
  • 78.Cui Y, Riedlinger G, Miyoshi K, Tang W, Li C, Deng CX, Robinson GW, Hennighausen L. Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation. Mol Cell Biol. 2004;24:8037–8047. doi: 10.1128/MCB.24.18.8037-8047.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J, Pratt KL, Bezrodnik L, Jasper H, Tepper A, et al. Growth hormone insensitivity associated with a STAT5b mutation. N Engl J Med. 2003;349:1139–1147. doi: 10.1056/NEJMoa022926. [DOI] [PubMed] [Google Scholar]
  • 80.Hwa V, Little B, Adiyaman P, Kofoed EM, Pratt KL, Ocal G, Berberoglu M, Rosenfeld RG. Severe growth hormone insensitivity resulting from total absence of signal transducer and activator of transcription 5b. J Clin Endocrinol Metab. 2005;90:4260–4266. doi: 10.1210/jc.2005-0515. [DOI] [PubMed] [Google Scholar]
  • 81.Vidarsdottir S, Walenkamp MJ, Pereira AM, Karperien M, van DJ, van Duyvenvoorde HA, White S, Breuning MH, Roelfsema F, Kruithof MF, et al. Clinical and biochemical characteristics of a male patient with a novel homozygous STAT5b mutation. J Clin Endocrinol Metab. 2006;91:3482–3485. doi: 10.1210/jc.2006-0368. [DOI] [PubMed] [Google Scholar]
  • 82.Walenkamp MJ, Vidarsdottir S, Pereira AM, Karperien M, van DJ, van Duyvenvoorde HA, Breuning MH, Roelfsema F, Kruithof MF, van DJ, et al. Growth hormone secretion and immunological function of a male patient with a homozygous STAT5b mutation. Eur J Endocrinol. 2007;156:155–165. doi: 10.1530/eje.1.02327. [DOI] [PubMed] [Google Scholar]
  • 83.Bernasconi A, Marino R, Ribas A, Rossi J, Ciaccio M, Oleastro M, Ornani A, Paz R, Rivarola MA, Zelazko M, et al. Characterization of immunodeficiency in a patient with growth hormone insensitivity secondary to a novel STAT5b gene mutation. Pediatrics. 2006;118:e1584–e1592. doi: 10.1542/peds.2005-2882. [DOI] [PubMed] [Google Scholar]
  • 84.Hwa V, Camacho-Hubner C, Little BM, David A, Metherell LA, El-Khatib N, Savage MO, Rosenfeld RG. Growth hormone insensitivity and severe short stature in siblings: a novel mutation at the exon 13-intron 13 junction of the STAT5b gene. Horm Res. 2007;68:218–224. doi: 10.1159/000101334. [DOI] [PubMed] [Google Scholar]
  • 85.Pugliese-Pires PN, Tonelli CA, Dora JM, Silva PC, Czepielewski M, Simoni G, Arnhold IJ, Jorge AA. A novel STAT5B mutation causing GH insensitivity syndrome associated with hyperprolactinemia and immune dysfunction in two male siblings. Eur J Endocrinol. 2010;163:349–355. doi: 10.1530/EJE-10-0272. [DOI] [PubMed] [Google Scholar]
  • 86.Liu L, Okada S, Kong XF, Kreins AY, Cypowyj S, Abhyankar A, Toubiana J, Itan Y, Audry M, Nitschke P, et al. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med. 2011;208:1635–1648. doi: 10.1084/jem.20110958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.van de Veerdonk FL, Plantinga TS, Hoischen A, Smeekens SP, Joosten LA, Gilissen C, Arts P, Rosentul DC, Carmichael AJ, Smits-van der Graaf CA, et al. STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med. 2011;365:54–61. doi: 10.1056/NEJMoa1100102. [DOI] [PubMed] [Google Scholar]
  • 88.Boisson-Dupuis S, Kong XF, Okada S, Cypowyj S, Puel A, Abel L, Casanova JL. Inborn errors of human STAT1: allelic heterogeneity governs the diversity of immunological and infectious phenotypes. Curr Opin Immunol. 2012;24:364–378. doi: 10.1016/j.coi.2012.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••89.Uzel G, Sampaio EP, Lawrence MG, Hsu AP, Hackett M, Dorsey MJ, Noel RJ, Verbsky JW, Freeman AF, Janssen E, et al. Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation-polyendocrinopathy-enteropathy-X-linked-like syndrome. J Allergy Clin Immunol. 2013;131:1611–1623. e1613. doi: 10.1016/j.jaci.2012.11.054. This report demonstrates a novel IPEX like phenotype caused by dominant gain of function mutations in STAT1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lohr NJ, Molleston JP, Strauss KA, Torres-Martinez W, Sherman EA, Squires RH, Rider NL, Chikwava KR, Cummings OW, Morton DH, et al. Human ITCH E3 ubiquitin ligase deficiency causes syndromic multisystem autoimmune disease. Am J Hum Genet. 2010;86:447–453. doi: 10.1016/j.ajhg.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Perry WL, Hustad CM, Swing DA, O’Sullivan TN, Jenkins NA, Copeland NG. The itchy locus encodes a novel ubiquitin protein ligase that is disrupted in a18H mice. Nat Genet. 1998;18:143–146. doi: 10.1038/ng0298-143. [DOI] [PubMed] [Google Scholar]
  • 92.Fang D, Elly C, Gao B, Fang N, Altman Y, Joazeiro C, Hunter T, Copeland N, Jenkins N, Liu YC. Dysregulation of T lymphocyte function in itchy mice: a role for Itch in TH2 differentiation. Nat Immunol. 2002;3:281–287. doi: 10.1038/ni763. [DOI] [PubMed] [Google Scholar]
  • 93.Gao M, Labuda T, Xia Y, Gallagher E, Fang D, Liu YC, Karin M. Jun turnover is controlled through JNK-dependent phosphorylation of the E3 ligase Itch. Science. 2004;306:271–275. doi: 10.1126/science.1099414. [DOI] [PubMed] [Google Scholar]
  • 94.Fang D, Kerppola TK. Ubiquitin-mediated fluorescence complementation reveals that Jun ubiquitinated by Itch/AIP4 is localized to lysosomes. Proc Natl Acad Sci U S A. 2004;101:14782–14787. doi: 10.1073/pnas.0404445101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Heissmeyer V, Macian F, Im SH, Varma R, Feske S, Venuprasad K, Gu H, Liu YC, Dustin ML, Rao A. Calcineurin imposes T cell unresponsiveness through targeted proteolysis of signaling proteins. Nat Immunol. 2004;5:255–265. doi: 10.1038/ni1047. [DOI] [PubMed] [Google Scholar]
  • 96.Venuprasad K, Huang H, Harada Y, Elly C, Subramaniam M, Spelsberg T, Su J, Liu YC. The E3 ubiquitin ligase Itch regulates expression of transcription factor Foxp3 and airway inflammation by enhancing the function of transcription factor TIEG1. Nat Immunol. 2008;9:245–253. doi: 10.1038/niXXXX. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES