Clinical, Immunological, and Molecular Heterogeneity of 173 Patients With the Phenotype of Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked (IPEX) Syndrome

Eleonora Gambineri, Sara Ciullini Mannurita, David Hagin, Marina Vignoli, Stephanie Anover-Sombke, Stacey DeBoer, Gesmar R S Segundo, Eric J Allenspach, Claudio Favre, Hans D Ochs, Troy R Torgerson, Eleonora Gambineri, Sara Ciullini Mannurita, David Hagin, Marina Vignoli, Stephanie Anover-Sombke, Stacey DeBoer, Gesmar R S Segundo, Eric J Allenspach, Claudio Favre, Hans D Ochs, Troy R Torgerson

Abstract

Background: Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-linked (IPEX) Syndrome is a rare recessive disorder caused by mutations in the FOXP3 gene. In addition, there has been an increasing number of patients with wild-type FOXP3 gene and, in some cases, mutations in other immune regulatory genes. Objective: To molecularly asses a cohort of 173 patients with the IPEX phenotype and to delineate the relationship between the clinical/immunologic phenotypes and the genotypes. Methods: We reviewed the clinical presentation and laboratory characteristics of each patient and compared clinical and laboratory data of FOXP3 mutation-positive (IPEX patients) with those from FOXP3 mutation-negative patients (IPEX-like). A total of 173 affected patients underwent direct sequence analysis of the FOXP3 gene while 85 IPEX-like patients with normal FOXP3 were investigated by a multiplex panel of "Primary Immune Deficiency (PID-related) genes." Results: Forty-four distinct FOXP3 variants were identified in 88 IPEX patients, 9 of which were not previously reported. Among the 85 IPEX-like patients, 19 different disease-associated variants affecting 9 distinct genes were identified. Conclusions: We provide a comprehensive analysis of the clinical features and molecular bases of IPEX and IPEX-like patients. Although we were not able to identify major distinctive clinical features to differentiate IPEX from IPEX-like syndromes, we propose a simple flow-chart to effectively evaluate such patients and to focus on the most likely molecular diagnosis. Given the large number of potential candidate genes and overlapping phenotypes, selecting a panel of PID-related genes will facilitate a molecular diagnosis.

Keywords: FOXP3; IPEX-like; X-linked (IPEX); enteropathy; immune dysregulation; immune regulatory genes; polyendocrinopathy; regulatory T cells.

Figures

Figure 1
Figure 1
FOXP3 mutations identified in our cohort of IPEX patients. Schematic representation of FOXP3 mRNA and protein showing the location and sizes of the predicted structural domains. Numbers along the top line indicate amino acid number (1–431). The position of the identified mutations along the mRNA is reported with different shape for each type of variation. Each symbol represents a single kindred. ZF, zinc finger; LZ, leucine zipper; FKH, forkhead box domain; N, amino terminus; C, carboxy terminus; PolyA, polyadenylation sequence.
Figure 2
Figure 2
(A)FOXP3 mutations in IPEX (symbols in yellow) in forkhead box domain. Amino acid sequence of FOXP3 protein aligned with other FOX proteins. Shows conserved residues (blue) and highly conservative residues (green). (B)FOXP3 mutations in IPEX (symbols in yellow) in the leucine zipper domain. Amino acid sequence of FOXP3 protein aligned with c-myc and v-myc leucine zippers (known to be highly conserved). Shows highly conserved residues (blue) and highly conservative residues (green).
Figure 3
Figure 3
(A) CD25+FOXP3+ expression in IPEX, IPEX-like patients and control subjects. (B) IPEX patients differ according to the type of FOXP3 mutations. Deletion = in frame deletion; frameshift = deletion with frameshift. Values are expressed as percentage of CD4 cells, median and 5–95 percentile are shown, circles, triangles, and cones represent singles patients out of 5–95 percentile (circles = IPEX patients, triangles = IPEX-like patients, cones = control subjects) (**p < 0.001; ***p < 0.0001).
Figure 4
Figure 4
CD25+FOXP3+ expression in IPEX-like patients differed by the type of mutated genes identified and control subjects. Values are expressed as percentage of CD4 cells, mean and SD are shown.
Figure 5
Figure 5
Clinical manifestations in the IPEX and IPEX-like cohorts (A). IPEX patient stratified for type of mutation showing classical triad of symptoms (B).
Figure 6
Figure 6
Heat map showing frequencies of clinical manifestations in our cohort of patients with LRBA, CTLA4, STAT1 GOF, STAT3 GOF, STAT5b, and IL2RA mutations.
Figure 7
Figure 7
IgE levels in IPEX and IPEX-like patients. Mean and SD are shown (****p < 0.0001).
Figure 8
Figure 8
Kaplan-Meier survival curve for overall survival of patients. (A) The overall survival in the whole cohort of patient is evaluated (n = 163); (B) IPEX (n = 85) vs. IPEX-like (n = 78) patients are compared; (C) IPEX patients stratified for type of mutation: frameshift (FS) n = 6, in frame deletion (IFD) n = 11, missense (MS) n = 43, splicing mutations (SP) n = 14 and mutations in polyadenylation site (POLA) n = 11 are compared; (D) IPEX-like without mutations (NA= not available) n = 57 and IPEX-like with mutation (Mut) n = 21 are compared; (E) IPEX patient stratified whether (n = 39) or not (n = 46) they underwent HSCT and (F) IPEX-like patient stratified whether (n = 10) or not (n = 68) they underwent HSCT.
Figure 9
Figure 9
Flow chart suggesting molecular analysis strategy based on patients' clinical phenotype and immunological markers.

References

    1. Allenspach EJ, Finn LS, Rendi MH, Eken A, Singh AK, Oukka M, et al. . Absence of functional fetal regulatory T cells in humans causes in utero organ-specific autoimmunity. J Allergy Clin Immunol. (2017) 140:616–9.e7. 10.1016/j.jaci.2017.02.017
    1. Powell BR, Buist NRM, Stenzel P. An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr. (1982) 100:731–7. 10.1016/S0022-3476(82)80573-8
    1. Bennett CL, Yoshioka R, Kiyosawa H, Barker DF, Fain PR, Shigeoka AO, et al. . X-Linked syndrome of polyendocrinopathy, immune dysfunction, and diarrhea maps to Xp11.23-Xq13.3. Am J Hum Genet. (2000) 66:461–8. 10.1086/302761
    1. Ferguson PJ, Blanton SH, Saulsbury FT, McDuffie MJ, Lemahieu V, Gastier JM, et al. . Manifestations and linkage analysis in X-linked autoimmunity-immunodeficiency syndrome. Am J Med Genet. (2000) 90:390–7. 10.1002/(SICI)1096-8628(20000228)90:5<390::AID-AJMG9>;2-M
    1. Chatila TA, Blaeser F, Ho N, Lederman HM, Voulgaropoulos C, Helms C, et al. . JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest. (2000) 106:R75–81. 10.1172/JCI11679
    1. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. . The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. (2001) 27:20–1. 10.1038/83713
    1. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. . X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. (2001) 27:18–20. 10.1038/83707
    1. Kobayashi I, Shiari R, Yamada M, Kawamura N, Okano M, Yara A, et al. . Novel mutations of FOXP3 in two Japanese patients with immune dysregulation, polyendocrinopathy, enteropathy, X linked syndrome (IPEX). J Med Genet. (2001) 38:874–6. 10.1136/jmg.38.12.874
    1. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko S-A, et al. . Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. (2001) 27:68–73. 10.1038/83784
    1. Russell WL, Russell LB, Gower JS. Exceptional inheritance of a sex-linked gene in the mouse explained on the basis that the X/O sex-chromosome constitution is female. Proc Natl Acad Sci USA. (1959) 45:554–60. 10.1073/pnas.45.4.554
    1. Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol. (1991) 138:1379–87.
    1. 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) 153:3764–74.
    1. Clark LB, Appleby MW, Brunkow ME, Wilkinson JE, Ziegler SF, Ramsdell F. Cellular and molecular characterization of the scurfy mouse mutant. J Immunol. (1999) 162:2546–54.
    1. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. (2003) 4:337–42. 10.1038/ni909
    1. Kanangat S, Blair P, Reddy R, Deheshia M, Godfrey V, Rouse BT, et al. . Disease in the scurfy (sf) mouse is associated with overexpression of cytokine genes. Eur J Immunol. (1996) 26:161–5. 10.1002/eji.1830260125
    1. Ohkura N, Kitagawa Y, Sakaguchi S. Development and maintenance of regulatory T cells. Immunity (2013) 38:414–23. 10.1016/j.immuni.2013.03.002
    1. Schubert LA, Jeffery E, Zhang Y, Ramsdell F, Ziegler SF. Scurfin (FOXP3) acts as a repressor of transcription and regulates T cell activation. J Biol Chem. (2001) 276:37672–9. 10.1074/jbc.M104521200
    1. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. (2003) 4:330–6. 10.1038/ni904
    1. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science (2003) 299:1057–61. 10.1126/science.1079490
    1. Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. (2006) 24:209–26. 10.1146/annurev.immunol.24.021605.090547
    1. Bin Dhuban K, Piccirillo CA. The immunological and genetic basis of immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Curr Opin Allergy Clin Immunol. (2015) 15:525–32. 10.1097/ACI.0000000000000214
    1. Alroqi FJ, Chatila TA. T regulatory cell biology in health and disease. Curr Allergy Asthma Rep. (2016)16:27. 10.1007/s11882-016-0606-9
    1. Bacchetta R, Barzaghi F, Roncarolo MG. From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann NY Acad Sci. (2018) 1417:5–22. 10.1111/nyas.13011
    1. Komatsu N, Mariotti-Ferrandiz ME, Wang Y, Malissen B, Waldmann H, Hori S. Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity. Proc Natl Acad Sci USA. (2009) 106:1903–8. 10.1073/pnas.0811556106
    1. Himmel ME, MacDonald KG, Garcia R V, Steiner TS, Levings MK. Helios+ and Helios- cells coexist within the natural FOXP3+ T regulatory cell subset in humans. J Immunol. (2013) 190:2001–8. 10.4049/jimmunol.1201379
    1. Attridge K, Walker LSK. Homeostasis and function of regulatory T cells (Tregs) in vivo: lessons from TCR-transgenic Tregs. Immunol Rev. (2014) 259:23–39. 10.1111/imr.12165
    1. Campbell DJ. Control of regulatory T cell migration, function, and homeostasis. J Immunol. (2015) 195:2507–13. 10.4049/jimmunol.1500801
    1. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, et al. . CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. (2006) 203:1701–11. 10.1084/jem.20060772
    1. Palomares O, Martín-Fontecha M, Lauener R, Traidl-Hoffmann C, Cavkaytar O, Akdis M, et al. . Regulatory T cells and immune regulation of allergic diseases: roles of IL-10 and TGF-β. Genes Immun. (2014) 15:511–20. 10.1038/gene.2014.45
    1. Allenspach E, Torgerson TR. Autoimmunity and primary immunodeficiency disorders. J Clin Immunol. (2016) 36:57–67. 10.1007/s10875-016-0294-1
    1. 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) 8:1353–62. 10.1038/ni1536
    1. Kanai T, Jenks J, Nadeau KC. The STAT5b pathway defect and autoimmunity. Front Immunol. (2012) 3:234. 10.3389/fimmu.2012.00234
    1. Goudy K, Aydin D, Barzaghi F, Gambineri E, Vignoli M, Mannurita SC, et al. . Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol. (2013) 146:248–61. 10.1016/j.clim.2013.01.004
    1. Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, et al. . Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science (2014) 345:1623–7. 10.1126/science.1255904
    1. Charbonnier LM, Janssen E, Chou J, Ohsumi TK, Keles S, Hsu JT, et al. . 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. (2015) 135:217–27. 10.1016/j.jaci.2014.10.019
    1. Angulo I, Vadas O, Garçon F, Banham-Hall E, Plagnol V, Leahy TR, et al. . Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage. Science (2013) 342:866–71. 10.1126/science.1243292
    1. Uzel G, Sampaio EP, Lawrence MG, Hsu AP, Hackett M, Dorsey MJ, 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) 131:1611–23. 10.1016/j.jaci.2012.11.054
    1. Deau MC, Heurtier L, Frange P, Suarez F, Bole-Feysot C, Nitschke P, et al. . A human immunodeficiency caused by mutations in the PIK3R1 gene. J Clin Invest. (2014) 124:3923–8. 10.1172/JCI75746
    1. Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, et al. . Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat Immunol. (2014) 15:88–97. 10.1038/ni.2771
    1. Haapaniemi EM, Kaustio M, Rajala HLM, Van Adrichem AJ, Kainulainen L, Glumoff V, et al. . Autoimmunity, hypogammaglobulinemia, lymphoproliferation, and mycobacterial disease in patients with activating mutations in STAT3. Blood (2015) 125:639–48. 10.1182/blood-2014-04-570101
    1. Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y, et al. . Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. (2015) 48:67–73. 10.1038/ng.3459
    1. Bennett CL. A rare polyadenylation signal mutation of the FOXP3 gene (AAUAAA → AAUGAA) leads to the IPEX syndrome. Immunogenetics (2001) 53:435–9. 10.1007/s002510100358
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. . The genome analysis toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. (2010) 20:1297–303. 10.1101/gr.107524.110
    1. Yang H, Wang K. Genomic variant annotation and prioritization with ANNOVAR and wANNOVAR. Nat Protoc. (2015) 10:1556–66. 10.1038/nprot.2015.105
    1. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, 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) 132:1705–17. 10.1053/j.gastro.2007.02.044
    1. Burroughs LM, Torgerson TR, Storb R, Carpenter PA, Rawlings DJ, Sanders J, et al. . Stable hematopoietic cell engraftment after low-intensity nonmyeloablative conditioning in patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. J Allergy Clin Immunol. (2010) 126:1000–5. 10.1016/j.jaci.2010.05.021
    1. Fuchizawa T, Adachi Y, Ito Y, Higashiyama H, Kanegane H, Futatani T, et al. . Developmental changes of FOXP3-expressing CD4+CD25+ regulatory T cells and their impairment in patients with FOXP3 gene mutations. Clin Immunol. (2007) 125:237–46. 10.1016/j.clim.2007.08.004
    1. Otsubo K, Kanegane H, Kamachi Y, Kobayashi I, Tsuge I, Imaizumi M, et al. . Identification of FOXP3-negative regulatory T-like (CD4 +CD25 +CD127 low) cells in patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Clin Immunol. (2011) 141:111–20. 10.1016/j.clim.2011.06.006
    1. Kobayashi I, Kubota M, Yamada M, Tanaka H, Itoh S, Sasahara Y, et al. . Autoantibodies to villin occur frequently in IPEX, a severe immune dysregulation, syndrome caused by mutation of FOXP3. Clin Immunol. (2011) 141:83–9. 10.1016/j.clim.2011.05.010
    1. Moudgil A, Perriello P, Loechelt B, Przygodzki R, Fitzerald W, Kamani N. Immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome: an unusual cause of proteinuria in infancy. Pediatr Nephrol. (2007) 22:1799–802. 10.1007/s00467-007-0532-0
    1. De Benedetti F, Insalaco A, Diamanti A, Cortis E, Muratori F, Lamioni A, et al. . Mechanistic associations of a mild phenotype of immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Clin Gastroenterol Hepatol. (2006) 4:653–9. 10.1016/j.cgh.2005.12.014
    1. Okou DT, Mondal K, Faubion WA, Kobrynski LJ, Denson LA, Mulle JG, et al. . Exome sequencing identifies a novel FOXP3 mutation in a 2-generation family with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. (2014) 58:561–8. 10.1097/MPG.0000000000000302
    1. Gambineri E, Perroni L, Passerini L, Bianchi L, Doglioni C, Meschi F, et al. . Clinical and molecular profile of a new series of patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome: Inconsistent correlation between forkhead box protein 3 expression and disease severity. J Allergy Clin Immunol. (2008) 122:1105–12. 10.1016/j.jaci.2008.09.027
    1. Baris S, Schulze I, Ozen A, Aydiner EK, Altuncu E, Karasu GT, et al. . Clinical heterogeneity of immunodysregulation, polyendocrinopathy, enteropathy, X-linked: pulmonary involvement as a non-classical disease manifestation. J Clin Immunol. (2014) 34:601–6. 10.1007/s10875-014-0059-7
    1. Chen CA, Chung WC, Chiou YY, Yang YJ, Lin YC, Ochs HD, et al. . Quantitative analysis of tissue inflammation and responses to treatment in immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, and review of literature. J Microbiol Immunol Infect. (2016) 49:775–82. 10.1016/j.jmii.2015.10.015
    1. Lopes JE, Torgerson TR, Schubert LA, Anover SD, Ocheltree EL, Ochs HD, et al. . Analysis of FOXP3 reveals multiple domains required for its function as a transcriptional repressor. J Immunol. (2006) 177:3133–42. 10.4049/jimmunol.177.5.3133
    1. Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, X linked (IPEX) syndrome. J Med Genet. (2002) 39:537–45. 10.1136/jmg.39.8.537
    1. Hashimura Y, Nozu K, Kanegane H, Miyawaki T, Hayakawa A, Yoshikawa N, et al. . Minimal change nephrotic syndrome associated with immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Pediatr Nephrol. (2009) 24:1181–6. 10.1007/s00467-009-1119-8
    1. Li B, Samanta A, Song X, Iacono KT, Brennan P, Chatila TA, 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) 19:825–35. 10.1093/intimm/dxm043
    1. Nademi Z, Slatter M, Gambineri E, Mannurita SC, Barge D, Hodges S, et al. . Single centre experience of haematopoietic SCT for patients with immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Bone Marrow Transplant. (2014) 49:310–2. 10.1038/bmt.2013.181
    1. Passerini L, Olek S, Di Nunzio S, Barzaghi F, Hambleton S, Abinun M, et al. . Forkhead box protein 3 (FOXP3) mutations lead to increased TH17 cell numbers and regulatory T-cell instability. J Allergy Clin Immunol. (2011) 128:1376–9. 10.1016/j.jaci.2011.09.010
    1. Harbuz R, Lespinasse J, Boulet S, Francannet C, Creveaux I, Benkhelifa M, et al. . Identification of new FOXP3 mutations and prenatal diagnosis of IPEX syndrome. Prenat Diagn. (2010) 30:1072–8. 10.1002/pd.2613
    1. Tsuda M, Torgerson TR, Selmi C, Gambineri E, Carneiro-Sampaio M, Mannurita SC, et al. . The spectrum of autoantibodies in IPEX syndrome is broad and includes anti-mitochondrial autoantibodies. J Autoimmun. (2010) 35:265–8. 10.1016/j.jaut.2010.06.017
    1. Bacchetta R, Passerini L, Gambineri E, Dai M, Allan SE, Perroni L, et al. . Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest. (2006) 116:1713–22. 10.1172/JCI25112
    1. McMurchy AN, Gillies J, Allan SE, Passerini L, Gambineri E, Roncarolo MG, et al. . Point mutants of forkhead box P3 that cause immune dysregulation, polyendocrinopathy, enteropathy, X-linked have diverse abilities to reprogram T cells into regulatory T cells. J Allergy Clin Immunol. (2010) 126:1242–51. 10.1016/j.jaci.2010.09.001
    1. Rubio-Cabezas O, Minton JAL, Caswell R, Shield JP, Deiss D, Sumnik Z, et al. . Clinical heterogeneity in patients with FOXP3 mutations presenting with permanent neonatal diabetes. Diabetes Care (2009) 32:111–6. 10.2337/dc08-1188
    1. Savova R, Arshinkova M, Houghton J, Konstantinova M, Gaydarova M, Georgieva E, et al. . Clinical case of immune dysregulation, polyendocrinopaty, enteropathy, X-linked (IPEX) syndrome with severe immune deficiency and late onset of endocrinopathy and enteropathy. Case Rep Med. (2014) 2014:1–5. 10.1155/2014/564926
    1. Tuijnenburg P, Cuadrado E, Bosch AM, Kindermann A, Jansen MH, Alders M, et al. . Humoral Immunodeficiency with hypotonia, feeding difficulties, enteropathy, and mild eczema caused by a classical FOXP3 mutation. Front Pediatr. (2017) 5:37. 10.3389/fped.2017.00037
    1. Colobran R, Álvarez de la Campa E, Soler-Palacín P, Martín-Nalda A, Pujol-Borrell R, de la Cruz X, et al. . Clinical and structural impact of mutations affecting the residue Phe367 of FOXP3 in patients with IPEX syndrome. Clin Immunol. (2016) 163:60–5. 10.1016/j.clim.2015.12.014
    1. An YF, Xu F, Wang M, Zhang ZY, Zhao XD. Clinical and molecular characteristics of immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome in China. Scand J Immunol. (2011) 74:304–9. 10.1111/j.1365-3083.2011.02574.x
    1. Nieves DS, Phipps RP, Pollock SJ, Ochs HD, Zhu Q, Scott GA, et al. . Dermatologic and immunologic findings in the immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Arch Dermatol. (2004) 140:466–72. 10.1001/archderm.140.4.466
    1. d'Hennezel E, Ben-Shoshan M, Ochs HD, Torgerson TR, Russell LJ, Lejtenyi C, et al. . FOXP3 forkhead domain mutation and regulatory T cells in the IPEX syndrome. N Engl J Med. (2009) 361:1710–3. 10.1056/NEJMc0907093
    1. Myers a K, Perroni L, Costigan C, Reardon W. Clinical and molecular findings in IPEX syndrome. Arch Dis Child. (2006) 91:63–4. 10.1136/adc.2005.078287
    1. Banerjee-Basu S, Baxevanis AD. Structural analysis of disease-causing mutations in the P-subfamily of forkhead transcription factors. Proteins Struct Funct Genet. (2004) 54:639–47. 10.1002/prot.10621
    1. Rao A, Kamani N, Filipovich A, Lee SM, Davies SM, Dalal J, et al. . Successful bone marrow transplantation for IPEX syndrome after reduced-intensity conditioning. Blood (2007) 109:383–5. 10.1182/blood-2006-05-025072
    1. Alkorta-Aranburu G, Carmody D, Cheng YW, Nelakuditi V, Ma L, Dickens JT, et al. . Phenotypic heterogeneity in monogenic diabetes: the clinical and diagnostic utility of a gene panel-based next-generation sequencing approach. Mol Genet Metab. (2014) 113:315–20. 10.1016/j.ymgme.2014.09.007
    1. Dorsey MJ, Petrovic A, Morrow MR, Dishaw LJ, Sleasman JW. FOXP3 expression following bone marrow transplantation for IPEX syndrome after reduced-intensity conditioning. Immunol Res. (2009) 44:179–84. 10.1007/s12026-009-8112-y
    1. Stray-Pedersen A, Sorte HS, Samarakoon P, Gambin T, Chinn IK, Coban Akdemir ZH, et al. . Primary immunodeficiency diseases: genomic approaches delineate heterogeneous Mendelian disorders. J Allergy Clin Immunol. (2017) 139:232–45. 10.1016/j.jaci.2016.05.042
    1. Takezaki S, Yamada M, Kato M, Park M-J, Maruyama K, Yamazaki Y, et al. . Chronic mucocutaneous candidiasis caused by a gain-of-function mutation in the STAT1 DNA-binding domain. J Immunol. (2012) 189:1521–6. 10.4049/jimmunol.1200926
    1. Toubiana J, Okada S, Hiller J, Oleastro M, Gomez ML, Becerra JCA, et al. . Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood (2016) 127:3154–64. 10.1182/blood-2015-11-679902
    1. Leiding JW, Okada S, Hagin D, Abinun M, Shcherbina A, Balashov DN, et al. . Hematopoietic stem cell transplantation in patients with gain-of-function signal transducer and activator of transcription 1 mutations. J Allergy Clin Immunol. (2018) 141:704–17. 10.1016/j.jaci.2017.03.049
    1. Hwa V, Camacho-Hübner C, Little BM, David A, Metherell LA, El-Khatib N, et al. . 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) 68:218–24. 10.1159/000101334
    1. Chen R, Giliani S, Lanzi G, Mias GI, Lonardi S, Dobbs K, et al. . Whole-exome sequencing identifies tetratricopeptide repeat domain 7A (TTC7A) mutations for combined immunodeficiency with intestinal atresias. J Allergy Clin Immunol. (2013) 132:656–664. 10.1016/j.jaci.2013.06.013
    1. Dinwiddie DL, Kingsmore SF, Caracciolo S, Rossi G, Moratto D, Mazza C, et al. . Combined DOCK8 and CLEC7A mutations causing immunodeficiency in 3 brothers with diarrhea, eczema, and infections. J Allergy Clin Immunol. (2013) 131:594–7.e3. 10.1016/j.jaci.2012.10.062
    1. Allan SE, Passerini L, Bacchetta R, Crellin N, Dai M, Orban PC, et al. . The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest. (2005) 115:3276–84. 10.1172/JCI24685
    1. Manavalan JS, Kim-Schulze S, Scotto L, Naiyer AJ, Vlad G, Colombo PC, et al. . Alloantigen specific CD8+CD28– FOXP3+ T suppressor cells induce ILT3+ ILT4+ tolerogenic endothelial cells, inhibiting alloreactivity. Int Immunol. (2004) 16:1055–68. 10.1093/intimm/dxh107
    1. Yagi H, Nomura T, Nakamura K, Yamazaki S, Kitawaki T, Hori S, et al. . Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol. (2004) 16:1643–56. 10.1093/intimm/dxh165
    1. Aarts-Riemens T, Emmelot ME, Verdonck LF, Mutis T. Forced overexpression of either of the two common human Foxp3 isoforms can iduce regulatory T cells from CD4+CD25- cells. Eur J Immunol. (2008) 38:1381–1390. 10.1002/eji.200737590
    1. Kristensen B, Hegedüs L, Madsen HO, Smith TJ, Nielsen CH. Altered balance between self-reactive T helper (Th)17 cells and Th10 cells and between full-length forkhead box protein 3 (FoxP3) and FoxP3 splice variants in Hashimoto's thyroiditis. Clin Exp Immunol. (2015) 180:58–69. 10.1111/cei.12557
    1. Serena G, Yan S, Camhi S, Patel S, Lima RS, Sapone A, et al. . Proinflammatory cytokine interferon-γ and microbiome-derived metabolites dictate epigenetic switch between forkhead box protein 3 isoforms in coeliac disease. Clin Exp Immunol. (2017) 187:490–506. 10.1111/cei.12911
    1. Smith EL, Finney HM, Nesbitt AM, Ramsdell F, Robinson MK. Splice variants of human FOXP3 are functional inhibitors of human CD4 + T-cell activation. Immunology (2006) 119:203–11. 10.1111/j.1365-2567.2006.02425.x
    1. Mailer RKW. Alternative splicing of FOXP3-virtue and vice. Front Immunol. (2018) 9:530. 10.3389/fimmu.2018.00530
    1. Curinha A, Braz SO, Pereira-Castro I, Cruz A, Moreira A. Implications of polyadenylation in health and disease. Nucleus (2014) 5:508–19. 10.4161/nucl.36360
    1. Alkhairy OK, Abolhassani H, Rezaei N, Fang M, Andersen KK, Chavoshzadeh Z, et al. . Spectrum of phenotypes associated with mutations in LRBA. J Clin Immunol. (2016) 36:33–45. 10.1007/s10875-015-0224-7
    1. Slatter MA, Engelhardt KR, Burroughs LM, Arkwright PD, Nademi Z, Skoda-Smith S, et al. . Hematopoietic stem cell transplantation for CTLA4 deficiency. J Allergy Clin Immunol. (2016) 138:615–619.e1. 10.1016/j.jaci.2016.01.045
    1. Rickert M, Wang X, Boulanger MJ, Goriatcheva N, Garcia KC. The structure of interleukin-2 complexed with its alpha rece. Science (2005) 2057:1477–81. 10.1126/science.1109745
    1. Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, et al. . Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. (2009) 361:2046–55. 10.1056/NEJMoa0905506
    1. Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. . Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol. (2009) 124:1289–302. 10.1016/j.jaci.2009.10.038
    1. Alroqi FJ, Charbonnier LM, Keles S, Ghandour F, Mouawad P, Sabouneh R, et al. . DOCK8 deficiency presenting as an IPEX-like disorder. J Clin Immunol. (2017) 37:811–9. 10.1007/s10875-017-0451-1
    1. Hartley JL, Zachos NC, Dawood B, Donowitz M, Forman J, Pollitt RJ, et al. . Mutations in TTC37 cause trichohepatoenteric syndrome (Phenotypic Diarrhea of Infancy). Gastroenterology (2010) 138:2388–98. 10.1053/j.gastro.2010.02.010
    1. Bettelli E, Dastrange M, Oukka M. Foxp3 interacts with nuclear factor of activated T cells and NF- kB to repress cytokine gene expression and effector functions of T helper cells. Proc Natl Acad Sci USA. (2005) 102:5138–43. 10.1073/pnas.0501675102
    1. Torgerson TR, Genin A, Chen C, Zhang M, Zhou B, Frank MB, et al. . FOXP3 inhibits activation-induced NFAT2 expression in T cells thereby limiting effector cytokine expression. J Immunol. (2009) 183:907–15. 10.4049/jimmunol.0800216
    1. Bandukwala HS, Wu Y, Feurer M, Chen Y, Ghosh S, Stroud JC, et al. . Structure of a domain-swapped FOXP3 dimer on DNA and its function in regulatory T cells. Immunity (2011) 34:479–91. 10.1016/j.immuni.2011.02.017
    1. Bin Dhuban K, D'Hennezel E, Nagai Y, Xiao Y, Shao S, Istomine R, et al. . Suppression by human FOXP3(+) regulatory T cells requires FOXP3-TIP60 interactions. Sci Immunol. (2017) 2:eaai9297. 10.1126/sciimmunol.aai9297
    1. Hsu AP, Fleisher TA, Niemela JE. Mutation analysis in primary immunodeficiency diseases: case studies. Curr Opin Allergy Clin Immunol. (2009) 9:517–24. 10.1097/ACI.0b013e3283328f59
    1. Higgs DR, Goodbourn SEY, Lamb J, Clegg JB, Weatherall DJ, Proudfoot NJ. α-Thalassaemia caused by a polyadenylation signal mutation. Nature (1983) 306:398–400. 10.1038/306398a0
    1. Jassim N, Al-Arrayed S, Gerard N, Al-Mukharraq H, Al-Ajami A, Ramasawmy R, et al. . A mismatched-primer polymerase chain reaction-restriction fragment length polymorphism strategy for rapid screening of the polyadenylation signal mutation α(T-Saudi) (AATAAA → AATAAG) in the α2-globin gene. Hemoglobin (1999) 23:213–20. 10.3109/03630269909005701
    1. Haddad F, Trabelsi N, Chaouch L, Darragi I, Oueslati M, Boudriga I, et al. . Homozygous mutation on the β-globin polyadenylation signal in a tunisian patient with β-thalassemia intermedia and coinheritance of Gilbert's syndrome. Hemoglobin (2017) 41:147–50. 10.1080/03630269.2017.1322610
    1. Losekoot M, Fodde R, Harteveld CL, van Heeren H, Giordano PC, Went LN, et al. . Homozygous beta+ thalassaemia owing to a mutation in the cleavage-polyadenylation sequence of the human beta globin gene. J Med Genet. (1991) 28:252–5. 10.1136/jmg.28.4.252
    1. Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol. (2012) 3:211. 10.3389/fimmu.2012.00211
    1. Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ, et al. . Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol. (2018) 141:1036–49. 10.1016/j.jaci.2017.10.041
    1. Ciullini Mannurita S, Gambineri E. Novel molecular defects associated with very early-onset inflammatory bowel. Curr Opin Allergy Clin Immunol. (2017) 17:317–24. 10.1097/ACI.0000000000000393
    1. Notarangelo LD. Multiple intestinal atresia with combined immune deficiency. Curr Opin Pediatr. (2014) 26:690–6. 10.1097/MOP.0000000000000159
    1. Lee W-I, Huang J-L, Chen C-C, Lin J-L, Wu R-C, Jaing T-H, et al. . Identifying mutations of the tetratricopeptide repeat domain 37 (TTC37) gene in infants with intractable diarrhea and a comparison of Asian and Non-Asian phenotype and genotype. Medicine (2016) 95:1–9. 10.1097/MD.0000000000002918

Source: PubMed

3
Abonnieren