Skip to main content

Gut Involvement in Cellular Immunodeficiencies

  • Chapter
  • First Online:
Cellular Primary Immunodeficiencies

Abstract

Primary cellular immunodeficiencies may often initially present with manifestations affecting the gastrointestinal tract. Not only infections but also inflammatory, autoimmune, and in some cases malignant manifestations may suggest the presence of a defect in the immune system.

Gastrointestinal manifestations are present in 5–50% of patients with primary immunodeficiency, which is not surprising since the intestine is the largest lymphoid organ.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Lai Ping So A, Mayer L (1997) Gastrointestinal manifestations of primary immunodeficiency disorders. Semin Gastrointest Dis 8(1):22–32

    CAS  PubMed  Google Scholar 

  2. Rosen FS (1986) Defects in cell-mediated immunity. Clin Immunol Immunopathol 41(1):1–7

    Article  CAS  PubMed  Google Scholar 

  3. Jarvis WR, Middleton PJ, Gelfand EW (1983) Significance of viral infections in severe combined immunodeficiency disease. Pediatr Infect Dis 2(3):187–192

    Article  CAS  PubMed  Google Scholar 

  4. Bakare N, Menschik D, Tiernan R, Hua W, Martin D (2010) Severe combined immunodeficiency (SCID) and rotavirus vaccination: reports to the Vaccine Adverse Events Reporting System (VAERS). Vaccine 28(40):6609–6612

    Article  PubMed  Google Scholar 

  5. Patil S, Rao RS, Majumdar B, Anil S (2015) Clinical appearance of oral candida infection and therapeutic strategies. Front Microbiol 6:1391

    Article  PubMed  PubMed Central  Google Scholar 

  6. Stephan JL, Vlekova V, Le Deist F, Blanche S, Donadieu J, De Saint-Basile G et al (1993) Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients. J Pediatr 123(4):564–572

    Article  CAS  PubMed  Google Scholar 

  7. Snover DC, Filipovich AH, Ramsay NK, Weisdorf SA, Kersey JH (1985) Graft-versus-host-disease-like histopathological findings in pre-bone-marrow transplantation biopsies of patients with severe T cell deficiency. Transplantation 39(1):95–97

    Article  CAS  PubMed  Google Scholar 

  8. Lee EY, Clouse RE, Aliperti G, DeSchryver-Kecskemeti K (1991) Small intestinal lesion resembling graft-vs-host disease. A case report in immunodeficiency and review of the literature. Arch Pathol Lab Med 115(5):529–532

    CAS  PubMed  Google Scholar 

  9. Washington K, Gossage DL, Gottfried MR (1993) Pathology of the liver in severe combined immunodeficiency and DiGeorge syndrome. Pediatr Pathol 13(4):485–504

    Article  CAS  PubMed  Google Scholar 

  10. Gilger MA, Matson DO, Conner ME, Rosenblatt HM, Finegold MJ, Estes MK (1992) Extraintestinal rotavirus infections in children with immunodeficiency. J Pediatr 120(6):912–917

    Article  CAS  PubMed  Google Scholar 

  11. Washington K, Gossage DL, Gottfried MR (1994) Pathology of the pancreas in severe combined immunodeficiency and DiGeorge syndrome: acute graft-versus-host disease and unusual viral infections. Hum Pathol 25(9):908–914

    Article  CAS  PubMed  Google Scholar 

  12. Sanal O, Jing H, Ozgur T, Ayvaz D, Strauss-Albee DM, Ersoy-Evans S et al (2012) Additional diverse findings expand the clinical presentation of DOCK8 deficiency. J Clin Immunol 32(4):698–708

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Engelhardt KR, Gertz ME, Keles S, Schäffer AA, Sigmund EC, Glocker C et al (2015) The extended clinical phenotype of 64 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol 136(2):402–412

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Alsum Z, Hawwari A, Alsmadi O, Al-Hissi S, Borrero E, Abu-Staiteh A et al (2013) Clinical, immunological and molecular characterization of DOCK8 and DOCK8-like deficient patients: single center experience of twenty-five patients. J Clin Immunol 33(1):55–67

    Article  CAS  PubMed  Google Scholar 

  15. Betts K, Abusleme L, Freeman AF, Sarmadi M, Fahle G, Pittaluga S et al (2015) A 17-year old patient with DOCK8 deficiency, severe oral HSV-1 and aggressive periodontitis—a case of virally induced periodontitis? J Clin Virol 63:46–50

    Article  CAS  PubMed  Google Scholar 

  16. Al-Herz W, Ragupathy R, Massaad MJ, Al-Attiyah R, Nanda A, Engelhardt KR et al (2012) Clinical, immunologic and genetic profiles of DOCK8-deficient patients in Kuwait. Clin Immunol 143(3):266–272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Shah T, Cale C, Hadzic N, Jones A (2014) Dedicator of cytokinesis 8 deficiency: a predisposition to sclerosing cholangitis. Clin Immunol 155(1):71–73

    Article  CAS  PubMed  Google Scholar 

  18. Al-Herz W, Chu JI, van der Spek J, Raghupathy R, Massaad MJ, Keles S et al (2016) Hematopoietic stem cell transplantation outcomes for 11 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol 138(3):852–859. e3

    Article  PubMed  PubMed Central  Google Scholar 

  19. Biggs CM, Keles S, Chatila TA (2017) DOCK8 deficiency: insights into pathophysiology, clinical features and management. Clin Immunol 181:75–82

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Alroqi FJ, Charbonnier LM, Keles S, Ghandour F, Mouawad P, Sabouneh R et al (2017) DOCK8 deficiency presenting as an IPEX-like disorder. J Clin Immunol 37(8):811–819

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Picard C, Casanova JL, Puel A (2011) Infectious diseases in patients with IRAK-4, MyD88, NEMO, or IκBα deficiency. Clin Microbiol Rev 24(3):490–497

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hanson EP, Monaco-Shawver L, Solt LA, Madge LA, Banerjee PP, May MJ et al (2008) Hypomorphic nuclear factor-kappaB essential modulator mutation database and reconstitution system identifies phenotypic and immunologic diversity. J Allergy Clin Immunol 122(6):1169–1177. e16

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cheng LE, Kanwar B, Tcheurekdjian H, Grenert JP, Muskat M, Heyman MB et al (2009) Persistent systemic inflammation and atypical enterocolitis in patients with NEMO syndrome. Clin Immunol 132(1):124–131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hönig M, Schwarz K (2006) Omenn syndrome: a lack of tolerance on the background of deficient lymphocyte development and maturation. Curr Opin Rheumatol 18(4):383–388

    Article  PubMed  Google Scholar 

  25. Puzenat E, Rohrlich P, Thierry P, Girardin P, Taghian M, Ouachee M et al (2007) Omenn syndrome: a rare case of neonatal erythroderma. Eur J Dermatol 17(2):137–139

    CAS  PubMed  Google Scholar 

  26. Desplantes C, Fremond ML, Beaupain B, Harousseau JL, Buzyn A, Pellier I et al (2014) Clinical spectrum and long-term follow-up of 14 cases with G6PC3 mutations from the French Severe Congenital Neutropenia Registry. Orphanet J Rare Dis 9:183

    Article  PubMed  PubMed Central  Google Scholar 

  27. Marciano BE, Rosenzweig SD, Kleiner DE, Anderson VL, Darnell DN, Anaya-O'Brien S et al (2004) Gastrointestinal involvement in chronic granulomatous disease. Pediatrics 114(2):462–468

    Article  PubMed  Google Scholar 

  28. Arnold DE, Heimall JR (2017) A review of chronic granulomatous disease. Adv Ther 34(12):2543–2557

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Marciano BE, Spalding C, Fitzgerald A, Mann D, Brown T, Osgood S et al (2015) Common severe infections in chronic granulomatous disease. Clin Infect Dis 60(8):1176–1183

    Article  CAS  PubMed  Google Scholar 

  30. McCabe RP (2002) Gastrointestinal Manifestations of Non-AIDS Immunodeficiency. Curr Treat Opt Gastroenterol 5(1):17–25

    Article  Google Scholar 

  31. Yu JE, De Ravin SS, Uzel G, Landers C, Targan S, Malech HL et al (2011) High levels of Crohn's disease-associated anti-microbial antibodies are present and independent of colitis in chronic granulomatous disease. Clin Immunol 138(1):14–22

    Article  CAS  PubMed  Google Scholar 

  32. Rieber N, Hector A, Kuijpers T, Roos D, Hartl D (2012) Current concepts of hyperinflammation in chronic granulomatous disease. Clin Dev Immunol 2012:252460

    Article  PubMed  Google Scholar 

  33. van den Berg JM, van Koppen E, Ahlin A, Belohradsky BH, Bernatowska E, Corbeel L et al (2009) Chronic granulomatous disease: the European experience. PLoS One 4(4):e5234

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Magnani A, Brosselin P, Beauté J, de Vergnes N, Mouy R, Debré M et al (2014) Inflammatory manifestations in a single-center cohort of patients with chronic granulomatous disease. J Allergy Clin Immunol 134(3):655–662. e8

    Article  PubMed  Google Scholar 

  35. Alimchandani M, Lai JP, Aung PP, Khangura S, Kamal N, Gallin JI et al (2013) Gastrointestinal histopathology in chronic granulomatous disease: a study of 87 patients. Am J Surg Pathol 37(9):1365–1372

    Article  PubMed  PubMed Central  Google Scholar 

  36. Angelino G, De Angelis P, Faraci S, Rea F, Romeo EF, Torroni F et al (2017) Inflammatory bowel disease in chronic granulomatous disease: an emerging problem over a twenty years’ experience. Pediatr Allergy Immunol 28(8):801–809

    Article  PubMed  Google Scholar 

  37. Winkelstein JA, Marino MC, Johnston RB, Boyle J, Curnutte J, Gallin JI et al (2000) Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore) 79(3):155–169

    Article  CAS  Google Scholar 

  38. Hussain N, Feld JJ, Kleiner DE, Hoofnagle JH, Garcia-Eulate R, Ahlawat S et al (2007) Hepatic abnormalities in patients with chronic granulomatous disease. Hepatology 45(3):675–683

    Article  PubMed  Google Scholar 

  39. Feld JJ, Hussain N, Wright EC, Kleiner DE, Hoofnagle JH, Ahlawat S et al (2008) Hepatic involvement and portal hypertension predict mortality in chronic granulomatous disease. Gastroenterology 134(7):1917–1926

    Article  PubMed  Google Scholar 

  40. Etzioni A (2009) Genetic etiologies of leukocyte adhesion defects. Curr Opin Immunol 21(5):481–486

    Article  CAS  PubMed  Google Scholar 

  41. Levy-Mendelovich S, Rechavi E, Abuzaitoun O, Vernitsky H, Simon AJ, Lev A et al (2016) Highlighting the problematic reliance on CD18 for diagnosing leukocyte adhesion deficiency type 1. Immunol Res 64(2):476–482

    Article  CAS  PubMed  Google Scholar 

  42. Roberts MW, Atkinson JC (1990) Oral manifestations associated with leukocyte adhesion deficiency: a five-year case study. Pediatr Dent 12(2):107–111

    CAS  PubMed  Google Scholar 

  43. Wolach B, Gavrieli R, Wolach O, Stauber T, Abuzaitoun O, Kuperman A et al (2019) Leucocyte adhesion deficiency—a multicentre national experience. Eur J Clin Investig 49(2):e13047

    Article  CAS  Google Scholar 

  44. D'Agata ID, Paradis K, Chad Z, Bonny Y, Seidman E (1996) Leucocyte adhesion deficiency presenting as a chronic ileocolitis. Gut 39(4):605–608

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Uzel G, Kleiner DE, Kuhns DB, Holland SM (2001) Dysfunctional LAD-1 neutrophils and colitis. Gastroenterology 121(4):958–964

    Article  CAS  PubMed  Google Scholar 

  46. Jain S, Gozdziak P, Morgan A, Burt RK (2013) Remission of Crohn's disease after cord blood transplantation for leukocyte adhesion deficiency type 1. Bone Marrow Transplant 48(7):1006–1007

    Article  CAS  PubMed  Google Scholar 

  47. Chatila TA, Blaeser F, Ho N, Lederman HM, Voulgaropoulos C, Helms C et al (2000) JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest 106(12):R75–R81

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wildin RS, Smyk-Pearson S, Filipovich AH (2002) Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, X linked (IPEX) syndrome. J Med Genet 39(8):537–545

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F et al (2007) Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology 132(5):1705–1717

    Article  CAS  PubMed  Google Scholar 

  50. Patey-Mariaud de Serre N, Canioni D, Ganousse S, Rieux-Laucat F, Goulet O, Ruemmele F et al (2009) Digestive histopathological presentation of IPEX syndrome. Mod Pathol 22(1):95–102

    Article  CAS  PubMed  Google Scholar 

  51. Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ et al (2018) Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol 141(3):1036–1049. e5

    Article  PubMed  Google Scholar 

  52. Bennett CL, Ochs HD (2001) IPEX is a unique X-linked syndrome characterized by immune dysfunction, polyendocrinopathy, enteropathy, and a variety of autoimmune phenomena. Curr Opin Pediatr 13(6):533–538

    Article  CAS  PubMed  Google Scholar 

  53. Yong PL, Russo P, Sullivan KE (2008) Use of sirolimus in IPEX and IPEX-like children. J Clin Immunol 28(5):581–587

    Article  CAS  PubMed  Google Scholar 

  54. Kobayashi I, Kawamura N, Okano M (2001) A long-term survivor with the immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. N Engl J Med 345(13):999–1000

    Article  CAS  PubMed  Google Scholar 

  55. Gambineri E, Ciullini Mannurita S, Robertson H, Vignoli M, Haugk B, Lionetti P et al (2015) Gut immune reconstitution in immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome after hematopoietic stem cell transplantation. J Allergy Clin Immunol 135(1):260–262

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Rao A, Kamani N, Filipovich A, Lee SM, Davies SM, Dalal J et al (2007) Successful bone marrow transplantation for IPEX syndrome after reduced-intensity conditioning. Blood 109(1):383–385

    Article  CAS  PubMed  Google Scholar 

  57. 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(2):482–487

    Article  CAS  PubMed  Google Scholar 

  58. Vignoli M, Ciullini Mannurita S, Fioravanti A, Tumino M, Grassi A, Guariso G et al (2019) CD25 deficiency: a new conformational mutation prevents the receptor expression on cell surface. Clin Immunol 201:15–19

    Article  CAS  PubMed  Google Scholar 

  59. Sharfe N, Dadi HK, Shahar M, Roifman CM (1997) Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor. Proc Natl Acad Sci U S A 94(7):3168–3171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Schwab C, Gabrysch A, Olbrich P, Patiño V, Warnatz K, Wolff D et al (2018) Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol 142(6):1932–1946

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A et al (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20(12):1410–1416

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. D'Elios MM, Rizzi M (2019) Humoral primary immunodeficiencies. Springer, Cham

    Book  Google Scholar 

  63. Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F et al (2009) Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med 361(21):2033–2045

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Glocker EO, Frede N, Perro M, Sebire N, Elawad M, Shah N et al (2010) Infant colitis—it's in the genes. Lancet 376(9748):1272

    Article  PubMed  Google Scholar 

  65. Engelhardt KR, Shah N, Faizura-Yeop I, Kocacik Uygun DF, Frede N, Muise AM et al (2013) Clinical outcome in IL-10- and IL-10 receptor-deficient patients with or without hematopoietic stem cell transplantation. J Allergy Clin Immunol 131(3):825–830

    Article  CAS  PubMed  Google Scholar 

  66. Kotlarz D, Beier R, Murugan D, Diestelhorst J, Jensen O, Boztug K et al (2012) Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology 143(2):347–355

    Article  CAS  PubMed  Google Scholar 

  67. Li Y, Führer M, Bahrami E, Socha P, Klaudel-Dreszler M, Bouzidi A et al (2019) Human RIPK1 deficiency causes combined immunodeficiency and inflammatory bowel diseases. Proc Natl Acad Sci U S A 116(3):970–975

    Article  CAS  PubMed  Google Scholar 

  68. Cuchet-Lourenço D, Eletto D, Wu C, Plagnol V, Papapietro O, Curtis J et al (2018) Biallelic. Science 361(6404):810–813

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Boland BS, Widjaja CE, Banno A, Zhang B, Kim SH, Stoven S et al (2015) Immunodeficiency and autoimmune enterocolopathy linked to NFAT5 haploinsufficiency. J Immunol 194(6):2551–2560

    Article  CAS  PubMed  Google Scholar 

  70. Kotlarz D, Marquardt B, Barøy T, Lee WS, Konnikova L, Hollizeck S et al (2018) Human TGF-β1 deficiency causes severe inflammatory bowel disease and encephalopathy. Nat Genet 50(3):344–348

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Blaydon DC, Biancheri P, Di WL, Plagnol V, Cabral RM, Brooke MA et al (2011) Inflammatory skin and bowel disease linked to ADAM17 deletion. N Engl J Med 365(16):1502–1508

    Article  CAS  PubMed  Google Scholar 

  72. Aguilar C, Lenoir C, Lambert N, Bègue B, Brousse N, Canioni D et al (2014) Characterization of Crohn disease in X-linked inhibitor of apoptosis-deficient male patients and female symptomatic carriers. J Allergy Clin Immunol 134(5):1131–1141. e9

    Article  CAS  PubMed  Google Scholar 

  73. Marsh RA, Madden L, Kitchen BJ, Mody R, McClimon B, Jordan MB et al (2010) XIAP deficiency: a unique primary immunodeficiency best classified as X-linked familial hemophagocytic lymphohistiocytosis and not as X-linked lymphoproliferative disease. Blood 116(7):1079–1082

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yang X, Kanegane H, Nishida N, Imamura T, Hamamoto K, Miyashita R et al (2012) Clinical and genetic characteristics of XIAP deficiency in Japan. J Clin Immunol 32(3):411–420

    Article  CAS  PubMed  Google Scholar 

  75. Worthey EA, Mayer AN, Syverson GD, Helbling D, Bonacci BB, Decker B et al (2011) Making a definitive diagnosis: successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet Med 13(3):255–262

    Article  PubMed  Google Scholar 

  76. Girardelli M, Arrigo S, Barabino A, Loganes C, Morreale G, Crovella S et al (2015) The diagnostic challenge of very early-onset enterocolitis in an infant with XIAP deficiency. BMC Pediatr 15:208

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Dziadzio M, Ammann S, Canning C, Boyle F, Hassan A, Cale C et al (2015) Symptomatic males and female carriers in a large Caucasian kindred with XIAP deficiency. J Clin Immunol 35(5):439–444

    Article  CAS  PubMed  Google Scholar 

  78. Tsuma Y, Imamura T, Ichise E, Sakamoto K, Ouchi K, Osone S et al (2015) Successful treatment of idiopathic colitis related to XIAP deficiency with allo-HSCT using reduced-intensity conditioning. Pediatr Transplant 19(1):E25–E28

    Article  PubMed  Google Scholar 

  79. Candotti F (2018) Clinical Manifestations and Pathophysiological Mechanisms of the Wiskott-Aldrich Syndrome. J Clin Immunol 38(1):13–27

    Article  CAS  PubMed  Google Scholar 

  80. Dupuis-Girod S, Medioni J, Haddad E, Quartier P, Cavazzana-Calvo M, Le Deist F et al (2003) Autoimmunity in Wiskott-Aldrich syndrome: risk factors, clinical features, and outcome in a single-center cohort of 55 patients. Pediatrics 111(5 Pt 1):e622–e627

    Article  PubMed  Google Scholar 

  81. Folwaczny C, Ruelfs C, Walther J, König A, Emmerich B (2002) Ulcerative colitis in a patient with Wiskott-Aldrich syndrome. Endoscopy 34(10):840–841

    Article  CAS  PubMed  Google Scholar 

  82. Hsieh KH, Chang MH, Lee CY, Wang CY (1988) Wiskott-Aldrich syndrome and inflammatory bowel disease. Ann Allergy 60(5):429–431

    CAS  PubMed  Google Scholar 

  83. Esmaeilzadeh H, Bordbar MR, Dastsooz H, Silawi M, Fard MAF, Adib A et al (2018) A novel splice site mutation in WAS gene in patient with Wiskott-Aldrich syndrome and chronic colitis: a case report. BMC Med Genet 19(1):123

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Root AW, Speicher CE (1963) The triad of thrombocytopenia, eczema, and recurrent infections (Wiskott-Aldrich syndrome) associated with milk antibodies, giant-cell pneumonia, and cytomegalic inclusion disease. Pediatrics 31:444–454

    Article  CAS  PubMed  Google Scholar 

  85. Orange JS, Stone KD, Turvey SE, Krzewski K (2004) The Wiskott-Aldrich syndrome. Cell Mol Life Sci 61(18):2361–2385

    Article  CAS  PubMed  Google Scholar 

  86. Ohya T, Yanagimachi M, Iwasawa K, Umetsu S, Sogo T, Inui A et al (2017) Childhood-onset inflammatory bowel diseases associated with mutation of Wiskott-Aldrich syndrome protein gene. World J Gastroenterol 23(48):8544–8552

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Buchbinder D, Nugent DJ, Fillipovich AH (2014) Wiskott-Aldrich syndrome: diagnosis, current management, and emerging treatments. Appl Clin Genet 7:55–66

    Article  PubMed  PubMed Central  Google Scholar 

  88. Sullivan KE, Mullen CA, Blaese RM, Winkelstein JA (1994) A multiinstitutional survey of the Wiskott-Aldrich syndrome. J Pediatr 125(6 Pt 1):876–885

    Article  CAS  PubMed  Google Scholar 

  89. McDonald-McGinn DM (2018) 22q11.2 deletion syndrome: a tiny piece leading to a big picture. Am J Med Genet A 176(10):2055–2057

    Article  PubMed  PubMed Central  Google Scholar 

  90. Digilio MC, Giannotti A, Castro M, Colistro F, Ferretti F, Marino B et al (2003) Screening for celiac disease in patients with deletion 22q11.2 (DiGeorge/velo-cardio-facial syndrome). Am J Med Genet A 121A(3):286–288

    Article  PubMed  Google Scholar 

  91. Giardino G, Cirillo E, Maio F, Gallo V, Esposito T, Naddei R et al (2014) Gastrointestinal involvement in patients affected with 22q11.2 deletion syndrome. Scand J Gastroenterol 49(3):274–279

    Article  PubMed  Google Scholar 

  92. Markert ML, Sarzotti M, Ozaki DA, Sempowski GD, Rhein ME, Hale LP et al (2003) Thymus transplantation in complete DiGeorge syndrome: immunologic and safety evaluations in 12 patients. Blood 102(3):1121–1130

    Article  CAS  PubMed  Google Scholar 

  93. Digilio MC, Marino B, Bagolan P, Giannotti A, Dallapiccola B (1999) Microdeletion 22q11 and oesophageal atresia. J Med Genet 36(2):137–139

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Kilic SS, Gurpinar A, Yakut T, Egeli U, Dogruyol H (2003) Esophageal atresia and tracheo-esophageal fistula in a patient with Digeorge syndrome. J Pediatr Surg 38(8):E21–E23

    Article  PubMed  Google Scholar 

  95. Campbell IM, Sheppard SE, Crowley TB, McGinn DE, Bailey A, McGinn MJ et al (2018) What is new with 22q? An update from the 22q and You Center at the Children's Hospital of Philadelphia. Am J Med Genet A 176(10):2058–2069

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Jackson O, Crowley TB, Sharkus R, Smith R, Jeong S, Solot C et al (2019) Palatal evaluation and treatment in 22q11.2 deletion syndrome. Am J Med Genet A 179(7):1184–1195

    CAS  PubMed  Google Scholar 

  97. Humbert L, Cornu M, Proust-Lemoine E, Bayry J, Wemeau JL, Vantyghem MC et al (2018) Chronic mucocutaneous candidiasis in autoimmune polyendocrine syndrome type 1. Front Immunol 9:2570

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mario Milco D’Elios .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Troilo, A., Frede, N., Della Bella, C., D’Elios, M.M. (2021). Gut Involvement in Cellular Immunodeficiencies. In: D'Elios, M.M., Baldari, C.T., Annunziato, F. (eds) Cellular Primary Immunodeficiencies. Rare Diseases of the Immune System. Springer, Cham. https://doi.org/10.1007/978-3-030-70107-9_13

Download citation

  • DOI: https://doi.org/10.1007/978-3-030-70107-9_13

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-030-70106-2

  • Online ISBN: 978-3-030-70107-9

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics